At the beginning of the 21st century, diarrheal disease continues to be a significant cause of morbidity and mortality worldwide. During the period of 1986 to 2000, an estimated 1.4 billion children younger than 5 years suffered an episode of acute diarrhea every year in developing countries; among these, 123.6 million required outpatient medical care, and 9 million required hospitalization. Approximately 2 million diarrhea-associated deaths occurred in this age group annually, primarily in the most impoverished areas of the world.1 These estimates are somewhat lower than the more than 3 million annual deaths from diarrhea reported in the prior 10 years,2 indicating progress in prevention and treatment of acute diarrhea. In the United States, approximately 400 childhood deaths per year were reported during the late 1980s,3, 4 although the actual number may be higher.4
Accurate incidence rates for acute diarrhea in neonates from different populations are not readily available. The relative sparing of the newborn probably results from low exposure to enteropathogens and protection associated with breast-feeding.5, 6, 7, 8 After the first few months of life, increasing interaction with other individuals and the environment, including introduction of artificial feeding, increases the risk of exposure to enteropathogens. For most pathogens, the incidence of acute diarrhea peaks in children between 6 months and 4 years old.9 Neonatal diarrhea is more common in underdeveloped areas, where low educational levels, crowding, and poor standards of medical care, environmental sanitation, and personal hygiene favor early contact with enteropathogens. As the incidence of neonatal gastroenteritis rises, there is a proportional increase in neonatal deaths because medical care for the poor often is inadequate.10, 11 For very low birth weight infants (<1500g), the death rate from diarrhea is 100-fold greater than for higher-birth-weight infants.12
This chapter discusses the pathogenesis, diagnosis, treatment, and prevention of gastroenteritis based on the available knowledge about pathogens that can cause neonatal diarrhea. Pathogens that rarely or never cause acute diarrhea in neonates are not discussed. After an overview of host defense mechanisms and protective factors in human milk, the remainder of the chapter is devoted to specific pathogens that cause inflammatory or noninflammatory diarrhea.
ENTERIC HOST DEFENSE MECHANISMS
The neonate is a host that is uniquely susceptible to enteric infections. Neonates have not had the opportunity to develop local or systemic immune responses, and in the first few days of life, they have not acquired the highly important enteric flora that protects the normal adult gastrointestinal tract.13, 14, 15, 16, 17, 18 Still less is known about the barrier effect of the neonate’s gastric acidity,19 intestinal mucus,20 or motility,21, 22 each of which provides protection against gastrointestinal tract infections in older infants, children, and adults.
The gastric acid barrier appears to be least effective during the first months of life. The average gastric pH level of the newborn is high (pH 4 to 7; mean, 6).23, 24 Although the pH falls to low levels by the end of the first day of life (pH 2 to 3),23 it subsequently rises again; by 7 to 10 days of life, the hydrochloric acid output of the neonatal stomach is far less than that of older infants and children.24, 25 The buffering action of frequent milk feedings and the short gastric emptying time26, 27, 28, 29 interpose additional factors in the neonate that would be expected to permit viable ingested organisms to reach the small intestine.
The intestinal epithelium serves as a nutrient absorptive machine, barrier to pathogen entry, and regulator of inflammation. Intestinal epithelial cells have receptors for bacterial products and produce chemokines (e.g., interleukin [IL]-8, monocyte chemotactic protein type 1 [MCP-1], granulocyte macrophage-cell stimulating factor [GM-CSF]) and proinflammatory cytokines (e.g., IL-6, tumor necrosis factor-α [TNF-α], IL-1) in response to invasion by enteropathogens.30 The gut epithelium orchestrates the immune response. However, in the newborn, phagocytic, chemotactic, and complement functions are immature. B and T lymphocyte functions are impaired, resulting in a preferential IgM production in response to antigenic stimulation. IgG is actively transferred from mother to infant across the placenta at about 32 weeks’ gestation and peaks by about 37 weeks; premature neonates, especially those born before 28 weeks’ gestation, are deficient in these maternally derived serum antibodies.31
PROTECTIVE FACTORS IN HUMAN MILK
The importance of breast-feeding infants for the prevention of diarrheal disease has long been emphasized.13, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45 Published studies reporting the association between breast-feeding and diarrhea are extensive and suggest that infants who are breast-fed suffer fewer episodes of diarrhea than those who are not. This protection is greatest during a child’s first 3 months of life and declines with increasing age. During the period of weaning, partial breast-feeding confers protection that is intermediate between that gained by infants who are exclusively breast-fed and that by those who are exclusively bottle-fed.
A striking demonstration of the protection afforded by breast-feeding of newborns has been provided by Mata and Urrutia13 in their studies of a population of infants born in a rural Guatemalan village. Despite extremely poor sanitation and the demonstration of fecal organisms in the colostrum and milk of almost one third of mothers,46 diarrheal disease did not occur in any newborns. The incidence of diarrhea rose significantly only after these infants reached 4 to 6 months old, at which time solids and other fluids were used to supplement the human milk feedings. At that time, Escherichia coli and gram-negative anaerobes (e.g., Bacteroides) were found to colonize the intestinal tract.13 In contrast, urban infants of a similar ethnic background who were partly or totally artificially fed frequently acquired diarrheal disease caused by enteropathogenic E. coli (EPEC).
Multiple mechanisms by which breast-feeding protects against diarrhea have been postulated. Breast-feeding confers protection by active components in milk and by decreased exposure to organisms present on or in contaminated bottles, food, or water. Many protective components have been identified in human milk and generally are classified as belonging to the major categories of cells, antibody, anti-inflammatory factors, and glycoconjugates and other nonantibody factors.47, 48, 49, 50 Examples of milk antibodies are summarized in Table 20–1. For any given pathogen, multiple milk factors may help protect the infant. Human milk typically targets a major pathogenic mechanism using multiple, redundant strategies. Redundancy of milk protective factors and targeting of complex virulence machinery have created a formidable barrier to enteropathogens. Despite the fact that pathogens can rapidly divide and mutate, milk continues to protect infants. For example, human milk has secretory antibodies to Shigella virulence antigens and lipopolysaccharides,51, 52 neutral glycolipid Gb3 to bind Shiga toxin,53, 54 and lactoferrin to disrupt and degrade the surface-expressed virulence antigens.55, 56, 57 In a similar way, milk contains antibodies directed toward the surface expressed virulence antigens of EPEC,58 oligosaccharides that block cell attachment,59 and lactoferrin that disrupts and degrades the surface expressed EPEC antigens.60 Human milk can initiate and maintain the growth of Bifidobacterium and low pH in the feces of newborn infants, creating an environment antagonistic to the growth of E. coli.13, 17, 18, 61
Table 20-1.
Association between Antibodies in Human Milk and Protection against Enteropathogens
| Organism | Antibody |
|---|---|
| Vibrio cholerae | Lipopolysaccharide, enterotoxin |
| Campylobacter jejuni | Surface proteins |
| Enteropathogenic Escherichia coli | Adherence proteins |
| Enterotoxigenic E. coli | Enterotoxin, adherence proteins |
| Shigatoxin producing E. coli | Adherence proteins |
| Shigella | Lipopolysaccharide, virulence plasmid–associated antigens |
| Giardia lamblia | Surface proteins |
The protective effect of human milk antibodies against enteropathogen-specific disease has been described for Vibrio cholerae, 62 Campylobacter jejuni, 63 EPEC,59 enterotoxigenic E. coli (ETEC),64, 65 Shigella, 66, 67 and Giardia lamblia 68, 69 and for bovine milk concentrate against ETEC,70 rotavirus,71 and Shigella. 72
In 1933, the nonlactose carbohydrate fraction of human milk was found to consist mainly of oligosaccharides.73 In 1960, Montreuil and Mullet74 determined that up to 2.4% of colostrum and up to 1.3% of mature milk are oligosaccharides. Human milk contains a larger quantity of the oligosaccharides than does milk from other mammals, and its composition is singularly complex.75 The metabolic fate of the oligosaccharides is of interest. Only water, lactose, and lipids are present in greater amounts than the oligosaccharides. Despite the fact that substantial energy must be expended by the mother to synthesize the many hundreds of different milk oligosaccharides, the infant does not use them as food. Most of the oligosaccharides pass through the gut undigested.76, 77 It is thought that they are present primarily to serve as receptor analogues that misdirect enteropathogen attachment factors away from gut epithelial carbohydrate receptors. Likewise, enteropathogens use the oligosaccharide portion of glycolipids and glycoproteins as targets for attachment of whole bacteria and toxins. Evidence is emerging that these glycoconjugates may have an important role in protection of the breast-fed infant from disease.48
Human milk protects suckling mice from the heat-stable enterotoxin (ST) of E. coli; on the basis of its chemical stability and physical properties, the protective factor has been deduced to be a neutral fucosyloligosaccharide.79, 80 Experiments have shown that EPEC attachment to HEp-2 cells can be inhibited by purified oligosaccharide fractions from human milk.59 Oligosaccharides also may be relevant to protection from Norwalk virus and other caliciviruses, because these viruses attach to human ABO, Lewis, and secretor blood group antigens.80, 81 Human milk contains large amounts of these carbohydrates. The ganglioside fraction in human milk has been shown to inhibit the action of heat-labile toxin (LT) and cholera toxin on ileal loops more effectively than secretory IgA.82, 83 Lactadherin in human milk has been shown to bind rotavirus and to inhibit viral replication in vitro and in vivo.84 A study of infants in Mexico showed that lactadherin in human milk protected infants from symptoms of rotavirus infection.72
ESCHERICHIA COLI
E. coli organisms promptly colonize the lower intestinal tracts of healthy infants in their first few days of life85, 86, 87, 88 and constitute the predominant aerobic coliform fecal flora throughout life in humans and in many animals. The concept that this species might cause enteric disease was first suggested in the late 19th and early 20th centuries, when several veterinary workers described the association of diarrhea (i.e., scours) in newborn calves with certain strains of E. coli. 89, 90, 91, 92, 93, 94
In 1905, Moro95 observed that Bacterium (now Escherichia) coli was found more often in the small intestines of children with diarrhea than in children without diarrhea. Adam96, 97 confirmed these findings and noted the similarity with Asiatic cholera and calf scours. He further extended these observations by suggesting that E. coli strains from patients with diarrhea could be distinguished from normal coliform flora by certain sugar fermentation patterns. Although he called these disease-producing organisms dyspepsicoli and introduced the important concept that E. coli could cause enteric disease, biochemical reactions have not proved to be a reliable means of distinguishing nonpathogenic from pathogenic E. coli strains. There are now at least six recognized enteric pathotypes of E. coli. 98 The pathotypes can be distinguished clinically, epidemiologically, and pathogenetically (Table 20–2).98, 99, 100, 101, 102, 103, 104
Table 20-2.
Predominant Serogroups, Mechanisms, and Gene Codes Associated with Enterotoxigenic, Enteroinvasive, Enteropathogenic, Enterohemorrhagic, and Enteroaggregative Escherichia coli
| ETEC | EIEC | EPEC | EHEC | EAEC |
|---|---|---|---|---|
| Class I Serogroup | ||||
| LT | O28ac | O157:H7 | O3:H2 | |
| O6:K15 | O29, O112 | O55:K59 (B5) | O26:H11/H- | O44 |
| O8:K40 | O115, O124 | O111ab:K88 (B4) | O128, O103:H2 | O78:H33 |
| O136, O144 | O119K6a (B14) | O39 | O15:H11 | |
| LT and ST | O147, O152 | O125ac:K70 (B15) | O111:K58:H8/H- | O77:H18 |
| O11:H27 | O164 | O126:K71 (B16) | O113:K75:H7/H21 | O51:H11 |
| O15, O20:K79 | O127a:K63 (B8) | O121:H-, O145:H- | And many others | |
| O25:K7 | O128abc:K67 (B12) | Rough | ||
| O27, O63 | O142, O158 | And many others | ||
| O80, O85, O139 | ||||
| Class II Serogroup | ||||
| ST | ||||
| O groups 78, 115 | O44:K74 | |||
| 128, 148, 149, 153 | O86a:K61 (B7) | |||
| 159, 166, 167 | O114:H2 | |||
| Mechanisms | ||||
| Adenylate or guanylate cyclase activation | Colonic invasiveness (e.g., Shigella) | Localized attachment and effacement | Shiga toxins block protein synthesis; attachment and effacement | Aggregative adherence and toxins |
| Gene Codes | ||||
| Plasmid | Chromosomal and plasmid | Chromosomal and plasmid | Phage and chromosomal | Plasmid and chromosomal |
| ETEC, enterotoxigenic Escherichia coli; EIEC, enteroinvasive E. coli; EPEC, enteropathogenic E. coli; EHEC, enterohemorrhagic E. coli; EAEC, enteroaggregative E. coli; LT, heat-labile toxin; ST, heat-stable toxin. | ||||
ETEC organisms are defined by their ability to secrete the LT or the ST enterotoxin, or both. LT is closely related to cholera toxin and similarly acts by means of intestinal adenylate cyclase,105, 106 prostaglandin synthesis,107, 108 and possibly platelet activating factor.109, 110 ST (particularly the variant STa) causes secretion by specifically activating intestinal mucosal guanylate cyclase.111, 112, 113 The STb toxin causes noncyclic, nucleotide-mediated bicarbonate secretion and appears to be important only in animals.114, 115, 116 Enteroinvasive E. coli (EIEC) has the capacity to invade the intestinal mucosa, thereby causing an inflammatory enteritis much like shigellosis.117, 118 EPEC elicits diarrhea by a signal transduction mechanism,98, 99, 100, 101, 102, 119, 120 which is accompanied by a characteristic attaching-and-effacing histopathologic lesion in the small intestine.121 Enterohemorrhagic E. coli (EHEC) also induces an attaching-and-effacing lesion, but in the colon.98 EHEC also secretes Shiga toxin, which gives rise to the sequela of hemolytic-uremic syndrome (HUS). Diffusely adherent E. coli 122 executes a signal transduction effect, which is accompanied by the induction of long cellular processes.123 Enteroaggregative E. coli (EAEC) adheres to the intestinal mucosa and elaborates enterotoxins and cytotoxins.98, 103, 125
A major problem in the recognition of ETEC, EIEC, EPEC, and EHEC strains of E. coli is that they are indistinguishable from normal coliform flora of the intestinal tract by the usual bacteriologic methods. Serotyping is of value in recognizing EPEC serotypes126 and EIEC, because these organisms tend to fall into a limited number of specific serogroups (see Table 20–2).126, 127 EIEC invasiveness is confirmed by inoculating fresh isolates into guinea pig conjunctivae, as described by Sereny.128 The ability of organisms to produce enterotoxins (LT or ST) is encoded by a transmissible plasmid that can be lost by one strain of E. coli or transferred to a previously unrecognized strain.129, 130, 131 Although the enterotoxin plasmids appear to prefer certain serogroups (different from EPEC or invasive serogroups),132 ETEC is not expected to be strictly limited to a particular set of serogroups. Instead, these strains can be recognized only by examining for the enterotoxin. This is done in ligated animal loops,133 in tissue culture,134, 135 or by enzyme-linked immunosorbent assay (ELISA)136 for LT or in suckling mice for ST.137, 138 Specific DNA probes also are available for LT and ST.98 Whether there are other mechanisms involved in the ability of the versatile E. coli species to cause enteric disease, such as by producing other types of enterotoxins139 or by fimbriate adherence traits alone,140, 141 remains to be elucidated.
Enterotoxigenic Escherichia coli
Although early work on the recognition of E. coli as a potential enteric pathogen focused on biochemical or serologic distinctions, there followed a shift in emphasis to the enterotoxins produced by previously recognized and entirely “new” strains of E. coli. Beginning in the mid-1950s with work by De and colleagues142, 143 in Calcutta, E. coli strains from patients with diarrhea were found to cause a fluid secretory response in ligated rabbit ileal loops analogous to that seen with V. cholerae. Work by Taylor and associates144, 145 showed that the viable E. coli strains were not required to produce this secretory response and that this enterotoxin production correlated poorly with classically recognized EPEC serotypes. In São Paulo, Trabulsi146 made similar observations with E. coli isolated from children with diarrhea, and several veterinary workers demonstrated that ETEC was associated with diarrhea in piglets and calves.147, 148, 149, 150 A similar pattern was described in 1971 with acute undifferentiated diarrhea in adults in Bengal from whom E. coli could be isolated from the upper small bowel only during acute illness.151, 152 These strains of E. coli produced a nondialyzable, LT, ammonium sulfate–precipitable enterotoxin.153 Analogous to the usually short-lived diarrheal illnesses of E. coli reported by several workers, a short-lived course of the secretory response to E. coli culture filtrates compared with the secretory response of cholera toxin was described.154 However, like responses to cholera toxin, secretory responses to E. coli were associated with activation of intestinal mucosal adenylate cyclase that paralleled the fluid secretory response.155, 156
The two types of enterotoxins produced by E. coli 157, 158, 159 have been found to be plasmid-encoded traits that are potentially separable from each other and from the equally important plasmid-encoded adherence traits for pathogenesis.129, 130, 131, 160 ST causes an immediate and reversible secretory response,133 whereas the effects of LT (e.g., cholera toxin) follow a lag period necessitated by its intracellular site of action.105, 106, 134 Only LT appears to cause fluid secretion by activating adenylate cyclase, which is accomplished by toxin-induced ADP-ribosylation of the Gsα signaling protein.98, 105 The activation of adenylate cyclase by LT and by cholera toxin is highly promiscuous, occurring in many cell types and resulting in development of nonintestinal tissue culture assay systems such as the Chinese hamster ovary (CHO) cell assay134 and Y1 adrenal cell assay.135 The antigenic similarity of LT and cholera toxin and their apparent binding to the monosialoganglioside GM1 have enabled development of ELISAs for detection of LT and cholera toxin.138, 161, 162, 163
ST is a much smaller molecule and is distinct antigenically from LT and cholera toxin.134, 137, 138 Although it fails to alter cAMP levels, ST increases intracellular intestinal mucosal cyclic guanosine monophosphate (cGMP) concentrations and specifically activates plasma membrane–associated intestinal guanylate cyclase.111, 112, 113 Like cAMP analogues, cGMP analogues cause intestinal secretion that mimics the response to ST.111 The receptor for STa responds to an endogenous ligand called guanylin, of which STa is a structural homologue.164 Because the capacity to produce an enterotoxin may be transmissible between different organisms by a plasmid or even a bacteriophage,129, 130, 131 interstrain gene transfer may be expected to be responsible for occasional toxigenic non–E. coli. Enterotoxigenic Klebsiella and Citrobacter strains have been associated with diarrhea in a few reports, often in the same patients with ETEC.165, 166 Likewise, certain strains of Salmonella appear to produce an LT, CHO cell– positive toxin that may play a similar role in the pathogenesis of the watery, noninflammatory diarrhea sometimes seen with Salmonella enteritidis infection.167, 168 At least equally important as enterotoxigenicity for E. coli to cause disease is the ability of these organisms to colonize the upper small bowel, where the enterotoxin produced has its greatest effect. A separable, plasmid-encoded colonization trait was first recognized in porcine E. coli. Veterinary workers demonstrated that the fimbriate K-88 surface antigen was necessary for ETEC to cause disease in piglets.160 An autosomal dominant allele appears to be responsible for the specific intestinal receptor in piglets. In elegant studies by Gibbons and co-workers,169 the homozygous recessive piglets lacked the receptor for K-88 and were resistant to scours caused by ETEC. At least 15 analogous colonization factors have been described for human E. coli isolates98, 170, 171 against which local IgA antibody may be produced. These antigens potentially may be useful in vaccine development.
Epidemiology and Transmission
Data on the epidemiology and transmission of ETEC remain scanty for the neonatal period. In the past 2 decades, these strains have been recognized among adults with endemic, cholera-like diarrhea in Calcutta, India, and in Dacca, Bangladesh,105, 151 and among travelers to areas such as Mexico and Central Africa.173, 174, 175
The isolation of ETEC is uncommon in sporadic diarrheal illnesses in temperate climates where sanitation facilities are good and where winter viral patterns of diarrhea predominate. ETEC is commonly isolated from infants and children with acute watery summer diarrhea in areas where sanitary facilities are less than optimal.35, 165, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187 These include areas such as Africa,165 Brazil,35, 175, 181, 186, 187 Argentina,177 Bengal,178, 179 Mexico,180 and Native American reservations in the southwestern United States.182, 183 In a multicenter study of acute diarrhea in 3640 infants and children in China, India, Mexico, Myanmar, and Pakistan, 16% of cases (versus 5% of 3279 controls) had ETEC.184 A case-control study from northwestern Spain showed a highly significant association of ETEC with 26.5% of neonatal diarrhea, often acquired in the hospital.185 Although all types of ETEC (LT and/or ST producers) are associated with cholera-like, non inflammatory, watery diarrhea in adults in these areas, they probably constitute the major cause (along with rotaviruses) of dehydrating diarrhea in infants and young children in these areas. In this setting, peaks of illnesses tend to occur in the summer or rainy season, and dehydrating illnesses may be life threatening, especially in infants and young children.35, 181, 186 Humans are probably the major reservoirs for the human strains of ETEC, and contaminated food and water probably constitute the principal vectors.188, 189 Although antitoxic immunity to LT and asymptomatic infection with LT-producing E. coli tends to increase with age, ST is poorly immunogenic, and ST-producing E. coli continues to be associated with symptomatic illnesses into adulthood in endemic areas.183, 187
The association of ETEC with outbreaks of diarrhea in newborn nurseries is well documented. Ryder and colleagues190 isolated an ST-producing E. coli from 72% of infants with diarrhea, from the environment, and in one instance, from an infant’s formula during a 7-month period in a prolonged outbreak in a special care nursery in Texas. Another ST-producing E. coli outbreak was reported in 1976 by Gross and associates191 from a maternity hospital in Scotland. ETEC and EPEC were significantly associated with diarrhea among infants younger than 1 year in Bangladesh.192
An outbreak of diarrhea in a newborn special care nursery that was associated with enterotoxigenic organisms that were not limited to the same serotype or even the same species has been reported.193 The short-lived ETEC, Klebsiella, and Citrobacter species in this outbreak raised the possibility that each infant’s indigenous bowel flora might become transiently toxigenic, possibly by receiving the LT genome from a plasmid or even a bacteriophage.
Clinical Manifestations
The clinical manifestations of ETEC diarrhea tend to be mild and self-limited, except in small or undernourished infants, in whom dehydration may constitute a major threat to life. In many parts of the developing world, acute diarrheal illnesses are the leading recognized causes of death. There is some suggestion that the diarrheal illnesses associated with ST-producing ETEC may be particularly severe.179 Most probably the best definition of the clinical manifestations of ETEC infection comes from volunteer studies with adults. Ingestion of 108 to 1010 human ETEC isolates that produce LT and ST or ST alone resulted in a 30% to 80% attack rate of mild to moderate diarrheal illnesses within 12 to 56 hours that lasted 1 to 3 days.117 These illnesses, typical for traveler’s diarrhea, were manifested by malaise, anorexia, abdominal cramps, and sometimes explosive diarrhea. Nausea and vomiting occur relatively infrequently, and up to one third of patients may have a low-grade fever. Although illnesses usually resolve spontaneously within 1 to 5 days, they occasionally may persist for 1 week or longer. The diarrhea is noninflammatory, without fecal leukocytes or blood. In outbreaks in infants and neonates, the duration has been in the same range (1 to 11 days), with a mean of approximately 4 days.
Pathology
As in cholera, the pathologic changes associated with ETEC infection are minimal. From animal experiments in which Thiry-Vella loops were infected with these organisms and at a time when the secretory and adenylate cyclase responses were present, there was only a mild discharge of mucus from goblet cells and otherwise no significant pathologic change in the intestinal tract.106 Unless terminal complications of severe hypotension ensue, ETEC organisms rarely disseminate beyond the intestinal tract. Like cholera, ETEC diarrhea is typically limited to being an intraluminal infection.
Diagnosis
The preliminary diagnosis of ETEC diarrhea can be suspected by the epidemiologic setting and the noninflammatory nature of stool specimens, which reveal few or no leukocytes. Although the ability of E. coli to produce enterotoxins may be lost or transmitted to other strains, there is a tendency for the enterotoxin plasmids to occur among certain predominant serotypes, as shown in Table 20–2.132 These serotypes differ from EPEC or invasive serotypes, but their demonstration does not prove that they are enterotoxigenic. The only definitive way to identify ETEC is to demonstrate the enterotoxin itself by a specific gene probe for the toxin codon, by a bioassay such as tissue culture or ileal loop assays for LT or the suckling mouse assay for ST, or in the case of LT, by immunoassay such as ELISA. However, even these sensitive bioassays are limited by the unavailability of any selective media for detecting ETEC by culture. Even though substantial improvements have been made in enterotoxin assay (particularly for LT), the necessary random selection of E. coli from a relatively nonselective stool culture plate resulted in a sensitivity of only 43% of epidemiologically incriminated cases in an outbreak when 5 to 10 isolates were randomly picked and tested for enterotoxigenicity.189 By also examining paired serum samples for antibody against LT, only 36% demonstrated significant serum antibody titer rises, for a total sensitivity of ETEC isolation or serum antibody titer rises of only 64%. Some have suggested that isolates may be pooled for LT or ST assay. The capacity to prove with radiolabeled or enzyme-tagged oligonucleotide gene sequences for the enterotoxins (LT or ST) further facilitates the identification of enterotoxigenic organisms.194, 195 A novel method of combining immunomagnetic separation (using antibody-coated magnetic beads) followed by DNA or polymerase chain reaction (PCR) probing may enhance the sensitivity of screening fecal or food specimens for ETEC or other pathogens.196, 197
Therapy and Prevention
The mainstay of treatment of any diarrheal illness is rehydration.198 This principle especially pertains to ETEC diarrhea, which is an intraluminal infection. The glucose absorptive mechanism remains intact in E. coli enterotoxin-induced secretion, much as it does in cholera, a concept that has resulted in the major advance of oral glucose-electrolyte therapy. This regimen can usually provide fully adequate rehydration in infants and children able to tolerate oral fluids, replacing the need for parenteral rehydration in most cases.199, 200 Its use is particularly critical in rural areas and developing nations, where early application before dehydration becomes severe may be lifesaving.
The standard World Health Organization solution contains 3.5g NaCl, 2.5g NaHCO3, 1.5g KCl, and 20g glucose per liter of clean or boiled drinking water.198 This corresponds to the following concentrations: 90mmol/L of sodium, 20mmol/L of potassium, 30mmol/L of bicarbonate, 80mmol/L of chloride, and 110mmol/L of glucose. A variety of recipes for homemade preparations have been described,201 but unless the cost is prohibitive, the premade standard solution is preferred. Each 4 ounces of this solution should be followed by 2 ounces of plain water. If there is concern about hypertonicity, especially in small infants in whom a high intake and constant direct supervision of feeding cannot be ensured, the concentration of salt can be reduced.202 A reduced osmolality solution with 60 mmol/L of sodium and 84mmol/L of glucose and a total osmolality of 224 (instead of 311) mOsm/kg has been found to reduce stool output by 28% and illness duration by 18% in a multicenter trial involving 447 children in four countries.203 Commercially available rehydration solutions are increasingly available worldwide.198
The role of antimicrobial agents in the treatment or prevention of ETEC is controversial. This infection usually resolves within 3 to 5 days in the absence of antibacterial therapy.198 Moreover, there is concern about the potential for coexistence of enterotoxigenicity and antibiotic resistance on the same plasmid, and co-transfer of multiple antibiotic resistance and enterotoxigenicity has been well documented.204 Widespread use of prophylactic antibiotics in areas where antimicrobial resistance is common has the potential for selecting for rather than against enterotoxigenic organisms. The prevention and control of ETEC infections would be similar to those discussed under EPEC serotypes. The use of breast-feeding should be encouraged.
Enteroinvasive Escherichia coli
EIEC causes diarrhea by means of Shigella-like intestinal epithelial invasion (discussed later).117, 118 The somatic antigens of these invasive strains have been identified and seem to fall into 1 of 10 recognized O groups (see Table 20–2). Most, if not all, of these bacteria share cell wall antigens with one or another of the various Shigella serotypes and produce positive reactions with antisera against the cross-reacting antigen.118 However, not all strains of E. coli belonging to the 10 serogroups associated with dysentery-like illness are pathogenic, because a large (140 MDa) invasive plasmid is also required.205 Additional biologic tests, including the guinea pig conjunctivitis (Sereny) test or a gene probe for the plasmid, are used to confirm the property of invasiveness.117
Although an outbreak of foodborne EIEC diarrhea has been well documented among adults who ate an imported cheese,118 little is known about the epidemiology and transmission of this organism, especially in newborns and infants. Whether the infectious dose may be as low as it is for Shigella is unknown; however, studies of adult volunteers suggest that attack rates may be somewhat lower after ingestion of even large numbers of EIEC than would be expected with Shigella. The outbreak of EIEC diarrhea resulted in a dysentery-like syndrome with an inflammatory exudate in stool and invasion and disruption of colonic mucosa.118 Descriptions of extensive and severe ileocolitis in infants dying with E. coli diarrhea indicate that neonatal disease also can be caused by invasive strains capable of mimicking the pathologic features of shigellosis.206 The immunofluorescent demonstration of E. coli together with an acute inflammatory infiltrate207 in the intestinal tissue of infants tends to support this impression, although it has been suggested that the organisms may have invaded the bowel wall in the postmortem period.117 There is still little direct evidence concerning the role of invasive strains of E. coli in the cause of neonatal diarrhea.175 The infrequency with which newborns manifest a dysentery-like syndrome makes it unlikely that this pathogen is responsible for a very large proportion of the diarrheal disease that occurs during the first month of life.
The diagnosis should be suspected in infants who have an inflammatory diarrhea as evidenced by fecal polymorphonuclear neutrophils or even bloody dysenteric syndromes from whom no other invasive pathogens, such as Campylobacter, Shigella, Salmonella, Vibrio, or Yersinia, can be isolated. In this instance, it may be appropriate to have the fecal E. coli isolated and serotyped or tested for invasiveness in the Sereny test. Plasmid pattern analysis and chromosomal restriction endonuclease digestion pattern analysis by pulsed-field gel electrophoresis have been used to evaluate strains involved in outbreaks.208 The management and prevention of EIEC diarrhea should be similar to those for acute Shigella or other E. coli enteric infections.
Enteropathogenic Escherichia coli: Classic Serotypes
The serologic distinction of E. coli strains associated with epidemic and sporadic infantile diarrhea was first suggested by Goldschmidt in 1933209 and confirmed by Dulaney and Michelson in 1935.205 These researchers found that certain strains of E. coli associated with institutional outbreaks of diarrhea would agglutinate with antisera on slides. In 1943, Bray211 isolated a serologically homogeneous strain of E. coli (subsequently identified as serogroup O111) from 95% of infants with summer diarrhea in England. He subsequently summarized a larger experience with this organism isolated from only 4% of asymptomatic controls but from 88% of infants with diarrhea, one half of which was hospital acquired.212 This strain (initially called E. coli-gomez by Varela in 1946) also was associated with infantile diarrhea in Mexico.213 A second type of E. coli (called beta by Giles in 1948 and subsequently identified as O55) was associated with an outbreak of infantile diarrhea in Aberdeen, Scotland.214, 215
From this early work primarily with epidemic diarrhea in infants has developed an elaborate serotyping system for certain E. coli strains that were clearly associated with infantile diarrhea.216, 217, 218 These strains first were called enteropathogenic E. coli by Neter and colleagues219 in 1955, and the association with particular serotypes can still be observed.220 As shown in Table 20–1, these organisms are distinct from the enterotoxigenic or enteroinvasive organisms or those that inhabit the normal gastrointestinal tract. They exhibit localized adherence to HEp-2 cells, a phenotype that has been suggested to be useful for diagnosis and pathogenesis research.119
Epidemiology and Transmission
EPEC is an important cause of diarrhea in infants in developing or transitional countries.98, 221, 222, 223 Outbreaks have become rare in the United States and other industrialized countries, but they still occur.224 Some have attributed the rarity of this recognition of illness in part to the declining severity of diarrheal disease caused by EPEC within the past 30 years, resulting in fewer cultures being obtained from infants with relatively mild symptoms.98, 225 However, several other variables influence the apparent incidence of this disease in the community. A problem arises with false-positive EPEC on the basis of the nonspecific cross-reactions seen with improper shortening of the serotyping procedure.226, 227 Because of their complexity and relatively low yield, neither slide agglutination nor HEp-2 cell adherence or DNA probe tests are provided as part of the routine identification of enteric pathogens by most clinical bacteriology laboratories. Failure to recognize the presence of EPEC in fecal specimens is the inevitable consequence.
The apparent incidence of EPEC gastroenteritis also varies with the epidemiologic circumstances under which stool cultures are obtained. The prevalence of enteropathogenic strains is higher among infants from whom cultures are obtained during a community epidemic compared with those obtained during sporadic diarrheal disease. Neither reflects the incidence of EPEC infection among infants involved in a nursery outbreak or hospital epidemic.
EPEC gastroenteritis is a worldwide problem, and socioeconomic conditions play a significant role in determining the incidence of this disease in different populations.228 For instance, it is unusual for newborn infants born in a rural environment to manifest diarrheal disease caused by EPEC; most infections of the gastrointestinal tract in these infants occur after the first 6 months of life.5, 229 Conversely, among infants born in large cities, the attack rate of EPEC is high during the first 3 months of life. This age distribution reflects in large part the frequency with which EPEC causes cross-infection outbreaks among nursery populations191, 230, 231, 232, 233, 234, 235, 236, 237; however, a predominance of EPEC in infants in the first 3 months of life also has been described in community epidemics238, 239, 240 and among sporadic cases of diarrhea acquired outside the hospital.241, 242, 243, 244, 245, 246, 247 The disparity in the incidence of neonatal EPEC infection between rural and urban populations has been ascribed to two factors: the trend away from breast-feeding among mothers in industrialized societies and the crowding together of susceptible newborns in nurseries in countries in which hospital deliveries predominate over home deliveries.5, 229, 248 Although the predominant serogroup can vary from year to year,239, 242, 243, 246, 249, 250 the same strains have been prevalent during the past 40 years in Great Britain,251 Puerto Rico,252 Guatemala,5 Panama,205 Israel,247 Newfoundland,240 Indonesia,244 Thailand,254 Uganda,255 and South Africa.256
When living conditions are poor and overcrowding of susceptible infants exists, there is a rise in the incidence of neonatal diarrhea in general257 and EPEC gastroenteritis in particular.215, 238, 258 A higher incidence of asymptomatic family carriers is found in such situations.238, 239
Newborn infants can acquire EPEC during the first days of life by one of several routes: (1) organisms from the mother ingested at the time of birth; (2) bacteria from other infants or toddlers with diarrheal disease or from asymptomatic adults colonized with the organism, commonly transmitted on the hands of nursery personnel or parents; (3) airborne or droplet infection; (4) fomites; or (5) organisms present in formulas or solid food supplements.259 Only the first two routes have been shown conclusively to be of any real significance in the transmission of disease or the propagation of epidemics.
Most neonates acquire EPEC at the time of delivery through ingestion of organisms residing in the maternal birth canal or rectum. Stool cultures taken from women before, during, or shortly after delivery have shown that 10% to 15% carry EPEC at some time during this period.85, 86, 88, 260, 261 Use of fluorescent antibody techniques261 or cultures during a community outbreak of EPEC gastroenteritis88 revealed twice this number of persons excreting the organism. Virtually none of the women carrying pathogenic strains of E. coli had symptoms referable to the gastrointestinal tract.
Many of the mothers whose stools contain EPEC transmit these organisms to their infants,85, 88 resulting in an asymptomatic infection rate of 2% to 5% among newborns cultured at random in nursery surveys.85, 86, 191, 262 These results must be considered conservative and are probably an artifact of the sampling technique. One study using 150 O antisera to identify as many E. coli as possible in fecal cultures showed a correlation between the coliform flora in 66% of mother-infant pairs.263 Of particular interest was the observation that the O groups of E. coli isolated from the infants’ mucus immediately after delivery correlated with those subsequently recovered from their stools, supporting the contention that these organisms were acquired orally at the time of birth. In mothers whose stools contained the same O group as their offspring, the mean time from rupture of membranes to delivery was about 2 hours longer than in those whose infants did not acquire the same serogroups, suggesting that ascending colonization before birth also can play a role in determining the newborn’s fecal flora.
The contours of the epidemiologic curves in nursery238, 264, 265, 266, 267, 268, 269 and community238, 239, 240 outbreaks are in keeping with a contact mode of spread. Transmission of organisms from infant to infant takes place by way of the fecal-oral route in almost all cases, most likely on the hands of persons attending to their care.86, 267, 269, 270 Ill infants represent the greatest risk to those around them because of the large numbers of organisms found in their stools271, 272, 273, 274 and vomitus.275, 276, 277 Cross-infection also has been initiated by infants who were healthy at the time of their admission to the nursery.264, 272, 278, 279, 280
A newborn exposed to EPEC is likely to acquire enteric infection if contact with a person excreting the organism is intimate and prolonged, as in a hospital or family setting. Stool culture surveys taken during outbreaks have shown that between 20% and 50% of term neonates residing in the nursery carry EPEC in their intestinal tracts.102, 230, 231, 234 Despite descriptions of nursery outbreaks in which virtually every neonate or low-birth-weight infant became infected,262, 264, 281 there is ample evidence that exposure to pathogenic strains of E. coli does not necessarily result in greater likelihood of illness for premature infants than for term infants.261, 272, 279, 282 Any increased prevalence of cross-infections that may exist among premature infants can be explained more readily by the prolonged hospital stays, their increased handling, and the clustering of infants born in different institutions than by a particular susceptibility to EPEC based on immature defense mechanisms.
The most extensive studies on the epidemiology of gastroenteritis related to E. coli have dealt with events that took place during outbreaks in newborn nurseries. Unfortunately, investigations of this sort frequently regard the epidemic as an isolated phenomenon and ignore the strong interdependence that exists between community- and hospital-acquired illness.280, 283, 284 Not surprisingly, the direction of spread is most often from the reservoir of disease within the community to the hospital. When the original source of a nursery outbreak can be established, frequently it is an infant born of a carrier mother who recently acquired her EPEC infection from a toddler living in the home. Cross-infection epidemics also can be initiated by infected newborns who have been admitted directly into a clean nursery unit from the surrounding district270, 272, 285 or have been transferred from a nearby hospital.278, 280, 286
After a nursery epidemic has begun, it generally follows one of two major patterns. Some are explosive, with rapid involvement of all susceptible infants and a duration that seldom exceeds 2 or 3 months.264, 265, 276, 287 The case-fatality rate in these epidemics may be very high. Other nursery outbreaks have an insidious onset with a few mild, unrecognized cases; the patients may not even develop illness until after discharge from the hospital. During the next few days to weeks, neonates with an increased number of loose stools are reported by the nurses; shortly thereafter, the appearance of the first severely ill infants makes it apparent that an epidemic has begun. Unless oral antimicrobial therapy is instituted (see “Therapy”), nursery outbreaks like these may continue for months266, 267, 268, 269 or years,270 with cycles of illness followed by periods of relative quiescence. This pattern can be caused by multiple strains (of different phage or antibiogram types) sequentially introduced into the nursery.278, 288, 289
The nursery can be a source of infection for the community. The release of infants who are in the incubation stages of their illness or are convalescent carriers about to relapse may lead to secondary cases of diarrheal disease among young siblings living in widely scattered areas.238, 239, 243 These children further disseminate infection to neighboring households, involving playmates of their own age, young infants, and mothers.238, 239, 242 As the sickest of these contact cases are admitted to different hospitals, they contaminate new susceptible persons, completing the cycle and compounding the outbreak. This feedback mechanism has proved to be a means of spreading infantile gastroenteritis through entire cities,238, 239, 242 counties,239, 285, 290 and even provinces.240 One major epidemic of diarrhea related to EPEC O111:B4 that occurred in the metropolitan Chicago and northwestern Indiana region during the winter of 1961 involved more than 1300 children and 29 community hospitals during a period of 9 months.240, 291 Almost all of the patients were younger than 2 years old, and 10% were younger than 1 month, producing an age-specific attack rate of nearly 4% of neonates in the community. The importance of the hospital as a source of cross-infection in this epidemic was demonstrated through interviews with patients’ families, indicating that a minimum of 40% of infants had direct or indirect contact with a hospital shortly before the onset of their illness.
It has been suggested, but not proved, that asymptomatic carriers of EPEC in close contact with a newborn infant, such as nursery personnel or family members, might play an important role in its transmission.280, 284, 292 Stool culture surveys have shown that at any one time about 1% of adults242, 293 and 1% to 5% of young children230, 238, 243 who are free of illness harbor EPEC strains. Higher percentages have been recorded during community epidemics.238, 239, 243 Because this intestinal carriage is transitory,238, 280 the number of individuals who excrete EPEC at one time or another during the year is far higher than the 1% figure recorded for single specimens.280, 293
Nursery personnel feed, bathe, and diaper a constantly changing population of newborns, about 2% to 5% of whom excrete EPEC.238, 280 Despite this constant exposure, intestinal carriage among nursery workers is surprisingly low. Even during outbreaks of diarrheal illness, when dissemination of organisms is most intense, less than 5% of the hospital personnel in direct contact with infected neonates are themselves excreting pathogenic strains of E. coli. 291, 294, 295
Although adult asymptomatic carriers generally excrete fewer organisms than patients with acute illness do,272 large numbers of pathogenic bacteria may nevertheless exist in their stools.242, 274 However, no nursery outbreak and few family cases240 have been traced to a symptomless carrier. Instead, passive transfer of bacteria from infant to infant by the hands of personnel appears to be of primary importance in these outbreaks.
EPEC can be recovered from the throat or nose of 5% to 80% of infants with diarrheal illness275, 294, 295 and from about 1% of asymptomatic infants.232, 243 The throat and nasal mucosa may represent a portal of entry or a source of transmission for EPEC. Environmental studies have shown that EPEC is distributed readily and widely in the vicinity of an infant with active diarrheal disease, often within 1 day of admission to the ward.232, 296 Massive numbers of organisms are shed in the diarrheal stool or vomitus of infected infants.249, 296 E. coli organisms may survive 2 to 4 weeks in dust243, 296 and can be found in the nursery air when the bedding or diapers of infected infants are disturbed during routine nursing procedures243, 296 or on floors, walls, cupboards, and nursery equipment such as scales, hand towels, bassinets, incubators, and oxygen tents of other infants.88, 243, 267 Documentation of the presence of EPEC in nursery air and dust does not establish the importance of this route as a source of cross-infection. One study presented evidence of the respiratory transmission of EPEC; however, even in the cases described, the investigators pointed out that fecal-oral transmission could not be completely ruled out.238 Additional clinical and experimental data are required to clarify the significance of droplet and environmental infection.
Coliform organisms have also been isolated in significant numbers from human milk,46, 297, 298 prebottled infant formulas,299 and formulas prepared in the home.292 EPEC in particular has been found in stool cultures obtained from donors of human milk and workers in a nursery formula room.260 In one instance, EPEC O111:B4 was isolated from a donor, and subsequently, the same serogroup was recovered in massive amounts in almost pure culture from her milk.260 Pathogenic strains of E. coli have also been isolated from raw cow’s milk300 and from drinking water.301 Likewise, EPEC has been isolated from flies during an epidemic, but this fact has not been shown to be of epidemiologic significance.211, 220
Pathogenesis
Infection of the newborn infant with EPEC takes place exclusively by the oral route. Attempts to induce disease in adult volunteers by rectal instillation of infected material have been unsuccessful.98 There are no reports of disease occurring after transplacental invasion of the fetal bloodstream by enteropathogenic or nonenteropathogenic strains of E. coli. Ascending intrauterine infection after prolonged rupture of the membranes has been reported only once; the neonate in this case suffered only from mild diarrhea.86
Bacterial cultures of the meconium and feces of newborns indicate that enteropathogenic strains of E. coli can colonize effectively the intestinal tract in the first days of life.85, 86, 87, 88 Although E. coli may disappear completely from stools of breast-fed children during the ensuing weeks, this disappearance is believed to be related to factors present in the human milk rather than the gastric secretions.5, 302, 303 The use of breast-feeding or expressed human milk has even been effective in terminating nursery epidemics caused by EPEC O111:B4, probably by reducing the incidence of cross-infections among infants.303, 304 Although dose-effect studies have not been performed among newborns, severe diarrhea has occurred after ingestion of 108 EPEC organisms by very young infants.305, 306 The high incidence of cross-infection outbreaks in newborn nurseries suggests that a far lower inoculum can often effect spread in this setting.
The role of circulating immunity in the prevention of gastrointestinal tract disease related to EPEC has not been clearly established. Virtually 100% of maternal sera have been found to contain hemagglutinating,219, 307, 308 bactericidal,305, 310 or bacteriostatic280, 312 antibodies against EPEC. The passive transfer of these antibodies across the placenta is extremely inefficient. Titers in blood of newborn infants are, on average, 4 to 100 times lower than those in the corresponding maternal sera. Group-specific hemagglutinating antibodies against the O antigen of EPEC are present in 10% to 20% of cord blood samples,219, 307, 308 whereas bactericidal307, 311 or bacteriostatic311 activity against these organisms can be found much more frequently. Tests for bacterial agglutination, which are relatively insensitive, are positive in only a small percentage of neonates.219, 311
The importance of circulating antibodies in the susceptibility of infants to EPEC infection is unknown. Experiments with suckling mice have failed to demonstrate any effect of humoral immunity on the establishment or course of duration of intestinal colonization with E. coli O127 in mothers or their infants.313 Similar observations have been made in epidemiologic studies among premature human infants using enteropathogenic (O127:B8)310 and nonenteropathogenic (O4:H5)269 strains of E. coli as the indicator organisms. In a cohort of 63 mothers and their infants followed from birth to 3 months old, Cooper and associates85 were able to show a far higher incidence of clinical EPEC disease in infants of EPEC-negative mothers than in infants born of mothers with EPEC isolated from stool cultures. This finding suggested to the investigators the possibility that mothers harboring EPEC in their gastrointestinal tracts transfer specific antibodies to their infants that confer some protection during the first weeks of life.
Protection against enteric infections in humans often correlates more closely with levels of local secretory than serum antibodies. Although it is known that colonization of newborns with E. coli leads to the production of coproantibodies against the ingested organisms,314, 315 the clinical significance of this intestinal immunity is uncertain. The previously mentioned experiment with mice showed no effect of active intestinal immunity on enteric colonization.313 In human infants, the frequency of bacteriologic and clinical relapse related to EPEC of the same serotype264, 265, 279 and the capacity of one strain of EPEC to superinfect a patient already harboring a different strain247, 258, 268 also cast some doubt on the ability of mucosal antibodies to inhibit or alter the course of intestinal infection. Studies of the protective effects of orally administered EPEC vaccines could help to resolve these questions.248
The mechanism by which EPEC causes diarrhea involves a complex array of plasmid and chromosomally encoded traits. EPEC serotypes usually do not make one of the recognized enterotoxins (LT or ST) as usually measured in tissue culture or animal models,316, 317, 318, 319, 320 nor do these serotypes cause a typical invasive colitis or produce a positive Sereny test result.315, 316 Only uncommonly do EPEC strains invade the bloodstream or disseminate.288 Nevertheless, EPEC strains that test negative in these tests are capable of causing diarrhea; inocula of 1010 E. coli O142 or O127 organisms caused diarrhea in 8 of 10 adult volunteers.320
Some EPEC strains may secrete weak enterotoxins,321, 322 but the consensus opinion is that the attaching and effacing lesion constitutes the critical secretory virulence phenotype.98, 121 Clinical pathologic reports reveal the characteristic attaching and effacing lesion in the small intestine of infected infants.324 The lesion is manifested by intimate (about 10nm) apposition of the EPEC to the enterocytes plasma membrane, with dissolution of the normal brush border and rearrangement of the cytoskeleton.121, 324 In some instances, the bacteria are observed to rise up on pedestal-like structures, which are diagnostic of the infection.121 Villus blunting, crypt hypertrophy, histiocytic infiltration in the lamina propria, and a reduction in the brush border enzymes may also be observed.324, 325
Two major EPEC virulence factors have been described; strains with both factors are designated as typical EPEC.98, 99, 323 One such factor is the locus of enterocyte effacement (LEE), a type III secretion system encoded by the LEE chromosomal pathogenicity island.326, 327, 328 The LEE secretion apparatus injects proteins directly from the cytoplasm of the infecting bacterium into the cytoplasm of the target enterocytes.327 The injected proteins constitute cytoskeletal toxins, which together elicit the close apposition of the bacterium to the cell, cause the effacement of microvilli, and most likely give rise to the net secretory state.98, 99, 121 One critical secreted protein, called Toll/interleukin-1 receptor (Tir),120 inserts into the plasma membrane of the epithelial cell, where it serves as the receptor for a LEE-encoded EPEC outer membrane protein called intimin.121 Animals infected with attaching and effacing pathogens mount antibody responses to intimin and Tir,329 and both are considered potential immunogens. The lack of protection from EPEC reinfection suggests that natural antibody responses to Tir and intimin are not protective.
The second major virulence factor of typical EPEC is the bundle-forming pilus (BFP),330 which is encoded on a partially conserved 60 MDa virulence plasmid called EPEC adherence factor (EAF).331 BFP, a member of the type IV pilus family, mediates aggregation of the bacteria to each other and probably to enterocytes themselves, thereby facilitating mucosal colonization.332 A BFP mutant was shown to be attenuated in adult volunteers.333
Pathology
The principal pathologic lesion with EPEC is the focal destructive adherence of the organism, effacing the microvillous brush border with villus blunting, crypt hypertrophy, histiocytic infiltration of the lamina propria, and reduced brush border enzymes. Rothbaum and colleagues324 described similar findings with dissolution of the glycocalyx and flattened microvilli with the nontoxigenic EPEC strain O119:B14. There has been a wide range of pathologic findings reported in infants dying of EPEC gastroenteritis. Most newborns dying with diarrheal disease caused by EPEC show no morphologic changes of the gastrointestinal tract by gross or microscopic examination of tissues.209, 210 Bray211 described such “meager” changes in the intestinal tract that “the impression received was that the term gastroenteritis is incorrect.” At the other extreme, extensive and severe involvement of the intestinal tract, although distinctly unusual among neonates with EPEC diarrhea, has been discussed in several reviews of the pathologic anatomy of this disease.247, 319, 334 Changes virtually identical to those found in infants dying with necrotizing enterocolitis have been reported.334 Drucker and co-workers319 found that among 17 infants who were dying of EPEC diarrhea, “intestinal gangrene, and/or perforation, and/or peritonitis were present in five, and intestinal pneumatosis in five.”
The reasons for such wide discrepancies in EPEC disease pathology are not clear. The severity of intestinal lesions at the time of death does not correlate with the birth weight of the patient, the age of onset of illness, the serogroup of the infecting strain, or the prior administration of oral or systemic antimicrobial agents. The suggestion that the intensity of inflammatory changes may depend on the duration of the diarrhea319 cannot be corroborated in autopsy studies215, 264, 335 or small intestinal biopsies.336, 337 It is difficult to reconcile such a thesis with the observation that a wide range of intestinal findings can be seen at autopsy among newborns infected by a single serotype of EPEC during an epidemic. The nonspecific pathologic picture described by some researchers includes capillary congestion and edema of the bowel wall and an increase in the number of eosinophils, plasma cells, macrophages, and mononuclear cells in the mucosa and submucosa.262, 319, 335 Villous patterns are generally well preserved, although some flattening and broadening of the villi are seen in the more severe cases. Almost complete absence of villi and failure of regeneration of small bowel mucosa have been reported in an extreme case.338 Edema in and around the myenteric plexuses of Auerbach, a common associated finding, has been suggested as a cause of the gastrointestinal tract dilatation often seen at autopsy in infants with EPEC infections.247, 335, 339 In general, the distal small intestine shows the most marked alterations; however, the reported pathologic findings may be found at all levels of the intestinal tract.
Several complications of EPEC infection have been reported. Candidal esophagitis accounted for significant morbidity in two series collected before210 and during247 the antibiotic era. Oral thrush has been seen in 50% of EPEC-infected infants treated with oral or systemic antibiotics.245, 264, 335 Some degree of fatty metamorphosis of the liver has been reported by several investigators210, 215 335; however, these changes are nonspecific and probably result from the poor caloric intake associated with persistent diarrhea or vomiting. Some degree of bronchopneumonia, probably a terminal event in most cases, exists in a large proportion of newborns dying of EPEC infection.210, 215, 339 In one reported series of infant cases, EPEC was demonstrated by immunofluorescent staining in the bronchi, alveoli, and interalveolar septa.
Mesenteric lymph nodes are often swollen and congested with reactive germinal centers in the lymphoid follicles.215, 262, 293 Severe lymphoid depletion, unrelated to the duration or severity of the antecedent illness, also has been described.285 The kidneys frequently show tubular epithelial toxic changes. Various degrees of tubular degeneration and cloudy swelling of convoluted tubules are common findings.215, 285, 335 Renal vein thrombosis or cortical necrosis may be observed in infants with disseminated intravascular coagulation in the terminal phases of the illness. The heart is grossly normal in most instances but may show minimal vacuolar changes of nonspecific toxic myocarditis on microscopic examination.335, 339 Candidal abscesses of the heart339 and kidneys285, 335, 339 have been described. With the exception of mild congestion of the pia arachnoid vessels and some edema of the meninges, examination of the central nervous system reveals few changes.215, 262 Despite the observation of Bray211 that “inflammation of the middle ear [is] exceptional,” strains of EPEC have been isolated from a significant number of specimens of the middle ear in case series in which dissection of the temporal bone has been performed.209, 215
Clinical Manifestations
Exposure of newborns to EPEC may be followed by one of several possible consequences: no infection, infection without illness, illness with gastroenteritis of variable severity and duration, and rarely, septicemia with or without metastatic foci of infection accompanying gastroenteritis.
When infants are exposed to EPEC, a significant number become colonized as temporary stool85, 88, 231 or pharyngeal238 carriers with no signs of clinical disease. Although Laurell340 showed that the percentage of asymptomatic infections rises steadily as age increases, this observation has not been confirmed by other investigators.214, 341 Similarly, the suggestion that prematurity per se is associated with a low incidence of inapparent EPEC infection has been documented in several clinical studies262, 264, 265 but refuted in others.252, 279 Most neonates who acquire infection with EPEC eventually show some clinical evidence of gastroenteritis. The incubation period is quite variable. Its duration has been calculated mostly from evidence in outbreaks in newborn nurseries, where the time of first exposure can be clearly defined in terms of birth or admission dates. In these circumstances, almost all infants show signs of illness between 2 and 12 days after exposure, and most cases show signs within the first 7 days.215, 231, 264 In some naturally acquired85, 86 and experimental306 infections with heavy exposure, the incubation period may be as short as 24 hours; the stated upper limit is 20 days.232, 343 The first positive stool culture and the earliest recognizable clinical signs of disease occur simultaneously in most infants,264, 266 although colonization may precede symptoms by 7 to 14 days.265, 266, 344 The gastroenteritis associated with EPEC infection in the newborn is notable for its marked variation in clinical pattern. Clinical manifestations vary from mild illness manifest only by transient anorexia and failure to gain weight to a sudden explosive fulminating diarrhea causing death within 12 hours of onset. Prematurity, underlying disease, and congenital anomalies often are associated with the more severe forms of illness.214, 233, 345, 346 Experienced clinicians have observed that the severity of EPEC gastroenteritis has declined markedly during the past 3 decades.225 The onset of illness usually is insidious, with vague signs of reluctance to feed, lethargy, spitting up of formula, mild abdominal distention, or even weight loss that may occur for 1 or 2 days before the first loose stool is passed. Diarrhea usually begins abruptly. It may be continuous and violent, or in milder infections, it may run an intermittent course with 1 or more days of normal stools followed by 1 or more days of diarrhea. Emesis sometimes is a prominent and persistent early finding. Stools are loose and bright yellow initially, later becoming watery, mucoid, and green. Flecks or streaks of blood, which are commonly seen with enterocolitis caused by Salmonella, Campylobacter, or Shigella, are rarely a feature of EPEC diarrheal disease. A characteristic seminal smell may pervade the environment of infants infected with EPEC O111:B34,232, 262, 347 and an odor variously described as “pungent,” “musty,” or “fetid” often surrounds patients excreting other strains in their stools.231, 256 Because the buttocks are repeatedly covered with liquid stools, excoriation of the perianal skin can be an early and persistent problem. Fever is an inconstant feature, and when it occurs, the patient’s temperature rarely rises above 39° C. Convulsions occur infrequently; their occurrence should alert the clinician to the possible presence of electrolyte disturbances, particularly hypernatremia. Prolonged hematochezia, distention, edema, and jaundice are ominous signs and suggest an unfavorable prognosis.215, 240, 285 Most infants receiving antimicrobial agents orally show a cessation of diarrhea, tolerate oral feedings, and resume weight gain within 3 to 7 days after therapy has been started.242, 245 Those with mild illness who receive no treatment can continue to have intermittent loose stools for 1 to 3 weeks. In one outbreak related to EPEC O142:K86, more than one third of the untreated or inappropriately treated infants had diarrhea for more than 14 days in the absence of a recognized enteric pathogen on repeated culturing.267 Recurrence of diarrhea and vomiting after a period of initial improvement is characteristic of EPEC enteritis.191, 239, 240 Although seen most often in newborns who have been treated inadequately or not treated at all, clinical relapses also occur after appropriate therapy. Occasionally, the signs of illness during a relapse can be more severe than those accompanying the initial attack of illness.215, 232, 285 Not all clinical relapses result from persistent infection. A significant number of relapses, particularly those that consistently follow attempts at reinstitution of formula feedings,262, 265 are caused by disaccharide intolerance rather than bacterial proliferation. Intestinal superinfections, caused by another serotype of EPEC283, 347, 348 or by completely different enteric pathogens, such as Salmonella or Shigella, 245 also can delay the resolution of symptoms. Rarely, infants suffer a “relapse” caused by an organism from the same O group as the original strain but differing in its H antigen. Unless complete serotyping is performed on all EPEC isolates, such an event easily could be dismissed as being a recurrence rather than a superinfection with a new organism.258, 268
Antimicrobial agents to which the infecting organisms are susceptible often may not eradicate EPEC,245, 265, 267 which may persist for weeks264, 283, 344 or months349 after the acute illness has subsided. Although reinfection cannot always be excluded, a significant number of infants are discharged from the hospital with positive rectal cultures.231, 233 Dehydration is the most common and serious complication of gastroenteritis caused by EPEC or a toxin-producing E. coli. Virtually all deaths directly attributable to the intestinal infection are caused by disturbances in fluids and electrolytes. When stools are frequent in number, large in volume, and violent in release, as they often are in severe infections with abrupt onset, a neonate can lose up to 15% of body weight in a few hours.232, 276 Rarely, fluid excretion into the lumen of the bowel proceeds so rapidly that reduction of circulating blood volume and shock may intervene before passage of even a single loose stool.262 Before the discovery of the etiologic agent, epidemic diarrhea of the newborn was also known by the term cholera infantum.
Mild disease, particularly when aggravated by poor fluid intake, can lead to a subtle but serious deterioration of an infant’s metabolic status. Sometimes, a week or more of illness elapses before it becomes apparent that an infant with borderline acidosis and dehydration who seemed to be responding to oral fluids alone requires parenteral therapy for improvement.272 It is incumbent on the clinician caring for small infants with gastroenteritis to follow them closely, with particular attention to serial weights, until full recovery can be confirmed.
There are few other complications, with the possible exception of aspiration pneumonia, directly related to EPEC gastroenteritis. Protracted diarrhea and nutritional failure may occur as a consequence of functional damage to the small intestinal mucosa, with secondary intolerance to dietary sugars.265, 338 Necrotizing enterocolitis, which occasionally results in perforation of the bowel and peritonitis, has not been causally related to infection with EPEC.247, 264, 266 A review of most of the large clinical series describing EPEC disease in infants who ranged in age from neonates to children aged 2 years revealed only three proven instances of bacteremia,265, 278 one possible urinary tract infection,265 and one documented case of meningitis in an infant of unspecified age.351 Focal infections among neonates were limited to several cases of otitis media247, 262 and a subcutaneous abscess294 from which EPEC was isolated. Additional complications include interstitial pneumonia,319 gastrointestinal bleeding with or without disseminated intravascular coagulation,334, 352 and methemoglobinemia caused by a mutant of EPEC O127:B8 that was capable of generating large quantities of nitrite from proteins present in the gastrointestinal tract.353
Diagnosis
The gold standard of EPEC diagnostics is identification in the stool of E. coli carrying genes for BFP and LEE. Identification of these genes can be accomplished by molecular methods (discussed later), but lack of access to these methods has led many labs to rely on surrogate markers, such as serotyping.19 Classic EPEC has been recovered from the vomitus, stool, or bowel contents of infected newborns. Isolation from bile233 and the upper respiratory tract85, 238, 239 has been described in those instances in which a specific search has been made. Less commonly, EPEC is isolated from ascetic fluid252 or purulent exudates209, 215, 294; occasionally, the organism has been recovered from blood cultures,265, 278 urine,265 and cerebrospinal fluid. Stool cultures generally are more reliable than rectal swabs in detecting the presence of enteric pathogens, although a properly obtained swab should be adequate to demonstrate EPEC in most cases.217, 296, 354 Specimens should be obtained as early in the course of the illness as possible because organisms are present in virtually pure culture during the acute phase of the enteritis but diminish in numbers during convalescence. Because of the preponderance of EPEC in diarrheal stools, two cultures are adequate for isolation of these pathogens in almost all cases of active disease. Studies using fluorescent antibody methods for identification of EPEC in stool specimens have demonstrated that during the incubation period of the illness, during convalescence, and among asymptomatic carriers of EPEC, organisms can be excreted in such small numbers that they escape detection by standard bacteriologic methods in a significant proportion of infants.245, 355, 356 As many as 3 to 10 specimens may be required to detect EPEC using methods that identify individual EPEC isolates in the stool.85, 346 After a stool specimen is received, it should be plated as quickly as possible onto noninhibiting media or placed in a preservative medium if it is to be held for longer periods. Deep freezing of specimens preserves viable EPEC when a prolonged delay in isolation is necessary.218 No selective media, biochemical reactions, or colonial variations permit differentiation of pathogenic and nonpathogenic strains. Certain features may aid in the recognition of two important serogroups. Cultures of serogroups O111:B4 and O55:B5, unlike many other coliforms, are sticky or stringy when picked with a wire loop and are rarely hemolytic on blood agar,217, 219 whereas O111:B4 colonies emit a distinctive evanescent odor commonly described as “seminal.”210, 214 This unusual odor first led Bray347 to suspect that specific strains of E. coli might be responsible for infantile gastroenteritis.
Because serotyping is simpler than molecular detection and because EPEC have long been known to belong to certain highly characteristic serotypes, serotyping can be used to identify likely EPEC strains, especially in outbreaks.216 E. coli, like other Enterobacteriaceae members, possesses cell wall somatic antigens (O), envelope or capsular antigens (K), and if motile, flagellar antigens (H). Many of the O groups may be further divided into two or more subgroups (a, b, c), and the K antigens are divisible into at least three varieties (B, L, A) on the basis of their physical behavior. Organisms that do not possess flagellar antigens are nonmotile (designated NM). The EPEC B capsular surface antigen prevents agglutination by antibodies directed against the underlying O antigen. Heating at 100°C for 1 hour inactivates the agglutinability and antigenicity of the B antigen.
Slide agglutination tests with polyvalent O or OB antiserum may be performed on suspensions of colonies typical of E. coli that have been isolated from infants with diarrhea, especially in nursery outbreaks. However, because of numerous false-positive “cross-reactions,” the O and K (or B) type must be confirmed by titration with the specific antisera.227 The presence of EPEC does not prove that EPEC is the cause of diarrhea in an individual patient. Mixed cultures with two or three serotypes of EPEC have been demonstrated in 1% to 10% of patients.244, 245, 352 This need not mean that two or three serotypes are causative agents. Secondary infection with hospital-acquired strains can occur during convalescence,173, 281, 283, 380 and some infants may have been asymptomatic carriers of one serotype at the time that another produced diarrheal disease. A similar explanation may pertain to mixed infections with EPEC and Salmonella or Shigella. 217, 220, 358 Nelson245 reported the presence of these pathogens in combination with EPEC in 14% of infants who were cultured as part of an antibiotic therapy trial. Salmonella and Shigella that had not been identified on cultures obtained at admission were isolated only after institution of oral therapy with neomycin. The investigator postulated that the alteration in bowel flora brought about by the neomycin facilitated the growth of these organisms, which had previously been suppressed and obscured by coliform overgrowth.245 The importance of seeking all enteric pathogens in primary and follow-up cultures of infantile diarrhea is apparent, particularly when the specimen originates from a patient in a newborn nursery or infants’ ward.
Although EPEC gastroenteritis was once considered to be synonymous with “summer diarrhea,” community outbreaks have occurred as frequently, if not more frequently, in the colder seasons.133, 160, 172 It has been suggested that the increased incidence at that time of year might be related to the heightened chance of contact between infants and toddlers that is bound to occur when children remain indoors in close contact.294 Nursery epidemics, which depend on the chance introduction and dissemination of EPEC within a relatively homogeneous population and stable environment, demonstrate no seasonal prevalence. Average relative humidity, temperature, and hours of daylight have no significant effect in determining whether an outbreak will follow the introduction of enteropathogenic strains of E. coli into a ward of infants.243
There are no clinical studies of the variations in peripheral leukocyte count, urine, or cerebrospinal fluid in neonatal enteritis caused by EPEC. Microscopic examination of stools of infants with acute diarrheal illness caused by these organisms usually has revealed an absence of fecal polymorphonuclear leukocytes,214, 256, 320, 359 although data on fecal lactoferrin in human volunteers suggest that an inflammatory process may be important in EPEC diarrhea.360, 361 Stool pH can be neutral, acid, or alkaline.61, 341 Serologic methods have not proved to be useful in attempting to establish a retrospective diagnosis of EPEC infection in neonates. Rising or significantly elevated agglutinin titers rarely could be demonstrated in early investigations210, 215, 231; hemagglutinating antibodies showed a significant response in no more than 10% to 20% of cases.245, 297 Fluorescent antibody techniques have shown promise for preliminary identification of EPEC in acute infantile diarrhea. This method is specific, with few false-positive results, and it is more sensitive than conventional plating and isolation techniques.282, 361, 362 The rapidity with which determinations can be performed makes them ideally suited for screening ill infants and possible carriers in determining the extent and progression of a nursery272, 282 or community238, 291 outbreak. Because immunofluorescence does not depend on the viability of organisms and is not affected by antibiotics that suppress growth on culture plates, it can be used to advantage in following bacteriologic responses and relapses in patients receiving oral therapy.210, 363 The use of fluorescent antibody techniques offers many advantages in the surveillance and epidemiologic control of EPEC gastroenteritis. Immunofluorescent methods should supplement but not replace standard bacteriologic and serologic methods for identification of enteric pathogens.
Specific gene probes and PCR primers for the BFP adhesin, the intimin-encoding gene (eae) and for a cryptic plasmid locus (EAF) are available.98 Detection of BFP or EAF are superior to detection of eae, because many non-EPEC, including nonpathogens, carry the eae gene.98, 364 PCR and gene probe analysis can be performed directly on the stools of suspect infants. However, confirmation of infection by the identification of the organism in pure culture should be pursued.
Before widespread use of molecular methods, the HEp-2 cell adherence assay was proposed for EPEC diagnosis.119 The presence of a focal or localized adherence (LA)119 pattern on the surface of HEp-2 or HeLa cells after 3-hour co-incubation is a highly sensitive and specific test for detection of EPEC.365 The requirement for cell culture and expertise in reading this assay limits its utility to the research setting. An ELISA for the BFP has been described but is not readily available.366 The capacity of LA + EPEC to polymerize F-actin can be detected in tissue culture cells stained with rhodamine-labeled phalloidin.367 This fluorescence-actin staining (FAS) test is cumbersome and impractical for routine clinical use.
Prognosis
The mortality rate recorded previously in epidemics of EPEC gastroenteritis is impressive for its variability. During the 1930s and 1940s, when organisms later recognized as classic enteropathogenic serotypes were infecting infants, the case-fatality ratio among neonates was about 50%.209, 210 During the 1950s and 1960s, many nursery epidemics still claimed about one of every four infected infants, but several outbreaks involving the same serotypes under similar epidemiologic circumstances had fatality rates of less than 3%.234, 241, 251 In the 1970s, reports appeared in the literature of a nursery epidemic with a 40% neonatal mortality rate285 and of an extensive outbreak in a nursery for premature infants with 4% fatalities265; another report stated that among “243 consecutive infants admitted to the hospital for EPEC diarrheal disease, none died of diarrheal disease per se.”368
A significant proportion of the infants who died during or shortly after an episode of gastroenteritis already were compromised by preexisting disease233, 283, 330 or by congenital malformations214, 231, 240 at the time they acquired their illness. These underlying pathologic conditions appear to exert a strongly unfavorable influence, probably by reducing the infant’s ability to respond to the added stresses imposed by the gastrointestinal tract infection. Although prematurity is often mentioned as a factor predisposing to a fatal outcome, the overall mortality rate among premature infants with EPEC gastroenteritis has not differed significantly over the years from that recorded for term infants.233, 262, 264
Therapy
The management of EPEC gastroenteritis should be directed primarily toward prevention or correction of problems caused by loss of fluids and electrolytes.198 Most neonates have a relatively mild illness that can be treated with oral rehydration. Infants who appear toxic, those with voluminous diarrhea and persistent vomiting, and those with increasing weight loss should be hospitalized for observation and treatment with parenteral fluids and careful maintenance of fluid and electrolyte balance and possibly with antimicrobial therapy. Clinical studies suggest that slow nasogastric infusion of an elemental diet can be valuable in treating infants who have intractable diarrhea that is unresponsive to standard modes of therapy.369
There is no evidence that the use of proprietary formulas containing kaolin or pectin is effective in reducing the number of diarrheal stools in neonates with gastroenteritis. Attempts to suppress the growth of enteric pathogens by feeding lactobacillus to the infant in the form of yogurt, powder, or granules have not been shown to be of value.370 A trial of cholestyramine in 15 newborns with EPEC gastroenteritis had no effect on the duration or severity of the diarrhea.265 The use of atropine-like drugs, paregoric, or loperamide to reduce intestinal motility or cramping should be avoided. Inhibition of peristalsis interferes with an efficient protective mechanism designed to rid the body of intestinal pathogens and may lead to fluid retention in the lumen of the bowel that may be sufficient to mask depletion of extracellular fluid and electrolytes.
The value of antimicrobial therapy in management of neonatal EPEC gastroenteritis, if any, is uncertain. There are no adequately controlled studies defining the benefits of any antibiotic in eliminating EPEC from the gastrointestinal tract, reducing the risk of cross-infection in community or nursery outbreaks, or modifying the severity of the illness. Proponents of the use of antimicrobial agents have based their claims for efficacy on anecdotal observations or comparative studies.245 Nonetheless, several clinical investigations have provided sufficient information to guide the physician faced with the dilemma of deciding whether to treat an individual infant or an entire nursery population suffering from EPEC diarrheal disease. It should be emphasized, however, that these guidelines must be considered tentative until rigidly controlled, double-blind studies have established the efficacy of antibiotics on a more rational and scientific basis.
Oral therapy with neomycin,234, 251 colistin,363 or chloramphenicol344 appears to be effective in rapidly reducing the number of susceptible EPEC organisms in the stool of infected infants. Studies comparing the responses of infants treated orally with neomycin,233 gentamicin,265 polymyxin,242 or kanamycin371 with the responses of infants receiving supportive therapy alone have shown that complete eradication of EPEC occurs more rapidly in those receiving an antimicrobial agent. In most cases, stool cultures are free of EPEC 2 to 4 days after the start of therapy.245, 363 Bacteriologic failure, defined as continued isolation of organisms during or after a course of an antimicrobial agent, can be expected to occur in 15% to 30% of patients.245, 265 Such relapses generally are not associated with a recurrence of symptoms.231, 234, 245 The effectiveness of oral antimicrobial therapy in reducing the duration of EPEC excretion serves to diminish environmental contamination and the spread of pathogenic organisms from one infant to another. Breaking the chain of fecal-oral transmission by administering antimicrobial agents simultaneously to all carriers of EPEC and their immediate contacts in the nursery has appeared to be valuable in terminating outbreaks that have failed to respond to more conservative measures.234, 264, 372 The apparent reduction in morbidity and mortality associated with oral administration of neomycin,230, 233, 234 colistin,246, 267, 285 polymyxin,242 or gentamicin246 during nursery epidemics has led to the impression that these drugs also exert a beneficial clinical effect in severely or moderately ill infants. Reports describing clinical,256 bacteriologic,265 or histopathologic319 evidence of tissue invasion by EPEC have persuaded some investigators to suggest the use of parenteral rather than oral drug therapy in debilitated or malnourished infants. On the basis of these data, there appears to be sufficient evidence to recommend oral administration of nonabsorbable antibiotics in the treatment of severely or moderately ill newborns with EPEC gastroenteritis. The drug most frequently used for initial therapy is neomycin sulfate in a dosage of 100mg/kg/day administered orally every 8 hours in three divided doses.245 In communities in which neomycin-resistant EPEC has been prevalent, treatment with colistin sulfate or polymyxin B in a dosage of 15 to 20mg/kg/day orally and divided into three equal doses may be appropriate. However, it is rarely necessary to use this approach.
Treatment should be continued only until stool cultures become negative for EPEC.245 Because of the unavoidable delay before cultures can be reported, most infants receive therapy for 3 to 5 days. If fluorescent antibody testing of rectal swab specimens is available, therapy can be discontinued as soon as EPEC no longer is identified in smears; this takes no more than 48 hours in more than 90% of cases.245 After diarrhea and vomiting have stopped and the infant tolerates formula feedings, shows a steady weight gain, and appears clinically well, discharge with outpatient follow-up is indicated. Bacteriologic relapses do not require therapy unless they are associated with illness or high epidemiologic risks to other young infants in the household. Because the infecting organisms in these recurrences generally continue to show in vitro susceptibility to the original drug, it should be reinstituted pending bacteriologic results.245
When clinical judgment suggests that a neonate may be suffering from bacterial sepsis and EPEC diarrheal disease, parenteral antimicrobial therapy is indicated after appropriate cultures have been obtained. The routine use of systemic therapy in severe cases of EPEC enteritis is not appropriate on the basis of current clinical experience.
Antimicrobial susceptibility patterns of EPEC are an important determinant of the success of therapy in infections with these organisms.233, 246, 247 These patterns are unpredictable, depending on the ecologic pressures exerted by local antibiotic usage246, 247 and on the incidence of transmissible resistance factors in the enteric flora of the particular population served by an institution.373, 374, 375, 376, 377, 378 For these reasons, variations in susceptibility patterns are apparent in different nurseries246, 376 and even from time to time within the same institution.247, 248, 250 Sudden changes in clinical response may even occur during the course of a single epidemic as drug-susceptible strains of EPEC are replaced by strains with multidrug resistance.233, 291, 375 Because differences can exist in the susceptibilities of different EPEC serogroups to various antimicrobial agents, regional susceptibility patterns should be reported on the basis of OB group or serotype rather than for EPEC as a whole.250 Knowledge of the resistance pattern in one’s area may help in the initial choice of antimicrobial therapy.
Prevention
The prevention of hospital outbreaks of EPEC gastroenteritis is best accomplished by careful attention to infection control policies for a nursery. All infants hospitalized with diarrhea should have a bacteriologic evaluation. If the laboratory is equipped and staffed to perform fluorescent antibody testing, infants transferred from another institution to a newborn, premature, or intensive care nursery and all infants with gastroenteritis on admission during an outbreak of EPEC diarrhea or in a highly endemic area can be held in an observation area for 1 or 2 hours until the results of the fluorescent antibody test or PCR are received. Because of the difficulty in diagnosing EPEC infection, reference laboratories, such as those at the Centers for Disease Control and Prevention (CDC), should be notified when an outbreak is suspected. Infants suspected to be excreting EPEC, even if healthy in appearance, then can be separated from others and given oral therapy until the test results are negative. Some experts have suggested that when the rapid results obtainable with fluorescent antibody procedures are not available, all infants admitted with diarrhea in a setting where EPEC is common may be treated as if they were excreting EPEC or some other enteric pathogen until contrary proof is obtained.372 Stool cultures should be obtained at admission, and contact precautions should be enforced among all who come into contact with the infant. Additional epidemiologic studies are needed to establish the advantages of careful isolation and nursing techniques, particularly in smaller community hospitals in which the number of infants in a “gastroenteritis ward” may be small. The use of prophylactic antibiotics has been shown to be of no value and can select for increased resistance.377, 378, 379
Unfortunately, it can be difficult to keep a nursery continuously free of EPEC. Specific procedures have been suggested for handling a suspected outbreak of bacterial enteritis in a newborn nursery or infant care unit.235, 355, 380 Evidence indicating that a significant proportion of E. coli enteritis may be caused by nontypeable strains has required some modification of these earlier recommendations. The following infection control measures may be appropriate:
-
1.
The unit is closed, when possible, to all new admissions.
-
2.
Cultures for enteric pathogens are obtained from nursing personnel assigned to the unit at the time of the outbreak.
-
3.
Stool specimens obtained from all infants in the nursery can be screened by the fluorescent antibody or another technique and cultured. Identification of a classic enteropathogenic serotype provides a useful epidemiologic marker; however, failure to isolate one of these strains does not eliminate the possibility of illness caused by a nontypeable EPEC.
-
4.
Antimicrobial therapy with oral neomycin or colistin can be considered for all infants with a positive fluorescent antibody test or culture result. The initial drug of choice depends on local patterns of susceptibility. Depending on the results of susceptibility tests, subsequent therapy may require modification.
-
5.
If an identifiable EPEC strain is isolated, second and third stool specimens from all infants in the unit are reexamined by the fluorescent antibody technique or culture at 48-hour intervals. If this is not practical, exposed infants should be carefully followed.
-
6.
Early discharge for healthy, mature, uninfected infants is advocated.
-
7.
An epidemiologic investigation should be performed to seek the factor or factors responsible for the outbreak. A surveillance system may be established for all those in contact with the nursery, including physicians and other health care personnel, housekeeping personnel, and postpartum mothers with evidence of enteric disease. A telephone, mail, or home survey may be conducted on all infants who were residing in the involved unit during the 2 weeks before the outbreak.
-
8.
When all patients and contacts are discharged and control of the outbreak is achieved, a thorough terminal disinfection of the involved nursery is mandatory.
Above all, personnel and parents should pay scrupulous attention to hand hygiene when handling infants.381
Enterohemorrhagic Escherichia coli
Since a multistate outbreak of enterohemorrhagic colitis was associated with E. coli O157:H7,382 Shiga toxin–producing E. coli (STEC) have been recognized as emerging gastrointestinal pathogens in most of the industrialized world. A particularly virulent subset of STEC, EHEC, causes frequent and severe outbreaks of gastrointestinal disease98, 383; the most virulent EHEC belong to serotype O157:H7. EHEC has a bovine reservoir and is transmitted by undercooked meat, unpasteurized milk, and contaminated vegetables such as lettuce, alfalfa sprouts, and radish sprouts (as occurred in more than 9000 schoolchildren in Japan).384, 387 It also spreads directly from person to person.387, 388 The clinical syndrome is that of bloody, noninflammatory (sometimes voluminous) diarrhea that is distinct from febrile dysentery with fecal leukocytes seen in shigellosis or EIEC infections.98 Most cases of EHEC infections have been recognized in outbreaks of bloody diarrhea or HUS in daycare centers, schools, nursing homes, and communities.388, 389, 390 Although EHEC infections often involve infants and young children, the frequency of this infection in neonates remains unclear; animal studies suggest that receptors for the Shiga toxin may be developmentally regulated and that susceptibility to disease may be age related.391
The capacity of EHEC to cause disease is related to the phage-encoded capacity of the organism to produce a Vero cell cytotoxin, subsequently shown to be one of the Shiga toxins.392, 393, 394 Shiga toxin 1 is neutralized by antiserum against Shiga toxin, whereas Shiga toxin 2, although biologically similar, is not neutralized by anti-Shiga toxin.395, 396 Like Shiga toxin made by Shigella dysenteriae, both E. coli Shiga toxins act by inhibiting protein synthesis by cleaving an adenosine residue from position 4324 in the 28S ribosomal RNA (rRNA) to prevent elongation factor-1–dependent aminoacyl transfer RNA (tRNA) from binding to the 60S rRNA.392, 393 The virulence of EHEC also may be determined in part by a 60-MDa plasmid that encodes for a fimbrial adhesin in O157 and O26.397, 398 This phenotype is mediated by the LEE pathogenicity island, which is highly homologous to the island present in EPEC strains.328
EHEC and other STEC infections should be suspected in neonates who have bloody diarrhea or who may have been exposed in the course of an outbreak among older individuals. Because most cases are caused by ingestion of contaminated food, neonates have a degree of epidemiologic protection from the illness. Diagnosis of STEC diarrhea is made by isolation and identification of the pathogen in the feces. E. coli O157:H7 does not ferment sorbitol, and this biochemical trait is commonly used in the detection of this serotype.98, 399 Because some nonpathogenic E. coli share this characteristic, confirmation of the serotype by slide agglutination is required. These techniques can be performed in most clinical laboratories. However, detection of non-O157 serotypes is problematic and relies on detection of the Shiga toxin; available methods include Shiga toxin ELISA, latex agglutination, and molecular methods.98, 399 These should be performed by a reference laboratory.
HUS in infants is not necessarily caused by STEC infection. Even in older patients, however, the stool is typically negative for STEC at the time the that HUS develops.400, 401 Serum and fecal detection of cytotoxin has been performed in such patients, but no diagnostic modality is definitive once HUS has supervened.400, 401
Antimicrobial therapy should not be administered to patients who may have STEC infection, although their role in inducing HUS remains controversial.402, 403 Management of the diarrhea and possible sequelae is supportive, with proper emphasis on fluid and electrolyte replacement. Aggressive rehydration is helpful in minimizing the frequency of serious sequelae.
Enteroaggregative Escherichia coli
The Hep-2 adherence assay is useful for the detection of EPEC, which exhibit a classic LA pattern.119 Two other adherence patterns can be discerned in this assay: aggregative (AA) and diffuse (DA). These two patterns have been suggested to define additional pathotypes of diarrheogenic E. coli. 98 Strains exhibiting the AA pattern (i.e., EAEC) are common pathogens of infants.125
EAEC cause diarrhea by colonization of the intestinal mucosa and elaboration of enterotoxins and cytotoxins.125, 404 Many strains can be shown to elicit secretion of inflammatory cytokines in vitro, which may contribute to growth retardation associated with prolonged otherwise asymptomatic colonization.103 Several virulence factors in EAEC are under the control of the virulence gene activator AggR.404 Presence of the AggR regulator or its effector genes has been proposed as a means of detecting truly virulent EAEC strains (called typical EAEC),404, 405 and an empirical gene probe long used for EAEC detection has been shown to correspond to one gene under AggR control.406, 407
Epidemiology and Transmission
The mode of transmission of EAEC has not been well established. In adult volunteer studies, the infectious dose is high (>108 colony-forming units [CFU]), suggesting that in adults at least, person-to-person transmission is unlikely.408, 409 Several outbreaks have been linked to consumption of contaminated food.410, 411 The largest of these outbreaks involved almost 2700 schoolchildren in Japan410; a contaminated school lunch was the implicated source of the outbreak. Some studies have demonstrated contamination of condiments or milk, which could represent vehicles of foodborne transmission.
Several nursery outbreaks of EAEC have been observed,412, 413 although in no case has the mechanism of transmission been established. The fist reported nursery outbreak involved 19 infants in Nis, Serbia, in 1995. Because these infants did not ingest milk from a common source, it is presumed that horizontal transmission by environmental contamination or hands of health care personnel was possible. Most of the infants were full term and previously well, and they were housed in two separate nursery rooms.
The earliest epidemiologic studies of EAEC implicated this organism as a cause of endemic diarrhea in developing countries.414, 415, 416 In this setting, EAEC as defined by the AA pattern of adherence to Hep-2 cells can be found in upward of 30% of the population at any one time.417 Newer molecular diagnostic modalities have revised this figure downward, although the organism remains highly prevalent in many areas. Several studies from the Indian subcontinent implicated EAEC among the most frequent enteric pathogens.414, 415, 418 Other sites reproducibly reporting high incidence rates include Mexico416 and Brazil.417, 419 There is evidence that EAEC may be emerging in incidence. A study from São Paulo, Brazil, implicated EAEC as the prevalent E. coli pathotypes in infants419; EPEC had previously been shown to be the most common pathogen in this community. Many other sites in developing countries of Africa,421 Asia,405, 422 and South America420 have described high endemic rates.
Several studies have suggested that EAEC is also a common cause of infant diarrhea in industrialized countries.423, 424, 425 Using molecular diagnostic methods, a large prospective study in the United Kingdom implicated EAEC as the second most common enteric bacterial pathogen after Campylobacter. 426 A similar study from Switzerland found EAEC to be the most common bacterial enteropathogen.423 Studies from the United States also have demonstrated a high rate of EAEC diarrhea in infants; using molecular diagnostic methods, EAEC was implicated in 11% and 8% of outpatient and inpatient diarrhea cohorts, respectively, compared with less than 2% of asymptomatic control infants (P < .05).427 Although epidemiologic studies have shown that EAEC can cause diarrhea in all age groups, several studies suggest that the infection is particularly common in infants younger than 12 months old.405, 420
Clinical Manifestations
Descriptions from outbreaks and volunteer studies suggest that EAEC diarrhea is watery in character with mucus but without blood or frank pus.408, 409, 412 Patients typically are afebrile. Several epidemiologic studies have suggested that many infants may have bloody diarrhea,416 but fecal leukocytes are uncommon.
The earliest reports of EAEC infection suggested that this pathogen may be particularly associated with persistent diarrhea (>14 days).414, 415, 416 However, later studies suggest that persistent diarrhea may occur in only a subset of infected infants.410 In the Serbian outbreak of 19 infected infants, the mean duration of diarrhea was 5.2 days412; diarrhea persisted more than 14 days in only three patients. Infants in this outbreak had frequent, green, odorless stools. In three cases, the stools had mucus, but none had visible blood. Eleven babies developed temperatures in excess of 38°C; only one had vomiting.
Despite a lack of clinical evidence suggesting inflammatory enteritis, several clinical studies have suggested that EAEC is associated with subclinical inflammation, including the shedding of fecal cytokines and lactoferrin.103, 428 Studies in Fortaleza, Brazil, suggest that children asymptomatically excreting EAEC may exhibit growth shortfalls compared with uninfected peers.103 A study from Germany reported an association between EAEC isolation and infant colic in infants without diarrhea.425 This observation has not been repeated. EAEC should be considered in the differential diagnosis of persistent diarrhea and failure to thrive in infants.
Diagnosis and Therapy
Diagnosis of EAEC requires identification of the organism in the patient’s feces. The HEp-2 adherence assay can be used for this purpose119; some reports suggest that the adherence phenotype can be observed using formalin-fixed cells,429, 430 thereby obviating the need to cultivate eukaryotic cells for each assay. PCR and gene probe for typical EAEC are available.
Successful antibiotic therapy has been reported using fluoroquinolones in adult patients,431 although preliminary studies suggest that azithromycin432 or rifaximin433 also may be effective. Therapy in infected infants should be guided by the results of susceptibility testing, as EAEC frequently is antibiotic resistant.421
Other Escherichia coli Pathotypes
Additional E. coli pathotypes have been described, including diffusely adherent E. coli (DAEC),434 and cytodetaching E. coli. 435 DAEC has been specifically associated with diarrhea outside of infancy, as infants may have some degree of inherent resistance to infection.436 Cytodetaching E. coli represent organisms that secrete the E. coli hemolysin.437 It is not clear whether these latter organisms are true enteric pathogens.
SALMONELLA
Nature of the Organism
Salmonella classification tends to be confusing. Although taxonomists classify Salmonella narrowly as a single species, with Salmonella typhi, Salmonella choleraesuis, and Salmonella enteritidis technically being serovars or subspecies, for clinical purposes, these subspecies conventionally are referred to as species. For example, clinical laboratories tend to use the shorthand S. typhimurium rather than the more formal designation Salmonella enterica serovar typhimurium. Biochemical traits are used routinely by hospital laboratories to differentiate S. typhi, S. choleraesuis, and S. enteritidis from each other. S. typhi is unlike other salmonellae in that it does not produce gas from glucose.438 Because there are several thousand serotypes included in the species S. enteritidis, serotyping of S. enteritidis is usually performed by state health departments rather than by hospital laboratories. The most common serogroups and representative serotypes are listed in Table 20–3. Infection of humans with the other serogroups (e.g., C3, D2, E2, E3, F, G, H, I) is uncommon.
Table 20-3.
Common Serotypes and Serogroups of Salmonella
| Serogroups | Serotypes |
|---|---|
| A | Paratyphi A |
| B | Agona |
| Derby | |
| Heidelberg | |
| Paratyphi B (schottmuelleri) | |
| Saint-paul | |
| Typhimurium | |
| C1 | Choleraesuis |
| Eimsbuettel | |
| Infantis | |
| Montevideo | |
| Oranienburg | |
| Paratyphi C (hirschfeldii) | |
| Thompson | |
| C2 | Blockley |
| Hadar | |
| Muenchen | |
| Newport | |
| C3 | Kentucky |
| D1 | Dublin |
| Enteritidis | |
| Javiana | |
| Panama | |
| Typhi | |
| D2 | Maarssen |
| E1 | Anatum |
| E2 | London Newington |
| E3 | Illinois |
| E4 | Krefeld |
| Senftenberg | |
There are differences in invasiveness of Salmonella strains related to serotype. S. typhi, S. choleraesuis, Salmonella heidelberg,439, 440 and Salmonella dublin 441 are particularly invasive, with bacteremia and extraintestinal focal infections occurring frequently. Salmonella species possess genes closely related to those for the Shigella invasion plasmid antigens; these genes are probably essential to intestinal infection.442, 443 Virulence plasmids, which increase invasiveness in some serotypes, have been recognized, although the precise mechanisms of virulence remain to be elucidated; resistance to complement-mediated bacteriolysis by inhibition of insertion of the terminal C5b-9 membrane attack complex into the outer membrane may be important.444, 445 Laboratory studies have demonstrated dramatic strain-related difference in the ability of S. typhimurium to evoke fluid secretion, to invade intestinal mucosa, and to disseminate beyond the gut.446 Production of an enterotoxin immunologically related to cholera toxin by about two thirds of Salmonella strains may be related to the watery diarrhea often seen.447 The significance of protein synthesis-inhibiting cytotoxins448 remains to be proved, although such toxins can damage gut epithelium, which could facilitate invasion. The cytotoxins produced by Salmonella are not immunologically related to Shiga toxin made by Shigella dysenteriae type 1449 or E. coli O157:H7.
Salmonellae have the ability to penetrate epithelial cells and reach the submucosa, where they are ingested by phagocytes.450 In phagocytes, salmonellae are resistant to killing, in part because of the properties of their lipopolysaccharides.451, 452 Persistence of the organism within phagolysosomes of phagocytic cells may occur with any species of Salmonella. It is not completely clear how the organisms have adapted to survive in the harsh intracellular environment, but their survival has major clinical significance. It accounts for relapses after therapy. It explains the inadequacy of some antimicrobial agents that do not penetrate phagolysosomes. It perhaps is the reason for prolonged febrile courses that occur even in the face of appropriate therapy. Although humoral immunity and cell-mediated immunity are stimulated during Salmonella infections, it is believed that cell-mediated immunity plays a greater role in eradication of the bacteria.453 T cell activation of macrophages appears to be important in killing intracellular Salmonella. 454 Defective interferon-γ production by monocytes of newborns in response to S. typhimurium lipopolysaccharide may explain in part the unusual susceptibility of infants to Salmonella infection.455 Studies in mice suggest that helper T cell (TH1) responses in Peyer’s patches and mesenteric lymph nodes may be central to protection of the intestinal mucosa.456 Humans who lack the IL-12 receptor and therefore have impaired TH1 responses and interferon-γ production are at increased risk for Salmonella infection.457
In typhoid fever, presence of an envelope antigen, Vi, is known to enhance virulence. Patients who develop classic enteric fever have positive stool cultures in the first few days after ingestion of the organism and again late in the course after a period of bacteremia. This course reflects early colonization of the gut, penetration of gut epithelium with infection of mesenteric lymph nodes, and reseeding of the gut during a subsequent bacteremic phase.458 Studies of S. typhimurium in monkeys suggest similar initial steps in pathogenesis (e.g., colonization of gut, penetration of gut epithelium, infection of mesenteric lymph nodes) but failure of the organism to cause a detectable level of bacteremia.459
Although both Salmonella and Shigella invade intestinal mucosa, the resultant pathologic changes are different. Shigella multiplies within and kills enterocytes with production of ulcerations and a brisk inflammatory response, whereas Salmonella passes through the mucosa and multiplies within the lamina propria, where the organisms are ingested by phagocytes; consequently, ulcer formation is less striking,446 although villus tip cells are sometimes sloughed. Acute crypt abscesses can be seen in the stomach and small intestine, but the most dramatic changes occur in the colon, where acute diffuse inflammation with mucosal edema and crypt abscesses are the most consistent findings.460, 461 With S. typhi there also is hyperplasia of Peyer’s patches in the ileum, with ulceration of overlying tissues.
Epidemiology and Transmission
Salmonella strains, with the exception of S. typhi, are well adapted to a variety of animal hosts; human infection often can be traced to infected meat, contaminated milk, or contact with a specific animal. Half of commercial poultry samples are contaminated with Salmonella. 462 Definition of the serotype causing infection can sometimes suggest the likely source. For example, S. dublin is closely associated with cattle; human cases occur with a higher-than-predicted frequency in people who drink raw milk.441 For S. typhimurium, which is the most common serotype and accounts for more than one third of all reported human cases, a single source has not been established, although there is an association with cattle. Despite the 1975 ban by the U.S. Food and Drug Administration (FDA) on interstate commercial distribution of small turtles, these animals continue to be associated with infection, as illustrated by a series of cases in Puerto Rico.463 Various pet reptiles are an important source of a variety of unusual Salmonella serotypes such as Salmonella marina, Salmonella chameleon, Salmonella arizonae, Salmonella java, Salmonella stanley, Salmonella poona, Salmonella jangwain, Salmonella tilene, Salmonella pomona, Salmonella miami, Salmonella manhattan, Salmonella litchfield, Salmonella rubislaw, and Salmonella wassenaar. 464, 465, 466 Salmonella organisms are hardy and capable of prolonged survival; organisms have been documented to survive in flour for nearly a year.467 Salmonella tennessee has been shown to remain viable for many hours on non-nutritive surfaces (i.e., glass, 48 hours; stainless steel, 68 hours; enameled surface, 114 hours; rubber mattress, 119 hours; linen, 192 hours; and rubber tabletop, 192 hours).468
Infection with Salmonella is, like most enteric infections, more common in young children than in adults. The frequency of infection is far greater in the first 4 years of life; roughly equal numbers of cases are reported during each decade beyond 4 years of age. Although the peak incidence occurs in the second through sixth months of life, infection in the neonate is relatively common. Researchers at the CDC have estimated the incidence of Salmonella infection in the first month of life at nearly 75 cases per 100,000 infants.469
Adult volunteer studies suggest that large numbers of Salmonella (105 to 109) need to be ingested to cause disease.470 However, it is likely that lower doses cause illness in infants. The occurrence of nursery outbreaks468, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496 and intrafamilial spread497 suggests that organisms are easily spread from person to person; this pattern is typical of low-inoculum diseases transmitted by the fecal-oral route. The neonate with Salmonella infection infrequently acquires the organism from his or her mother during delivery. Although the index case in an outbreak can often be traced to a mother,474, 475, 476, 477, 495 subsequent cases result from contaminated objects in the nursery environment498, 499 serving as a reservoir coming in contact with hands of attending personnel.468, 483 The mother of an index case may be symptomatic479, 480, 500, 501 or asymptomatic with preclinical infection,484 convalescent infection,477, 481, 502 or chronic carriage.503 The risk of the newborn becoming infected once Salmonella is introduced into a nursery has been reported to be as high as 20% to 27%,487, 493 but the frequency of infection may be lower because isolated cases without a subsequent epidemic are unlikely to be reported.
Gastric acidity is an important barrier to Salmonella infection. Patients with anatomic or functional achlorhydria are at increased risk of developing salmonellosis.504, 505 The hypochlorhydria25 and rapid gastric emptying typical of early life28 may in part explain the susceptibility of infants to Salmonella. Premature and low-birth-weight infants appear to be at higher risk of acquiring Salmonella infection than term neonates.483, 485 Whether this reflects increased exposure because of prolonged hospital stays or increased susceptibility on the basis of intestinal or immune function is unclear. Contaminated food or water is often the source of Salmonella infection in older patients; the limited diet of the infant makes contaminated food a less likely source of infection. Although human milk,506, 507, 508 raw milk,509 powdered milk,510, 511, 512 formula,493 and cereal513 have been implicated in transmission to infants, more often fomites, such as delivery room resuscitators,471 rectal thermometers,486, 514 oropharyngeal suction devices,515, 516, 517 water baths for heating formula,517 soap dispensers,518 scales,468, 472, 519“clean” medicine tables,468 air-conditioning filters,468 mattresses, radiant warmers,498 and dust,472 serve as reservoirs. One unusual outbreak involving 394 premature and 122 term infants was traced to faulty plumbing, which caused massive contamination of environment and personnel.493 After Salmonella enters a nursery, it is difficult to eradicate. Epidemics lasting 6 to 7 weeks,486, 491 17 weeks,468 6 months,485, 490 1 year,480 and 27 to 30 months487, 493 have been reported. Spread to nearby pediatric wards has occurred.488, 494
The incubation period in nursery outbreaks has varied widely in several studies where careful attention has been paid to this variable. In one outbreak of Salmonella oranienburg involving 35 newborns, 97% of cases occurred within 4 days of birth.487 In an outbreak of S. typhimurium, each of the ill infants presented within 6 days of birth.477 These incubation periods are similar to those reported for Salmonella newport in older children and adults, 95% of whom have been reported to be ill within 8 days of exposure.520, 521 Conversely, one outbreak of Salmonella nienstedten involving newborns was characterized by incubation periods of 7 to 18 days.488
The usual incubation period associated with fecal-oral nursery transmission is not found with congenital typhoid. During pregnancy, typhoid fever is associated with bacteremic infection of the fetus. The congenitally infected infants are symptomatic at birth. They are usually born during the second to fourth week of untreated maternal illness.522 Usually, the mother is a carrier; fecal-oral transmission of S. typhi can occur with delayed illness in the newborn.523
Clinical Manifestations
Several major clinical syndromes occur with nontyphoidal Salmonella infection in young infants. Colonization without illness may be the most common outcome of ingestion of Salmonella by the neonate. Such colonization usually is detected when an outbreak is under investigation. Most infected infants who become ill have abrupt onset of loose, green, mucus-containing stools, or they have bloody diarrhea; an elevated temperature is also a common finding in Salmonella gastroenteritis in the first months of life.440 Grossly bloody stools are found in the minority of patients, although grossly bloody stools can occur in the first 24 hours of life. Hematochezia is more typically associated with noninfectious causes (e.g., swallowed maternal blood, intestinal ischemia, hemorrhagic diseases, anorectal fissures) at this early age.524 There appear to be major differences in presentation related to the serotype of S. enteritidis causing infection. For example, in one epidemic of S. oranienburg 487 involving 46 newborns, 76% had grossly bloody stools, 11% were febrile, 26% had mucus in their stools, and only 11% were healthy. In a series of S. newport infections involving 11 premature infants,474 90% of infants with gastroenteritis had blood in their stools, 10% had fever, 10% had mucus in their stools, and 9% were asymptomatic. In an outbreak of S. typhimurium 477 involving 11 ill and 5 healthy infants, none had bloody stools; all of the symptomatic infants were febrile and usually had loose, green stools. Of 26 infants infected by Salmonella virchow, 42% were asymptomatic; the rest had mild diarrhea.482 Seals and colleagues488 described 12 infants with S. nienstedten, all of whom had watery diarrhea and low-grade fever; none had bloody stools. In a large outbreak in Zimbabwe of S. heidelberg infection reported by Bannerman,485 38% of 100 infants were asymptomatic, 42% had diarrhea, 16% had fever, 15% had pneumonia, and 2% developed meningitis. An outbreak of Salmonella worthington was characterized primarily by diarrhea, fever, and jaundice, although 3 of 18 infants developed meningitis and 17% died.515 In dramatic contrast to these series, none of 27 infants with positive stool cultures for S. tennessee had an illness in a nursery found to be contaminated with that organism.469 A few infants with Salmonella gastroenteritis have developed necrotizing enterocolitis,492, 525 but it is not clear whether Salmonella was the cause.
Although gastroenteritis is usually self-limited, chronic diarrhea has sometimes been attributed to Salmonella. 503, 526 Whether chronic diarrhea is caused by Salmonella is uncertain. Although some infants develop carbohydrate intolerance after a bout of Salmonella enteritis527, 528 and Salmonella is typically listed as one of the causes of postinfectious protracted diarrhea,529 it is difficult to be sure that the relationship is causal. The prolonged excretion of Salmonella after a bout of gastroenteritis may sometimes cause nonspecific chronic diarrhea to be erroneously attributed to Salmonella.
Major extraintestinal complications of Salmonella infection may develop in the neonate who becomes bacteremic. Extraintestinal spread may develop in infants who initially present with diarrhea and in some who have no gastrointestinal tract signs. Bacteremia appears to be more common in the neonate than in the older child.530 A study of more than 800 children with Salmonella infection showed that extraintestinal infection occurred significantly more often (8.7% versus 3.6%) in the first 3 months of life.531 Several retrospective studies suggest that infants in the first month of life may have a risk of bacteremia as high as 30% to 50%.440 One retrospective study439 suggests that the risk is not increased in infancy and estimates that the risk of bacteremia in childhood Salmonella gastroenteritis is between 8.5% and 15.6%. Prospective studies of infants in the first year of life suggest that the risk of bacteremia is 1.8% to 6.0%.532, 533 Although selection biases in these studies limit the reliability of these estimates, the risk is substantial. The Salmonella species isolated from infants include some serotypes that appear to be more invasive in the first 2 months of life than in older children or healthy adults (S. newport, S. agona, S. blockley, S. derby, S. enteritidis, S. heidelberg, S. infantis, S. javiana, S. saint-paul, and S. typhimurium) and serotypes that are aggressive in every age group (S. choleraesuis and S. dublin). Other serotypes appear more likely to cause bacteremia in adults (S. typhi, S. paratyphi A, and S. paratyphi B).530
Virtually any Salmonella serotype can cause bacteremic disease in neonates. A few infants with Salmonella gastroenteritis have died with E. coli or Pseudomonas aeruginosa sepsis,494 but the role of Salmonella in these cases is unclear. Unlike the situation in older children in whom bacteremic salmonellosis often is associated with underlying medical conditions, bacteremia may occur in infants who have no immunocompromising conditions.534 Salmonella bacteremia is often not suspected clinically because the syndrome is not usually distinctive.439, 440 Even afebrile, well-appearing children with Salmonella gastroenteritis have been documented to have bacteremia that persists for several days.535 Although infants with bacteremia may have spontaneous resolution without therapy,536 a sufficient number develop complications to warrant empirical antimicrobial therapy when bacteremia is suspected. The frequency of complications is highest in the first month of life. Meningitis is the most feared complication of bacteremic Salmonella disease. Between 50% and 75% of all cases of nontyphoidal Salmonella meningitis occur in the first 4 months of life.537 The serotypes associated with neonatal meningitis (S. typhimurium, S. heidelberg, S. enteritidis, S. saint-paul, S. newport, and S. panama) 497 are serotypes frequently associated with bacteremia. Meningitis has a high mortality rate, in part because of the high relapse rates. Relapse has been reported in up to 64% of cases.538 In some studies, more than 90% of patients with meningitis have died,539 although more typically, 30% to 60% of infants die.540, 541 The survivors suffer the expected complications of gram-negative neonatal meningitis, including hydrocephalus, seizures, ventriculitis, abscess formation, subdural empyema, and permanent neurologic impairment. Neurologic sequelae have included retardation, hemiparesis, epilepsy, visual impairment, and athetosis.537
In large nursery outbreaks, it is common to find infants whose course is complicated by pneumonia,485 osteomyelitis,542, 543 or septic arthritis.483, 485 Other rare complications of salmonellosis include pericarditis,544 pyelitis,545 peritonitis,477 otitis media,477 mastitis,546 cholecystitis,547 endophthalmitis,548 cutaneous abscesses,492 and infected cephalohematoma.542 Other focal infections seen in older children and adults, such as endocarditis and infected aortic aneurysms, rarely or never have been reported in neonates.537, 549 Although the mortality rate in two reviews of nursery outbreaks was 3.7% to 7.0%,495, 496 in some series, it reached 18%.485
Enteric fever, most often related to S. typhi but also occurring with S. paratyphi A, S. paratyphi B, S. paratyphi C, and other Salmonella species, is reported much less commonly in infants than in older patients. Infected infants develop typical findings of neonatal sepsis and meningitis. Current data suggest that mortality is about 30%.550 In utero infection with S. typhi has been described. Typhoid fever518, 551 and nontyphoidal Salmonella infections552 during pregnancy put women at risk of aborting the fetus. Premature labor usually occurs during the second to the fourth week of maternal typhoid if the woman is untreated.522 In a survey of typhoid fever in pregnancy during the preantibiotic era, 24 of 60 women with well-documented cases delivered prematurely, with resultant fetal death; the rest delivered at term, although only 17 infants survived.553 The outlook for carrying the pregnancy to term and delivering a healthy infant appears to have improved dramatically during the antibiotic era. However, one of seven women with typhoid in a series still delivered a dead fetus with extensive liver necrosis.554 In the preantibiotic era, about 14% of pregnant women with typhoid fever died.555 With appropriate antimicrobial therapy, pregnancy does not appear to put the woman at increased risk of death. Despite these well-described cases, typhoid fever is rare early in life.
Of 1500 cases of typhoid fever that Osler and McCrae556 reported, only 2 were in the first year of life. In areas where typhoid fever is still endemic, systematic search for infants with enteric fever has failed to find many cases. The few infections with S. typhi documented in children in the first year of life often present as a brief nondescript “viral syndrome” or as pneumonitis.557, 558 Fever, diarrhea, cough, vomiting, rash, and splenomegaly may occur; the fever may be high, and the duration of illness may be many weeks.522
Diagnosis
The current practice of early discharge of newborn infants, although potentially decreasing the risk of exposure, can make recognition of a nursery outbreak difficult. Diagnosis of neonatal salmonellosis should trigger an investigation for other cases. Other than diarrhea, signs of neonatal Salmonella infection are similar to the nonspecific findings seen in most neonatal infections. Lethargy, poor feeding, pallor, jaundice, apnea, respiratory distress, weight loss, and fever are common. Enlarged liver and spleen are common in those neonates with positive blood cultures. Laboratory studies are required to establish the diagnosis because the clinical picture is not distinct. The fecal leukocyte examination reveals polymorphonuclear leukocytes in 36% to 82%359, 559 of persons with Salmonella infection, but it has not been evaluated in neonates. Obviously, the presence of fecal leukocytes is consistent with colitis of any cause and therefore is a nonspecific finding. Routine stool cultures usually detect Salmonella if two or three different enteric media (i.e., MacConkey’s, eosin-methylene blue, Salmonella-Shigella, Tergitol 7, xylose-lysine-deoxycholate, brilliant green, or bismuth sulfite agar) are used. Stool, rather than rectal swab material, is preferable for culture, particularly if the aim of culture is to detect carriers.560 On the infrequent occasions when proctoscopy is performed, mucosal edema, hyperemia, friability, and hemorrhages may be seen.461 Infants who are bacteremic often do not appear sufficiently toxic to raise the suspicion of bacteremia.561 Blood cultures should be obtained as a routine part of evaluation of neonates with suspected or documented Salmonella infection. Ill neonates with Salmonella infection should have a cerebrospinal fluid examination performed. Bone marrow cultures also may be indicated when enteric fever is suspected. There are no consistent abnormalities in the white blood cell count. Serologic studies are not helpful in establishing the diagnosis, although antibodies to somatic562, 563 and flagellar antigens487 develop in many infected newborns.
If an outbreak of salmonellosis is suspected, further characterization of the organism is imperative.464 Determination of somatic and flagellar antigens to characterize the specific serotype may be critical to investigation of an outbreak. When the serotype found during investigation of an outbreak is a common one (e.g., S. typhimurium), antimicrobial resistance testing475, 565 and use of molecular techniques such as plasmid characterization565 can be helpful in determining whether a single-strain, common-source outbreak is in progress.
Therapy
As in all enteric infections, attention to fluid and electrolyte abnormalities is the first issue that must be addressed by the physician. Specific measures to eradicate Salmonella intestinal infection have met with little success. Multiple studies show that antibiotic treatment of Salmonella gastroenteritis prolongs the excretion of Salmonella. 566, 567, 568, 569, 570, 571, 572, 573 Almost one half of the infected children in the first 5 years of life continue to excrete Salmonella 12 weeks after the onset of infection; more than 5% have positive cultures at 1 year.574 No benefit of therapy has been shown in comparisons of ampicillin or neomycin versus placebo,570 chloramphenicol versus no antibiotic treatment,569 neomycin versus placebo,571 ampicillin or trimethoprim-sulfamethoxazole versus no antibiotic,568 and ampicillin or amoxicillin versus placebo.572 In contrast to these studies, data suggest that there may be a role for quinolone antibiotics in adults and children,573, 575 but these drugs are not approved for use in neonates, and resistance has been encountered.576 Because these studies have few data as to the risk-benefit ratio of therapy in the neonate, it is uncertain whether they should influence treatment decisions in neonates. Studies that have included a small number of neonates suggest little benefit from antimicrobial therapy.477, 487, 568, 577, 578 However, because bacteremia is common in neonates, antimicrobial therapy for infants younger than 3 months who have Salmonella gastroenteritis often is recommended,532, 533, 561 especially if the infant appears toxic. Premature infants and those who have other significant debilitating conditions also should probably be treated. The duration of therapy is debatable but should probably be no more than 3 to 5 days if the infant is not seriously ill and if blood cultures are sterile. If toxicity, clinical deterioration, or documented bacteremia complicates gastroenteritis, prolonged treatment is indicated. Even with antimicrobial therapy, some infants develop complications. The relatively low risk of extraintestinal dissemination must be balanced against the well-documented risk of prolonging the carrier state. For infants who develop chronic diarrhea and malnutrition, hyperalimentation may be required; the role of antimicrobial agents in this setting is unclear. The infant with typhoid fever should be treated with an antimicrobial agent; relapses sometimes occur after therapy.
Colonized healthy infants discovered by stool cultures during evaluation of an outbreak ought to be isolated but probably should not receive antimicrobial therapy. Such infants should be discharged from the nursery as early as possible and followed carefully as outpatients.
Antimicrobial treatment of neonates who have documented extraintestinal dissemination must be prolonged. Bacteremia without localization is generally treated with at least a 10-day course of therapy. Therapy for Salmonella meningitis must be given for at least 4 weeks to lessen the risk of relapse. About three fourths of patients who have relapses have been treated for three weeks or less.537 Similar to meningitis, treatment for osteomyelitis must be prolonged to be adequate. Although cures have been reported with 3 weeks of therapy, 4 to 6 weeks of therapy is recommended.
In vitro susceptibility data for Salmonella isolates must be interpreted with caution. The aminoglycosides show good in vitro activity but poor clinical efficacy, perhaps because of the low pH of the phagolysosome. Aminoglycosides have poor activity in an acid environment. The stability of some drugs in this acid environment also may explain in vitro and in vivo disparities. The intracellular localization and survival of Salmonella within phagocytic cells also presumably explains the relapses encountered with virtually every regimen. Resistance to antibiotics has long been a problem with Salmonella infection.566, 579, 580 There has been a steady increase in resistance to Salmonella in the United States over the last 20 years.581 With the emergence of typhimurium type DT 104, resistance to ampicillin, chloramphenicol, streptomycin, sulfonamides, and tetracycline has increased from 0.6% in 1979 and 1980 to 34% in 1996.582 Resistance plasmids have been selected and transmitted, partly because therapy has been given for mild illness that should not have been treated566 and partly because of use of antibiotics in animal feeds. Resistance to chloramphenicol and ampicillin has made trimethoprim-sulfamethoxazole increasingly important for the treatment of Salmonella infection in those patients who require therapy. However, with increasing resistance to all three of these agents in Asia,583 the Middle East,584 Africa,585 Europe,586, 587 Argentina,580 and North America,579, 588, 589 the third-generation cephalosporins and quinolones represent drugs of choice for invasive salmonellosis. The quinolones currently are not approved for persons younger than 18 years. Cefotaxime, ceftriaxone, and cefoperazone represent acceptable alternative drugs for typhoidal and nontyphoidal salmonellosis when resistance is encountered.590, 591 Because the second-generation cephalosporins, such as cefuroxime, are less active in vitro than the third-generation cephalosporins and are not consistently clinically effective, they should not be used.590, 592 Data suggest that cefoperazone may sterilize blood and cause patients with typhoid fever to become afebrile more rapidly than with chloramphenicol,593 perhaps because cefoperazone is excreted into bile in high concentrations.594 The third-generation cephalosporins may have higher cure and lower relapse rates than ampicillin or chloramphenicol in children with Salmonella meningitis.595 The doses of ampicillin, chloramphenicol, or cefotaxime used in infants with gastroenteritis pending results of blood cultures are the same as those used in treatment of sepsis. Because of the risk of gray baby syndrome, chloramphenicol should not be used in neonates unless other effective agents are not available. Trimethoprim-sulfamethoxazole, although useful in older children and adults, is not used in neonates because of the risk of kernicterus. Nosocomial infection with strains of Salmonella resistant to multiple antibiotics, including third-generation cephalosporins, has emerged as a problem in South America.580
Nonantibiotic interventions are important in the control of Salmonella infections. Limited data suggest that intravenous immune globulin (IGIV) (500mg/kg on days 1, 2, 3, and 8 of therapy) along with antibiotic therapy may decrease the risk of bacteremia and death in preterm infants with Salmonella gastroenteritis.596
Prevention
Early recognition and intervention in nursery outbreaks of Salmonella are crucial to control. When a neonate develops salmonellosis, a search for other infants who have been in the same nursery should be undertaken. When two or more cases are recognized, environmental cultures, cultures of all infants, cohorting and contact isolation of infected infants, rigorous enforcement of hand hygiene, early discharge of infected infants, and thorough cleaning of all possible fomites in the nursery and delivery rooms are important elements of control. If cases continue to occur, the nursery should be closed to further admissions. Cultures of nursery personnel are likely to be helpful in the unusual situation of an S. typhi outbreak in which a chronic carrier may be among the caretakers. Culture of health care personnel during outbreaks of salmonellosis caused by other Salmonella species is debatable, although often recommended. Data suggest that nurses infected with Salmonella rarely infect patients in the hospital setting.597 The fact that nursing personnel are sometimes found to be colonized during nursery outbreaks468, 474, 487, 489, 490 may be a result rather than a cause of those epidemics.
The potential role of vaccines in control of neonatal disease is minimal. For the vast number of non–S. typhi serotypes, there is no prospect for an immunization strategy. Multiple doses of the commercially available oral live attenuated vaccine (Ty21a; Vivotif, Berna), has been shown in Chilean schoolchildren to reduce typhoid fever cases by more than 70%.598, 599 However, the vaccine is not recommended for persons younger than 6 years, in part because immunogenicity of Ty21a is age dependent; children younger than 24 months fail to respond with development of immunity.600 Vi capsular polysaccharide vaccine is available for children older than 2 years and is effective in a single dose. Whether some degree of protection of infants could occur if stool carriage were reduced or could be transferred to infants by the milk of vaccinated mothers’ remains to be studied. Data suggest that breast-feeding may decrease the risk of other Salmonella infections.601
SHIGELLA
Nature of the Organism
On the basis of DNA relatedness, shigellae and E. coli organisms belong to the same species.602 However, for historical reasons and because of their medical significance, shigellae have been maintained as separate species. Shigellae are gram-negative bacilli that are unlike typical E. coli in that they do not metabolize lactose or do so slowly, are nonmotile, and generally produce no gas during carbohydrate use. They are classically divided into four species (serogroups) on the basis of metabolic and antigenic characteristics (Table 20–4). The mannitol nonfermenters usually are classified as S. dysenteriae. Although the lipopolysaccharide antigens of the 13 recognized members of this group are not related to each other antigenically, these serotypes are grouped together as serogroup A. Serogroup D (Shigella sonnei) are ornithine decarboxylase positive and slow lactose fermenters. All S. sonnei share the same lipopolysaccharide (O antigen). Those shigellae that ferment mannitol (unlike S. dysenteriae) but do not decarboxylate ornithine or ferment lactose (S. sonnei) belong to serogroups B and C. Of these, the strains that have lipopolysaccharide antigens immunologically related to each other are grouped together as serogroup B (Shigella flexneri), whereas those whose O antigens are not related to each other or to other shigellae are included in serogroup C (Shigella boydii). There are six major serotypes of S. flexneri and 13 subserotypes (1a, 1b, 2a, 2b, 3a, 3b, 4a, 4b, 5a, 5b, 6, X and Y variant). There are 19 antigenically distinct serotypes of S. boydii. For S. dysenteriae and S. boydii serogroup confirmation, pools of polyvalent antisera are used.
Table 20-4.
Shigella Serogroups
| Serogroups | Species | No. of Serotypes |
|---|---|---|
| A | S. dysenteriae | 13 |
| B | S. flexneri | 15 (including subtypes) |
| C | S. boydii | 18 |
| D | S. sonnei | 1 |
The virulence of shigellae has been studied extensively since their recognition as major pathogens at the beginning of the 20th century. The major determinants of virulence are encoded by a 120- to 140-MDa plasmid.603, 604 This plasmid, which is found in all virulent shigellae, encodes the synthesis of proteins that are required for invasion of mammalian cells and for the vigorous inflammatory response that is characteristic of the disease.605, 606 Shigellae that have lost this plasmid, have deletions of genetic material from the region involved in synthesis of these proteins, or have the plasmid inserted into the chromosome lose the ability to invade eukaryotic cells and become avirulent607; maintenance of the plasmid can be detected in the clinical microbiology lab by ability to bind Congo red. The ability to invade cells is the basic pathogenic property shared by all shigellae608, 609 and by the Shigella-like invasive E. coli, which also possesses the Shigella virulence plasmid.205, 605, 606, 610, 611 In the laboratory, Shigella invasiveness is studied in tissue culture (HeLa cell invasion), in animal intestine, or in rabbit or guinea pig eye, where instillation of the organism causes keratoconjunctivitis (Sereny test).128 Animal model studies have shown that bacteria penetrate and kill colonic mucosal cells and then elicit a brisk inflammatory response.
In addition to the virulence plasmid, several chromosomal loci enhance virulence.612, 613 This has been best studied in S. flexneri in which multiple virulence-enhancing regions of the chromosome have been defined.604, 612, 613, 614 The specific gene products of some of the chromosomal loci are not known; one chromosomal virulence segment encodes for synthesis of the O repeat units of lipopolysaccharide. Intact lipopolysaccharide is necessary but not sufficient to cause virulence.612, 615 At least two cell-damaging cytotoxins that also are chromosomally encoded are produced by shigellae. One of these toxins (Shiga toxin) is made in large quantities by S. dysenteriae serotype 1 (the Shiga bacillus) and is made infrequently by other shigellae.616 Shiga toxin is a major virulence factor in S. dysenteriae, enhancing virulence at the colonic mucosa and also giving rise to sequelae similar to those caused by STEC (discussed earlier). This toxin kills cells by interfering with peptide elongation during protein synthesis.617, 618, 619 Additional toxins may also be secreted by shigellae, although their roles in virulence are not established.620
Epidemiology
Although much of the epidemiology of shigellosis is predictable based on its infectious dose, certain elements are unexplained. Shigellae, like other organisms transmitted by the fecal-oral route, are commonly spread by food and water, but the low infecting inoculum allows person-to-person spread. Because of this low inoculum, Shigella is one of the few enteric pathogens that can infect swimmers.621 The dose required to cause illness in adult volunteers is as low as 10 organisms for S. dysenteriae serotype 1,622 about 200 organisms for S. flexneri, 623 and 500 organisms for S. sonnei. 624 Person-to-person transmission of infection probably explains the continuing occurrence of Shigella in the developed world. Enteropathogens that require large inocula and hence are best spread by food or drinking water are less common in industrialized societies because of sewage disposal facilities, water treatment, and food-handling practices. In the United States, daycare centers currently serve as a major focus for acquisition of shigellosis.625 Numerous outbreaks of shigellosis related to crowding, poor sanitation, and the low dose required for diseases have occurred in this setting.
Given the ease of transmission, it is not surprising that the peak incidence of disease is in the first 4 years of life. It is, however, paradoxical that symptomatic infection is uncommon in the first year of life.626, 627, 628, 629 The best data on the age-related incidence of shigellosis come from Mata’s625 prospective studies of Guatemalan infants. In these studies, stool cultures were performed weekly on a group of children followed from birth to 3 years old. The rate of infection was more than 60-fold lower in the first 6 months of life than between 2 and 3 years (Fig. 20–1).626 The same age-related incidence has been described in the United States629 and in a rural Egyptian village.628 This anomaly has been explained by the salutary effects of breast-feeding.630, 631, 632 However, it is likely that breast-feeding alone does not explain the resistance of infants to shigellosis.
Figure 20-1.

Age-related incidence of Shigella infection.
(Data from Mata LG: The Children of Santa Maria Cauque: A Prospective Field Study of Health and Growth. Cambridge, Mass, MIT Press, 1978.)
A review of three large case series633, 634, 635 suggests that about 1.6% (35 of 2225) of shigellosis cases occur in infants in the neonatal period. The largest series of neonatal shigellosis632 suggests that the course, complications, and etiologic serogroups are different in neonates than in older children. Although newborns are routinely contaminated by maternal feces, neonatal shigellosis is rare.
Other aspects of the epidemiology of shigellosis elude simple explanation. The seasonality (summer-fall peak in the United States, rainy season peak in the tropics) is not well explained. The geographic variation in species causing infection likewise is not well understood. In the United States, most Shigella infections are caused by S. sonnei or, less commonly, S. flexneri. In most of the developing world, the relative importance of these two species is reversed, and other Shigella serotypes, especially S. dysenteriae serotype 1, are identified more frequently. As hygiene improves, the proportion of S. sonnei increases and that of S. flexneri decreases.636 Data from Bangladesh suggest that S. dysenteriae is less common in neonates, but S. sonnei and S. boydii are more common.632
Clinical Manifestations
There appear to be some important differences in the relative frequencies of various complications of Shigella infection related to age. Some of these differences and estimates are based on data that are undoubtedly compromised by reporting biases. S. dysenteriae serotype 1 characteristically causes a more severe illness than other shigellae with more complications, including pseudomembranous colitis, hemolysis, and HUS. However, illnesses caused by various Shigella serotypes usually are indistinguishable from each other and conventionally are discussed together.
The incubation period of shigellosis is related to the number of organisms ingested, but in general, it is between 12 and 48 hours. Volunteer studies have shown that after ingestion, illness may be delayed for a week or more. Neonatal shigellosis seems to have a similar incubation period. More than one half of the neonatal cases occur within 3 days of birth, consistent with fecal-oral transmission during parturition. Mothers of infected neonates are sometimes carriers, although more typically they are symptomatic during the perinatal period. Intrauterine infection is rare. In the older child, the initial signs are usually high fever, abdominal pain, vomiting, toxicity, and large-volume watery stools; diarrhea may be bloody or may become bloody. Painful defecation and severe, crampy abdominal pain associated with frequent passage of small-volume stools with gross blood and mucus are characteristic findings in older children or adults who develop severe colitis. Many children, however, never develop bloody diarrhea. Adult volunteer studies have demonstrated that variations in presentation and course are not related to the dose ingested because some patients develop colitis with dysentery but others develop only watery diarrhea after ingestion of the same inoculum.623 The neonate with shigellosis may have a mild diarrheal syndrome or a severe colitis.633, 637, 638, 639, 640, 641, 642, 643, 644, 645 Fever in neonates is usually low grade (<102°F) if the course is uncomplicated. The neonate has less bloody diarrhea, more dehydration, more bacteremia, and a greater likelihood of death than the older child.632 Physical examination of the neonate may show signs of toxicity and dehydration, although fever, abdominal tenderness, and rectal findings are less striking than in the older child.634
Complications of shigellosis are common.646 Although the illness is self-limited in the normal host, resolution may be delayed for a week or more. In neonates and malnourished children, chronic diarrhea may follow a bout of shigellosis.637, 645 Between 10% and 35% of hospitalized children with Shigella have convulsions before or during the course of diarrhea.646, 647, 648 Usually, the seizures are brief, generalized, and associated with high fever. Seizures are uncommon in the first 6 months of life, although neonates have been described with seizures.639, 649 The cerebrospinal fluid generally reveals normal values in these children, but a few have mild cerebrospinal fluid pleocytosis. The neurologic outcome generally is good even with focal or prolonged seizures, but fatalities do occasionally occur, often associated with toxic encephalopathy.650 Although the seizures had been postulated to result from the neurotoxicity of Shiga toxin, this explanation was proved to be incorrect because most shigellae make little or no Shiga toxin and the strains isolated from children with neurologic symptoms do not produce Shiga toxin.616, 651 Hemolysis with or without development of uremia is a complication primarily of S. dysenteriae serotype 1 infection.652
Sepsis during the course of shigellosis may be caused by the Shigella itself or by other gut flora that gain access to the bloodstream through damaged mucosa.632, 653, 654 The risk of sepsis is higher in the first year of life, particularly in neonates,632, 637, 638, 639, 649, 656 in malnourished children, and in those with S. dysenteriae serotype 1 infection.654 Sepsis occurs in up to 12% of neonates with shigellosis.631, 646, 653, 655 Given the infrequency of neonatal shigellosis, it is striking that 9% of reported cases of Shigella sepsis have involved infants in the first month of life.657 One of the infants with bacteremia658 reportedly had no discernible illness. Disseminated intravascular coagulation may develop in those patients whose course is complicated by sepsis. Meningitis has been described in a septic neonate. Colonic perforation has occurred in neonates,630, 659 older children,660 and adults.661 Although this complication of toxic megacolon is rare, it appears to be more common in neonates than in older individuals. Bronchopneumonia may complicate the course of shigellosis, but shigellae are rarely isolated from lungs or tracheal secretions.662 The syndrome of sudden death in the setting of extreme toxicity with hyperpyrexia and convulsions but without dehydration or sepsis (i.e., Ekiri syndrome)663, 664, 665 is rare in neonates. In the nonbacteremic child, other extraintestinal foci of infection, including vagina666, 667 and eye,668 rarely occur. Reiter’s syndrome, which rarely complicates the illness in children, has not been reported in neonates.
Although infection is less common in infants than in toddlers, case fatality rates are highest in infants.669, 670 The mortality rate in newborns appears to be about twice that of older children.632 In industrialized societies, less than 1% of children with shigellosis die, whereas in developing countries, up to 30% die. These differences in mortality rates are related to nutrition,633 availability of medical care, antibiotic resistance of many shigellae, the frequency of sepsis, and the higher frequency of S. dysenteriae serotype 1 infection in the less-developed world.654
Diagnosis
Although the diagnosis of shigellosis can be suspected on clinical grounds, other enteropathogens can cause illnesses that are impossible to distinguish clinically. Shigellosis in the neonate is rare. The neonate with watery diarrhea is more likely to be infected with E. coli, Salmonella or rotavirus than Shigella. Infants presenting with bloody diarrhea may have necrotizing enterocolitis or infection with Salmonella, EIEC, Yersinia enterocolitica, C. jejuni, or Entamoeba histolytica. Before cultures establish a diagnosis, clinical and laboratory data may aid in making a presumptive diagnosis. Abdominal radiographs demonstrating pneumatosis intestinalis suggest the diagnosis of necrotizing enterocolitis. A history of several weeks of illness not associated with fever and with few fecal leukocytes suggests E. histolytica rather than Shigella infection.671
The definitive diagnosis of shigellosis depends on isolation of the organism from stool. Unfortunately, culture may be insensitive.672 In volunteer studies, daily stool cultures failed to detect shigellae in about 20% of symptomatic subjects.623 Optimal recovery is achieved by immediate inoculation of stool (as opposed to rectal swabs) onto culture media. Use of transport media in general decreases the yield of cultures positive for Shigella 673 when compared with immediate inoculation.
Examination of stool for leukocytes as an indication of colitis is useful in support of the clinical suspicion of shigellosis. The white blood cell count and differential count also are used as supporting evidence for the diagnosis. Leukemoid reactions (white blood cells > 50,000/mm3) occur in almost 15% of children with S. dysenteriae serotype 1 but in less than 2% of children with other shigellae.651 Leukemoid reactions are more frequent in infants than in older children.652 Even when the total white blood cell count is not dramatically elevated, there may be a striking left shift. Almost 30% of children with shigellosis have greater than 25% bands on the differential cell count.674, 675, 676 Few reports address the white blood cell count in newborns, but those that do suggest that normal or low rather than elevated counts are more common. Although serum and fecal antibodies develop to lipopolysaccharides and the virulence plasmid-associated polypeptides,677 serologic studies are not useful in the diagnosis of shigellosis. PCR can identify Shigella and EIEC in feces.678 Colonoscopy typically shows inflammatory changes that are most severe in the distal segments of colon.679
Therapy
Because dehydration is particularly common in neonatal shigellosis, attention to correction of fluid and electrolyte disturbances is always the first concern when the illness is suspected. Although debate continues over the indications for antimicrobial therapy in the patient with shigellosis, the benefits of therapy generally appear to outweigh the risks. The chief disadvantages of antimicrobial therapy include cost, drug toxicity, and emergence of antibiotic-resistant shigellae. Because of the self-limited nature of shigellosis, it has been argued that less severe illness should not be treated. However, children can feel quite ill during the typical bout of shigellosis, and appropriate antimicrobial therapy shortens the duration of illness and eliminates shigellae from stool, decreasing secondary spread. Complications are probably decreased by antibiotics. Given the high mortality rates of neonatal shigellosis, therapy should not be withheld.
The empirical choice of an antimicrobial agent is dictated by susceptibility data for strains circulating in the community at the time the patient’s infection occurs. Multiresistant shigellae complicate the choice of empirical therapy before availability of susceptibility data for the patient’s isolate. Plasmid-encoded resistance (R factors) for multiple antibiotics has been observed frequently in S. dysenteriae serotype 1 outbreaks682 and with other shigellae.683, 684, 685 Antimicrobial resistance patterns fluctuate from year to year in a given locale.686 However, despite the guesswork involved, early preemptive therapy is indicated when an illness is strongly suggestive of shigellosis. In vitro susceptibility does not always adequately predict therapeutic responses. Cefaclor,687 furazolidone,688 cephalexin,689 amoxicillin,690 kanamycin,691 and cefamandole692 all are relatively ineffective agents.
The optimal duration of therapy is debatable. Studies in children older than 2 years and in adults suggest that single-dose regimens may be as effective in relieving symptoms as courses given for 5 days. The single-dose regimens generally are not as effective in eliminating shigellae from the feces as are the longer courses. A third-generation cephalosporin, such as ceftriaxone, may be the best empirical choice. Optimal doses for newborns with shigellosis have not been established. Trimethoprim at a dose of 10mg/kg/day (maximum, 160mg/day) and sulfamethoxazole at a dose of 50mg/kg/day (maximum, 800mg/day) in two divided doses for a total of 5 days are recommended for the older child if the organism is susceptible.693, 694, 695 If the condition of the infant does not permit orally administration, the drug usually is divided into three doses given intravenously over 1 hour.696 Ampicillin at a dose of 100mg/kg/day in four divided doses taken orally for 5 days may be used if the strain is susceptible.676
For the rare newborn who acquires shigellosis, appropriate therapy often is delayed until susceptibility data are available. This occurs because shigellosis is so rare in newborns that it is almost never the presumptive diagnosis of the child with watery or bloody diarrhea. Although a sulfonamide is as efficacious as ampicillin when the infecting strain is susceptible,675 sulfonamides are avoided in neonates because of concern about the potential risk of kernicterus. The risk of empirical ampicillin therapy is that shigellae are frequently resistant to the drug; 50% of shigellae currently circulating in the United States are ampicillin resistant.696, 697 For the neonate infected with ampicillin-resistant Shigella, there are few data on which to base a recommendation. Ceftriaxone is generally active against shigellae, but in the neonate, this drug can displace bilirubin-binding sites and elicit clinically significant cholestasis. Data on children and adults suggest that clinical improvement occurs with ceftriaxone.698, 699 Quinolones, such as ciprofloxacin and ofloxacin, have been shown to be effective agents for treating shigellosis700, 701 in adults, but they are not approved for use in children younger than 18 years. Other drugs sometimes used to treat diarrhea pose special risks to the infant with shigellosis. The antimotility agents, in addition to their intoxication risk, may pose a special danger in dysentery. In adults, diphenoxylate hydrochloride with atropine has been shown to prolong fever and excretion of the organism.702
The response to appropriate antibiotic therapy is generally gratifying. Improvement is often obvious in less than 24 hours. Complete resolution of diarrhea may not occur until a week or more after the start of treatment. In those who have severe colitis or those infected by S. dysenteriae serotype 1, the response to treatment is somewhat delayed.
Prevention
For most of the developing world, the best strategy for prevention of shigellosis during infancy is prolonged breast-feeding. Specific antibodies in milk appear to prevent symptomatic shigellosis66, 68; nonspecific modification of gut flora and the lack of bacterial contamination of human milk also may be important. Breast-feeding, even when other foods are consumed, decreases the risk of shigellosis; children who continue to consume human milk into the third year of life are still partially protected from illness.703 In the United States, the best means of preventing infection in the infant is good hand hygiene when an older sibling or parent develops diarrhea. Even in unsanitary environments, secondary spread of shigellae can be dramatically decreased by hand hygiene after defecation and before meals.704 Spread of shigellae in the hospital nursery can presumably be prevented by the use of contact isolation for infants with diarrhea and attention to thorough hand hygiene. Although nursery personnel have acquired shigellosis from infected newborns,685 further transmission to other infants in the nursery, although described,705 is rare. In contrast to Salmonella, large outbreaks of nosocomial shigellosis in neonates are rare.
Unfortunately, good hygiene is a particularly difficult problem in daycare centers. The gathering of susceptible children, breakdown in hand hygiene, failure to use different personnel for food preparation and diaper changing, and difficulty controlling the behavior of toddlers all contribute to daycare-focused outbreaks of shigellosis.
Immunization strategies have been studied since the turn of the 20th century, but no satisfactory immunization has been developed. Even if immunizations are improved, a role in managing neonates seems unlikely.
CAMPYLOBACTER
Nature of the Organism
Campylobacter was first recognized in an aborted sheep fetus in the early 1900s706 and was named Vibrio fetus by Smith and Taylor in 1919.707 This organism subsequently was identified as a major venereally transmitted cause of abortion and sterility and as a cause of scours in cattle, sheep, and goats.708, 709 It was not until 1947, when it was isolated from the blood culture of a pregnant woman who subsequently aborted at 6 months’ gestation, that the significance of Campylobacter as a relatively rare cause of bacteremia and perinatal infections in humans was appreciated.710, 711, 712 During the 1970s, Campylobacter was recognized to be an opportunistic pathogen in debilitated patients.713, 714 In 1963, V. fetus and related organisms were separated from the vibrios (such as V. cholerae and V. parahaemolyticus) and placed in a new genus, Campylobacter (Greek word for “curved rod”).715 Since 1973, several Campylobacter species have been recognized as a common cause of enteritis716, 717, 718, 719, 720, 721, 722, 723, 724, 725, 726, 727, 728, 729, 730, 731, 732 and, in some cases, extraintestinal infections.
The genus Campylobacter contains 15 species, most of which are recognized as animal and human pathogens. The most commonly considered causes of human disease are Campylobacter fetus, Campylobacter jejuni, Campylobacter coli, Campylobacter lari, and Campylobacter upsaliensis (Table 20–5),730, 731, 732 although Campylobacter mucosalis has been isolated from stool of children with diarrhea.733 DNA hybridization studies have shown that these species are distinct, sharing less than 35% DNA homology under stringent hybridization conditions.734, 735 Helicobacter pylori was originally named Campylobacter pylori, but because of differences in DNA, it was reclassified and is no longer considered in the Campylobacter genus.
Table 20-5.
Campylobacter Species That Infect Humans
| Current Nomenclature | Previous Nomenclature | Usual Disease Produced |
|---|---|---|
| C. fetus | Vibrio fetus | Bacteremia, meningitis, perinatal infection, intravascular infection |
| V. fetus var. intestinalis | ||
| C. fetus subsp. intestinalis | ||
| C. jejuni | Vibrio jejuni | Diarrhea |
| C. coli | C. fetus subsp. jejuni | Diarrhea |
| C. lari | Grouped with C. jejuni, nalidixic acid–resistant, thermophilic Campylobacter, C. laridis I | Diarrhea, bacteremia |
| C. upsaliensis | None | Diarrhea, bacteremia |
| C. hyointestinalis | None | Diarrhea, bacteremia |
| C. concisus | None | Diarrhea |
Strains of C. fetus are divided into two subspecies: C. fetus subsp. fetus and C. fetus subsp. venerealis. The first subspecies causes sporadic abortion in cattle and sheep736, 737; in the human fetus and newborn, it causes perinatal and neonatal infections that result in abortion, premature delivery, bacteremia, and meningitis.710, 711, 712, 736, 737, 738, 739, 740, 741, 742, 743, 744, 745, 746, 747, 748 Outside the newborn period, Campylobacter is a relatively infrequent cause of bacteremia, usually infecting those with impaired host defenses, including the elderly or the debilitated; less frequently, it causes intravascular infection.713, 714, 715, 749, 750
By far the most common syndrome caused by a Campylobacter species is enteritis. C. jejuni and C. coli cause gastroenteritis and generally are referred to collectively as C. jejuni, although DNA hybridization studies show them to be different. In the laboratory, C. jejuni can be differentiated from C. coli because it is capable of hydrolyzing hippurate, whereas C. coli is not. Most isolates that are associated with diarrhea (61% to 100%) are identified as C. jejuni, 751, 752, 753, 754 and in some cases, individuals have been shown to be simultaneously infected with C. jejuni and C. coli. 752
Because of the fastidious nature of C. jejuni, which is difficult to isolate from fecal flora, its widespread occurrence was not recognized until 1973.716, 717, 718, 719, 720, 721, 722, 723, 724, 725, 726, 727, 728, 729, 730, 731, 732 Previously called related vibrios by King,749 this organism had been associated with bloody diarrhea and colitis in infants and adults only when it had been associated with a recognized bacteremia.755, 756, 757 In the late 1970s, development of selective fecal culture methods for C. jejuni enabled its recognition worldwide as one of the most common causes of enteritis in persons of all ages. It is an uncommon infection in neonates who generally develop gastroenteritis when infected.718, 758, 759, 760, 761, 762, 763, 764, 765, 766, 767, 768, 769, 770, 771, 772, 773, 774, 775, 776, 777, 778, 779, 780, 781, 782, 783, 784, 785, 786, 787, 788, 789, 790, 791, 792, 793, 794, 795, 796, 797, 798, 799, 800, 801, 802, 803, 804, 805, 806, 807, 808, 809, 810, 811, 812, 813, 814, 815, 816, 817, 818, 819, 820, 821, 822, 823, 824, 825, 826, 827, 828, 829, 830, 831, 832, 833, 834, 835, 836, 837, 838, 839, 840, 841, 842, 843, 844, 845, 846, 847, 848, 849, 850, 851, 852, 853, 854, 855, 856, 857, 858, 859, 860, 861, 862, 863, 864, 865, 866, 867, 868, 869, 870, 871, 872, 873, 874, 875, 876, 877, 878, 879, 880, 881 Bacteremia with C. jejuni enteritis also is uncommon.718, 759, 764, 769, 772, 773, 774, 775, 776, 777, 778 Maternal symptoms considered to be related to C. jejuni infection generally are mild and include fever (75%) and diarrhea (30%). In contrast to the serious disease in newborns that is caused by C. fetus, neonatal infections with C. jejuni usually result in a mild illness,758, 760, 762, 763, 765, 766, 767, 768, 771, 778 although meningitis occurs in rare instances.761, 769 Third trimester infection related to C. fetus or C. jejuni may results in abortion or stillbirth.
Pathogenesis
C. fetus does not produce recognized enterotoxins or cytotoxins and does not appear to be locally invasive by the Sereny test.714, 731 Instead, these infections may be associated with penetration of the organism through a relatively intact intestinal mucosa to the reticuloendothelial system and bloodstream.714 Whether this reflects a capacity to resist serum factors or to multiply intracellularly remains to be determined.
C. jejuni is capable of producing illness by several mechanisms. These organisms have been shown to produce an LT enterotoxin and a cytotoxin.779, 780, 781, 782 This enterotoxin is known to be a heat-labile protein with a molecular mass of 60 to 70 MDa.779, 782 It shares functional and immunologic properties with cholera toxin and E. coli LT. C. jejuni and C. coli also elaborate a cytotoxin that is toxic for a number of mammalian cells.783, 784, 785 The toxin is heat labile, trypsin sensitive, and not neutralized by immune sera to Shiga toxin or the cytotoxin of Clostridium difficile. The role of these toxins as virulence factors in diarrheal disease remains unproved.779, 784
Several animal models have been tested for use in the study of this pathogen.786 Potential models for the study of C. jejuni enteritis include dogs, which may acquire symptomatic infection787; 3- to 8-day old chicks788, 789, 790; chicken embryo cells, which are readily invaded by C. jejuni 725; rhesus monkeys791; and rabbits by means of the removable intestinal tie adult rabbit technique. An established small mammal model that mimics human disease in the absence of previous treatment or surgical procedure has not been successful in adult mice.792 An infant mouse model793, 794 and a hamster model795 of diarrhea appear promising. C. jejuni is negative in the Sereny test for invasiveness,796 and most investigators report no fluid accumulation in ligated rabbit ileal loops.
Pathology
The pathologic findings of C. fetus infection in the perinatal period include placental necrosis711 and, in the neonate, widespread endothelial proliferation, intravascular fibrin deposition, perivascular inflammation, and hemorrhagic necrosis in the brain.797 The tendency for intravascular location and hepatosplenomegaly in adults infected with C. fetus has been shown.714
The pathologic findings in infants and children infected with C. fetus can include an acute inflammatory process in the colon or rectum, as evidenced by the tendency for patients to have bloody diarrhea with numerous fecal leukocytes.798 There also can be crypt abscess formation and an ulcerative colitis or pseudomembranous colitis–like appearance799, 800 or a hemorrhagic jejunitis or ileitis.717, 725, 801, 802 Mesenteric lymphadenitis, ileocolitis and acute appendicitis also have been described.
Epidemiology
Infection with Campylobacter species occurs after ingestion of contaminated food, including unpasteurized milk, poultry, and contaminated water.726, 803, 804, 805, 806, 807, 808, 809, 810, 811, 812 Many farm animals and pets, such as chickens,813 dogs,814, 815 and cats (especially young animals), are potential sources. The intrafamilial spread of infection in households,717, 816 the occurrence of outbreaks in nurseries,769, 770, 817 and the apparent laboratory acquisition of C. jejuni 818 all suggest that C. jejuni infection may occur after person-to-person transmission of the organism. Outbreaks of C. jejuni in the child daycare setting are not common. Volunteer studies819 have shown a variable range in the infecting dose, with many volunteers developing no illness. The report of illness after ingestion of 106 organisms in a glass of milk728 and production of illness in a single volunteer by 500 organisms819 substantiate the variation in individual susceptibility. The potential for low-inoculum disease has significant implications for the importance of strict enteric precautions when infected persons are hospitalized, particularly in maternity and nursery areas. When diarrhea in neonates caused by C. jejuni has been reported,758, 759, 760, 761, 762, 763, 764, 765, 766, 767, 768, 769, 770, 771 maternal-infant transmission during labor has generally been documented.758, 759, 760, 761, 762, 763, 765, 766, 767, 768, 770, 771 The Lior serotyping system, restriction length polymorphism, and pulse-field gel electrophoresis 820 have been used to confirm the identity of the infant and maternal isolates. Most mothers gave no history of diarrhea during pregnancy.762, 763, 765, 766 Outbreaks have occurred in neonatal intensive care units because of person-to-person spread.820
The frequency of asymptomatic carriage of C. jejuni ranges from 0% to 1.3%716, 717 to as high as 13% to 85%.716, 717, 732, 821, 822, 823 In a cohort study in Mexico, 66% of all infections related to C. jejuni were asymptomatic.732 Infected children, if untreated, can be expected to excrete the organisms for 3 or 4 weeks; however, more than 80% are culture negative after 5 weeks.721, 722 Asymptomatic excreters pose a significant risk in the neonatal period, in which acquisition from an infected mother can be clinically important.718, 760, 762, 766 C. jejuni has increasingly been recognized as a cause of watery and inflammatory diarrhea in temperate and tropical climates throughout the world. It has been isolated from 2% to 11% of all fecal cultures from patients with diarrheal illnesses in various parts of the world.716, 717, 718, 719, 720, 721, 722, 723, 724, 728, 824, 825, 826, 827, 828, 829 There is a tendency for C. jejuni enteritis to occur in the summer in countries with temperate climates.825
The reservoir of Campylobacter is the gastrointestinal tract of domestic and wild birds and animals. It infects sheep, cattle, goats, antelope, swine, chickens, domestic turkeys, and pet dogs. C. fetus often is carried asymptomatically in the intestinal or biliary tracts of sheep and cattle. During the course of a bacteremic illness in pregnant animals, C. fetus organisms, which have a high affinity for placental tissue, invade the uterus and multiply in the immunologically immature fetus. The infected fetuses generally are aborted. Whether this organism is acquired by humans from animals or is carried asymptomatically for long periods in humans, who may then transmit the organism through sexual contact as appears to occur in animals, is unclear. It is believed that this subspecies rarely is found in the human intestine and that it is not a cause of human enteritis.725 C. fetus infections predominantly occur in older men with a history of farm or animal exposure and in pregnant women in their third trimester.710, 711, 716, 717 Symptomatically or asymptomatically infected women may have recurrent abortions or premature deliveries and are the source of organisms associated with life-threatening perinatal infections of the fetus or newborn infant.710, 739, 740, 741, 742, 743, 744, 745, 746, 747, 748, 830 In several instances of neonatal sepsis and meningitis, C. fetus was isolated from culture of maternal cervix or vagina.712, 747, 795 A nosocomial nursery outbreak has been associated with carriage in some healthy infants.831 Other outbreaks have been associated with meningitis 832, 833 Cervical cultures have remained positive in women who have had recurrent abortions and whose husbands have antibody titer elevations.738
The most commonly incriminated reservoir of C. jejuni is poultry.808, 812, 834, 835 Most chickens in several different geographic locations had a large number (mean, 4 × 106/g) of C. jejuni in the lower intestinal tract or feces. This occurred in some instances despite the use of tetracycline, to which the Campylobacter was susceptible in vitro, in the chicken feed.828 The internal cavities of chickens remain positive for Campylobacter even after they have been cleaned, packaged, and frozen.834 However, unlike Salmonella, C. jejuni organisms that survive usually do not multiply to high concentrations.725 Domestic puppies or kittens with C. jejuni diarrhea also can provide a source for spread, especially to infants or small children.717, 755, 812, 836, 837, 838
C. jejuni enteritis also has been associated in a number of outbreaks with consumption of unpasteurized milk.725, 809, 810, 811, 839, 840, 841 In retrospect, the first reported human cases of C. jejuni enteritis were probably in a milk-borne outbreak reported in 1946.842 Because Campylobacter infections of the udder are not seen, milk is probably contaminated from fecal shedding of the organism. These organisms are killed by adequate heating.
Fecally contaminated water is a potential vehicle for C. jejuni infections.843 Several phenotypic and genotypic methods have been used for distinguishing C. jejuni strains from animals and humans involved in epidemics.844 C. jejuni is associated with traveler’s diarrhea among those traveling from England or the United States.720
Clinical Manifestations
Clinical manifestations of infection caused by Campylobacter depend on the species involved (see Table 20–5). Human infections with C. fetus are rare and generally are limited to bacteremia in patients with predisposing conditions749, 750 or to bacteremia or uterine infections with prolonged fever and pneumonitis that lasts for several weeks in women during the third trimester of pregnancy. Unless appropriately treated, symptoms usually resolve only after abortion or delivery of an infected infant.710, 712, 739, 740, 741, 742, 743, 744, 745, 746, 747, 748, 750 These infected neonates, who are often premature, develop signs suggesting sepsis, including fever, cough, respiratory distress, vomiting, diarrhea, cyanosis, convulsions, and jaundice. The condition typically progresses to meningitis, which may be rapidly fatal or may result in serious neurologic sequelae.712 Additional systemic manifestations include pericarditis, pneumonia, peritonitis, salpingitis, septic arthritis, and abscesses.823
C. jejuni infection typically involves the gastrointestinal tract, producing watery diarrhea or a dysentery-like illness with fever and abdominal pain and stools that contain blood and mucus.715, 732, 800 Older infants and children generally are affected, but neonates with diarrhea have been reported. Infection in neonates generally is not clinically apparent or is mild. Stools can contain blood, mucus, and pus712, 721, 762, 763; fever often is absent.721, 762 The illness usually responds to appropriate antimicrobial therapy,760, 762, 816 which shortens the period of fecal shedding.845 Extraintestinal infections related to C. jejuni other than bacteremia are rare but include cholecystitis,846 urinary tract infection,847 and meningitis.761 Bacteremia is a complication of gastrointestinal infection,848 especially in malnourished children.849 Meningitis that appears to occur secondary to intestinal infection also has been reported in premature infants who have had intraventricular needle aspirations for neonatal hydrocephalus.712 Complications in older children and adults that have been associated with C. jejuni enteritis include Reiter’s syndrome,850 Guillain-Barré syndrome,851, 852 and reactive arthritis.853, 854 Persistent C. jejuni infections have been described in patients infected with human immunodeficiency virus.855 Extraintestinal manifestations generally occur in patients who are immunosuppressed or at the extremes of age.714 Campylobacter lari has caused chronic diarrhea and bacteremia in a neonate.856
Diagnosis
Most important in the diagnosis of Campylobacter infection is a high index of suspicion based on clinical grounds. C. fetus and C. jejuni are fastidious and may be overlooked on routine fecal cultures. Isolation of Campylobacter from blood or other sterile body sites does not represent the same problem as isolation from stool. Growth occurs with standard blood culture media, but it may be slow. In the case of C. fetus infecting the bloodstream or central nervous system, blood culture flasks should be blindly subcultured and held for at least 7 days or the organism may not be detected because of slow or inapparent growth.742 The diagnosis of C. fetus infection should be considered when there is an unexplained febrile illness in the third trimester of pregnancy or in the event of recurrent abortion, prematurity, or neonatal sepsis with or without meningitis. A high index of suspicion and prompt, appropriate antimicrobial therapy may prevent the potentially serious neonatal complications that may follow maternal C. fetus infection.
Campylobacter is distinguished from the Vibrio organisms by its characteristics of carbohydrate nonfermentation and by its different nucleotide base composition.715, 733, 734, 735, 738 Campylobacter is 0.2 to 0.5 fm wide and 0.5 to 8.0 long. It is a fastidious, microaerophilic, curved, motile gram-negative bacillus that has a single polar flagellum and is oxidase and catalase positive, except for C. upsaliensis, which is generally catalase negative or weakly positive. C. jejuni and C. fetus are separated by growth temperature (C. fetus grows best at 25°C but can be cultured at 37°C; C. jejuni grows best at 42°C) and by nalidixic acid and cephalosporin susceptibilities, because C. jejuni is susceptible to nalidixic acid and resistant to cephalosporins. C. jejuni grows best in a microaerobic environment of 5% oxygen and 10% carbon dioxide at 42°C. It grows on a variety of media, including Brucella and Mueller-Hinton agars, but optimal isolation requires the addition of selective and nutritional supplements. Growth at 42° C in the presence of cephalosporins is used to culture selectively for C. jejuni from fecal specimens. In a study of six media, charcoal-based selective media and a modified charcoal cefoperazone deoxycholate agar were the most selective for identification of Campylobacter species. Extending the incubation time from 48 to 72 hours led to an increase in the isolation rate regardless of the medium used.857 Its typical darting motility may provide a clue to identification, even in fresh fecal specimens, when viewed by phase-contrast microscopy.721, 858
When the organism has been cultured, it is presumptively identified by motility and by its curved, sometimes sea gull– like appearance on carbolfuchsin stain. Polymorphonuclear leukocytes are usually found in stools when bloody diarrhea occurs and indicate the occurrence of colitis.762, 798 To avoid potentially serious C. jejuni infection in the newborn infant, careful histories of any diarrheal illnesses in the family should be obtained, and pregnant women with any enteric illness should have cultures for this and other enteric pathogens. Detection of C. jejuni and C. coli by PCR has been reported859 and in the future may be useful for the rapid and reliable identification of this organism.
The differential diagnosis of C. fetus infections include the numerous agents that cause neonatal sepsis or meningitis, especially gram-negative bacilli. Diagnostic considerations for inflammatory or bloody enteritis include necrotizing enterocolitis, allergic proctitis, and Salmonella; rarely Shigella, and other infectious agents occur. Agglutination, complement fixation, bactericidal, immunofluorescence, and ELISA tests have been used for serologic diagnosis of C. jejuni infection and to study the immune response, but these assays are of limited value in establishing the diagnosis during an acute infection.731
Therapy
The prognosis is grave in newborn infants with sepsis or meningitis caused by C. fetus. In infants with C. jejuni gastroenteritis, limited data suggest that appropriate, early antimicrobial therapy results in improvement and rapid clearance of the organism from stool.845 Campylobacter species are often resistant to β-lactams, including ampicillin and cephalosporins.860, 861 Most strains are susceptible to erythromycin, gentamicin, tetracycline, chloramphenicol, and the newer quinolones, although resistance to these agents has been reported.862, 863 It appears that a parenteral aminoglycoside is the drug of choice for C. fetus infections, pending in vitro susceptibility studies. In the case of central nervous system involvement, cefotaxime and chloramphenicol are potential alternative drugs. Depending on in vitro susceptibilities, which vary somewhat with locale, erythromycin is the drug of choice for treating C. jejuni enteritis.717, 721, 722 If erythromycin therapy is initiated within the first 4 days of illness, a reduction in excretion of the organism and resolution of symptoms occur.845 Although data regarding treatment of asymptomatic or convalescent carriers are not available, it seems appropriate to treat colonized pregnant women in the third trimester of pregnancy when there is a risk of perinatal or neonatal infection. The failure of prophylactic parenteral gentamicin in a premature infant has been documented, followed by successful resolution of symptoms and fecal shedding with erythromycin. Because there appears to be an increased risk of toxicity with erythromycin estolate during pregnancy and infancy,864 other forms of erythromycin should probably be used in these settings. Azithromycin appears to be effective if the organism is susceptible.865 Strains that are erythromycin resistant often are resistant to azithromycin.866 Campylobacter tends to have higher minimal inhibitory concentrations for clarithromycin than for azithromycin.867 Furazolidone has been used in children and ciprofloxacin in nonpregnant patients older than 17 years.
Prevention
Contact precautions should be employed during any acute diarrheal illness and until the diarrhea has subsided. Hand hygiene after handling raw poultry and washing cutting boards and utensils with soap and water after contact with raw poultry may decrease risk of infection. Pasteurization of milk and chlorination of water are critically important. Infected food handlers and hospital employees who are asymptomatic pose no known hazard for disease transmission if proper personal hygiene measures are maintained. Ingestion of human milk that contains anti–C. jejuni antibodies has been shown to protect infants from diarrhea due to C. jejuni. 63, 868
CLOSTRIDIUM DIFFICILE
Nature of the Organism and Pathophysiology
C. difficile is a spore-forming, gram-positive, anaerobic bacillus that produces two toxins. In the presence of antibiotic pressure, C. difficile colonic overgrowth and toxin production occur. The virulence properties of C. difficile are related to production of an enterotoxin that causes fluid secretion (toxin A) and a cytotoxin detectable by its cytopathic effects in tissue culture (toxin B).869, 870 Both toxin genes have been cloned and sequenced, revealing that they encode proteins with estimated molecular masses of 308kDa for toxin A and 270kDa for toxin B.871
A wide variety of antibacterial, antifungal, antituberculosis, and antineoplastic agents have been associated with C. difficile colitis, although penicillin, clindamycin, and cephalosporins are associated most frequently. Rarely, no precipitating drug has been given.872, 873, 874, 875, 876 C. difficile and its toxins can be demonstrated in up to one third of patients with antibiotic-associated diarrhea and in about 98% of patients with pseudomembranous colitis.877
Epidemiology
C. difficile can be isolated from soil and frequently exists in the hospital environment. Spores of C. difficile are acquired from the environment or by fecal-oral transmission from colonized individuals or from items in the environment such as thermometers and feeding tubes.878, 879, 880, 881, 882, 883 C. difficile has been demonstrated to persist on a contaminated floor for 5 months.879 Nosocomial spread is related to organisms on the hands of personnel879, 880, 884 and to contaminated surfaces, which may serve as reservoirs.885, 886 Although all groups are susceptible to infection, newborn infants represent a special problem. Less than 5% of healthy children older than 2 years887 and healthy adults carry C. difficile, 877 but more than 50% of neonates can be demonstrated to have C. difficile and its cytotoxin in their stools, usually in the absence of clinical findings.880, 886, 888, 889, 890 Infants in neonatal intensive care units have high rates of colonization, in part because of frequent use of antimicrobial agents in these units.889, 890 Clustering of infected infants suggests that much of the colonization of newborn infants represents nosocomial spread886 rather than acquisition of maternal flora. The number of C. difficile organisms present in stools of well infants is similar to that found in older patients with pseudomembranous colitis.890 The high frequency of colonization has led to justified skepticism about the pathogenic potential of this organism in the very young.891 Although some episodes of diarrhea in early infancy may be caused by C. difficile, 892 the diagnostic criteria used in older children and adults are inadequate to establish a definite diagnosis in this age group.
Clinical Manifestations
The usual manifestations of C. difficile disease in older children and adults include watery diarrhea, abdominal pain and tenderness, nausea, vomiting, and low-grade fever. Grossly bloody diarrhea is unusual, although occult fecal blood is common. Leukocytosis is present during severe illness. Diarrhea usually begins 4 to 9 days into a course of antimicrobial therapy but may be delayed until several weeks after completion of the therapeutic course. Usually, the illness is mild and self-limited if the offending drug is discontinued. Severe colitis with pseudomembranes is less common now than in previous years because the risk of diarrhea developing during antimicrobial therapy is recognized and the antimicrobial agent typically is stopped.
It is unclear whether this organism causes disease in newborns. One study from a newborn intensive care unit suggests that toxin A in stools is associated with an increased frequency of abnormal stools.893
Diagnosis
Endoscopic findings of pseudomembranes and hyperemic, friable rectal mucosa suggest the diagnosis of pseudomembranous colitis. Pseudomembranes are not always present in C. difficile colitis; mild cases are often described as nonspecific colitis. Several noninvasive techniques are used to establish the diagnosis, including enzyme immunoassay (EIA) for toxin detection and PCR.893, 894, 895, 896 Isolation of C. difficile from stool does not distinguish between toxigenic and nontoxigenic isolates. If C. difficile is isolated, testing for toxin by cell culture or EIA should be performed to confirm the presence of a toxigenic strain. There are multiple commercially available EIAs that detect either toxin A or both toxins A and B.893, 894, 895 These assays are sensitive and easy to perform. Other assays are available for epidemiologic investigation of outbreaks of disease due to C. difficile. 896
In older children and adults, the diagnosis is confirmed by culture of C. difficile and demonstration of toxin in feces. In neonates, these data are inadequate to prove that an illness is related to C. difficile. When the clinical picture is consistent, the stool studies are positive for C. difficile and no other cause for illness is found, a diagnosis of “possible” C. difficile is made. A favorable response to eradication of C. difficile is supportive evidence that the diagnosis is correct.872 Because of the uncertainty implicit in the ambiguity of neonatal diagnostic criteria, other diagnoses must be considered.
Therapy
When the decision is made that a neonate’s illness might be related to C. difficile, the initial approach should include fluid and electrolyte therapy and discontinuation of the offending antimicrobial agent. If the illness persists or worsens or if the patient has severe diarrhea, specific therapy with metronidazole886, 897 should be instituted. Metronidazole is considered to be the treatment of choice for most patients with C. difficile colitis.898 Rarely is there a need to consider orally administered vancomycin or bacitracin in neonates.899, 900
After initiation of therapy, signs of illness generally resolve within several days, titers decrease, and fecal toxins disappear eventually. Recurrence of colitis after discontinuation of metronidazole or vancomycin has been documented in 10% to 20% of adults.901 Relapses are treated with a second course of metronidazole or vancomycin. Drugs that decrease intestinal motility should not be administered.
Neutralizing antibody against C. difficile cytotoxin has been demonstrated in human colostrum.902 Secretory component of sIgA binds to toxin A to inhibit its binding to receptors 903 Data show that there are nonantibody factors present in milk that interfere with the action of toxin B in addition to secretory IgA directed at toxin A.904 Breast-feeding appears to decrease the frequency of colonization by C. difficile. 905
Prevention
In addition to standard precautions, contact precautions are recommended for the duration of illness. Meticulous hand hygiene techniques, proper handling of contaminated waste and fomites, and limiting the use of antimicrobial agents are the best available methods for control of C. difficile infection.
VIBRIO CHOLERAE
Nature of the Organism
V. cholerae is a gram-negative, curved bacillus with a polar flagellum. Of the many serotypes, only enterotoxin-producing organisms of serotype O1 and O139 cause epidemics. V. cholerae O1 is divided into two serotypes, Inaba and Ogawa, and two biotypes, classic and E1 Tor; the latter is the predominant biotype. Nontoxigenic O1 strains and non-O1 strains of V. cholerae can cause diarrhea and sepsis but do not cause outbreaks.906, 907, 908
Pathogenesis
V. cholerae O group 1 is the classic example of an enteropathogen whose virulence is caused by enterotoxin production. Cholera toxin is an 84-MDa protein whose five B subunits cause toxin binding to the enterocyte membrane ganglioside GM1 and whose A subunit causes adenosine diphosphate ribosylation of a guanosine triphosphate–binding regulatory subunit of adenylate cyclase.105, 908 The elevated cAMP levels that result from stimulation of enterocytes by cholera toxin cause secretion of salt and water with concomitant inhibition of absorption. Two other toxins are also encoded within the virulence cassette that encodes cholera toxin. These toxins, zona occludens toxin (zot) and accessory cholera toxin (ace), are consistently found in illness-causing strains of O1 and O139 but not usually in V. cholerae organisms that are less virulent.
Epidemiology
Since 1960, V. cholerae O1, biotype El Tor, has spread from India and Southeast Asia to Africa, the Middle East, southern Europe, and the southern, western, and central Pacific islands (Oceania). In late January of 1991, toxigenic V. cholerae O1, serotype Inaba, biotype El Tor, appeared in several coastal cities of Peru.907, 908 It rapidly spread to most countries in South and North America. In reported cases, travel from the United States to Latin America or Asia and ingestion of contaminated food transported from Latin America or Asia have been incriminated. V. cholerae O139 (Bengal) arose on the Indian subcontinent as a new cause of epidemic cholera in 1993.910, 911, 912, 913, 914, 915 It rapidly spread through Asia and continues to periodically reemerge as a cause of epidemic cholera. In the United States, an endemic focus of a unique strain of toxigenic V. cholerae O1 exists on the Gulf Coast of Louisiana and Texas.906, 916 This strain is different from the one associated with the epidemic in South America. Most cases of disease associated with the strain endemic to the U.S. Gulf Coast have resulted from the consumption of raw or undercooked shellfish. Humans are the only documented natural host, but free-living V. cholerae organisms can exist in the aquatic environment. The usual reported vehicles of transmission have included contaminated water or ice; contaminated food, particularly raw or undercooked shellfish; moist grains held at ambient temperature; and raw or partially dried fish. The usual mode of infection is ingestion of contaminated food or water. Boiling water or treating it with chlorine or iodine and adequate cooking of food kill the organism.907 Asymptomatic infection of family contacts is common but direct person-to-person transmission of disease has not been documented. Persons with low gastric acidity are at increased risk for cholera infection.
Clinical Manifestations
Cholera acquired during pregnancy, particularly in the third trimester, is associated with a high incidence of fetal death.917 Miscarriage can be attributed to a fetal acidosis and hypoxemia resulting from the marked metabolic and circulatory changes that this disease induces in the mother. It is not surprising that the likelihood of delivering a stillborn child is closely correlated with the severity of the maternal illness. The inability to culture V. cholerae from stillborn infants of infected mothers, together with the usual absence of bacteremia in cholera, suggests that transplacental fetal infection is not a cause of intrauterine death.
Neonatal cholera is a rare disease. This generalization also applies to the new O139 strains, although mild918 and severe forms of illness have rarely been described in newborns.919 Among 242 neonates admitted to a cholera research hospital in Dacca, Bangladesh, there were 25 infants ill with cholera.920 Even infants born to mothers with active diarrheal disease may escape infection, despite evidence that rice-water stools, almost certain to be ingested during the birth process, may contain as many as 109 organisms/mL.920 The reason for this apparently low attack rate among newborns is not certain; however, it probably can be attributed in large part to the protection conferred by breast-feeding.921 Human milk contains antibodies62 and receptor-like glycoprotein that inhibit adherence of V. cholerae 64 and gangliosides that bind cholera toxin.65 The role of transplacentally acquired vibriocidal maternal antibodies has not been determined.922 Because V. cholerae causes neither bacteremia nor intestinal invasion, protection against illness is more likely to be a function of mucosal rather than serum antibodies.923, 924 Additional factors that may reduce the incidence of neonatal cholera include the large inoculum required for infection925 and the limited exposure of the newborn to the contaminated food and water.229
Diagnosis
Clinicians should request that appropriate cultures be performed for stool specimens from persons suspected of having cholera. The specimen is plated on thiosulfate citrate bile salts sucrose agar directly or after enrichment in alkaline peptone water. Isolates of V. cholerae should be confirmed at a state health department and then sent to the CDC for testing for production of cholera toxin. A fourfold rise in vibriocidal antibody titers between acute and convalescent serum samples or a fourfold decline in titers between early and late (>2 months) convalescent serum specimens can confirm the diagnosis. Probes have been developed to test for cholera toxin.926, 927
Therapy and Prevention
The most important modality of therapy is administration of oral or parenteral rehydration therapy to correct dehydration and electrolyte imbalance and maintain hydration.907 Antimicrobial therapy can eradicate vibrios, reduce the duration of diarrhea, and reduce requirements for fluid replacement. One cholera vaccine, which is administered parenterally, is licensed in the United States but is of very limited value. Several experimental oral vaccines are being tested.928, 929, 930
YERSINIA ENTEROCOLITICA
Nature of the Organism, Epidemiology, and Pathogenesis
Y. enterocolitica is a major cause of enteritis in much of the industrialized world.931, 932 Enteritis due to this organism primarily occurs in infants and young children, and infections in the United States are reported to be more common in the North than in the South.933, 934, 935, 936, 937, 938 Animals, especially swine, have been shown to serve as the reservoir for Y. enterocolitica. A history of recent exposure to chitterlings (i.e., pig intestine) is common. Transmission has also occurred after ingestion of contaminated milk and infusion of contaminated blood products.939, 940
Virulence of Y. enterocolitica is related primarily to a virulence plasmid, which is closely related to the virulence plasmids of Yersinia pseudotuberculosis and Yersinia pestis. 941, 942 An ST enterotoxin, which is closely related to the ST of ETEC,943 may also be important.
Clinical Manifestations
Infection with Y. enterocolitica is recognized as one of the causes of bacterial gastroenteritis in young children, but knowledge of neonatal infection with this organism is fragmentary. Even in large series, isolation of Yersinia from newborns is rare.931, 932, 944
The youngest infants whose clinical course has been described in detail were 11 days to several months old at the onset of their illness.932, 944, 945, 946, 947, 948, 949, 950, 951, 952 There were no features of the gastroenteritis to distinguish it from that caused by other invasive enteric pathogens such as Shigella or Salmonella. Infants presented with watery diarrhea or with stools containing mucus with streaks of blood. Sepsis was common in these infants particularly in the first 3 months of life when 28% of enteritis was complicated by sepsis.948, 949, 953, 954 Fever is not a consistent finding in children with bacteremia, and meningitis is rare. In older children, fever and right lower quadrant pain mimicking appendicitis are often found.940
Diagnosis
Y. enterocolitica can be recovered from throat swabs, mesenteric lymph nodes, peritoneal fluid, blood, and stool. Because laboratory identification of organisms from stool requires special techniques, laboratory personnel should be notified when Yersinia is suspected. Because avirulent environmental isolates occur, biotyping and serotyping are useful in assessing the clinical relevance of isolates. PCR has been used to detect pathogenic strains.955, 956
Therapy
The effect of antimicrobial therapy on the outcome of gastrointestinal infection is uncertain. It has been recommended that antibiotics be reserved for sepsis or prolonged and severe gastroenteritis931; however, there are no prospective studies comparing the efficacy of various antimicrobial agents with each other or with supportive therapy alone. Most strains of Y. enterocolitica are susceptible to trimethoprim-sulfamethoxazole, the aminoglycosides, piperacillin, imipenem, third-generation cephalosporins, amoxicillin-clavulanate potassium, and chloramphenicol, and resistant to amoxicillin, ampicillin, carbenicillin, ticarcillin, and macrolides.957, 958, 959 Therapy in individual cases should be guided by in vitro susceptibility testing, although cefotaxime has been successfully used in bacteremic infants.954
AEROMONAS HYDROPHILA
Nature of the Organism, Epidemiology, and Pathogenesis
Aeromonas hydrophila is widely distributed in animals and the environment. Although wound infection, pneumonia, and sepsis (especially in immunocompromised hosts) represent typical Aeromonas infections, gastroenteritis increasingly is being recognized. The organism is a gram-negative, oxidase-positive, facultatively anaerobic bacillus belonging to the family Vibrionaceae. Like other members of this family, it produces an enterotoxin960 that causes fluid secretion in rabbit ileal loops.961 Some strains cause fluid accumulation in the suckling mouse model,962 whereas other strains are invasive963 or cytotoxic.964 The enterotoxin is not immunologically related to cholera toxin or the heat LT of E. coli. 965
Although volunteer studies and studies with monkeys have failed to provide supportive evidence for enteropathogenicity,966, 967 there is good reason to believe that A. hydrophila does cause diarrhea in children. The earliest description of Aeromonas causing diarrhea was an outbreak that occurred in a neonatal unit.968 Although several studies have failed to show an association with diarrhea,969, 970, 971, 972, 973, 974 most studies have found more Aeromonas isolates among children with gastroenteritis than among controls.974, 975, 976 Part of the controversy may be caused by strain differences; some strains possess virulence traits related to production of gastroenteritis, whereas others do not.970, 977
The diarrhea described in children is a disease of summer, primarily affecting children in the first 2 years of life. In one study, 7 (13%) of 55 cases of Aeromonas detected during a 20-month period occurred in infants younger than 1 month.
Clinical Manifestations
Typically, watery diarrhea with no fever has been described; although there are descriptions of watery diarrhea with fever.978 However, in 22%, a dysentery-like illness occurred. Dysentery-like illness has been described in the neonate.979 In one third of children, diarrhea has been reported to last for more than 2 weeks.970 There may be species-related differences in clinical features of Aeromonas-associated gastroenteritis in children.980 Organisms that were formerly classified as A. hydrophila are now sometimes labeled as Aeromonas sobria or Aeromonas caviae. 981, 982 Fever and abdominal pain appear to be particularly common with A. sobria. One series of A. hydrophila isolates from newborns in Dallas showed more blood cultures than stool cultures positive for Aeromonas. 983
Diagnosis and Therapy
Diagnosis of enteric infection associated with Aeromonas often is not made because this organism is not routinely sought in stool cultures. When the organism is suspected, the laboratory should be notified so that oxidase testing can be performed. The organism is usually susceptible to aztreonam, imipenem, meropenem, third-generation cephalosporins, trimethoprim-sulfamethoxazole, and chloramphenicol.984, 985, 986
PLESIOMONAS SHIGELLOIDES
Plesiomonas shigelloides is a gram-negative, facultative anaerobic bacillus that, like Aeromonas, is a member of the Vibrionaceae family. It is widely disseminated in the environment; outbreaks of disease are usually related to ingestion of contaminated water or seafood.987 Although it has been associated with outbreaks of diarrheal disease988 and has been found more commonly in ill than well controls, the role of P. shigelloides in diarrheal disease has remained controversial.989 If it is a true enteropathogen, the mechanism by which it causes disease is unclear.990, 991 The role of this organism in neonatal diarrhea has not been extensively investigated. Infections of neonates have been reported,992, 993, 994, 995 but most cases of enteric disease currently reported in the United States are in adults.987 Typical illness consists of watery diarrhea and cramps; sometimes, fever, bloody stools, and emesis occur and last for 3 to 42 days.
Diagnosis is not usually made by clinical microbiology laboratory testing because, as with Aeromonas, coliforms can be confused with P. shigelloides unless an oxidase test is performed.996 The true frequency of infection is unknown. The organism has antibiotic susceptibilities similar to those of Aeromonas. 997, 998
OTHER BACTERIAL AGENTS AND FUNGI
Proving that an organism causes diarrhea is difficult, particularly when it may be present in large numbers in stools of healthy persons. Bacteria that have been associated with acute gastroenteritis may be considered causative when the following criteria are met:
-
1.
A single specific strain of the organism should be found as the predominant organism in most affected infants by different investigators in outbreaks of enteric disease in different communities.
-
2.
This strain should be isolated in a significantly lower percentage and in smaller numbers from stool specimens of healthy infants.
-
3.
Available methods must be used to exclude other recognized enteropathogens, including viruses and parasites, enterotoxigenic agents, and fastidious organisms such as Campylobacter.
-
4.
Demonstration of effective specific antimicrobial therapy and specific antibody responses and, ultimately, production of experimental disease in volunteers are helpful in establishing the identity of a microorganism as a pathogen.
Optimally, the putative pathogen should have virulence traits that can be demonstrated in model systems. Most bacteria that have been suggested as occasional causes of gastroenteritis in neonates fail to fulfill one or more of these criteria. Their role in the cause of diarrheal disease is questionable. This is particularly true of microorganisms described in early reports in which the possibility of infection with more recently recognized agents could not be excluded. Much of the clinical, bacteriologic, and epidemiologic data collected earlier linking unusual enteropathogens to infantile diarrhea must be reevaluated in light of current knowledge and methodology.
Several reports of acute gastroenteritis believed to have been caused by Klebsiella suggest that, rather than playing an etiologic role, these organisms had probably proliferated within an already inflamed bowel.999, 1000, 1001 The recovery of Klebsiella-Enterobacter in pure culture from diarrheal stools has led several investigators to suggest that these bacteria may occasionally play a causative role in infantile gastroenteritis and enterocolitis.1002, 1003, 1004, 1005, 1006, 1007 Ingestion of infant formula contaminated with Enterobacter sakazakii has been associated with development of bloody diarrhea and sepsis.1008 However, Klebsiella species also may be isolated in pure culture from stools of newborns with no enteric symptoms.1009, 1010, 1011 In one study, certain capsular types of Klebsiella were more often isolated from infants with diarrheal disease than from normal infants.1002 Later work has shown that Klebsiella pneumoniae, Enterobacter cloacae, and Citrobacter species are capable of producing enterotoxins.139, 166, 167, 175, 193, 1008, 1012, 1013 Reports of isolation of Citrobacter species, such as those of Klebsiella species, describe associations with enteric illnesses in up to 7% of cases.1014, 1015, 1016 There is inadequate evidence to define the roles of Klebsiella, Enterobacter, and Citrobacter species as etiologic agents of enteric illnesses.
Listeria monocytogenes, one of the classic causes of neonatal sepsis and meningitis (see Chapter 14), has been linked to outbreaks of febrile diarrheal disease in immunocompetent adults and children.1017, 1018, 1019, 1020, 1021 Seventy-two percent of ill individuals have had fever.1022 Outbreaks have been related to ingestion of contaminated foods. Listeria has rarely been described as a cause of neonatal gastroenteritis.1023, 1024, 1025, 1026
Infection with enterotoxin-producing Bacteroides fragilis has been associated with mild watery diarrhea.1027 These infections have a peak incidence in 2- to 3-year-old infants.1028 These toxin-producing organisms cannot be detected in routine hospital laboratories.
A variety of organisms has been isolated from infant stools during episodes of diarrhea. Most of these reports have failed to associate illness with specific organisms in a way that has stood the test of time. For example, P. aeruginosa 1029, 1030, 1031, 1032, 1033, 1034 and Proteus 1017, 1035, 1036, 1037, 1038, 1039, 1040, 1041 have been associated with diarrhea, but there are few convincing data suggesting that either is a true enteropathogen. These organisms generally are recovered as frequently from healthy infants as from infants with diarrheal disease, suggesting that their presence in stool cultures is significant.273, 1042, 1043, 1044, 1045, 1046 An association between Providencia and neonatal enteritis has been substantiated largely by anecdotal reports of nursery outbreaks.215, 271, 1016, 1047 These bacteria are rarely isolated from infants with sporadic or community-acquired diarrheal disease.1042, 1043, 1044, 1048, 1049, 1050
Candida albicans usually is acquired during passage through the birth canal and is considered a normal, although minor, component of the fecal flora of the neonate (see Chapter 33).1051 Intestinal overgrowth of these organisms frequently accompanies infantile gastroenteritis,211, 233, 1051, 1054 particularly after antimicrobial therapy.233, 247, 1052, 1053, 1054, 1055 The upper small gut may become colonized with Candida in malnourished children with diarrhea1056; whether the presence of the organism is cause or effect is unclear. Stool cultures obtained from infants with diarrheal disease are therefore inconclusive, and although Candida enteritis has been reported in adults,1057 the importance of this organism as a primary cause of neonatal gastroenteritis has been difficult to prove. Clinical descriptions of nursery epidemics of candidal enteritis are poorly documented, generally preceding the recognition of EPEC and rotaviruses as a cause of neonatal diarrhea. Even well studied cases of intestinal involvement add little in the way of substantive proof because secondary invasion of Candida has been shown to be a complication of coliform enteritis.211, 233, 247
Although diarrhea has sometimes been described as a finding in neonatal disseminated candidiasis, more typically, gastrointestinal tract involvement with disseminated Candida is associated with abdominal distention and bloody stools mimicking necrotizing enterocolitis.247, 1056, 1057, 1058, 1059, 1060, 1061 Typically, affected infants are premature and have courses complicated by antibiotic administration, intravascular catheter use, and surgical procedures during the first several weeks of life. A trial of oral anticandidal therapy may be helpful in neonates suffering from diarrhea in the presence of oral or cutaneous candidiasis. If the therapy is appropriate, a response should be forthcoming within 2 to 5 days.
Diarrhea sometimes occurs as a manifestation of systemic infection. Patients with staphylococcal toxic shock syndrome, for example, often have diarrhea. Loose stools sometimes occur in sepsis, but it is unclear whether the diarrhea is a cause or an effect. The organisms isolated from blood cultures in a group of Bangladeshi infants and children with diarrhea included Staphylococcus aureus, Haemophilus influenzae, Streptococcus pneumoniae, P. aeruginosa, and various gram-negative enteric bacilli.1062 It is unknown whether the bacteriology of sepsis associated with diarrhea is similar in the well-nourished infants seen in industrialized countries.
PARASITES
Acute diarrhea associated with intestinal parasites is infrequent during the neonatal period. In areas with high endemicity, infection of the newborn is likely to be associated with inadequate maternal and delivery care, insufficient environmental sanitation, and poor personal hygiene standards. The occurrence of symptomatic intestinal parasitic infection during the first month of life requires acquisition of the parasite during the first days or weeks; the incubation period for E. histolytica and G. lamblia is 1 to 4 weeks, and for Cryptosporidium parvum, it is 7 to 14 days. The newborn can be infected during delivery by contact with maternal feces,1063 in the hospital through contact with the mother or personnel, or in the household through contact with infected individuals in close contact with the child. Contaminated water can be an important source of infection for G. lamblia and C. parvum.
Entamoeba histolytica
Organisms formerly identified as E. histolytica have been reclassified into two species that are morphologically identical but genetically distinct: E. histolytica and E. dispar. The former can cause acute nonbloody and bloody diarrhea, necrotizing enterocolitis, ameboma, and liver abscess, and the latter is a noninvasive parasite that does not cause disease. Early acquisition of disease tends to be more severe in young infants; rarely, amebic liver abscess and rapidly fatal colitis have been reported in infants.1064, 1065, 1066, 1067, 1068, 1069, 1070 For example, a 19-day-old child from India who presented with 10 to 12 episodes of watery and mucous diarrhea, lethargy, jaundice, and mildly elevated liver enzymes has been described; the child recovered completely after 10 days of intravenous omidazole.1064 However, asymptomatic colonization of neonates with various species of ameba is common in areas of high endemicity.1071
Diagnosis can be established by stool examination for cysts and trophozoites and by serologic studies.1072 Through the use of PCR, isoenzyme analysis, and antigen detection assays, E. histolytica and E. dispar can be differentiated.1073, 1074 Serum antibody assays may be helpful in establishing the diagnosis of amebic dysentery and extraintestinal amebiasis with liver involvement. The efficacy of treatment with metronidazole for colitis or liver abscess has not been established for the newborn period, although this therapy has been used with success.1065 Patients with colitis or liver abscess caused by E. histolytica are treated also with iodoquinol, as are asymptomatic carriers.
Giardia lamblia
G. lamblia is a binucleate, flagellated protozoan parasite with trophozoite and cyst stages. It is spread by the fecal-oral route through ingestion of cysts. Child-care center outbreaks reflecting person-to-person spread have demonstrated high infectivity.1075, 1076, 1077, 1078 Foodborne transmission and waterborne transmission also occur. Infection is often asymptomatic or mildly symptomatic; cases of severe symptomatic infection during the immediate newborn period have not been reported. Symptoms in giardiasis are related to the age of the patient, with diarrhea, vomiting, anorexia, and failure to thrive typical in the youngest children. Seroprevalence studies have demonstrated evidence of past or current infection in 40% of Peruvian children by the age of 6 months.1081 In a study of lactating Bangladeshi mothers and their infants, 82% of women and 42% of infants excreted Giardia once during the study; in some infants, this occurred before they were 3 months old.1082 Of these infected infants, 86% had diarrhea, suggesting that the early exposure to the parasite resulted in disease. In a prospective study of diarrhea conducted in Mexico, infants frequently were infected with Giardia from birth to 2 months, with a crude incidence rate of first Giardia infection of 1.4 infections per child-year in this age group.6 The symptom status of these children was not reported but this study strongly suggests that G. lamblia may be more common than currently recognized among newborns living in developing areas.
The diagnosis of giardiasis can be made on the basis of demonstration of antigen by EIA or by microscopy of feces, duodenal fluid or, less frequently, duodenal biopsy.1079, 1080 Breast-feeding is believed to protect against symptomatic giardiasis.6, 69, 1083 This protection may be mediated by cellular and humoral immunity67, 1084, 1085 and nonspecifically by the antigiardial effects of unsaturated fatty acids.1086 Giardia infections causing severe diarrhea may respond to metronidazole or furazolidone.1080
Cryptosporidium
C. parvum is a coccidian protozoon related to Toxoplasma gondii, Isospora belli, and Plasmodium species.1087, 1088 The life cycle involves ingestion of thick-walled oocysts; release of sporozoites, which penetrate intestinal epithelium; and development of merozoites. There is asexual and sexual reproduction, with the latter resulting in formation of new oocysts that can be passed in stools.
Cryptosporidium species are ubiquitous. Infection often occurs in persons traveling to endemic areas.1089 Because Cryptosporidium infects a wide variety of animal species, there is often a history of animal contact among infected individuals.1090 Person-to-person spread, particularly in household contacts1091, 1092, 1093, 1094 and daycare centers,1095, 1096 is well documented and suggests that the organism is highly infectious. Waterborne outbreaks of cryptosporidiosis occur and can be of massive proportions.1097
The clinical manifestations of illness in immunocompetent persons resemble those of Giardia infection but are somewhat shorter in duration1098; asymptomatic carriage is rare. Symptoms and signs include watery diarrhea, abdominal pain, myalgia, fever, and weight loss.1089, 1090, 1095, 1096, 1098, 1099 Infection in the first month of life has been described.1100, 1101 Because symptoms resolve before excretion of oocysts ceases, a newborn whose mother has been ill with cryptosporidiosis in the month before delivery might be at risk even if the mother is asymptomatic at the time of the child’s birth.1102 With the increasing frequency of human immunodeficiency virus infection, it is likely that women with symptomatic cryptosporidiosis occasionally will deliver an infant who will become infected. Infants infected early in life may develop chronic diarrhea and malnutrition.1103
The diagnosis of cryptosporidiosis is most typically made by examination of fecal smears using the Giemsa stain, Ziehl-Neelsen stain, auramine-rhodamine stain, Sheather’s sugar flotation, an immunofluorescence procedure, a modified concentration-sugar flotation method, or an EIA.1104, 1105 Nitazoxanide is effective therapy of immunocompetent adults and children ill with cryptosporidiosis.1106 Because illness is usually self-limited in the normal host, attention to fluid, electrolyte, and nutritional status usually suffices. Enteric isolation of hospitalized infants with this illness is appropriate because of the high infectivity. Several studies suggest that the risk of infection early in life may be decreased by breast-feeding.1101, 1107
VIRUSES
Enteric Viruses
Viruses that infect the intestinal mucosa and cause primarily gastroenteritis are referred to as enteric viruses; they should not be confused with enteroviruses, members of Picornaviridae family that are associated primarily with systemic illnesses. Enteric viruses include rotaviruses, enteric adenoviruses, human caliciviruses, and astroviruses. Other viruses such as coronaviruses, Breda viruses, pestiviruses, parvoviruses, toroviruses, and picobirnaviruses have been sporadically associated with acute diarrhea but are currently considered of uncertain relevance. Extensive reviews on the role of enteric viruses in childhood diarrhea can be found elsewhere.1108, 1109, 1110, 1111
All four enteric viruses could conceivably infect the newborn, but the extent of exposure and clinical manifestations are largely unknown for astrovirus, enteric adenovirus, and human caliciviruses. Rotavirus is the most extensively studied enteric virus. Neonatal rotavirus infections have similar virologic and clinical characteristics to infection in older children, although some differences exist.
Rotavirus
Rotavirus is a 75-nm, nonenveloped virus composed of three concentric protein shells: a segmented genome (11 segments), an RNA-dependent polymerase, and enzymes required for messenger RNA synthesis are located within the inner core. Each segment codes for at least one viral protein (VP). The VP can be part of the structure of the virus, or it may be a nonstructural protein (NSP) required for replication, viral assembly, budding, determination of host range, or viral pathogenesis.1110
Six distinct rotavirus groups (A through F) have been identified serologically based on common group antigens,1112, 1113 of which three (A, B, and C) have been identified in humans.1108 Because group A rotaviruses represent more than 95% of isolated strains in humans worldwide, further discussion focuses on this group. Group A rotaviruses are subclassified into serotypes based on neutralization epitopes located on the outer capsid. Both rotavirus surface proteins, VP4 and VP7, can induce production of neutralizing antibodies.1114, 1115 At least 10 VP7 types (G serotypes: G1 to G6, G8 to G10, and G12) and nine VP4 types (P serotypes: P1A, P1B, P2A, P3, P3B, P4, P5, P8, and P12) have been detected among human rotaviruses.1116, 1117, 1118, 1119, 1120, 1121, 1122, 1123 By sequencing the VP4-coding gene, eight genomic P types (genotypes) have been identified that correspond to one or more of the described P antigenic types (genotype 8 to antigenic type P1A, 4 to P1B, 6 to P2A, 9 to P3, 13 to P3B, 10 to P4, 3 to P5, and 11 to P8).1110 Combining G antigenic with P antigenic and genetic typing, a specific rotavirus strain can be identified: P antigenic type (P genetic type), G type. As an example, the human neonatal M37 strain is described as P2A[6], G1.
Four combined GP types: P1A[8], G1; P1B[4], G2; P1A[8], G3; and P1A[8], G4 account for more than 95% of the organisms isolated from children, and of these, P1A[8], G1 represents the single most common type.1118, 1119, 1120, 1121, 1122, 1123 Isolation of less common types appears to be more frequent among neonates with nosocomial rotavirus infections.1123, 1124, 1125, 1126, 1127, 1128, 1129, 1130 Some of these strains seem to be associated with occurrence of asymptomatic infections, although the existence of naturally acquired asymptomatic strains is controversial. Strains P2A[6], G9; P2A[6], G4; P2A[6], G2; and P2A[6], G8 have been reported1128, 1129, 1130, 1131 from newborn nurseries, some of which seem to be endemic to the newborn units with high rates of asymptomatic infection,1129, 1130, 1131 and less commonly, outbreaks of symptomatic infection.1128 These findings suggest that specific conditions of the newborn environment (e.g., child, nursery, personnel) may increase the possibility of reassortments between human strains; such strains may persist in these settings possibly through constant transmission involving asymptomatic newborns, adults, and contaminated surfaces.
Pathogenesis
Rotavirus primarily infects mature enterocytes located in the mid and upper villous epithelium.1132, 1133, 1134, 1135, 1136 Lactase, which is present only on the brush border of the differentiated epithelial cells at these sites, may act as a combined receptor and uncoating enzyme for the virus, permitting transfer of the particles into the cell.1137 Perhaps for this reason, infection is limited to the mature columnar enterocytes; crypt cells and crypt-derived cuboidal cells, which lack a brush border, appear to be resistant to rotaviral infection.1137, 1138 This concept also may explain why rotavirus infection is less common in infants younger than 32 weeks’ gestational age than in more mature infants1139; between 26 and 34 weeks’ gestational age, lactase activity is approximately 30% of that found in term infants.1140
The upper small intestine is most commonly involved, although lesions may extend to the distal ileum and rarely to the colon.1141, 1142 Interaction between intestinal cell and rotavirus structural and nonstructural proteins occurs, resulting in death of infected villous enterocytes.1143 Once infected, the villous enterocyte is sloughed, resulting in an altered mucosal architecture that becomes stunted and flattened. The gross appearance of the bowel is usually normal; however, under the dissecting microscope, scattered focal lesions of the mucosal surface are apparent in most cases. Light microscopy also shows patchy changes in villous morphology, compatible with a process of infection, inflammation, and accelerated mucosal renewal. The villi take on a shortened and blunt appearance as tall columnar cells are shed and replaced by less mature cuboidal enterocytes.1133, 1135, 1144 Ischemia may also play a role in the loss and stunting of villi1145 and activation of the enteric nervous system; active secretion of fluid and electrolytes may be another pathogenic mechanism.1146 During the recovery phase, the enteroblastic cells mature and reconstruct the villous structure. Because of the loss of mature enterocytes on the tips of the villi, the surface area of the intestine is reduced. Diarrhea that occurs may be a result of this decrease in surface area, disruption in epithelial integrity, transient disaccharidase deficiency, or altered countercurrent mechanisms and net secretion of water and electrolytes.1133, 1140, 1142, 1146, 1147, 1148 NSP4 has been found to induce age-dependent diarrhea in CD1 mice by triggering calcium-dependent chloride and water secretion.1149 The potential role of this “viral enterotoxin” in human disease is not yet clear.1150, 1151
Infection and Immunity
Infants with asymptomatic rotavirus infections in the nursery are less likely than uninfected nursery mates to experience severe rotavirus infection later in life1152, 1153; this finding suggested protective immunity and supported vaccine development. Most studies have indicated that serum and intestinal antirotavirus antibody levels are correlated with protection against infection1153, 1154, 1155, 1156, 1157, 1158, 1159, 1160, 1161 although this correlation has not been universal.1162, 1163 Breast-feeding protects against rotavirus disease during the first year of life,57 probably including newborns.1146 The high prevalence of antirotaviral antibodies in colostrum and human milk has been demonstrated by numerous investigators in widely diverse geographic areas.8 Maternal rotavirus infection or immunization is accompanied by the appearance of specific antibodies in milk, probably through stimulation of the enteromammary immune system.1164, 1165, 1166, 1167, 1168, 1169 Between 90% and 100% of women examined in London, Bangladesh, Guatemala, Costa Rica, and the United States had antirotaviral IgA antibodies in their milk for up to 2 years of lactation.8, 1164, 1165, 1166, 1167, 1168, 1169, 1170 Rotavirus-specific IgG antibodies have been found during the first few postpartum days in about one third of human milk samples assayed,1164, 1167 whereas IgM antibodies were detectable in about one half.1167
Glycoproteins in human milk have been shown to prevent rotavirus infection in vitro and in an animal model.1170 The concentration of one milk glycoprotein, lactadherin, was found to be significantly higher in human milk ingested by infants who developed asymptomatic rotavirus infection than in milk ingested by infants who developed symptomatic infection.45
Epidemiology
Rotaviruses probably infect neonates more commonly than previously recognized, although most infections seem to be asymptomatic or mildly symptomatic.1128, 1129, 1130, 1131, 1172, 1173, 1174, 1175, 1176, 1177, 1178, 1179, 1180, 1181, 1182, 1183, 1184, 1185, 1186, 1187 In a prospective study, the prevalence of rotavirus infection among neonatal intensive care unit patients was 18.4%. Rotavirus has a mean incubation period of 2 days, with a range of 1 to 3 days in children and in experimentally infected adults. Fecal excretion of virus often begins a day or so before illness and maximal excretion usually occurs during the third and fourth days, and generally diminishes by the end of the first week, although low concentrations of virus have been detected in neonates for up to 8 weeks.1140, 1186, 1187, 1188, 1189
Rotavirus infections are markedly seasonal (autumn and winter) in many areas of the world, although in some countries seasonality is less striking; the reason for this is unclear.1190, 1191, 1192, 1193, 1194, 1195 In nurseries in which persisting endemic infection has permitted long-term surveillance of large numbers of neonates, rotavirus excretion can follow the seasonal pattern of the community but can also show no seasonal fluctuation.1196, 1197, 1198 It is not clear how units in which infection remains endemic for months or years differ from those with a low incidence of rotavirus. Some nurseries are free of rotavirus infection1198, 1199, 1200 or minimally affected45, 1201 whereas others have rotavirus diarrheal disease throughout the year or in outbreaks that involve 10% to 40% of neonates.1128, 1139, 1179, 1202, 1203
Low birth weight does not seem to be an important factor in determining the attack rate among infants at risk but may be important in mortality.1204 Infants in premature or special-care nurseries, despite their prolonged stays and the increased handling necessary for their care, do not demonstrate a higher susceptibility to infection; data regarding shedding of the virus are inconsistent.45, 1200
After infection is introduced into a nursery, rotavirus probably will spread steadily and remain endemic until the nursery is closed to new admissions or nursing practices permit interruption of the cycle.1205 Exactly how the virus is introduced and transmitted is uncertain, although limited observations and experience with other types of enteric disease in maternity units suggest several possibilities. The early appearance of virus in stools of some neonates indicates that infection probably was acquired at delivery. Virus particles can be detected on the first45, 1186 or second1198 day of life in a significant number of infected infants. By day 3 or 4, most infected infants who will shed virus, with or without signs of illness, are doing so.1174, 1186, 1198 The large numbers of virus particles excreted1174, 1198 suggest a fairly large and early oral inoculum. It is unlikely that contamination from any source other than maternal feces could provide an inoculum large enough to cause infection by the second day.
Transfer of particles from infant to infant on the hands of nursing and medical staff is probably the most important means of viral spread. With 108 to 1011 viral particles usually present in 1 g of stool, the hands of personnel easily could become contaminated after infection is introduced into a nursery. There are numerous reports of nosocomial and daycare center rotavirus gastroenteritis outbreaks that attest to the ease with which this agent spreads through a hospital or institutional setting.1108 Admission of a symptomatic child usually is the initiating event, although transfer of a neonate with inapparent infection from one ward to another also has been incriminated. The most important factors influencing the incidence of rotavirus diarrhea in a nursery are the proximity to other newborns and the frequency of handling.1187 During a 4-month study, infants cared for by nursing staff and kept in communal nurseries experienced three epidemics of diarrhea with attack rates between 20% and 50%. During the same period, only 2% of infants rooming in with their mothers became ill, even though they had frequent contact with adult relatives and siblings.
There is no clear evidence of airborne or droplet infection originating in the upper respiratory tract or spread by aerosolization of diarrheal fluid while diapers are changed. Indirect evidence of airborne transmission includes the high infection rate in closed settings, the isolation of the virus from respiratory secretions,1206 and the experimental observation of transmission by aerosol droplets in mice.1207 However, the respiratory isolation achieved by placing an infant in a closed incubator is not fully protective.1187 No evidence indicates that transplacental or ascending intrauterine infection occurs. Transmission of virus through contaminated fomites, formula, or food is possible but has not been documented in newborns. Rotavirus particles have not been found in human milk or colostrum.1166, 1170
Clinical Manifestations
Exposure of a newborn to rotavirus can result in asymptomatic infection or cause mild or severe gastroenteritis.1129, 1130, 1173, 1179, 1196, 1197, 1201, 1208 Outbreaks with high attack rates as measured by rotavirus excretion have been described but the extent of symptomatic infection varies.1175, 1177, 1186, 1198, 1203 Severe rotavirus infection is seldom reported during the newborn period1203 but the extent of underreporting of severe disease, especially in the less developed areas of the world, has not been evaluated.
It has been hypothesized that asymptomatic infections during the newborn period are the result of naturally attenuated strains circulating in this environment. RNA electrophoretic patterns of rotaviruses found in certain nurseries have shown uniform patterns1180, 1182, 1184, 1208, and it has been suggested that these strains may be attenuated. The presence of unusual antigenic types such as the P2A[6] type within nurseries also suggests “less virulent strains.” At least 10 rotavirus strains were documented to co-circulate in a tertiary care center during a 2-month period1209 and in a different setting the same rotavirus strains by electropherotype produced asymptomatic infection in neonates and symptomatic infection in older infants.1183 Newborns within a nursery exposed to a given rotavirus strain can develop symptomatic or asymptomatic infection.1130, 1210, 1211 Because newborns routinely have frequent relatively loose stools, it is possible that mild diarrhea episodes caused by rotavirus are being wrongly labeled as asymptomatic episodes.
No clinical feature is pathognomonic of rotaviral gastroenteritis. Early signs of illness, such as lethargy, irritability, vomiting, and poor feeding, usually are followed in a few hours by the passage of watery yellow or green stools free of blood but sometimes containing mucus.1187, 1212, 1213, 1214 Diarrhea usually decreases by the second day of illness and is much improved by the third or fourth day. Occasionally, intestinal fluid loss and poor weight gain may continue for 1 or 2 weeks, particularly in low-birth-weight infants.1175 Although reducing substances frequently are present in early fecal samples1139, 175, 1176, 1187 this finding is not necessarily abnormal in neonates, particularly those who are breast-fed.1215 Nevertheless, infants with prolonged diarrhea should be investigated for monosaccharide or disaccharide malabsorption or intolerance to cow’s milk protein or both.1216 In a prospective study,1185 49% of newborns with gastrointestinal symptoms in a neonatal intensive care unit had rotavirus detected in their stools. Frequent stooling (present in 60%), bloody mucoid stool (42%), and watery stools (24%) were risk factors for a rotavirus infection. Bloody mucoid stools, intestinal dilatation, and abdominal distention were significantly more common in preterm infants, but severe outcomes such as necrotizing enterocolitis and death did not differ among infected term and preterm infants.
Longitudinal studies in newborn nurseries and investigations of outbreaks among neonates rarely describe a severe adverse outcome or death.1139, 1168, 1187 Because these infants are under constant observation, early detection of excessive fluid losses and the availability of immediate medical care are probably major factors in determining outcome. Rotavirus gastroenteritis causes almost 400,000 deaths of infants every year,1217 concentrated largely in the poorest regions of the world. It is likely that in places where hospital-based care is uncommon, rotavirus causes neonatal deaths secondary to dehydration.
Group A rotavirus has been associated with a wide array of diseases in infants and children; Reye syndromes, encephalitis-aseptic meningitis, sudden infant death syndrome, inflammatory bowel disease, and Kawasaki syndrome have been described but not systematically studied.1108 Case reports and small case series have associated neonatal rotavirus infection with necrotizing enterocolitis.1218, 1219 Rotavirus infection may play a role in a small proportion of cases of necrotizing enterocolitis, although it probably represents one of many potential triggering factors. A significant association between neonatal rotavirus infection and bradycardia-apnea episodes was detected in one prospective study.1220 The possible association between natural rotavirus infection and intussusception1221, 1222, 1223 gained support after the association was made between the human-simian reassortant vaccine and intussusception in infants older than 2 months (attributable risk ≈ 1:10,000).1224 Intussusception is extremely uncommon in the newborn; it is highly unlikely that rotavirus triggers this disease in neonates.
Diagnosis
There are many methods used for detection of rotavirus in stool specimens, including electron microscopy, immune electron microscopy, ELISA, latex agglutination, gel electrophoresis, culture of the virus, and reverse transcriptase– polymerase chain reaction. ELISA and latex agglutination currently are the most widely used diagnostic techniques for detection of rotavirus in clinical samples. Many commercial kits are available that differ in specificity and sensitivity.1225, 1226, 1227, 1228, 1229 In general, latex agglutination assays are more rapid than ELISAs but are less sensitive. The sensitivity and specificity of the commercially available ELISAs surpass 90%. Checking of the ELISA by another method such as gel electrophoresis or PCR amplification may be desirable if there is concern about false-positive results.
Fecal material for detection of rotavirus infection should be obtained during the acute phase of illness. Whole-stool samples are preferred, although suspensions of rectal swab specimens have been adequate for detection of rotavirus by ELISA.1230, 1231 Rotavirus are relatively resistant to environmental temperatures, even tropical temperatures,1232 although 4° C is desirable for short-term storage and −70° C for prolonged storage.1108 Excretion of viral particles may precede signs of illness by several days1190; maximal excretion by older infants and children usually occurs 3 to 4 days after onset of symptoms.1233 Neonates can shed virus for 1 to 2 weeks after onset of symptoms.
Therapy and Prevention
The primary goal of therapy is restoration and maintenance of fluid and electrolyte balance. Despite the documented defect in carbohydrate digestion with rotavirus diarrhea, rehydration often can be accomplished with glucose-electrolyte or sucrose-electrolyte solutions given orally.199, 1234, 1235, 1236 Intravenous fluids may be needed in neonates who are severely dehydrated, who have ileus, or who refuse to feed. Persistent or recurrent diarrhea after introduction of milk-based formulas or human milk warrants investigation for secondary carbohydrate or milk protein intolerance.1139, 1217 Disaccharidase levels and xylose absorption return to normal within a few days1144 to weeks after infection.1133
Intractable diarrhea related to severe morphologic and enzymatic changes of the bowel mucosa is possible although rare in the newborn; it may require an elemental diet or parenteral nutrition. Efficacy of anti-rotavirus antibodies (e.g., hyperimmune colostrum, antibody-supplemented formula, human serum immunoglobulin) and of probiotics has been postulated,1238, 1239, 1240, 1241 although not convincingly shown1242; the widespread clinical use of these measures seems remote. One study suggests that use of lactobacillus during the diarrheal episode may decrease the duration of rotavirus-associated hospital stays, especially when used early in the course of the disease, although more studies are needed before recommending widespread use.1241
Hand hygiene before and after contact with each infant remains the single most important means of preventing the spread of infection. Because rotavirus is often excreted several days before illness is recognized, isolation of an infant with diarrhea may be too late to prevent cross-infection unless all nursing personnel and medical staff have adhered to this fundamental precaution. Infants who develop gastroenteritis should be moved out of the nursery area if adequate facilities are available and the infant’s condition permits transfer. The use of an incubator is of value in reducing transmission of disease only by serving as a reminder that proper hand-hygiene and glove techniques are required, but is of little value as a physical barrier to the spread of virus.1187 Encouraging rooming-in of infants with their mothers has been shown to be helpful in preventing or containing nursery epidemics.1243 Temporary closure of the nursery may be required for clinically significant outbreaks that cannot be controlled with other measures.1128
Vaccines
Development of rotavirus vaccines began in the early 1980s. Candidate vaccines included bovine and rhesus monkey attenuated strains, human attenuated strains, and bovine-human and rhesus-human reassortant strains.1109 In August 1998, the first licensed rotavirus vaccine, Rotashield, an oral formulation of a simian-human quadrivalent reassortant vaccine, was recommended for use in children when they were 2, 4, and 6 months old. After approximately 500,000 children were vaccinated with more than 1 million doses, a significantly increased risk of intussusception was observed among vaccinated children, with an overall odds ratio of 1.8.1244 Use of this vaccine was terminated. Two new vaccine candidates are undergoing phase III clinical trials: a “pentavalent” bovine-human reassortant vaccine including G types G1-G4 and P type P1A[8] and a monovalent human attenuated P1A[8]G1 vaccine. The epidemiology of rotavirus infection will change significantly if one or both candidates become widely available in the future. The impact on neonatal infection will depend on the effect of herd immunity in decreasing circulation of rotavirus strains.
DIFFERENTIAL DIAGNOSIS
Stools from breast-fed neonates are typically watery and yellow, green, or brown. The frequency of stooling can vary from one every other day to eight evacuations per day. In an active, healthy infant who is feeding well, has no vomiting, and has a soft abdomen, these varied patterns of stooling are not a cause for concern. Physicians need to consider the child’s previous frequency and consistency of stools and establish a diagnosis of acute diarrhea on an individual basis. Close follow-up of weight increase in infants with nonformed stools can help confirm the clinical impression. A normal weight gain should direct medical action away from stool exams or treatment.
Diarrhea during the neonatal period is a clinical manifestation of a wide variety of disorders (Table 20–6). The most common initiating factor is a primary infection of the gastrointestinal tract that is mild to moderate in severity, self-limited, and responsive to supportive measures. Acute diarrhea can also be an initial manifestation of a systemic infection, including bacterial and viral neonatal sepsis. Infants with moderate to severe diarrhea require close monitoring until the etiologic diagnosis and the clinical evolution are clarified. There are noninfectious diseases leading to chronic intractable diarrhea that may result in severe nutritional disturbances or even death unless the specific underlying condition is identified and treated appropriately. The differential diagnosis of a diarrheal illness requires a careful clinical examination to determine whether the child has a localized or a systemic process. Lethargy, abnormalities in body temperature, hypothermia or hyperthermia, decreased feeding, abdominal distention, vomiting, pallor, respiratory distress, apnea, cyanosis, hemodynamic instability, hypotension, hepatomegaly or splenomegaly, coagulation or bleeding disorders, petechiae, and exanthemas should lead to an intense laboratory investigation directed at systemic viral or bacterial infection. If the process is deemed a localized intestinal infection, initial evaluation can be focused on differentiating an inflammatory-invasive pathogen from those that cause a noninflammatory process. For this, stool examination for fecal leukocytes, red blood cells, and lactoferrin can be a helpful indicator of the former.
Table 20-6.
Differential Diagnosis of Neonatal Diarrhea
| Diagnosis | Reference(s) |
|---|---|
| Anatomic Disorders | |
| Microvillous inclusion disease | 1245 |
| Hirschsprung’s disease | 1246 |
| Massive intestinal resection (short-bowel syndrome) | 1247 |
| Intestinal lymphangiectasis | 1248 |
| Metabolic and Enzymatic Disorders | |
| Congenital disaccharidase deficiency (lactase, sucrase-isomaltase deficiency) | 1249,1250 |
| Congenital glucose-galactose malabsorption | 1251,1252 |
| Secondary disaccharide, monosaccharide malabsorption | 337,528,1252,1253,1254,1255,1256,1257,1258 |
| After gastrointestinal surgery | |
| After infection | |
| With milk-soy protein sensitivity | |
| Cystic fibrosis | 1259 |
| Syndrome of pancreatic insufficiency and bone marrow dysfunction (Shwachman’s syndrome) | 1260 |
| Physiologic deficiency of pancreatic amylase | 1261 |
| Intestinal enterokinase deficiency | 1262 |
| Congenital bile acid deficiency syndrome | 1263 |
| Alpha/beta-lipoproteinemia | 1264 |
| Acrodermatitis enteropathica | 1265,1266 |
| Congenital chloride diarrhea | 1267,1268 |
| Primary hypomagnesemia | 1269 |
| Congenital adrenal hyperplasia | 1270 |
| Intestinal hormone hypersecretion | 1271,1272 |
| Non–beta islet cell hyperplasia (Wolman’s disease) | 1273 |
| Transcobalamin II deficiency | 1274 |
| Congenital iron storage | 1275 |
| Hartnup’s disease | 1276 |
| Congenital Na+ diarrhea | 1277 |
| Inflammatory Disorders | |
| Cow’s milk protein intolerance | 1278 |
| Soy protein intolerance | 1279,1280 |
| Regional enteritis | 1281 |
| Ulcerative colitis | 1282,1283 |
| Primary Immunodeficiency Disorders | |
| Wiskott-Aldrich syndrome | 1284 |
| Thymic dysplasia | 1284 |
| Acquired immunodeficiency syndrome | 1285 |
| Miscellaneous | |
| Irritable colon of childhood (chronic nonspecific diarrhea) | 1286 |
| Phototherapy for hyperbilirubinemia | 1287 |
| Familial dysautonomia (Riley-Day syndrome) | 1288 |
| Familial enteropathy | 1289,1290 |
| High sulfates in water | 1291 |
| Phenolphthalein poisoning/child abuse | 1292 |
Inflammatory diarrhea can be caused by Shigella, Salmonella, Campylobacter, V. parahaemolyticus, Y. enterocolitica, EIEC, EAEC, C. difficile, necrotizing enterocolitis, antibiotic-associated colitis, and allergic colitis (i.e., milk or soy intolerance). Noninflammatory causes of diarrhea include ETEC, EPEC, rotaviruses, enteric adenoviruses, calicivirus, astrovirus, G. lamblia and Cryptosporidium. Although supportive fluid therapy is mandatory for all types of diarrhea, the brief examination for fecal leukocytes and red blood cells can direct the diagnostic and therapeutic approach. Pathogens such as Shigella, Salmonella, and EHEC can cause watery or bloody diarrhea, depending on the specific host-pathogen interaction and the pathogenic mechanisms involved. Some of the noninfectious diseases responsible for neonatal diarrhea are listed in Table 20–6.337, 528, 1245, 1246, 1247, 1248, 1249, 1250, 1251, 1252, 1253, 1254, 1255, 1256, 1257, 1258, 1259, 1260, 1261, 1262, 1263, 1264, 1265, 1266, 1267, 1268, 1269, 1270, 1271, 1272, 1273, 1274, 1275, 1276, 1277, 1278, 1279, 1280, 1281, 1282, 1283, 1284, 1285, 1286, 1287, 1288, 1289, 1290, 1291, 1292 The evaluation and management of persistent infantile diarrhea has been reviewed.1293
REFERENCES
- 1.Prashar UD, Hummelman EG, Bresee JS. Global illness and deaths caused by rotavirus disease in children. Emerg Infect Dis. 2003;9:565. doi: 10.3201/eid0905.020562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Guerrant RL. Lessons from diarrheal diseases: demography to molecular pharmacology. J Infect Dis. 1994;169:1206. doi: 10.1093/infdis/169.6.1206. [DOI] [PubMed] [Google Scholar]
- 3.Ho MS, Glass RI, Pinsky PF. Diarrheal deaths in American children: are they preventable? JAMA. 1988;260:3281. [PubMed] [Google Scholar]
- 4.Cohen ML. The epidemiology of diarrheal disease in the United States. Infect Dis Clin North Am. 1988;2:557. [PubMed] [Google Scholar]
- 5.Mata LJ, Urrutia JJ. Intestinal colonization of breastfed children in a rural area of low socioeconomic level. Ann N Y Acad Sci. 1971;176:93. [Google Scholar]
- 6.Morrow AL, Reves RR, West MS. Protection against infection with Giardia lamblia by breast-feeding in a cohort of Mexican infants. J Pediatr. 1992;121:363. doi: 10.1016/s0022-3476(05)81787-1. [DOI] [PubMed] [Google Scholar]
- 7.Velásquez FR, Matson DO, Lourdes Guerrero M. Serum antibody as a marker of protection against natural rotavirus infection and disease. J Infect Dis. 2000;182:1602. doi: 10.1086/317619. [DOI] [PubMed] [Google Scholar]
- 8.Morrow AL, Pickering LK. Human milk protection against diarrheal disease. Semin Pediatr Infect Dis. 1994;5:236. [Google Scholar]
- 9.Prado V, O’Ryan M. Acute Gastroenteritis in Latin America. Infect Dis Clin North Am. 1994;8:77. [PubMed] [Google Scholar]
- 10.Guerrant RL, Hughes JM, Lima NL. Diarrhea in developed and developing countries: magnitude, special settings, and etiologies. Rev Infect Dis. 1990;12:S41. doi: 10.1093/clinids/12.Supplement_1.S41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.World Health Organization . Implementation of the Global Strategy for Health for All by the Year 2000, Second Evaluation, and Eighth Report of the World Health Situation. Forty-Fifth World Health Assembly. World Health Organization; Geneva: 1992. p. A45/3. [Google Scholar]
- 12.Parashar UD, Kilgore PE, Holman RC. Diarrheal mortality in US infants. Arch Pediatr Adolesc Med. 1998;152:47. doi: 10.1001/archpedi.152.1.47. [DOI] [PubMed] [Google Scholar]
- 13.Mata LJ, Urrutia JJ. Intestinal colonization of breastfed children in a rural area of low socioeconomic level. Ann N Y Acad Sci. 1971;176:93. [Google Scholar]
- 14.Guerrant LR, Steiner TS, Lima AAM, Bobak DA. How intestinal bacteria cause disease. J Infect Dis. 1999;179:S331. doi: 10.1086/513845. [DOI] [PubMed] [Google Scholar]
- 15.Pickering LK, Cleary TG. Approach to patients with gastrointestinal tract infections and food poisoning. In: Feigin RD, Cherry JC, editors. Textbook of Pediatric Infectious Diseases. 4th ed. WB Saunders; Philadelphia: 1997. p. 567. [Google Scholar]
- 16.Bettelheim KA, Lennox-King SMJ. The acquisition of Escherichia coli by newborn babies. Infection. 1976;4:174. doi: 10.1007/BF01638945. [DOI] [PubMed] [Google Scholar]
- 17.Gorden J, Small PLC. Acid resistance in enteric bacteria. Infect Immun. 1993;61:364. doi: 10.1128/iai.61.1.364-367.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Pickering LK. Biotherapeutic agents and disease in infants. Adv Exp Med Biol. 2001;501:365. doi: 10.1007/978-1-4615-1371-1_45. [DOI] [PubMed] [Google Scholar]
- 19.Giannella RA, Broitman SA, Zamcheck N. Influence of gastric acidity on bacterial and parasitic enteric infections: a perspective. Ann Intern Med. 1973;78:271. doi: 10.7326/0003-4819-78-2-271. [DOI] [PubMed] [Google Scholar]
- 20.Schrager J. The chemical composition and function of gastrointestinal mucus. Gut. 1970;11:450. doi: 10.1136/gut.11.5.450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Challacombe DN, Richardson JM, Anderson CM. Bacterial microflora of the upper gastrointestinal tract in infants without diarrhea. Arch Dis Child. 1974;49:264. doi: 10.1136/adc.49.4.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Furuta GT, Walker WA. Nonimmune defense mechanisms of the gastrointestinal tract. In: Blaser MJ, Smith PD, Ravdin JI, editors. Infections of the Gastrointestinal Tract. Raven Press; New York: 1995. pp. 89–97. [Google Scholar]
- 23.Avery G, Randolph JG, Weaver T. Gastric acidity in the first day of life. Pediatrics. 1966;37:1005. [PubMed] [Google Scholar]
- 24.Harries JT, Fraser AJ. The acidity of the gastric contents of premature babies during the first fourteen days of life. Biol Neonate. 1968;12:186. doi: 10.1159/000240105. [DOI] [PubMed] [Google Scholar]
- 25.Agunod M, Yamaguchi N, Lopez R. Correlative study of hydrochloric acid, pepsin, and intrinsic factor secretion in newborns and infants. Am J Dig Dis. 1969;14:400. doi: 10.1007/BF02239360. [DOI] [PubMed] [Google Scholar]
- 26.Cavel B. Gastric emptying in infants. Acta Paediatr Scand. 1971;60:371. [PubMed] [Google Scholar]
- 27.Blumenthal I, Ebel A, Pildes RS. Effect of posture on the pattern of stomach emptying in the newborn. Pediatrics. 1980;66:482. [PubMed] [Google Scholar]
- 28.Silverio J. Gastric emptying time in the newborn and nursling. Am J Med Sci. 1964;247:732. doi: 10.1097/00000441-196406000-00015. [DOI] [PubMed] [Google Scholar]
- 29.Cavell B. Gastric emptying in preterm infants. Acta Paediatr Scand. 1979;68:725. doi: 10.1111/j.1651-2227.1979.tb18446.x. [DOI] [PubMed] [Google Scholar]
- 30.Eckmann L, Kagnoff M, Fierer J. Intestinal epithelial cells as watchdogs for the natural immune system. Trends Microbiol. 1995;3:118. doi: 10.1016/s0966-842x(00)88894-0. [DOI] [PubMed] [Google Scholar]
- 31.Bernt K, Walker W. Human milk as a carrier of biochemical messages. Acta Paediatr Suppl. 1999;88:27. doi: 10.1111/j.1651-2227.1999.tb01298.x. [DOI] [PubMed] [Google Scholar]
- 32.Fallot ME, Boyd JL, Oski FA. Breast-feeding reduces incidence of hospital admissions for infection in infants. Pediatrics. 1980;65:1121. [PubMed] [Google Scholar]
- 33.Larsen SA, Homer DR. Relation of breast versus bottle feeding to hospitalization for gastroenteritis in a middle-class U.S. population. J Pediatr. 1978;92:417. doi: 10.1016/s0022-3476(78)80430-2. [DOI] [PubMed] [Google Scholar]
- 34.Cushing AH, Anderson L. Diarrhea in breast-fed and non–breast-fed infants. Pediatrics. 1982;70:921. [PubMed] [Google Scholar]
- 35.Guerrant RL, Kirchhoff LV, Shields DS. Prospective study of diarrheal illnesses in northeastern Brazil: patterns of disease, nutritional impact, etiologies and risk factors. J Infect Dis. 1983;148:986. doi: 10.1093/infdis/148.6.986. [DOI] [PubMed] [Google Scholar]
- 36.Myers MG, Fomon SJ, Koontz FP. Respiratory and gastrointestinal illnesses in breast- and formula-fed infants. Am J Dis Child. 1984;138:629. doi: 10.1001/archpedi.1984.02140450011003. [DOI] [PubMed] [Google Scholar]
- 37.Kovar MG, Serdula MK, Marks JS. Review of the epidemiologic evidence for an association between infant feeding and infant health. Pediatrics. 1984;74:615. [PubMed] [Google Scholar]
- 38.Feachem RG, Koblinsky MA. Interventions for the control of diarrhoeal diseases among young children: promotion of breast-feeding. Bull World Health Organ. 1984;62:271. [PMC free article] [PubMed] [Google Scholar]
- 39.Forman MR, Graubard BI, Hoffman HJ. The Pima infant feeding study: breastfeeding and gastroenteritis in the first year of life. Am J Epidemiol. 1984;119:335. doi: 10.1093/oxfordjournals.aje.a113752. [DOI] [PubMed] [Google Scholar]
- 40.Leventhal JM, Shapiro ED, Aten CB. Does breastfeeding protect against infections in infants less than 3 months of age? Pediatrics. 1986;78:8896. [PubMed] [Google Scholar]
- 41.Rubin DH, Leventhal JM, Krsilnikoff PA. Relationship between infant feeding and infectious illness: a prospective study of infants during the first year of life. Pediatrics. 1989;85:464. [PubMed] [Google Scholar]
- 42.Victora CG, Smith PG, Vaughan JP. Infant feeding and deaths due to diarrhea. Am J Epidemiol. 1989;129:1032. doi: 10.1093/oxfordjournals.aje.a115207. [DOI] [PubMed] [Google Scholar]
- 43.Popkin BM, Adair L, Akin JS. Breast-feeding and diarrheal morbidity. Pediatrics. 1990;86:874. [PubMed] [Google Scholar]
- 44.Morrow AL, Pickering LK. Human milk and infectious diseases. In: Long SS, Pickering LK, Prober CG, editors. Principles and Practice of Pediatric Infectious Diseases. Churchill-Livingstone; New York: 1997. pp. 87–95. [Google Scholar]
- 45.Newburg DS, Peterson JA, Ruiz-Palacios GM. High levels of lactadherin in human milk are associated with protection against symptomatic rotavirus infection amongst breast-fed infants. Lancet. 1998;351:1160. doi: 10.1016/s0140-6736(97)10322-1. [DOI] [PubMed] [Google Scholar]
- 46.Wyatt RG, Mata LJ. Bacteria in colostrum and milk in Guatemalan Indian women. J Trop Pediatr. 1969;15:159. doi: 10.1093/tropej/15.4.159. [DOI] [PubMed] [Google Scholar]
- 47.Grazioso CF, Werner AL, Alling DW. Antiinflammatory effects of human milk on chemically induced colitis in rats. Pediatr Res. 1997;42:639. doi: 10.1203/00006450-199711000-00015. [DOI] [PubMed] [Google Scholar]
- 48.Newburg DS. Oligosaccharides and glycoconjugates in human milk: their role in host defense. J Mammary Gland Biol Neoplasia. 1996;1:271. doi: 10.1007/BF02018080. [DOI] [PubMed] [Google Scholar]
- 49.Morrow AL, Pickering LK. Human milk protection against diarrheal disease. Semin Pediatr Infect Dis. 1994;5:236. [Google Scholar]
- 50.Pickering LK, Granoff DM, Erickson JE. Modulation of the immune system by human milk and infant formula containing nucleotides. Pediatrics. 1998;101:242. doi: 10.1542/peds.101.2.242. [DOI] [PubMed] [Google Scholar]
- 51.Hayani K, Guerrero M, Morrow A. Concentration of milk secretory immunoglobulin A against Shigella virulence plasmid-associated antigens as a predictor of symptom status in Shigella-infected breast-fed infants. J Pediatr. 1992;121:852. doi: 10.1016/s0022-3476(05)80327-0. [DOI] [PubMed] [Google Scholar]
- 52.Hayani K, Guerrero M, Ruiz-Palacios G. Evidence for long-term memory of the mucosal immune system: milk secretory immunoglobulin A against Shigella lipopolysaccharides. J Clin Microbiol. 1991;29:2599. doi: 10.1128/jcm.29.11.2599-2603.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Newburg D, Ashkenazi S, Cleary T. Human milk contains the Shiga toxin and Shiga-like toxin receptor glycolipid Gb3. J Infect Dis. 1992;166:832. doi: 10.1093/infdis/166.4.832. [DOI] [PubMed] [Google Scholar]
- 54.Herrera-Insua I, Gomez H, Diaz-Gonzalez V. Human milk lipids bind Shiga toxin. Adv Exp Med Biol. 2001;501:333. doi: 10.1007/978-1-4615-1371-1_41. [DOI] [PubMed] [Google Scholar]
- 55.Gomez H, Ochoa T, Carlin L, Cleary T. Human lactoferrin impairs virulence of Shigella flexneri. J Infect Dis. 2003;187:87. doi: 10.1086/345875. [DOI] [PubMed] [Google Scholar]
- 56.Gomez H, Ochoa T, Herrera-Insua I. Lactoferrin protects rabbits from Shigella flexneri–induced inflammatory enteritis. Infect Immun. 2002;70:7050. doi: 10.1128/IAI.70.12.7050-7053.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Gomez H, Herrera-Insua I, Siddiqui M. Protective role of human lactoferrin against invasion of Shigella flexneri M90T. Adv Exp Med Biol. 2001;501:457. doi: 10.1007/978-1-4615-1371-1_57. [DOI] [PubMed] [Google Scholar]
- 58.Noguera-Obenza M, Ochoa T, Gomez H. Human milk secretory antibodies against attaching and effacing Escherichia coli antigens. Emerg Infect Dis. 2003;9:545. doi: 10.3201/eid0905.020441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Cravioto A, Tello A, Villafan H. Inhibition of localized adhesion of enteropathogenic Escherichia coli to HEp-2 cells by immunoglobulin and oligosaccharide fractions of human colostrum and breast milk. J Infect Dis. 1991;163:1247. doi: 10.1093/infdis/163.6.1247. [DOI] [PubMed] [Google Scholar]
- 60.Ochoa T, Noguera-Obenza M, Ebel F. Lactoferrin impairs type III secretory system function in Enteropathogenic Escherichia coli. Infect Immun. 2003;71:5149. doi: 10.1128/IAI.71.9.5149-5155.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ross CA, Dawes EA. Resistance of the breast fed infant to gastroenteritis. Lancet. 1954;1:994. doi: 10.1016/s0140-6736(54)92395-8. [DOI] [PubMed] [Google Scholar]
- 62.Glass RI, Svennerholm A, Stoll BJ. Milk antibodies protect breastfed children against cholera. N Engl J Med. 1983;308:1389. doi: 10.1056/NEJM198306093082304. [DOI] [PubMed] [Google Scholar]
- 63.Ruiz-Palacios GM, Calva JJ, Pickering LK. Protection of breastfed infants against Campylobacter diarrhea by antibodies in human milk. J Pediatr. 1990;116:707. doi: 10.1016/s0022-3476(05)82652-6. [DOI] [PubMed] [Google Scholar]
- 64.Holmgren J, Svennerholm AM, Lindblad M. Receptor-like glycocompounds in human milk that inhibit classical and El Tor Vibrio cholerae cell adherence (hemagglutination) Infect Immun. 1983;39:147. doi: 10.1128/iai.39.1.147-154.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Laegreid A, Otnaess ABK, Fuglesang J. Human and bovine milk: comparison of ganglioside composition and enterotoxin-inhibitory activity. Pediatr Res. 1986;20:416. doi: 10.1203/00006450-198605000-00008. [DOI] [PubMed] [Google Scholar]
- 66.Hayani KC, Guerrero ML, Morrow AL. Concentration of milk secretory immunoglobulin A against Shigella virulence plasmid-associated antigens as a predictor of symptom status in Shigella-infected breast-fed infants. J Pediatr. 1992;121:852. doi: 10.1016/s0022-3476(05)80327-0. [DOI] [PubMed] [Google Scholar]
- 67.Miotti PG, Gilman RH, Pickering LK. Prevalence of serum and milk antibodies to Giardia lamblia in different populations of lactating women. J Infect Dis. 1985;152:1025. doi: 10.1093/infdis/152.5.1025. [DOI] [PubMed] [Google Scholar]
- 68.Hayani KC, Guerrero ML, Ruiz-Palacios GM. Evidence for long-term memory of the mucosal immune system: milk secretory immunoglobulin A against Shigella lipopolysaccharides. J Clin Microbiol. 1991;29:2599. doi: 10.1128/jcm.29.11.2599-2603.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Walterspiel JN, Morrow AL, Guerrero ML. Protective effect of secretory anti-Giardia lamblia antibodies in human milk against diarrhea. Pediatrics. 1994;93:28. [PubMed] [Google Scholar]
- 70.Tacket CO, Losonsky G, Link H. Protection by milk immunoglobulin concentrate against oral challenge with enterotoxigenic E. coli. N Engl J Med. 1988;318:1240. doi: 10.1056/NEJM198805123181904. [DOI] [PubMed] [Google Scholar]
- 71.Davidson GP, Whyte PBD, Daniels E. Passive immunization of children with bovine colostrum containing antibodies to human rotavirus. Lancet. 1989;2:709. doi: 10.1016/s0140-6736(89)90771-x. [DOI] [PubMed] [Google Scholar]
- 72.Tacket CO, Binion SB, Bostwick E. Efficacy of bovine milk immunoglobulin concentrate in preventing illness after Shigella flexneri challenge. Am J Trop Med Hyg. 1992;47:276. doi: 10.4269/ajtmh.1992.47.276. [DOI] [PubMed] [Google Scholar]
- 73.Polonovsky M, Lespagnol A. Nouvelles acquisitions sur les composés glucidiques du lait de femme. Bull Soc Chem Biol. 1933;15:320. [Google Scholar]
- 74.Montreuil J, Mullet S. Etude des variations des constituants glucidiques du lait de femme au cours de la lactation. Bull Soc Chem Biol. 1960;42:365. [PubMed] [Google Scholar]
- 75.Kobata A. Milk glycoproteins and oligosaccharides. In: Horowitz MI, Pigman W, editors. The Glycoconjugates. I. Academic Press; New York: 1978. p. 423. [Google Scholar]
- 76.Gnoth M, Kunz C, Kinne-Saffran E, Rudloff S. Human milk oligosaccharides are minimally digested in vitro. J Nutr. 2000;130:3014. doi: 10.1093/jn/130.12.3014. [DOI] [PubMed] [Google Scholar]
- 77.Chaturvedi P, Warren C, Buescher C. Survival of human milk oligosaccharides in the intestine of infants. Adv Exp Med Biol. 2001;501:315. doi: 10.1007/978-1-4615-1371-1_39. [DOI] [PubMed] [Google Scholar]
- 78.Cleary TG, Chambers JP, Pickering LK. Protection of suckling mice from heat-stable enterotoxin of Escherichia coli by human milk. J Infect Dis. 1983;148:1114. doi: 10.1093/infdis/148.6.1114. [DOI] [PubMed] [Google Scholar]
- 79.Newburg DS, Pickering LK, McCluer RH. Fucosylated oligosaccharides of human milk protect suckling mice from heat-stable enterotoxin of Escherichia coli. J Infect Dis. 1990;162:1075. doi: 10.1093/infdis/162.5.1075. [DOI] [PubMed] [Google Scholar]
- 80.Huang P, Farkas T, Marionneau S. Noroviruses bind to human ABO, Lewis, and secretor histo-blood group antigens: identification of 4 distinct strain-specific patterns. J Infect Dis. 2003;188:19. doi: 10.1086/375742. [DOI] [PubMed] [Google Scholar]
- 81.Lindesmith L, Moe C, Marionneau S. Human susceptibility and resistance to Norwalk virus infection. Nat Med. 2003;9:548. doi: 10.1038/nm860. [DOI] [PubMed] [Google Scholar]
- 82.Otnaess AB, Svennerholm AM. Non-immunoglobulin fraction of human milk protects rabbits against enterotoxin-induced intestinal fluid secretion. Infect Immun. 1982;35:738. doi: 10.1128/iai.35.2.738-740.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Otnaess ABK, Laegreid A, Ertesvag K. Inhibition of enterotoxin from Escherichia coli and Vibrio cholerae by gangliosides from human milk. Infect Immun. 1983;40:563. doi: 10.1128/iai.40.2.563-569.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yolken RH, Peterson JA, Vonderfecht SL. Human milk mucin inhibits rotavirus replication and prevents experimental gastroenteritis. J Clin Invest. 1992;90:1984. doi: 10.1172/JCI116078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cooper ML, Keller HM, Walters EW. Isolation of enteropathogenic Escherichia coli from mothers and newborn infants. Am J Dis Child. 1959;97:255. doi: 10.1001/archpedi.1959.02070010257001. [DOI] [PubMed] [Google Scholar]
- 86.Ocklitz HW, Schmidt EF. Enteropathogenic Escherichia coli serotypes: infection of the newborn through mother. BMJ. 1957;2:1036. doi: 10.1136/bmj.2.5052.1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gareau FE, Mackel DC, Boring JR., III The acquisition of fecal flora by infants from their mothers during birth. J Pediatr. 1959;54:313. doi: 10.1016/s0022-3476(59)80005-6. [DOI] [PubMed] [Google Scholar]
- 88.Rosner R. Antepartum culture findings of mothers in relation to infantile diarrhea. Am J Clin Pathol. 1966;45:732. doi: 10.1093/ajcp/45.6.732. [DOI] [PubMed] [Google Scholar]
- 89.Nocard E, Leclainche E. Les Maladies Microbiennes des Animaux. 2nd ed. Masson; Paris: 1898. p. 106. [Google Scholar]
- 90.Joest E. Untersuchungen über Kalberruhr. Z Tiermed. 1903;7:377. [Google Scholar]
- 91.Titze C, Weichel A. Die Ätiologie der Kalberruhr. Berl Tierarztl Wochenschr. 1908;26:457. [Google Scholar]
- 92.Jensen CO. vol 6. G Fischer; Jena: 1913. p. 131. (Handbuch der pathogenen Microorganismen). [Google Scholar]
- 93.Smith T, Orcutt ML. The bacteriology of the intestinal tract of young calves with special reference to the early diarrhea (“scours”) J Exp Med. 1925;41:89. doi: 10.1084/jem.41.1.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Neonatal enteric infections caused by Escherichia coli. Tennant B, editor. Ann N Y Acad Sci. 1971;176:1. [Google Scholar]
- 95.Moro E. Quoted in Adam A. Über die Biologie der Dyspepsiecoli und ihre Beziehungen zur Pathogenese der Dyspepsie und Intoxikation. Jahrb Kinderheilkd. 1923;101:295. [Google Scholar]
- 96.Adam A. Über die Biologie der Dyspepsiecoli und ihre Beziehungen zur Pathogenese der Dyspepsie und Intoxikation. Jahrb Kinderheilkd. 1923;101:295. [Google Scholar]
- 97.Adam A. Zur Frage der bakteriellen Ätiologie der sogenannten alimentaren Intoxikation. Jahrb Kinderheilkd. 1927;116:8. [Google Scholar]
- 98.Nataro JP, Kaper JB. Diarrheagenic Escherichia coli. Clin Microbiol Rev. 1998;11:142. doi: 10.1128/cmr.11.1.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Donnenberg MS, Kaper JB. Enteropathogenic Escherichia coli. Infect Immun. 1992;60:3953. doi: 10.1128/iai.60.10.3953-3961.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Levine MM. Escherichia coli that cause diarrhea: enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorrhagic, and enteroadherent. J Infect Dis. 1987;155:377. doi: 10.1093/infdis/155.3.377. [DOI] [PubMed] [Google Scholar]
- 101.Guerrant RL, Thielman NM. Types of Escherichia coli enteropathogens. In: Blaser MJ, Smith PD, Ravdin JL, editors. Infections of the Gastrointestinal Tract. Raven Press; New York: 1995. p. 687. [Google Scholar]
- 102.Schlager TA, Guerrant RL. Seven possible mechanisms for Escherichia coli diarrhea. Infect Dis Clin North Am. 1988;2:607. [PubMed] [Google Scholar]
- 103.Steiner TS, Lima AM, Nataro JP, Guerrant R. Enteroaggregative Escherichia coli produce intestinal inflammation and growth impairment and cause interleukin-8 release from intestinal epithelial cells. J Infect Dis. 1998;177:88. doi: 10.1086/513809. [DOI] [PubMed] [Google Scholar]
- 104.Guerrant R, Steiner T. Principles and syndromes of enteric infections. In: Mandell GL, Bennett J, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 5th ed. WB Saunders; Philadelphia: 1999. [Google Scholar]
- 105.Spangler BD. Structure and function of cholera toxin and the related Escherichia coli heat-labile enterotoxin. Microbiol Rev. 1992;56:622. doi: 10.1128/mr.56.4.622-647.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Guerrant RL, Ganguly U, Casper AGT. Effect of Escherichia coli on fluid transport across canine small bowel: mechanism and time-course with enterotoxin and whole bacterial cells. J Clin Invest. 1973;52:1707. doi: 10.1172/JCI107352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Peterson JW, Ochoa G. Role of prostaglandins and cAMP in the secretory effects of cholera toxin. Science. 1989;245:857. doi: 10.1126/science.2549637. [DOI] [PubMed] [Google Scholar]
- 108.Peterson JW, Reitmeyer JC, Jackson CA. Protein synthesis is required for cholera toxin-induced stimulation or arachidonic acid metabolism. Biochim Biophys Acta. 1991;1092:79. doi: 10.1016/0167-4889(91)90179-2. [DOI] [PubMed] [Google Scholar]
- 109.Thielman NM, Marcinkiewics M, Sarosiek J. The role of platelet activating factor in Chinese hamster ovary cell responses to cholera toxin. J Clin Invest. 1997;99:1999. doi: 10.1172/JCI119368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Guerrant RL, Fang GD, Thielman NM, Fonteles MC. Role of platelet activating factor (PAF) in the intestinal epithelial secretory and Chinese hamster ovary (CHO) cell cytoskeletal responses to cholera toxin. Proc Natl Acad Sci U S A. 1994;91:9655. doi: 10.1073/pnas.91.20.9655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Hughes JM, Murad F, Chang B. Role of cyclic GMP in the action of heat-stable enterotoxin of Escherichia coli. Nature. 1978;271:755. doi: 10.1038/271755a0. [DOI] [PubMed] [Google Scholar]
- 112.Field M, Graf LH, Jr, Laird WJ. Heat-stable enterotoxin of Escherichia coli: in vitro effects on guanylate cyclase activity, cyclic GMP concentration, and ion transport in small intestine. Proc Natl Acad Sci U S A. 1978;75:2800. doi: 10.1073/pnas.75.6.2800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Guerrant RL, Hughes JM, Chang B. Activation of intestinal guanylate cyclase by heat-stable enterotoxin of Escherichia coli: studies of tissue specificity, potential receptors and intermediates. J Infect Dis. 1980;142:220. doi: 10.1093/infdis/142.2.220. [DOI] [PubMed] [Google Scholar]
- 114.Kennedy DJ, Greenberg RN, Dunn JA. Effects of E. coli heat stabile enterotoxin STb on intestines of mice, rats, rabbits, and piglets. Infect Immun. 1984;46:639. doi: 10.1128/iai.46.3.639-643.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Weikel CS, Nellans HN, Guerrant RL. In vivo and in vitro effects of a novel enterotoxin, STb, produced by E. coli. J Infect Dis. 1986;153:893. doi: 10.1093/infdis/153.5.893. [DOI] [PubMed] [Google Scholar]
- 116.Weikel CS, Tiemens KM, Moseley SL. Species specificity and lack of production of STb enterotoxin by E. coli strains isolated from humans with diarrheal illness. Infect Immun. 1986;52:323. doi: 10.1128/iai.52.1.323-325.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.DuPont HL, Formal SB, Hornick RB. Pathogenesis of Escherichia coli diarrhea. N Engl J Med. 1971;285:1. doi: 10.1056/NEJM197107012850101. [DOI] [PubMed] [Google Scholar]
- 118.Tulloch EF, Ryan KJ, Formal SB. Invasive enteropathic Escherichia coli dysentery. An outbreak in 28 adults. Ann Intern Med. 1973;79:13. doi: 10.7326/0003-4819-79-1-13. [DOI] [PubMed] [Google Scholar]
- 119.Nataro JP, Kaper JB, Robins-Browne R. Patterns of adherence of diarrheagenic E. coli to HEp-2 cells. Pediatr Infect Dis J. 1987;6:829. doi: 10.1097/00006454-198709000-00008. [DOI] [PubMed] [Google Scholar]
- 120.Kenny B, DeVinney R, Stein M. Enteropathogenic E. coli (EPEC) transfers its receptor for intimate adherence into mammalian cells. Cell. 1997;91:511. doi: 10.1016/s0092-8674(00)80437-7. [DOI] [PubMed] [Google Scholar]
- 121.Nougayrede JP, Fernandes PJ, Donnenberg MS. Adhesion of enteropathogenic Escherichia coli to host cells. Cell Microbiol. 2003;5:359. doi: 10.1046/j.1462-5822.2003.00281.x. [DOI] [PubMed] [Google Scholar]
- 122.Giron JA, Fry J, Frankel G. Diffuse-adhering Escherichia coli (DAEC) as a putative cause of diarrhea in Mayan children in Mexico. J Infect Dis. 1991;163:507. doi: 10.1093/infdis/163.3.507. [DOI] [PubMed] [Google Scholar]
- 123.Peiffer I, Bernet-Camard MF, Rousset M, Servin AL. Impairments in enzyme activity and biosynthesis of brush border-associated hydrolases in human intestinal Caco-2/TC7 cells infected by members of the Afa/Dr family of diffusely adhering Escherichia coli. Cell Microbiol. 2001;3:341. doi: 10.1046/j.1462-5822.2001.00121.x. [DOI] [PubMed] [Google Scholar]
- 124.Wanke CA, Schorling JB, Barrett LJ. Adherence traits of Escherichia coli, alone and in association with other stool pathogens: potential role in pathogenesis of persistent diarrhea in an urban Brazilian slum. Pediatr J Infect Dis. 1991;10:746. doi: 10.1097/00006454-199110000-00006. [DOI] [PubMed] [Google Scholar]
- 125.Okeke IN, Nataro JP. Enteroaggregative Escherichia coli. Lancet Infect Dis. 2001;1:304. doi: 10.1016/S1473-3099(01)00144-X. [DOI] [PubMed] [Google Scholar]
- 126.Rowe B, Scotland SM, Gross RJ. Enterotoxigenic Escherichia coli causing infantile enteritis in Britain. Lancet. 1977;1:90. doi: 10.1016/s0140-6736(77)91099-6. [DOI] [PubMed] [Google Scholar]
- 127.Trabulsi LR, Fernandes MFR, Zuliani ME. Novas bacterias patogenicas para o intestino do homen. Rev Inst Med Trop São Paulo. 1967;9:31. [PubMed] [Google Scholar]
- 128.Sereny B. Experimental Shigella keratoconjunctivitis: a preliminary report. Acta Microbiol Acad Sci Hung. 1955;2:293. [PubMed] [Google Scholar]
- 129.Skerman FJ, Formal SB, Falkow S. Plasmid-associated enterotoxin production in a strain of Escherichia coli isolated from humans. Infect Immun. 1972;56:22. doi: 10.1128/iai.5.4.622-624.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Takeda Y, Murphy J. Bacteriophage conversion of heat-labile enterotoxin in Escherichia coli. J Bacteriol. 1978;133:172. doi: 10.1128/jb.133.1.172-177.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lathe R, Hirth P. Cell-free synthesis of enterotoxin of E. coli from a cloned gene. Nature. 1980;284:473. doi: 10.1038/284473a0. [DOI] [PubMed] [Google Scholar]
- 132.Merson MH, Rowe B, Black RE. Use of antisera for identification of enterotoxigenic Escherichia coli. Lancet. 1980;2:222. doi: 10.1016/s0140-6736(80)90118-x. [DOI] [PubMed] [Google Scholar]
- 133.Evans DG, Evans DJ, Jr, Pierce NF. Differences in the response of rabbit small intestine to heat-labile and heat-stable enterotoxins of Escherichia coli. Infect Immun. 1973;7:873. doi: 10.1128/iai.7.6.873-880.1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Sears CL, Kaper JB. Enteric bacterial toxins: mechanisms of action and linkage to intestinal secretion. Microbiol Rev. 1996;60:167. doi: 10.1128/mr.60.1.167-215.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Donta ST, Moon HW, Whipp SC. Detection of heat-labile Escherichia coli enterotoxin with the use of adrenal cells in tissue cultures. Science. 1974;183:334. doi: 10.1126/science.183.4122.334. [DOI] [PubMed] [Google Scholar]
- 136.Yolken RH, Greenberg HB, Merson MH. Enzyme-linked immunosorbent assay for detection of Escherichia coli heat-labile enterotoxin. J Clin Microbiol. 1977;6:439. doi: 10.1128/jcm.6.5.439-444.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Dean AG, Ching YC, Williams RG. Test for Escherichia coli enterotoxin using infant mice: application in a study of diarrhea in children in Honolulu. J Infect Dis. 1972;125:407. doi: 10.1093/infdis/125.4.407. [DOI] [PubMed] [Google Scholar]
- 138.Giannella RA. Suckling mouse model for detection of heat-stable Escherichia coli enterotoxin: characteristics of the model. Infect Immun. 1976;14:95. doi: 10.1128/iai.14.1.95-99.1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Klipstein FA, Holdeman LV, Corcino JJ. Enterotoxigenic intestinal bacteria in tropical sprue. Ann Intern Med. 1973;79:632. doi: 10.7326/0003-4819-79-5-632. [DOI] [PubMed] [Google Scholar]
- 140.Smith HW, Halls S. Observations by ligated intestinal segment and oral inoculation methods on Escherichia coli infections in pigs, calves, lambs and rabbits. J Pathol Bacteriol. 1967;93:499. doi: 10.1002/path.1700930211. [DOI] [PubMed] [Google Scholar]
- 141.Evans DG, Silver RP, Evans DJ., Jr Plasmid-controlled colonization factor associated with virulence in Escherichia coli enterotoxigenic for humans. Infect Immun. 1975;12:656. doi: 10.1128/iai.12.3.656-667.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.De SN, Chatterjee DN. An experimental study of the mechanism of action of Vibrio cholerae on the intestinal mucous membrane. J Pathol Bacteriol. 1953;66:559. doi: 10.1002/path.1700660228. [DOI] [PubMed] [Google Scholar]
- 143.De SN, Bhattachaya K, Sakar JK. A study of the pathogenicity of strains of Bacterium coli from acute and chronic enteritis. J Pathol Bacteriol. 1956;71:201. doi: 10.1002/path.1700710126. [DOI] [PubMed] [Google Scholar]
- 144.Taylor J, Wilkins MP, Payne JM. Relation of rabbit gut reaction to enteropathogenic Escherichia coli. Br J Exp Pathol. 1961;42:43. [PMC free article] [PubMed] [Google Scholar]
- 145.Taylor J, Bettelheim KA. The action of chloroform-killed suspensions of enteropathogenic Escherichia coli on ligated rabbit gut segments. J Gen Microbiol. 1966;42:309. doi: 10.1099/00221287-42-2-309. [DOI] [PubMed] [Google Scholar]
- 146.Trabulsi LR. Revelacao de colibacilos associados as diarreias infantis pelo metodo da infeccao experimental de alca ligade do intestino do coehlo. Rev Inst Med Trop São Paulo. 1964;6:197. [PubMed] [Google Scholar]
- 147.Moon HW, Sorensen DK, Sautter JH. Association of Escherichia coli with diarrheal disease of the newborn pig. Am J Vet Res. 1966;27:1107. [PubMed] [Google Scholar]
- 148.Smith HW, Halls S. Studies on Escherichia coli enterotoxin. J Pathol Bacteriol. 1967;93:531. doi: 10.1002/path.1700930212. [DOI] [PubMed] [Google Scholar]
- 149.Truszcynski M, Pilaszek J. Effects of injection of enterotoxin, endotoxin or live culture of Escherichia coli into the small intestine of pigs. Res Vet Sci. 1969;10:469. [PubMed] [Google Scholar]
- 150.Gyles CL, Barnum DA. A heat-labile enterotoxin from strains of Escherichia coli enteropathogenic for pigs. J Infect Dis. 1969;120:419. doi: 10.1093/infdis/120.4.419. [DOI] [PubMed] [Google Scholar]
- 151.Gorbach SL, Banwell JG, Chatterjee BD. Acute undifferentiated human diarrhea in the tropics. I. Alterations in intestinal microflora. J Clin Invest. 1971;50:881. doi: 10.1172/JCI106560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Banwell JG, Gorbach SL, Pierce NF. Acute undifferentiated human diarrhea in the tropics. II. Alterations in intestinal fluid and electrolyte movements. J Clin Invest. 1971;50:890. doi: 10.1172/JCI106561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sack RB, Gorbach SL, Banwell JG. Enterotoxigenic Escherichia coli isolated from patients with severe cholera-like disease. J Infect Dis. 1971;123:278. doi: 10.1093/infdis/123.4.378. [DOI] [PubMed] [Google Scholar]
- 154.Pierce NF, Wallace CK. Stimulation of jejunal secretion by a crude Escherichia coli enterotoxin. Gastroenterology. 1972;63:439. [PubMed] [Google Scholar]
- 155.Guerrant RL, Carpenter CCJ, Pierce NF. Experimental E. coli diarrhea: effects of viable bacteria and enterotoxin. Trans Assoc Am Physicians. 1973;86:111. [PubMed] [Google Scholar]
- 156.Kantor HS, Tao P, Gorbach SL. Stimulation of intestinal adenyl cyclase by Escherichia coli enterotoxin: comparison of strains from an infant and an adult with diarrhea. J Infect Dis. 1974;129:1. doi: 10.1093/infdis/129.1.1. [DOI] [PubMed] [Google Scholar]
- 157.Smith HW, Gyles CL. The relationship between two apparently different enterotoxins produced by enteropathogenic strains of Escherichia coli of porcine origin. J Med Microbiol. 1970;3:387. doi: 10.1099/00222615-3-3-387. [DOI] [PubMed] [Google Scholar]
- 158.Kohler EM. Observations on enterotoxins produced by enteropathogenic Escherichia coli. Ann N Y Acad Sci. 1971;176:212. [Google Scholar]
- 159.Moon HW, Whipp SC. Systems for testing the enteropathogenicity of Escherichia coli. Ann N Y Acad Sci. 1971;176:197. [Google Scholar]
- 160.Smith HW, Linggood MA. Observations on the pathogenic properties of the K88, HLY and ENT plasmids of Escherichia coli with particular reference to porcine diarrhea. J Med Microbiol. 1971;4:467. doi: 10.1099/00222615-4-4-467. [DOI] [PubMed] [Google Scholar]
- 161.Holmgren J, Svennerholm AM. Enzyme-linked immunosorbent assays for cholera serology. Infect Immun. 1973;7:759. doi: 10.1128/iai.7.5.759-763.1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Svennerholm AM, Holmgren J. Identification of Escherichia coli heat-labile enterotoxin by means of a ganglioside immunosorbent assay (Gm1 ELISA) procedure. Curr Microbiol. 1978;1:19. [Google Scholar]
- 163.Sack DA, Huda S, Neogi PKB. Microtiter ganglioside enzyme-linked immunosorbent assay for Vibrio and Escherichia coli heat-labile enterotoxins and antitoxin. J Clin Microbiol. 1980;11:35. doi: 10.1128/jcm.11.1.35-40.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Currie MG, Fok KF, Kato J. Guanylin: an endogenous activator of intestinal guanylate cyclase. Proc Natl Acad Sci U S A. 1992;89:947. doi: 10.1073/pnas.89.3.947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wadstrom T, Aust-Kettis A, Habte D. Enterotoxin-producing bacteria and parasites in stools of Ethiopian children with diarrhoeal disease. Arch Dis Child. 1976;51:865. doi: 10.1136/adc.51.11.865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Wachsmuth K, Wells J, Shipley P. Heat-labile enterotoxin production in isolates from a shipboard outbreak of human diarrheal illness. Infect Immun. 1979;24:793. doi: 10.1128/iai.24.3.793-797.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Sandefur PD, Peterson JW. Isolation of skin permeability factors from culture filtrates of Salmonella typhimurium. Infect Immun. 1976;14:671. doi: 10.1128/iai.14.3.671-679.1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Sandefur PD, Peterson JW. Neutralization of Salmonella toxin–induced elongation of Chinese hamster ovary cells by cholera antitoxin. Infect Immun. 1977;15:988. doi: 10.1128/iai.15.3.988-992.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Gibbons RA, Sellwood R, Burrows M. Inheritance of resistance to neonatal E. coli diarrhoea in the pig: examination of the genetic system. Theor Appl Genet. 1977;51:65. doi: 10.1007/BF00299479. [DOI] [PubMed] [Google Scholar]
- 170.Wolf MK. Occurrence, distribution and associations of O and H serogroups, colonization factor antigens, and toxins of enterotoxigenic Escherichia coli. Clin Microbiol Rev. 1997;10:569. doi: 10.1128/cmr.10.4.569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Cassels FJ, Wolf MK. Colonization factors of diarrheagenic E. coli and their intestinal receptors. J Ind Microbiol. 1995;15:214. doi: 10.1007/BF01569828. [DOI] [PubMed] [Google Scholar]
- 172.Katz DE, DeLorimier AJ, Wolf MK. Oral immunization of adult volunteers with microencapsulated enterotoxigenic Escherichia coli (ETEC) CS6 antigen. Vaccine. 2003;21:341. doi: 10.1016/s0264-410x(02)00613-8. [DOI] [PubMed] [Google Scholar]
- 173.Sack DA, Kaminsky DC, Sack RB. Enterotoxigenic Escherichia coli diarrhea of travelers: a prospective study of American Peace Corps volunteers. Johns Hopkins Med J. 1977;141:63. [PubMed] [Google Scholar]
- 174.Guerrant RL, Rouse JD, Hughes JM. Turista among members of the Yale Glee Club in Latin America. Am J Trop Med Hyg. 1980;29:895. doi: 10.4269/ajtmh.1980.29.895. [DOI] [PubMed] [Google Scholar]
- 175.Guerrant RL, Moore RA, Kirschenfeld PM. Role of toxigenic and invasive bacteria in acute diarrhea of childhood. N Engl J Med. 1975;293:567. doi: 10.1056/NEJM197509182931201. [DOI] [PubMed] [Google Scholar]
- 176.Echeverria P, Blacklow NR, Smith DH. Role of heat-labile toxigenic Escherichia coli and reovirus-like agent in diarrhoea in Boston children. Lancet. 1975;2:1113. doi: 10.1016/s0140-6736(75)91004-1. [DOI] [PubMed] [Google Scholar]
- 177.Viboud GI, Binsztein N, Svennerholm AM. Characterization of monoclonal antibodies against putative colonization factors of enterotoxigenic Escherichia coli and their use in an epidemiological study. J Clin Microbiol. 1993;31:558. doi: 10.1128/jcm.31.3.558-564.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Ryder RW, Sack DA, Kapikian AZ. Enterotoxigenic Escherichia coli and reovirus-like agent in rural Bangladesh. Lancet. 1976;1:659. doi: 10.1016/s0140-6736(76)92776-8. [DOI] [PubMed] [Google Scholar]
- 179.Nalin DR, McLaughlin JC, Rahaman M. Enterotoxigenic Escherichia coli and idiopathic diarrhea in Bangladesh. Lancet. 1975;2:1116. doi: 10.1016/s0140-6736(75)91005-3. [DOI] [PubMed] [Google Scholar]
- 180.Lopez-Vidal Y, Calva JJ, Trujillo A. Enterotoxins and adhesins of enterotoxigenic Escherichia coli: are they risk factors for acute diarrhea in the community? J Infect Dis. 1990;162:442. doi: 10.1093/infdis/162.2.442. [DOI] [PubMed] [Google Scholar]
- 181.McLean M, Brennan R, Hughes JM. Etiology and oral rehydration therapy of childhood diarrhea in northeastern Brazil. Bull Pan Am Health Organ. 1981;15:318. [PubMed] [Google Scholar]
- 182.Sack RB, Hirschhorn N, Brownlee I. Enterotoxigenic Escherichia coli associated diarrheal disease in Apache children. N Engl J Med. 1975;292:1041. doi: 10.1056/NEJM197505152922001. [DOI] [PubMed] [Google Scholar]
- 183.Hughes JM, Rouse JD, Barada AF. Etiology of summer diarrhea among the Navajo. Am J Trop Med Hyg. 1980;29:613. doi: 10.4269/ajtmh.1980.29.613. [DOI] [PubMed] [Google Scholar]
- 184.Huilan S, Zhen LG, Mathan MM. Etiology of acute diarrhea among children in developing countries: a multicentre study in five countries. Bull World Health Organ. 1991;69:549. [PMC free article] [PubMed] [Google Scholar]
- 185.Blanco J, Gonzalez EA, Blanco M. Enterotoxigenic Escherichia coli associated with infant diarrhoea in Galicia, northwestern Spain. J Med Microbiol. 1991;35:162. doi: 10.1099/00222615-35-3-162. [DOI] [PubMed] [Google Scholar]
- 186.Nations MK, de Sousa MA, Correia LL, daSilva DM. Brazilian popular healers as effective promoters of oral rehydration therapy (ORT) and related child survival strategies. Bull Pan Am Health Organ. 1988;22:335. [PubMed] [Google Scholar]
- 187.Korzeniowski OM, Dantas W, Trabulsi CR. A controlled study of endemic sporadic diarrhea among adult residents of southern Brazil. Trans R Soc Trop Med Hyg. 1984;78:363. doi: 10.1016/0035-9203(84)90123-8. [DOI] [PubMed] [Google Scholar]
- 188.Kudoh Y, Hiroshi ZY, Matsushita S. Outbreaks of acute enteritis due to heat-stable enterotoxin-producing strains of Escherichia coli. Microbiol Immunol. 1977;21:175. doi: 10.1111/j.1348-0421.1977.tb00278.x. [DOI] [PubMed] [Google Scholar]
- 189.Rosenberg ML, Koplan JP, Wachsmuth IK. Epidemic diarrhea at Crater Lake from enterotoxigenic Escherichia coli: a large waterborne outbreak. Ann Intern Med. 1977;86:714. doi: 10.7326/0003-4819-86-6-714. [DOI] [PubMed] [Google Scholar]
- 190.Ryder RW, Wachsmuth IK, Buxton AE. Infantile diarrhea produced by heat-stable enterotoxigenic Escherichia coli. N Engl J Med. 1976;295:849. doi: 10.1056/NEJM197610142951601. [DOI] [PubMed] [Google Scholar]
- 191.Gross RJ, Rowe B, Henderson A. A new Escherichia coli O-group, O159, associated with outbreaks of enteritis in infants. Scand J Infect Dis. 1976;8:195. doi: 10.3109/inf.1976.8.issue-3.14. [DOI] [PubMed] [Google Scholar]
- 192.Albert MJ, Faruque SM, Faruque AS. Controlled study of Escherichia coli diarrheal infections in Bangladeshi children. J Clin Microbiol. 1995;33:973. doi: 10.1128/jcm.33.4.973-977.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Guerrant RL, Dickens MD, Wenzel RP. Toxigenic bacterial diarrhea: nursery outbreak involving multiple bacterial strains. J Pediatr. 1976;89:885. doi: 10.1016/s0022-3476(76)80591-4. [DOI] [PubMed] [Google Scholar]
- 194.Abe A, Komase K, Bangtrakulnonth A. Trivalent heat-labile- and heat-stable-enterotoxin probe conjugated with horseradish peroxidase for detection of enterotoxigenic Escherichia coli by hybridization. J Clin Microbiol. 1990;28:2616. doi: 10.1128/jcm.28.12.2616-2620.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Sommerfelt H, Svennerholm AM, Kallard KH. Comparative study of colony hybridization with synthetic oligonucleotide probes and enzyme-linked immunosorbent assay for identification of enterotoxigenic E. coli. J Clin Microbiol. 1988;26:530. doi: 10.1128/jcm.26.3.530-534.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Lund A, Wasteson W, Olsvik O. Immunomagnetic separation and DNA hybridization for detection of enterotoxigenic Escherichia coli in a piglet model. J Clin Microbiol. 1991;29:2259. doi: 10.1128/jcm.29.10.2259-2262.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Hornes E, Wasteson W, Olsvik O. Detection of Escherichia coli heat-stable enterotoxin genes in pig stool specimens by an immobilized, calorimetric, nested polymerase chain reaction. J Clin Microbiol. 1991;29:2375. doi: 10.1128/jcm.29.11.2375-2379.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Guerrant RL, Van Gilder T, Steiner TS. Practice guidelines for the management of infectious diarrhea. Clin Infect Dis. 2001;32:331. doi: 10.1086/318514. [DOI] [PubMed] [Google Scholar]
- 199.Pizarro D, Posada G, Mata L. Oral rehydration of neonates with dehydrating diarrheas. Lancet. 1979;2:1209. doi: 10.1016/s0140-6736(79)92332-8. [DOI] [PubMed] [Google Scholar]
- 200.Santosham M, Daum RS, Dillman L. Oral rehydration therapy for infantile diarrhea. A controlled study of well nourished children hospitalized in the United States and Panama. N Engl J Med. 1982;306:1070. doi: 10.1056/NEJM198205063061802. [DOI] [PubMed] [Google Scholar]
- 201.Molla AM, Molla A, Nath SK. Food based oral rehydration salt solutions for acute childhood diarrhoea. Lancet. 1989;2:429. doi: 10.1016/s0140-6736(89)90601-6. [DOI] [PubMed] [Google Scholar]
- 202.Walker SH, Gahol VP, Quintero BA. Sodium and water content of feedings for use in infants with diarrhea. Clin Pediatr. 1981;20:199. doi: 10.1177/000992288102000305. [DOI] [PubMed] [Google Scholar]
- 203.International Study Group on Reduced Osmolality ORS Solution Multicentre evaluation of reduced-osmolality oral rehydration salts solution. Lancet. 1995;345:282. [PubMed] [Google Scholar]
- 204.Echeverria P, Ulyangco CV, Ho MT. Antimicrobial resistance and enterotoxin production among isolates of Escherichia coli in the Far East. Lancet. 1978;2:589. doi: 10.1016/s0140-6736(78)92820-9. [DOI] [PubMed] [Google Scholar]
- 205.Harris JR, Wachsmuth IK, Davis BR. High-molecular-weight plasmid correlates with Escherichia coli invasiveness. Infect Immun. 1982;37:1295. doi: 10.1128/iai.37.3.1295-1298.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.De Assis A. Shigella guanabara, tipo serologico destacado do grupo B ceylonensis-dispar. O Hospital. 1948;33:508. [PubMed] [Google Scholar]
- 207.Lapatsanis PD, Irving IM. A study of specific E. coli infections occurring in a unit for surgical neonates. Acta Paediatr. 1963;52:436. doi: 10.1111/j.1651-2227.1963.tb03803.x. [DOI] [PubMed] [Google Scholar]
- 208.Gordillo ME, Reeve GR, Pappas J. Molecular characterization of strains of enteroinvasive Escherichia coli O143, including isolates from a large outbreak in Houston, Texas. J Clin Microbiol. 1992;30:889. doi: 10.1128/jcm.30.4.889-893.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Goldschmidt R. Untersuchungen zur Ätiologie der Durchfallserkrankungen des Säuglings. Jahrb Kinderheilkd. 1933;139:318. [Google Scholar]
- 210.Dulaney AD, Michelson ID. A study of E. coli mutable from an outbreak of diarrhea in the new-born. Am J Public Health. 1935;25:1241. doi: 10.2105/ajph.25.11.1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Bray J. Isolation of antigenically homogenous strains of Bact. coli neopolitanum from summer diarrhea of infants. J Pathol Bacteriol. 1945;57:239. [Google Scholar]
- 212.Bray J, Beaven TED. Slide agglutination of Bacterium coli var. neopolitanum in summer diarrhea. J Pathol Bacteriol. 1948;60:395. [Google Scholar]
- 213.Olarte J, Varela G. A complete somatic antigen common to Salmonella adelaide, Escherichia coli-gomez and Escherichia coli O111:B4. J Lab Clin Med (Lond) 1952;40:252. [PubMed] [Google Scholar]
- 214.Giles C, Sangster G. An outbreak of infantile gastroenteritis in Aberdeen. J Hyg. 1948;46:1. doi: 10.1017/s0022172400036007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Giles C, Sangster G, Smith J. Epidemic gastroenteritis of infants in Aberdeen during 1947. Arch Dis Child. 1949;24:45. doi: 10.1136/adc.24.117.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Kaufman F, Dupont A. Escherichia strains from infantile epidemic gastroenteritis. Acta Pathol Microbiol Scand. 1950;27:552. doi: 10.1111/j.1699-0463.1950.tb04927.x. [DOI] [PubMed] [Google Scholar]
- 217.Edwards PR, Ewing WH. Identification of Enterobacteriaceae. 3rd ed. Burgess Publishing; Minneapolis, Minn: 1972. [Google Scholar]
- 218.Neter E, Korns RF, Trussell RF. Association of Escherichia coli serogroup O111 with two hospital outbreaks of epidemic diarrhea of the newborn infant in New York State during 1947. Pediatrics. 1953;12:377. [PubMed] [Google Scholar]
- 219.Neter E, Westphal O, Luderitz O. Demonstration of antibodies against enteropathogenic Escherichia coli in sera of children of various ages. Pediatrics. 1955;16:801. [PubMed] [Google Scholar]
- 220.Gronroos JA. Investigations on certain Escherichia coli serotypes, with special reference to infantile diarrhoea. Ann Med. 1954;32:9. [PubMed] [Google Scholar]
- 221.Donnenberg MS, Whittam TS. Pathogenesis and evolution of virulence in enteropathogenic and enterohemorrhagic Escherichia coli. J Clin Invest. 2001;107:539. doi: 10.1172/JCI12404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Trabulsi LR, Keller R, Tardelli Gomes TA. Typical and atypical enteropathogenic Escherichia coli. Emerg Infect Dis. 2002;8:508. doi: 10.3201/eid0805.010385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Moyenuddin M, Rahman KM. Enteropathogenic Escherichia coli diarrhea in hospitalized children in Bangladesh. J Clin Microbiol. 1985;22:838. doi: 10.1128/jcm.22.5.838-840.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Bower JR, Congeni BL, Cleary TG. Escherichia coli O114: nonmotile as a pathogen in an outbreak of severe diarrhea associated with a day care center. J Infect Dis. 1989;160:243. doi: 10.1093/infdis/160.2.243. [DOI] [PubMed] [Google Scholar]
- 225.Neter E. Discussion. Ann N Y Acad Sci. 1971;176:136. [Google Scholar]
- 226.Marker SC, Blazevic DJ. Enteropathogenic serotypes of E. coli. J Pediatr. 1977;90:1037. doi: 10.1016/s0022-3476(77)80600-8. [DOI] [PubMed] [Google Scholar]
- 227.Farmer JJ, Davis BR, Cherry WB. “Enteropathogenic serotypes” of Escherichia coli which really are not. J Pediatr. 1977;90:1047. doi: 10.1016/s0022-3476(77)80617-3. [DOI] [PubMed] [Google Scholar]
- 228.Gordon JE. Diarrheal disease of early childhood—worldwide scope of the problem. Ann N Y Acad Sci. 1971;176:9. [Google Scholar]
- 229.Gordon JE, Chitkara ID, Wyon JB. Weanling diarrhea. Am J Med Sci. 1963;245:345. [PubMed] [Google Scholar]
- 230.Bernet CP, Graber CD, Anthony CW. Association of Escherichia coli O127:B8 with an outbreak of infantile gastroenteritis and its concurrent distribution in the pediatric population. J Pediatr. 1955;47:287. doi: 10.1016/s0022-3476(55)80002-9. [DOI] [PubMed] [Google Scholar]
- 231.Cooper ML, Walters EW, Keller HM. Epidemic diarrhea among infants associated with the isolation of a new serotype of Escherichia coli: E. coli O127:B8. Pediatrics. 1955;16:215. [PubMed] [Google Scholar]
- 232.Laurell G, Magnusson JH, Frisell E. Epidemic infantile diarrhea and vomiting. Acta Paediatr. 1951;40:302. doi: 10.1111/j.1651-2227.1951.tb15797.x. [DOI] [PubMed] [Google Scholar]
- 233.Martineau B, Raymond R, Jeliu G. Bacteriological and clinical study of gastroenteritis and enteropathogenic Escherichia coli O127:B8. Can Med Assoc J. 1958;79:351. [PMC free article] [PubMed] [Google Scholar]
- 234.Wheeler WE, Wainerman B. The treatment and prevention of epidemic infantile diarrhea due to E. coli O111 by the use of chloramphenicol and neomycin. Pediatrics. 1954;14:357. [PubMed] [Google Scholar]
- 235.Kaslow RA, Taylor A, Jr, Dweck HS. Enteropathogenic Escherichia coli infection in a newborn nursery. Am J Dis Child. 1974;128:797. doi: 10.1001/archpedi.1974.02110310045007. [DOI] [PubMed] [Google Scholar]
- 236.Boyer KM, Peterson NJ, Farzaneh I. An outbreak of gastroenteritis due to E. coli O142 in a neonatal nursery. J Pediatr. 1975;86:919. doi: 10.1016/s0022-3476(75)80230-7. [DOI] [PubMed] [Google Scholar]
- 237.Masembe RN. The pattern of bacterial diarrhea of the newborn in Mulago Hospital (Kampala) J Trop Pediatr. 1977;23:61. doi: 10.1093/tropej/23.2.61. [DOI] [PubMed] [Google Scholar]
- 238.Boris M, Thomason BM, Hines VD. A community epidemic of enteropathogenic Escherichia coli O126:B16:NM gastroenteritis associated with asymptomatic respiratory infection. Pediatrics. 1964;33:18. [PubMed] [Google Scholar]
- 239.Kessner DM, Shaughnessy HJ, Googins J. An extensive community outbreak of diarrhea due to enteropathogenic Escherichia coli O111:B4. I. Epidemiologic studies. Am J Hyg. 1962;76:27. doi: 10.1093/oxfordjournals.aje.a120262. [DOI] [PubMed] [Google Scholar]
- 240.Severs D, Fardy P, Acres S. Epidemic gastroenteritis in Newfoundland during 1963 associated with E. coli O111:B4. Can Med Assoc J. 1966;94:373. [PMC free article] [PubMed] [Google Scholar]
- 241.Cooper ML, Keller HM, Walters EW. Comparative frequency of detection of enteropathogenic E. coli, Salmonella and Shigella in rectal swab cultures from infants and young children. Pediatrics. 1957;19:411. [PubMed] [Google Scholar]
- 242.Hinton NA, MacGregor RR. A study of infections due to pathogenic serogroups of Escherichia coli. Can Med Assoc J. 1958;79:359. [PMC free article] [PubMed] [Google Scholar]
- 243.Hutchinson RI. Escherichia coli (O-types 111, 55, and 26) and their association with infantile diarrhea. A five-year study. J Hyg. 1957;55:27. doi: 10.1017/s0022172400061246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Joe LK, Sahab K, Yauw GS. Diarrhea among infants and children in Djakarta, Indonesia, with special reference to pathogenic Escherichia coli. Am J Trop Med Hyg. 1960;9:626. doi: 10.4269/ajtmh.1960.9.626. [DOI] [PubMed] [Google Scholar]
- 245.Nelson JD. Duration of neomycin therapy for enteropathogenic Escherichia coli diarrheal disease: a comparative study of 113 cases. Pediatrics. 1971;48:248. [PubMed] [Google Scholar]
- 246.Riley HD., Jr Antibiotic therapy in neonatal enteric disease. Ann N Y Acad Sci. 1971;176:360. [Google Scholar]
- 247.Rozansky R, Berant M, Rosenmann E. Enteropathogenic Escherichia coli infections in infants during the period from 1957 to 1962. Pediatrics. 1964;64:521. doi: 10.1016/s0022-3476(64)80341-3. [DOI] [PubMed] [Google Scholar]
- 248.South MA. Enteropathogenic Escherichia coli disease: new developments and perspectives. J Pediatr. 1971;79:1. doi: 10.1016/s0022-3476(71)80051-3. [DOI] [PubMed] [Google Scholar]
- 249.Linzenmeier G. Wandel im Auftreten und Verhalten enteropathogene Colitypen. Z Bakteriol. 1962;184:74. [Google Scholar]
- 250.Nicolopoulos D, Arseni A. Susceptibility of enteropathogenic E. coli to various antibiotics. Letter to the editor. J Pediatr. 1972;81:426. doi: 10.1016/s0022-3476(72)80359-7. [DOI] [PubMed] [Google Scholar]
- 251.Ironside AG, Tuxford AF, Heyworth B. A survey of infantile gastroenteritis. BMJ. 1970;2:20. doi: 10.1136/bmj.3.5713.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Riley HD., Jr Clinical rounds. Enteropathogenic E. coli gastroenteritis. Clin Pediatr. 1964;3:93. doi: 10.1177/000992286400300208. [DOI] [PubMed] [Google Scholar]
- 253.Kourany M, Vasquez MA. Enteropathogenic bacteria associated with diarrhea among infants in Panama. Am J Trop Med Hyg. 1969;18:930. doi: 10.4269/ajtmh.1969.18.930. [DOI] [PubMed] [Google Scholar]
- 254.Gaines S, Achavasmith U, Thareesawat M. Types and distribution of enteropathogenic Escherichia coli in Bangkok, Thailand. Am J Hyg. 1964;80:388. doi: 10.1093/oxfordjournals.aje.a120488. [DOI] [PubMed] [Google Scholar]
- 255.Buttner DW, Lado-Kenyi A. Prevalence of Salmonella, Shigella, and enteropathogenic Escherichia coli in young children in Kampala, Uganda. Tropenmed Parasitol. 1973;24:259. [PubMed] [Google Scholar]
- 256.Coetzee M, Leary PM. Gentamicin in Esch. coli gastroenteritis. Arch Dis Child. 1971;46:646. doi: 10.1136/adc.46.249.646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Kahn E. The aetiology of summer diarrhoea. S Afr Med J. 1957;31:47. [PubMed] [Google Scholar]
- 258.Taylor J. The diarrhoeal diseases in England and Wales. With special reference to those caused by Salmonella, Escherichia, and Shigella. Bull World Health Organ. 1960;23:763. [PMC free article] [PubMed] [Google Scholar]
- 259.Epidemiological Research Laboratory of the Public Health Laboratory Service, United Kingdom and Republic of Ireland E. coli gastroenteritis from food. BMJ. 1976;1:911. [Google Scholar]
- 260.Ocklitz HW, Schmidt E. F. Über das Vorkommen von Dispepsie-Coli bei Erwachsenen. Helv Paediatr Acta. 1955;10:450. [PubMed] [Google Scholar]
- 261.Schaffer J, Lewis V, Nelson J. Antepartum survey for enteropathogenic Escherichia coli. Detection by cultural and fluorescent antibody methods. Am J Dis Child. 1963;106:170. doi: 10.1001/archpedi.1963.02080050172009. [DOI] [PubMed] [Google Scholar]
- 262.Kirby AC, Hall EG, Coackley W. Neonatal diarrhoea and vomiting. Outbreaks in the same maternity unit. Lancet. 1950;2:201. doi: 10.1016/s0140-6736(50)91189-5. [DOI] [PubMed] [Google Scholar]
- 263.Bettelheim KA, Breaden A, Faiers MC. The origin of O-serotypes of Escherichia coli in babies after normal delivery. J Hyg. 1974;72:67. doi: 10.1017/s0022172400023226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Stulberg CS, Zuelzer WW, Nolke AC. An epidemic of diarrhea of the newborn caused by Escherichia coli O111:B4. Pediatrics. 1954;14:133. [PubMed] [Google Scholar]
- 265.Farmer K, Hassall IB. An epidemic of E. coli type O55:K59(B5) in a neonatal unit. N Z Med J. 1973;77:372. [PubMed] [Google Scholar]
- 266.Hugh-Jones K, Ross GIM. Epidemics of gastroenteritis associated with E. coli O119 infection. Arch Dis Child. 1958;33:543. doi: 10.1136/adc.33.172.543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Senerwa D, Olsvik O, Mutanda LN. Colonization of neonates in a nursery ward with enteropathogenic Escherichia coli and correlation to the clinical histories of the children. J Clin Microbiol. 1989;27:2539. doi: 10.1128/jcm.27.11.2539-2543.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Wright J, Roden AT. Escherichia coli O55B5 infection in a gastroenteritis ward. Epidemiological applications of H antigen type determinations. Am J Hyg. 1953;58:133. doi: 10.1093/oxfordjournals.aje.a119595. [DOI] [PubMed] [Google Scholar]
- 269.Balassanian N, Wolinsky E. Epidemiologic and serologic studies of E. coli O4:115 in a premature nursery. Pediatrics. 1968;41:463. [PubMed] [Google Scholar]
- 270.Jameson JE, Mann TP, Rothfield NJ. Hospital gastroenteritis. An epidemiological survey of infantile diarrhea and vomiting contracted in a children’s hospital. Lancet. 1954;2:459. doi: 10.1016/s0140-6736(54)91876-0. [DOI] [PubMed] [Google Scholar]
- 271.Thomson S. The role of certain varieties of Bacterium coli in gastroenteritis in babies. J Hyg. 1955;53:357. doi: 10.1017/s002217240000084x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Page RH, Stulberg CS. Immunofluorescence in epidemiologic control of E. coli diarrhea. Incidence, cross-infections, and control in a children’s hospital. Am J Dis Child. 1962;104:149. doi: 10.1001/archpedi.1962.02080030151007. [DOI] [PubMed] [Google Scholar]
- 273.Bertrams J, Pfortner M, Neusel H. Colienteritis des Säuglings. Quantitative und fluoreszenzserologische Verlaufsuntersuchungen. Munch Med Wochenschr. 1970;112:38. [PubMed] [Google Scholar]
- 274.Thomson S. The numbers of pathogenic bacilli in faeces in intestinal diseases. J Hyg. 1955;53:217. doi: 10.1017/s0022172400000681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Herweg JC, Middlekamp JN, Thornton HK. Escherichia coli diarrhea. The relationship of certain serotypes of Escherichia coli to sporadic and epidemic cases of infantile diarrhea. J Pediatr. 1956;49:629. doi: 10.1016/s0022-3476(56)80151-0. [DOI] [PubMed] [Google Scholar]
- 276.Belnap WD, O’Donnell JJ. Epidemic gastroenteritis due to Escherichia coli 0–111. A review of the literature, with the epidemiology, bacteriology, and clinical findings of a large outbreak. J Pediatr. 1955;47:178. doi: 10.1016/s0022-3476(55)80029-7. [DOI] [PubMed] [Google Scholar]
- 277.Rogers KB, Koegler SJ. Inter-hospital cross-infection epidemic infantile gastroenteritis associated with type strains of Bacterium coli. J Hyg. 1951;49:152. doi: 10.1017/s0022172400044041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Stock AH, Shuman ME. Gastroenteritis in infants associated with specific serotypes of Escherichia coli. II. An epidemic of Escherichia coli O111:B4 gastroenteritis involving multiple institutions. Pediatrics. 1956;17:196. [PubMed] [Google Scholar]
- 279.Stulberg CS, Zuelzer WW, Nolke AC. Escherichia coli O127:B8, a pathogenic strain causing infantile diarrhea. Epidemiology and bacteriology of a prolonged outbreak in a premature nursery. Am J Dis Child. 1955;90:125. doi: 10.1001/archpedi.1955.04030010127001. [DOI] [PubMed] [Google Scholar]
- 280.Thomson S, Watkins AG, Grapy PO. Escherichia coli gastroenteritis. Arch Dis Child. 1956;31:340. doi: 10.1136/adc.31.159.340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Olarte J, Ramos-Alvarez M. Epidemic diarrhea in premature infants. Etiological significance of a newly recognized type of Escherichia coli (O142:K86:H6) Am J Dis Child. 1965;109:436. [PubMed] [Google Scholar]
- 282.Nelson JD, Whitaker JA, Hempstead B. Epidemiological application of the fluorescent antibody technique. Study of a diarrhea outbreak in a premature nursery. JAMA. 1961;176:26. doi: 10.1001/jama.1961.03040140028008. [DOI] [PubMed] [Google Scholar]
- 283.Buttiaux R, Nicolle P, LeMinor S. Etude épidémiologique des gastroentéritis à Escherichia coli dans un service hospitalier du nord de la France. Arch Mal Appar Dig Mal Nutr. 1956;45:225. [PubMed] [Google Scholar]
- 284.Harris AH, Yankauer A, Green DC. Control of epidemic diarrhea of the newborn in hospital nurseries and pediatric wards. Ann N Y Acad Sci. 1956;66:118. doi: 10.1111/j.1749-6632.1956.tb40110.x. [DOI] [PubMed] [Google Scholar]
- 285.Jacobs SI, Holzel A, Wolman B. Outbreak of infantile gastroenteritis caused by Escherichia coli O114. Arch Dis Child. 1970;46:656. doi: 10.1136/adc.45.243.656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Curtin M, Clifford SH. Incidence of pathogenic serologic types of Escherichia coli among neonatal patients in the New England area. N Engl J Med. 1956;255:1090. doi: 10.1056/NEJM195612062552308. [DOI] [PubMed] [Google Scholar]
- 287.Greene DC, Albrecht RM. Recent developments in diarrhea of the newborn. N Y State J Med. 1955;55:2764. [PubMed] [Google Scholar]
- 288.Wheeler WE. Spread and control of Escherichia coli diarrheal disease. Ann N Y Acad Sci. 1956;66:112. doi: 10.1111/j.1749-6632.1956.tb40109.x. [DOI] [PubMed] [Google Scholar]
- 289.Buttiaux RR, Nicolle P, LeMinor L. Etudes sur les E. coli de gastroentérite infantile. Ann Inst Pasteur. 1956;91:799. [PubMed] [Google Scholar]
- 290.Love WC, Gordon AM, Gross RJ. Infantile gastroenteritis due to Escherichia coli O142. Lancet. 1972;2:355. doi: 10.1016/s0140-6736(72)91738-2. [DOI] [PubMed] [Google Scholar]
- 291.Shaughnessy HJ, Lesko M, Dorigan F. An extensive community outbreak of diarrhea due to enteropathogenic Escherichia coli O111:B4. Am J Hyg. 1962;76:44. [PubMed] [Google Scholar]
- 292.Kendall N, Vaughan VC, III, Kusakcioglu A. A study of preparation of infant formulas. A medical and sociocultural appraisal. Am J Dis Child. 1971;122:215. doi: 10.1001/archpedi.1971.02110030071006. [DOI] [PubMed] [Google Scholar]
- 293.Gamble DR, Rowson KEK. The incidence of pathogenic Escherichia coli in routine fecal specimens. Lancet. 1957;2:619. doi: 10.1016/s0140-6736(57)91946-3. [DOI] [PubMed] [Google Scholar]
- 294.Taylor J, Powell BW, Wright J. Infantile diarrhoea and vomiting. A clinical and bacteriological investigation. BMJ. 1949;2:117. doi: 10.1136/bmj.2.4619.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Modica RI, Ferguson WW, Ducey EF. Epidemic infantile diarrhea associated with Escherichia coli O111, B4. J Lab Clin Med. 1952;39:122. [PubMed] [Google Scholar]
- 296.Rogers KB. The spread of infantile gastroenteritis in a cubicled ward. J Hyg. 1951;49:140. doi: 10.1017/s002217240004403x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Mossel DAA, Weijers HA. Uitkomsten, verkregen bij bacteriologisch onderzoek van vrouwenmelk van diverse herkomst en de betekenis daarvan de pediatrische praktijk. Maandschr Kindergeneeskd. 1957;25:37. [PubMed] [Google Scholar]
- 298.Rantasalo I, Kauppinen MA. The occurrence of Staphylococcus aureus in mother’s milk. Ann Chir Gynaecol. 1959;48:246. [PubMed] [Google Scholar]
- 299.Edwards LD, Tan-Gatue LG, Levin S. The problem of bacteriologically contaminated infant formulas in a newborn nursery. Clin Pediatr. 1974;13:63. doi: 10.1177/000992287401300109. [DOI] [PubMed] [Google Scholar]
- 300.Thomson S. Is infantile gastroenteritis fundamentally a milk-borne infection? J Hyg. 1956;54:311. doi: 10.1017/s0022172400044545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Danielssen D, Laurell G. Fluorescent antibody technique in the diagnosis of enteropathogenic Escherichia coli, with special reference to sensitivity and specificity. Acta Pathol Microbiol Scand. 1969;76:601. doi: 10.1111/j.1699-0463.1969.tb03291.x. [DOI] [PubMed] [Google Scholar]
- 302.Bullen CL, Willis AT. Resistance of the breast-fed infant to gastroenteritis. BMJ. 1971;2:338. doi: 10.1136/bmj.3.5770.338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Svirsky-Gross S. Pathogenic strains of coli (0,111) among prematures and the use of human milk in controlling the outbreak of diarrhea. Ann Paediatr. 1958;190:109. [PubMed] [Google Scholar]
- 304.Tassovatz B, Kotsitch A. Le lait de femme et son action de protection contre les infections intestinales chez le nouveau-né. Ann Paediatr. 1961;8:285. [Google Scholar]
- 305.Adam A. Fortschritte in der Pathogenese und Therapie der Ernährungsstörungen. Arztl Forschung. 1952;6:59. [Google Scholar]
- 306.Neter E, Shumway CN. E. coli serotype D433; occurrence in intestinal and respiratory tracts, cultural characteristics, pathogenicity, sensitivity to antibiotics. Proc Soc Exp Biol Med. 1950;75:504. doi: 10.3181/00379727-75-18246. [DOI] [PubMed] [Google Scholar]
- 307.Arnon H, Salzberger M, Olitzki AL. The appearance of antibacterial and antitoxic antibodies in maternal sera, umbilical cord blood and milk: observations on the specificity of antibacterial antibodies in human sera. Pediatrics. 1959;23:86. [PubMed] [Google Scholar]
- 308.Kenny JF, Boesman MI, Michaels RH. Bacterial and viral coproantibodies in breast-fed infants. Pediatrics. 1967;39:202. [PubMed] [Google Scholar]
- 309.Sussman S. The passive transfer of antibodies to Escherichia coli O111:B4 from mother to offspring. Pediatrics. 1961;27:308. [Google Scholar]
- 310.Stulberg CS, Zuelzer WW. Infantile diarrhea due to Escherichia coli. Ann N Y Acad Sci. 1956;66:90. doi: 10.1111/j.1749-6632.1956.tb40106.x. [DOI] [PubMed] [Google Scholar]
- 311.Yeivin R, Salzberger M, Olitzki AL. Development of antibodies to enteric pathogens: placental transfer of antibodies and development of immunity in childhood. Pediatrics. 1956;18:19. [PubMed] [Google Scholar]
- 312.Dancis J, Kunz HW. Studies of the immunology of the newborn infant. VI. Bacteriostatic and complement activity of the serum. Pediatrics. 1954;13:339. [PubMed] [Google Scholar]
- 313.Kenny JF, Weinert DW, Gray JA. Enteric infection with Escherichia coli O127 in the mouse. II. Failure of specific immunity to alter intestinal colonization of infants and adults. J Infect Dis. 1974;129:10. doi: 10.1093/infdis/129.1.10. [DOI] [PubMed] [Google Scholar]
- 314.Lodinova R, Jouja V, Wagner V. Serum immunoglobulins and coproantibody formation in infants after artificial intestinal colonization with Escherichia coli O83 and oral lysozyme administration. Pediatr Res. 1973;7:659. doi: 10.1203/00006450-197307000-00009. [DOI] [PubMed] [Google Scholar]
- 315.McNeish AS, Gaze H. The intestinal antibody response in infants with enteropathic E. coli gastroenteritis. Acta Paediatr Scand. 1974;63:663. [Google Scholar]
- 316.Goldschmidt MC, DuPont HL. Enteropathogenic Escherichia coli: lack of correlation of serotype with pathogenicity. J Infect Dis. 1976;133:153. doi: 10.1093/infdis/133.2.153. [DOI] [PubMed] [Google Scholar]
- 317.Echeverria PD, Chang CP, Smith D. Enterotoxigenicity and invasive capacity of “enteropathogenic” serotypes of Escherichia coli. J Pediatr. 1976;89:8. doi: 10.1016/s0022-3476(76)80917-1. [DOI] [PubMed] [Google Scholar]
- 318.Gross RJ, Scotland SM, Rowe B. Enterotoxin testing of Escherichia coli causing epidemic infantile enteritis in the U.K. Lancet. 1976;1:629. doi: 10.1016/s0140-6736(76)90429-3. [DOI] [PubMed] [Google Scholar]
- 319.Drucker MM, Pollack A, Yeivin R. Immunofluorescent demonstration of enteropathogenic Escherichia coli in tissues of infants dying with enteritis. Pediatrics. 1970;46:855. [PubMed] [Google Scholar]
- 320.Levine MM, Bergquist EJ, Nalin DR. Escherichia coli strains that cause diarrhea but do not produce heat-labile or heat-stable enterotoxins and are non-invasive. Lancet. 1978;1:1119. doi: 10.1016/s0140-6736(78)90299-4. [DOI] [PubMed] [Google Scholar]
- 321.Wade WG, Thom BT, Evans N. Cytotoxic enteropathogenic Escherichia coli. Lancet. 1979;2:1235. doi: 10.1016/s0140-6736(79)92349-3. [DOI] [PubMed] [Google Scholar]
- 322.Mellies JL, Navarro-Garcia F, Okeke I. EspC pathogenicity island of Enteropathogenic Escherichia coli encodes an enterotoxin. Infect Immun. 2001;69:315. doi: 10.1128/IAI.69.1.315-324.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Vallance BA, Chan C, Robertson ML, Finlay BB. Enteropathogenic and enterohemorrhagic Escherichia coli infections: emerging themes in pathogenesis and prevention. Can J Gastroenterol. 2002;16:771. doi: 10.1155/2002/410980. [DOI] [PubMed] [Google Scholar]
- 324.Rothbaum RJ, Partin JC, McAdams AJ. Enterocyte adherent E. coli O119:B14: a novel mechanism of infant diarrhea. Gastroenterology. 1981;80:1265. [Google Scholar]
- 325.Polotsky Y, Dragunskaya EM, Seliverstova VG. Pathogenic effect of enterotoxigenic Escherichia coli and Escherichia coli causing infantile diarrhea. Acta Microbiol Acad Sci Hung. 1977;24:221. [PubMed] [Google Scholar]
- 326.McDaniel TK, Jarvis KG, Donnenberg MS, Kaper JB. A genetic locus of enterocyte effacement conserved among diverse enterobacterial pathogens. Proc Natl Acad Sci U S A. 1995;92:1664. doi: 10.1073/pnas.92.5.1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327.McDaniel TK, Kaper JB. A cloned pathogenicity island from enteropathogenic Escherichia coli confers the attaching and effacing phenotype on E. coli K-12. Mol Microbiol. 1997;23:399. doi: 10.1046/j.1365-2958.1997.2311591.x. [DOI] [PubMed] [Google Scholar]
- 328.Elliott SJ, Wainwright LA, McDaniel TK. The complete sequence of the locus of enterocyte effacement (LEE) from enteropathogenic Escherichia coli E2348/69. Mol Microbiol. 1998;28:1. doi: 10.1046/j.1365-2958.1998.00783.x. [DOI] [PubMed] [Google Scholar]
- 329.Ghaem-Maghami M, Simmons CP, Daniell S. Intimin-specific immune responses prevent bacterial colonization by the attaching-effacing pathogen Citrobacter rodentium. Infect Immun. 2001;69:5597. doi: 10.1128/IAI.69.9.5597-5605.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Giron JA, Ho ASY, Schoolnik GK. An inducible bundle-forming pilus of enteropathogenic Escherichia coli. Science. 1992;254:710. doi: 10.1126/science.1683004. [DOI] [PubMed] [Google Scholar]
- 331.Stone KD, Zhang HZ, Carlson LK, Donnenberg MS. A cluster of fourteen genes from enteropathogenic Escherichia coli is sufficient for the biogenesis of a type IV pilus. Mol Microbiol. 1996;20:325. doi: 10.1111/j.1365-2958.1996.tb02620.x. [DOI] [PubMed] [Google Scholar]
- 332.Knutton S, Shaw RK, Anantha RP. The type IV bundle-forming pilus of enteropathogenic Escherichia coli undergoes dramatic alterations in structure associated with bacterial adherence, aggregation and dispersal. Mol Microbiol. 1999;33:499. doi: 10.1046/j.1365-2958.1999.01495.x. [DOI] [PubMed] [Google Scholar]
- 333.Bieber D, Ramer SW, Wu CY. Type IV pili, transient bacterial aggregates, and virulence of enteropathogenic Escherichia coli. Science. 1998;280:2114. doi: 10.1126/science.280.5372.2114. [DOI] [PubMed] [Google Scholar]
- 334.Hopkins GB, Gould VE, Stevenson JK. Necrotizing enterocolitis in premature infants. A clinical and pathologic evaluation of autopsy material. Am J Dis Child. 1970;120:229. doi: 10.1001/archpedi.1970.02100080113009. [DOI] [PubMed] [Google Scholar]
- 335.Rho Y, Josephson JE. Epidemic enteropathogenic Escherichia coli. Newfoundland, 1963: autopsy study of 16 cases. Can Med Assoc J. 1967;96:392. [PMC free article] [PubMed] [Google Scholar]
- 336.Shwachman H, Lloyd-Still JD, Khaw KT. Protracted diarrhea of infancy treated by intravenous alimentation. II. Studies of small intestinal biopsy results. Am J Dis Child. 1973;125:365. doi: 10.1001/archpedi.1973.04160030039007. [DOI] [PubMed] [Google Scholar]
- 337.Lucking T, Gruttner R. Chronic diarrhea and severe malabsorption in infancy following infections with pathogenic E. coli. Acta Paediatr Scand. 1974;63:167. [Google Scholar]
- 338.Handforth CP, Sorger K. Failure of regeneration of small bowel mucosa following epidemic infantile gastroenteritis. Can Med Assoc J. 1961;84:425. [PMC free article] [PubMed] [Google Scholar]
- 339.McKay DG, Wahle GH., Jr Epidemic gastroenteritis due to Escherichia coli O111:B4. II. Pathologic anatomy (with special reference to the presence of the local and generalized Shwartzman phenomena) Arch Pathol. 1955;60:679. [PubMed] [Google Scholar]
- 340.Laurell G. Quoted in Ordway NK. Diarrhoeal disease and its control. Bull World Health Organ. 1960;23:93. [PMC free article] [PubMed] [Google Scholar]
- 341.Braun OH, Henckel H. Über epidemische Säuglingsenteritis. Z Kinderheilkd. 1951;70:33. [PubMed] [Google Scholar]
- 342.Neter E. Enteritis due to enteropathogenic Escherichia coli. Present day status and unsolved problems. J Pediatr. 1959;55:223. doi: 10.1016/s0022-3476(59)80091-3. [DOI] [PubMed] [Google Scholar]
- 343.Rogers KB, Cracknell VM. Epidemic infantile gastroenteritis due to Escherichia coli type 0,114. J Pathol Bacteriol. 1956;72:27. [PubMed] [Google Scholar]
- 344.Todd RM, Hall EG. Chloramphenicol in prophylaxis of infantile gastroenteritis. BMJ. 1953;1:1359. doi: 10.1136/bmj.1.4824.1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345.Gastroenteritis due to Escherichia coli. Editorial. Lancet. 1968;1:32. [PubMed] [Google Scholar]
- 346.Senerwa D, Olsvik O, Mutanda LN. Enteropathogenic Escherichia coli serotype O111:HNT isolated from preterm neonates in Nairobi, Kenya. J Clin Microbiol. 1989;27:1307. doi: 10.1128/jcm.27.6.1307-1311.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347.Bray J. Bray’s discovery of pathogenic Esch. coli as a cause of infantile gastroenteritis. Arch Dis Child. 1973;48:923. doi: 10.1136/adc.48.12.923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348.Ironside AG, Brennand J, Mandal BK. Cross-infection in infantile gastroenteritis. Arch Dis Child. 1971;46:815. doi: 10.1136/adc.46.250.815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 349.Linetskaya-Novgorodskaya EM. Acute intestinal infections of non-dysenteric etiology. Bull World Health Organ. 1959;21:299. [PMC free article] [PubMed] [Google Scholar]
- 350.Lloyd-Still JD, Shwachman H, Filler RM. Protracted diarrhea of infancy treated by intravenous alimentation. 1. Clinical studies of 16 infants. Am J Dis Child. 1973;125:358. doi: 10.1001/archpedi.1973.04160030032006. [DOI] [PubMed] [Google Scholar]
- 351.Drimmer-Hernheiser H, Olitzki AL. The association of Escherichia coli (serotypes O111:B4 and O55:B5) with cases of acute infantile gastroenteritis in Jerusalem. Acta Med Orient. 1951;10:219. [PubMed] [Google Scholar]
- 352.Linde K, Kodizt H, Funk G. Die Mehrfachinfektionen mit Dyspepsie-Coli, ihre Beurteilung in statistischer, bakteriologischer und klinischer Sicht. Z Hyg. 1960;147:94. [Google Scholar]
- 353.Fandre M, Coffin R, Dropsy G. Epidemic of infantile gastroenteritis due to Escherichia coli O127:B8 with methemoglobinemic cyanosis. Arch Fr Pediatr. 1962;19:1129. [Google Scholar]
- 354.Garcia de Olarte D, Trujillo H, Agudelo ON. Treatment of diarrhea in malnourished infants and children. A double-blind study comparing ampicillin and placebo. Am J Dis Child. 1974;127:379. doi: 10.1001/archpedi.1974.02110220077011. [DOI] [PubMed] [Google Scholar]
- 355.Yow MD. Centers for Disease Control Proceedings of the International Conference on Nosocomial Infections, August 3–6, 1970. American Hospital Association; Chicago: 1971. Prophylactic antimicrobial agents-panel. Statement of panelist; pp. 315–316. [Google Scholar]
- 356.Bettelheim KA, Faiers M, Sheeter RA. Serotypes of Escherichia coli in normal stools. Lancet. 1972;2:1227. [PubMed] [Google Scholar]
- 357.Stock AH, Shuman ME. Gastroenteritis in infants associated with specific serotypes of Escherichia coli. I. Incidence of specific Escherichia coli serotypes O111:B4 and O55:B5 in the Pittsburgh area. Pediatrics. 1956;17:192. [PubMed] [Google Scholar]
- 358.Mushin R. Multiple intestinal infection. Med J Aust. 1953;1:807. doi: 10.5694/j.1326-5377.1953.tb84931.x. [DOI] [PubMed] [Google Scholar]
- 359.Harris JC, DuPont HL, Hornick RB. Fecal leukocytes in diarrheal illness. Ann Intern Med. 1972;76:697. doi: 10.7326/0003-4819-76-5-697. [DOI] [PubMed] [Google Scholar]
- 360.Guerrant RL, Araujo V, Cooper WH. Measurement of fecal lactoferrin as a marker of fecal leukocytes and inflammatory enteritis. J Clin Microbiol. 1992;30:1238. doi: 10.1128/jcm.30.5.1238-1242.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 361.Miller JR, Barrett LJ, Kotloff K, Guerrant RL. A rapid test for infectious and inflammatory enteritis. Arch Intern Med. 1994;154:2660. doi: 10.1001/archinte.1994.00420230043006. [DOI] [PubMed] [Google Scholar]
- 362.Cherry WB, Thomason BM. Fluorescent antibody techniques for Salmonella and other enteric pathogens. Public Health Rep. 1969;84:887. [PMC free article] [PubMed] [Google Scholar]
- 363.Murray WA, Kheder J, Wheeler WE. Colistin suppression of Escherichia coli in stools. I. Control of a nosocomial outbreak of diarrhea caused by neomycin-resistant Escherichia coli O111:B4. Am J Dis Child. 1964;108:274. doi: 10.1001/archpedi.1964.02090010276009. [DOI] [PubMed] [Google Scholar]
- 364.Bokete TN, Whittam TS, Wilson RA. Genetic and phenotypic analysis of Escherichia coli with enteropathogenic characteristics isolated from Seattle children. J Infect Dis. 1997;175:1382. doi: 10.1086/516470. [DOI] [PubMed] [Google Scholar]
- 365.Vial PA, Mathewson JJ, DuPont HL, Guers L, Levine MM. Comparison of two assay methods for patterns of adherence to HEp-2 cells of Escherichia coli from patients with diarrhea. J Clin Microbiol. 1990;28:882. doi: 10.1128/jcm.28.5.882-885.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366.Albert MJ, Ansaruzzaman M, Faruque SM. An ELISA for the detection of localized adherent classic enteropathogenic Escherichia coli serogroups. J Infect Dis. 1991;164:986. doi: 10.1093/infdis/164.5.986. [DOI] [PubMed] [Google Scholar]
- 367.Knutton S, Baldwin T, Williams PH. Actin accumulation at sites of bacterial adhesion to tissue culture cells: basis of a new diagnostic test for enteropathogenic and enterohemorrhagic Escherichia coli. Infect Immun. 1989;57:1290. doi: 10.1128/iai.57.4.1290-1298.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 368.Nelson JD. Comment. In: Gellis S, editor. Yearbook of Pediatrics, 1973. Mosby–Year Book; St Louis: 1973. [Google Scholar]
- 369.Sherman JO, Hamly CA, Khachadurian AK. Use of an oral elemental diet in infants with severe intractable diarrhea. J Pediatr. 1975;86:518. doi: 10.1016/s0022-3476(75)80140-5. [DOI] [PubMed] [Google Scholar]
- 370.Pearce JL, Hamilton JR. Controlled trial of orally administered lactobacilli in acute infantile diarrhea. J Pediatr. 1974;84:261. doi: 10.1016/s0022-3476(74)80618-9. [DOI] [PubMed] [Google Scholar]
- 371.Marie J, Hennequet A, Roux C. La kanamycin “per os” dans le traitement des gastroentérites à colibacilles du nourrison. Ann Paediatr. 1962;9:97. [PubMed] [Google Scholar]
- 372.Valman HB, Wilmers MJ. Use of antibiotics in acute gastroenteritis among infants in hospital. Lancet. 1969;1:1122. doi: 10.1016/s0140-6736(69)91643-2. [DOI] [PubMed] [Google Scholar]
- 373.Dailey KM, Sturtevant AB, Feary TW. Incidence of antibiotic resistance and R-factors among gram-negative bacteria isolated from the neonatal intestine. J Pediatr. 1972;80:198. doi: 10.1016/s0022-3476(72)80578-x. [DOI] [PubMed] [Google Scholar]
- 374.Neu HC, Cherubin C, Vogt M. Antibiotic resistance of fecal Escherichia coli. A comparison of samples from children of low and high socioeconomic groups. Am J Dis Child. 1973;126:174. doi: 10.1001/archpedi.1973.02110190152008. [DOI] [PubMed] [Google Scholar]
- 375.Watanabe T. Transferable antibiotic resistance in Enterobacteriaceae: relationship to the problems of treatment and control of coliform enteritis. Ann N Y Acad Sci. 1971;176:371. [Google Scholar]
- 376.McCracken GH. Changing pattern of the antimicrobial susceptibilities of Escherichia coli in neonatal infections. J Pediatr. 1971;78:942. doi: 10.1016/s0022-3476(71)80422-5. [DOI] [PubMed] [Google Scholar]
- 377.Kunin CM. Resistance to antimicrobial drugs—a worldwide calamity. Ann Intern Med. 1993;118:559. doi: 10.7326/0003-4819-118-7-199304010-00011. [DOI] [PubMed] [Google Scholar]
- 378.Silver LL, Bostian KA. Discovery and development of new antibiotics: the problem of antibiotic resistance. Antimicrob Agents Chemother. 1993;37:377. doi: 10.1128/aac.37.3.377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 379.Nelson JD. Commentary. J Pediatr. 1976;89:471. [Google Scholar]
- 380.Committee on Fetus and Newborn . Guidelines for Perinatal Care. 3rd ed. American Academy of Pediatrics and American College of Obstetricians and Gynecologists; 1992. [Google Scholar]
- 381.Sprunt K, Redman W, Leidy G. Antibacterial effectiveness of routine handwashing. Pediatrics. 1973;52:264. [PubMed] [Google Scholar]
- 382.Multistate outbreak of Escherichia coli O157:H7 infections from hamburgers. MMWR Morb Mortal Wkly Rep. 1993;42:258. [PubMed] [Google Scholar]
- 383.MacDonald KL, Osterholm MT. The emergence of Escherichia coli O157:H7 infection in the United States. JAMA. 1993;269:2264. [PubMed] [Google Scholar]
- 384.Watanabe H, Wadam A, Inagaki Y. Outbreaks of enterohemorrhagic Escherichia coli O157:H7 infection by two different genotype strains in Japan. Lancet. 1996;348:831. doi: 10.1016/s0140-6736(05)65257-9. [DOI] [PubMed] [Google Scholar]
- 385.Slutsker L, Ries AA, Maloney K. A nationwide case-control study of Escherichia coli O157:H7 infection in the United States. J Infect Dis. 1998;177:962. doi: 10.1086/515258. [DOI] [PubMed] [Google Scholar]
- 386.Rasmussen MA, Casey TA. Environmental and food safety aspects of Escherichia coli O157:H7 infections in cattle. Crit Rev Microbiol. 2001;27:57. doi: 10.1080/20014091096701. [DOI] [PubMed] [Google Scholar]
- 387.Ochoa TJ, Cleary TG. Epidemiology and spectrum of disease of Escherichia coli O157. Curr Opin Infect Dis. 2003;16:259. doi: 10.1097/00001432-200306000-00013. [DOI] [PubMed] [Google Scholar]
- 388.Hemolytic-uremic syndrome associated with Escherichia coli O157:H7 enteric infections—United States. MMWR Morb Mortal Wkly Rep. 1985;34:20. [PubMed] [Google Scholar]
- 389.Belongia EA, Osterholm MT, Soler JT. Transmission of Escherichia coli O157:H7 infection in Minnesota child day-care facilities. JAMA. 1993;269:883. [PubMed] [Google Scholar]
- 390.Besser RE, Lett SM, Weber JT. An outbreak of diarrhea and hemolytic uremic syndrome from Escherichia coli O157:H7 in fresh-pressed apple cider. JAMA. 1993;269:2217. [PubMed] [Google Scholar]
- 391.Mobasellah M, Donohue-Rolfe A, Jacewicz M. Pathogenesis of Shigella diarrhea: evidence for a developmentally regulated glycolipid receptor for Shigatoxin involved in the fluid secretory response of rabbit small intestine. J Infect Dis. 1988;157:1023. doi: 10.1093/infdis/157.5.1023. [DOI] [PubMed] [Google Scholar]
- 392.Sandvig K. Shiga toxins. Toxicon. 2001;39:1629. doi: 10.1016/s0041-0101(01)00150-7. [DOI] [PubMed] [Google Scholar]
- 393.Ray PE, Liu XH. Pathogenesis of Shiga toxin–induced hemolytic uremic syndrome. Pediatr Nephrol. 2001;16:823. doi: 10.1007/s004670100660. [DOI] [PubMed] [Google Scholar]
- 394.Schmidt H. Shiga-toxin-converting bacteriophages. Res Microbiol. 2001;152:687. doi: 10.1016/s0923-2508(01)01249-9. [DOI] [PubMed] [Google Scholar]
- 395.Scotland SM, Smith HR, Rowe B. Two distinct toxins active on Vero cells from Escherichia coli O157. Lancet. 1985;2:885. doi: 10.1016/s0140-6736(85)90146-1. [DOI] [PubMed] [Google Scholar]
- 396.Karmali MA, Petric M, Louie S. Antigenic heterogeneity of Escherichia coli verotoxins. Lancet. 1986;1:164. doi: 10.1016/s0140-6736(86)92307-x. [DOI] [PubMed] [Google Scholar]
- 397.Karch H, Heeseman J, Laufs R. A plasmid of enterohemorrhagic Escherichia coli O157:H7 is required for expression of a new fimbrial antigen and for adhesion to epithelial cells. Infect Immun. 1987;55:455. doi: 10.1128/iai.55.2.455-461.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 398.Levine MM, Jian-guo XU, Kaper JB. A DNA probe to identify enterohemorrhagic Escherichia coli of O157:H7 and other serotypes that cause hemorrhagic colitis and hemolytic uremic syndrome. J Infect Dis. 1987;156:175. doi: 10.1093/infdis/156.1.175. [DOI] [PubMed] [Google Scholar]
- 399.Paton JC, Paton AW. Methods for detection of STEC in humans. An overview. Methods Mol Med. 2003;73:9. doi: 10.1385/1-59259-316-x:09. [DOI] [PubMed] [Google Scholar]
- 400.Klein EJ, Stapp JR, Clausen CR. Shiga toxin–producing Escherichia coli in children with diarrhea: a prospective point-of-care study. J Pediatr. 2002;141:172. doi: 10.1067/mpd.2002.125908. [DOI] [PubMed] [Google Scholar]
- 401.Tarr PI. Escherichia coli O157:H7: clinical, diagnostic, and epidemiological aspects of human infection. Clin Infect Dis. 1995;20:1. doi: 10.1093/clinids/20.1.1. [DOI] [PubMed] [Google Scholar]
- 402.Wong CS, Jelacic S, Habeeb RL. The risk of the hemolytic-uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 infections. N Engl J Med. 2000;342:1930. doi: 10.1056/NEJM200006293422601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 403.Safdar N, Said A, Gangnon RE, Maki DG. Risk of hemolytic uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 enteritis: a meta-analysis. JAMA. 2002;288:996. doi: 10.1001/jama.288.8.996. [DOI] [PubMed] [Google Scholar]
- 404.Nataro JP. Enteroaggregative Escherichia coli. In: Hughes J, editor. Emerging Infections 6. American Society for Microbiology Press; Washington, DC: 2003. [Google Scholar]
- 405.Sarantuya J, Nishi J, Wakimoto K, et al. Typical enteroaggregative Escherichia coli are the most prevalent pathotypes causing diarrhea in Mongolian children. J Clin Microbiol. In press. [DOI] [PMC free article] [PubMed]
- 406.Baudry B, Savarino SJ, Vial P. A sensitive and specific DNA probe to identify enteroaggregative Escherichia coli, a recently discovered diarrheal pathogen. J Infect Dis. 1990;161:1249. doi: 10.1093/infdis/161.6.1249. [DOI] [PubMed] [Google Scholar]
- 407.Nishi J, Sheikh J, Mizuguchi K. The export of coat protein from enteroaggregative Escherichia coli by a specific ATP-binding cassette transporter system. J Biol Chem. 2003;278:45680. doi: 10.1074/jbc.M306413200. [DOI] [PubMed] [Google Scholar]
- 408.Mathewson JJ, Johnson PC, DuPont HL. Pathogenicity of enteroadherent Escherichia coli in adult volunteers. J Infect Dis. 1986;154:524. doi: 10.1093/infdis/154.3.524. [DOI] [PubMed] [Google Scholar]
- 409.Nataro JP, Deng Y, Cookson S. Heterogeneity of enteroaggregative Escherichia coli virulence demonstrated in volunteers. J Infect Dis. 1995;17:465. doi: 10.1093/infdis/171.2.465. [DOI] [PubMed] [Google Scholar]
- 410.Itoh Y, Nagano I, Kunishima M, Ezaki T. Laboratory investigation of enteroaggregative Escherichia coli O untypeable:H10 associated with a massive outbreak of gastrointestinal illness. J Clin Microbiol. 1997;35:2546. doi: 10.1128/jcm.35.10.2546-2550.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 411.Smith HR, Cheasty T, Rowe B. Enteroaggregative Escherichia coli and outbreaks of gastroenteritis in UK. Lancet. 1997;350:814. doi: 10.1016/s0140-6736(05)62611-6. [DOI] [PubMed] [Google Scholar]
- 412.Cobeljic M, Miljkovic-Selimovic B, Paunovic-Todosijevic D. Enteroaggregative E. coli associated with an outbreak of diarrhea in a neonatal nursery ward. Epidemiol Infect. 1996;117:11. doi: 10.1017/s0950268800001072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 413.Eslava CE, Villaseca J, Morales R, et al. Identification of a protein with toxigenic activity produced by enteroaggregative Escherichia coli. Abstracts of the General Meeting of the American Society for Microbiology, 1993 (abstract B-105).
- 414.Bhan MK, Raj P, Levine MM. Enteroaggregative Escherichia coli associated with persistent diarrhea in a cohort of rural children in India. J Infect Dis. 1989;159:1061. doi: 10.1093/infdis/159.6.1061. [DOI] [PubMed] [Google Scholar]
- 415.Bhan MK, Khoshoo V, Sommerfelt H. Enteroaggregative Escherichia coli and Salmonella associated with non-dysenteric persistent diarrhea. Pediatr Infect Dis J. 1989;8:499. doi: 10.1097/00006454-198908000-00005. [DOI] [PubMed] [Google Scholar]
- 416.Cravioto A, Tello A, Navarro A. Association of Escherichia coli HEp-2 adherence patterns with type and duration of diarrhoea. Lancet. 1991;337:262. doi: 10.1016/0140-6736(91)90868-p. [DOI] [PubMed] [Google Scholar]
- 417.Lima AAM, Fang G, Schorling JB. Persistent diarrhea in Northeast Brazil: etiologies and interactions with malnutrition. Acta Paediatr Scand. 1992;381:39. doi: 10.1111/j.1651-2227.1992.tb12370.x. [DOI] [PubMed] [Google Scholar]
- 418.Dutta S, Pal S, Chakrabarti S, Dutta P, Manna B. Use of PCR to identify enteroaggregative Escherichia coli as an important cause of acute diarrhoea among children living in Calcutta, India. J Med Microbiol. 1999;48:1011. doi: 10.1099/00222615-48-11-1011. [DOI] [PubMed] [Google Scholar]
- 419.Scaletsky IC, Fabbricotti SH, Silva SO. HEp-2-adherent Escherichia coli strains associated with acute infantile diarrhea, São Paulo, Brazil. Emerg Infect Dis. 2002;8:855. doi: 10.3201/eid0808.010492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420.Gonzalez R, Diaz C, Marino M. Age-specific prevalence of Escherichia coli with localized and aggregative adherence in Venezuelan infants with acute diarrhea. J Clin Microbiol. 1997;35:1103. doi: 10.1128/jcm.35.5.1103-1107.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 421.Okeke IN, Lamikanra A, Czeczulin J. Heterogeneous virulence of enteroaggregative Escherichia coli strains isolated from children in Southwest Nigeria. J Infect Dis. 2000;181:252. doi: 10.1086/315204. [DOI] [PubMed] [Google Scholar]
- 422.Bouzari S, Jafari A, Farhoudi-Moghaddam AA, Shokouhi F, Parsi M. Adherence of non-enteropathogenic Escherichia coli to HeLa cells. J Med Microbiol. 1994;40:95. doi: 10.1099/00222615-40-2-95. [DOI] [PubMed] [Google Scholar]
- 423.Pabst WL, Altwegg M, Kind C. Prevalence of enteroaggregative Escherichia coli among children with and without diarrhea in Switzerland. J Clin Microbiol. 2003;41:2289. doi: 10.1128/JCM.41.6.2289-2293.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 424.Presterl E, Nadrchal R, Wolf D. Enteroaggregative and enterotoxigenic Escherichia coli among isolates from patients with diarrhea in Austria. Eur J Clin Microbiol Infect Dis. 1999;18:209. doi: 10.1007/s100960050261. [DOI] [PubMed] [Google Scholar]
- 425.Huppertz HI, Rutkowski S, Aleksic S, Karch H. Acute and chronic diarrhoea and abdominal colic associated with enteroaggregative Escherichia coli in young children living in western Europe. Lancet. 1997;349:1660. doi: 10.1016/S0140-6736(96)12485-5. [DOI] [PubMed] [Google Scholar]
- 426.Tompkins DS, Hudson MJ, Smith HR. A study of infectious intestinal disease in England: microbiological findings in cases and controls. Commun Dis Public Health. 1999;2:108. [PubMed] [Google Scholar]
- 427.Cohen M, Nataro JP. Unpublished observations, 2005.
- 428.Bouckenooghe AR, Dupont HL, Jiang ZD. Markers of enteric inflammation in enteroaggregative Escherichia coli diarrhea in travelers. Am J Trop Med Hyg. 2000;62:711. doi: 10.4269/ajtmh.2000.62.711. [DOI] [PubMed] [Google Scholar]
- 429.Miqdady MS, Jiang ZD, Nataro JP, DuPont HL. Detection of enteroaggregative Escherichia coli with formalin-preserved HEp-2 cells. J Clin Microbiol. 2002;40:3066. doi: 10.1128/JCM.40.8.3066-3067.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 430.Spencer J, Chart H, Smith HR, Rowe B. Improved detection of enteroaggregative Escherichia coli using formalin-fixed HEp-2 cells. Lett Appl Microbiol. 1997;25:325. doi: 10.1046/j.1472-765x.1997.00242.x. [DOI] [PubMed] [Google Scholar]
- 431.Wanke CA, Gerrior J, Blais V. Successful treatment of diarrheal disease associated with enteroaggregative Escherichia coli in adults infected with human immunodeficiency virus. J Infect Dis. 1998;178:1369. doi: 10.1086/314443. [DOI] [PubMed] [Google Scholar]
- 432.Adachi JA, Ericsson CD, Jiang ZD. Azithromycin found to be comparable to levofloxacin for the treatment of US travelers with acute diarrhea acquired in Mexico. Clin Infect Dis. 2003;37:1165. doi: 10.1086/378746. [DOI] [PubMed] [Google Scholar]
- 433.DuPont HL, Jiang ZD, Ericsson CD. Rifaximin versus ciprofloxacin for the treatment of traveler’s diarrhea: a randomized, double-blind clinical trial. Clin Infect Dis. 2001;33:1807. doi: 10.1086/323814. [DOI] [PubMed] [Google Scholar]
- 434.Bilge SS, Clausen CR, Lau W. Molecular characterization of a fimbrial adhesin, F1845, mediating diffuse adherences of diarrhea-associated Escherichia coli to HEp-2 cells. J Bacteriol. 1989;171:4281. doi: 10.1128/jb.171.8.4281-4289.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 435.Gunzburg ST, Chang BJ, Elliott SJ. Diffuse and enteroaggregative patterns of adherence of enteric Escherichia coli isolated from aboriginal children from the Kimberley region of western Australia. J Infect Dis. 1993;167:755. doi: 10.1093/infdis/167.3.755. [DOI] [PubMed] [Google Scholar]
- 436.Baqui AH, Sack RB, Black RE. Enteropathogens associated with acute and persistent diarrhea in Bangladeshi children <5 years of age. J Infect Dis. 1992;166:792. doi: 10.1093/infdis/166.4.792. [DOI] [PubMed] [Google Scholar]
- 437.Elliott SJ, Srinivas S, Albert MJ. Characterization of the roles of hemolysin and other toxins in enteropathy caused by alpha-hemolytic Escherichia coli linked to human diarrhea. Infect Immun. 1998;66:2040. doi: 10.1128/iai.66.5.2040-2051.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 438.Ewing WH. Edwards and Ewing’s Identification of Enterobacteriaceae. 4th ed. Elsevier; New York: 1986. [Google Scholar]
- 439.Meadow WL, Schneider H, Beem MO. Salmonella enteritidis bacteremia in childhood. J Infect Dis. 1985;152:185. doi: 10.1093/infdis/152.1.185. [DOI] [PubMed] [Google Scholar]
- 440.Hyams JS, Durbin WA, Grand RJ. Salmonella bacteremia in the first year of life. J Pediatr. 1980;96:57. doi: 10.1016/s0022-3476(80)80325-8. [DOI] [PubMed] [Google Scholar]
- 441.Taylor DN, Bied JM, Munro JS. Salmonella dublin infections in the United States, 1979–1980. J Infect Dis. 1982;146:322. doi: 10.1093/infdis/146.3.322. [DOI] [PubMed] [Google Scholar]
- 442.Zhou D, Galan J. Salmonella entry into host cells: the work in concert of type III secreted effector proteins. Microbes Infect. 2001;3:1293. doi: 10.1016/s1286-4579(01)01489-7. [DOI] [PubMed] [Google Scholar]
- 443.Zhang S, Kingsley RA, Santos RL. Molecular pathogenesis of Salmonella enterica serotype typhimurium-induced diarrhea. Infect Immun. 2003;71:1. doi: 10.1128/IAI.71.1.1-12.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 444.Waterman SR, Holden DW. Functions and effectors of the Salmonella pathogenicity island 2 type III secretion system. Cell Microbiol. 2003;5:501. doi: 10.1046/j.1462-5822.2003.00294.x. [DOI] [PubMed] [Google Scholar]
- 445.Fierer J, Krause M, Tauxe R. Salmonella typhimurium bacteremia: association with the virulence plasmid. J Infect Dis. 1992;166:639. doi: 10.1093/infdis/166.3.639. [DOI] [PubMed] [Google Scholar]
- 446.Giannella RA, Formal SB, Dammin GJ. Pathogenesis of salmonellosis: studies of fluid secretion, mucosal invasion, and morphologic reaction in the rabbit ileum. J Clin Invest. 1973;52:441. doi: 10.1172/JCI107201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 447.Jiwa SF. Probing for enterotoxigenicity among the salmonellae: an evaluation of biological assays. J Clin Microbiol. 1981;14:463. doi: 10.1128/jcm.14.5.463-472.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 448.Koo FCW, Peterson JW, Houston CW. Pathogenesis of experimental salmonellosis: inhibition of protein synthesis by cytotoxin. Infect Immun. 1984;43:93. doi: 10.1128/iai.43.1.93-100.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 449.Ashkenazi S, Cleary T, Murray BE. Cytotoxin production by Salmonella strains: quantitative analysis and partial characterization. Infect Immun. 1988;56:3089. doi: 10.1128/iai.56.12.3089-3094.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 450.Takeuchi A. Electron microscope studies of experimental Salmonella infection. I. Penetration into the intestinal epithelium by S. typhimurium. Am J Pathol. 1967;50:109. [PMC free article] [PubMed] [Google Scholar]
- 451.Modrazakowski MC, Spitznagel JK. Bactericidal activity of fractionated granule contents from human polymorphonuclear leukocytes: antagonism of granule cationic proteins by lipopolysaccharide. Infect Immun. 1979;25:597. doi: 10.1128/iai.25.2.597-602.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 452.Weiss J, Victor M, Elsbach P. Role of charge and hydrophobic interaction in the action of the bactericidal/permeability increasing protein of neutrophils on gram-negative bacteria. J Clin Invest. 1983;71:540. doi: 10.1172/JCI110798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 453.Mackaness GV. Resistance to intracellular infection. J Infect Dis. 1971;123:439. doi: 10.1093/infdis/123.4.439. [DOI] [PubMed] [Google Scholar]
- 454.Mackaness GV, Blander RV, Collins FM. Host parasite relations in mouse typhoid. J Exp Med. 1966;124:573. doi: 10.1084/jem.124.4.573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 455.McKenzie SE, Kline J, Douglas SD, Polin RA. Enhancement in vitro of the low interferon-gamma production of leukocytes from human newborn infants. J Leukoc Biol. 1993;53:691. doi: 10.1002/jlb.53.6.691. [DOI] [PubMed] [Google Scholar]
- 456.George A. Generation of gamma interferon responses in murine Peyer’s patches following oral immunization. Infect Immun. 1996;64:4606. doi: 10.1128/iai.64.11.4606-4611.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 457.de Jong R, Altare F, Haagen IA. Severe mycobacterial and Salmonella infections in interleukin-12 receptor-deficient patients. Science. 1998;280:1435. doi: 10.1126/science.280.5368.1435. [DOI] [PubMed] [Google Scholar]
- 458.Hornick RB, Greisman SE, Woodward TE. Typhoid fever: pathogenesis and immunologic control. N Engl J Med. 1970;283:686. doi: 10.1056/NEJM197009242831306. [DOI] [PubMed] [Google Scholar]
- 459.Kent TH, Formal SB, LaBrec EH. Salmonella gastroenteritis in rhesus monkeys. Arch Pathol. 1966;82:272. [PubMed] [Google Scholar]
- 460.Boyd JF. Pathology of the alimentary tract of S. typhimurium food poisoning. Gut. 1985;26:935. doi: 10.1136/gut.26.9.935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 461.Day DW, Mandel BK, Morson BC. The rectal biopsy appearances in Salmonella colitis. Histopathology. 1978;2:117. doi: 10.1111/j.1365-2559.1978.tb01700.x. [DOI] [PubMed] [Google Scholar]
- 462.Wilder AN, MacCready RA. Isolation of Salmonella from poultry, poultry products and poultry processing plants in Massachusetts. N Engl J Med. 1966;274:1453. doi: 10.1056/NEJM196606302742601. [DOI] [PubMed] [Google Scholar]
- 463.Pet turtle-associated salmonellosis—Puerto Rico. MMWR Morb Mortal Wkly Rep. 1984;33:141. [PubMed] [Google Scholar]
- 464.Sanyal D, Douglas T, Roberts R. Salmonella infection acquired from reptilian pets. Arch Dis Child. 1997;77:345. doi: 10.1136/adc.77.4.345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 465.Mermin J, Hoar B, Angulo FJ. Iguanas and Salmonella marina infection in children: a reflection of the increasing incidence of reptile-associated salmonellosis in the United States. Pediatrics. 1997;99:399. doi: 10.1542/peds.99.3.399. [DOI] [PubMed] [Google Scholar]
- 466.Woodward DL, Khakhria R, Johnson WM. Human salmonellosis associated with exotic pets. J Clin Microbiol. 1997;35:2786. doi: 10.1128/jcm.35.11.2786-2790.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 467.Thomson S. Paratyphoid fever and Baker’s confectionery: analysis of epidemic in South Wales 1952. Monthly Bull Ministry Health Public Health Serv. 1953;12:187. [PubMed] [Google Scholar]
- 468.Watt J, Wegman ME, Brown OW. Salmonellosis in a premature nursery unaccompanied by diarrheal diseases. Pediatrics. 1958;22:689. [PubMed] [Google Scholar]
- 469.Hargrett-Bean NT, Pavia AT, Tauxe RV. Salmonella isolates from humans in the United States, 1984–1986. MMWR Morb Mortal Wkly Rep. 1988;37:25SS. [PubMed] [Google Scholar]
- 470.Blaser MJ, Newman LS. A review of human salmonellosis. I. Infective dose. Rev Infect Dis. 1982;4:1096. doi: 10.1093/clinids/4.6.1096. [DOI] [PubMed] [Google Scholar]
- 471.Rubinstein AD, Fowler RN. Salmonellosis of the newborn with transmission by delivery room resuscitators. Am J Public Health. 1955;45:1109. doi: 10.2105/ajph.45.9.1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 472.Bate JG, James U. Salmonella typhimurium infection dustborne in a children’s ward. Lancet. 1958;2:713. doi: 10.1016/s0140-6736(58)91332-1. [DOI] [PubMed] [Google Scholar]
- 473.Rubbo SD. Cross-infection in hospital due to Salmonella derby. J Hyg. 1948;46:158. doi: 10.1017/s002217240003624x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 474.Lamb VA, Mayhall CG, Spadora AC. Outbreak of S. typhimurium gastroenteritis due to an imported strain resistant to ampicillin, chloramphenicol, and trimethoprim/sulfamethoxazole in a nursery. J Clin Microbiol. 1984;20:1076. doi: 10.1128/jcm.20.6.1076-1079.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 475.Abramas IF, Cochran WD, Holmes LB. A Salmonella newport outbreak in a premature nursery with a one year follow up. Pediatrics. 1966;37:616. [PubMed] [Google Scholar]
- 476.Epstein HC, Hochwald A, Agha R. Salmonella infections of the newborn infant. J Pediatr. 1951;38:723. doi: 10.1016/s0022-3476(51)80134-3. [DOI] [PubMed] [Google Scholar]
- 477.Abramson H. Infections with S. typhimurium in the newborn. Am J Dis Child. 1947;74:576. doi: 10.1001/archpedi.1947.02030010590003. [DOI] [PubMed] [Google Scholar]
- 478.Leeder FS. An epidemic of S. panama infections in infants. Ann N Y Acad Sci. 1956;66:54. doi: 10.1111/j.1749-6632.1956.tb40102.x. [DOI] [PubMed] [Google Scholar]
- 479.Watt J, Carlton E. Studies of the acute diarrheal diseases. XVI. An outbreak of S. typhimurium infection among newborn premature infants. Public Health Rep. 1945;60(Pt 1):734. [Google Scholar]
- 480.Foley AR. An outbreak of paratyphoid B fever in a nursery of a small hospital. Can J Public Health. 1947;38:73. [PubMed] [Google Scholar]
- 481.Seligman E. Mass invasion of salmonellae in a babies’ ward. Ann Paediatr. 1949;172:406. [PubMed] [Google Scholar]
- 482.Rowe B, Giles C, Brown GL. Outbreak of gastroenteritis due to S. virchow in a maternity hospital. BMJ. 1969;3:561. doi: 10.1136/bmj.3.5670.561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 483.Sasidharan CK, Rajagopal KC, Jayaram CK. S. typhimurium epidemic in newborn nursery. Indian J Pediatr. 1983;50:599. doi: 10.1007/BF02957725. [DOI] [PubMed] [Google Scholar]
- 484.Borecka J, Hocmannova M, van Leeuwen WJ. Nosocomial infection of nurslings caused by multiple drug-resistant strain of S. typhimurium-utilization of a new typing method based on lysogeny of strains. Z Bakteriol. 1976;2336:262. [PubMed] [Google Scholar]
- 485.Bannerman CHS. Heidelberg enteritis—an outbreak in the neonatal unit of Harare Central Hospital. Cent Afr J Med. 1985;31:1. [PubMed] [Google Scholar]
- 486.McAllister TA, Roud JA, Marshall A. Outbreak of S. eimsbuettel in newborn infants spread by rectal thermometers. Lancet. 1986;1:1262. doi: 10.1016/s0140-6736(86)91397-8. [DOI] [PubMed] [Google Scholar]
- 487.Szanton VL. Epidemic salmonellosis: a 30-month study of 80 cases of S. oranienburg infection. Pediatrics. 1957;20:794. [PubMed] [Google Scholar]
- 488.Seals JE, Parrott PL, McGowan JE. Nursery salmonellosis: delayed recognition due to unusually long incubation period. Infect Control Hosp Epidemiol. 1983;4:205. doi: 10.1017/s0195941700058239. [DOI] [PubMed] [Google Scholar]
- 489.Hering E, Fuenzalida O, Lynch B. Analises clinico-epidemiologica de un brote de infeccion por S. bredeney en recien nacidos. Rev Clin Pediatr. 1979;50:81. [PubMed] [Google Scholar]
- 490.Kumari S, Gupta R, Bhargava SK. A nursery outbreak with S. newport. Indian Pediatr. 1980;17:11. [PubMed] [Google Scholar]
- 491.Omland T, Gardborg O. Salmonella enteritidis infections in infancy with special reference to a small nosocomial epidemic. Acta Paediatr Belg. 1960;49:583. doi: 10.1111/j.1651-2227.1960.tb07773.x. [DOI] [PubMed] [Google Scholar]
- 492.Puri V, Thirupuram S, Khalil A. Nosocomial S. typhimurium epidemic in a neonatal special care unit. Indian Pediatr. 1980;17:233. [PubMed] [Google Scholar]
- 493.Mendis NMP, de la Motte PU, Gunatillaka PDP. Protracted infection with S. bareilly in a maternity hospital. J Trop Med Hyg. 1976;79:142. [PubMed] [Google Scholar]
- 494.Marzetti G, Laurenti F, deCaro M. Salmonella muenchen infections in newborns and small infants. Clin Pediatr. 1973;12:93. doi: 10.1177/000992287301200211. [DOI] [PubMed] [Google Scholar]
- 495.Baine WB, Gangarosa EJ, Bennett JV. Institutional salmonellosis. J Infect Dis. 1973;128:357. doi: 10.1093/infdis/128.3.357. [DOI] [PubMed] [Google Scholar]
- 496.Schroeder SA, Aserkoff B, Brachman PS. Epidemic salmonellosis in hospitals and institutions. N Engl J Med. 1968;279:674. doi: 10.1056/NEJM196809262791303. [DOI] [PubMed] [Google Scholar]
- 497.Wilson R, Feldman RA, Davis J. Salmonellosis in infants: the importance of intrafamilial transmission. Pediatrics. 1982;69:436. [PubMed] [Google Scholar]
- 498.Newman MJ. Multiple-resistant Salmonella group G outbreak in a neonatal intensive care unit. West Afr J Med. 1996;15:165. [PubMed] [Google Scholar]
- 499.Mahajan R, Mathur M, Kumar A. Nosocomial outbreak of Salmonella typhimurium infection in a nursery intensive care unit (NICU) and paediatric ward. J Commun Dis. 1995;27:10. [PubMed] [Google Scholar]
- 500.Martyn-Jones DM, Pantin GC. Neonatal diarrhea due to S. paratyphi B. J Clin Pathol. 1956;9:128. doi: 10.1136/jcp.9.2.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 501.Rubinstein AD, Feemster RF, Smith HM. Salmonellosis as a public health problem in wartime. Am J Public Health. 1944;34:841. doi: 10.2105/ajph.34.8.841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 502.Neter E. Observation on the transmission of salmonellosis in man. Am J Public Health. 1950;40:929. doi: 10.2105/ajph.40.8.929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 503.Sanders DY, Sinal SH, Morrison L. Chronic salmonellosis in infancy. Clin Pediatr. 1974;13:640. doi: 10.1177/000992287401300804. [DOI] [PubMed] [Google Scholar]
- 504.Waddell WR, Kunz LJ. Association of Salmonella enteritis with operations on the stomach. N Engl J Med. 1956;255:555. doi: 10.1056/NEJM195609202551203. [DOI] [PubMed] [Google Scholar]
- 505.Gray JA, Trueman AM. Severe Salmonella gastroenteritis associated with hypochlorhydria. Scott Med J. 1971;16:255. doi: 10.1177/003693307101600504. [DOI] [PubMed] [Google Scholar]
- 506.Fleischhacker G, Vutue C, Werner H-P. Infektion eines Neugeborenen durch S. typhimurium-haltige Muttermilch. Wien Klin Wochenschr. 1972;24:394. [PubMed] [Google Scholar]
- 507.Ryder RW, Crosby-Ritchie A, McDonough B. Human milk contaminated with S. kottbus. A cause of nosocomial illness in infants. JAMA. 1977;238:1533. [PubMed] [Google Scholar]
- 508.Revathi G, Mahajan R, Faridi MM. Transmission of lethal Salmonella senftenberg from mother’s breast-milk to her baby. Ann Trop Paediatr. 1995;15:159. doi: 10.1080/02724936.1995.11747765. [DOI] [PubMed] [Google Scholar]
- 509.Small RG, Sharp JCM. A milkborne outbreak of S. dublin. J Hyg. 1979;82:95. doi: 10.1017/s0022172400025511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 510.Weissman JB, Deen RMAD, Williams M. An island-wide epidemic of salmonellosis in Trinidad traced to contaminated powdered milk. West Indian Med J. 1977;26:135. [PubMed] [Google Scholar]
- 511.Salmonella anatum infection in infants linked to dried milk. Commun Dis Rep CDR Wkly. 1997;7:33. [PubMed] [Google Scholar]
- 512.Usera MA, Echeita A, Aladuena A. Interregional foodborne salmonellosis outbreak due to powdered infant formula contaminated with lactose-fermenting Salmonella virchow. Eur J Epidemiol. 1996;12:377. doi: 10.1007/BF00145301. [DOI] [PubMed] [Google Scholar]
- 513.Silverstope L, Plazikowski U, Kjellander J. An epidemic among infants caused by S. muenchen. J Appl Bacteriol. 1961;24:134. [Google Scholar]
- 514.Im SWK, Chow K, Chau PY. Rectal thermometer-mediated cross-infection with S. wadsworth in a pediatric ward. J Hosp Infect. 1981;2:171. doi: 10.1016/0195-6701(81)90026-8. [DOI] [PubMed] [Google Scholar]
- 515.Khan MA, Abdur-Rab M, Israr N. Transmission of S. worthington by oropharyngeal suction in hospital neonatal unit. Pediatr Infect Dis J. 1991;10:668. doi: 10.1097/00006454-199109000-00009. [DOI] [PubMed] [Google Scholar]
- 516.Umasankar S, Mridha EU, Hannan MM. An outbreak of Salmonella enteritidis in a maternity and neonatal intensive care unit. J Hosp Infect. 1996;34:117. doi: 10.1016/s0195-6701(96)90136-x. [DOI] [PubMed] [Google Scholar]
- 517.Riley LW, Cohen ML. Plasmid profiles and Salmonella epidemiology. Lancet. 1982;1:573. doi: 10.1016/s0140-6736(82)92088-8. [DOI] [PubMed] [Google Scholar]
- 518.Michel J, Malpeach G, Godeneche P. Etude clinique et bactériologique d’une épidémie de salmonellose en milieu hospitalier (S. oranienburg) Pediatrie. 1970;25:13. [PubMed] [Google Scholar]
- 519.Adler JL, Anderson RL, Boring JR. A protracted hospital-associated outbreak of salmonellosis due to a multiple antibiotic-resistant strain of S. indiana. J Pediatr. 1970;77:970. doi: 10.1016/s0022-3476(70)80079-8. [DOI] [PubMed] [Google Scholar]
- 520.Bilie BO, Mellbin T, Nordbring F. An extensive outbreak of gastroenteritis caused by S. newport. Acta Med Scand. 1964;175:557. doi: 10.1111/j.0954-6820.1964.tb00607.x. [DOI] [PubMed] [Google Scholar]
- 521.Horwitz M, Pollard R, Merson M. A large outbreak of foodborne salmonellosis on the Navajo Nation Indian Reservation: epidemiology and secondary transmission. Am J Public Health. 1977;67:1071. doi: 10.2105/ajph.67.11.1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 522.Griffith JPC, Ostheimer M. Typhoid fever in children. Am J Med Sci. 1902;124:868. [Google Scholar]
- 523.Freedman ML, Christopher P, Boughton CR. Typhoid carriage in pregnancy with infection of neonate. Lancet. 1970;1:310. doi: 10.1016/s0140-6736(70)90689-6. [DOI] [PubMed] [Google Scholar]
- 524.Chhabra RS, Glaser JH. Salmonella infection presenting as hematochezia on the first day of life. Pediatrics. 1994;94:739. [PubMed] [Google Scholar]
- 525.Stein H, Beck J, Solomon A. Gastroenteritis with necrotizing enterocolitis in premature babies. BMJ. 1972;1:616. doi: 10.1136/bmj.2.5814.616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 526.Guarino A, Spagnuolo MI, Russo S. Etiology and risk factors of severe and protracted diarrhea. J Pediatr Gastroenterol Nutr. 1995;20:173. doi: 10.1097/00005176-199502000-00006. [DOI] [PubMed] [Google Scholar]
- 527.Lifshitz F, Coello-Ramirez P, Gutirrez-Topete G. Monosaccharide intolerance and hypoglycemia in infants with diarrhea. I. Clinical course of 23 infants. J Pediatr. 1970;77:595. doi: 10.1016/s0022-3476(70)80200-1. [DOI] [PubMed] [Google Scholar]
- 528.Iyngkaran N, Abdin Z, Davis K. Acquired carbohydrate intolerance and cow milk protein-sensitive enteropathy in young infants. J Pediatr. 1979;95:373. doi: 10.1016/s0022-3476(79)80509-0. [DOI] [PubMed] [Google Scholar]
- 529.Lo CW, Walker WA. Chronic protracted diarrhea of infancy: a nutritional disease. Pediatrics. 1983;72:786. [PubMed] [Google Scholar]
- 530.Blaser MJ, Feldman RA. Salmonella bacteremia: reports to the Centers for Disease Control, 1968–1979. J Infect Dis. 1981;143:743. doi: 10.1093/infdis/143.5.743. [DOI] [PubMed] [Google Scholar]
- 531.Schutze GE, Schutze SE, Kirby RS. Extraintestinal salmonellosis in a children’s hospital. Pediatr Infect Dis J. 1997;16:482. doi: 10.1097/00006454-199705000-00005. [DOI] [PubMed] [Google Scholar]
- 532.Davis RC. Salmonella sepsis in infancy. Am J Dis Child. 1981;135:1096. doi: 10.1001/archpedi.1981.02130360004003. [DOI] [PubMed] [Google Scholar]
- 533.Torrey S, Fleisher G, Jaffe D. Incidence of Salmonella bacteremia in infants with Salmonella gastroenteritis. J Pediatr. 1986;108:718. doi: 10.1016/s0022-3476(86)81050-2. [DOI] [PubMed] [Google Scholar]
- 534.Sirinavin S, Jayanetra P, Lolekha S. Predictors for extraintestinal infection of Salmonella enteritis in Thailand. Pediatr Infect Dis J. 1988;7:44. doi: 10.1097/00006454-198801000-00011. [DOI] [PubMed] [Google Scholar]
- 535.Katz BZ, Shapiro ED. Predictors of persistently positive blood cultures in children with “occult” Salmonella bacteremia. Pediatr Infect Dis J. 1986;5:713. doi: 10.1097/00006454-198611000-00029. [DOI] [PubMed] [Google Scholar]
- 536.Yamamoto LG, Ashton MJ. Salmonella infections in infants in Hawaii. Pediatr Infect Dis J. 1988;7:48. doi: 10.1097/00006454-198801000-00012. [DOI] [PubMed] [Google Scholar]
- 537.Cohen JI, Bartlett JA, Corey GR. Extraintestinal manifestations of Salmonella infections. Medicine (Baltimore) 1987;66:349. doi: 10.1097/00005792-198709000-00003. [DOI] [PubMed] [Google Scholar]
- 538.West SE, Goodkin R, Kaplan AM. Neonatal Salmonella meningitis complicated by cerebral abscesses. West J Med. 1977;127:142. [PMC free article] [PubMed] [Google Scholar]
- 539.Applebaum PC, Scragg J. Salmonella meningitis in infants. Lancet. 1977;1:1052. doi: 10.1016/s0140-6736(77)91280-6. [DOI] [PubMed] [Google Scholar]
- 540.Cherubin CE, Marr JS, Sierra MF. Listeria and gram-negative bacillary meningitis in New York City 1973–1979. Am J Med. 1981;71:199. doi: 10.1016/0002-9343(81)90106-6. [DOI] [PubMed] [Google Scholar]
- 541.Low LC, Lam BC, Wong WT. Salmonella meningitis in infancy. Aust Paediatr J. 1984;20:225. doi: 10.1111/j.1440-1754.1984.tb00084.x. [DOI] [PubMed] [Google Scholar]
- 542.Diwan N, Sharma KB. Isolation of S. typhimurium from cephalohematoma and osteomyelitis. Indian J Med Res. 1978;67:27. [PubMed] [Google Scholar]
- 543.Konzert W. Über eine Salmonella-Osteomyelitis in Rahmen einer S. typhimurium Epidemie auf einer Neugeborenen Station. Wien Klin Wochenschr. 1969;81:713. [PubMed] [Google Scholar]
- 544.McKinlay B. Infectious diarrhea of the newborn caused by an unclassified species of Salmonella. Am J Dis Child. 1937;54:1252. [Google Scholar]
- 545.Szumness W, Sikorska J, Szymanek E. The microbiological and epidemiological properties of infections caused by S. enteritidis. J Hyg. 1966;64:9. doi: 10.1017/s0022172400040298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 546.Nelson JD. Suppurative mastitis in infants. Am J Dis Child. 1973;125:458. doi: 10.1001/archpedi.1973.04160030108024. [DOI] [PubMed] [Google Scholar]
- 547.Guthrie KJ, Montgomery GI. Infections with Bacterium enteritidis in infancy with the triad of enteritis, cholecystitis and meningitis. J Pathol Bacteriol. 1939;49:393. [Google Scholar]
- 548.Corman LI, Poirier RH, Littlefield CA. Endophthalmitis due to S. enteritidis. J Pediatr. 1979;95:1001. doi: 10.1016/s0022-3476(79)80295-4. [DOI] [PubMed] [Google Scholar]
- 549.Haggman DL, Rehm SJ, Moodie DS. Nontyphoidal Salmonella pericarditis: a case report and review of the literature. Pediatr Infect Dis J. 1986;5:259. [PubMed] [Google Scholar]
- 550.Reed RP, Klugman KP. Neonatal typhoid fever. Pediatr Infect Dis J. 1994;13:774. doi: 10.1097/00006454-199409000-00004. [DOI] [PubMed] [Google Scholar]
- 551.Stuart BM, Pullen RL. Typhoid: clinical analysis of 360 cases. Arch Intern Med. 1946;78:629. doi: 10.1001/archinte.1946.00220060002001. [DOI] [PubMed] [Google Scholar]
- 552.Sengupta B, Ramachander N, Zamah N. Salmonella septic abortion. Int Surg. 1980;65:183. [PubMed] [Google Scholar]
- 553.Diddle AW, Stephens RL. Typhoid fever in pregnancy. Am J Obstet Gynecol. 1939;38:300. [Google Scholar]
- 554.Riggall F, Salkind G, Spellacy W. Typhoid fever complicating pregnancy. Obstet Gynecol. 1974;44:117. [PubMed] [Google Scholar]
- 555.Hicks HT, French H. Typhoid fever and pregnancy with special references to fetal infection. Lancet. 1905;1:1491. [Google Scholar]
- 556.Osler W, McCrae T. Osler W. Principles and Practice of Medicine. 8th ed. D Appleton; New York: 1912. Typhoid fever; pp. 1–46. [Google Scholar]
- 557.Ferreccio C, Levine MM, Manterola A. Benign bacteremia caused by S. typhi and S. paratyphi in children younger than two years. J Pediatr. 1984;104:899. doi: 10.1016/s0022-3476(84)80492-8. [DOI] [PubMed] [Google Scholar]
- 558.Thisyakorn U, Mansuwan P, Taylor DN. Typhoid and paratyphoid fever in 192 hospitalized children in Thailand. Am J Dis Child. 1987;141:862. doi: 10.1001/archpedi.1987.04460080048025. [DOI] [PubMed] [Google Scholar]
- 559.Pickering LK, DuPont HL, Olarte J. Fecal leukocytes in enteric infections. Am J Clin Pathol. 1977;68:562. doi: 10.1093/ajcp/68.5.562. [DOI] [PubMed] [Google Scholar]
- 560.McCall CE, Martin WT Boring JR Efficiency of cultures of rectal swabs and fecal specimens in detecting Salmonella carriers: correlation with numbers of Salmonella excreted. J Hyg. 1966;64:261. doi: 10.1017/s0022172400040547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 561.Raucher HS, Eichenfield AH, Hodes HL. Treatment of Salmonella gastroenteritis in infants. The significance of bacteremia. Clin Pediatr. 1983;22:601. doi: 10.1177/000992288302200901. [DOI] [PubMed] [Google Scholar]
- 562.Gotoff SP, Cochran WD. Antibody response to the somatic antigen of S. newport in premature infants. Pediatrics. 1966;37:610. [PubMed] [Google Scholar]
- 563.Hodes HL, Zepp HD, Ainbender E. Production of O and H agglutinins by a newborn infant infected with S. saint-paul. J Pediatr. 1966;68:780. doi: 10.1016/s0022-3476(66)80453-5. [DOI] [PubMed] [Google Scholar]
- 564.Taylor DN, Bopp C, Birkness K. An outbreak of salmonellosis associated with a fatality in a healthy child: a large dose and severe illness. Am J Epidemiol. 1984;119:907. doi: 10.1093/oxfordjournals.aje.a113813. [DOI] [PubMed] [Google Scholar]
- 565.Rivera MJ, Rivera N, Castillo J. Molecular and epidemiological study of Salmonella clinical isolates. J Clin Microbiol. 1991;29:927. doi: 10.1128/jcm.29.5.927-932.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 566.Aserkoff B, Bennett JV. Effect of antibiotic therapy in acute salmonellosis on the fecal excretion of salmonellae. N Engl J Med. 1969;281:636. doi: 10.1056/NEJM196909182811202. [DOI] [PubMed] [Google Scholar]
- 567.Dixon JMS. Effect of antibiotic treatment on duration of excretion of S. typhimurium by children. BMW. 1965;2:1343. doi: 10.1136/bmj.2.5474.1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 568.Kazemi M, Bumpert TG, Marks MI. A controlled trial comparing trimethoprim/sulfamethoxazole, ampicillin, and no therapy in the treatment of Salmonella gastroenteritis in children. J Pediatr. 1973;83:646. doi: 10.1016/s0022-3476(73)80234-3. [DOI] [PubMed] [Google Scholar]
- 569.Neill MA, Opal SM, Heelan J. Failure of ciprofloxacin to eradicate convalescent fecal excretion after acute salmonellosis: experience during an outbreak in health care workers. Ann Intern Med. 1991;114:195. doi: 10.7326/0003-4819-114-3-195. [DOI] [PubMed] [Google Scholar]
- 570.Pettersson T, Klemola E, Wager O. Treatment of acute cases of Salmonella infection and Salmonella carriers with ampicillin and neomycin. Acta Med Scand. 1964;175:185. doi: 10.1111/j.0954-6820.1964.tb00565.x. [DOI] [PubMed] [Google Scholar]
- 571.Association for Study of Infectious Diseases Effect of neomycin in noninvasive Salmonella infections of the gastrointestinal tract. Lancet. 1970;2:1159. [PubMed] [Google Scholar]
- 572.Nelson JD, Kusmiesz H, Jackson LH. Treatment of Salmonella gastroenteritis with ampicillin, amoxicillin, or placebo. Pediatrics. 1980;65:1125. [PubMed] [Google Scholar]
- 573.Asperilla MO, Smego RA, Scott LK. Quinolone antibiotics in the treatment of Salmonella infections. Rev Infect Dis. 1990;12:873. doi: 10.1093/clinids/12.5.873. [DOI] [PubMed] [Google Scholar]
- 574.Buchwald DS, Blaser MJ. A review of human salmonellosis. II. Duration of excretion following infection with nontyphi Salmonella. Rev Infect Dis. 1984;6:345. doi: 10.1093/clinids/6.3.345. [DOI] [PubMed] [Google Scholar]
- 575.Dutta P, Rasaily R, Saha MR. Ciprofloxacin for treatment of severe typhoid fever in children. Antimicrob Agents Chemother. 1993;37:1197. doi: 10.1128/aac.37.5.1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 576.Piddock LJV, Griggs DJ, Hall MC. Ciprofloxacin resistance in clinical isolates of Salmonella typhimurium obtained from two patients. Antimicrob Agents Chemother. 1993;37:662. doi: 10.1128/aac.37.4.662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 577.Edgar WM, Lacey BW. Infection with S. heidelberg. An outbreak presumably not foodborne. Lancet. 1963;1:161. [Google Scholar]
- 578.Rice PA, Craven PC, Wells JG. S. heidelberg enteritis and bacteremia. An epidemic on two pediatric wards. Am J Med. 1976;60:509. doi: 10.1016/0002-9343(76)90717-8. [DOI] [PubMed] [Google Scholar]
- 579.MacDonald KL, Cohen ML, Hargrett-Bean NT. Changes in antimicrobial resistance of Salmonella isolated from humans in the United States. JAMA. 1987;258:1496. [PubMed] [Google Scholar]
- 580.Maiorini E, Lopez EL, Morrow AL. Multiply resistant nontyphoidal Salmonella gastroenteritis in children. Pediatr Infect Dis J. 1993;12:139. doi: 10.1097/00006454-199302000-00007. [DOI] [PubMed] [Google Scholar]
- 581.Lee LA, Puhr ND, Maloney EK. Increase in antimicrobial-resistant Salmonella infections in the United States, 1989–1990. J Infect Dis. 1994;170:128. doi: 10.1093/infdis/170.1.128. [DOI] [PubMed] [Google Scholar]
- 582.Glynn MK, Bopp C, Dewitt W. Emergence of multidrug-resistant Salmonella enterica serotype typhimurium DT104 infections in the United States. N Engl J Med. 1998;338:1333. doi: 10.1056/NEJM199805073381901. [DOI] [PubMed] [Google Scholar]
- 583.Koshi G. Alarming increases in multi-drug resistant S. typhimurium in Southern India. Indian J Med Res. 1981;74:635. [PubMed] [Google Scholar]
- 584.Anderson ES, Threlfall EJ, Carr JM. Clonal distribution of resistance plasmid carrying S. typhimurium, mainly in the Middle East. J Hyg. 1977;79:425. doi: 10.1017/s0022172400053286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 585.Wamola IA, Mirza NB. Problems of Salmonella infections in a hospital in Kenya. East Afr Med J. 1981;58:677. [PubMed] [Google Scholar]
- 586.Falbo V, Capriola A, Moncello F. Antimicrobial resistance among Salmonella isolates from hospitals in Rome. J Hyg. 1982;88:275. doi: 10.1017/s0022172400070133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 587.Threlfall EJ, Ward LR, Ashley AS. Plasmid encoded trimethoprim resistance in multi-resistant epidemic S. typhimurium phagotypes 204 and 193 in Britain. BMJ. 1980;280:1210. doi: 10.1136/bmj.280.6225.1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 588.French GI, Lowry MF. Trimethoprim-resistant Salmonella. Lancet. 1978;2:375. doi: 10.1016/s0140-6736(77)90595-5. [DOI] [PubMed] [Google Scholar]
- 589.Smith SM, Palumbo PE, Edelson PJ. Salmonella strains resistant to multiple antibiotics: therapeutic implications. Pediatr Infect Dis J. 1984;3:455. doi: 10.1097/00006454-198409000-00017. [DOI] [PubMed] [Google Scholar]
- 590.Soe GB, Overturf GD. Treatment of typhoid fever and other systemic salmonellosis with cefotaxime, ceftriazone, cefoperazone, and other newer cephalosporins. Rev Infect Dis. 1987;9:719. doi: 10.1093/clinids/9.4.719. [DOI] [PubMed] [Google Scholar]
- 591.Moosa A, Rubidge CJ. Once daily ceftriaxone vs. chloramphenicol for treatment of typhoid fever in children. Pediatr Infect Dis J. 1989;8:696. doi: 10.1097/00006454-198910000-00007. [DOI] [PubMed] [Google Scholar]
- 592.deCarvalho EM, Martinelli R, de Oliveira MMG. Cefamandole treatment of Salmonella bacteremia. Antimicrob Agents Chemother. 1982;21:334. doi: 10.1128/aac.21.2.334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 593.Pape JW, Gerdes H, Oriol L. Typhoid fever: successful therapy with cefoperazone. J Infect Dis. 1986;153:272. doi: 10.1093/infdis/153.2.272. [DOI] [PubMed] [Google Scholar]
- 594.Demmerich B, Lode H, Boner K. Biliary excretion and pharmacokinetics of cefoperazone in humans. J Antimicrob Chemother. 1983;12:27. doi: 10.1093/jac/12.1.27. [DOI] [PubMed] [Google Scholar]
- 595.Kinsella TR, Yogev R, Shulman ST. Treatment of Salmonella meningitis and brain abscess with the new cephalosporins: two case reports and a review of the literature. Pediatr Infect Dis J. 1987;6:476. doi: 10.1097/00006454-198705000-00014. [DOI] [PubMed] [Google Scholar]
- 596.Gokalp AS, Toksoy HB, Turkay S. Intravenous immunoglobulin in the treatment of Salmonella typhimurium infections in preterm neonates. Clin Pediatr (Phila) 1994;33:349. doi: 10.1177/000992289403300607. [DOI] [PubMed] [Google Scholar]
- 597.Tauxe RV, Hassan LF, Findeisen KO. Salmonellosis in nurses: lack of transmission to patients. J Infect Dis. 1988;157:370. doi: 10.1093/infdis/157.2.370. [DOI] [PubMed] [Google Scholar]
- 598.Levine MM, Ferraccio C, Cryz S. Comparison of enteric coated capsules and liquid formulation of Ty21a typhoid vaccine in randomised controlled field trial. Lancet. 1990;336:4. doi: 10.1016/0140-6736(90)92266-k. [DOI] [PubMed] [Google Scholar]
- 599.Cryz SJ, Jr, Vanprapar N, Thisyakorn U. Safety and immunogenicity of Salmonella typhi Ty21a vaccine in young Thai children. Infect Immun. 1993;61:1149. doi: 10.1128/iai.61.3.1149-1151.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 600.Murphy JR, Grez L, Schlesinger L. Immunogenicity of S. typhi Ty21a vaccine for young children. Infect Immun. 1991;59:4291. doi: 10.1128/iai.59.11.4291-4293.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 601.France GL, Marmer DJ, Steele RW. Breast-feeding and Salmonella infection. Am J Dis Child. 1980;134:147. doi: 10.1001/archpedi.1980.02130140021007. [DOI] [PubMed] [Google Scholar]
- 602.Brenner DJ, Fannin GR, Skerman FJ. Polynucleotide sequence divergence among strains of E. coli and closely related organisms. J Bacteriol. 1972;109:953. doi: 10.1128/jb.109.3.953-965.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 603.Sansonetti PJ, Kopecko DJ, Formal SB. Involvement of a plasmid in the invasive ability of Shigella flexneri. Infect Immun. 1982;35:852. doi: 10.1128/iai.35.3.852-860.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 604.Sansonetti P. Host-pathogen interactions: the seduction of molecular cross talk. Gut. 2002;50(Suppl 3):III2. doi: 10.1136/gut.50.suppl_3.iii2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 605.Fernandez MI, Sansonetti PJ. Shigella interaction with intestinal epithelial cells determines the innate immune response in shigellosis. Int J Med Microbiol. 2003;293:55. doi: 10.1078/1438-4221-00244. [DOI] [PubMed] [Google Scholar]
- 606.Hale TL, Oaks EV, Formal SB. Identification and characterization of virulence associated, plasmid-coded proteins of Shigella spp. and enteroinvasive E. coli. Infect Immun. 1985;50:620. doi: 10.1128/iai.50.3.620-629.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 607.Sasakawa C, Kamata K, Sakai T. Molecular alteration of the 140-megadalton plasmid associated with the loss of virulence and Congo red binding activity in Shigella flexneri. Infect Immun. 1986;51:470. doi: 10.1128/iai.51.2.470-475.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 608.LaBrec EH, Schneider H, Magnani TJ. Epithelial cell penetration as an essential step in the pathogenesis of bacillary dysentery. J Bacteriol. 1964;88:1503. doi: 10.1128/jb.88.5.1503-1518.1964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 609.Ogawa H. Experimental approach in studies on pathogenesis of bacillary dysentery—with special reference to the invasion of bacilli into intestinal mucosa. Acta Pathol Jpn. 1970;20:261. doi: 10.1111/j.1440-1827.1970.tb03071.x. [DOI] [PubMed] [Google Scholar]
- 610.Sansonetti PJ, d’Hauteville H, Formal SB. Plasmid-mediated invasiveness of “Shigella-like” Escherichia coli. Ann Microbiol. 1982;133A:351. [PubMed] [Google Scholar]
- 611.Sansonetti PJ, d’Hauteville H, Ecobiochon C. Molecular comparison of virulence plasmids in Shigella and enteroinvasive Escherichia coli. Ann Microbiol. 1983;134A:295. [PubMed] [Google Scholar]
- 612.Sansonetti PJ, Hale TL, Dammin GI. Alterations in the pathogenicity of Escherichia coli K-12 after transfer of plasmids and chromosomal genes from Shigella flexneri. Infect Immun. 1983;39:1392. doi: 10.1128/iai.39.3.1392-1402.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 613.Wei J, Goldberg MB, Burland V. Complete genome sequence and comparative genomics of Shigella flexneri serotype 2a strain 2457T. Infect Immun. 2003;71:2775. doi: 10.1128/IAI.71.5.2775-2786.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 614.Okada N, Sasakawa C, Tobe T. Virulence associated chromosomal loci of S. flexneri identified by random Tn5 insertion mutagenesis. Mol Microbiol. 1991;5:187. doi: 10.1111/j.1365-2958.1991.tb01839.x. [DOI] [PubMed] [Google Scholar]
- 615.Okamura N, Nagai T, Nakaya R. HeLa cell invasiveness and O antigen of Shigella flexneri as separate and prerequisite attributes of virulence to evoke keratoconjunctivitis in guinea pigs. Infect Immun. 1983;39:505. doi: 10.1128/iai.39.2.505-513.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 616.Bartlett AV, Prado D, Cleary TG. Production of Shiga toxin and other cytotoxins by serogroups of Shigella. J Infect Dis. 1986;154:996. doi: 10.1093/infdis/154.6.996. [DOI] [PubMed] [Google Scholar]
- 617.Olenick JG, Wolfe AD. Shigella toxin inhibition of binding and translation of polyuridylic acid by Escherichia coli ribosomes. J Bacteriol. 1980;141:1246. doi: 10.1128/jb.141.3.1246-1250.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 618.Brown JE, Rothman SW, Doctor BP. Inhibition of protein synthesis in intact HeLa cells by Shigella dysenteriae 1 toxin. Infect Immun. 1980;29:98. doi: 10.1128/iai.29.1.98-107.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 619.Al-Hasani K, Henderson IR, Sakellaris H. The sigA Gene which is borne on the she pathogenicity island of Shigella flexneri 2a encodes an exported cytopathic protease involved in intestinal fluid accumulation. Infect Immun. 2000;68:2457. doi: 10.1128/iai.68.5.2457-2463.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 620.Prado D, Cleary TG, Pickering LK. The relation between production of cytotoxin and clinical features in shigellosis. J Infect Dis. 1986;154:149. doi: 10.1093/infdis/154.1.149. [DOI] [PubMed] [Google Scholar]
- 621.Makintubee S, Mallonee J, Istre GR. Shigellosis outbreak associated with swimming. Am J Public Health. 1987;77:166. doi: 10.2105/ajph.77.2.166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 622.Levine MM, DuPont HL, Formal SB. Pathogenesis of Shigella dysenteriae 1 (Shiga) dysentery. J Infect Dis. 1973;127:261. doi: 10.1093/infdis/127.3.261. [DOI] [PubMed] [Google Scholar]
- 623.DuPont HL, Hornick RB, Dawkins AT. The response of man to virulent Shigella flexneri 2a. J Infect Dis. 1969;119:296. doi: 10.1093/infdis/119.3.296. [DOI] [PubMed] [Google Scholar]
- 624.Levine MM. Shigella infections and vaccines: experiences from volunteer and controlled field studies. In: Rahaman MM, Greenough WB, Novak NR, editors. Shigellosis: A Continuing Global Problem. International Centre for Diarrhoeal Disease Research; Dacca, Bangladesh: 1983. p. 208. [Google Scholar]
- 625.Pickering LK, Hadler SC. Management and prevention of infectious diseases in day care. In: Feigin RD, Cherry JC, editors. Textbook of Pediatric Infectious Diseases. WB Saunders; Philadelphia: 1992. p. 2308. [Google Scholar]
- 626.Mata LG. The Children of Santa Maria Cauque: A Prospective Field Study of Health and Growth. MIT Press; Cambridge, Mass: 1978. [Google Scholar]
- 627.Stoll BJ, Glass RI, Huq MI. Surveillance of patients attending a diarrhoeal disease hospital in Bangladesh. BMJ. 1982;285:1185. doi: 10.1136/bmj.285.6349.1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 628.Floyd T, Higgins AR, Kader M. A. Studies in shigellosis. V. The relationship of age to the incidence of Shigella infections in Egyptian children, with special reference to shigellosis in the newborn and infant in the first six months of life. Am J Trop Med Hyg. 1956;5:119. doi: 10.4269/ajtmh.1956.5.119. [DOI] [PubMed] [Google Scholar]
- 629.Summary of notifiable diseases, United States, 1981. MMWR Morb Mortal Wkly Rep. 1992;40:10. [PubMed] [Google Scholar]
- 630.Clemens JS, Stanton B, Stoll B. Breast-feeding as a determinant of severity in shigellosis. Am J Epidemiol. 1986;123:710. doi: 10.1093/oxfordjournals.aje.a114291. [DOI] [PubMed] [Google Scholar]
- 631.Mata LJ, Urrutia JM, Garcia B. Shigella infections in breast fed Guatemalan Indian neonates. Am J Dis Child. 1969;117:142. doi: 10.1001/archpedi.1969.02100030144004. [DOI] [PubMed] [Google Scholar]
- 632.Huskins WC, Griffiths JK, Faruque AS, Bennish ML. Shigellosis in neonates and young infants. J Pediatr. 1994;125:14. doi: 10.1016/s0022-3476(94)70115-6. [DOI] [PubMed] [Google Scholar]
- 633.Haltalin KC. Neonatal shigellosis. Am J Dis Child. 1967;114:603. doi: 10.1001/archpedi.1967.02090270059004. [DOI] [PubMed] [Google Scholar]
- 634.Scragg JN, Rubidge CJ, Appelbaum PC. Shigella infection in African and Indian children with special reference to Shigella septicemia. J Pediatr. 1978;93:796. doi: 10.1016/s0022-3476(78)81081-6. [DOI] [PubMed] [Google Scholar]
- 635.Burry VF, Thurn AN, Co TG. Shigellosis: an analysis of 239 cases in a pediatric population. Mo Med. 1968;65:671. [PubMed] [Google Scholar]
- 636.Enteric infection due to Campylobacter, Yersinia, Salmonella and Shigella. Bull World Health Organ. 1980;58:519. [PMC free article] [PubMed] [Google Scholar]
- 637.Kraybill EN, Controni G. Septicemia and enterocolitis due to S. sonnei in a newborn infant. Pediatrics. 1968;42:529. [PubMed] [Google Scholar]
- 638.Moore EE. Shigella sonnei septicemia in a neonate. BMJ. 1974;1:22. doi: 10.1136/bmj.1.5896.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 639.Aldrich JA, Flowers RP, Hall FK. S. sonnei septicemia in a neonate: a case report. J Am Osteopath Assoc. 1979;79:93. [PubMed] [Google Scholar]
- 640.Barton LL, Pickering LK. Shigellosis in the first week of life. Pediatrics. 1973;52:437. [PubMed] [Google Scholar]
- 641.Landsberger M. Bacillary dysentery in a newborn infant. Arch Pediatr. 1942;59:330. [Google Scholar]
- 642.Neter E. S. sonnei infection at term and its transfer to the newborn. Obstet Gynecol. 1961;17:517. [PubMed] [Google Scholar]
- 643.McIntire MS, Jahr HM. An isolated case of shigellosis in the newborn nursery. Nebr State Med J. 1954;39:425. [PubMed] [Google Scholar]
- 644.Greenberg M, Frant S, Shapiro R. Bacillary dysentery acquired at birth. J Pediatr. 1940;17:363. [Google Scholar]
- 645.Emanuel B, Sherman JO. Shigellosis in a neonate. Clin Pediatr. 1975;14:725. doi: 10.1177/000992287501400806. [DOI] [PubMed] [Google Scholar]
- 646.Barret-Connor E, Connor JD. Extraintestinal manifestations of shigellosis. Am J Gastroenterol. 1970;53:234. [PubMed] [Google Scholar]
- 647.Fischler E. Convulsions as a complication of shigellosis in children. Helv Paediatr Acta. 1962;4:389. [PubMed] [Google Scholar]
- 648.Ashkenazi S, Dinari G, Zevalunov A. Convulsions in childhood shigellosis. Am J Dis Child. 1987;141:208. doi: 10.1001/archpedi.1987.04460020098036. [DOI] [PubMed] [Google Scholar]
- 649.Whitfield C, Humphries JM. Meningitis and septicemia due to Shigella in a newborn infant. J Pediatr. 1967;70:805. doi: 10.1016/s0022-3476(67)80335-4. [DOI] [PubMed] [Google Scholar]
- 650.Goren A, Freier S, Passwell JH. Lethal toxic encephalopathy due to childhood shigellosis in a developed country. Pediatrics. 1992;89:1189. [PubMed] [Google Scholar]
- 651.Ashkenazi S, Cleary KR, Pickering LK. The association of Shiga toxin and other cytotoxins with the neurologic manifestations of shigellosis. J Infect Dis. 1990;161:961. doi: 10.1093/infdis/161.5.961. [DOI] [PubMed] [Google Scholar]
- 652.Rahaman MM, Jamiul Alam AKM, Islam MR. Shiga bacillus dysentery associated with marked leukocytosis and erythrocyte fragmentation. Johns Hopkins Med J. 1975;136:65. [PubMed] [Google Scholar]
- 653.Neglia TG, Marr TJ, Davis AT. Shigella dysentery with secondary Klebsiella sepsis. J Pediatr. 1976;63:253. doi: 10.1016/s0022-3476(76)80458-1. [DOI] [PubMed] [Google Scholar]
- 654.Struelens MJ, Patte D, Kabir I. Shigella septicemia: prevalence, presentation, risk factors, and outcome. J Infect Dis. 1985;152:784. doi: 10.1093/infdis/152.4.784. [DOI] [PubMed] [Google Scholar]
- 655.Haltalin KC, Nelson JD. Coliform septicemia complicating shigellosis in children. JAMA. 1965;192:441. doi: 10.1001/jama.1965.03080190007001. [DOI] [PubMed] [Google Scholar]
- 656.Levin SE. Shigella septicemia in the newborn infant. J Pediatr. 1967;71:917. doi: 10.1016/s0022-3476(67)80027-1. [DOI] [PubMed] [Google Scholar]
- 657.Martin T, Habbick BF, Nyssen J. Shigellosis with bacteremia: a report of two cases and a review of the literature. Pediatr Infect Dis J. 1983;2:21. [PubMed] [Google Scholar]
- 658.Raderman JW, Stoller KP, Pomerance JJ. Blood-stream invasion with S. sonnei in an asymptomatic newborn infant. Pediatr Infect Dis J. 1986;5:379. doi: 10.1097/00006454-198605000-00024. [DOI] [PubMed] [Google Scholar]
- 659.Starke JR, Baker CJ. Neonatal shigellosis with bowel perforation. Pediatr Infect Dis J. 1985;4:405. doi: 10.1097/00006454-198507000-00016. [DOI] [PubMed] [Google Scholar]
- 660.Azad MAK, Islam M. Colonic perforation in Shigella dysenteriae 1 infection. Pediatr Infect Dis J. 1986;5:103. doi: 10.1097/00006454-198601000-00019. [DOI] [PubMed] [Google Scholar]
- 661.O’Connor JH, O’Callaghan U. Fatal S. sonnei septicemia in an adult complemented by marrow aplasia and intestinal perforation. J Infect. 1981;3:277. doi: 10.1016/s0163-4453(81)90980-4. [DOI] [PubMed] [Google Scholar]
- 662.Alam AN, Chowdhurg AAKM, Kabir IAKM. Association of pneumonia with under-nutrition and shigellosis. Indian Pediatr. 1984;21:609. [PubMed] [Google Scholar]
- 663.Hoefnagel D. Fulminating, rapidly fatal shigellosis in children. N Engl J Med. 1958;258:1256. doi: 10.1056/NEJM195806192582506. [DOI] [PubMed] [Google Scholar]
- 664.Sakamoto A, Kamo S. Clinical, statistical observations on Ekiri and bacillary dysentery. A study of 785 cases. Ann Paediatr. 1956;186:1. [PubMed] [Google Scholar]
- 665.Dodd K, Buddingh GJ, Rapoport S. The etiology of Ekiri, a highly fatal disease of Japanese children. Pediatrics. 1949;3:9. [PubMed] [Google Scholar]
- 666.Davis TC. Chronic vulvovaginitis in children due to S. flexneri. Pediatrics. 1975;56:41. [PubMed] [Google Scholar]
- 667.Murphy TV, Nelson JD. Shigella vaginitis: report on 38 patients and review of the literature. Pediatrics. 1979;63:511. [PubMed] [Google Scholar]
- 668.Tobias JD, Starke JR, Tosi MF. Shigella keratitis: a report of two cases and a review of the literature. Pediatr Infect Dis J. 1987;6:79. [PubMed] [Google Scholar]
- 669.Butler T, Dunn D, Dahms B. Causes of death and the histopathologic findings in fatal shigellosis. Pediatr Infect Dis J. 1989;8:767. doi: 10.1097/00006454-198911000-00008. [DOI] [PubMed] [Google Scholar]
- 670.Bennish ML, Harris JR, Wojtyniak BJ. Death in shigellosis: incidence and risk factors in hospitalized patients. J Infect Dis. 1990;161:500. doi: 10.1093/infdis/161.3.500. [DOI] [PubMed] [Google Scholar]
- 671.Speelman P, McGlaughlin R, Kabir I. Differential clinical features and stool findings in shigellosis and amoebic dysentery. Trans R Soc Trop Med Hyg. 1987;81:549. doi: 10.1016/0035-9203(87)90402-0. [DOI] [PubMed] [Google Scholar]
- 672.Taylor WI, Harris B. Isolation of shigellae. II. Comparison of plating media and enrichment broths. Am J Clin Pathol. 1965;44:476. [PubMed] [Google Scholar]
- 673.Stypulkowska-Misiurewics H. Problems in bacteriological diagnosis of shigellosis. In: Rahaman MM, Greenough WB, Novak NR, editors. Shigellosis: A Continuing Global Problem. International Centre for Diarrhoeal Disease Research; Dacca, Bangladesh: 1983. p. 87. [Google Scholar]
- 674.Haltalin KC, Nelson JD, Ring R., III Double-blind treatment study of shigellosis comparing ampicillin, sulfadiazine, and placebo. J Pediatr. 1967;70:970. doi: 10.1016/s0022-3476(67)80275-0. [DOI] [PubMed] [Google Scholar]
- 675.Haltalin KC, Nelson JD, Kusmiesz HT. Optimal dosage of ampicillin in shigellosis. J Pediatr. 1969;74:626. doi: 10.1016/s0022-3476(69)80051-x. [DOI] [PubMed] [Google Scholar]
- 676.Haltalin KC, Nelson JD, Kusmiesz HT. Comparative efficacy of nalidixic acid and ampicillin for severe shigellosis. Arch Dis Child. 1973;48:305. doi: 10.1136/adc.48.4.305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 677.Oaks EV, Hale TL, Formal SB. Serum immune response to Shigella protein antigens in rhesus monkeys and humans infected with Shigella spp. Infect Immun. 1986;53:57. doi: 10.1128/iai.53.1.57-63.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 678.Frankel G, Riley L, Giron JA. Detection of Shigella in feces using DNA amplification. J Infect Dis. 1990;161:1252. doi: 10.1093/infdis/161.6.1252. [DOI] [PubMed] [Google Scholar]
- 679.Speelman P, Kabir I, Islam M. Distribution and spread of colonic lesions in shigellosis: a colonoscopic study. J Infect Dis. 1984;150:899. doi: 10.1093/infdis/150.6.899. [DOI] [PubMed] [Google Scholar]
- 680.Formal SB, Kent TH, Austin S. Fluorescent antibody and histological studies of vaccinated control monkeys challenged with Shigella flexneri. J Bacteriol. 1966;91:2368. doi: 10.1128/jb.91.6.2368-2376.1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 681.LaBrec EH, Formal SB. Experimental Shigella infections. IV. Fluorescent antibody studies of an infection in guinea pigs. J Immunol. 1961;87:562. [PubMed] [Google Scholar]
- 682.Frost JA, Rowe B, Vandepitte J. Plasmid characterization in the investigation of an epidemic caused by multiply resistant S. dysenteriae type 1 in Central Africa. Lancet. 1981;2:1074. doi: 10.1016/s0140-6736(81)91277-0. [DOI] [PubMed] [Google Scholar]
- 683.Haider K, Hug MI, Samadi AR. Plasmid characterization of Shigella spp. isolated from children with shigellosis and asymptomatic excretors. J Antimicrob Chemother. 1985;16:691. doi: 10.1093/jac/16.6.691. [DOI] [PubMed] [Google Scholar]
- 684.Tauxe RV, Puhr ND, Wells JG. Antimicrobial resistance of Shigella isolates in the USA: the importance of international travelers. J Infect Dis. 1990;162:1107. doi: 10.1093/infdis/162.5.1107. [DOI] [PubMed] [Google Scholar]
- 685.Salzman TC, Scher CD, Moss R. Shigellae with transferable drug resistance: outbreak in a nursery for premature infants. J Pediatr. 1967;71:21. doi: 10.1016/s0022-3476(67)80225-7. [DOI] [PubMed] [Google Scholar]
- 686.Bennish ML, Salam MA, Hossain MA. Antimicrobial resistance of Shigella isolates in Bangladesh, 1983–1990: increasing frequency of strains multiply resistant to ampicillin, trimethoprim/sulfamethoxazole, and nalidixic acid. Clin Infect Dis. 1992;14:1055. doi: 10.1093/clinids/14.5.1055. [DOI] [PubMed] [Google Scholar]
- 687.Ostrower VG. Comparison of cefaclor and ampicillin in the treatment of shigellosis. Postgrad Med J. 1979;55:82. [PubMed] [Google Scholar]
- 688.Haltalin KC, Nelson JD. Failure of furazolidone therapy on shigellosis. Am J Dis Child. 1972;123:40. doi: 10.1001/archpedi.1972.02110070090011. [DOI] [PubMed] [Google Scholar]
- 689.Nelson JD, Haltalin KC. Comparative efficacy of cephalexin and ampicillin for shigellosis and other types of acute diarrhea in infants and children. Antimicrob Agents Chemother. 1975;7:415. doi: 10.1128/aac.7.4.415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 690.Nelson JD, Haltalin KC. Amoxicillin less effective than ampicillin against Shigella in vitro and in vivo: relationship of efficacy to activity in serum. J Infect Dis. 1974;129:S222. doi: 10.1093/infdis/129.supplement_2.s222. [DOI] [PubMed] [Google Scholar]
- 691.Tong MJ, Martin DG, Cunningham JJ. Clinical and bacteriological evaluation of antibiotic treatment in shigellosis. JAMA. 1970;214:1841. [PubMed] [Google Scholar]
- 692.Orenstein WA, Ross L, Overturf GD. Antibiotic treatment of acute shigellosis: failure of cefamandole compared to trimethoprim/sulfamethoxazole and ampicillin. Am J Med Sci. 1981;282:27. doi: 10.1097/00000441-198107000-00004. [DOI] [PubMed] [Google Scholar]
- 693.Yunus MD, Rahaman MM, Faruque ASG. Comparative treatment of shigellosis with trimethoprim/sulfamethoxazole and ampicillin. In: Rahaman MM, Greenough WB, Novak NR, editors. Shigellosis: A Continuing Global Problem. International Centre for Diarrhoeal Disease Research; Dacca, Bangladesh: 1983. p. 166. [Google Scholar]
- 694.Nelson JD, Kusmiesz H, Jackson LH. Trimethoprim/sulfamethoxazole therapy for shigellosis. JAMA. 1976;235:1239. [PubMed] [Google Scholar]
- 695.Nelson JD, Kusmiesz H, Jackson LH. Comparison of trimethoprim/sulfamethoxazole and ampicillin for shigellosis in ambulatory patients. J Pediatr. 1976;89:491. doi: 10.1016/s0022-3476(76)80561-6. [DOI] [PubMed] [Google Scholar]
- 696.Nelson JD, Kusmiesz H, Shelton S. Oral or intravenous trimethoprim/sulfamethoxazole therapy for shigellosis. Rev Infect Dis J. 1982;4:546. doi: 10.1093/clinids/4.2.546. [DOI] [PubMed] [Google Scholar]
- 697.Gilman RH, Spira W, Rabbani H. Single dose ampicillin therapy for severe shigellosis in Bangladesh. J Infect Dis. 1981;143:164. doi: 10.1093/infdis/143.2.164. [DOI] [PubMed] [Google Scholar]
- 698.Varsano I, Elditz-Marcus T, Nussinovitch M. Comparative efficacy of ceftriaxone and ampicillin for treatment of severe shigellosis in children. J Pediatr. 1991;118:627. doi: 10.1016/s0022-3476(05)83392-x. [DOI] [PubMed] [Google Scholar]
- 699.Kabir T, Butler T, Khanam A. Comparative efficacies of single intravenous doses of ceftriaxone and ampicillin for shigellosis in a placebo-controlled trial. Antimicrob Agents Chemother. 1986;29:645. doi: 10.1128/aac.29.4.645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 700.Bennish ML, Salam MA, Khan WA. Treatment of shigellosis. III. Comparison of one- or two-dose ciprofloxacin with standard 5-day therapy. Ann Intern Med. 1992;117:727. doi: 10.7326/0003-4819-117-9-727. [DOI] [PubMed] [Google Scholar]
- 701.John JF, Jr, Atkins LT, Maple PAH. Activities of new fluoroquinolones against Shigella sonnei. Antimicrob Agents Chemother. 1992;36:2346. doi: 10.1128/aac.36.10.2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 702.DuPont HL, Hornick RB. Adverse effect of Lomotil therapy in shigellosis. JAMA. 1973;226:1525. [PubMed] [Google Scholar]
- 703.Ahmed F, Clemens JD, Rao MR. Community based evaluation of the effect of breast feeding on the risk of microbiologically confirmed or clinically presumptive shigellosis in Bangladeshi children. Pediatrics. 1992;90:406. [PubMed] [Google Scholar]
- 704.Khan MU. Interruption of shigellosis by handwashing. Trans R Soc Trop Med Hyg. 1982;76:164. doi: 10.1016/0035-9203(82)90266-8. [DOI] [PubMed] [Google Scholar]
- 705.Farrar WE, Edison M, Guerry P. Interbacterial transfer of R-factor in the human intestine: in vitro acquisition of R-factor mediated kanamycin resistance by a multi-resistant strain of S. sonnei. J Infect Dis. 1972;126:27. doi: 10.1093/infdis/126.1.27. [DOI] [PubMed] [Google Scholar]
- 706.McFadyean F, Stockman S. vol 3. His Majesty’s Stationery Office; London: 1909. (Report of the Departmental Committee Appointed by the Board of Agriculture and Fisheries to Inquire into Epizootic Abortion). [Google Scholar]
- 707.Smith T, Taylor MS. Some morphological and biochemical characters of the spirilla (Vibrio fetus, n. spp.) associated with disease of the fetal membranes in cattle. J Exp Med. 1919;30:200. doi: 10.1084/jem.30.4.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 708.Jones FS, Orcutt M, Little RB. Vibrios (Vibrio jejuni, n. spp.) associated with intestinal disorders of cows and calves. J Exp Med. 1931;53:853. doi: 10.1084/jem.53.6.853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 709.Bryner JH, Estes PC, Foley JW. Infectivity of three Vibrio fetus biotypes for gallbladder and intestines of cattle, sheep, rabbits, guinea pigs, and mice. Am J Vet Res. 1971;32:465. [PubMed] [Google Scholar]
- 710.Vinzent R, Dumas J, Picard N. Septicémie grave au cours de la grossesse due à un vibrion. Avortement consécutif. Bull Acad Natl Med. 1947;131:90. [Google Scholar]
- 711.Eden AN. Perinatal mortality caused by Vibrio fetus: review and analysis. J Pediatr. 1966;68:297. [Google Scholar]
- 712.Torphy DE, Bond WW. Campylobacter fetus infections in children. Pediatrics. 1979;64:898. [PubMed] [Google Scholar]
- 713.Bokkenheuser V. Vibrio fetus infection in man. I. Ten new cases and some epidemiologic observations. Am J Epidemiol. 1970;91:400. doi: 10.1093/oxfordjournals.aje.a121150. [DOI] [PubMed] [Google Scholar]
- 714.Guerrant RL, Lahita RG, Winn WC. Campylobacteriosis in man: pathogenic mechanisms and review of 91 bloodstream infections. Am J Med. 1978;65:584. doi: 10.1016/0002-9343(78)90845-8. [DOI] [PubMed] [Google Scholar]
- 715.Sebald M, Veron M. Teneur en bases de l’ADN et classification des vibrions. Ann Inst Pasteur. 1963;105:897. [PubMed] [Google Scholar]
- 716.Butzler JP, Dekeyser P, Detrain M. Related vibrio in stools. J Pediatr. 1973;82:493. doi: 10.1016/s0022-3476(73)80131-3. [DOI] [PubMed] [Google Scholar]
- 717.Skirrow MB. Campylobacter enteritis: a “new” disease. BMJ. 1977;2:9. doi: 10.1136/bmj.2.6078.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 718.Communicable Disease Surveillance Centre and the Communicable Diseases (Scotland) Unit Campylobacter infections in Britain 1977. BMJ. 1978;1:1357. [PMC free article] [PubMed] [Google Scholar]
- 719.De Mol P, Bosmans E. Campylobacter enteritis in Central Africa. Lancet. 1978;1:604. doi: 10.1016/s0140-6736(78)91044-9. [DOI] [PubMed] [Google Scholar]
- 720.Lindquist B, Kjellander J, Kosunen T. Campylobacter enteritis in Sweden. BMJ. 1978;1:303. doi: 10.1136/bmj.1.6108.303-d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 721.Karmali MA, Fleming PC. Campylobacter enteritis in children. J Pediatr. 1979;94:527. doi: 10.1016/s0022-3476(79)80004-9. [DOI] [PubMed] [Google Scholar]
- 722.Pai CM, Sorger S, Lackman L. Campylobacter gastroenteritis in children. J Pediatr. 1979;94:589. doi: 10.1016/s0022-3476(79)80016-5. [DOI] [PubMed] [Google Scholar]
- 723.Blaser MJ, Reller LB. Campylobacter enteritis. N Engl J Med. 1981;305:1444. doi: 10.1056/NEJM198112103052404. [DOI] [PubMed] [Google Scholar]
- 724.Dekeyser P, Gossuin-Detrain M, Butzler JP. Acute enteritis due to related Vibrio: first positive stool cultures. J Infect Dis. 1972;125:390. doi: 10.1093/infdis/125.4.390. [DOI] [PubMed] [Google Scholar]
- 725.Butzler JP, Skirrow MB. Campylobacter enteritis. Clin Gastroenterol. 1979;8:737. [PubMed] [Google Scholar]
- 726.Blaser MJ, Berkowitz ID, LaForce FM. Campylobacter enteritis: clinical and epidemiologic features. Ann Intern Med. 1979;91:179. doi: 10.7326/0003-4819-91-2-179. [DOI] [PubMed] [Google Scholar]
- 727.Karmali MA, Fleming PC. Campylobacter enteritis. Can Med Assoc J. 1979;120:1525. [PMC free article] [PubMed] [Google Scholar]
- 728.Steele TW, McDermott S. Campylobacter enteritis in South Australia. Med J Aust. 1978;2:404. doi: 10.5694/j.1326-5377.1978.tb76814.x. [DOI] [PubMed] [Google Scholar]
- 729.Guandalini S, Cucchiara S, deRitis G. Campylobacter colitis in infants. J Pediatr. 1983;102:72. doi: 10.1016/s0022-3476(83)80291-1. [DOI] [PubMed] [Google Scholar]
- 730.Walker RI, Caldwell MB, Lee EC. Pathophysiology of Campylobacter enteritis. Microbiol Rev. 1986;50:81. doi: 10.1128/mr.50.1.81-94.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 731.Penner JL. The genus Campylobacter: a decade of progress. Clin Microbiol Rev. 1988;1:157. doi: 10.1128/cmr.1.2.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 732.Calva JJ, Ruiz-Palacios GM, Lopez-Vidal AB. Cohort study of intestinal infection with Campylobacter in Mexican children. Lancet. 1988;1:503. doi: 10.1016/s0140-6736(88)91297-4. [DOI] [PubMed] [Google Scholar]
- 733.Figura N, Guglielmetti P, Zanchi A. Two cases of Campylobacter mucosalis enteritis in children. J Clin Microbiol. 1993;31:727. doi: 10.1128/jcm.31.3.727-728.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 734.Harvey SM, Greenwood JR. Relationships among catalase-positive campylobacters determined by deoxyribonucleic acid-deoxyribonucleic acid hybridization. Int J Syst Bacteriol. 1983;33:275. [Google Scholar]
- 735.Owen RJ. Nucleic acids in the classification of campylobacters. Eur J Clin Microbiol. 1983;2:367. doi: 10.1007/BF02019473. [DOI] [PubMed] [Google Scholar]
- 736.Hoffer MA. Bovine campylobacteriosis: a review. Can Vet J. 1981;22:327. [PMC free article] [PubMed] [Google Scholar]
- 737.Grant CA. Bovine vibriosis: a brief review. Can J Comp Med. 1955;19:156. [PMC free article] [PubMed] [Google Scholar]
- 738.Vinzent R. Une affection méconnue de la grossesse: l’infection placentaire à “Vibrio fetus.”. Presse Med. 1949;57:1230. [PubMed] [Google Scholar]
- 739.Wong S-N, Tam Y-CA, Yeun K-Y. Campylobacter infection in the neonate: case report and review of the literature. Pediatr Infect Dis J. 1990;9:665. [PubMed] [Google Scholar]
- 740.Hood M, Todd JM. Vibrio fetus—a cause of human abortion. Am J Obstet Gynecol. 1960;80:506. doi: 10.1016/s0002-9378(16)36505-x. [DOI] [PubMed] [Google Scholar]
- 741.van Wering RF, Esseveld H. Vibrio fetus. Ned Tijdschr Geneeskd. 1963;107:119. [PubMed] [Google Scholar]
- 742.Burgert W, Jr, Hagstrom JWC. Vibrio fetus meningoencephalitis. Arch Neurol. 1964;10:196. doi: 10.1001/archneur.1964.00460140082010. [DOI] [PubMed] [Google Scholar]
- 743.Willis MD, Austin WJ. Human Vibrio fetus infection: report of two dissimilar cases. Am J Dis Child. 1966;112:459. doi: 10.1001/archpedi.1966.02090140131012. [DOI] [PubMed] [Google Scholar]
- 744.Smith JP, Marymont JH, Jr, Schweers J. Septicemia due to Campylobacter fetus in a newborn infant with gastroenteritis. Am J Med Technol. 1977;43:38. [PubMed] [Google Scholar]
- 745.West SE, Houghton DJ, Crock S. Campylobacter spp. isolated from the cervix during septic abortion. Case report. Br J Obstet Gynaecol. 1982;89:771. doi: 10.1111/j.1471-0528.1982.tb05108.x. [DOI] [PubMed] [Google Scholar]
- 746.Lee MM, Welliver RC, LaScolea LJ. Campylobacter meningitis in childhood. Pediatr Infect Dis J. 1985;4:544. doi: 10.1097/00006454-198509000-00021. [DOI] [PubMed] [Google Scholar]
- 747.Simor AE, Karmali MA, Jadavji T. Abortion and perinatal sepsis associated with Campylobacter infection. Rev Infect Dis J. 1986;8:397. doi: 10.1093/clinids/8.3.397. [DOI] [PubMed] [Google Scholar]
- 748.Forbes JC, Scheifele DW. Early onset Campylobacter sepsis in a neonate. Pediatr Infect Dis J. 1987;6:494. [PubMed] [Google Scholar]
- 749.King EO. Human infections with Vibrio fetus and a closely related vibrio. J Infect Dis. 1957;101:119. doi: 10.1093/infdis/101.2.119. [DOI] [PubMed] [Google Scholar]
- 750.Francioli P, Herzstein J, Grobe JP. Campylobacter fetus subspecies fetus bacteremia. Arch Intern Med. 1985;145:289. [PubMed] [Google Scholar]
- 751.Karmali MA, Penner JL, Fleming PC. The biotype and biotype distribution of clinical isolates of Campylobacter jejuni and Campylobacter coli over a three-year period. J Infect Dis. 1983;147:243. doi: 10.1093/infdis/147.2.243. [DOI] [PubMed] [Google Scholar]
- 752.Albert MJ, Leach A, Asche V. Serotype distribution of Campylobacter jejuni and Campylobacter coli isolated from hospitalized patients with diarrhea in Central Australia. J Clin Microbiol. 1992;30:207. doi: 10.1128/jcm.30.1.207-210.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 753.Riley LW, Finch MJ. Results of the first year of national surveillance of Campylobacter infections in the United States. J Infect Dis. 1985;151:956. doi: 10.1093/infdis/151.5.956. [DOI] [PubMed] [Google Scholar]
- 754.Georges-Courbot MC, Baya C, Beraud AM. Distribution and serotypes of Campylobacter jejuni and Campylobacter coli in enteric Campylobacter strains isolated from children in the Central African Republic. J Clin Microbiol. 1986;23:592. doi: 10.1128/jcm.23.3.592-594.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 755.Wheeler WE, Borchers J. Vibrionic enteritis in infants. Am J Dis Child. 1961;101:60. doi: 10.1001/archpedi.1961.04020020062010. [DOI] [PubMed] [Google Scholar]
- 756.Middlekamp JN, Wolf HA. Infection due to a “related” Vibrio. J Pediatr. 1961;59:318. doi: 10.1016/s0022-3476(61)80283-7. [DOI] [PubMed] [Google Scholar]
- 757.Ruben FL, Wolinsky E. Human infection with Vibrio fetus. In: Hobby GL, editor. Antimicrobial Agents and Chemotherapy. American Society for Microbiology; Bethesda, Md: 1967. p. 143. [PubMed] [Google Scholar]
- 758.Mawer SL, Smith BAM. Campylobacter infection of premature baby. Lancet. 1979;1:1041. doi: 10.1016/s0140-6736(79)92803-4. [DOI] [PubMed] [Google Scholar]
- 759.Campylobacter in a mother and baby. Commun Dis Rep CDR. 1979;7917:4. [Google Scholar]
- 760.Karmali MA, Tan YC. Neonatal Campylobacter enteritis. Can Med Assoc J. 1980;122:192. [PMC free article] [PubMed] [Google Scholar]
- 761.Thomas K, Chan KN, Riberiro CD. Campylobacter jejuni/coli meningitis in a neonate. BMJ. 1980;280:1301. doi: 10.1136/bmj.280.6227.1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 762.Anders BJ, Lauer BA, Paisley JW. Campylobacter gastroenteritis in neonates. Am J Dis Child. 1981;135:900. doi: 10.1001/archpedi.1981.02130340012005. [DOI] [PubMed] [Google Scholar]
- 763.Vesikari T, Huttunen L, Maki R. Perinatal Campylobacter fetus ss. jejuni enteritis. Acta Paediatr Scand. 1981;70:261. doi: 10.1111/j.1651-2227.1981.tb05554.x. [DOI] [PubMed] [Google Scholar]
- 764.Miller RC, Guard RW. A case of premature labour due to Campylobacter jejuni infection. Aust N Z Obstet Gynaecol. 1982;22:118. doi: 10.1111/j.1479-828x.1982.tb01420.x. [DOI] [PubMed] [Google Scholar]
- 765.Buck GE, Kelly MT, Pichanick AM. Campylobacter jejuni in newborns: a cause of asymptomatic bloody diarrhea. Am J Dis Child. 1982;136:744. doi: 10.1001/archpedi.1982.03970440088027. [DOI] [PubMed] [Google Scholar]
- 766.Karmali MA, Norrish B, Lior H. Campylobacter enterocolitis in a neonatal nursery. J Infect Dis. 1984;149:874. doi: 10.1093/infdis/149.6.874. [DOI] [PubMed] [Google Scholar]
- 767.Youngs ER, Roberts C, Davidson DC. Campylobacter enteritis and bloody stools in the neonate. Arch Dis Child. 1985;60:480. doi: 10.1136/adc.60.5.480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 768.Terrier A, Altwegg M, Bader P. Hospital epidemic of neonatal Campylobacter jejuni infection. Lancet. 1985;2:1182. doi: 10.1016/s0140-6736(85)92698-4. [DOI] [PubMed] [Google Scholar]
- 769.Goossens H, Henocque G, Kremp L. Nosocomial outbreak of Campylobacter jejuni meningitis in newborn infants. Lancet. 1986;2:146. doi: 10.1016/s0140-6736(86)91956-2. [DOI] [PubMed] [Google Scholar]
- 770.DiNicola AF. Campylobacter jejuni diarrhea in a 3-day old male neonate. Pediatr Forum. 1986;140:191. doi: 10.1001/archpedi.1986.02140170017013. [DOI] [PubMed] [Google Scholar]
- 771.Hershkowici S, Barak M, Cohen A. An outbreak of Campylobacter jejuni infection in a neonatal intensive care unit. J Hosp Infect. 1987;9:54. doi: 10.1016/0195-6701(87)90096-x. [DOI] [PubMed] [Google Scholar]
- 772.Gribble MJ, Salit IE, Isaac-Renton J. Campylobacter infections in pregnancy: case report and literature review. Am J Obstet Gynecol. 1981;140:423. doi: 10.1016/0002-9378(81)90038-7. [DOI] [PubMed] [Google Scholar]
- 773.Gilbert GL, Davoren RA, Cole ME. Midtrimester abortion associated with septicaemia caused by Campylobacter jejuni. Med J Aust. 1981;1:585. doi: 10.5694/j.1326-5377.1981.tb135843.x. [DOI] [PubMed] [Google Scholar]
- 774.Jost PM, Galvin MC, Brewer JH. Campylobacter septic abortion. South Med J. 1984;77:924. doi: 10.1097/00007611-198407000-00033. [DOI] [PubMed] [Google Scholar]
- 775.Pearce CT. Campylobacter jejuni infection as a cause of septic abortion. Aust J Med Lab Sci. 1981;2:107. [Google Scholar]
- 776.Pines A, Goldhammer E, Bregman J. Campylobacter enteritis associated with recurrent abortions in agammaglobulinemia. Acta Obstet Gynecol Scand. 1983;62:279. doi: 10.3109/00016348309155809. [DOI] [PubMed] [Google Scholar]
- 777.Kist M, Keller KM, Niebling W. Campylobacter coli septicaemia associated with septic abortion. Infection. 1984;12:88. doi: 10.1007/BF01641678. [DOI] [PubMed] [Google Scholar]
- 778.Reina J, Borrell N, Fiol M. Rectal bleeding caused by Campylobacter jejuni in a neonate. Pediatr Infect Dis J. 1992;6:500. [PubMed] [Google Scholar]
- 779.Ruiz-Palacios GM, Torres J, Escamilla NI. Cholera-like enterotoxin produced by Campylobacter jejuni: characterization and clinical significance. Lancet. 1983;2:250. doi: 10.1016/s0140-6736(83)90234-9. [DOI] [PubMed] [Google Scholar]
- 780.Johnson WM, Lior H. Toxins produced by Campylobacter jejuni and Campylobacter coli. Lancet. 1984;1:229. doi: 10.1016/s0140-6736(84)92155-x. [DOI] [PubMed] [Google Scholar]
- 781.Guerrant RL, Wanke CA, Pennie RA. Production of a unique cytotoxin by Campylobacter jejuni. Infect Immun. 1987;55:2526. doi: 10.1128/iai.55.10.2526-2530.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 782.McCardell BA, Madden JM, Lee EC. Campylobacter jejuni and Campylobacter coli production of a cytotonic toxin immunologically similar to cholera toxin. J Food Prot. 1984;47:943. doi: 10.4315/0362-028X-47.12.943. [DOI] [PubMed] [Google Scholar]
- 783.Klipstein FA, Engert RF. Purification of Campylobacter jejuni enterotoxin. Lancet. 1984;1:1123. doi: 10.1016/s0140-6736(84)92536-4. [DOI] [PubMed] [Google Scholar]
- 784.Klipstein FA, Engert RF, Short H. Pathogenic properties of Campylobacter jejuni: assay and correlation with clinical manifestations. Infect Immun. 1985;50:43. doi: 10.1128/iai.50.1.43-49.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 785.Lee A, Smith SC, Coloe PJ. Detection of a novel Campylobacter cytotoxin. J Appl Microbiol. 2001;89:719. doi: 10.1046/j.1365-2672.2000.01175.x. [DOI] [PubMed] [Google Scholar]
- 786.Newell DG. Experimental studies of Campylobacter enteritis. In: Butzler JP, editor. Campylobacter Infection in Man and Animals. CRC Press; Boca Raton, Fla: 1984. p. 113. [Google Scholar]
- 787.Prescott JF, Barker IK, Manninen KI. Campylobacter jejuni colitis in gnotobiotic dogs. Can J Comp Med. 1981;45:377. [PMC free article] [PubMed] [Google Scholar]
- 788.Ruiz-Palacios GM, Escamilla E, Torres N. Experimental Campylobacter diarrhea in chickens. Infect Immun. 1981;34:250. doi: 10.1128/iai.34.1.250-255.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 789.Sanyal SC, Islam KM, Neogy PKB. Campylobacter jejuni diarrhea model in infant chickens. Infect Immun. 1984;43:931. doi: 10.1128/iai.43.3.931-936.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 790.Welkos SL. Experimental gastroenteritis in newly hatched chicks infected with Campylobacter jejuni. J Med Microbiol. 1984;18:233. doi: 10.1099/00222615-18-2-233. [DOI] [PubMed] [Google Scholar]
- 791.Fitzgeorge RB, Baskerville A, Lander KP. Experimental infection of rhesus monkeys with a human strain of Campylobacter jejuni. J Hyg. 1981;86:343. doi: 10.1017/s0022172400069096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 792.Field LH, Underwood JL, Berry LJ. The role of gut flora and animal passage in the colonization of adult mice with Campylobacter jejuni. J Med Microbiol. 1984;17:59. doi: 10.1099/00222615-17-1-59. [DOI] [PubMed] [Google Scholar]
- 793.Jesudason MV, Hentges DJ, Pongpeeh P. Colonization of mice by Campylobacter jejuni. Infect Immun. 1989;57:2279. doi: 10.1128/iai.57.8.2279-2282.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 794.Kazmi SU, Roberson BS, Stern NJ. Animal-passed, virulence-enhanced Campylobacter jejuni causes enteritis in neonatal mice. Curr Microbiol. 1984;11:159. [Google Scholar]
- 795.Humphrey CD, Montag DM, Pittman FE. Experimental infection of hamsters with Campylobacter jejuni. J Infect Dis. 1985;151:485. doi: 10.1093/infdis/151.3.485. [DOI] [PubMed] [Google Scholar]
- 796.Manninen KI, Prescott JF, Dohoo IR. Pathogenicity of C. jejuni isolates from animals and humans. Infect Immun. 1982;38:46. doi: 10.1128/iai.38.1.46-52.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 797.Eden AN. Vibrio fetus meningitis in a newborn infant. J Pediatr. 1962;61:33. doi: 10.1016/s0022-3476(62)80226-1. [DOI] [PubMed] [Google Scholar]
- 798.Maki M, Maki R, Vesikari T. Fecal leucocytes in Campylobacter-associated diarrhoea in infants. Acta Paediatr Scand. 1979;68:271. doi: 10.1111/j.1651-2227.1979.tb05000.x. [DOI] [PubMed] [Google Scholar]
- 799.Lambert ME, Schofield PF, Ironside AG. Campylobacter colitis. BMJ. 1979;1:857. doi: 10.1136/bmj.1.6167.857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 800.Blaser MJ, Parsons RB, Wang WL. Acute colitis caused by Campylobacter fetus spp. jejuni. Gastroenterology. 1980;78:448. [PubMed] [Google Scholar]
- 801.King EO. The laboratory recognition of Vibrio fetus and a closely related Vibrio isolated from cases of human vibriosis. Ann N Y Acad Sci. 1962;78:700. [Google Scholar]
- 802.Evans RG, Dadswell JV. Human vibriosis. BMJ. 1967;3:240. [Google Scholar]
- 803.Butzler JP, Oosterom J. Campylobacter: pathogenicity and significance in foods. Int J Food Microbiol. 1991;12:1. doi: 10.1016/0168-1605(91)90043-o. [DOI] [PubMed] [Google Scholar]
- 804.Gill CO, Harris LM. Contamination of red meat carcasses by Campylobacter fetus subsp. jejuni. Appl Environ Microbiol. 1982;43:977. doi: 10.1128/aem.43.5.977-980.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 805.Palmer SR, Gulley PR, White JM. Water-borne outbreak of Campylobacter gastroenteritis. Lancet. 1983;1:287. doi: 10.1016/s0140-6736(83)91698-7. [DOI] [PubMed] [Google Scholar]
- 806.Rogol M, Sechter I, Falk H. Water-borne outbreaks of Campylobacter enteritis. Eur J Clin Microbiol. 1983;2:588. doi: 10.1007/BF02016571. [DOI] [PubMed] [Google Scholar]
- 807.Shankers S, Rosenfield JA, Davey GR. Campylobacter jejuni: incidence in processed broilers and biotype distribution in human and broiler isolates. Appl Environ Microbiol. 1982;43:1219. doi: 10.1128/aem.43.5.1219-1220.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 808.Hood AM, Pearson AD, Shahamat M. The extent of surface contamination of retailed chickens with Campylobacter jejuni serogroups. Epidemiol Infect. 1988;100:17. doi: 10.1017/s0950268800065511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 809.Harris NV, Kimball TJ, Bennett P. Campylobacter jejuni enteritis associated with raw goat’s milk. Am J Epidemiol. 1987;126:179. doi: 10.1093/aje/126.2.179. [DOI] [PubMed] [Google Scholar]
- 810.Korlath JA, Osterholm MT, Judy LA. A point-source outbreak of campylobacteriosis associated with consumption of raw milk. J Infect Dis. 1985;152:592. doi: 10.1093/infdis/152.3.592. [DOI] [PubMed] [Google Scholar]
- 811.Klein BS, Vergeront JM, Blaser MJ. Campylobacter infection associated with raw milk. JAMA. 1986;255:361. doi: 10.1001/jama.255.3.361. [DOI] [PubMed] [Google Scholar]
- 812.Deming MS, Tauxe RV, Blake PA. Campylobacter enteritis at a university: transmission from eating chicken and from cats. Am J Epidemiol. 1987;126:526. doi: 10.1093/oxfordjournals.aje.a114685. [DOI] [PubMed] [Google Scholar]
- 813.Tenkate TD, Stafford RJ. Risk factors for Campylobacter infection in infants and young children: a matched case-controled study. Epidemiol Infect. 2001;127:399. doi: 10.1017/s0950268801006306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 814.Hallett AF, Botha PL, Logan A. Isolation of Campylobacter fetus from recent cases of human vibriosis. J Hyp. 1977;79:381. doi: 10.1017/s0022172400053225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 815.Wolfs TF, Duim B, Geelen Sp. Neonatal sepsis by Campylobacter jejuni: genetically proven transmission from a household puppy. Clin Infect Dis. 2001;31:e97. doi: 10.1086/319224. [DOI] [PubMed] [Google Scholar]
- 816.Blaser MJ, Waldman RJ, Barrett T. Outbreaks of Campylobacter enteritis in two extended families: evidence for person-to-person transmission. J Pediatr. 1981;98:254. doi: 10.1016/s0022-3476(81)80652-x. [DOI] [PubMed] [Google Scholar]
- 817.Cadranel S, Rodesch P, Butzler JP. Enteritis due to “related Vibrio” in children. Am J Dis Child. 1973;126:152. doi: 10.1001/archpedi.1973.02110190134004. [DOI] [PubMed] [Google Scholar]
- 818.Prescott JF, Karmali MA. Attempts to transmit Campylobacter enteritis to dogs and cats. Can Med Assoc J. 1978;119:1001. [PMC free article] [PubMed] [Google Scholar]
- 819.Black RE, Levine MM, Clements ML. Experimental Campylobacter jejuni infection in humans. J Infect Dis. 1988;157:472. doi: 10.1093/infdis/157.3.472. [DOI] [PubMed] [Google Scholar]
- 820.Llovo J, Mateo E, Munoz A. Molecular typing of Campylobacter jejuni isolates involved in a neonatal outbreak indicates nosocomial transmission. J Clin Microbiol. 2003;41:3926. doi: 10.1128/JCM.41.8.3926-3928.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 821.Bokkenheuser VD, Richardson NJ, Bryner JH. Detection of enteric campylobacteriosis in children. J Clin Microbiol. 1979;9:227. doi: 10.1128/jcm.9.2.227-232.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 822.Georges-Courbot MC, Beraud-Cassel AM, Gouandjika I. Prospective study of enteric Campylobacter infections in children from birth to 6 months in the Central African Republic. J Clin Microbiol. 1987;25:836. doi: 10.1128/jcm.25.5.836-839.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 823.Rettig PJ. Campylobacter infections in human beings. J Pediatr. 1979;94:855. doi: 10.1016/s0022-3476(79)80202-4. [DOI] [PubMed] [Google Scholar]
- 824.Blaser MJ, Berkowitz ID, LaForce FM. Campylobacter enteritis: clinical and epidemiological features. Ann Intern Med. 1979;91:179. doi: 10.7326/0003-4819-91-2-179. [DOI] [PubMed] [Google Scholar]
- 825.Butzler JP. Related vibrios in Africa. Lancet. 1973;2:858. doi: 10.1016/s0140-6736(73)90915-x. [DOI] [PubMed] [Google Scholar]
- 826.Lauwers S, DeBoeck M, Butzler JP. Campylobacter enteritis in Brussels. Lancet. 1978;1:604. doi: 10.1016/s0140-6736(78)91045-0. [DOI] [PubMed] [Google Scholar]
- 827.Severin WP. Campylobacter en enteritis. Ned Tijdschr Geneeskd. 1978;122:499. [PubMed] [Google Scholar]
- 828.Ribeiro CD. Campylobacter enteritis. Lancet. 1978;2:270. doi: 10.1016/s0140-6736(78)91789-0. [DOI] [PubMed] [Google Scholar]
- 829.Richardson NJ, Koornhof HJ. Campylobacter infections in Soweto. S Afr Med J. 1979;55:73. [PubMed] [Google Scholar]
- 830.Gribble MJ, Salit EI, Isaac-Renton J. Campylobacter infections in pregnancy. Case report and literature review. Am J Obstet Gynecol. 1981;140:423. doi: 10.1016/0002-9378(81)90038-7. [DOI] [PubMed] [Google Scholar]
- 831.Morooka T, Umeda A, Fujita M. Epidemiologic application of pulsed-field gel electrophoresis to an outbreak of Campylobacter fetus meningitis in a neonatal intensive care unit. Scand J Infect Dis. 1996;28:269. doi: 10.3109/00365549609027170. [DOI] [PubMed] [Google Scholar]
- 832.Morooka T, Takeo H, Yasumoto S, Mimatsu T. Nosocomial meningitis due to Campylobacter fetus subspecies fetus in a neonatal intensive care unit. Acta Paediatr Jpn. 1992;34:350. doi: 10.1111/j.1442-200x.1992.tb01001.x. [DOI] [PubMed] [Google Scholar]
- 833.Norooka T, Takeo H, Yasumoto S. Nosocomial meningitis due to Campylobacter fetus subspecies fetus in a neonatal intensive care unit. Acta Paediatr Jpn. 1992;34:530. doi: 10.1111/j.1442-200x.1992.tb01001.x. [DOI] [PubMed] [Google Scholar]
- 834.Smith MV, Muldoon AJ. Campylobacter fetus subspecies jejuni (Vibrio fetus) from commercially processed poultry. Appl Microbiol. 1974;27:995. doi: 10.1128/am.27.5.995-996.1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 835.Grant IH, Richardson NJ, Bokkenheuser VD. Broiler chickens as potential source of Campylobacter infections in humans. J Clin Microbiol. 1980;2:508. doi: 10.1128/jcm.11.5.508-510.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 836.Blaser MJ, Weiss SH, Barrett TJ. Campylobacter enteritis associated with a healthy cat. JAMA. 1982;247:816. [PubMed] [Google Scholar]
- 837.Skirrow MB, Turnbull GL, Walker RE. Campylobacter jejuni enteritis transmitted from cat to man. Lancet. 1980;1:1188. doi: 10.1016/s0140-6736(80)91644-x. [DOI] [PubMed] [Google Scholar]
- 838.Blaser MJ, Cravens J, Powers BW. Campylobacter enteritis associated with canine infection. Lancet. 1978;2:979. doi: 10.1016/s0140-6736(78)92541-2. [DOI] [PubMed] [Google Scholar]
- 839.Taylor PR, Weinstein WM, Bryner JH. Campylobacter fetus infection in human subjects: association with raw milk. Am J Med. 1979;66:779. doi: 10.1016/0002-9343(79)91116-1. [DOI] [PubMed] [Google Scholar]
- 840.Campylobacter enteritis in a household—Colorado. MMWR Morb Mortal Wkly Rep. 1979;27:273. [Google Scholar]
- 841.Robinson DA, Edgar WM, Gibson GL. Campylobacter enteritis associated with the consumption of unpasteurized milk. BMJ. 1979;1:1171. doi: 10.1136/bmj.1.6172.1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 842.Levy AJ. A gastroenteritis outbreak probably due to bovine strain of Vibrio. Yale J Biol Med. 1946;18:243. [PMC free article] [PubMed] [Google Scholar]
- 843.Vogt RL, Sours HE, Barrett T. Campylobacter enteritis associated with contaminated water. Ann Intern Med. 1982;96:292. doi: 10.7326/0003-4819-96-3-292. [DOI] [PubMed] [Google Scholar]
- 844.Patton CM, Wachsmuth IK, Evins GM. Evaluation of 10 methods to distinguish epidemic-associated Campylobacter strains. J Clin Microbiol. 1991;29:680. doi: 10.1128/jcm.29.4.680-688.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 845.Salazar-Lindo E, Sack B, Chea-Woo E. Early treatment with erythromycin of Campylobacter jejuni associated dysentery in children. J Pediatr. 1986;109:355. doi: 10.1016/s0022-3476(86)80404-8. [DOI] [PubMed] [Google Scholar]
- 846.Darling WM, Peel RN, Skirrow MB. Campylobacter cholecystitis. Lancet. 1979;1:1302. doi: 10.1016/s0140-6736(79)92269-4. [DOI] [PubMed] [Google Scholar]
- 847.Davis JS, Penfold JB. Campylobacter urinary tract infection. Lancet. 1979;1:1091. doi: 10.1016/s0140-6736(79)92995-7. [DOI] [PubMed] [Google Scholar]
- 848.Hossain MA, Kabir I, Albert MJ. Campylobacter jejuni bacteraemia in children with diarrhea in Bangladesh: report of six cases. J Diarrhoeal Dis Res. 1992;10:101. [PubMed] [Google Scholar]
- 849.Reed RP, Friedland IR, Wegerhoff FO, Khoosal M. Campylobacter bacteremia in children. Pediatr Infect Dis J. 1996;15:345. doi: 10.1097/00006454-199604000-00012. [DOI] [PubMed] [Google Scholar]
- 850.Johonsen K, Ostensen M, Christine A. HLA-B27-negative arthritis related to Campylobacter jejuni enteritis in three children and two adults. Acta Med Scand. 1983;214:165. doi: 10.1111/j.0954-6820.1983.tb08589.x. [DOI] [PubMed] [Google Scholar]
- 851.Kaldor J, Speed BR. Guillain-Barré syndrome and Campylobacter jejuni: a serological study. BMJ. 1984;288:1867. doi: 10.1136/bmj.288.6434.1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 852.Kuroki S, Haruta T, Yoshioka M. Guillain-Barré syndrome associated with Campylobacter infection. Pediatr Infect Dis J. 1991;10:149. doi: 10.1097/00006454-199102000-00014. [DOI] [PubMed] [Google Scholar]
- 853.Ebright JR, Ryay LM. Acute erosive reactive arthritis associated with Campylobacter jejuni–induced colitis. Am J Med. 1984;76:321. doi: 10.1016/0002-9343(84)90794-0. [DOI] [PubMed] [Google Scholar]
- 854.Schaad UB. Reactive arthritis associated with Campylobacter enteritis. Pediatr Infect Dis J. 1982;1:328. doi: 10.1097/00006454-198209000-00009. [DOI] [PubMed] [Google Scholar]
- 855.Perlman DM, Ampel NM, Schifman RB. Persistent Campylobacter jejuni infections in patients infected with human immunodeficiency virus (HIV) Ann Intern Med. 1988;108:540. doi: 10.7326/0003-4819-108-4-540. [DOI] [PubMed] [Google Scholar]
- 856.Chiu CH, Kuo CY, Ou JT. Chronic diarrhea and bacteremia caused by Campylobacter lari in a neonate. Clin Infect Dis. 1995;21:700. doi: 10.1093/clinids/21.3.700. [DOI] [PubMed] [Google Scholar]
- 857.Endtz HP, Ruijs GJHM, Zwinderman AH. Comparison of six media, including a semisolid agar for the isolation of various Campylobacter species from stool specimens. J Clin Microbiol. 1991;29:1007. doi: 10.1128/jcm.29.5.1007-1010.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 858.Paisley JW, Mirrett S, Lauer BA. Darkfield microscopy of human feces for presumptive diagnosis of Campylobacter fetus subsp. jejuni enteritis. J Clin Microbiol. 1982;15:61. doi: 10.1128/jcm.15.1.61-63.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 859.Oyofo BA, Thornton SA, Burr DH. Specific detection of Campylobacter jejuni and Campylobacter coli by using polymerase chain reaction. J Clin Microbiol. 1992;30:2613. doi: 10.1128/jcm.30.10.2613-2619.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 860.Kiehlbauch JA, Baker CN, Wachsmuth IK. In vitro susceptibilities of aerotolerant Campylobacter isolates to 22 antimicrobial agents. Antimicrob Agents Chemother. 1992;36:717. doi: 10.1128/aac.36.4.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 861.LaChance N, Gaudreau C, Lamothe F. Susceptibilities of β-lactamase-positive and -negative strains of Campylobacter coli to β-lactam agents. Antimicrob Agents Chemother. 1993;37:1174. doi: 10.1128/aac.37.5.1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 862.Yan W, Taylor DE. Characterization of erythromycin resistance in Campylobacter jejuni and Campylobacter coli. Antimicrob Agents Chemother. 1991;35:1989. doi: 10.1128/aac.35.10.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 863.Segretti J, Gootz TD, Goodman LJ. High-level quinolone resistance in clinical isolates of Campylobacter jejuni. J Infect Dis. 1992;165:667. doi: 10.1093/infdis/165.4.667. [DOI] [PubMed] [Google Scholar]
- 864.Krowchuk D, Seashore JH. Complete biliary obstruction due to erythromycin estolate administration in an infant. Pediatrics. 1979;64:956. [PubMed] [Google Scholar]
- 865.Kuschner RA, Trofa AF, Thomas RJ. Use of azithromycin for the treatment of Campylobacter enteritis in travelers to Thailand, an area where ciprofloxacin resistance is prevalent. Clin Infect Dis. 1995;21:536. doi: 10.1093/clinids/21.3.536. [DOI] [PubMed] [Google Scholar]
- 866.Rautelin H, Renkonen OV, Kosunen TU. Azithromycin resistance in Campylobacter jejuni and Campylobacter coli. Eur J Clin Microbiol Infect Dis. 1993;12:864. doi: 10.1007/BF02000410. [DOI] [PubMed] [Google Scholar]
- 867.Endtz HP, Broeren M, Mouton RP. In vitro susceptibility of quinolone-resistant Campylobacter jejuni to new macrolide antibiotics. Eur J Clin Microbiol Infect Dis. 1993;12:48. doi: 10.1007/BF01997058. [DOI] [PubMed] [Google Scholar]
- 868.Nachamkin I, Fischer SH, Yang XH. Immunoglobulin A antibodies directed against Campylobacter jejuni flagellin present in breast-milk. Epidemiol Infect. 1994;112:359. doi: 10.1017/s0950268800057769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 869.Borriello SP, Davies HA, Kamiya S. Virulence factors of Clostridium difficile. Rev Infect Dis. 1990;12:S185. doi: 10.1093/clinids/12.supplement_2.s185. [DOI] [PubMed] [Google Scholar]
- 870.Sears CL, Kaper JB. Enteric bacterial toxins: mechanisms of action and linkage to intestinal secretion. Microbiol Rev. 1996;60:167. doi: 10.1128/mr.60.1.167-215.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 871.Wren BW. Molecular characterisation of Clostridium difficile toxins A and B. Rev Med Microbiol. 1992;3:21. [Google Scholar]
- 872.Hyams JS, Berman MM, Helgason H. Nonantibiotic-associated enterocolitis caused by Clostridium difficile in an infant. J Pediatr. 1981;99:750. doi: 10.1016/s0022-3476(81)80402-7. [DOI] [PubMed] [Google Scholar]
- 873.Larson HE, Price AB. Pseudomembranous colitis: presence of clostridial toxin. Lancet. 1977;1:1312. doi: 10.1016/s0140-6736(77)90363-4. [DOI] [PubMed] [Google Scholar]
- 874.Peikin SR, Galdibin J, Bartlett JG. Role of Clostridium difficile in a case of nonantibiotic-associated pseudomembranous colitis. Gastroenterology. 1980;79:948. [PubMed] [Google Scholar]
- 875.Wald A, Mendelow H, Bartlett JG. Nonantibiotic-associated pseudomembranous colitis due to toxin producing clostridia. Ann Intern Med. 1980;92:798. doi: 10.7326/0003-4819-92-6-798. [DOI] [PubMed] [Google Scholar]
- 876.Adler SP, Chandrika T, Berman WF. Clostridium difficile associated with pseudomembranous colitis. Am J Dis Child. 1981;135:820. [PubMed] [Google Scholar]
- 877.Willey S, Bartlett JG. Cultures for C. difficile in stools containing a cytotoxin neutralized by C. sordellii antitoxin. J Clin Microbiol. 1979;10:880. doi: 10.1128/jcm.10.6.880-884.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 878.Tabaqchali S. Epidemiologic markers of Clostridium difficile. Rev Infect Dis. 1990;12:S192. doi: 10.1093/clinids/12.supplement_2.s192. [DOI] [PubMed] [Google Scholar]
- 879.Kim KH, Fekety R, Batts D. Isolation of C. difficile from the environment and contacts of patients with antibiotic-associated colitis. J Infect Dis. 1981;143:42. doi: 10.1093/infdis/143.1.42. [DOI] [PubMed] [Google Scholar]
- 880.Sheretz RJ, Sarubb FA. The prevalence of C. difficile and toxin in a nursery population: a comparison between patients with necrotizing enterocolitis and an asymptomatic group. J Pediatr. 1982;100:435. doi: 10.1016/s0022-3476(82)80455-1. [DOI] [PubMed] [Google Scholar]
- 881.Clabots CR, Johnson S, Olson MM. Acquisition of Clostridium difficile by hospitalized patients: evidence for colonized new admissions as a source of infection. J Infect Dis. 1992;166:561. doi: 10.1093/infdis/166.3.561. [DOI] [PubMed] [Google Scholar]
- 882.Bliss DZ, Johnson S, Savik K. Acquisition of Clostridium difficile and Clostridium difficile-associated diarrhea in hospitalized patients receiving tube feeding. Ann Intern Med. 1998;129:1012. doi: 10.7326/0003-4819-129-12-199812150-00004. [DOI] [PubMed] [Google Scholar]
- 883.Jernigan JA, Siegman-Igra Y, Guerrant RC, Farr BM. A randomized crossover study of disposable thermometers for prevention of Clostridium difficile and other nosocomial infections. Infect Control Hosp Epidemiol. 1998;19:494. doi: 10.1086/647855. [DOI] [PubMed] [Google Scholar]
- 884.Johnson S, Gerding DN, Olson NM. Prospective controlled study of vinyl glove use to interrupt Clostridium difficile transmission. Am J Med. 1990;88:137. doi: 10.1016/0002-9343(90)90462-m. [DOI] [PubMed] [Google Scholar]
- 885.Zedd AJ, Sell TL, Schabert DR. Nosocomial C. difficile reservoir in a neonatal intensive care unit. Pediatr Infect Dis J. 1984;3:429. doi: 10.1097/00006454-198409000-00008. [DOI] [PubMed] [Google Scholar]
- 886.Johnson S, Gerding DN. Clostridium difficile–associated diarrhea. Clin Infect Dis. 1998;26:1027. doi: 10.1086/520276. [DOI] [PubMed] [Google Scholar]
- 887.Fekety R, Shah AB. Diagnosis and treatment of Clostridium difficile colitis. JAMA. 1993;269:71. [PubMed] [Google Scholar]
- 888.Kelber M, Ament ME. Shigella dysenteriae 1. A forgotten cause of pseudomembranous colitis. J Pediatr. 1976;89:595. doi: 10.1016/s0022-3476(76)80394-0. [DOI] [PubMed] [Google Scholar]
- 889.Donta ST, Myers MG. C. difficile toxin in asymptomatic neonates. J Pediatr. 1982;100:431. doi: 10.1016/s0022-3476(82)80454-x. [DOI] [PubMed] [Google Scholar]
- 890.Al-Jumaili I, Shibley M, Lishman AH. Incidence and origin of C. difficile in neonates. J Clin Microbiol. 1984;19:77. doi: 10.1128/jcm.19.1.77-78.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 891.Welch DF, Marks MT. Is C. difficile pathogenic in infants? J Pediatr. 1982;100:393. doi: 10.1016/s0022-3476(82)80436-8. [DOI] [PubMed] [Google Scholar]
- 892.Donta ST, Stuppy MS, Myers MG. Neonatal antibiotic-associated colitis. Am J Dis Child. 1981;135:181. doi: 10.1001/archpedi.1981.02130260069022. [DOI] [PubMed] [Google Scholar]
- 893.Enad D, Meislich D, Bodsky NL, Hurt H. Is Clostridium difficile a pathogen in the newborn intensive care unit? A prospective evaluation. J Perinatol. 1997;17:355. [PubMed] [Google Scholar]
- 894.Lyerly DM, Neville LM, Evans DT. Multicenter evaluation of the Clostridium difficile TOX A/B TEST. J Clin Microbiol. 1998;36:184. doi: 10.1128/jcm.36.1.184-190.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 895.Kato H, Kato N, Watanabe K. Identification of toxin A-negative, toxin B-positive Clostridium difficile by PCR. J Clin Microbiol. 1998;36:2178. doi: 10.1128/jcm.36.8.2178-2182.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 896.Rafferty ME, Baltch AL, Smith RP. Comparison of restriction enzyme analysis, arbitrarily primed PCR, and protein profile analysis typing for epidemiologic investigation of an ongoing Clostridium difficile outbreak. J Clin Microbiol. 1998;36:2957. doi: 10.1128/jcm.36.10.2957-2963.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 897.Teasley DG, Gerding DN, Olson MM. Prospective randomized trial of metronidazole vs. vancomycin for C. difficile associated diarrhoea and colitis. Lancet. 1983;2:1043. doi: 10.1016/s0140-6736(83)91036-x. [DOI] [PubMed] [Google Scholar]
- 898.Wenisch C, Parschalk B, Hasenhündl M. Comparison of vancomycin, teicoplanin, metronidazole, and fusidic acid for the treatment of Clostridum difficile-associated diarrhea. Clin Infect Dis. 1996;22:813. doi: 10.1093/clinids/22.5.813. [DOI] [PubMed] [Google Scholar]
- 899.Young GP, Ward PB, Bayley N. Antibiotic-associated colitis due to C. difficile: double-blind comparison of vancomycin with bacitracin. Gastroenterology. 1985;89:1039. doi: 10.1016/0016-5085(85)90206-9. [DOI] [PubMed] [Google Scholar]
- 900.Dudley MN, McLaughlin JC, Carrington G. Oral bacitracin vs. vancomycin therapy for C. difficile–induced diarrhea. Arch Intern Med. 1986;146:1101. [PubMed] [Google Scholar]
- 901.Bartlett JG, Tedesco FJ, Shull S. Relapse following oral vancomycin therapy of antibiotic-associated pseudomembranous colitis. Gastroenterology. 1980;78:431. [PubMed] [Google Scholar]
- 902.Wada N, Nishida N, Iwak S. Neutralizing activity against C. difficile toxin in the supernatants of cultures of colostral cells. Infect Immun. 1980;29:545. doi: 10.1128/iai.29.2.545-550.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 903.Dallas S, Rolfe R. Binding of Clostridium difficile toxin A to human milk secretory component. J Med Microbiol. 1998;47:879. doi: 10.1099/00222615-47-10-879. [DOI] [PubMed] [Google Scholar]
- 904.Kim K, Pickering LK, DuPont HL. In vitro and in vivo neutralizing activity of C. difficile purified toxins A and B by human colostrum and milk. J Infect Dis. 1984;150:57. doi: 10.1093/infdis/150.1.57. [DOI] [PubMed] [Google Scholar]
- 905.Cooperstock MS, Steffen E, Yolken R. C. difficile in normal infants and sudden death syndrome: an association with infant formula feeding. Pediatrics. 1982;70:91. [PubMed] [Google Scholar]
- 906.Levine WC, Griffin PM, and the Gulf Coast Vibrio Working Group Vibrio infections on the Gulf Coast: results of first year of regional surveillance. J Infect Dis. 1993;167:479. doi: 10.1093/infdis/167.2.479. [DOI] [PubMed] [Google Scholar]
- 907.Swerdlow DL, Ries AA. Cholera in the Americas: guidelines for the clinician. JAMA. 1992;267:1495. [PubMed] [Google Scholar]
- 908.Glass RI, Libel M, Brandling-Bennett AD. Epidemic cholera in the Americas. Science. 1992;256:1524. doi: 10.1126/science.1598586. [DOI] [PubMed] [Google Scholar]
- 909.Wachsmuth IK, Evins GM, Fields PI. The molecular epidemiology of cholera in Latin America. J Infect Dis. 1993;167:621. doi: 10.1093/infdis/167.3.621. [DOI] [PubMed] [Google Scholar]
- 910.Siddique AK, Zaman K, Akram K. Emergence of a new epidemic strain of Vibrio cholerae in Bangladesh. An epidemiological study. Trop Geogr Med. 1994;46:147. [PubMed] [Google Scholar]
- 911.Fisher-Hoch SP, Khan A, Inam-ul-Haq Vibrio cholerae O139 in Karachi, Pakistan. Lancet. 1993;342:1422. doi: 10.1016/0140-6736(93)92780-w. [DOI] [PubMed] [Google Scholar]
- 912.Chongsa-Nguan M, Chaicumpa W, Moolasart P. Vibrio cholerae O139 Bengal in Bangkok. Lancet. 1993;342:430. doi: 10.1016/0140-6736(93)92841-g. [DOI] [PubMed] [Google Scholar]
- 913.Cholera Working Group, International Centre for Diarrhoeal Diseases Research, Bangladesh Large epidemic of cholera-like disease in Bangladesh caused by Vibrio cholerae O139 synonym Bengal. Lancet. 1993;342:387. [PubMed] [Google Scholar]
- 914.Bhattacharya SK, Bhattacharya MK, Nair GB. Clinical profile of acute diarrhoea cases infected with the new epidemic strain of Vibrio cholerae O139: designation of the disease as cholera. J Infect. 1993;27:11. doi: 10.1016/0163-4453(93)93488-p. [DOI] [PubMed] [Google Scholar]
- 915.Garg S, Saha PK, Ramamurthy T. Nationwide prevalence of the new epidemic strain of Vibrio cholerae O139 Bengal in India. J Infect. 1993;27:108. doi: 10.1016/0163-4453(93)94398-u. [DOI] [PubMed] [Google Scholar]
- 916.Blake PA, Allegra DT, Snyder JD. Cholera—a possible endemic focus in the United States. N Engl J Med. 1980;302:305. doi: 10.1056/NEJM198002073020601. [DOI] [PubMed] [Google Scholar]
- 917.Hirschhorn N, Chowdhury AAKM, Lindenbaum J. Cholera in pregnant women. Lancet. 1969;1:1230. doi: 10.1016/s0140-6736(69)92115-1. [DOI] [PubMed] [Google Scholar]
- 918.Khan AM, Bhattacharyl MK, Albert MJ. Neonatal diarrhea caused by Vibrio cholerae O139 Bengal. Diagn Microbiol Infect Dis. 1995;23:155. doi: 10.1016/0732-8893(95)00186-7. [DOI] [PubMed] [Google Scholar]
- 919.Lumbiganon P, Kosalaraksa P, Kowsuwan P. Vibrio cholerae O139 diarrhea and acute renal failure in a three day old infant. Pediatr Infect Dis J. 1995;14:1105. doi: 10.1097/00006454-199512000-00017. [DOI] [PubMed] [Google Scholar]
- 920.Haider R, Kabir I, Fuchs GJ, Habte D. Neonatal diarrhea in a diarrhea treatment center in Bangladesh: clinical presentation, breastfeeding management and outcome. Indian Pediatr. 2000;37:37. [PubMed] [Google Scholar]
- 921.Gunn RA, Kimball AM, Pollard RA. Bottle feeding as a risk factor for cholera in infants. Lancet. 1979;2:730. doi: 10.1016/s0140-6736(79)90653-6. [DOI] [PubMed] [Google Scholar]
- 922.Ahmed A, Bhattacharjee AK, Mosley WH. Characteristics of the serum vibriocidal and agglutinating antibodies in cholera cases and in normal residents of the endemic and non-endemic cholera areas. J Immunol. 1970;105:431. [PubMed] [Google Scholar]
- 923.Merson MH, Black RE, Sack DA. Maternal cholera immunisation and secretory IgA in breast milk. Lancet. 1980;1:931. doi: 10.1016/s0140-6736(80)90860-0. [DOI] [PubMed] [Google Scholar]
- 924.Cash RA, Music SI, Libonati JP. Response of man to infection with Vibrio cholerae. I. Clinical, serologic and bacteriologic responses to a known inoculum. J Infect Dis. 1974;129:45. doi: 10.1093/infdis/129.1.45. [DOI] [PubMed] [Google Scholar]
- 925.Nalin DR, Levine RJ, Levine MM. Cholera, non–Vibrio cholera, and stomach acid. Lancet. 1978;2:856. doi: 10.1016/s0140-6736(78)91568-4. [DOI] [PubMed] [Google Scholar]
- 926.Wright AC, Guo Y, Johnson JA. Development and testing of a nonradioactive DNA oligonucleotide probe that is specific for Vibrio cholerae cholera toxin. J Clin Microbiol. 1992;30:2302. doi: 10.1128/jcm.30.9.2302-2306.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 927.Yoh M, Miyagi K, Matsumoto Y. Development of an enzyme-labeled oligonucleotide probe for the cholera toxin gene. J Clin Microbiol. 1993;31:1312. doi: 10.1128/jcm.31.5.1312-1314.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 928.Kotloff KL, Wasserman SS, O’Donnell S. Safety and immunogenicity in North Americans of a single dose of live oral cholera vaccine CVD 103-HgR: results of a randomized, placebo-controlled, double-blind crossover trial. Infect Immun. 1992;60:4430. doi: 10.1128/iai.60.10.4430-4432.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 929.Clemens JD, Sack DA, Rao MR. Evidence that inactivated oral cholera vaccines both prevent and mitigate Vibrio cholerae O1 infections in a cholera-endemic area. J Infect Dis. 1992;166:1029. doi: 10.1093/infdis/166.5.1029. [DOI] [PubMed] [Google Scholar]
- 930.Levine MM, Noriega F. A review of the current status of enteric vaccines. P N G Med J. 1995;38:325. [PubMed] [Google Scholar]
- 931.Kohl S. Yersinia enterocolitica infections in children. Pediatr Clin North Am. 1979;26:433. doi: 10.1016/s0031-3955(16)33715-4. [DOI] [PubMed] [Google Scholar]
- 932.Marks MI, Pai CH, Lafleur L. Yersinia enterocolitica gastroenteritis: a prospective study of clinical, bacteriologic, and epidemiologic features. J Pediatr. 1980;96:26. doi: 10.1016/s0022-3476(80)80318-0. [DOI] [PubMed] [Google Scholar]
- 933.Lee LA, Gerber AR, Lonsway DR. Yersinia enterocolitica 0:3 infections in infants and children, associated with the household preparation of chitterlings. N Engl J Med. 1990;322:984. doi: 10.1056/NEJM199004053221407. [DOI] [PubMed] [Google Scholar]
- 934.Krogstad P, Mendelman PM, Miller VL. Clinical and microbiologic characteristics of cutaneous infection with Yersinia enterocolitica. J Infect Dis. 1992;165:740. doi: 10.1093/infdis/165.4.740. [DOI] [PubMed] [Google Scholar]
- 935.Lee LA, Taylor J, Carter GP. Yersinia enterocolitica 0:3: an emerging cause of pediatric gastroenteritis in the United States. J Infect Dis. 1991;163:660. doi: 10.1093/infdis/163.3.660. [DOI] [PubMed] [Google Scholar]
- 936.Morris JG, Jr, Prado V, Ferreccio C. Yersinia enterocolitica isolated from two cohorts of young children in Santiago, Chile: incidence of and lack of correlation between illness and proposed virulence factors. J Clin Microbiol. 1991;29:2784. doi: 10.1128/jcm.29.12.2784-2788.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 937.Metchock B, Lonsway DR, Carter GP. Yersinia enterocolitica: a frequent seasonal stool isolate from children at an urban hospital in the southeast United States. J Clin Microbiol. 1991;29:2868. doi: 10.1128/jcm.29.12.2868-2869.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 938.Kane DR, Reuman PD. Yersinia enterocolitica causing pneumonia and empyema in a child and a review of the literature. Pediatr Infect Dis J. 1992;11:591. [PubMed] [Google Scholar]
- 939.Black RE, Jackson RJ, Tsai T. Epidemic Yersinia enterocolitica infection due to contaminated chocolate milk. N Engl J Med. 1978;298:76. doi: 10.1056/NEJM197801122980204. [DOI] [PubMed] [Google Scholar]
- 940.Pietersz RNI, Reesink HW, Pauw W. Prevention of Yersinia enterocolitica growth in red blood cell concentrates. Lancet. 1992;340:755. doi: 10.1016/0140-6736(92)92296-r. [DOI] [PubMed] [Google Scholar]
- 941.Kapperud G, Namork E, Skurnik M. Plasmid-mediated surface fibrillae of Y. pseudotuberculosis and Y. enterocolitica. Relationship to the outer membrane protein YOP1 and possible importance for pathogenesis. Infect Immun. 1987;55:2247. doi: 10.1128/iai.55.9.2247-2254.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 942.Brubaker RR. Factors promoting acute and chronic diseases caused by yersiniae. Clin Microbiol Rev. 1991;4:309. doi: 10.1128/cmr.4.3.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 943.Takao T, Tominaga N, Shimoniski Y. Primary structure of heat-stable enterotoxin produced by Y. enterocolitica. Biochem Biophys Res Commun. 1984;125:845. doi: 10.1016/0006-291x(84)91360-3. [DOI] [PubMed] [Google Scholar]
- 944.Paisley JW, Lauer BA. Neonatal Yersinia enterocolitica enteritis. Pediatr Infect Dis J. 1992;11:331. doi: 10.1097/00006454-199204000-00014. [DOI] [PubMed] [Google Scholar]
- 945.Shapiro ED. Yersinia enterocolitica septicemia in normal infants. Am J Dis Child. 1981;135:477. doi: 10.1001/archpedi.1981.02130290073025. [DOI] [PubMed] [Google Scholar]
- 946.Chester B, Sanderson T, Zeller DJ. Infections due to Yersinia enterocolitica serotypes 0:2, 3 and 0:4 acquired in South Florida. J Clin Microbiol. 1981;13:885. doi: 10.1128/jcm.13.5.885-887.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 947.Rodriguez WJ, Controni G, Cohen GJ. Y. enterocolitica enteritis in children. JAMA. 1979;242:1978. doi: 10.1001/jama.242.18.1978. [DOI] [PubMed] [Google Scholar]
- 948.Challapalli M, Cunningham DG. Yersinia enterocolitica septicemia in infants younger than three months of age. Pediatr Infect Dis J. 1993;12:168. doi: 10.1097/00006454-199302000-00018. [DOI] [PubMed] [Google Scholar]
- 949.Paisley JW, Lauer BA. Neonatal Yersinia enterocolitica enteritis. Pediatr Infect Dis J. 1992;11:332. doi: 10.1097/00006454-199204000-00014. [DOI] [PubMed] [Google Scholar]
- 950.Antonio-Santiago MT, Kaul A, Lue Y. Yersinia enterocolitica septicemia in an infant presenting as fever of unknown origin. Clin Pediatr. 1986;25:213. [PubMed] [Google Scholar]
- 951.Sutton JM, Pasquariell PS. Yersinia enterocolitica septicemia in a normal child. Am J Dis Child. 1983;137:305. doi: 10.1001/archpedi.1983.02140290085031. [DOI] [PubMed] [Google Scholar]
- 952.Kohl S, Jacobson JA, Nahmias A. Yersinia enterocolitica infections in children. J Pediatr. 1976;89:77. doi: 10.1016/s0022-3476(76)80932-8. [DOI] [PubMed] [Google Scholar]
- 953.Naqvi S, Swierkosz E, Gerard J, Mills J. Presentation of Yersinia enterocolitica enteritis in children. Pediatr Infect Dis J. 1993;12:386. doi: 10.1097/00006454-199305000-00007. [DOI] [PubMed] [Google Scholar]
- 954.Abdel-Haq N, Asmar B, Abuhammour W, Brown W. Yersinia enterocolitica infection in children. Pediatr Infect Dis J. 2000;19:954. doi: 10.1097/00006454-200010000-00002. [DOI] [PubMed] [Google Scholar]
- 955.Ibrahim A, Liesack W, Stackebrandt E. Polymerase chain reaction– gene probe detection system specific for pathogenic strains of Yersinia enterocolitica. J Clin Microbiol. 1992;30:1942. doi: 10.1128/jcm.30.8.1942-1947.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 956.Kwaga J, Iversen JO, Misra V. Detection of pathogenic Yersinia enterocolitica by polymerase chain reaction and digoxigenin-labeled polynucleotide probes. J Clin Microbiol. 1992;30:2668. doi: 10.1128/jcm.30.10.2668-2673.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 957.Stolk-Engelaar VM, Meis JF, Mulder JA. In-vitro antimicrobial susceptibility of Yersinia enterocolitica isolates from stools of patients in the Netherlands from 1982–1991. J Antimicrob Chemother. 1995;36:839. doi: 10.1093/jac/36.5.839. [DOI] [PubMed] [Google Scholar]
- 958.Alzugaray R, Gonzalez Hevia MA, Landeras E, Mendoza MC. Yersinia enterocolitica 0:3. Antimicrobial resistance patterns, virulence profiles and plasmids. New Microbiol. 1995;18:215. [PubMed] [Google Scholar]
- 959.Preston MA, Brown S, Borczyk AA. Antimicrobial susceptibility of pathogenic Yersinia enterocolitica isolated in Canada from 1972 to 1990. Antimicrob Agents Chemother. 1994;38:2121. doi: 10.1128/aac.38.9.2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 960.James C, Dibley M, Burke V. Immunological cross-reactivity of enterotoxins of A. hydrophila and cholera toxin. Clin Exp Immunol. 1982;47:34. [PMC free article] [PubMed] [Google Scholar]
- 961.Sanyal SC, Singh SJ, Sen PC. Enteropathogenicity of A. hydrophila and P. shigelloides. J Med Microbiol. 1975;8:195. doi: 10.1099/00222615-8-1-195. [DOI] [PubMed] [Google Scholar]
- 962.Kirov SM, Rees B, Wellock RC. Virulence characteristics of Aeromonas spp. in relation to source and biotype. J Clin Microbiol. 1986;24:827. doi: 10.1128/jcm.24.5.827-834.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 963.Watson IM, Robinson JO, Burke V. Invasiveness of Aeromonas spp. in relation to biotype, virulence factors, and clinical features. J Clin Microbiol. 1985;22:48. doi: 10.1128/jcm.22.1.48-51.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 964.Kindshuh M, Pickering LK, Cleary TG. Clinical and biochemical significance of toxin production by A. hydrophila. J Clin Microbiol. 1987;25:916. doi: 10.1128/jcm.25.5.916-921.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 965.Ljungh A, Eneroth P, Wadstrom T. Cytotonic enterotoxin from Aeromonas hydrophila. Toxicon. 1982;20:787. doi: 10.1016/0041-0101(82)90126-x. [DOI] [PubMed] [Google Scholar]
- 966.Morgan D, Johnson PC, DuPont HL. Lack of correlation between known virulence properties of A. hydrophila and enteropathogenicity for humans. Infect Immun. 1985;50:62. doi: 10.1128/iai.50.1.62-65.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 967.Pitarangsi C, Echeverria P, Whitemire R. Enteropathogenicity of A. hydrophila and P. shigelloides: prevalence among individuals with and without diarrhea in Thailand. Infect Immun. 1982;35:666. doi: 10.1128/iai.35.2.666-673.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 968.Martinez-Silva R, Guzmann-Urrego M, Caselitz FH. Zur Frage der Bedeutung von Aeromonasstammen bei Saüglingsenteritis. Z Tropenmed Parasitol. 1961;12:445. [PubMed] [Google Scholar]
- 969.Figura N, Marri L, Verdiani S. Prevalence, species differentiation, and toxigenicity of Aeromonas strains in cases of childhood gastroenteritis and in controls. J Clin Microbiol. 1986;23:595. doi: 10.1128/jcm.23.3.595-599.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 970.Gracey M, Burke V, Robinson J. Aeromonas-associated gastroenteritis. Lancet. 1982;2:1304. doi: 10.1016/s0140-6736(82)91510-0. [DOI] [PubMed] [Google Scholar]
- 971.Shread P, Donovan TJ, Lee JV. A survey of the incidence of Aeromonas in human feces. Soc Gen Microbiol. 1981;8:184. [Google Scholar]
- 972.Escheverria P, Blacklow NR, Sanford LB. Travelers’ diarrhea among American Peace Corps volunteers in rural Thailand. J Infect Dis. 1981;143:767. doi: 10.1093/infdis/143.6.767. [DOI] [PubMed] [Google Scholar]
- 973.Bhat P, Shanthakumari S, Rajan D. The characterization and significance of P. shigelloides and A. hydrophila isolated from an epidemic of diarrhea. Indian J Med Res. 1974;62:1051. [PubMed] [Google Scholar]
- 974.Agger WA, McCormick JD, Gurwith MJ. Clinical and microbiological features of A. hydrophila–associated diarrhea. J Clin Microbiol. 1985;21:909. doi: 10.1128/jcm.21.6.909-913.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 975.Agger WA. Diarrhea associated with A. hydrophila. Pediatr Infect Dis J. 1986;5:S106. doi: 10.1097/00006454-198601001-00018. [DOI] [PubMed] [Google Scholar]
- 976.Deodhar LP, Saraswathi K, Varudkar A. Aeromonas spp. and their association with human diarrheal disease. J Clin Microbiol. 1991;29:853. doi: 10.1128/jcm.29.5.853-856.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 977.Santoso H, Agung IGN, Robinson J. Faecal Aeromonas spp. in Balinese children. J Gastroenterol Hepatol. 1986;1:115. [Google Scholar]
- 978.Gomez CJ, Munozz P, Lopez F. Gastroenteritis due to Aeromonas in pediatrics. An Esp Pediatr. 1996;44:548. [PubMed] [Google Scholar]
- 979.Diaz A, Velasco AC, Hawkins F. A. hydrophila–associated diarrhea in a neonate. Pediatr Infect Dis J. 1986;5:704. doi: 10.1097/00006454-198611000-00022. [DOI] [PubMed] [Google Scholar]
- 980.San Joaquin VH, Pickett DA. Aeromonas-associated gastroenteritis in children. Pediatr Infect Dis J. 1988;7:53. doi: 10.1097/00006454-198801000-00013. [DOI] [PubMed] [Google Scholar]
- 981.George WL, Jones MJ, Nakata MM. Phenotypic characteristics of Aeromonas species isolated from adult humans. J Clin Microbiol. 1986;23:1026. doi: 10.1128/jcm.23.6.1026-1029.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 982.Janda JM. Recent advances in the study of the taxonomy, pathogenicity, and infectious syndromes associated with the genus Aeromonas. Clin Microbiol Rev. 1991;4:397. doi: 10.1128/cmr.4.4.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 983.Freij BJ. Aeromonas: biology of the organism and diseases in children. Pediatr Infect Dis J. 1984;3:164. [PubMed] [Google Scholar]
- 984.Fainstein V, Weaver S, Bodey GP. In vitro susceptibilities of A. hydrophila against new antibiotics. Antimicrob Agents Chemother. 1982;22:513. doi: 10.1128/aac.22.3.513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 985.San Joaquin VH, Scribner RK, Pickett DA. Antimicrobial susceptibility of Aeromonas species isolated from patients with diarrhea. Antimicrob Agents Chemother. 1986;30:794. doi: 10.1128/aac.30.5.794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 986.Jones BL, Wilcox MH. Aeromonas infections and their treatment. J Antimicrob Chemother. 1995;35:453. doi: 10.1093/jac/35.4.453. [DOI] [PubMed] [Google Scholar]
- 987.Holmberg SD, Wachsmuth IK, Hickmann-Brenner FW. Plesiomonas enteric infections in the United States. Ann Intern Med. 1986;105:690. doi: 10.7326/0003-4819-105-5-690. [DOI] [PubMed] [Google Scholar]
- 988.Tsukamoto T, Kinoshita Y, Shimada T. Two epidemics of diarrhoeal disease possibly caused by P. shigelloides. J Hyg. 1978;80:275. doi: 10.1017/s0022172400053638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 989.Holmberg SD, Farmer JJ. A. hydrophila and P. shigelloides as causes of intestinal infections. Rev Infect Dis. 1984;6:633. doi: 10.1093/clinids/6.5.633. [DOI] [PubMed] [Google Scholar]
- 990.Herrington DA, Tzipori S, Robins-Browne RM. In vitro and in vivo pathogenicity of P. shigelloides. Infect Immun. 1987;55:979. doi: 10.1128/iai.55.4.979-985.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 991.Brenden RA, Miller MA, Janda JM. Clinical disease spectrum and pathogenic factors associated with P. shigelloides infections in humans. Rev Infect Dis. 1988;10:303. doi: 10.1093/clinids/10.2.303. [DOI] [PubMed] [Google Scholar]
- 992.Pathak A, Custer JR, Levy J. Neonatal septicemia and meningitis due to Plesiomonas shigelloides. Pediatrics. 1983;71:389. [PubMed] [Google Scholar]
- 993.Fujita K, Shirai M, Ishioka T, Kakuya F. Neonatal Plesiomonas shigelloides septicemia and meningitis: a case review. Acta Paediatr Jpn. 1994;36:450. doi: 10.1111/j.1442-200x.1994.tb03222.x. [DOI] [PubMed] [Google Scholar]
- 994.Terpeluk C, Goldmann A, Bartmann P, Pohlandt F. Plesiomonas shigelloides sepsis and meningoencephalitis in a neonate. Eur J Pediatr. 1992;151:499. doi: 10.1007/BF01957753. [DOI] [PubMed] [Google Scholar]
- 995.Billiet J, Kuypers S, Van Lierde S, Verhaegen J. Plesiomonas shigelloides meningitis and septicaemia in a neonate: report of a case and review of the literature. J Infect. 1989;19:267. doi: 10.1016/s0163-4453(89)90809-8. [DOI] [PubMed] [Google Scholar]
- 996.Alabi SA, Odugbemi T. Biochemical characteristics and a simple scheme for the identification of Aeromonas species and Plesiomonas shigelloides. J Trop Med Hyg. 1990;93:166. [PubMed] [Google Scholar]
- 997.Reinhardt JF, George WL. Comparative in vitro activities of selected antimicrobial agents against Aeromonas species and P. shigelloides. Antimicrob Agents Chemother. 1985;27:643. doi: 10.1128/aac.27.4.643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 998.Visitsunthorn N, Komolpis P. Antimicrobial therapy in Plesiomonas shigelloides–associated diarrhea in Thai children. Southeast Asian J Trop Med Public Health. 1995;26:86. [PubMed] [Google Scholar]
- 999.Jampolis M, Howell KM, Calvin JK. Bacillus mucosus infection of the newborn. Am J Dis Child. 1932;43:70. [Google Scholar]
- 1000.Olarte J, Ferguson WW, Henderson ND. Klebsiella strains isolated from diarrheal infants. Human volunteer studies. Am J Dis Child. 1961;101:763. doi: 10.1001/archpedi.1961.04020070077011. [DOI] [PubMed] [Google Scholar]
- 1001.Murdoch MM, Janovski NA, Joseph S. Klebsiella pseudomembranous enterocolitis. Report of two cases. Med Ann Dist Columbia. 1969;38:137. [PubMed] [Google Scholar]
- 1002.Ujvary G, Angyal T, Voros S. Beobachtungen Über die Ätiologie der Gastroenterocolitiden des Säuglings- und Kindesalters. II. Untersuchung der Rolle der Klebsiella-Stamme. Acta Microbiol Acad Sci Hung. 1964;10:241. [PubMed] [Google Scholar]
- 1003.Gergely K. Über eine Enteritis-Epidemie bei Frühgeborenen, verursacht durch den Bacillus Klebsiella. Kinderarztl Prax. 1964;9:385. [PubMed] [Google Scholar]
- 1004.Walcher DN. “Bacillus mucosus capsulatus” in infantile diarrhea. J Clin Invest. 1946;25:103. [PubMed] [Google Scholar]
- 1005.Cass JM. Bacillus lactis aerogenes infection in the newborn. Lancet. 1941;1:346. [Google Scholar]
- 1006.Sternberg SD, Hoffman C, Zweifler BM. Stomatitis and diarrhea in infants caused by Bacillus mucosus capsulatus. J Pediatr. 1951;38:509. doi: 10.1016/s0022-3476(51)80038-6. [DOI] [PubMed] [Google Scholar]
- 1007.Worfel MT, Ferguson WW. A new Klebsiella type (capsular type 15) isolated from feces and urine. Am J Clin Pathol. 1951;21:1097. doi: 10.1093/ajcp/21.11_ts.1097. [DOI] [PubMed] [Google Scholar]
- 1008.Simmons BP, Gelfand MS, Haas M. Enterobacter sakazakii infections in neonates associated with intrinsic contamination of a powdered infant formula. Infect Control Hosp Epidemiol. 1989;10:398. doi: 10.1086/646060. [DOI] [PubMed] [Google Scholar]
- 1009.Ayliffe GAJ, Collins BJ, Pettit F. Contamination of infant feeds in a Milton milk kitchen. Lancet. 1970;1:559. doi: 10.1016/s0140-6736(70)90783-x. [DOI] [PubMed] [Google Scholar]
- 1010.Adler JL, Shulman JA, Terry PM. Nosocomial colonization with kanamycin-resistant Klebsiella pneumoniae, types 2 and 11, in a premature nursery. J Pediatr. 1970;77:376. doi: 10.1016/s0022-3476(70)80004-x. [DOI] [PubMed] [Google Scholar]
- 1011.Hill HR, Hunt CE, Matsen JM. Nosocomial colonization with Klebsiella, type 16, in a neonatal intensive-care unit associated with an outbreak of sepsis, meningitis, and necrotizing enterocolitis. J Pediatr. 1974;85:415. doi: 10.1016/s0022-3476(74)80133-2. [DOI] [PubMed] [Google Scholar]
- 1012.Panigrahi D, Roy P, Chakrabarti A. Enterotoxigenic Klebsiella pneumoniae in acute childhood diarrhea. Indian J Med Res. 1991;93:293. [PubMed] [Google Scholar]
- 1013.Guarino A, Capano G, Malamisura B. Production of E. coli STa-like heat stable enterotoxin by Citrobacter freundii isolated from humans. J Clin Microbiol. 1987;25:110. doi: 10.1128/jcm.25.1.110-114.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1014.Lipsky BA, Hook EW, Smith AA. Citrobacter infections in humans: experience at the Seattle Veterans Administration Medical Center and a review of the literature. Rev Infect Dis. 1980;2:746. doi: 10.1093/clinids/2.5.746. [DOI] [PubMed] [Google Scholar]
- 1015.Kahlich R, Webershinke J. A contribution to incidence and evaluation of Citrobacter findings in man. Cesk Epidemiol Mikrobiol Imunol. 1963;12:55. [PubMed] [Google Scholar]
- 1016.Parida SN, Verma IC, Deb M. An outbreak of diarrhea due to Citrobacter freundii in a neonatal special care nursery. Indian J Pediatr. 1980;47:81. doi: 10.1007/BF02900180. [DOI] [PubMed] [Google Scholar]
- 1017.Heitmann M, Gerner-Smidt P, Heltberg O. Gastroenteritis caused by Listeria monocytogenes in a private day-care facility. Pediatr Infect Dis J. 1997;16:827. doi: 10.1097/00006454-199708000-00025. [DOI] [PubMed] [Google Scholar]
- 1018.Sim J, Hood D, Finnie L. Series of incidents of Listeria monocytogenes non-invasive febrile gastroenteritis involving ready-to-eat meats. Lett Appl Microbiol. 2002;35:409. doi: 10.1046/j.1472-765x.2002.01207.x. [DOI] [PubMed] [Google Scholar]
- 1019.Schlech W. Listeria gastroenteritis—old syndrome, new pathogen. N Engl J Med. 1997;336:130. doi: 10.1056/NEJM199701093360211. [DOI] [PubMed] [Google Scholar]
- 1020.Wing E, Gregory S. Listeria monocytogenes: clinical and experimental update. J Infect Dis. 2002;185:S18. doi: 10.1086/338465. [DOI] [PubMed] [Google Scholar]
- 1021.Aureli P, Fiorucci G, Caroli D. An outbreak of febrile gastroenteritis associated with corn contaminated by Listeria monocytogenes. N Engl J Med. 2000;342:1236. doi: 10.1056/NEJM200004273421702. [DOI] [PubMed] [Google Scholar]
- 1022.Dalton C, Austin C, Sobel J. An outbreak of gastroenteritis and fever due to Listeria monocytogenes in milk. N Engl J Med. 1997;336:100. doi: 10.1056/NEJM199701093360204. [DOI] [PubMed] [Google Scholar]
- 1023.Hof H, Lampidis R, Bensch J. Noscomial Listeria gastroenteritis in a newborn, confirmed by random amplification of polymorphic DNA. Clin Microbiol Infect. 2000;6:683. doi: 10.1046/j.1469-0691.2000.00111.x. [DOI] [PubMed] [Google Scholar]
- 1024.Larsson S, Cederberg A, Ivarsson S. Listeria monocytogenes causing hospital-acquired enterocolitis and meningitis in newborn infants. Br Med J. 1978;2:473. doi: 10.1136/bmj.2.6135.473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1025.Edelbroek M, De Nef J, Rajnherc J. Listeria meningitis presenting as enteritis in a previously healthy infant: a case report. Eur J Pediatr. 1994;153:179. doi: 10.1007/BF01958980. [DOI] [PubMed] [Google Scholar]
- 1026.Norys H. Fetal chronic nonspecific enterocolitis with peritonitis in uniovular twins after Listeria infection in the mother. Monatsschr Kinderheilkd. 1960;108:59. [PubMed] [Google Scholar]
- 1027.Sack R, Albert M, Alam K. Isolation of enterotoxigenic Bacteroides fragilis from Bangladeshi children with diarrhea: a controlled study. J Clin Microbiol. 1994;32:960. doi: 10.1128/jcm.32.4.960-963.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1028.Sack R, Myers L, Almeido-Hill J. Enterotoxigenic Bacteroides fragilis: epidemiologic studies of its role as a human diarrhoeal pathogen. J Diarrhoeal Dis Res. 1992;10:4. [PubMed] [Google Scholar]
- 1029.Kubota Y, Liu PV. An enterotoxin of Pseudomonas aeruginosa. J Infect Dis. 1971;123:97. doi: 10.1093/infdis/123.1.97. [DOI] [PubMed] [Google Scholar]
- 1030.Bassett DCJ, Thompson SAS, Page B. Neonatal infections with Pseudomonas aeruginosa associated with contaminated resuscitation equipment. Lancet. 1965;1:781. doi: 10.1016/s0140-6736(65)92955-7. [DOI] [PubMed] [Google Scholar]
- 1031.Ensign PR, Hunter CA. An epidemic of diarrhea in the newborn nursery caused by a milk-borne epidemic in the community. J Pediatr. 1946;29:620. doi: 10.1016/s0022-3476(46)80129-x. [DOI] [PubMed] [Google Scholar]
- 1032.Falcao DP, Mendonca CP, Scrassolo A. Nursery outbreak of severe diarrhoea due to multiple strains of Pseudomonas aeruginosa. Lancet. 1972;2:38. doi: 10.1016/s0140-6736(72)91291-3. [DOI] [PubMed] [Google Scholar]
- 1033.Henderson A, Maclaurin J, Scott JM. Pseudomonas in a Glasgow baby unit. Lancet. 1969;2:316. doi: 10.1016/s0140-6736(69)90071-3. [DOI] [PubMed] [Google Scholar]
- 1034.Jellard CH, Churcher GM. An outbreak of Pseudomonas aeruginosa (pyocyanea) infection in a premature baby unit, with observations on the intestinal carriage of Pseudomonas aeruginosa in the newborn. J Hyg. 1967;65:219. doi: 10.1017/s0022172400045708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1035.Rowe B, Gross RJ, Allen HA. Citrobacter koseri II. Serological and biochemical examination of Citrobacter koseri strains from clinical specimens. J Hyg. 1975;75:129. doi: 10.1017/s0022172400047148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1036.Kalashnikova GK, Lokosova AK, Sorokina RS. Concerning the etiological role of bacteria belonging to Citrobacter and Hafnia genera in children suffering from diseases accompanied by diarrhea, and some of their epidemiological peculiarities. Zh Mikrobiol Epidemiol Immunobiol. 1974;6:78. [PubMed] [Google Scholar]
- 1037.Graber CD, Dodd MC. The role of Paracolobactrum and Proteus in infantile diarrhea. Ann N Y Acad Sci. 1956;66:136. doi: 10.1111/j.1749-6632.1956.tb40112.x. [DOI] [PubMed] [Google Scholar]
- 1038.Neter E, Goodale ML. Peritonitis due to the Morgani bacillus. With a brief review of literature on the pathogenicity of this organism. Am J Dis Child. 1938;56:1313. [Google Scholar]
- 1039.Neter ER, Farrar RH. Proteus vulgaris and Proteus morgani in diarrheal disease of infants. Am J Dig Dis. 1943;10:344. [Google Scholar]
- 1040.Neter E, Bender NC. Bacillus morgani, type I, in enterocolitis of infants. J Pediatr. 1941;19:53. [Google Scholar]
- 1041.Ujvary G, Lanyi B, Gregacs M. Beobachtungen über die Ätiologie der Gastroenterocolitiden des Säuglings- und Kindesalters. III. Untersuchung der Rolle der Proteus vulgaris- und der Proteus mirabilis-Stamme. Acta Microbiol Acad Sci Hung. 1964;10:315. [PubMed] [Google Scholar]
- 1042.Moffet HL, Shulenberger HK, Burkholder ER. Epidemiology and etiology of severe infantile diarrhea. J Pediatr. 1968;72:1. doi: 10.1016/s0022-3476(68)80394-4. [DOI] [PubMed] [Google Scholar]
- 1043.Mohieldin MS, Gabr M, el-Hefny A. Bacteriological and clinical studies in infantile diarrhoea. II. Doubtful pathogens: Enterobacteriaceae, Pseudomonas, Alcaligenes and Aeromonas. J Trop Pediatr. 1966;11:88. doi: 10.1093/oxfordjournals.tropej.a057181. [DOI] [PubMed] [Google Scholar]
- 1044.Singer JM, Bar-Hay J, Hoenigsberg R. The intestinal flora in the etiology of infantile infectious diarrhea. Am J Dis Child. 1955;89:531. doi: 10.1001/archpedi.1955.02050110645002. [DOI] [PubMed] [Google Scholar]
- 1045.Williams S. The bacteriological considerations of infantile enteritis in Sydney. Med J Aust. 1951;2:137. doi: 10.5694/j.1326-5377.1951.tb68173.x. [DOI] [PubMed] [Google Scholar]
- 1046.Ujvary G, Voros S, Angyal T. Beobachtungen über die Ätiologie der Gastroenterocolitiden des Säuglings- und Kindesalters. IV. Untersuchung der Rolle der Proteus morgani-Stamme. Acta Microbiol Acad Sci Hung. 1964;10:327. [PubMed] [Google Scholar]
- 1047.Sharpe ME. Group D streptococci in the faeces of healthy infants and of infants with neonatal diarrhea. J Hyg. 1952;50:209. doi: 10.1017/s0022172400019550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1048.Kohler H, Kite P. Neonatal enteritis due to Providencia organisms. Arch Dis Child. 1970;45:709. doi: 10.1136/adc.45.243.709-c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1049.Ridge LEL, Thomas MEM. Infection with the Providence type of Paracolon bacillus in a residential nursery. J Pathol Bacteriol. 1955;69:335. [PubMed] [Google Scholar]
- 1050.Bhat P, Myers RM, Feldman RA. Providence group of organisms in the aetiology of juvenile diarrhoea. Indian J Med Res. 1971;59:1010. [PubMed] [Google Scholar]
- 1051.Bishop RF, Barnes GL, Townley RRW. Microbial flora of stomach and small intestine in infantile gastroenteritis. Acta Paediatr Scand. 1974;63:418. doi: 10.1111/j.1651-2227.1974.tb04820.x. [DOI] [PubMed] [Google Scholar]
- 1052.Chaudhury A, Nath G, Shukla B. Diarrhoea associated with Candida spp.: incidence and seasonal variation. J Diarrhoeal Dis Res. 1996;14:110. [PubMed] [Google Scholar]
- 1053.Enweani I, Obi C, Jokpeyibo M. Prevalence of Candida species in Nigerian children with diarrhea. J Diarrhoeal Dis Res. 1994;12:133. [PubMed] [Google Scholar]
- 1054.Klingspor L, Stitzing G, Johansen K. Infantile diarrhea and malnutrition associated with Candida in a developing community. Mycoses. 1993;36:19. doi: 10.1111/j.1439-0507.1993.tb00682.x. [DOI] [PubMed] [Google Scholar]
- 1055.Ponnuvel K, Rajkumar R, Menon T, Sankaranarayanan V. Role of Candida in indirect pathogenesis of antibiotic associated diarrhea in infants. Mycopathologia. 1996;135:145. doi: 10.1007/BF00632335. [DOI] [PubMed] [Google Scholar]
- 1056.Omoike IU, Abiodun PO. Upper small intestine microflora in diarrhea and malnutrition in Nigerian children. J Pediatr Gastroenterol Nutr. 1989;9:314. doi: 10.1097/00005176-198910000-00009. [DOI] [PubMed] [Google Scholar]
- 1057.Kane JG, Chretien JH, Garagusi VF. Diarrhea caused by Candida. Lancet. 1976;1:335. doi: 10.1016/s0140-6736(76)90087-8. [DOI] [PubMed] [Google Scholar]
- 1058.VonGerloczy F, Schmidt K, Scholz M. Beiträge zur Frage der Moniliasis in Säuglingsalter. Ann Pediatr (Paris) 1956;187:119. [PubMed] [Google Scholar]
- 1059.Hill HR, Mitchell TG, Matsen JM. Recovery from disseminated candidiasis in a premature neonate. Pediatrics. 1974;53:748. [PubMed] [Google Scholar]
- 1060.Faix RG. Systemic Candida infections in infants in intensive care nurseries: high incidence of central nervous system involvement. J Pediatr. 1984;105:616. doi: 10.1016/s0022-3476(84)80433-3. [DOI] [PubMed] [Google Scholar]
- 1061.Baley JE, Kliegman RM, Fanaroff AA. Disseminated fungal infections in very low birth weight infants: clinical manifestations and epidemiology. Pediatrics. 1984;73:144. [PubMed] [Google Scholar]
- 1062.Struelens MJ, Bennish ML, Mondal G. Bacteremia during diarrhea: incidence, etiology, risk factors, and outcome. Am J Epidemiol. 1991;133:451. doi: 10.1093/oxfordjournals.aje.a115912. [DOI] [PubMed] [Google Scholar]
- 1063.Rodriguez-García R, Rodriguez-Guzman LM, Sanchez-Maldonado MI. Prevalence and risk factors associated with intestinal parasitoses in pregnant women and their relation to the infant’s birth weight. Ginecol Obstet Mex. 2002;70:338. [PubMed] [Google Scholar]
- 1064.Guven A. Amebiasis in the newborn. Indian J Pediatr. 2003;70:437. doi: 10.1007/BF02723620. [DOI] [PubMed] [Google Scholar]
- 1065.Axton JHM. Amoebic proctocolitis and liver abscess in a neonate. S Afr Med J. 1972;46:258. [PubMed] [Google Scholar]
- 1066.Botman T, Rusy PJ. Amoebic appendicitis in a newborn infant. Trop Geogr Med. 1963;15:221. [PubMed] [Google Scholar]
- 1067.Hsiung CC. Amebiasis of the newborn: report of three cases. Chin J Pathol. 1958;4:14. [Google Scholar]
- 1068.Dykes AC, Ruebush TK, Gorelkin L. Extraintestinal amebiasis in infancy: report of three patients and epidemiologic investigations of their families. Pediatrics. 1980;65:799. [PubMed] [Google Scholar]
- 1069.Gomez NA, Cozzarelli R, Alvarez LR. Amebic liver abscess in newborn. Report of a case. Acta Gastroenterol Latinoam. 1999;29:115. [PubMed] [Google Scholar]
- 1070.Rennert W, Ray C. Fuliminant amebic colitis in a ten-day-old infant. Arch Pediatr. 1997;4:92. doi: 10.1097/00006454-200011000-00023. [DOI] [PubMed] [Google Scholar]
- 1071.Kotcher E, Mata LJ, Esquivel R. Acquisition of intestinal parasites in newborn human infants. Fed Proc. 1965;24:442. (abstract). [Google Scholar]
- 1072.Ravdin JI. Amebiasis. Clin Infect Dis. 1995;20:1453. doi: 10.1093/clinids/20.6.1453. [DOI] [PubMed] [Google Scholar]
- 1073.Mirelman D, Nuchamowitz Y, Stolarsky T. Comparison of use of enzyme-linked immunosorbent assay–based kits and PCR amplification of rRNA genes for simultaneous detection of Entamoeba histolytica and E. dispar. J Clin Microbiol. 1997;35:2405. doi: 10.1128/jcm.35.9.2405-2407.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1074.Haque R, Ali IKM, Petri WA., Jr Comparison of PCR, isoenzyme analysis, and antigen detection for diagnosis of Entamoeba histolytica infection. J Clin Microbiol. 1998;36:449. doi: 10.1128/jcm.36.2.449-452.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1075.Black RE, Dykes AC, Sinclair SP. Giardiasis in day care centers. Evidence of person-to-person transmission. Pediatrics. 1977;60:486. [PubMed] [Google Scholar]
- 1076.Keystone JS, Krajden S, Warren MR. Person-to-person transmission of G. lamblia in day care nurseries. Can Med Assoc J. 1978;119:241. [PMC free article] [PubMed] [Google Scholar]
- 1077.Pickering LK, Evans DG, DuPont HL. Diarrhea caused by Shigella, rotavirus, and Giardia in day care centers: prospective study. J Pediatr. 1981;99:51. doi: 10.1016/s0022-3476(81)80956-0. [DOI] [PubMed] [Google Scholar]
- 1078.Pickering LK, Woodward WE, DuPont HL. Occurrence of G. lamblia in children in day care centers. J Pediatr. 1984;104:522. doi: 10.1016/s0022-3476(84)80540-5. [DOI] [PubMed] [Google Scholar]
- 1079.Adam RD. The biology of Giardia spp. Microbiol Rev. 1991;55:706. doi: 10.1128/mr.55.4.706-732.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1080.Pickering LK, Engelkirk PG. Giardia lamblia. Pediatr Clin North Am. 1988;35:565. doi: 10.1016/s0031-3955(16)36472-0. [DOI] [PubMed] [Google Scholar]
- 1081.Miotti PG, Gilman RH, Santosham M. Age-related rate of seropositivity and antibody to Giardia lamblia in four diverse populations. J Clin Microbiol. 1986;24:972. doi: 10.1128/jcm.24.6.972-975.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1082.Islam A, Stoll BJ, Ljungstrom I. Giardia lamblia infections in a cohort of Bangladeshi mothers and infants followed for one year. J Pediatr. 1983;103:996. doi: 10.1016/s0022-3476(83)80739-2. [DOI] [PubMed] [Google Scholar]
- 1083.Gendrel D, Richard-Lenoble D, Kombila M. Giardiasis and breastfeeding in urban Africa. Pediatr Infect Dis J. 1989;8:58. doi: 10.1097/00006454-198901000-00018. [DOI] [PubMed] [Google Scholar]
- 1084.Stevens DP, Frank DM. Local immunity in murine giardiasis: is milk protective at the expense of maternal gut? Trans Assoc Am Physicians. 1978;91:268. [PubMed] [Google Scholar]
- 1085.Andrews JS, Jr, Hewlett EL. Protection against infection with Giardia muris by milk containing antibody to Giardia. J Infect Dis. 1981;143:242. doi: 10.1093/infdis/143.2.242. [DOI] [PubMed] [Google Scholar]
- 1086.Rohrer L, Winterhalter KH, Eckert J. Killing of G. lamblia by human milk mediated by unsaturated fatty acids. Antimicrob Agents Chemother. 1986;30:254. doi: 10.1128/aac.30.2.254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1087.Current WL, Garcia LS. Cryptosporidiosis. Clin Microbiol Rev. 1991;4:325. doi: 10.1128/cmr.4.3.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1088.Heyworth MF. Immunology of Giardia and Cryptosporidium infections. J Infect Dis. 1992;166:465. doi: 10.1093/infdis/166.3.465. [DOI] [PubMed] [Google Scholar]
- 1089.Wolfson IS, Richter JM, Waldron MA. Cryptosporidiosis in immunocompetent patients. N Engl J Med. 1985;312:1278. doi: 10.1056/NEJM198505163122002. [DOI] [PubMed] [Google Scholar]
- 1090.Tzipori S. Cryptosporidiosis in animals and humans. Microbiol Rev. 1983;47:84. doi: 10.1128/mr.47.1.84-96.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1091.Stehr-Green JK, McCaig L, Remsen HM. Shedding of oocysts in immunocompetent individuals infected with Cryptosporidium. Am J Trop Med Hyg. 1987;36:338. doi: 10.4269/ajtmh.1987.36.338. [DOI] [PubMed] [Google Scholar]
- 1092.Soave R, Ma P. Cryptosporidiosis travelers’ diarrhea in two families. Arch Intern Med. 1985;145:70. doi: 10.1001/archinte.145.1.70. [DOI] [PubMed] [Google Scholar]
- 1093.Collier AC, Miller RA, Meyers JD. Cryptosporidiosis after marrow transplantation, person-to-person transmission and treatment with spiramycin. Ann Intern Med. 1984;101:205. doi: 10.7326/0003-4819-101-2-205. [DOI] [PubMed] [Google Scholar]
- 1094.Navin TR. Cryptosporidiosis in humans: review of recent epidemiologic studies. Eur J Epidemiol. 1985;1:77. doi: 10.1007/BF00141796. [DOI] [PubMed] [Google Scholar]
- 1095.Alpert G, Bell LM, Kirkpatrick CE. Outbreak of cryptosporidiosis in a day care center. Pediatrics. 1986;77:152. [PubMed] [Google Scholar]
- 1096.Taylor JP, Perdue JN, Dingley D. Cryptosporidiosis outbreak in a day care center. Am J Dis Child. 1986;139:1023. doi: 10.1001/archpedi.1985.02140120069029. [DOI] [PubMed] [Google Scholar]
- 1097.Hoxie NJ, Davis JP, Vergeront JM. Cryptosporidiosis-associated mortality following a massive waterborne outbreak in Milwaukee, Wisconsin. Am J Public Health. 1997;87:2032. doi: 10.2105/ajph.87.12.2032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1098.Jokipii L, Pohiola S, Jokipii AM. Cryptosporidium: a frequent finding in patients with gastrointestinal symptoms. Lancet. 1983;2:358. doi: 10.1016/s0140-6736(83)90341-0. [DOI] [PubMed] [Google Scholar]
- 1099.Current WL, Reese NC, Ernst JV. Human cryptosporidiosis in immunocompetent and immunodeficient persons: studies of an outbreak and experimental transmission. N Engl J Med. 1983;308:1252. doi: 10.1056/NEJM198305263082102. [DOI] [PubMed] [Google Scholar]
- 1100.Enriquez FJ, Avila CR, Santos JI. Cryptosporidium infections in Mexican children: clinical, nutritional, enteropathogenic, and diagnostic evaluations. Am J Trop Med Hyg. 1997;56:254. doi: 10.4269/ajtmh.1997.56.254. [DOI] [PubMed] [Google Scholar]
- 1101.Mata L, Bolanos H, Pizarro D. Cryptosporidiosis in children from some highland Costa Rican rural and urban areas. Am J Trop Med Hyg. 1984;33:24. doi: 10.4269/ajtmh.1984.33.24. [DOI] [PubMed] [Google Scholar]
- 1102.Jokipii L, Jokipii AMM. Timing of symptoms and oocyst excretion in human cryptosporidiosis. N Engl J Med. 1986;313:1643. doi: 10.1056/NEJM198612253152604. [DOI] [PubMed] [Google Scholar]
- 1103.Sallon S, Deckelbaum RI, Schmid II. Cryptosporidium, malnutrition and chronic diarrhea in children. Am J Dis Child. 1988;142:312. doi: 10.1001/archpedi.1988.02150030086027. [DOI] [PubMed] [Google Scholar]
- 1104.Garcia LS, Shimizu RY. Evaluation of nine immunoassay kits (enzyme immunoassay and direct fluorescence) for detection of Giardia lamblia and Cryptosporidium parvum in human fecal specimens. J Clin Microbiol. 1997;35:1526. doi: 10.1128/jcm.35.6.1526-1529.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1105.MacPherson DW, McQueen R. Cryptosporidiosis: multiattribute evaluation of six diagnostic methods. J Clin Microbiol. 1993;31:198. doi: 10.1128/jcm.31.2.198-202.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1106.Rossignol FJ, Ayoub A, Ayers MS. Treatment of diarrhea caused by Cryptosporidium parvum: a prospective of randomized, double-blind, placebo-controlled study of nitazoxanide. J Infect Dis. 2001;184:103. doi: 10.1086/321008. [DOI] [PubMed] [Google Scholar]
- 1107.Agnew DG, Lima AAM, Newman RD. Cryptosporidiosis in northeastern Brazilian children: association with increased diarrhea morbidity. J Infect Dis. 1998;177:754. doi: 10.1086/514247. [DOI] [PubMed] [Google Scholar]
- 1108.Matson DO, O’Ryan ML, Jiang X, Mitchell DK. Rotavirus, enteric adenoviruses, caliciviruses, astroviruses, and other viruses causing gastroenteritis. In: Spector S, Hodinka RL, Young SA, editors. Clinical Virology Manual, 3rd ed. ASM Press; Washington, DC: 2000. p. 270. [Google Scholar]
- 1109.Kapikian AZ, Chanock RM. Rotaviruses. In: Fields BN, Knipe DM, Howley PM, editors. Fields Virology. 3rd ed. Lippincott-Raven Press; Philadelphia: 1996. p. 1657. [Google Scholar]
- 1110.Estes MK. Rotaviruses and their replication. In: Fields BN, Knipe DM, Howley PM, editors. Fields Virology. 3rd ed. Lippincott-Raven Press; Philadelphia: 1996. p. 1625. [Google Scholar]
- 1111.Wilhelmi I, Roman E, Sanchez-Fauquier A. Viruses causing gastroenteritis. Clin Microbiol Infect. 2003;9:247. doi: 10.1046/j.1469-0691.2003.00560.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1112.Bridger JC. Non-group A rotavirus. In: Farthing M, editor. Viruses in the Gut. Smith Kline & French; Welwyn Garden City, UK: 1988. p. 79. [Google Scholar]
- 1113.Desselberger U. Molecular epidemiology of rotavirus. In: Farthing M, editor. Viruses in the Gut. Smith Kline & French; Welwyn Garden City, UK: 1988. p. 55. [Google Scholar]
- 1114.Hoshino Y, Saif LJ, Sereno MM. Infection immunity of piglets to either VP3 or VP7 outer capsid protein confers resistance to challenge with a virulent rotavirus bearing the corresponding antigen. J Virol. 1988;62:74. doi: 10.1128/jvi.62.3.744-748.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1115.Offit PA, Clark HF, Blavat G. Reassortant rotavirus containing structural proteins VP3 and VP7 from different parents are protective against each parental strain. J Virol. 1986;57:376. doi: 10.1128/jvi.60.2.491-496.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1116.Zhou Y, Li L, Okitsu S. Distribution of human rotaviruses, especially G9 strains, in Japan from 1996 to 2000. Microbiol Immunol. 2003;47:591. doi: 10.1111/j.1348-0421.2003.tb03422.x. [DOI] [PubMed] [Google Scholar]
- 1117.O’Ryan ML, Matson DO, Estes MK, Pickering LK. Molecular epidemiology of rotavirus in children attending day care centers in Houston. J Infect Dis. 1990;162:810. doi: 10.1093/infdis/162.4.810. [DOI] [PubMed] [Google Scholar]
- 1118.Ramachandran M, Das B, Vij A. Unusual diversity of human rotavirus G and P genotypes in India. J Clin Microbiol. 1996;34:436. doi: 10.1128/jcm.34.2.436-439.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1119.Unicomb L, Podder G, Gentsch J. Evidence of high-frequency genomic reassortment of group A rotavirus strains in Bangladesh: emergence of type G9 in 1995. J Clin Microbiol. 1999;37:1885. doi: 10.1128/jcm.37.6.1885-1891.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1120.Santos N, Lima R, Pereira C, Gouvea V. Detection of rotavirus types G8 and G10 among Brazilian children with diarrhea. J Clin Microbiol. 1998;36:2727. doi: 10.1128/jcm.36.9.2727-2729.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1121.Palombo E, Bishop R. Genetic an antigenetic characterization of a serotype G6 human rotavirus isolated in Melbourne, Australia. J Med Virol. 1995;47:348. doi: 10.1002/jmv.1890470410. [DOI] [PubMed] [Google Scholar]
- 1122.Pongsuwanna Y, Guntapong R, Chiwakul M. Detection of a human rotavirus with G12 and P[9] specificity in Thailand. J Clin Microbiol. 2002;40:1390. doi: 10.1128/JCM.40.4.1390-1394.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1123.Gentsch JR, Woods PA, Ramachandran M. Review of G and P typing results from a global collection of rotavirus strains: implications for vaccine development. J Infect Dis. 1996;174(Suppl 1):S30. doi: 10.1093/infdis/174.supplement_1.s30. [DOI] [PubMed] [Google Scholar]
- 1124.Kilgore P, Unicomb L, Gentsch R. Neonatal rotavirus infection in Bangladesh strain characterization and risk factors for nosocomial infection. Pediatr Infect Dis J. 1996;15:672. doi: 10.1097/00006454-199608000-00007. [DOI] [PubMed] [Google Scholar]
- 1125.Tam JS, Zeng BJ, Lo SK. Distinct population of rotaviruses circulating among neonates and older infants. J Clin Microbiol. 1990;28:1033. doi: 10.1128/jcm.28.5.1033-1038.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1126.Jain V, Prashar UD, Glass RI, Bhan MK. Epidemiology of rotavirus in India. Indian J Pediatr. 2001;68:855. doi: 10.1007/BF02762113. [DOI] [PubMed] [Google Scholar]
- 1127.Mascarenhas JD, Linhares AC, Gabbay YB, Leite JP. Detection and characterization of rotavirus G and P types from children participating in a rotavirus vaccine trial in Belen, Brazil. Mem Inst Oswald Cruz. 2002;97:113. doi: 10.1590/s0074-02762002000100020. [DOI] [PubMed] [Google Scholar]
- 1128.Widdowson MA, van Doornum GJ, van der Poel WH. An outbreak of diarrhea in a neonatal medium care unit caused by a novel strain of rotavirus: investigation using both epidemiologival and microbiological methods. Infect Control Hosp Epidemiol. 2002;23:665. doi: 10.1086/501991. [DOI] [PubMed] [Google Scholar]
- 1129.Steele D, Reynecke E, de Beer M. Characterization of rotavirus infection in a hospital neonatal unit in Pretoria, South Africa. J Trop Pediatr. 2002;48:161. doi: 10.1093/tropej/48.3.167. [DOI] [PubMed] [Google Scholar]
- 1130.Linhares AC, Mascarenhas JD, Gusmao RH. Neonatal rotavirus infection in Belem, northern Brazil: nosocomial transmission of a P[6] G2 strain. J Med Virol. 2002;67:418. doi: 10.1002/jmv.10089. [DOI] [PubMed] [Google Scholar]
- 1131.Cunliffe NA, Rogerson S, Dove W. Detection and characterization of rotaviruses in hospitalized neonates in Blantyre, Malawi. J Clin Microbiol. 2002;40:1534. doi: 10.1128/JCM.40.4.1534-1537.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1132.Davidson GP, Bishop RF, Townley RR, Holmes IH. Importance of a new virus in acute sporadic enteritis in children. Lancet. 1975;1:242. doi: 10.1016/s0140-6736(75)91140-x. [DOI] [PubMed] [Google Scholar]
- 1133.Bishop RF, Davidson GP, Holmes IH. Virus particles in epithelial cells of duodenal mucosa from children with acute nonbacterial gastroenteritis. Lancet. 1973;2:1281. doi: 10.1016/s0140-6736(73)92867-5. [DOI] [PubMed] [Google Scholar]
- 1134.Holmes IH, Ruck BJ, Bishop RF. Infantile enteritis viruses: morphogenesis and morphology. J Virol. 1975;16:937. doi: 10.1128/jvi.16.4.937-943.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1135.Suzuki H, Konno T. Reovirus-like particles in jejunal mucosa of a Japanese infant with acute infectious nonbacterial gastroenteritis. Tohoku J Exp Med. 1975;115:199. doi: 10.1620/tjem.115.199. [DOI] [PubMed] [Google Scholar]
- 1136.Graham DY, Estes MK. Comparison of methods for immunocytochemical detection of rotavirus infections. Infect Immun. 1979;26:686. doi: 10.1128/iai.26.2.686-689.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1137.Holmes IH, Rodger SM, Schnagl RD. Is lactase the receptor and uncoating enzyme for infantile enteritis (rota) viruses? Lancet. 1976;1:1387. doi: 10.1016/s0140-6736(76)93032-4. [DOI] [PubMed] [Google Scholar]
- 1138.Shepherd RW, Butler DG, Cutz E. The mucosal lesion in viral enteritis. Extent and dynamics of the epithelial response to virus invasion in transmissible gastroenteritis of piglets. Gastroenterology. 1979;76:770. doi: 10.1016/S0016-5085(79)80177-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1139.Cameron DJS, Bishop RF, Veenstra A. Noncultivable viruses and neonatal diarrhea. Fifteen-month survey in a newborn special care nursery. J Clin Microbiol. 1978;8:93. doi: 10.1128/jcm.8.1.93-98.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1140.Lebenthal E. Lactose malabsorption and milk consumption in infants and children. Am J Dis Child. 1979;133:21. doi: 10.1001/archpedi.1979.02130010027001. [DOI] [PubMed] [Google Scholar]
- 1141.Philipps AD. Mechanisms of mucosal injury: human studies. In: Farthing M, editor. Viruses in the Gut. Smith Kline & French; Welwyn Garden City, UK: 1988. p. 30. [Google Scholar]
- 1142.Shepherd RW, Gall DG, Butler DG. Determinants of diarrhea in viral enteritis. The role of ion transport and epithelial changes in the ileum in transmissible gastroenteritis in piglets. Gastroenterology. 1979;76:20. doi: 10.1016/S0016-5085(79)80122-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1143.Hoshino Y, Saif LJ, Kang SY. Identification of group A rotavirus genes associated with virulence of a porcine rotavirus and host range restriction of a human rotavirus in the gnotobiotic piglet model. Virology. 1995;209:274. doi: 10.1006/viro.1995.1255. [DOI] [PubMed] [Google Scholar]
- 1144.Saulsbury FT, Winklestein JA, Yolken RH. Chronic rotavirus infection in immunodeficiency. J Pediatr. 1980;97:61. doi: 10.1016/s0022-3476(80)80131-4. [DOI] [PubMed] [Google Scholar]
- 1145.Stephen J. Functional abnormalities in the intestine. In: Farthing M, editor. Viruses in the Gut. Smith Kline & French; Welwyn Garden City, UK: 1988. p. 41. [Google Scholar]
- 1146.Lundgren O, Peregrin AT, Persson K. Role of the enteric nervous system in the fluid and electrolyte secretion of rotavirus diarrhea. Science. 2000;287:491. doi: 10.1126/science.287.5452.491. [DOI] [PubMed] [Google Scholar]
- 1147.Kerzner B, Kelly MH, Gall DG. Transmissible gastroenteritis: sodium transport and the intestinal epithelium during the course of viral gastroenteritis. Gastroenterology. 1977;72:457. [PubMed] [Google Scholar]
- 1148.Gall DG, Chapman D, Kelly M. Na+ transport in jejunal crypt cells. Gastroenterology. 1977;72:452. [PubMed] [Google Scholar]
- 1149.Ball JM, Tian P, Zeng CQ. Age-dependent diarrhea induced by a rotaviral nonstructural glycoprotein. Science. 1996;272:101. doi: 10.1126/science.272.5258.101. [DOI] [PubMed] [Google Scholar]
- 1150.Ward RL, Mason B, Bernstein D. Attenuation of a human rotavirus vaccine candidate did not correlate with mutations in the NSP4 protein gene. J Virol. 1997;71:6267. doi: 10.1128/jvi.71.8.6267-6270.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1151.Zhang M, Zeng CQ, Dong Y. Mutations in nonstructural glycoprotein NSP4 are associated with altered virus virulence. J Virol. 1998;72:3666. doi: 10.1128/jvi.72.5.3666-3672.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1152.Bishop RF, Barnes GL, Cipriani E. Clinical immunity after neonatal rotavirus infection. A prospective longitudinal study in young children. N Engl J Med. 1983;309:72. doi: 10.1056/NEJM198307143090203. [DOI] [PubMed] [Google Scholar]
- 1153.Bhan MK, Lew JF, Sazawal S. Protection conferred by neonatal rotavirus infection against subsequent rotavirus diarrhea. J Infect Dis. 1993;168:282. doi: 10.1093/infdis/168.2.282. [DOI] [PubMed] [Google Scholar]
- 1154.Chiba S, Nakata S, Urasawa T. Protective effect of naturally acquired homotypic and heterotypic rotavirus antibodies. Lancet. 1986;1:417. doi: 10.1016/s0140-6736(86)92133-1. [DOI] [PubMed] [Google Scholar]
- 1155.Greene KY, Kapikian AZ. Identification of VP7 epitopes associated with protection against human rotavirus illness or shedding in volunteers. J Virol. 1992;66:548. doi: 10.1128/jvi.66.1.548-553.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1156.Hjelt K, Grauballe PC, Paerregaard A. Protective effect of pre-existing rotavirus-specific immunoglobulin A against naturally acquired rotavirus infection in children. J Med Virol. 1987;21:39. doi: 10.1002/jmv.1890210106. [DOI] [PubMed] [Google Scholar]
- 1157.Matson DO, O’Ryan ML, Estes MK. Characterization of serum antibody responses to natural rotavirus infections in children by VP7-specific epitope-blocking assays. J Clin Microbiol. 1992;30:1056. doi: 10.1128/jcm.30.5.1056-1061.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1158.Ward RL, Knowlton DR, Schiff GM. Relative concentrations of serum neutralizing antibody to VP3 and VP7 protein in adults infected with human rotavirus. J Virol. 1988;62:1543. doi: 10.1128/jvi.62.5.1543-1549.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1159.Clemens JD, Ward RL, Rao MR. Seroepidemiologic evaluation of antibodies to rotavirus as correlates of the risk of clinically significant rotavirus diarrhea in rural Bangladesh. J Infect Dis. 1992;165:161. doi: 10.1093/infdis/165.1.161. [DOI] [PubMed] [Google Scholar]
- 1160.Matson DO, O’Ryan M, Herrera I. Fecal antibody responses to symptomatic and asymptomatic rotavirus infections. J Infect Dis. 1993;167:557. doi: 10.1093/infdis/167.3.577. [DOI] [PubMed] [Google Scholar]
- 1161.O’Ryan M, Matson DO, Estes MK, Pickering LK. Anti-rotavirus G type-specific and isotype-specific antibodies in children with natural rotavirus infections. J Infect Dis. 1994;169:504. doi: 10.1093/infdis/169.3.504. [DOI] [PubMed] [Google Scholar]
- 1162.Zheng BJ, Lo SK, Tam J., Jr Prospective study of community-acquired rotavirus infection. J Clin Microbiol. 1989;27:2083. doi: 10.1128/jcm.27.9.2083-2090.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1163.Ward RL, Clemens JD, Knowlton DR. Evidence that protection against rotavirus diarrhea after natural infection is not dependent on serotype-specific neutralizing antibody. J Infect Dis. 1992;166:1251. doi: 10.1093/infdis/166.6.1251. [DOI] [PubMed] [Google Scholar]
- 1164.Totterdell BM, Chrystie IL, Banatvala JE. Cord blood and breast milk antibodies in neonatal rotavirus infection. BMJ. 1980;1:828. doi: 10.1136/bmj.280.6217.828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1165.Yolken RH, Wyatt RG, Zissis G. Epidemiology of human rotavirus types 1 and 2 as studied by enzyme-linked immunosorbent assay. N Engl J Med. 1978;299:1156. doi: 10.1056/NEJM197811232992103. [DOI] [PubMed] [Google Scholar]
- 1166.McLean B, Holmes IH. Transfer of anti-rotaviral antibodies from mothers to their infants. J Clin Microbiol. 1980;12:320. doi: 10.1128/jcm.12.3.320-325.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1167.McLean BS, Holmes IH. Effects of antibodies, trypsin, and trypsin inhibitors on susceptibility of neonates to rotavirus infection. J Clin Microbiol. 1981;13:22. doi: 10.1128/jcm.13.1.22-29.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1168.Brhssow H, Sidoti J, Lerner L. Antibodies to seven rotavirus serotypes in cord sera, maternal sera, and colostrum of German women. J Clin Microbiol. 1991;29:2856. doi: 10.1128/jcm.29.12.2856-2859.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1169.Brhssow H, Benitez O, Uribe F. Rotavirus-inhibitory activity in serial milk samples from Mexican women and rotavirus infections in their children during their first year of life. J Clin Microbiol. 1993;31:593. doi: 10.1128/jcm.31.3.593-597.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1170.Yolken RH, Wyatt RG, Mata L. Secretory antibody directed against rotavirus in human milk-measurement by means of enzyme-linked immunosorbent assay. J Pediatr. 1978;93:916. doi: 10.1016/s0022-3476(78)81211-6. [DOI] [PubMed] [Google Scholar]
- 1171.Yolken RH, Peterson JA, Vonderfecht SL. Human milk mucin inhibits rotavirus replication and prevents experimental gastroenteritis. J Clin Invest. 1992;90:1984. doi: 10.1172/JCI116078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1172.Bishop RF, Barnes GL, Cipriani E. Clinical immunity after neonatal rotavirus infection. A prospective longitudinal study in young children. N Engl J Med. 1983;309:72. doi: 10.1056/NEJM198307143090203. [DOI] [PubMed] [Google Scholar]
- 1173.Santosham M. Neonatal rotavirus infection. Lancet. 1982;1:1070. doi: 10.1016/S0140-6736(82)92123-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1174.Madeley CR, Cosgrove BP, Bell EJ. Stool viruses in babies in Glasgow. 2. Investigation of normal newborns in hospital. J Hyg. 1978;81:285. doi: 10.1017/s0022172400025122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1175.Glasgow JFT, McClure BG, Connolly J. Nosocomial rotavirus gastroenteritis in a neonatal nursery. Ulster Med J. 1978;47:50. [PMC free article] [PubMed] [Google Scholar]
- 1176.Cameron DJS, Bishop RF, Davidson G. New virus associated with diarrhoea in neonates. Med J Aust. 1976;1:85. [PubMed] [Google Scholar]
- 1177.Bryden AS, Thouless ME, Hall CJ. Rotavirus infections in a special-care baby unit. J Infect. 1982;4:43. doi: 10.1016/s0163-4453(82)90988-4. [DOI] [PubMed] [Google Scholar]
- 1178.Grillner L, Broberger U, Chrystie I. Rotavirus infections in newborns: an epidemiological and clinical study. Scand J Infect Dis. 1985;17:349. doi: 10.3109/13813458509058774. [DOI] [PubMed] [Google Scholar]
- 1179.Tufvesson B, Polberger L, Svanberg L. A prospective study of rotavirus infections in neonatal and maternity wards. Acta Paediatr Scand. 1986;75:211. doi: 10.1111/j.1651-2227.1986.tb10186.x. [DOI] [PubMed] [Google Scholar]
- 1180.Hoshino Y, Wyatt RG, Flores J. Serotypic characterization of rotaviruses derived from asymptomatic human neonatal infections. J Clin Microbiol. 1985;21:425. doi: 10.1128/jcm.21.3.425-430.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1181.Crewe E, Murphy AM. Further studies on neonatal rotavirus infection. Med J Aust. 1980;1:61. doi: 10.5694/j.1326-5377.1980.tb134626.x. [DOI] [PubMed] [Google Scholar]
- 1182.Perez-Schael I, Daoud G, White L. Rotavirus shedding by newborn children. J Med Virol. 1984;14:127. doi: 10.1002/jmv.1890140206. [DOI] [PubMed] [Google Scholar]
- 1183.Vial PA, Kotloff KL, Losonsky GA. Molecular epidemiology of rotavirus infection in a room for convalescing newborns. J Infect Dis. 1988;157:668. doi: 10.1093/infdis/157.4.668. [DOI] [PubMed] [Google Scholar]
- 1184.Haffejee IE. Neonatal rotavirus infections. Rev Infect Dis. 1991;13:957. doi: 10.1093/clinids/13.5.957. [DOI] [PubMed] [Google Scholar]
- 1185.Rodriguez WJ, Kim HW, Brandt CD. Rotavirus: a cause of nosocomial infection in a nursery. J Pediatr. 1982;101:274. doi: 10.1016/s0022-3476(82)80140-6. [DOI] [PubMed] [Google Scholar]
- 1186.Jesudoss ES, John TJ, Maiya PP. Prevalence of rotavirus infection in neonates. Indian J Med Res. 1979;70:863. [PubMed] [Google Scholar]
- 1187.Bishop RF, Cameron DJS, Veenstra AA. Diarrhea and rotavirus infection associated with differing regimens for postnatal care of newborn babies. J Clin Microbiol. 1979;9:525. doi: 10.1128/jcm.9.4.525-529.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1188.Pickering LK, Bartlett AV, Reves RR. Asymptomatic rotavirus before and after rotavirus diarrhea in children in day care centers. J Pediatr. 1988;112:361. doi: 10.1016/s0022-3476(88)80313-5. [DOI] [PubMed] [Google Scholar]
- 1189.Vesikari T, Sarkkinen HK, Maki M. Quantitative aspects of rotavirus excretion in childhood diarrhoea. Acta Paediatr Scand. 1981;70:717. doi: 10.1111/j.1651-2227.1981.tb05774.x. [DOI] [PubMed] [Google Scholar]
- 1190.Konno T, Suzuki H, Katsushima N. Influence of temperature and relative humidity on human rotavirus infection in Japan. J Infect Dis. 1983;147:125. doi: 10.1093/infdis/147.1.125. [DOI] [PubMed] [Google Scholar]
- 1191.Bartlett AV, III, Reeves RR, Pickering LK. Rotavirus in infant-toddler day care centers: epidemiology relevant to disease control strategies. J Pediatr. 1988;113:435. doi: 10.1016/S0022-3476(88)80624-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1192.Matson DO, Estes MK, Burns JW. Serotype variation of human group A rotaviruses in two regions of the United States. J Infect Dis. 1990;162:605. doi: 10.1093/infdis/162.3.605. [DOI] [PubMed] [Google Scholar]
- 1193.Ryan MJ, Ramsay M, Brown D. Hospital admissions attributable to rotavirus infection in England. J Infect Dis. 1996;174(Suppl 1):S12. doi: 10.1093/infdis/174.supplement_1.s12. [DOI] [PubMed] [Google Scholar]
- 1194.Glass R, Kilgore PE, Holmans RC. The epidemiology of rotavirus diarrhea in the United States: surveillance and estimates of disease burden. J Infect Dis. 1996;74(Suppl 1):S5. doi: 10.1093/infdis/174.supplement_1.s5. [DOI] [PubMed] [Google Scholar]
- 1195.O’Ryan M, Pérez-Schael I, Mamani N. Rotavirus-associated medical visits and hospitalizations in South America: a prospective study at three large sentinel hospitals. Pediatr Infect Dis J. 2001;20:685. doi: 10.1097/00006454-200107000-00009. [DOI] [PubMed] [Google Scholar]
- 1196.Duffy LC, Riepenhoff-Talty M, Byers TE. Modulation of rotavirus enteritis during breast-feeding. Am J Dis Child. 1986;140:1164. doi: 10.1001/archpedi.1986.02140250090041. [DOI] [PubMed] [Google Scholar]
- 1197.van Renterghem L, Borre P, Tilleman J. Rotavirus and other viruses in the stool of premature babies. J Med Virol. 1980;5:137. doi: 10.1002/jmv.1890050207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1198.Murphy AM, Albrey MB, Crewe EB. Rotavirus infections in neonates. Lancet. 1977;2:1149. doi: 10.1016/S0140-6736(77)91538-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1199.Soenarto Y, Sebodo T, Ridho R. Acute diarrhea and rotavirus infection in newborn babies and children in Yogyakarta, Indonesia from June 1978 to June 1979. J Clin Microbiol. 1981;14:123. doi: 10.1128/jcm.14.2.123-129.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1200.Appleton H, Buckley M, Robertson MH. A search for faecal viruses in newborn and other infants. J Hyg. 1978;81:279. doi: 10.1017/s0022172400025110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1201.Schnagl RD, Morey F, Holmes IH. Rotavirus and coronavirus-like particles in aboriginal and non-aboriginal neonates in Kalgoorlie and Alice Springs. Med J Aust. 1979;2:178. doi: 10.5694/j.1326-5377.1979.tb142026.x. [DOI] [PubMed] [Google Scholar]
- 1202.Grillner L, Broberger U, Chrystie I. Rotavirus infections in newborns: an epidemiological and clinical study. Scand J Infect Dis. 1985;17:349. doi: 10.3109/13813458509058774. [DOI] [PubMed] [Google Scholar]
- 1203.Dearlove J, Latham P, Dearlove B. Clinical range of neonatal rotavirus gastroenteritis. BMJ. 1983;286:1473. doi: 10.1136/bmj.286.6376.1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1204.Parashar UD, Holman RC, Bresee JS. Epidemiology of diarrheal disease among children enrolled in four West Coast health maintenance organizations. Pediatr Infect Dis J. 1998;17:605. doi: 10.1097/00006454-199807000-00006. [DOI] [PubMed] [Google Scholar]
- 1205.Leece JC, King MW, Dorsey WE. Rearing regimen producing piglet diarrhea (rotavirus) and its relevance to acute infantile diarrhea. Science. 1978;199:776. doi: 10.1126/science.203032. [DOI] [PubMed] [Google Scholar]
- 1206.Santosham M, Yolken RH, Quiroz E. Detection of rotavirus in respiratory secretions of children with pneumonia. J Pediatr. 1983;103:583. doi: 10.1016/s0022-3476(83)80591-5. [DOI] [PubMed] [Google Scholar]
- 1207.Prince DS, Astry C, Vonderfecht S. Aerosol transmission of experimental rotavirus infection. Pediatr Infect Dis J. 1986;5:218. doi: 10.1097/00006454-198603000-00012. [DOI] [PubMed] [Google Scholar]
- 1208.Steele AD, Alexander JJ. Molecular epidemiology of rotavirus in black infants in South Africa. J Clin Microbiol. 1987;25:2384. doi: 10.1128/jcm.25.12.2384-2387.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1209.Rodriguez WJ, Kim HW, Brandt CD. Use of electrophoresis of RNA from human rotavirus to establish the identity of stains involved in outbreaks in a tertiary care nursery. J Infect Dis. 1983;148:34. doi: 10.1093/infdis/148.1.34. [DOI] [PubMed] [Google Scholar]
- 1210.Srivinasan G, Azarcon E, Muldoon Rotavirus infection in a normal nursery: epidemic and surveillance. Infect Control. 1984;5:478. doi: 10.1017/s0195941700060884. [DOI] [PubMed] [Google Scholar]
- 1211.Gerna G, Forster J, Parea M. Nosocomial outbreak of neonatal gastroenteritis caused by a new serotype 4, subtype 4B human rotavirus. J Med Virol. 1990;31:175. doi: 10.1002/jmv.1890310302. [DOI] [PubMed] [Google Scholar]
- 1212.Tallet S, MacKenzie C, Middleton P. Clinical, laboratory, and epidemiologic features of a viral gastroenteritis in infants and children. Pediatrics. 1977;60:217. [PubMed] [Google Scholar]
- 1213.Hieber JP, Shelton S, Nelson JD. Comparison of human rotavirus disease in tropical and temperate settings. Am J Dis Child. 1978;132:853. doi: 10.1001/archpedi.1978.02120340029004. [DOI] [PubMed] [Google Scholar]
- 1214.Mutanda LN. Epidemiology of acute gastroenteritis in early childhood in Kenya. VI. Some clinical and laboratory characteristics relative to the aetiological agents. East Afr Med J. 1980;57:599. [PubMed] [Google Scholar]
- 1215.Whyte RK, Homes R, Pennock CA. Faecal excretion of oligosaccharides and other carbohydrates in normal neonates. Arch Dis Child. 1978;53:913. doi: 10.1136/adc.53.11.913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1216.Hyams JS, Krause PJ, Gleason PA. Lactose malabsorption following rotavirus infection in young children. J Pediatr. 1981;99:916. doi: 10.1016/s0022-3476(81)80020-0. [DOI] [PubMed] [Google Scholar]
- 1217.Prashar UD, Hummelman EG, Breese J. Global illness and deaths caused by rotavirus disease in children. Emerg Infect Dis. 2003;9:565. doi: 10.3201/eid0905.020562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1218.Dani C, Trevisanuto D, Cantarutti F, Zanardo V. A case of neonatal necrotizing enterocolitis due to rotavirus. Pediatr Med Chir. 1994;16:185. [PubMed] [Google Scholar]
- 1219.Goma Brufau AR, Vega Romero M, Martinez Ubieto P. Epidemic outbreak of necrotizing enterocolitis coincident with an epidemic of neonatal rotavirus gastroenteritis. An Esp Pediatr. 1988;29:307. [PubMed] [Google Scholar]
- 1220.Riedel F, Kroener T, Stein K. Rotavirus infection and bradycardia-apnoea-episodes in the neonate. Eur J Pediatr. 1996;155:36. doi: 10.1007/BF02115624. [DOI] [PubMed] [Google Scholar]
- 1221.Konno T, Suzuki H, Kutsuzawa T. Human rotavirus infection in infants and young children with intussusception. J Med Virol. 1978;2:265. doi: 10.1002/jmv.1890020310. [DOI] [PubMed] [Google Scholar]
- 1222.Mulcahy DL, Kamath KR, de Silva LM. A two-part study of the aetiological role of rotavirus in intussusception. J Med Virol. 1982;9:51. doi: 10.1002/jmv.1890090108. [DOI] [PubMed] [Google Scholar]
- 1223.Nicolas JC, Ingrand D, Fortier B, Bricout F. A one-year virological survey of acute intussusception in childhood. J Med Virol. 1982;9:267. doi: 10.1002/jmv.1890090404. [DOI] [PubMed] [Google Scholar]
- 1224.Murphy TV, Gargiullo PM, Massoudi MS. Intussusception among infants given an oral rotavirus vaccine. N Engl J Med. 2001;22:564. doi: 10.1056/NEJM200102223440804. [DOI] [PubMed] [Google Scholar]
- 1225.Dennehy P, Hartin M, Nelson S, Reising S. Evaluation of the inmunocardstat. Rotavirus assay for detection of group A rotavirus in fecal specimens. J Clin Microbiol. 1999;37:1977. doi: 10.1128/jcm.37.6.1977-1979.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1226.Gilchrist MJR, Bretl TS, Moultney K. Comparison of seven kits for detection of rotavirus in fecal specimens with a sensitive, specific enzyme immunoassay. Diagn Microbiol Infect Dis. 1987;8:221. doi: 10.1016/0732-8893(87)90053-8. [DOI] [PubMed] [Google Scholar]
- 1227.Knisley CV, Bednarz-Prashad A, Pickering LK. Detection of rotavirus in stool specimens with monoclonal and polyclonal antibody-based assay systems. J Clin Microbiol. 1986;23:897. doi: 10.1128/jcm.23.5.897-900.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1228.Thomas EE, Puterman ML, Kawano E. Evaluation of seven immunoassays for detection of rotavirus in pediatric stool samples. J Clin Microbiol. 1988;26:1189. doi: 10.1128/jcm.26.6.1189-1193.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1229.Miotti PG, Eiden J, Yolken RH. Comparative efficacy of commercial immunoassays for the diagnosis of rotavirus gastroenteritis during the course of infection. J Clin Microbiol. 1985;22:693. doi: 10.1128/jcm.22.5.693-698.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1230.Brandt CD, Kim HW, Rodriguez WJ. Comparison of direct electron microscopy, immune electron microscopy, and rotavirus enzyme-linked immunosorbent assay for detection of gastroenteritis viruses in children. J Clin Microbiol. 1981;13:976. doi: 10.1128/jcm.13.5.976-981.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1231.Yolken RH, Kim HW, Clem T. Enzyme-linked immunosorbent assay (ELISA) for detection of human reovirus-like agent of infantile gastroenteritis. Lancet. 1977;2:263. doi: 10.1016/s0140-6736(77)90951-5. [DOI] [PubMed] [Google Scholar]
- 1232.Fischer TK, Steinsland H, Valentiner-Branth P. Rotavirus particles can survive storage in ambient tropical temperatures for more than 2 months. J Clin Microbiol. 2002;40:4763. doi: 10.1128/JCM.40.12.4763-4764.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1233.Viera de Torres B, Mazzali de Ilja R, Esparza J. Epidemiological aspects of rotavirus infection in hospitalized Venezuelan children with gastroenteritis. Am J Trop Med Hyg. 1978;27:567. doi: 10.4269/ajtmh.1978.27.567. [DOI] [PubMed] [Google Scholar]
- 1234.Provisional Committee on Quality Improvement, Subcommittee on Acute Gastroenteritis Practice parameter: the management of acute gastroenteritis in young children. Pediatrics. 1996;97:424. [PubMed] [Google Scholar]
- 1235.Sack DA, Eusof A, Merson MH. Oral hydration in rotavirus diarrhoea: a double-blind comparison of sucrose with glucose electrolyte solution. Lancet. 1978;2:280. doi: 10.1016/s0140-6736(78)91687-2. [DOI] [PubMed] [Google Scholar]
- 1236.Black RE, Merson MH, Taylor PR. Glucose vs. sucrose in oral rehydration solutions for infants and young children with rotavirus-associated diarrhea. Pediatrics. 1981;67:79. [PubMed] [Google Scholar]
- 1237.Saulsbury FT, Winklestein JA, Yolken RH. Chronic rotavirus infection in immunodeficiency. J Pediatr. 1980;97:61. doi: 10.1016/s0022-3476(80)80131-4. [DOI] [PubMed] [Google Scholar]
- 1238.Ebina T, Ohta M, Kanamura Y. Passive immunizations of suckling mice and infants with bovine colostrum containing antibodies to human rotavirus. J Med Virol. 1992;38:117. doi: 10.1002/jmv.1890380209. [DOI] [PubMed] [Google Scholar]
- 1239.Guarino A, Guandalini S, Albano F. Enteral immunoglobulins for treatment of protracted rotaviral diarrhea. Pediatr Infect Dis J. 1991;10:612. doi: 10.1097/00006454-199108000-00010. [DOI] [PubMed] [Google Scholar]
- 1240.Brunser O, Espinoza J, Figueroa G. Field trial of an infant formula containing anti-rotavirus and anti–Escherichia coli milk antibodies from hyperimmunized cows. J Pediatr Gastroenterol Nutr. 1992;15:63. doi: 10.1097/00005176-199207000-00010. [DOI] [PubMed] [Google Scholar]
- 1241.Rosenfeldt V, Fleischer K, Jakobsen M. Effect of probiotic Lactobacillus strains in young children hospitalized with acute diarrhea. Pediatr Infect Dis J. 2002;21:411. doi: 10.1097/00006454-200205000-00012. [DOI] [PubMed] [Google Scholar]
- 1242.Mohan P, Haque K. Oral immunoglobulin for the treatment of rotavirus infection in low birth weight infants. Cochrane Database Syst Rev. 2003;3:CD003742. doi: 10.1002/14651858.CD003740. [DOI] [PubMed] [Google Scholar]
- 1243.Birch CJ, Lewis FA, Kennett ML. A study of the prevalence of rotavirus infection in children with gastroenteritis admitted to an infectious disease hospital. J Med Virol. 1977;1:69. doi: 10.1002/jmv.1890010109. [DOI] [PubMed] [Google Scholar]
- 1244.Kombo LA, Gerber MA, Pickering LK. Intussusception, infection, and immunization: summary of a workshop on rotavirus. Pediatrics. 2001;108:E37. doi: 10.1542/peds.108.2.e37. [DOI] [PubMed] [Google Scholar]
- 1245.Wilson W, Scott RB, Pinto A, Robertson MA. Intractable diarrhea in a newborn infant: microvillous inclusion disease. Can J Gastroenterol. 2001;15:61. doi: 10.1155/2001/743925. [DOI] [PubMed] [Google Scholar]
- 1246.Stockdale EM, Miller CA. Persistent diarrhea as the predominant symptom of Hirschsprung’s disease (congenital dilatation of colon) Pediatrics. 1957;19:91. [PubMed] [Google Scholar]
- 1247.Wilmore DW. Factors correlating with a successful outcome following extensive intestinal resection in newborn infants. J Pediatr. 1972;80:88. doi: 10.1016/s0022-3476(72)80459-1. [DOI] [PubMed] [Google Scholar]
- 1248.Fried D, Gotlieb A, Zaidel L. Intractable diarrhea of infancy due to lymphangiectasis. Am J Dis Child. 1974;127:416. doi: 10.1001/archpedi.1974.02110220114017. [DOI] [PubMed] [Google Scholar]
- 1249.Lebenthal E. Small intestinal disaccharidase deficiency. Pediatr Clin North Am. 1975;22:757. doi: 10.1016/s0031-3955(16)33206-0. [DOI] [PubMed] [Google Scholar]
- 1250.Ament ME, Perera DR, Esther LJ. Sucrase-isomaltose deficiency—a frequently misdiagnosed disease. J Pediatr. 1973;83:721. doi: 10.1016/s0022-3476(73)80362-2. [DOI] [PubMed] [Google Scholar]
- 1251.Marks JF, Norton JB, Fordtran JS. Glucose-galactose malabsorption. J Pediatr. 1969;69:225. doi: 10.1016/s0022-3476(66)80324-4. [DOI] [PubMed] [Google Scholar]
- 1252.Burke V, Anderson CM. Sugar intolerance as a cause of protracted diarrhea following surgery of the gastrointestinal tract in neonates. Aust Paediatr J. 1966;2:219. [Google Scholar]
- 1253.Bishop RF, Davidson GP, Holmes IH. Virus particles in epithelial cells of duodenal mucosa from children with acute non-bacterial gastroenteritis. Lancet. 1973;2:1281. doi: 10.1016/s0140-6736(73)92867-5. [DOI] [PubMed] [Google Scholar]
- 1254.Coello-Ramirez P, Lifshitz F, Zuniga V. Enteric microflora and carbohydrate intolerance in infants with diarrhea. Pediatrics. 1972;49:233. [PubMed] [Google Scholar]
- 1255.Akesode F, Lifshitz F, Hoffman KM. Transient monosaccharide intolerance in a newborn infant. Pediatrics. 1973;51:891. [PubMed] [Google Scholar]
- 1256.Iyngkaran N, Davis K, Robinson MJ. Cow’s milk protein-sensitive enteropathy. An important contributing cause of secondary sugar intolerance in young infants with acute infective enteritis. Arch Dis Child. 1979;54:39. doi: 10.1136/adc.54.1.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1257.Ament ME. Malabsorption syndromes in infancy and childhood. I, II. J Pediatr. 1972;81:685. doi: 10.1016/s0022-3476(72)80087-8. 867. [DOI] [PubMed] [Google Scholar]
- 1258.Whyte RK, Homer R, Pennock CA. Faecal excretion of oligosaccharides and other carbohydrates in normal neonates. Arch Dis Child. 1978;53:913. doi: 10.1136/adc.53.11.913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1259.Schwachman H, Redmond A, Khaw KT. Studies in cystic fibrosis. Report of 130 patients diagnosed under 3 months of age over a 20 year period. Pediatrics. 1970;46:335. [PubMed] [Google Scholar]
- 1260.Aggett PJ, Cavanagh NPC, Matthew DJ. Schwachman’s syndrome. A review of 21 cases. Arch Dis Child. 1980;55:331. doi: 10.1136/adc.55.5.331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1261.Lilibridge CB, Townes PL. Physiologic deficiency of pancreatic amylase in infancy: a factor in iatrogenic diarrhea. J Pediatr. 1973;82:279. doi: 10.1016/s0022-3476(73)80167-2. [DOI] [PubMed] [Google Scholar]
- 1262.Lebenthal E, Antonowicz I, Schwachman H. Enterokinase and trypsin activities in pancreatic insufficiency and diseases of the small intestine. Gastroenterology. 1979;70:508. [PubMed] [Google Scholar]
- 1263.Powell GK, Jones LA, Richardson J. A new syndrome of bile acid deficiency—a possible synthetic defect. J Pediatr. 1973;83:758. doi: 10.1016/s0022-3476(73)80366-x. [DOI] [PubMed] [Google Scholar]
- 1264.Lloyd JK. Disorders of the serum lipoproteins. I. Lipoprotein deficiency states. Arch Dis Child. 1968;43:393. doi: 10.1136/adc.43.230.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1265.Cash R, Berger CK. Acrodermatitis enteropathica: defective metabolism of unsaturated fatty acids. J Pediatr. 1969;74:717. doi: 10.1016/s0022-3476(69)80134-4. [DOI] [PubMed] [Google Scholar]
- 1266.Garretts M, Molokhia M. Acrodermatitis enteropathica without hypozincemia. J Pediatr. 1977;91:492. doi: 10.1016/s0022-3476(77)81333-4. [DOI] [PubMed] [Google Scholar]
- 1267.McReynolds EW, Roy S, III, Etteldorf JN. Congenital chloride diarrhea. Am J Dis Child. 1974;127:566. doi: 10.1001/archpedi.1974.02110230112020. [DOI] [PubMed] [Google Scholar]
- 1268.Minford AMB, Barr DGD. Prostaglandin synthetase inhibitor in an infant with congenital chloride diarrhea. Arch Dis Child. 1980;55:70. doi: 10.1136/adc.55.1.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1269.Woodard JC, Webster PD, Carr AA. Primary hypomagnesemia with secondary hypocalcemia, diarrhea and insensitivity to parathyroid hormone. Am J Dig Dis. 1972;17:612. doi: 10.1007/BF02231747. [DOI] [PubMed] [Google Scholar]
- 1270.Iversen T. Congenital adrenal hyperplasia with disturbed electrolyte regulation. Pediatrics. 1955;16:875. [PubMed] [Google Scholar]
- 1271.Iida Y, Nose O, Kai H. Watery diarrhoea with a vasoactive intestinal peptide–producing ganglioneuroblastoma. Arch Dis Child. 1980;55:929. doi: 10.1136/adc.55.12.929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1272.Ghishan FK, Soper RT, Nassif EG. Chronic diarrhea of infancy: nonbeta islet cell hyperplasia. Pediatrics. 1979;64:46. [PubMed] [Google Scholar]
- 1273.Storm W, Wendel U, Sprenkamp M, Seidler A. Wolman’s disease in an infant. Monatsschr Kinderheilkd. 1990;138:88. [PubMed] [Google Scholar]
- 1274.Hakami N, Neiman PE, Canellos GP. Neonatal megaloblastic anemia due to inherited transcobalamin II deficiency in 2 siblings. N Engl J Med. 1971;285:1163. doi: 10.1056/NEJM197111182852103. [DOI] [PubMed] [Google Scholar]
- 1275.Verloes A, Lombert J, Lambert Y. Tricho-hepato-enteric syndrome: further delineation of a distinct syndrome with neonatal hemochromatosis phenotype, intractable diarrhea, and hair anomalies. Am J Med Genet. 1997;68:391. doi: 10.1002/(sici)1096-8628(19970211)68:4<391::aid-ajmg3>3.0.co;2-p. [DOI] [PubMed] [Google Scholar]
- 1276.Jonas AJ, Butler IJ. Circumvention of defective neutral amino acid transport in Hartnup disease using tryptophan ethyl ester. J Clin Invest. 1989;84:200. doi: 10.1172/JCI114141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1277.Holmberg C, Perheentipa J. Congenital Na+ diarrhea: a new type of secretory diarrhea. J Pediatr. 1985;106:56. doi: 10.1016/s0022-3476(85)80465-0. [DOI] [PubMed] [Google Scholar]
- 1278.Bayna SL, Heiner DC. Cow’s milk allergy: manifestations, diagnosis and management. Adv Pediatr. 1978;25:1. [PubMed] [Google Scholar]
- 1279.Halpin TC, Byrne WJ, Ament ME. Colitis, persistent diarrhea, and soy protein intolerance. J Pediatr. 1977;91:404. doi: 10.1016/s0022-3476(77)81308-5. [DOI] [PubMed] [Google Scholar]
- 1280.Powell GK. Milk- and soy-induced enterocolitis of infancy. Clinical features and standardization of challenge. J Pediatr. 1978;93:553. doi: 10.1016/s0022-3476(78)80887-7. [DOI] [PubMed] [Google Scholar]
- 1281.Miller RC, Larsen E. Regional enteritis in early infancy. Am J Dis Child. 1971;122:301. doi: 10.1001/archpedi.1971.02110040085006. [DOI] [PubMed] [Google Scholar]
- 1282.Avery GB, Harkness M. Bloody diarrhea in the newborn infant of a mother with ulcerative colitis. Pediatrics. 1964;34:875. [PubMed] [Google Scholar]
- 1283.Ein SH, Lynch MJ, Stephens CA. Ulcerative colitis in children under one year: a twenty-year review. J Pediatr Surg. 1971;6:264. doi: 10.1016/0022-3468(71)90466-0. [DOI] [PubMed] [Google Scholar]
- 1284.Sunshine P, Sinatra FR, Mitchell CH. Intractable diarrhoea of infancy. Clin Gastroenterol. 1977;6:445. [PubMed] [Google Scholar]
- 1285.Scott GB, Buck BE, Leterman JG. Acquired immunodeficiency syndrome in infants. N Engl J Med. 1984;310:76. doi: 10.1056/NEJM198401123100202. [DOI] [PubMed] [Google Scholar]
- 1286.Davidson M, Wasserman R. The irritable colon of childhood (chronic nonspecific diarrhea syndrome) J Pediatr. 1966;69:1027. doi: 10.1016/s0022-3476(66)80292-5. [DOI] [PubMed] [Google Scholar]
- 1287.Ebbesen F, Edelsten D, Hertel J. Gut transit time and lactose malabsorption during phototherapy. I, II. Acta Paediatr Scand. 1980;69:65. doi: 10.1111/j.1651-2227.1980.tb07031.x. [DOI] [PubMed] [Google Scholar]
- 1288.Perlman M, Benady S, Saggi E. Neonatal diagnosis of familial dysautonomia. Pediatrics. 1979;63:238. [PubMed] [Google Scholar]
- 1289.Davidson GP, Cutz E, Hamilton JR. Familial enteropathy: a syndrome of protracted diarrhea from birth, failure to thrive, and hypoplastic villous atrophy. Gastroenterology. 1978;75:783. [PubMed] [Google Scholar]
- 1290.Candy DCA, Larcher VF, Cameron DJS. Lethal familial protracted diarrhea. Arch Dis Child. 1981;56:15. doi: 10.1136/adc.56.1.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 1291.Chien L, Robertson H, Gerrard JW. Infantile gastroenteritis due to water with high sulfate content. Can Med Assoc J. 1968;99:102. [PMC free article] [PubMed] [Google Scholar]
- 1292.Fleisher D, Ament ME. Diarrhea, red diapers, and child abuse. Clin Pediatr (Phila) 1978;17:820. doi: 10.1177/000992287701600916. [DOI] [PubMed] [Google Scholar]
- 1293.Ochoa TJ, Salazar-Lindo E, Cleary TG. Management of children with infection-associated persistent diarrhea. Semin Pediatr Infect Dis. 2004;15:229. doi: 10.1053/j.spid.2004.07.003. [DOI] [PubMed] [Google Scholar]
