Skip to main content
Frontiers in Medicine logoLink to Frontiers in Medicine
. 2016 Nov 29;3:60. doi: 10.3389/fmed.2016.00060

A Dormant Microbial Component in the Development of Preeclampsia

Douglas B Kell 1,2,3,*, Louise C Kenny 4,5
PMCID: PMC5126693  PMID: 27965958

Abstract

Preeclampsia (PE) is a complex, multisystem disorder that remains a leading cause of morbidity and mortality in pregnancy. Four main classes of dysregulation accompany PE and are widely considered to contribute to its severity. These are abnormal trophoblast invasion of the placenta, anti-angiogenic responses, oxidative stress, and inflammation. What is lacking, however, is an explanation of how these themselves are caused. We here develop the unifying idea, and the considerable evidence for it, that the originating cause of PE (and of the four classes of dysregulation) is, in fact, microbial infection, that most such microbes are dormant and hence resist detection by conventional (replication-dependent) microbiology, and that by occasional resuscitation and growth it is they that are responsible for all the observable sequelae, including the continuing, chronic inflammation. In particular, bacterial products such as lipopolysaccharide (LPS), also known as endotoxin, are well known as highly inflammagenic and stimulate an innate (and possibly trained) immune response that exacerbates the inflammation further. The known need of microbes for free iron can explain the iron dysregulation that accompanies PE. We describe the main routes of infection (gut, oral, and urinary tract infection) and the regularly observed presence of microbes in placental and other tissues in PE. Every known proteomic biomarker of “preeclampsia” that we assessed has, in fact, also been shown to be raised in response to infection. An infectious component to PE fulfills the Bradford Hill criteria for ascribing a disease to an environmental cause and suggests a number of treatments, some of which have, in fact, been shown to be successful. PE was classically referred to as endotoxemia or toxemia of pregnancy, and it is ironic that it seems that LPS and other microbial endotoxins really are involved. Overall, the recognition of an infectious component in the etiology of PE mirrors that for ulcers and other diseases that were previously considered to lack one.

Keywords: preeclampsia, dormancy, infection, biomarkers, sepsis, coagulopathies, amyloidoses

Introduction

Preeclampsia

Preeclampsia is a multisystem disorder of pregnancy, characterized and indeed defined by the presence of hypertension after 20 weeks’ gestation and before the onset of labor, or postpartum, with either proteinuria or any multisystem complication (19). It is a common condition, affecting some 3–5% of nulliparous pregnant women (8, 10) and is characterized by high mortality levels (1114). There is no known cure other than delivery, and consequently, preeclampsia (PE) also causes significant perinatal morbidity and mortality secondary to iatrogenic prematurity. There are a variety of known risk factors (Table 1) that may be of use in predicting a greater likelihood of developing PE, albeit there are so many, with only very modest correlations, that early-stage (especially the first-trimester) prediction of late-stage PE remains very difficult (8, 1517).

Table 1.

Some known risk factors for preeclampsia [based on part in Ref. (3, 7, 1820)].

Risk factor Comments Selected reference
Nulliparous women Epidemiological observation, suggested biochemical explanations include soluble fms-like tyrosine kinase 1 (sFlt1):placental growth factor (PlGF) ratio (3, 21)
Increased maternal age (especially >35 years) Epidemiological observation, though may be related to existing age-related disease (20, 2225)
History of preeclampsia in previous pregnancy Epidemiological observation, virtually akin to recurrence; among the strongest factors (18, 26)
Multi-fetal gestation, i.e., twins, etc. Extra demands on mother’s circulation; larger placenta, danger of ischemia? Relative risk ~3.5× in nulliparous (2729)
Obesity (especially BMI > 35) Can affect blood pressure (BP) directly, also via intra-abdominal pressure; diabetogenic and inflammatory; possible role for asymmetric dimethylarginine (3033)
Booking diastolic BP > 80 mm Hg An essential part of the later syndrome (19)
Booking proteinuria on at least 1 occasion, or ≥0.3 g/24 h An essential part of the later syndrome (19)
Family history of preeclampsia (mother or sister) Twofold to fivefold increase in likelihood. Genetic factors are said to account for some 50% of the variance, though few properly controlled monozygotic/dizygotic twin studies exist; when done the heritability of PE can be lower to negligible (3439)
Pre-existing medical conditions, including chronic hypertension, diabetes mellitus, antiphospholipid syndrome, thrombophilia, autoimmune disease, renal disease, systemic lupus erythematosus, and infertility These are mainly seen as (other) vascular diseases or comorbidities; however, antiphospholipid antibodies (Hughes’ syndrome) are of special interest as they can have an infectious origin; one-third of women with them will develop PE, and they cause recurrent pregnancy loss (6, 4051)
Urinary tract infection (UTI) An infectious origin for PE is the focus here, and not just from UTI (5254) and see below

See also http://bestpractice.bmj.com/best-practice/monograph/326/diagnosis.html. Note that most of these are risk factors that might and do pre-dispose for other diseases (or are themselves diseases).

It is striking that most of the “risk factors” of Table 1 are, in fact, risk factors for multiple vascular or metabolic diseases, i.e., they merely pre-dispose the individual to a greater likelihood of manifesting the disease or syndrome (in this case PE). Indeed, some of them are diseases. This would be consistent with the well-known comorbidities, e.g., between PE and later cardiovascular disease [e.g., Ref. (5565)], between PE and intracerebral hemorrhage during pregnancy [OR 10.39 (66)], and between PE and stroke postpartum (67, 68). The penultimate row of Table 1 lists a series of diseases that amount to comorbidities, although our interest was piqued by the observation that one-third of patients with antiphospholipid syndrome have PE, and infectious agents with known cross-reacting antigens are certainly one original (external) source of the triggers that cause the antiphospholipid antibodies (43, 44, 47, 69) (and see below). Similarly, in the case of urinary tract infection (UTI), the “risk” factor is a genuine external trigger, a point [following the call by Mignini and colleagues (70) for systematic reviews] that we shall expand on considerably here. [A preprint has been lodged at bioRxiv (71).]

In recent decades, intense investigation has led to the development of a two-stage etiological model for PE, first proposed by Redman (72), in which inadequate remodeling of the spiral arteries in early gestation results in poor placental development (stage one) and the resultant ischemia/reperfusion injury and oxidative stress (73) eventually leads to maternal vascular endothelial cell dysfunction and the maternal manifestations of the disease (stage 2) (72, 7477). However, many clinical inconsistencies challenge the simplicity of this model. For example, while the association between poor placentation and PE is well established, it is not specific. Poor placentation and fetal growth restriction (FGR) frequently present without maternal signs of PE. Moreover, FGR is not a consistent feature of PE. While it is commonly seen in PE presenting at earlier gestations, in PE presenting at term, neonates are not growth restricted and may even be large for dates (78).

Thus, the two-stage model has been further refined by Roberts and others (77, 79, 80) to take into account the heterogeneous nature of PE and the varying contribution from mother and infant to the disorder. We now appreciate that normal pregnancy is characterized by a low-grade systemic inflammatory response and specific metabolic changes, and that virtually all of the features of normal pregnancy are simply exaggerated in PE (8183). There is also widespread acceptance that maternal constitutional and environmental factors (such as obesity) can interact to modulate the risk of PE. Thus, with profoundly reduced placental perfusion (or significant “placental loading”), the generation of stage 2 may require very little contribution from the mother to provide sufficient stress to elicit the maternal syndrome. In this setting, almost any woman will develop PE. Conversely, the woman with extensive predisposing constitutional sensitivity could develop PE with very little reduced perfusion, or minimal “placental loading.” As with many complex disorders, multiple factors can affect disease development positively or negatively, with a convenient representation of the two main negative sources (fetal and maternal) being that of a see-saw (84), as in Figure 1.

Figure 1.

Figure 1

Two main sources (fetal and maternal) can drive a pregnancy toward preeclampsia.

While this explains the inconsistencies of the two-stage model, the precise mechanisms (1) underlying the initial poor placentation and (2) linking placental stress and the maternal syndrome have still not been fully elucidated.

Much recent research in PE has focused on various angiogenic factors, including the pro-angiogenic factors vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) and the two anti-angiogenic proteins, such as soluble endoglin (sEng) and soluble fms-like tyrosine kinase 1 (sFlt-1). Recent data suggest that alterations in circulating angiogenic factors play a pathogenic role in PE. These angiogenic factors tightly regulate angiogenesis and are also essential for maintenance of normal vessel health. Consequently, the synthesis and action of these factors and their receptors in the uterine bed and placenta are essential for normal placental development and pregnancy (85, 86). In PE, increased levels of the anti-angiogenic sFlt-1 and sEng trap circulating VEGF, PlGF, and transforming growth factor-β (TGF-β), respectively. A myriad of data support the idea that circulating levels of these factors alone, or in combination, can be used to predict PE (8789) (and see below under PE biomarkers), but in line with the heterogeneous nature of PE, these data are somewhat inconsistent and their performance as biomarkers seems limited to disease with significant placental loading (8). Therefore, angiogenic dysregulation would appear unlikely to be the sole link between the stressed placenta and endothelial dysfunction and the clinical manifestations of the disease.

Notwithstanding these many inconsistencies, the central role of the placenta as a source of “toxin,” in a condition regarded, and indeed often named, as “toxemia of pregnancy” (9092) cannot be refuted. The uncertainty regarding the nature of the toxin continues, and other placental sources of endothelial dysfunction include syncytiotrophoblast basement membrane fragments (STBM) (93) and endothelial progenitor cells (EPC) (94); an increase of reactive oxygen species over scavenging by antioxidants (95, 96) has also been promoted.

The Bradford Hill criteria for causation of a disease Y by an environmental factor X (97) are as follows: (1) strength of association between X and Y, (2) consistency of association between X and Y, (3) specificity of association between X and Y, (4) experiments verify the relationship between X and Y, (5) modification of X alters the occurrence of Y, and (6) biologically plausible cause and effect relationship.

In general terms (98), if we see that two things (A and B) co-vary in different circumstances, we might infer that A causes B, that B causes A, or that something else (C) causes both B and A, whether in series or parallel. To disentangle temporal relations requires a longitudinal study. The job of the systems biologist doing systems medicine is to uncover the chief actors and the means by which they interact (99), in this way fulfilling the Bradford Hill postulates, a topic to which we shall return at the end.

In infection microbiology, and long predating the Bradford Hill criteria, the essentially equivalent metrics are known [widely, but somewhat inaccurately (100)] as the Koch or Henle–Koch postulates (i.e., criteria). They involve assessing the correlation of a culturable organism with the presence of a disease, the cure of the disease (and its symptoms) upon removal of the organism, and the development of the disease with (re)inoculation of the organism. They are of great historical importance but present us with three main difficulties here. The first is that we cannot apply the third of them to humans for obvious ethical reasons. The second (see also below) and related one is that we cannot usefully apply them in animal models because none of the existing models recapitulates human PE well. Finally, as widely recognized (100107), they cannot be straightforwardly applied when dealing with dormant bacteria or bacteria that are otherwise refractory to culture.

Our solution to this is twofold: (i) we can assess the first two using molecular methods if culturing does not work and (ii) we exploit the philosophy of science principle known as “coherence” (108112). This states that if a series of ostensibly unrelated findings are brought together into a self-consistent narrative and that narrative is thereby strengthened. Our systems approach purposely represents a “coherence” in the sense given.

Overall, known biochemical associations with PE come into four main categories, such as abnormal trophoblast invasion, oxidative stress, inflammation and altered immune response, and anti-angiogenic responses (Figure 2). Each of these can contribute directly to PE, and although they can interact with each other (black arrows), no external or causal source is apparent. Figure 2 has been redrawn from a very nice review by Pennington and colleagues (113), which indicates four main generally accepted “causes” (or at least accompaniments) of PE as the four outer colored circles. As illustrated with the black two-way arrows, many of these also interact with each other. What is missing, in a sense, is then what causes these causes, and that is the nub of our argument here. Since we now know (and describe below) that microbes can affect each of these four general mechanisms, we have added these routes to Figure 1 (using pink arrows) where dormant, resuscitating, or growing microbes are known to contribute.

Figure 2.

Figure 2

There are four main “causes” of preeclampsia, represented by the colored outer circles, and these can also interact with each other. That part of the figure is redrawn from Pennington et al. (113). In addition, we note here, as the theme of this review, that microbes can themselves cause each of the features in the outer colored circles to manifest.

In a similar vein, Magee and colleagues (114) have nicely set down their related analysis of the causes and consequences of PE, with a central focus (redrawn in Figure 3) on endothelial cell activation. While bearing much similarity in terms of overall content to the analysis of Pennington and colleagues (113), and ours above, it again lacks a microbial or infection component as a causative element, but importantly does note that infection and/or inflammation can serve to lower the threshold for PE in cases of inadequate placentation. In our view, microbes can also enter following normal placentation if their dormant microbiome begins to wake up and/or to shed inflammagens.

Figure 3.

Figure 3

Another detailed representation of factors known to cause or accompany PE, redrawn from Magee et al. (114).

Heritability

The question of the extent of heritability of PE (susceptibility) is of interest. Although this seems to vary widely in different studies (Table 1), a number of candidate gene studies (37, 115118) imply that a susceptibility to PE is at least partly heritable, consistent with the variance in all the other “risk factors” of Table 1 [and see Ref. (6)]. As with all the other gene association studies where phenotypic (“lifestyle”) information is absent (119121), it is not possible to ascribe the heritability to genetics alone, as opposed to an interaction of a genetic susceptibility (e.g., in the HLA system) with environmental factors (117), such as cytomegalovirus infection (122).

Inflammation

Preeclampsia is accompanied by oxidative stress (123, 124) and inflammation and, thus, shares a set of observable properties with many other (and hence related) inflammatory diseases, be they vascular (e.g., atherosclerosis), neurodegenerative (e.g., Alzheimer’s, Parkinson’s), or “metabolic” (type 1 and 2 diabetes). It is thus at least plausible that they share some common etiologies, as we argue here, and that knowledge of the etiology of these diseases may give us useful clues for PE.

As well as raised levels of inflammatory cytokines that constitute virtually a circular definition of inflammation, we and others have noted that all of these diseases are accompanied by dysregulation of iron metabolism (84, 125, 126), hypercoagulability and hypofibrinolysis (127, 128), blood microparticles (126), and changes in the morphology of fibrin fibers [e.g., Ref. (129134)] and of erythrocytes [e.g., Ref. (127, 132137)].

In addition, we and others have recognized the extensive evidence for the role of a dormant blood and/or tissue microbiome in these (138143) and related (144147) diseases, coupled in part with the shedding of highly inflammagenic bacterial components such as Gram-negative lipopolysaccharide (LPS) and their Gram-positive cell wall equivalents such as lipoteichoic acids (148). (We shall often use the term “LPS” as a “shorthand,” to be illustrative of all of these kinds of highly inflammagenic molecules.)

The purpose of this review, outlined as a “mind map” in Figure 4, is thus to summarize the detailed and specific lines of evidence suggesting a very important role of a dormant microbial component in the etiology of PE [and see also Ref. (138)]. To do this, we must start by rehearsing what is meant by microbial dormancy.

Figure 4.

Figure 4

A mind map of the overall structure of the review.

Bacterial Viability

In microbiology, we usually consider microbes as being in one of the three “physiological macrostates” (Figure 5). The definition of a “viable” bacterium is normally based on its ability to replicate, i.e., “viability” = culturability (149151). In this sense, classical microbiology has barely changed since the time of Robert Koch, with the presence of a “viable” microorganism in a sample being assessed via its ability to form a visible colony on an agar plate containing suitable nutrients. However, it is well known, especially in environmental microbiology [“the great plate count anomaly” (152)], that only a small percentage of cells observable microscopically is typically culturable on agar plates. In principle, this could be because they are/were “irreversibly” non-culturable (operationally “dead”), or because our culture media either kill them (153) or such media lack nutrients or signaling molecules necessary for their regrowth (154, 155) from an otherwise dormant state (156, 157). These statements are true even for microbes that appear in culture collections and (whose growth requirements) would be regarded as “known.”

Figure 5.

Figure 5

The chief physiological macrostates exhibited by microorganisms.

However, it is common enough in clinical microbiology that we detect the existence or presence of “novel” microbial pathogens with obscure growth requirements before we learn to culture them; this is precisely what happened in the case of Legionella pneumophila (158161), Tropheryma whipplei [Whipple’s disease (162, 163)], and Coxiella burnetii [the causative agent of Q fever (164, 165)]. Even Helicobacter pylori was finally brought into culture on agar plates only because an unusually long Easter holiday break meant that the plates were incubated for an extended period of 5 days (rather than the normal 2) before being thrown out (166, 167)! Consequently, there is ample precedent for the presence of “invisible” microbes to go unremarked before they are discovered as the true cause of a supposedly non-infectious disease, even when they are perfectly viable (culturable) according to standard analyses.

Dormancy

Dormancy for a microbe is defined operationally as a state, commonly of low metabolic activity, in which the organism appears not to be viable in that it is unable to form a colony but where it is not dead in that it may revert to a state in which it can do so, via a process known as resuscitation (156, 157). However, an important issue (and see above) is that dormant bacteria do not typically fulfill the Koch–Henle postulates (100103), and in order for them to do so it is necessary that they be grown or resuscitated. This is precisely what was famously done by Marshall and Warren when they showed that the supposedly non-infectious disease of gastric ulcers was, in fact, caused by a “novel” organism called H. pylori (168, 169). One of the present authors showed in laboratory cultures of actinobacteria that these too could enter a state of true dormancy (170, 171) [as is well known for Mycobacterium tuberculosis, e.g., Ref. (172176)], and could be resuscitated by a secreted growth factor called Rpf (177181). This RPF family has a very highly conserved motif that is extremely immunogenic (182, 183), and it is presently under trials as a vaccine against M. bovis.

Prevalence of Dormant, Persistent, or Latent Bacteria in Infection Microbiology

It is worth stressing here that the presence of dormant or latent bacteria in infection microbiology is well established; one-third of humans carry dormant M. tuberculosis [e.g., Ref. (175, 184187)], most without reactivation, while probably 50–100% are infected with H. pylori, most without getting ulcers or worse (188, 189). As with the risk factors in Table 1, the organisms are merely or equivalently “risk factors” for these infectious diseases and are effectively seen as causative only when the disease is actually manifest.

In a similar vein, so-called persisters are phenotypic variants of infectious microbes that resist antibiotics and can effectively lie in hiding to resuscitate subsequently. This is also very well established [e.g., Ref. (139, 190203)]. In many cases, they can hide intracellularly (204), where antibiotics often penetrate poorly (205) because the necessary transporters (206209) are absent. This effectively provides for reservoirs of reinfection, e.g., for Staphylococcus aureus (210), Bartonella spp. (211), and – most pertinently here – for the Escherichia coli involved in urinary tract (re)infection (212215). The same intracellular persistence is true for parasites such as Toxoplasma gondii (216).

Thus, the main point of the extensive prevalence of microbial dormancy and persistence is that microbes can appear to be absent when they are, in fact, present at high concentrations. This is true not only in cases where infection is recognized as the cause of disease but, as we here argue, such microbes may be an important part of diseases presently thought to lack an infectious component.

Iron and Inflammation

It is well known that [with the possible exception of Borrelia (217, 218)] a lack of free iron normally limits microbial growth in vivo [e.g., Ref. (219243)], and we have reviewed previously (84, 125, 126) the very clear iron dysregulation accompanying PE [e.g., Ref. (90, 244256)].

This has led to the recognition (128, 139, 141) that the source of the continuing inflammation might be iron-based resuscitation of dormant microbes that could release well known and highly potent inflammagens such as LPS. Indeed, we have shown that absolutely tiny (highly substoichiometric) amounts of LPS can have a massive effect on the blood clotting process (257), potentially inducing β-amyloid formation directly (258, 259) [something, interestingly, that can be mimicked in liquid crystals (260, 261)]. The overall series of interactions envisaged [see also Kell et al. (139)] is shown in Figure 6.

Figure 6.

Figure 6

An 11-stage systems biology model of the factors that we consider cause initially formant microbes to manifest the symptoms (and disease) of preeclampsia.

As pointed out by a referee, worldwide, iron deficiency anemia is associated with increased perinatal morbidity and mortality, predominantly in low resource settings. This is most recently and comprehensively reviewed by Rahman and colleagues (262). This meta-analysis (262) found that in low- and middle-income countries, maternal hemoglobin concentrations <10 or <11 g dL−1 or hematocrit values <33 or <34% accounted for 12% of low birth weight, 19% of preterm births (PTBs), and 18% of perinatal mortality. However, the independent contribution of iron deficiency is difficult to define, especially since in low resource settings anemia is often not found in isolation but as part of a wider spectrum of nutritional insufficiencies (especially vitamins and folic acid) or as a result of parasitic infection. These can increase susceptibility to other insults such a hemorrhage or infection, also common in low resources settings. In addition, the exact speciation and hence availability of iron may be important (not just the total amount). Thus, the hypothesis presented here is that iron in excess or under certain physiological conditions can be, and indeed is, equally detrimental to maternal health. There is a likely a “Goldilocks” concentration of iron which is optimal for maternal and neonatal health, and the authors are proposing the selected use of treatments such as statins or iron chelators in the at-risk population, rather than as a broad panacea. Furthermore, the use of statins to prevent pregnancy complications is far from controversial (see also below). There are at least two registered current trials of statins to prevent PE (ISRCTN17787139 and ISRCTN23410175).

Detecting Dormant Microbes

By definition, dormant bacteria escape detection by classical methods of assessing viability that involve replication on agar plates. Other growth-associated methods include measurements involving changes in turbidity (263), including an important but now rather uncommon technique referred to as the “most probable number” (MPN). The MPN involves diluting samples serially and assessing by turbidity changes in the presence of growth/no growth. Look-up tables based on Poisson statistics enable estimation of the number of cells or propagules that were present. A particular virtue is that they allow dormant and “initially viable” cells to be discriminated via “dilution to extinction” (171), thereby avoiding many artifacts (157). As mentioned earlier, preincubation in a weak nutrient broth (171, 264) was instrumental in allowing the discovery (177) of an autocrine “wake-up” molecule necessary for the growth of many actinobacteria.

Other more classical means of detecting microbes, but not whether they were culturable, involved microscopy (190, 265268) or flow cytometry (269) with or without various stains that reflected the presence or otherwise of an intact cell wall/membrane (170, 270277). These stains are sometimes referred to as “viability” stains, but this is erroneous as they do not measure “culturability.” Readers may also come upon the term “viable-but-not-culturable”; however, since viable = culturable, this is an oxymoron that we suggest is best avoided (157). Other methods involved measurement of microbial products, e.g., CO2 (278, 279), or changes in the conductivity or impedance of the growth medium (263, 280282).

Most importantly, however, dormant (as well as culturable) cells may be detected by molecular means, nowadays most commonly through PCR and/or sequencing of the DNA encoding their small subunit ribosomal RNA (colloquially “16S”) (283297) or other suitable genes. It is clear that such methods will have a major role to play in detecting, identifying, and quantifying the kinds of microbes that we argue lie at the heart of PE etiology.

A Dormant Blood Microbiome

Of course, actual bacteremia, the presence of replicable bacteria in blood, is highly life-threatening (298), but – as emphasized – viability assays do not detect dormant bacteria. When molecular detection methods are applied to human blood, it turns out that blood does indeed harbor a great many dormant bacteria [e.g., Ref. (299309)]; they may also be detected ultramicroscopically [e.g., Ref. (139141, 190, 267, 300, 310)] or by flow cytometry (311), and dormant blood and tissue microbes probably underpin a great many chronic, inflammatory diseases normally considered to lack a microbial component (139141, 144147, 190, 267, 268, 302, 312321). Multiple arguments serve to exclude “contaminants” as the source of the bacterial DNA (141): (1) there are significant differences between the blood microbiomes of individuals harboring disease states and nominally healthy controls, despite the fact that samples are treated identically; (2) the morphological type of organism (e.g., coccus vs. bacillus) seems to be characteristic of particular diseases; (3) in many cases, relevant organisms lurk intracellularly, which is hard to explain by contamination; (4) there are just too many diseases where bacteria have been found to play a role in the pathogenesis, that all of them may be caused by contamination; (5) the actual numbers of cells involved seem far too great to be explicable by contamination; given that blood contains ~5 × 109 erythrocytes mL−1, if there was just one bacterial cell per 50,000 erythrocytes this will equate to 105 bacteria mL−1. These are big numbers, and if the cells were culturable, that number of cells would be the same as that ordinarily defining bacteriuria.

A recent study by Damgaard and colleagues (306) is of particular interest here. Recognizing the strong mismatch between the likelihood of an infection post-transfusion [very high (306)] and the likelihood of detecting culturable microbes in blood bank units (negligible, ca. 0.1%) (306, 322), Damgaard et al. reasoned that our methods of detecting and culturing these microbes might be the problem. Certainly, taking cells from a cooled blood bag and placing them onto an agar plate at room temperature that is directly exposed to atmospheric levels of gaseous O2 is a huge stress leading to the production of “reactive oxygen species” (125, 323), that might plausibly kill any dormant, injured, or even viable microbes. Thus, they incubated samples from blood on a rich medium (trypticase soy agar) for a full week, both aerobically and anaerobically. Subsequent PCR and sequencing allowed them to identify specific microbes in some 35–53% of the samples. Thus, very careful methods need to be deployed to help resuscitate bacteria from physiological states that normally resist culture, even when these bacteria are well-established species. This is very much beginning to happen in environmental microbiology [e.g., Ref. (154, 324326)], and such organisms are rightly seen as important sources of novel bioactives (327, 328).

As reviewed previously (139143), the chief sources of these blood microbes are the gut microbiome, the oral microbiome [periodontitis (329)], and via UTIs. Consequently, if we are to argue that there is indeed a microbial component to PE, we should expect to see some literature evidence for it (53, 54, 138, 330332). In what follows we shall rehearse the fact that it is voluminous.

Direct Evidence for a Role of Infectious Agents in PE

Although we recognize that many of the more molecular methods cannot distinguish culturable from dormant microbes, quite a number of studies have explicitly identified infection as a cause of PE (Table 2). The commonest microbe seems to be H. pylori; while it is most famously associated with gastric ulcers (168, 169, 333), there are many other extragastric manifestations [e.g., Ref. (334342)]. The odds ratio of no less than 26 in PE vs. controls when the strains can produce CagA antigens is especially striking, not least because it provides a mechanistic link to poor trophoblast invasion via a mechanism involving host antibodies to CagA cross-reacting with trophoblasts (343, 344), and circulating (345) in microparticles (346) or endosomes (347, 348).

Table 2.

Many studies have identified a much greater prevalence of infectious agents in the blood or urine of these exhibiting PE than in matched controls.

Microbes Comments Reference
Chlamydia pneumoniae IgG seroprevalence and gDNA associated with PE (p < 0.0001) (349)
IgG (but not IgA or IgM) associated with PE, OR = 3.1 (350)
Significantly greater numbers with PE, and reversion under antichlamydial treatment (351)
Chlamydia trachomatis Increased risk of PE, OR = 7.2 or 1.6 based on serology (352, 353)
Cytomegalovirus RR for PE 1.5 if infected with CMV (354) [see also Xie et al. (355)]
Helicobacter pylori Seropositivity or DNA. OR = 2.7 or 26 if CagA seropositivity (343) and editorial (356)
IgG seropositivity 54% PE vs. 21% controls (357)
Anti-CagA antibodies cross-react with trophoblasts and could inhibit placentation (358)
2.8× greater seropositivity in PE group (359)
OR = 2.86 for seropositivity in PE, correlated with high malondialdehyde levels (360)
Wide-ranging review of many studies showing PE more prevalent after Hp infection (361)
Seropositivity PE:control = 84:32% (p < 0.001) (362)
OR for seropositivity 1.83 (p < 0.001) (363)
Seropositivity PE:control 86:43% (p < 0.001) (364)
Human papillomavirus High-risk human papillomavirus (HR-HPV) presence implies an OR of 2.18 for PE (365)
Meta-analyses Incidence of PE 19% with asymptomatic bacteriuria vs. 3% (primigravid) or 6% (multigravid) controls (p < 0.005) (366)
UTI more than twice as likely in severe preeclamptics than in controls (367)
OR of 1.6 for PE if UTI present (368)
Increased risk of PE OR 1.57 for UTI, 1.76 for periodontal disease (53)
Early application of antibiotics in infection reduced PE by 52% (330)
Any overt infection led to an RR of 2 for PE (54)
UTI has OR of 3.2 for PE; OR = 4.3 if in third trimester (369)
UTI has OR of 1.3 for mild/moderate and 1.8 for severe PE (370)
Increased risk of PE with UTI (OR 1.22) or antibiotic prescription (OR 1.28) (371)
OR of 6.8 for symptomatic bacteriuria in PE vs. controls (372)
OR 1.3–1.8 of mild or severe PE if exposed to UTI (373)
OR 1.4 for PE following UTI (374)
OR 1.3 for PE after UTI (375)
Meta-analyses showing associations between PD and PE (376378)
Plasmodium falciparum (malaria) Indications that infection with malaria is associated with PE (379)
1.5 RR for PE if malarial (380)
Seasonality: 5.4-fold increase in eclampsia during malaria season (381)
Preeclampsia was significantly associated with malaria infection during pregnancy (p < 0.03) and 69.7% of cases of preeclampsia with infected placenta might be attributable to malaria infection (382)

In contrast to the situation in PE, albeit severe PE is associated with iatrogenic PTBs, there is a widespread recognition [e.g., Ref. (383410)] that infection is a common precursor to PTB in the absence of PE. The failure of antibiotics to help can be ascribed to their difficulty of penetrating to the trophoblasts and placental regions. Unfortunately, no proteomic biomarkers have yet been observed as predictive of PTB (411, 412). In a similar vein, and if we are talking about a time of parturition that is very much more “preterm,” we are in the realm of miscarriages and spontaneous abortions and stillbirths, where infection again remains a major cause (413416). Here, we note that early or pre-emptive antibiotic therapy has also proved of considerable value in improving outcomes after multiple spontaneous abortions (417).

Vaginal, Placental, and Amniotic Fluid Microbiomes in PE

It might be natural to assume that the placenta is a sterile organ, like blood is supposed to be. However, various studies [modulo the usual issues of contamination (418)] have shown the presence of microbes in tissues including the placenta (400, 409, 419432), vagina (393, 433440), uterus (391, 441, 442), amniotic fluid (430, 443448), and follicular fluid (449, 450), and how these may vary significantly in PE [we do not discuss other pregnancy disorders such as small for gestational age (SGA) and intrauterine growth restriction (IUGR)]. We list some of these in Table 3.

Table 3.

Evidence for microbes in placental tissues, including those with PE.

Organisms Comments Reference
Multiple, including Actinobacillus actinomycetemcomitans, Fusobacterium nucleatum Many more in PE placentas relative to controls (p ≤ 0.0055) (451)
Multiple Half of second-trimester pregnancies have culturable or PCR-detectable bacteria/mycoplasmas (419)
Multiple 38% of placental samples were positive for selected bacteria and viruses (452)
Bifidobacterium spp. and Lactobacillus rhamnosus Bifidobacteria and L. rhamnosus (from gut) detected in 31/34 and 33/34 placental samples (420)
Multiple Detectable in 27% of all placentas and 54% of spontaneous preterm delivery (421)
Multiple 16S/NGS, major review (400)
Multiple From 16S and NGS analysis of placental tissue of 7 PE patients (12.5%) (controls all negative) (453)
E. coli and L. monocytogenes When added ex vivo can migrate to extravillous trophoblasts (422)
Multiple Review, with some focus on preterm birth (423)
Multiple Overview, some focus on preterm birth (425)
Multiple Good recent overview, with possible implication of a physiological role (426)
Multiple 320 placentas; changed microbiome as a function of excess gestational weight gain (427)
Multiple One-third of placentas from preterm births were culture-positive (428)
Multiple Major differences in placental microbiome in preterm birth (432)
Plasmodium falciparum (malaria) Increased likelihood of PE (380)
Plasmodium falciparum (malaria) Reviews of placental malaria (454, 455)
Porphyromonas gingivalis OR of PE = 6.3 if detected in umbilical cord (456)
Porphyromonas gingivalis OR 7.59 in placental tissues with hypertensive disorders (457)
Treponema denticola OR 9.39 in placental tissues with hypertensive disorders (457)
Meta-analysis Widespread occurrence of microbes in female genital tract during pregnancy (403)

Origins of a Blood and Tissue Microbiome

As assessed previously (139141) over a large literature, the chief source of blood microbes is the gut (426), with another major entry point being via the oral microbiome (especially in periodontitis, see below). For rheumatoid arthritis (142, 458460) and diseases of pregnancy, UTI (see below and Table TT) also provides a major source.

Gut Origins of Blood Microbes and LPS

We have recently rehearsed these issues elsewhere (139141), so a brief summary will suffice. Clearly, the gut holds trillions of microbes, with many attendant varieties of LPS (461), so even low levels of translocation [e.g., Ref. (462464)], typically via Peyer’s patches and M cells, provide a major source of the blood microbiome. This may be exacerbated by intra-abdominal hypertension and overeating (465467) that can indeed stimulate the translocation of LPS (468). For reasons of space and scope, we do not discuss the origins and translocation of microbes in breast milk (469) nor the important question of the establishment of a well-functioning microbiome in the fetus and neonate (470), and the physiological role of the mother therein.

Preeclampsia and Periodontal Disease

One potential origin of microbes that might be involved in, or represent a major cause of, PE is the oral cavity, and in particular when there is oral disease (such as periodontitis and gum bleeding) that can allow microbes to enter the bloodstream. If this is a regular occurrence one would predict that PE would be much more prevalent in patients with pre-existing periodontitis [but cf. Ref. (471) for those in pregnancy] than in matched controls; this is indeed the case (Table 4). As with many of the tables herein, the odds ratios are far beyond anything that might weakly be referred to as an “association.”

Table 4.

Periodontal disease (PD) and preeclampsia.

Organisms Comments Reference
Meta-analyses OR of PE increased 3.69-fold if PD before 32 weeks (472)
OR of 3 for the development of PE if ureaplasmas present at first antenatal visit (473)
OR 5.56 for PD preceding PE (474)
OR 2.1 for preceding PE (475)
Extensive overview of role of oral health and periodontal disease in PE (476)
OR 3.71 for PE if history of periodontal treatment (477)
Excellent overview of likely relationship between PD and PE (478)
OR = 8.6 or 2.03 for PE if PD was present vs. controls (479)
Strong association between PD and PE (p < 0.01) (480)
Overview with many references (481)
OR for association between PD and PE = 3.73. No correlation with TNF-α or IL (482)
OR 2.46 PE:controls (483)
Excellent overviews, focusing on means of transport of microbes from mouth to reproductive tissue (484) [see also Bobetsis et al. (485)]
Relationship between C-reactive protein, PE, and severity of PD (486)
Adjusted PE RR 5.8 for Women with periodontal disease and CRP >75th percentile compared to women without periodontal disease (487)
PD prevalence 65.5% and significantly higher (p < 0.0001) in females with hypertension (RR = 1.5) (488)
Meta-analysis (489)
Periodontal bacteria “much more prevalent” in PE than controls, but OR not given (490)
Overview, stressing role of LPS (491)
Overview and meta-analysis of 25 studies (492)
OR 4.79–6.6 for PE is PD (493)
Porphyromonas gingivalis Its LPS inhibits trophoblast invasion (494)
OR = 3 overall (495)
Not stated Significantly higher periodontal probing depth and clinical attachment level scores in the preeclamptic group compared with controls (2.98 vs. 2.11 and 3.33 vs. 2.30, respectively). (496)

Urinary Tract Infections

A particular feature of UTIs is the frequency of reinfection (497504). This is because the organisms can effectively “hide” in bladder epithelial cells as the so-called “quiescent intracellular reservoirs” (212, 501, 503, 505509) of (presumably) dormant cells that can resuscitate. This is why reinfection is often from the same strains that caused the original infection (510514). Other complications can include renal scarring (515). Bacteriuria (often asymptomatic) is a frequent occurrence in pregnancy [e.g., Ref. (373, 375, 473, 516522)], and the frequency of UTI as a source of microbes causing PE is clear from Table 2.

From Blood to and from the Placenta: A Role for Microparticles

We and others have noted the fact that many chronic, inflammatory disease are accompanied by the shedding of various antigens and other factors; typically they pass through the bloodstream as microparticles (126, 140, 523530), sometimes known as endosomes (345, 347, 348, 524, 531) [and see later under microRNAs (miRNAs)]. Similarly, LPS is normally bound to proteins such as the LPS-binding protein and apoE (140). Given their prevalence, their role in simply finding their way from maternal blood to placenta, and the fact that we discussed them extensively in two previous reviews (126, 140), we do not discuss them further here.

Evidence from Antibiotic Therapies

Antibiotic drug prescriptions (532534) may be seen as a proxy for maternal infection, so if dormant (and resuscitating and growing) bacteria are a major part of PE etiology one might imagine an association between antibiotic prescriptions and PE. According to an opposite argument, antibiotics and antibiotic prescriptions given for nominally unrelated infections (UTI, chest, etc., and in particular diseases requiring long-term anti-infective medication that might even last throughout a pregnancy) might have the beneficial side-effect of controlling the proliferation of dormant cells as they seek to resuscitate. There is indeed some good evidence for both of these, implying that it is necessary to look quite closely at the nature, timing, and duration of the infections and of the anti-infective therapy relative to pregnancy. A summary is given in Table 5. A confounding factor can be that some (e.g., the antiretroviral) therapies are themselves quite toxic (535, 536); while the OR for avoiding PE was 15.3 in one study of untreated HIV-infected individuals vs. controls, implying (as is known) a strong involvement of the immune system in PE, the “advantage” virtually disappeared upon triple-antiretroviral therapy (537). Overall, it is hard to draw conclusions from antiretrovirals (538, 539). However, we have included one HIV study in the table. Despite a detailed survey, we found no reliable studies with diseases such as Lyme disease or tuberculosis, where treatment regimens are lengthy, that allowed a fair conclusion as to whether antibiotic treatment was protective against PE. However, we do highlight the absolutely stand-out study of Todros and colleagues (540), who noted that extended spiramycin treatment (of patients with T. gondii) gave a greater than 10-fold protection against PE, when the parasite alone had no effect (541). This makes such an endeavor (assessing the utility of early or pre-emptive antibiotics in PE) potentially highly worthwhile.

Table 5.

Examples of decreased PE following antibiotic therapies given for various reasons.

Target organisms Comments Reference
HIV OR of 0.65 for patients treated with mono- or triple antiretroviral therapy (542)
Toxoplasma gondii Massive (OR = 0.092) protection against PE in patients treated with spiramycin (540)
Various organisms 52% decrease in PE following 10-day antibiotic therapy (330)

Role of LPS in PE

It is exceptionally well known that LPS (sensu lato) is highly inflammagenic, and since one of us recently reviewed that literature in extenso (140) this is not directly rehearsed here. However, since we are arguing that it has a major role in PE naturally or in vivo, we do need to ask whether the literature is consistent with this more focused question. The answer is, of course, a resounding “yes.” Notwithstanding that only primates, and really only humans, are afflicted by “genuine” PE, so the genuine utility of rodent models is questionable (543), even if some can recapitulate elements of the disease (544, 545). Hence, it is somewhat ironic that there are a number of animal models in which LPS (also known as “endotoxin”) is used experimentally to induce a condition resembling PE [e.g., Ref. (546551) and also see Ref. (552)]. We merely argue that it is not a coincidence that exogenous administration of LPS has these effects, because we consider that it is, in fact, normally one of the main mediators of PE. Also note, in the context of gestational diabetes, that serum levels of LPS are raised significantly in both type 1 (466, 553) and type 2 (554556) diabetes.

The standard sequelae of LPS activation, e.g., TLR signaling and cytokine production, also occur in PE (557559), bolstering the argument that this is precisely what is going on. In a similar vein, double-stranded RNA-mediated activation of TLR3 and TLR7/8 can play a key role in the development of PE (560562). What is new here is our recognition that LPS and other inflammagens [e.g., Ref. (563565)] may continue to be produced and shed by dormant and resuscitating bacteria that are generally invisible to classical microbiology.

Effects of LPS and Other Microbial Antigens on Disrupting Trophoblast Invasion and/or Stimulating Parturition

As with other cases of cross-reactivity such as that of various antigens in Proteus spp. that can cause disease in rheumatoid arthritis (458460), the assumption is that various microbial antigens can lead to the production of (auto-)antibodies that attack the host, in the present case of interest by stopping the placentation by trophoblasts. This is commonly referred to as “molecular mimicry” [e.g., Ref. (566569)] and may extend between molecular classes, e.g., peptide/carbohydrate (570, 571). Table 6 shows some molecular examples where this has been demonstrated.

Table 6.

Molecular examples of bacterial antigens that can elicit antibodies that stop successful trophoblast implantation or stimulate parturition.

Organism Antigen and comments Reference
Gram-negatives LPS can stimulate parturition, via corticotropin-releasing hormone (572)
Gram-negatives LPS can stimulate parturition, via MAPKinase (559)
Helicobacter pylori Anti-CagA antibodies cross-react with trophoblasts and could inhibit placentation (344, 358)
Porphyromonas gingivalis LPS inhibits trophoblast invasion (494)
Various Antiphospholipid antibodies (that can be induced by microbes, see above) (573576)

In many cases, the actual (and possibly microbial) antigens are unknown, and clearly the microbial elicitation of antibodies to anything that might contribute to PE points to multiple potential origins. To this end, we note that PE has also been associated with antibodies to angiotensin receptors (577590), to smooth muscle (591, 592) [such blocking may be anti-inflammatory (593595)], to adrenoceptors (596), to the M2 muscarinic receptor (597), and to Th17 (598) [and see Ref. (599)]. It is not unreasonable that epitope scanning of the antibody targets coupled with comparative sequence analysis of potential microbes might light up those responsible. In the case of angiotensin II type 1 receptor antibodies, the epitope is considered (600) to be AFHYESQ, an epitope that also appears on parvovirus B19 capsid proteins; in the event, parvoviruses seem not to be the culprits here (601). However, the role of these antibodies in activating the angiotensin receptor is also considered to underpin the lowering of the renin–angiotensin system that is commonly seen in PE (602605), but which is typically raised during normal pregnancy.

Th-17 is of especial interest here, since these are the helper T (Th)-cell subset that produces IL-17. IL-17 is probably best known for its role in inflammation and autoimmunity (599, 606610). However, it also has an important role in induction of the protective immune response against extracellular bacteria or fungal pathogens at mucosal surfaces (608, 611623). Th17 cells seem to participate in successful pregnancy processes and can be lower in PE (624626), although more studies show them as higher (599, 627635) or unchanged (636, 637). One interpretation, consistent with the present thesis, is that the antimicrobial effects of placental IL-17 relative to Treg cells are compromised during PE (599, 633, 638).

A Note on the Terminology of Sepsis

As one may suppose from the name, sepsis (and the use of words like “antiseptic”) was originally taken to indicate the presence of culturable organisms in (or in a sample taken from) a host, e.g., as in bacteremia. Recognizing that it is the products of bacteria, especially cell wall components, that cause the cytokine storms that eventually lead to death from all kinds of infection (639643), “sepsis” nowadays has more come to indicate the latter, as a stage (in the case of established infection) on a road that leads to septic shock and (eventually) to death [with a shockingly high mortality, and many failures of initially promising treatments, e.g., Ref. (644, 645), and despite the clear utility of iron chelation (84, 125, 646648)]. In most cases, significant numbers of culturable microbes are either unmeasured or absent, and like most authors, we shall use “sepsis” to imply the results of an infection whether the organisms are detected or otherwise. Overall, it is possible to see the stages of PE as a milder form of the sepsis cascade on the left-hand side of Figure 7. Figure 7 compares the classical route of sepsis-induced death with the milder versions that we see in PE; they are at least consistent with the idea that PE is strongly related to the more classical sepsis in degree rather than in kind.

Figure 7.

Figure 7

Preeclampsia bears some similarities to and may be considered as a milder form of, the changes that occur during genuine sepsis leading to a systematic inflammatory response syndrome, septic shock, and multiple organ dysfunction.

Preeclampsia and Neonatal Sepsis

If PE is really based on infectious agents, it is reasonable that one might expect to see a greater incidence of neonatal sepsis (i.e., infection) following PE. While there are clearly other possible explanations (e.g., simply a weakened immune system, sometimes expressed as neutropenia, after PE), there is certainly evidence that this is consistent with this suggestion (649653).

PE Biomarkers and Infection

Because of the lengthy development of PE during pregnancy, there has long been a search for biomarkers (somewhat equivalent to the “risk factors” discussed earlier) that might have predictive power, and some of these, at both metabolome (15, 654661) and proteome (662667) level, are starting to come forward. The typical experimental design is a case–control, in which markers that are raised or lowered significantly relative to the age-matched controls are considered to be candidate markers of PE. However, just as noted with leukocyte markers (81) and polycystic ovary syndrome (PCOS) (668) that does not mean that they might not also be markers for other things too, such as infection (669)!

Thus, one prediction is that if dormant and resuscitating bacteria are responsible for PE then at least some of these biomarkers should also be (known to be) associated with infection. However, one obvious point is that the markers may appear only after infection, and this may itself be after the first-trimester; clearly then these would not then be seen as “first-trimester” biomarkers! There are many well-known inflammatory biomarkers that are part of the innate [and possibly trained (670)] immune response, such as the inflammatory cytokines CRP [cf. Ref. (671, 672)], IL-6 (673), IL-1β (674), TNF-α (675), and macrophage migration inhibitory factor (MIF) (676), which are also all biomarkers of infection (677681). Certainly, the fact that these increase in PE is consistent with a role for an infectious component. However, we shall mainly look at other biomarkers that are known to increase with PE, and see if they are also known to be biomarkers for (or at least changed in the presence of) infection (and see Th17/IL-17 above), and we next examine this. We shall see that pretty well every biomarker that is changed significantly in PE is also known to be changed following infection, a series of findings that we consider adds very strong weight to our arguments.

Proteomic and Similar Biomarkers – Circulating and Placental

What is really needed is a full systems biology strategy [see, e.g., Ref. (99, 682684)] that brings together the actors that interact then parametrizes the nature of these interactions in a suitable encoding [e.g., SBML (685)] that permits their modeling, at least as an ODE model using software such as CellDesigner (686), COPASI (687), or Cytoscape (688). Thus, to take a small example, “agonistic autoantibodies against the angiotensin II type 1 receptor autoantibodies (AT1-AA) are described. They induce NADPH oxidase and the MAPK/ERK pathway leading to NF-κB and tissue factor activation. AT1-AA are detectable in animal models of PE and are responsible for elevation of soluble fms-related tyrosine kinase-1 (sFlt1) and soluble endoglin (sEng), oxidative stress, and endothelin-1, all of which are enhanced in pre-eclamptic women. AT1-AA can be detected in pregnancies with abnormal uterine perfusion” (589). Many such players have been invoked, and we next list some.

Activin A

Activin A is a member of the TGF-β superfamily. Its levels are raised significantly in PE (118, 689691). However, activin A is also well established as a biomarker of infection (692695).

Calretinin

In a proteomic study of preclamptic vs. normal placentas (696), calretinin was one of the most differentially upregulated proteins (p = 1.6 × 10−13 for preterm PE vs. controls, p = 8.9 × 10−7 for term PE vs. controls), and in a manner that correlated with the severity of disease. While calretinin [normally more expressed in neural tissue and mesotheliomas (697)] is not normally seen as a marker of infection, it is, in fact, raised significantly when Chlamydia pneumoniae infects human mesothelial cells (698).

Chemerin

Chemerin is a relatively recently discovered adipokine, whose level can increase dramatically in the first-trimester of preeclamptic pregnancies (699), and beyond (700). Its levels are related to the severity of the PE (701703). Specifically, an ROC curve (704) analysis showed that a serum chemerin level >183.5 ng mL−1 predicted PE with 87.8% sensitivity and 75.7% specificity (AUC, 0.845; 95% CI, 0.811–0.875) (699). Papers showing that chemerin is also increased by infection (hence inflammation) include (705, 706); it even has antibacterial properties (707, 708) and was protective in a skin model of infection (709, 710). In a study of patients with sepsis (711), circulating chemerin was increased 1.69-fold compared with controls (p = 0.012) and was also protective as judged by survival. These seem like particularly potent argument for a role of chemerin as a marker of infection rather than of PE per se, and for the consequent fact that PE follows infection and not vice versa.

Copeptin

Copeptin, a glycosylated polypeptide consisting of the 39 C-terminal amino acids of arginine vasopressin, has been suggested as “a new biomarker that is specific for preeclampsia” (712), and certainly changes during its development (713). However, it turns out that it is also essentially a measure of all kinds of stresses and adverse events (714719), including those caused by infection (720729).

Cystatin C

Not least because kidney function is impaired in PE, low molecular weight (MW) proteins may serve as biomarkers for it. To this end, cystatin C (13 kDa) has been found to be raised significantly in PE (730736); it also contributed to the marker set in the SCOPE study (8, 17). Notably, although it certainly can be raised during infection (737), it seems to be more of a marker of inflammation or kidney function (738, 739).

d-Dimer

d-dimer” is a term used to describe quite varying forms of fibrin degradation products (740). Given that PE is accompanied by coagulopathies, it is probably not surprising that d-dimer levels are raised in PE (741745), although this is true for many conditions (746), and some of the assays would bear improvement (747, 748). Needless to say, however, raised d-dimer levels are also a strong marker for infection (749, 750).

Endocan

Endothelial cell-specific molecule-1 (ESM-1), known as endocan, is a cysteine-rich dermatan sulfate proteoglycan expressed (and sometimes released) by endothelial cells. It has been suggested to be a new biomarker for endothelial dysfunction and PE (751, 752). It would appear, however, to be a rather less specific inflammatory biomarker (753, 754) and is associated with a variety of diseases, including chronic kidney disease (755, 756) and cardiovascular disease (755). Most pertinently from our perspective, it is also raised strongly during sepsis (757760).

Endoglin

Endoglin is the product of a gene implicated (761, 762) in the rare disease Hereditary Hemorrhagic Telangiectasia. The role of endoglin remains somewhat enigmatic (763). However, endoglin levels were 2.5-fold higher in preeclamptic placentas compared to normal pregnancies (15.4 ± 2.6 vs. 5.7 ± 1.0, p < 0.01). After the onset of clinical disease, the mean serum level of sEng in women with preterm PE was 46.4 ng mL−1, as compared with 9.8 ng mL−1 in controls (p < 0.001) (88). Women with a particular endoglin polymorphism (AA) were 2.29 times more likely to develop PE than those with the GG genotype (p = 0.008) (764), and endoglin is seen as a reasonably good marker for PE (88, 691, 765768) [cf. Ref. (769)]. Again, endoglin levels are raised following infection by a variety of organisms (770773), with a particularly clear example that it is a marker of infection coming from the fact that there is raised endoglin only in infected vs. aseptic loosening in joints following arthroplasty (774). In general, it seems likely that these circulating (anti)angiogenic factors are more or less markers of endothelial cell damage, just as we have described for serum ferritin (126).

Ferritin

The natural iron transporter in blood is transferrin [e.g., Ref. (775780)], present at ca. 1–2 g L−1, with ferritin being an intracellular iron storage molecule, so one is led to wonder why there is even any serum ferritin at all (126, 781). The answer is almost certainly that it is a leakage molecule from damaged cells (126), and when in serum it is found to have lost its iron content (782785). Serum ferritin is, as expected, raised during PE (244, 246, 249, 253, 255, 786, 787) and in many other inflammatory diseases (126), including infection [e.g., Ref. (788, 789) and above].

microRNAs

microRNAs are a relatively novel and highly important class of ~22 nt non-coding, regulatory molecules (790793). Some are placenta specific, and those in the circulation [often in endo/exosomes (794796)] can be identified during pregnancy (797800), potentially providing a minimally invasive readout of placental condition (801803). There is aberrant expression of placenta-specific miRNAs in PE including miR-517a/b and miR-517c (804810) and miR-1233 (811). C19MC is one of the largest miRNA gene clusters in humans, maps to chromosome 19q13.41, and spans a ~100 kb long region. C19MC miRNAs are processed from the cluster (812), are primate-specific, conserved in humans, and comprise 46 miRNA genes, including the miR-517 family (813). miR-517 is known to be antiviral (814, 815), while miR-517a overexpression is apoptotic (816) and can inhibit trophoblast invasion (817). Importantly for our argument, miR-517 molecules are overexpressed following infection (818, 819).

Neuropeptide Y

Although, as its name suggests, neuropeptide Y is a neurotransmitter, it is also correlated with stress. Certainly, it is related to noradrenaline (see below) that may itself be responsible for the raised blood pressure (BP) in PE (820). It is also raised in sepsis, where it is considered to counterbalance the vasodilation characteristic of septic shock [e.g., Ref. (821, 822)]. The apparent paradox of a raised BP in PE and a lowered one in septic shock is considered to be related to the very different concentrations of endotoxin involved (Figure 7).

NGAL (Lipocalin 2, Siderocalin)

Neutrophil gelatinase-associated lipocalin (NGAL) is a lipocalin that is capable of binding catecholate-based siderophores (125, 823, 824). As such it is antimicrobial and is also an inflammatory or sepsis biomarker (825, 826). Given our interest in iron, it is not surprising that it is changed during PE. While one study suggested it to be decreased in PE (827), a great many other studies showed it to be increased significantly in PE, and typically in a manner that correlated with PE severity (735, 828837). Pertinently to PE, it is also well established as an early biomarker of acute kidney injury (AKI) (838841). However, it is not a specific biomarker for AKI vs. sepsis (839, 842850) and its origin in sepsis differs (851, 852). Of course, it can be the sepsis that leads to the AKI (853, 854). Fairly obviously, while it does tend to be increased during PE, we again see its direct role as an antimicrobial and marker of sepsis as highly supportive of our present thesis.

Placental Growth Factor

This is a member of the VEGF Family that despite its name has a great many activities (855). It is often considered in parallel with endoglin and sFlt, with a high sFlt:PlGF ratio being considered as especially discriminatory for PE (89, 856869), i.e., a lower PlGF can be diagnostic of PE (769, 870873). PlGF tends to be raised in sepsis unrelated to pregnancy (874, 875), while its lowering in PE may be due to the excess sFLT that decreases it (855, 876, 877). In one study of a patient with CMV infection and PE, it was, in fact, raised (878), while, in the case of IUGR, it was massively lowered (879). PlGF alone is thus probably not a useful general marker for either PE or sepsis if one is trying to disentangle them, although it has clear promise when PE is superimposed on CKD (872, 880).

Procalcitonin

Procalcitonin is the 116 amino acid polypeptide precursor of calcitonin, a calcium regulatory hormone. It is another marker that has been observed to be raised (according to severity) in preeclamptics (742, 881, 882) [but cf. Ref. (713)]. However, it is also a known marker of bacterial infections or sepsis (881, 883891).

Serum Amyloid A

This is an inflammatory biomarker, which was shown to increase fourfold in PE in one study (892), was significantly raised in another (882), but not in a third (893). However, it is a well-established (and potent) biomarker for infection/sepsis [e.g., Ref. (894907)]. Defective amyloid processing may be a hallmark of PE more generally (908), and of course amyloid can be induced by various microbes (317, 319, 909, 910) and their products (257).

Soluble fms-Like Tyrosine Kinase 1

The sFlt receptor is a splice variant of the VEGF receptor (766). It is raised considerably in PE (691, 767, 856, 862, 864, 911914) and may be causal (545, 590, 915918). Needless to say, by now, we can see that it is also a very clear marker of infection (767, 919, 920), whose levels even correlate with the severity of sepsis (921923). Of particular note is the fact that sFLT is actually anti-inflammatory (922).

Thrombomodulin

Soluble thrombomodulin was recognized early as an endothelial damage biomarker and is raised in PE (924934). Interestingly, it has been found to have significant efficacy in the treatment of sepsis [-based disseminated intravascular coagulation (DIC)] (935943).

TLR4

TLR4 upregulation in preeclamptic placentas (944) is entirely consistent with infection and the “danger model” as applied to PE (945). As well as LPS activation [reviewed in Kell and Pretorius (140)], the heat shock protein 60 of Chlamydia also activates TLR4 (138).

Visfatin

Visfatin is another adipokine that is raised in PE, approximately 2-fold in the study of Fasshauer and colleagues (946), and 1.5-fold in that of Adali and colleagues (947). However, it was little different in a third study (948), while in a different study, it was rather lower in PE than in controls (949). This kind of phenomenon rather lights up the need for excellent quality studies, including ELISA reagents, when making assessments of this type.

Fairly obviously, the conclusion that this long list of biomarkers that are raised in PE might be specific “PE” biomarkers is challenged very strongly by the finding that they are, in fact, all known markers of infection, a finding that in our view strongly bolsters the case for an infectious component in PE.

In a similar vein, there are a number of other sepsis markers (where sepsis is varied via, or occurs as, an independent variable) that we would predict are likely to be visible as raised in PE patient. These might include (680, 950) PAI-1, sE-selectin (951), and sVCAM-1 (921). In particular, Presepsin looks like a potentially useful marker for sepsis (888, 889, 952961), but we can find no literature on its use as a PE biomarker, where we predict that it may also be raised.

Metabolomic Biomarkers

For fundamental reasons connected with metabolic control and its formal, mathematical analysis (962966), changes in the metabolome are both expected (967) and found (968971) to be amplified relative to those in the transcriptome and proteome. For similar reasons, and coupled with evolution’s selection for robustness (972978) (i.e., homeostasis) in metabolic networks, we do not normally expect to find single metabolic biomarkers for a complex disease or syndrome. Since our initial metabolomic analyzes (654), the technology has improved considerably (979982), a full human metabolic network reconstruction has been published (978, 983985) in the style of that done for yeast (986), and a number of candidate metabolomics biomarkers for PE have been identified reproducibly on an entirely separate validation set (15, 655).

This latter, LC-MS-based, study (15) found a cohort of 14 metabolites from the first-trimester that when combined gave an OR of 23 as being predictive of third-trimester PE. For convenience, we list them in Table 7. Note that because they were characterized solely via their mass, there are some uncertainties in the exact identification in some cases, and that untargeted metabolomics of this type has a moderately high limit of detection (maybe 10 μM) such that many potentially discriminatory metabolites are below the limit of detection.

Table 7.

Fourteen metabolites contributing to a preeclamptic “signature” (15).

Metabolite Up or down in PE Average mass (Da) Chemspider identifier
5-hydroxytryptophan Down 220.225 141
Monosaccharide Up Unspecified Unspecified
Decanoylcarnitine Up 315.448 8420677
Methylglutaric and/or adipic acid Down 146.141 11549/191
Oleic acid Up 282.461 393217
Docosahexaenoic acid and/or docosatriynoic acid Up 328.488 393183/absent
γ-Butyrolactone and/or oxolan-3-one (dihydrofuran-3-one) Up 86.089 7029/461367
2-Oxovaleric acid and/or oxo-methylbutanoic acid Up 116.115 67142/absent
Acetoacetic acid Up 102.089 94
Hexadecenoyleicosatetraenoyl-sn-glycerol Up n/a Absent
Di-(octadecadienoyl)-sn-glycerol Up 616.954 4942782
Sphingosine-1-phosphate Up 379.472 4446673
Sphinganine 1-phosphate Up 381.488 559277
Vitamin D3 derivatives Up n/a Unspecified

A number of features of interest emerge from this:

  1. All the markers save 5-hydroxytryptophan and adipic/methylglutaric acid that were raised in PE; 5-hydroxytryptophan is a precursor of serotonin [which in some studies (987) has been seen to be mildly elevated in PE].

  2. Markers came from multiple classes of metabolite or areas of metabolism, including amino acids, carbohydrates, carnitines, dicarboxylic acids, fatty acids (especially), (phospho)lipids, and sterols.

  3. γ-Butyrolactone derivatives can act as signaling molecules for a variety of bacteria (988, 989).

  4. In common with many other inflammatory diseases (145), Vitamin D3 levels [usually measured as 25(OH)vitD or calcidiol] are often lower in PE (990994) [cf. Ref. (995997)], consistent with the levels of their derivatives being raised. However, the direction of causality inflammation ←→ vitamin D levels is not yet known (998) [see also Ref. (143, 145, 996)].

  5. None of these metabolites was among four metabolites proposed as first-trimester biomarkers in two other (smaller) studies from different groups (656, 999).

  6. Sphingolipid metabolism can be deranged in PE (1000) [also in Parkinson’s (1001)].

As well as the non-targeted metabolomics noted above, a number of other small molecule biomarkers have been turned up by more conventional measurements.

Noradrenaline (Norepinephrine)

An interesting early study (1002) found that venous plasma noradrenaline was raised by 67% in preeclamptics vs. controls. Similar data were found by others (1003). This is of particular interest in the present context since noradrenaline is well established as highly growth stimulatory to Gram-negative microorganisms [e.g., Ref. (10041008)], in part by acting as a siderophore (10091011). It also raises the levels of neuropeptide Y (820), and as a stress hormone (1012), is of course well known for its role in raising BP, a hallmark of PE.

There is relatively little metabolomics work in sepsis, but in one study, carnitine and sphingolipid metabolism were also modified during sepsis (1013), while in another (1014), a suite of molecules were decreased during acute sepsis. However, the patients involved here were quite close to death, so it is not clear that comparisons between the metabolome in PE and in dying patients are that worthwhile.

We also note a recent and rather interesting suggestion by Eggers (1015) that the maternal release of adrenaline (rather than noradrenaline) may have an important etiological role in PE, although as with the rest of our thesis here it is not there indicated as to what causes the adrenaline to rise (although infection and inflammation can of course do so).

Uric Acid

Hyperuricemia is a moderately common finding in preeclamptic pregnancies and may even be involved in its pathogenesis [see, e.g., Ref. (10161022)]. However, it does not seem to be very specific (10231027) and is seemingly not an early biomarker [and it did not appear in our own study (15)]. Its lack of specificity is illustrated by the fact that there is considerable evidence for the roles of purinergic signaling (1028), and especially the role of uric acid, in Alzheimer’s and Parkinson’s disease (10291031), as well as in a variety of other kinds of inflammatory processes, including pro-inflammatory cytokine production (1032, 1033), the Plasmodium falciparum-induced inflammatory response (1034), the mechanistic basis for the action of alum as an adjuvant (1035), and even peanut allergy (10361038). As is common in case–control studies when just one disease (e.g., PE) is studied, artificially high levels of sensitivity and (especially) specificity may appear when other patients with other diseases are not considered.

Clotting, Coagulopathies, and Fibrinogen in PE

In much of our previous work [e.g., Ref. (126134)], we have noted that each of these chronic, inflammatory diseases is accompanied by changes in fibrin fiber morphologies, coagulopathies, and changes in erythrocytes that are both substantial and characteristic. They can variously be mimicked by adding unliganded iron or LPS. As is well known, LPS itself is a strong inducer of coagulation, whether via tissue factor or otherwise [e.g., Ref. (10391048)], and will bind to fibrin strongly (259, 1049). The morphological methods have not yet, to our knowledge, been performed on blood from preeclamptics, whether as a diagnostic or a prognostic, although we note that clotting factors came top in one GWAS looking for gene–PE associations (117). Fibrinogen itself is a TLR4 ligand (1050), is raised in PE (10511055), and we note the extensive evidence for coagulopathies during pregnancies with PE [e.g., Ref. (64, 128, 527, 741, 10561068)]. In the worst cases, these are the very frightening DIC (1047, 10691073) that can, of course, also emerge as a consequence of sepsis (10741080). Variations in the plasminogen activator inhibitor-1 may contribute to the hypofibrinolysis observed (10811083).

We recently showed that LPS can potently induce amyloid formation in fibrin (258, 259, 1084, 1085). Thus, in addition, we note the increasing recognition that amyloid proteins themselves, that may occur as a result of coagulopathies, are themselves both inflammatory [e.g., Ref. (565, 669, 10861091)] and cytotoxic [e.g., Ref. (257, 10921096)], and that this can of itself contribute strongly to the death of, e.g., trophoblasts.

Related to clotting parameters are three other “old” but easily measured variables that probably reflect inflammation (1097), that have been suggested to differ in PE from normotensives, and may have some predictive power. The first two are the erythrocyte sedimentation rate (ESR) (1098, 1099) and the red cell distribution width (RDW) (1100) [but cf. Ref. (1101)]. Interestingly, the former was the only variable that was predictive of a subsequent stroke following sub-arachnoid hemorrhage (1102). The third relates to the morphology of erythrocytes (that may in part underpin the other two). We and others have shown in a series of studies [e.g., Ref. (134136, 11031106)] that erythrocyte morphology diverges very considerably from that “classical” discoid shape adopted by normal healthy cells, and that this can be a strong indicator of disease (137). In extreme cases [e.g., Ref. (133, 11071112)], including following infection (1113), this results in eryptosis, the suicidal death of erythrocytes. It is of interest that ceramide, a precursor of sphingosine-1-phosphate (S1P) (Table 7), is raised in various diseases such as Parkinson’s and may serve to stimulate eryptosis (1114). Although we know of no direct measurements to date, there is evidence that eryptosis may play a significant role in PE (1115).

Prevention Strategies

Apart from low-dose aspirin [that may have little effect (11161119) unless initiated relatively early in pregnancy (11201124)], and low-dose calcium (1125), there are relatively few treatment options in present use (11261129). [Magnesium sulfate (11301132) has been used as a treatment for eclampsia and, interestingly, prevents LPS-induced cell death in an in vitro model of the human placenta (1133).]

In the history of science or medicine, some treatments are empirical, while others are considered to have a mechanistic basis. The general assumption is that the more we know about the originating etiology of a disease or syndrome the more likely we are to be able to treat its causes effectively, and not just its symptoms. Clearly, also, clinicians are rightly loth to give complex and potentially teratogenic treatments to pregnant women when this can be avoided (11341137). However, the surprising lack of systematic data with antibiotics (1138), modulo one particularly spectacular success (540), suggests that we ought to be performing trials with safe antibiotics on women at special risk (1139). These must take care to avoid any Jarisch–Herxheimer reaction (11401143) due to the release from microbes induced by antibiotics of inflammagens such as LPS (11441147). A related strategy recognizes that some FDA-approved drugs can actually exert powerful antibiotic effects in vivo (but not on petri plates) by modifying the host (1148).

Because of the known oxidative stress accompanying PE, it had been assumed that antioxidants such as vitamin C (ascorbate) might be preventive; however, this turned out not to be the case (even the opposite) for ascorbate (1118, 1149). Probably, this is because in the presence of unliganded iron, ascorbate is, in fact, pro-oxidant (125). However, polyphenolic antioxidants that actually act by chelating iron (84, 125) seem to be more effective (1150).

Another area that we and others have previously highlighted recognizes the ability of non-siderophoric iron chelators to act as iron-withholding agents and thereby limit the growth of bacteria. Again, a prediction is that women with iron overload diseases should be more susceptible to PE, a prediction that is borne out for α-thalassemia (1151, 1152) though not apparently for hereditary hemochromatosis (1153). However, the extent of use of chelators and degree of control of free iron, thereby obtained is rarely recorded in any detail, so in truth it is difficult to draw conclusions.

The general benefits of nutritional iron chelators such as blueberries and other fruits and vegetables containing anthocyanins have been discussed elsewhere [e.g., Ref. (84, 125, 1154)].

How significant coagulopathies are to the etiology of PE development (as opposed to providing merely an accompaniment) is not entirely clear, but on the basis that they are then anticoagulants would potentially assist, just as thrombomodulin does in DIC accompanying sepsis (942, 943, 1079, 1080). Of course, one of many effects of low-dose aspirin is to act as an anticoagulant. There is also evidence for the efficacy of heparin (6, 1127, 11551160), which is especially interesting given our highlighting of the role of coagulopathies in PE. These anticoagulants that avoid bleeding (1161) are obviously of particular interest, while anything stopping the fibrin forming β-amyloid (258, 259) should serve as an especially useful anti-inflammatory anticoagulant.

With a change in focus from function-first to target-first-based drug discovery (976), there has been an assumption that because a drug is (i) found to bind potently to a molecular target and (ii) has efficacy at a physiological level in vivo, the first process is thus responsible for the second. This has precisely no basis in logic [it is a logical fault known variously as “affirming the consequent” or “post hoc ergo propter hoc” (1162)]. This is because the drug might be acting physiologically by any other means, since drug binding to proteins is typically quite promiscuous [e.g., Ref. (11631167)]. Indeed, the average known number of binding sites for marketed drugs is six (208, 1168). In particular, it is likely, from a network or systems pharmacology perspective [e.g., Ref. (978, 11691172)], that successful drugs (like aspirin) are successful precisely because they hit multiple targets. The so-called “statins” provide a particularly good case in point (125).

It had long been known that the enzyme HMGCoA reductase exerted strong control on the biosynthetic flux to cholesterol, and that inhibiting it might lower the flux and steady-state cholesterol levels (as indeed it does). Notwithstanding that cholesterol alone is a poor predictor of cardiovascular disease (11731175), especially in the normal range, HMGCoA reductase inhibitors have benefits in terms of decreasing the adverse events of various types of cardiovascular disease (1176). Following an original discovery of natural products such as compactin (mevastatin) and lovastatin containing a group related to hydroxymethylglutaric acid (rather than a CoA version) that inhibited the enzyme (1177), many variants with this (hydroxyl)methylglutaric substructure came to be produced, with the much larger “rest” of the molecule being considerably divergent [see Figure 8, where the MW values vary from 390.5 (mevastatin) to 558.6 (atorvastatin)]. Despite this wide structural diversity (Figure 8), they are still collectively known as “statins,” and despite the wildly illogical assumption that they might all work in the same way(s). The fact that different statins can cause a variety of distinct expression profiles (1178) is anyway utterly inconsistent with a unitary mode of action. In particular, in this latter study, statins clustered into whether they were (fluvastatin, lovastatin, and simvastatin) or were not (atorvastatin, pravastatin, and rosuvastatin) likely to induce the side-effect of rhabdomyolysis or any other myopathy. Clearly, any choice of “statin” should come from the latter group, with pravastatin and rosuvastatin being comparatively hydrophilic.

Figure 8.

Figure 8

Some structures of various statins.

The epidemiological fact of improved survival despite the comparative irrelevance of cholesterol levels to atherosclerotic plaque formation and heart disease in the normal range provides an apparent paradox (1179). This is easily solved by the recognition [e.g., Ref. (11801193), and many other references and reviews] that “statins” are, in fact, anti-inflammatory. They may also be antimicrobial/antiseptic, whether directly or otherwise (11941198), and we also note the role of cholesterol in mopping up endotoxin (1199). Finally, here, it needs to be recognized that statins do themselves serve to lower iron levels (12001202), and (while oddly this seems not to have been tested directly) simple inspection of their structures (Figure 8) implies that the better ones (with their multiple OH groups) might, in fact, chelate iron directly.

In consequence, a number of authors have indicated the potential utility of statins in treating PE (113, 545, 12031213), and pravastatin has been the subject of a number of favorable studies (545, 1204, 1206, 1209, 1211, 1214, 1215), including in humans (1204, 12161218). Pravastatin seems more than ripe for a proper, randomized clinical trial (1203).

Another “vascular” class of drugs that has been proposed for treating PE is represented by those of the family of vasodilatory phosphodiesterase 5 inhibitors such as sildenafil (Viagra) and vardenafil (Levitra), as it is reasonable that they might improve endothelial function, especially if started early in pregnancy (1219). Thus, vardefanil restores endothelial function by increasing PlGF (1220), and sildenafil has shown promise in a number of animal studies (12211226) and in human tissues (1227, 1228), with a clinical trial ongoing (1229). In particular (1226), it was able to normalize the metabolomics changes observed in a mouse model (the COMT−/− model) of PE.

Anti-hypertensive therapy for PE has been reviewed by Abalos and colleagues (1230) and Magee and colleagues (114). Anti-hypertensives did halve the incidence of hypertension but had no effect on PE. Methyldopa is one of the most commonly used anti-hypertensives in pregnancy, but it may also stimulate eryptosis (1231); alternative drugs were considered to be better (1230) for hypertension. Nifedipine (1232) and labetalol (1233) are considered a reasonable choice. There was also a slight reduction in the overall risk of developing proteinuria/PE when beta blockers and calcium channel blockers considered together (but not alone) were compared with methyldopa (1230). In mice, olmesartan (together with captopril) proved usefully anti-hypertensive (1234); this is of interest because olmesartan is also an agonist of the vitamin D receptor (1235). However, it was not mentioned in either Ref. (1230) or Ref. (114).

Lipopolysaccharide itself has long been recognized as a target of inflammatory diseases. Unfortunately, despite initially promising trials of an anti-LPS antibody known as centoxin (1236), it was eventually withdrawn, apparently because of a combination of ineffectiveness (1237, 1238) and toxicity (1239, 1240). LPS is rather hydrophobic, and thus it is hard to make even monoclonal antibodies very selective for such targets, such that the toxicity was probably because of its lack of specificity between lipid A and other hydrophobic ligands (1241). Other possible treatments based on LPS, such as “sushi peptides” (12421249) [or variants (1250, 1251)], and LPS-binding protein were covered elsewhere (140).

If an aberrant or dysbiotic gut microbiome is the source of the microbes that underpin PE, it is at least plausible that the gut microbiome should be predictive of PE (378), but we know of no suitably powered study that has been done to assess this, and this would clearly be worthwhile. However, in a study of primiparous women, the OR for getting severe PE was only 0.6 if probiotic milk drinks containing lactobacilli were consumed daily (1252). This is a very significant effects, such that this too seems an area well worth following up.

From a metabolomics point of view, the molecules seen to be raised in PE may either be biomarkers of the disease etiology or of the body’s attempts to respond to the disease [and this is true generally (1253)]. Thus, it is of great interest that S1P was raised in PE [see Kenny (15) and Table 7]. S1P is mainly vasoconstrictive (1254, 1255), but agonists of the S1P-1 receptor (that is involved in endothelial cell function) seemed to have considerable value in combatting the cytokine storm that followed infection-driven sepsis (12561261). The detailed mechanism seems not to be known, but in the context of infection, a need for S1P and other sphingolipids for successful pregnancies (1262, 1263) [see also Parkinson’s (1001)], and the induction of PE by its disruption (1000, 12641268), some serious investigation of the potential protective effects of S1PR1 agonists seems highly warranted.

Among other small molecules, melatonin has shown some promise in the treatment of septic shock, by lowering inflammatory cytokine production (1269) [and see Gitto et al. (1270) for neonatal oxidative stress], and a trial is in prospect for PE (1271).

Lipoxin A4 (LXA4) is considered to be an endogenous stop signal in inflammation. While recognizing the difficulties with rodent PE models (above), we note that in one study, the effect of BML-111 (a synthetic analog of LXA4) was tested on experimental PE induced in rats by low-dose endotoxin (LPS), and showed highly beneficial effects (549).

Coda – A Return to the Bradford Hill Criteria

Returning to the Bradford Hill criteria for ascribing causation of a disease to an environmental factor (97), we can now ask whether a detectable (if largely dormant) microbiome X, that is more likely to replicate with free iron, and that can anyway secrete or shed a variety of inflammatory components such as LPS, represents a plausible and major etiological factor for PE (Y):

  • (1)

    what is the strength of association between X and Y? We found an overwhelming co-occurrence of microbes or their products and PE.

  • (2)

    what is the consistency of association between X and Y? Almost wherever we looked, whether via periodontal disease (PD), UTI, or other means of ingress, we could find a microbial component in PE.

  • (3)

    what is the specificity of association between X and Y? Insufficient data are available to ascribe PE solely to one type of organism; however, these data clearly indicate that a variety of microbes, each capable of shedding inflammatory molecules such as LPS, can serve to stimulate or exacerbate PE.

  • (4)

    experiments verify the relationship between X and Y. It is unethical to do these in humans in terms of purposely infecting pregnant women, but data from antibiotics show the expected improvements.

  • (5)

    modification of X alters the occurrence of Y; this is really as (4).

  • (6)

    biologically plausible cause and effect relationship. Yes, this is where we think the ideas set down here are entirely consistent with current thinking on the main causes of PE. What we add in particular is the recognition that bacteria (and other microbes) that may be invisible to culture are both present and responsible, by established means, for the inflammation and other sequelae (and especially the coagulopathies) seen as causative accompaniments to PE.

Other Predictions

Classical clinical microbiology, involving mainly replication-based methods, is evolving rapidly to assess the microbial content of samples on the basis of DNA sequences (296, 1272), including 16S rDNA (287, 288, 290, 292, 293, 295, 297, 1273), suitable protein-encoding housekeeping genes [e.g., Ref. (12741279)], and, increasingly, full genome sequences (1280). In the future, we can thus expect a considerable increase in molecular assessments of the microbiological content of blood, urine, and tissues, and this will obviously be a vital part of the experimental assessment and development of the ideas presented here. Molecular methods will also be used to assess maternal circulating DNA (12811283) and RNA (1284) in terms of both its presence and sequencing, as well as the use of digital PCR (1285).

Since PE has such a strong vascular component, we also predict that measurements designed to detect coagulopathies will increase in importance, for both diagnosis and prognosis, and for assessing treatments.

New drugs designed to kill non-growing bacteria (12861290) or to overcome amyloid coagulopathies (12911295) will be needed and will come to the fore.

Finally, we consider that real progress in understanding PE from a systems biology perspective means that it must be modeled accordingly, and this must be a major goal.

Concluding Remarks

We have brought together a large and widely dispersed literature to make the case that an important etiological role in PE is played by dormant microbes, or at least ones that are somewhat refractory to culture, and that these can awaken, shed inflammagens such as LPS, and thereby initiate inflammatory cascades. (The sequelae of these, involving cytokines, coagulopathies, and so on, are well enough accepted.) The case is founded on a large substructure of interlocking evidence, but readers might find the following elements as discussed above, especially persuasive and/or worthy of follow-up:

  • the regular presence of detectable microbes in preeclamptic placentas [e.g., Ref. (400, 426, 427)].

  • the fact that endotoxin (LPS) can act as such a mimic for invoking PE in experimental models.

  • the fact that every known proteomic biomarker suggested for PE has also been shown to increase during infection.

  • the significant number of papers reviewing a link between infection and PE [e.g., Ref. (53, 54, 138, 371)].

  • the almost complete absence (one case) of PE in patients treated with spiramycin (540).

Any and all of these provide powerful strategies for testing whether PE is, in fact, such as gastric ulcers (166, 168, 169, 1296), essentially initiated as an infectious disease.

Author Notes

This is the paper number 8 of the series “The dormant blood microbiome in chronic, inflammatory diseases”. The other articles of the series can be found in the reference list with the numbers 141; 139; 140; 257; 147; 143; 1084; 258; 142.

Author Contributions

Both authors made substantial, direct, and intellectual contributions to the work and approved it for publication.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Funding

DK thanks the Biotechnology and Biological Sciences Research Council (grant BB/L025752/1) for financial support. LK is a Science Foundation Ireland Principal Investigator (grant number 08/IN.1/B2083). LK is also The Director of the Science Foundation Ireland-funded INFANT Research Centre (grant no. 12/RC/2272).

References

  • 1.Grill S, Rusterholz C, Zanetti-Dällenbach R, Tercanli S, Holzgreve W, Hahn S, et al. Potential markers of preeclampsia – a review. Reprod Biol Endocrinol (2009) 7:70. 10.1186/1477-7827-7-70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Steegers EAP, Von Dadelszen P, Duvekot JJ, Pijnenborg R. Pre-eclampsia. Lancet (2010) 376:631–44. 10.1016/S0140-6736(10)60279-6 [DOI] [PubMed] [Google Scholar]
  • 3.North RA, Mccowan LM, Dekker GA, Poston L, Chan EH, Stewart AW, et al. Clinical risk prediction for pre-eclampsia in nulliparous women: development of model in international prospective cohort. BMJ (2011) 342:d1875. 10.1136/bmj.d1875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Uzan J, Carbonnel M, Piconne O, Asmar R, Ayoubi JM. Pre-eclampsia: pathophysiology, diagnosis, and management. Vasc Health Risk Manag (2011) 7:467–74. 10.2147/VHRM.S20181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kenny L. Improving diagnosis and clinical management of pre-eclampsia. MLO Med Lab Obs (2012) 44(12):14. [PubMed] [Google Scholar]
  • 6.Desai P. Obstetric Vasculopathies. New Delhi: Jaypee; (2013). [Google Scholar]
  • 7.Chaiworapongsa T, Chaemsaithong P, Yeo L, Romero R. Pre-eclampsia part 1: current understanding of its pathophysiology. Nat Rev Nephrol (2014) 10:466–80. 10.1038/nrneph.2014.102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kenny LC, Black MA, Poston L, Taylor R, Myers JE, Baker PN, et al. Early pregnancy prediction of preeclampsia in nulliparous women, combining clinical risk and biomarkers: the screening for pregnancy endpoints (SCOPE) international cohort study. Hypertension (2014) 64:644–52. 10.1161/HYPERTENSIONAHA.114.03578 [DOI] [PubMed] [Google Scholar]
  • 9.Sircar M, Thadhani R, Karumanchi SA. Pathogenesis of preeclampsia. Curr Opin Nephrol Hypertens (2015) 24:131–8. 10.1097/MNH.0000000000000105 [DOI] [PubMed] [Google Scholar]
  • 10.Abalos E, Cuesta C, Grosso AL, Chou D, Say L. Global and regional estimates of preeclampsia and eclampsia: a systematic review. Eur J Obstet Gynecol Reprod Biol (2013) 170:1–7. 10.1016/j.ejogrb.2013.05.005 [DOI] [PubMed] [Google Scholar]
  • 11.Khan KS, Wojdyla D, Say L, Gulmezoglu AM, Van Look PF. WHO analysis of causes of maternal death: a systematic review. Lancet (2006) 367:1066–74. 10.1016/S0140-6736(06)68397-9 [DOI] [PubMed] [Google Scholar]
  • 12.Duley L. The global impact of pre-eclampsia and eclampsia. Semin Perinatol (2009) 33:130–7. 10.1053/j.semperi.2009.02.010 [DOI] [PubMed] [Google Scholar]
  • 13.Ghulmiyyah L, Sibai B. Maternal mortality from preeclampsia/eclampsia. Semin Perinatol (2012) 36:56–9. 10.1053/j.semperi.2011.09.011 [DOI] [PubMed] [Google Scholar]
  • 14.Abalos E, Cuesta C, Carroli G, Qureshi Z, Widmer M, Vogel JP, et al. Pre-eclampsia, eclampsia and adverse maternal and perinatal outcomes: a secondary analysis of the World Health Organization Multicountry Survey on Maternal and Newborn Health. BJOG (2014) 121(Suppl 1):14–24. 10.1111/1471-0528.12629 [DOI] [PubMed] [Google Scholar]
  • 15.Kenny LC, Broadhurst DI, Dunn W, Brown M, Francis-Mcintyre S, North RA, et al. Robust early pregnancy prediction of later preeclampsia using metabolomic biomarkers. Hypertension (2010) 56:741–9. 10.1161/HYPERTENSIONAHA.110.157297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bartsch E, Medcalf KE, Park AL, Ray JG, High Risk of Pre-Eclampsia Identification Group . Clinical risk factors for pre-eclampsia determined in early pregnancy: systematic review and meta-analysis of large cohort studies. BMJ (2016) 353:i1753. 10.1136/bmj.i1753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Karumanchi SA, Granger JP. Preeclampsia and pregnancy-related hypertensive disorders. Hypertension (2016) 67:238–42. 10.1161/HYPERTENSIONAHA.115.05024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Duckitt K, Harrington D. Risk factors for pre-eclampsia at antenatal booking: systematic review of controlled studies. Br Med J (2005) 330:565–7. 10.1136/bmj.38380.674340.E0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Baker PN, Kenny LC. Obstetrics by Ten Teachers. Boca Raton, FL: CRC Press; (2011). [Google Scholar]
  • 20.Ananth CV, Keyes KM, Wapner RJ. Pre-eclampsia rates in the United States, 1980–2010: age-period-cohort analysis. BMJ (2013) 347:f6564. 10.1136/bmj.f6564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bdolah Y, Elchalal U, Natanson-Yaron S, Yechiam H, Bdolah-Abram T, Greenfield C, et al. Relationship between nulliparity and preeclampsia may be explained by altered circulating soluble fms-like tyrosine kinase 1. Hypertens Pregnancy (2014) 33:250–9. 10.3109/10641955.2013.858745 [DOI] [PubMed] [Google Scholar]
  • 22.Saftlas AF, Olson DR, Franks AL, Atrash HK, Pokras R. Epidemiology of preeclampsia and eclampsia in the United-States, 1979-1986. Am J Obstet Gynecol (1990) 163:460–5. 10.1016/0002-9378(90)91176-D [DOI] [PubMed] [Google Scholar]
  • 23.Zhang J, Zeisler J, Hatch MC, Berkowitz G. Epidemiology of pregnancy-induced hypertension. Epidemiol Rev (1997) 19:218–32. 10.1093/oxfordjournals.epirev.a017954 [DOI] [PubMed] [Google Scholar]
  • 24.Lamminpää R, Vehvilainen-Julkunen K, Gissler M, Heinonen S. Preeclampsia complicated by advanced maternal age: a registry-based study on primiparous women in Finland 1997-2008. BMC Pregnancy Childbirth (2012) 12:47. 10.1186/1471-2393-12-47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Carolan M. Maternal age >= 45 years and maternal and perinatal outcomes: a review of the evidence. Midwifery (2013) 29:479–89. 10.1016/j.midw.2012.04.001 [DOI] [PubMed] [Google Scholar]
  • 26.Trogstad L, Magnus P, Stoltenberg C. Pre-eclampsia: risk factors and causal models. Best Pract Res Clin Obstet Gynaecol (2011) 25:329–42. 10.1016/j.bpobgyn.2011.01.007 [DOI] [PubMed] [Google Scholar]
  • 27.Coonrod DV, Hickok DE, Zhu KM, Easterling TR, Daling JR. Risk-factors for preeclampsia in twin pregnancies – a population-based cohort study. Obstet Gynecol (1995) 85:645–50. 10.1016/0029-7844(95)00049-W [DOI] [PubMed] [Google Scholar]
  • 28.Campbell DM, MacGillivray I. Preeclampsia in twin pregnancies: incidence and outcome. Hypertens Pregnancy (1999) 18:197–207. 10.3109/10641959909016193 [DOI] [PubMed] [Google Scholar]
  • 29.Bdolah Y, Lam C, Rajakumar A, Shivalingappa V, Mutter W, Sachs BP, et al. Twin pregnancy and the risk of preeclampsia: bigger placenta or relative ischemia? Am J Obstet Gynecol (2008) 198:428.e1–6. 10.1016/j.ajog.2007.10.783 [DOI] [PubMed] [Google Scholar]
  • 30.Bodnar LM, Ness RB, Harger GF, Roberts JM. Inflammation and triglycerides partially mediate the effect of prepregnancy body mass index on the risk of preeclampsia. Am J Epidemiol (2005) 162:1198–206. 10.1093/aje/kwi334 [DOI] [PubMed] [Google Scholar]
  • 31.Roberts JM, Bodnar LM, Patrick TE, Powers RW. The role of obesity in preeclampsia. Pregnancy Hypertens (2011) 1:6–16. 10.1016/j.preghy.2010.10.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jeyabalan A. Epidemiology of preeclampsia: impact of obesity. Nutr Rev (2013) 71:S18–25. 10.1111/nure.12055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sugerman HJ. Effect of obesity on incidence of preeclampsia. Am J Obstet Gynecol (2014) 210:375–375. 10.1016/j.ajog.2013.10.014 [DOI] [PubMed] [Google Scholar]
  • 34.Thornton JG, MacDonald AM. Twin mothers, pregnancy hypertension and pre-eclampsia. Brit J Obstet Gynaecol (1999):570–5. 10.1111/j.1471-0528.1999.tb08326.x [DOI] [PubMed] [Google Scholar]
  • 35.Ros HS, Lichtenstein P, Lipworth L, Cnattingius S. Genetic effects on the liability of developing pre-eclampsia and gestational hypertension. Am J Med Genet (2000) 91:256–60. [DOI] [PubMed] [Google Scholar]
  • 36.Williams PJ, Broughton Pipkin F. The genetics of pre-eclampsia and other hypertensive disorders of pregnancy. Best Pract Res Clin Obstet Gynaecol (2011) 25:405–17. 10.1016/j.bpobgyn.2011.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Valenzuela FJ, Perez-Sepulveda A, Torres MJ, Correa P, Repetto GM, Illanes SE. Pathogenesis of preeclampsia: the genetic component. J Pregnancy (2012) 2012:632732. 10.1155/2012/632732 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Boyd HA, Tahir H, Wohlfahrt J, Melbye M. Associations of personal and family preeclampsia history with the risk of early-, intermediate- and late-onset preeclampsia. Am J Epidemiol (2013) 178:1611–9. 10.1093/aje/kwt189 [DOI] [PubMed] [Google Scholar]
  • 39.Roten LT, Thomsen LCV, Gundersen AS, Fenstad MH, Odland ML, Strand KM, et al. The Norwegian preeclampsia family cohort study: a new resource for investigating genetic aspects and heritability of preeclampsia and related phenotypes. BMC Pregnancy Childbirth (2015) 15:319. 10.1186/s12884-015-0754-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Asherson RA, Shoenfeld Y. The role of infection in the pathogenesis of catastrophic antiphospholipid syndrome – molecular mimicry? J Rheumatol (2000) 27:12–4. [PubMed] [Google Scholar]
  • 41.Asherson RA, Cervera R. Antiphospholipid antibodies and infections. Ann Rheum Dis (2003) 62:388–93. 10.1136/ard.62.5.388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Blank M, Asherson RA, Cervera R, Shoenfeld Y. Antiphospholipid syndrome infectious origin. J Clin Immunol (2004) 24:12–23. 10.1023/B:JOCI.0000018058.28764.ce [DOI] [PubMed] [Google Scholar]
  • 43.Harel M, Aron-Maor A, Sherer Y, Blank M, Shoenfeld Y. The infectious etiology of the antiphospholipid syndrome: links between infection and autoimmunity. Immunobiology (2005) 210:743–7. 10.1016/j.imbio.2005.10.004 [DOI] [PubMed] [Google Scholar]
  • 44.Shoenfeld Y, Blank M, Cervera R, Font J, Raschi E, Meroni PL. Infectious origin of the antiphospholipid syndrome. Ann Rheum Dis (2006) 65:2–6. 10.1136/ard.2005.045443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Clark EA, Silver RM, Branch DW. Do antiphospholipid antibodies cause preeclampsia and HELLP syndrome? Curr Rheumatol Rep (2007) 9:219–25. 10.1007/s11926-007-0035-9 [DOI] [PubMed] [Google Scholar]
  • 46.Espinosa G, Cervera R, Asherson RA. Catastrophic antiphospholipid syndrome and sepsis. A common link? J Rheumatol (2007) 34:923–6. [PubMed] [Google Scholar]
  • 47.Sène D, Piette JC, Cacoub P. Antiphospholipid antibodies, antiphospholipid syndrome and infections. Autoimmun Rev (2008) 7:272–7. 10.1016/j.autrev.2007.10.001 [DOI] [PubMed] [Google Scholar]
  • 48.Zinger H, Sherer Y, Goddard G, Berkun Y, Barzilai O, Agmon-Levin N, et al. Common infectious agents prevalence in antiphospholipid syndrome. Lupus (2009) 18:1149–53. 10.1177/0961203309345738 [DOI] [PubMed] [Google Scholar]
  • 49.Kutteh WH. Antiphospholipid antibody syndrome and reproduction. Curr Opin Obstet Gynecol (2014) 26:260–5. 10.1097/Gco.0000000000000086 [DOI] [PubMed] [Google Scholar]
  • 50.Kutteh WH, Hinote CD. Antiphospholipid antibody syndrome. Obstet Gyn Clin N Am (2014) 41:113–32. 10.1016/j.ogc.2013.10.004 [DOI] [PubMed] [Google Scholar]
  • 51.O’Gorman N, Wright D, Syngelaki A, Akolekar R, Wright A, Poon LC, et al. Competing risks model in screening for preeclampsia by maternal factors and biomarkers at 11-13 weeks gestation. Am J Obstet Gynecol (2016) 214:103.e1–12. 10.1016/j.ajog.2015.08.034 [DOI] [PubMed] [Google Scholar]
  • 52.Schieve LA, Handler A, Hershow R, Davis F. Urinary-tract infection during pregnancy – its association with maternal morbidity and perinatal outcome. Am J Public Health (1994) 84:405–10. 10.2105/Ajph.84.3.405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Conde-Agudelo A, Villar J, Lindheimer M. Maternal infection and risk of preeclampsia: systematic review and metaanalysis. Am J Obstet Gynecol (2008) 198:7–22. 10.1016/j.ajog.2007.07.040 [DOI] [PubMed] [Google Scholar]
  • 54.Rustveld LO, Kelsey SF, Sharma R. Association between maternal infections and preeclampsia: a systematic review of epidemiologic studies. Matern Child Health J (2008) 12:223–42. 10.1007/s10995-007-0224-1 [DOI] [PubMed] [Google Scholar]
  • 55.Irgens HU, Reisaeter L, Irgens LM, Lie RT. Long term mortality of mothers and fathers after pre-eclampsia: population based cohort study. BMJ (2001) 323:1213–7. 10.1136/bmj.323.7323.1213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Bellamy L, Casas JP, Hingorani AD, Williams DJ. Pre-eclampsia and risk of cardiovascular disease and cancer in later life: systematic review and meta-analysis. BMJ (2007) 335:974. 10.1136/bmj.39335.385301.BE [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Craici IM, Wagner SJ, Hayman SR, Garovic VD. Pre-eclamptic pregnancies: an opportunity to identify women at risk for future cardiovascular disease. Womens Health (Lond Engl) (2008) 4:133–5. 10.2217/17455057.4.2.133 [DOI] [PubMed] [Google Scholar]
  • 58.Romundstad PR, Magnussen EB, Smith GD, Vatten LJ. Hypertension in pregnancy and later cardiovascular risk: common antecedents? Circulation (2010) 122:579–84. 10.1161/CIRCULATIONAHA.110.943407 [DOI] [PubMed] [Google Scholar]
  • 59.Powe CE, Levine RJ, Karumanchi SA. Preeclampsia, a disease of the maternal endothelium: the role of antiangiogenic factors and implications for later cardiovascular disease. Circulation (2011) 123:2856–69. 10.1161/CIRCULATIONAHA.109.853127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Skjaerven R, Wilcox AJ, Klungsøyr K, Irgens LM, Vikse BE, Vatten LJ, et al. Cardiovascular mortality after pre-eclampsia in one child mothers: prospective, population based cohort study. BMJ (2012) 345:e7677. 10.1136/bmj.e7677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ananth CV, Lawrence Cleary K. Pre-eclampsia and cardiovascular disease: more questions than answers? BJOG (2013) 120:920–3. 10.1111/1471-0528.12215 [DOI] [PubMed] [Google Scholar]
  • 62.Brennan LJ, Morton JS, Davidge ST. Vascular dysfunction in preeclampsia. Microcirculation (2014) 21:4–14. 10.1111/micc.12079 [DOI] [PubMed] [Google Scholar]
  • 63.Chen CW, Jaffe IZ, Karumanchi SA. Pre-eclampsia and cardiovascular disease. Cardiovasc Res (2014) 101:579–86. 10.1093/cvr/cvu018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tannetta DS, Hunt K, Jones CI, Davidson N, Coxon CH, Ferguson D, et al. Syncytiotrophoblast extracellular vesicles from pre-eclampsia placentas differentially affect platelet function. PLoS One (2015) 10:e0142538. 10.1371/journal.pone.0142538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mol BWJ, Roberts CT, Thangaratinam S, Magee LA, De Groot CJM, Hofmeyr GJ. Pre-eclampsia. Lancet (2016) 387:999–1011. 10.1016/S0140-6736(15)00070-7 [DOI] [PubMed] [Google Scholar]
  • 66.Bateman BT, Schumacher HC, Bushnell CD, Pile-Spellman J, Simpson LL, Sacco RL, et al. Intracerebral hemorrhage in pregnancy: frequency, risk factors, and outcome. Neurology (2006) 67:424–9. 10.1212/01.wnl.0000228277.84760.a2 [DOI] [PubMed] [Google Scholar]
  • 67.Kajantie E, Eriksson JG, Osmond C, Thornburg K, Barker DJP. Pre-eclampsia is associated with increased risk of stroke in the adult offspring: the Helsinki birth cohort study. Stroke (2009) 40:1176–80. 10.1161/STROKEAHA.108.538025 [DOI] [PubMed] [Google Scholar]
  • 68.Tang CH, Wu CS, Lee TH, Hung ST, Yang CY, Lee CH, et al. Preeclampsia-eclampsia and the risk of stroke among peripartum in Taiwan. Stroke (2009) 40:1162–8. 10.1161/STROKEAHA.108.540880 [DOI] [PubMed] [Google Scholar]
  • 69.Sherer Y, Blank M, Shoenfeld Y. Antiphospholipid syndrome (APS): where does it come from? Best Pract Res Clin Rheumatol (2007) 21:1071–8. 10.1016/j.berh.2007.09.005 [DOI] [PubMed] [Google Scholar]
  • 70.Mignini LE, Villar J, Khan KS. Mapping the theories of preeclampsia: the need for systematic reviews of mechanisms of the disease. Am J Obstet Gynecol (2006) 194:317–21. 10.1016/j.ajog.2005.08.065 [DOI] [PubMed] [Google Scholar]
  • 71.Kell DB, Kenny LC. A dormant microbial component in the development of pre-eclampsia. BioRxiv preprint. bioRxiv (2016) 057356. 10.1101/057356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Redman CWG. Current topic: pre-eclampsia and the placenta. Placenta (1991) 12:301–8. 10.1016/0143-4004(91)90339-H [DOI] [PubMed] [Google Scholar]
  • 73.Genc H, Uzun H, Benian A, Simsek G, Gelisgen R, Madazli R, et al. Evaluation of oxidative stress markers in first trimester for assessment of preeclampsia risk. Arch Gynecol Obstet (2011) 284:1367–73. 10.1007/s00404-011-1865-2 [DOI] [PubMed] [Google Scholar]
  • 74.Burton GJ, Jauniaux E. Placental oxidative stress: from miscarriage to preeclampsia. J Soc Gynecol Investig (2004) 11:342–52. 10.1016/j.jsgi.2004.03.003 [DOI] [PubMed] [Google Scholar]
  • 75.Redman CWG, Sargent IL. Latest advances in understanding preeclampsia. Science (2005) 308:1592–4. 10.1126/science.1111726 [DOI] [PubMed] [Google Scholar]
  • 76.Roberts JM, Bell MJ. If we know so much about preeclampsia, why haven’t we cured the disease? J Reprod Immunol (2013) 99:1–9. 10.1016/j.jri.2013.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Redman CW, Sargent IL, Staff AC. IFPA Senior Award Lecture: making sense of pre-eclampsia – two placental causes of preeclampsia? Placenta (2014) 35(Suppl):S20–5. 10.1016/j.placenta.2013.12.008 [DOI] [PubMed] [Google Scholar]
  • 78.Xiong X, Demianczuk NN, Saunders LD, Wang FL, Fraser WD. Impact of preeclampsia and gestational hypertension on birth weight by gestational age. Am J Epidemiol (2002) 155:203–9. 10.1093/aje/155.3.203 [DOI] [PubMed] [Google Scholar]
  • 79.Roberts JM, Hubel CA. The two stage model of preeclampsia: variations on the theme. Placenta (2009) 30(Suppl A):S32–7. 10.1016/j.placenta.2008.11.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Redman CWG. The six stages of pre-eclampsia. Pregnancy Hypertens (2014) 4:246. 10.1016/j.preghy.2014.04.020 [DOI] [PubMed] [Google Scholar]
  • 81.Sacks GP, Studena K, Sargent K, Redman CWG. Normal pregnancy and preeclampsia both produce inflammatory changes in peripheral blood leukocytes akin to those of sepsis. Am J Obstet Gynecol (1998) 179:80–6. 10.1016/S0002-9378(98)70254-6 [DOI] [PubMed] [Google Scholar]
  • 82.Redman CWG, Sargent IL. Pre-eclampsia, the placenta and the maternal systemic inflammatory response – a review. Placenta (2003) 24(Suppl A):S21–7. 10.1053/plac.2002.0930 [DOI] [PubMed] [Google Scholar]
  • 83.Hubel CA. Dyslipidemia and pre-eclampsia. In: Belfort MA, Lydall F, editors. Pre-Eclampsia-Aetiology and Clinical Practice. Cambridge: Cambridge University Press; (2006). p. 164–82. [Google Scholar]
  • 84.Kell DB. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson’s, Huntington’s, Alzheimer’s, prions, bactericides, chemical toxicology and others as examples. Arch Toxicol (2010) 577:825–89. 10.1007/s00204-010-0577-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature (2000) 407:242–8. 10.1038/35025215 [DOI] [PubMed] [Google Scholar]
  • 86.Maynard S, Epstein FH, Karumanchi SA. Preeclampsia and angiogenic imbalance. Annu Rev Med (2008) 59:61–78. 10.1146/annurev.med.59.110106.214058 [DOI] [PubMed] [Google Scholar]
  • 87.Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, et al. Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med (2004) 350:672–83. 10.1056/NEJMoa031884 [DOI] [PubMed] [Google Scholar]
  • 88.Levine RJ, Lam C, Qian C, Yu KF, Maynard SE, Sachs BP, et al. Soluble endoglin and other circulating antiangiogenic factors in preeclampsia. N Engl J Med (2006) 355:992–1005. 10.1056/NEJMoa055352 [DOI] [PubMed] [Google Scholar]
  • 89.Palomaki GE, Haddow JE, Haddow HR, Salahuddin S, Geahchan C, Cerdeira AS, et al. Modeling risk for severe adverse outcomes using angiogenic factor measurements in women with suspected preterm preeclampsia. Prenat Diagn (2015) 35:386–93. 10.1002/pd.4554 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Entman SS, Richardson LD. Clinical applications of the altered iron kinetics of toxemia of pregnancy. Am J Obstet Gynecol (1983) 146:568–74. 10.1016/0002-9378(83)90804-9 [DOI] [PubMed] [Google Scholar]
  • 91.MacDonald AB. Human fetal borreliosis, toxemia of pregnancy, and fetal death. Zentralbl Bakteriol Mikrobiol Hyg A (1986) 263:189–200. [DOI] [PubMed] [Google Scholar]
  • 92.Loudon I. Some historical aspects of toxaemia of pregnancy. A review. Br J Obstet Gynaecol (1991) 98:853–8. 10.1111/j.1471-0528.1991.tb13505.x [DOI] [PubMed] [Google Scholar]
  • 93.Meziani F, Tesse A, David E, Martinez MC, Wangesteen R, Schneider F, et al. Shed membrane particles from preeclamptic women generate vascular wall inflammation and blunt vascular contractility. Am J Pathol (2006) 169:1473–83. 10.2353/ajpath.2006.051304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Sugawara J, Mitsui-Saito M, Hayashi C, Hoshiai T, Senoo M, Chisaka H, et al. Decrease and senescence of endothelial progenitor cells in patients with preeclampsia. J Clin Endocrinol Metab (2005) 90:5329–32. 10.1210/jc.2005-0532 [DOI] [PubMed] [Google Scholar]
  • 95.Wang Y, Walsh SW. Placental mitochondria as a source of oxidative stress in pre-eclampsia. Placenta (1998) 19:581–6. 10.1016/S0143-4004(98)90018-2 [DOI] [PubMed] [Google Scholar]
  • 96.Schumacker PT. Current paradigms in cellular oxygen sensing. Adv Exp Med Biol (2003) 543:57–71. 10.1007/978-1-4419-8997-0_5 [DOI] [PubMed] [Google Scholar]
  • 97.Bradford Hill A. Environment and disease: association or causation? Proc R Soc Med (1965) 58:295–300. [PMC free article] [PubMed] [Google Scholar]
  • 98.Karmon A, Pilpel Y. Biological causal links on physiological and evolutionary time scales. Elife (2016) 5:e14424. 10.7554/eLife.14424 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kell DB. Metabolomics, modelling and machine learning in systems biology: towards an understanding of the languages of cells. The 2005 Theodor Bücher lecture. FEBS J (2006) 273:873–94. 10.1111/j.1742-4658.2006.05136.x [DOI] [PubMed] [Google Scholar]
  • 100.Gradmann C. A spirit of scientific rigour: Koch’s postulates in twentieth-century medicine. Microbes Infect (2014) 16:885–92. 10.1016/j.micinf.2014.08.012 [DOI] [PubMed] [Google Scholar]
  • 101.Falkow S. Molecular Koch’s postulates applied to microbial pathogenicity. Rev Infect Dis (1988) 10(Suppl 2):S274–6. 10.1093/cid/10.Supplement_2.S274 [DOI] [PubMed] [Google Scholar]
  • 102.Fredricks DN, Relman DA. Sequence-based identification of microbial pathogens – a reconsideration of Koch’s postulates. Clin Microbiol Rev (1996) 9:18–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Falkow S. Molecular Koch’s postulates applied to bacterial pathogenicity – a personal recollection 15 years later. Nat Rev Microbiol (2004) 2:67–72. 10.1038/nrmicro799 [DOI] [PubMed] [Google Scholar]
  • 104.Lowe AM, Yansouni CP, Behr MA. Causality and gastrointestinal infections: Koch, Hill, and Crohn’s. Lancet Infect Dis (2008) 8:720–6. 10.1016/S1473-3099(08)70257-3 [DOI] [PubMed] [Google Scholar]
  • 105.Seal JB, Morowitz M, Zaborina O, An G, Alverdy JC. The molecular Koch’s postulates and surgical infection: a view forward. Surgery (2010) 147:757–65. 10.1016/j.surg.2010.03.008 [DOI] [PubMed] [Google Scholar]
  • 106.Segre JA. What does it take to satisfy Koch’s postulates two centuries later? Microbial genomics and Propionibacteria acnes. J Invest Dermatol (2013) 133:2141–2. 10.1038/jid.2013.260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Byrd AL, Segre JA. Adapting Koch’s postulates. Science (2016) 351:224–6. 10.1126/science.aad6753 [DOI] [PubMed] [Google Scholar]
  • 108.Thagard P. Explanatory coherence. Behav Brain Sci (1989) 12:435–502. 10.1017/S0140525X00057046 [DOI] [Google Scholar]
  • 109.Thagard P, Verbeurgt K. Coherence as constraint satisfaction. Cogn Sci (1998) 22:1–24. 10.1207/s15516709cog2201_1 [DOI] [Google Scholar]
  • 110.Thagard P. How Scientists Explain Disease. Princeton, NJ: Princeton University Press; (1999). [Google Scholar]
  • 111.Thagard P. Coherence, truth, and the development of scientific knowledge. Philos Sci (2007) 74:28–47. 10.1086/520941 [DOI] [Google Scholar]
  • 112.Thagard P. Explanatory coherence. Reasoning: Studies of Human Inference and Its Foundations. (2008). p. 471–513. [Google Scholar]
  • 113.Pennington KA, Schlitt JM, Jackson DL, Schulz LC, Schust DJ. Preeclampsia: multiple approaches for a multifactorial disease. Dis Model Mech (2012) 5:9–18. 10.1242/dmm.008516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Magee LA, Pels A, Helewa M, Rey E, Von Dadelszen P, Canadian Hypertensive Disorders of Pregnancy Working Group . Diagnosis, evaluation, and management of the hypertensive disorders of pregnancy. Pregnancy Hypertens (2014) 4:105–45. 10.1016/j.preghy.2014.01.003 [DOI] [PubMed] [Google Scholar]
  • 115.Goddard KAG, Tromp G, Romero R, Olson JM, Lu Q, Xu Z, et al. Candidate-gene association study of mothers with pre-eclampsia, and their infants, analyzing 775 SNPs in 190 genes. Hum Hered (2007) 63:1–16. 10.1159/000097926 [DOI] [PubMed] [Google Scholar]
  • 116.Jebbink J, Wolters A, Fernando F, Afink G, van der Post J, Ris-Stalpers C. Molecular genetics of preeclampsia and HELLP syndrome – a review. Biochim Biophys Acta (2012) 1822:1960–9. 10.1016/j.bbadis.2012.08.004 [DOI] [PubMed] [Google Scholar]
  • 117.Fong FM, Sahemey MK, Hamedi G, Eyitayo R, Yates D, Kuan V, et al. Maternal genotype and severe preeclampsia: a HuGE review. Am J Epidemiol (2014) 180:335–45. 10.1093/aje/kwu151 [DOI] [PubMed] [Google Scholar]
  • 118.Williamson RD, O’Keeffe GW, Kenny LC. Activin signalling and pre-eclampsia: from genetic risk to pre-symptomatic biomarker. Cytokine (2015) 71:360–5. 10.1016/j.cyto.2014.11.017 [DOI] [PubMed] [Google Scholar]
  • 119.Maher B. The case of the missing heritability. Nature (2008) 456:18–21. 10.1038/456018a [DOI] [PubMed] [Google Scholar]
  • 120.Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, et al. Finding the missing heritability of complex diseases. Nature (2009) 461:747–53. 10.1038/nature08494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Zuk O, Hechter E, Sunyaev SR, Lander ES. The mystery of missing heritability: genetic interactions create phantom heritability. Proc Natl Acad Sci U S A (2012) 109:1193–8. 10.1073/pnas.1119675109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Carreiras M, Montagnani S, Layrisse Z. Preeclampsia: a multifactorial disease resulting from the interaction of the feto-maternal HLA genotype and HCMV infection. Am J Reprod Immunol (2002) 48:176–83. 10.1034/j.1600-0897.2002.01076.x [DOI] [PubMed] [Google Scholar]
  • 123.McCarthy CM, Kenny LC. Immunostimulatory role of mitochondrial DAMPs: alarming for pre-eclampsia? Am J Reprod Immunol (2016) 76:341–7. 10.1111/aji.12526 [DOI] [PubMed] [Google Scholar]
  • 124.McCarthy CM, Kenny LC. Mitochondrial [dys]function; culprit in pre-eclampsia? Clin Sci (Lond) (2016) 130:1179–84. 10.1042/CS20160103 [DOI] [PubMed] [Google Scholar]
  • 125.Kell DB. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med Genomics (2009) 2:2. 10.1186/1755-8794-2-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Kell DB, Pretorius E. Serum ferritin is an important disease marker, and is mainly a leakage product from damaged cells. Metallomics (2014) 6:748–73. 10.1039/C3MT00347G [DOI] [PubMed] [Google Scholar]
  • 127.Pretorius E, Kell DB. Diagnostic morphology: biophysical indicators for iron-driven inflammatory diseases. Integr Biol (2014) 6:486–510. 10.1039/c4ib00025k [DOI] [PubMed] [Google Scholar]
  • 128.Kell DB, Pretorius E. The simultaneous occurrence of both hypercoagulability and hypofibrinolysis in blood and serum during systemic inflammation, and the roles of iron and fibrin(ogen). Integr Biol (2015) 7:24–52. 10.1039/c4ib00173g [DOI] [PubMed] [Google Scholar]
  • 129.Pretorius E, Oberholzer HM, van der Spuy WJ, Swanepoel AC, Soma P. Qualitative scanning electron microscopy analysis of fibrin networks and platelet abnormalities in diabetes. Blood Coagul Fibrinol (2011) 22:463–7. 10.1097/MBC.0b013e3283468a0d [DOI] [PubMed] [Google Scholar]
  • 130.Pretorius E, Steyn H, Engelbrecht M, Swanepoel AC, Oberholzer HM. Differences in fibrin fiber diameters in healthy individuals and thromboembolic ischemic stroke patients. Blood Coagul Fibrinolysis (2011) 22:696–700. 10.1097/MBC.0b013e32834bdb32 [DOI] [PubMed] [Google Scholar]
  • 131.Pretorius E, Swanepoel AC, Oberholzer HM, van der Spuy WJ, Duim W, Wessels PF. A descriptive investigation of the ultrastructure of fibrin networks in thrombo-embolic ischemic stroke. J Thromb Thrombolysis (2011) 31:507–13. 10.1007/s11239-010-0538-5 [DOI] [PubMed] [Google Scholar]
  • 132.Pretorius E, Du Plooy J, Soma P, Gasparyan AY. An ultrastructural analysis of platelets, erythrocytes, white blood cells, and fibrin network in systemic lupus erythematosus. Rheumatol Int (2014) 34:1005–9. 10.1007/s00296-013-2817-x [DOI] [PubMed] [Google Scholar]
  • 133.Pretorius E, Swanepoel AC, Buys AV, Vermeulen N, Duim W, Kell DB. Eryptosis as a marker of Parkinson’s disease. Aging (2014) 6:788–819. 10.18632/aging.100695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Pretorius E, Bester J, Vermeulen N, Alummoottil S, Soma P, Buys AV, et al. Poorly controlled type 2 diabetes is accompanied by significant morphological and ultrastructural changes in both erythrocytes and in thrombin-generated fibrin: implications for diagnostics. Cardiovasc Diabetol (2015) 13:30. 10.1186/s12933-015-0192-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Bester J, Buys AV, Lipinski B, Kell DB, Pretorius E. High ferritin levels have major effects on the morphology of erythrocytes in Alzheimer’s disease. Front Aging Neurosci (2013) 5:00088. 10.3389/fnagi.2013.00088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Pretorius E, Bester J, Vermeulen N, Lipinski B, Gericke GS, Kell DB. Profound morphological changes in the erythrocytes and fibrin networks of patients with hemochromatosis or with hyperferritinemia, and their normalization by iron chelators and other agents. PLoS One (2014) 9:e85271. 10.1371/journal.pone.0085271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Pretorius E, Olumuyiwa-Akeredolu OO, Mbotwe S, Bester J. Erythrocytes and their role as health indicator: using structure in a patient-orientated precision medicine approach. Blood Rev (2016) 30:263–74. 10.1016/j.blre.2016.01.001 [DOI] [PubMed] [Google Scholar]
  • 138.Todros T, Vasario E, Cardaropoli S. Preeclampsia as an infectious disease. Exp Rev Obstet Gynecol (2007) 2:735–41. 10.1586/17474108.2.6.735 [DOI] [Google Scholar]
  • 139.Kell DB, Potgieter M, Pretorius E. Individuality, phenotypic differentiation, dormancy and ‘persistence’ in culturable bacterial systems: commonalities shared by environmental, laboratory, and clinical microbiology. F1000Research (2015) 4:179. 10.12688/f1000research.6709.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kell DB, Pretorius E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: the central roles of LPS and LPS-induced cell death. Integr Biol (2015) 7:1339–77. 10.1039/C5IB00158G [DOI] [PubMed] [Google Scholar]
  • 141.Potgieter M, Bester J, Kell DB, Pretorius E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS Microbiol Rev (2015) 39:567–91. 10.1093/femsre/fuv013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Pretorius E, Akeredolu O-O, Soma P, Kell DB. Major involvement of bacterial components in rheumatoid arthritis and its accompanying oxidative stress, systemic inflammation and hypercoagulability. Exp Biol Med (Forthcoming 2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Pretorius E, Bester J, Kell DB. A bacterial component to Alzheimer-type dementia seen via a systems biology approach that links iron dysregulation and inflammagen shedding to disease. J Alzheimers Dis (2016) 53:1237–56. 10.3233/JAD-160318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Proal AD, Albert PJ, Marshall TG. The human microbiome and autoimmunity. Curr Opin Rheumatol (2013) 25:234–40. 10.1097/BOR.0b013e32835cedbf [DOI] [PubMed] [Google Scholar]
  • 145.Mangin M, Sinha R, Fincher K. Inflammation and vitamin D: the infection connection. Inflamm Res (2014) 63:803–19. 10.1007/s00011-014-0755-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Proal AD, Albert PJ, Marshall TG. Inflammatory disease and the human microbiome. Discov Med (2014) 17:257–65. [PubMed] [Google Scholar]
  • 147.Itzhaki RF, Lathe R, Balin BJ, Ball MJ, Braak H, Bearer EL, et al. Microbes and Alzheimer’s disease. J Alzheimers Dis (2016) 51:979–84. 10.3233/JAD-160152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Morath S, Geyer A, Hartung T. Structure-function relationship of cytokine induction by lipoteichoic acid from Staphylococcus aureus. J Exp Med (2001) 193:393–7. 10.1084/jem.193.3.393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Postgate JR. Viability measurements and the survival of microbes under minimum stress. Adv Micro Physiol (1967) 1:1–23. 10.1016/S0065-2911(08)60248-9 [DOI] [Google Scholar]
  • 150.Postgate JR. Viable counts and viability. Methods Microbiol (1969) 1:611–28. 10.1016/S0580-9517(08)70149-1 [DOI] [Google Scholar]
  • 151.Postgate JR. Death in microbes and macrobes. In: Gray TRG, Postgate JR, editors. In the Survival of Vegetative Microbes. Cambridge: Cambridge University Press; (1976). p. 1–19. [Google Scholar]
  • 152.Staley JT, Konopka A. Measurement of in situ activities of nonphotosynthetic microorganisms in aquatic and terrestrial habitats. Annu Rev Microbiol (1985) 39:321–46. 10.1146/annurev.mi.39.100185.001541 [DOI] [PubMed] [Google Scholar]
  • 153.Tanaka T, Kawasaki K, Daimon S, Kitagawa W, Yamamoto K, Tamaki H, et al. A hidden pitfall in the preparation of agar media undermines microorganism cultivability. Appl Environ Microbiol (2014) 80:7659–66. 10.1128/AEM.02741-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Nichols D, Cahoon N, Trakhtenberg EM, Pham L, Mehta A, Belanger A, et al. Use of ichip for high-throughput in situ cultivation of “uncultivable” microbial species. Appl Environ Microbiol (2010) 76:2445–50. 10.1128/AEM.01754-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Browne HP, Forster SC, Anonye BO, Kumar N, Neville BA, Stares MD, et al. Culturing of ‘unculturable’ human microbiota reveals novel taxa and extensive sporulation. Nature (2016) 533:543–6. 10.1038/nature17645 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Kaprelyants AS, Gottschal JC, Kell DB. Dormancy in non-sporulating bacteria. FEMS Microbiol Rev (1993) 10:271–86. 10.1111/j.1574-6968.1993.tb05871.x [DOI] [PubMed] [Google Scholar]
  • 157.Kell DB, Kaprelyants AS, Weichart DH, Harwood CL, Barer MR. Viability and activity in readily culturable bacteria: a review and discussion of the practical issues. Antonie Van Leeuwenhoek (1998) 73:169–87. 10.1023/A:1000664013047 [DOI] [PubMed] [Google Scholar]
  • 158.Saito A, Rolfe RD, Edelstein PH, Finegold SM. Comparison of liquid growth media for Legionella pneumophila. J Clin Microbiol (1981) 14:623–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Meyer RD. Legionella infections – a review of 5 years of research. Rev Infect Dis (1983) 5:258–78. 10.1093/clinids/5.2.258 [DOI] [PubMed] [Google Scholar]
  • 160.Barker J, Farrell ID, Hutchison JG. Factors affecting growth of Legionella pneumophila in liquid media. J Med Microbiol (1986) 22:97–100. 10.1099/00222615-22-2-97 [DOI] [PubMed] [Google Scholar]
  • 161.Molinari J. Legionella and human disease: part 1: a path of scientific and community discovery. Compend Contin Educ Dent (1997) 18:556–9. [PubMed] [Google Scholar]
  • 162.Maiwald M, Schuhmacher F, Ditton HJ, Vonherbay A. Environmental occurrence of the Whipple’s disease bacterium (Tropheryma whippelii). Appl Environ Microbiol (1998) 64:760–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Maiwald M, Relman DA. Whipple’s disease and Tropheryma whippelii: secrets slowly revealed. Clin Infect Dis (2001) 32:457–63. 10.1086/318512 [DOI] [PubMed] [Google Scholar]
  • 164.Omsland A, Cockrell DC, Howe D, Fischer ER, Virtaneva K, Sturdevant DE, et al. Host cell-free growth of the Q fever bacterium Coxiella burnetii. Proc Natl Acad Sci U S A (2009) 106:4430–4. 10.1073/pnas.0812074106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Omsland A. Axenic growth of Coxiella burnetii. Adv Exp Med Biol (2012) 984:215–29. 10.1007/978-94-007-4315-1_11 [DOI] [PubMed] [Google Scholar]
  • 166.Marshall BJ. One hundred years of discovery and rediscovery of Helicobacter pylori and its association with peptic ulcer disease. In: Mobley HLT, Mendz GL, Hazell SL, editors. Helicobacter pylori: Physiology and Genetics. Washington, DC: ASM Press; (2001). p. 19–24. [PubMed] [Google Scholar]
  • 167.Marshall B. Helicobacter connections. ChemMedChem (2006) 1:783–802. 10.1002/cmdc.200600153 [DOI] [PubMed] [Google Scholar]
  • 168.Marshall BJ, Warren JR. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet (1984) 1:1311–5. 10.1016/S0140-6736(84)91816-6 [DOI] [PubMed] [Google Scholar]
  • 169.Marshall BJ, Armstrong JA, Mcgechie DB, Glancy RJ. Attempt to fulfil Koch’s postulates for pyloric Campylobacter. Med J Aust (1985) 142:436–9. [DOI] [PubMed] [Google Scholar]
  • 170.Kaprelyants AS, Kell DB. Dormancy in stationary-phase cultures of Micrococcus luteus: flow cytometric analysis of starvation and resuscitation. Appl Environ Microbiol (1993) 59:3187–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Kaprelyants AS, Mukamolova GV, Kell DB. Estimation of dormant Micrococcus luteus cells by penicillin lysis and by resuscitation in cell-free spent medium at high dilution. FEMS Microbiol Lett (1994) 115:347–52. 10.1111/j.1574-6968.1994.tb06662.x [DOI] [Google Scholar]
  • 172.Wayne LG. Dormancy of Mycobacterium tuberculosis and latency of disease. Eur J Clin Microbiol Infect Dis (1994) 13:908–14. 10.1007/BF02111491 [DOI] [PubMed] [Google Scholar]
  • 173.Kana BD, Gordhan BG, Downing KJ, Sung N, Vostroktunova G, Machowski EE, et al. The resuscitation-promoting factors of Mycobacterium tuberculosis are required for virulence and resuscitation from dormancy but are collectively dispensable for growth in vitro. Mol Microbiol (2008) 67:672–84. 10.1111/j.1365-2958.2007.06078.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Gengenbacher M, Kaufmann SHE. Mycobacterium tuberculosis: success through dormancy. FEMS Microbiol Rev (2012) 36:514–32. 10.1111/j.1574-6976.2012.00331.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Alnimr AM. Dormancy models for Mycobacterium tuberculosis: a minireview. Braz J Microbiol (2015) 46:641–7. 10.1590/S1517-838246320140507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Shleeva M, Kondratieva T, Rubakova E, Vostroknutova G, Kaprelyants A, Apt A. Reactivation of dormant “non-culturable” Mycobacterium tuberculosis developed in vitro after injection in mice: both the dormancy depth and host genetics influence the outcome. Microb Pathog (2015) 78:63–6. 10.1016/j.micpath.2014.11.016 [DOI] [PubMed] [Google Scholar]
  • 177.Mukamolova GV, Kaprelyants AS, Young DI, Young M, Kell DB. A bacterial cytokine. Proc Natl Acad Sci U S A (1998) 95:8916–21. 10.1073/pnas.95.15.8916 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Mukamolova GV, Turapov OA, Kazarian K, Telkov M, Kaprelyants AS, Kell DB, et al. The rpf gene of Micrococcus luteus encodes an essential secreted growth factor. Mol Microbiol (2002) 46:611–21. 10.1046/j.1365-2958.2002.03183.x [DOI] [PubMed] [Google Scholar]
  • 179.Mukamolova GV, Turapov OA, Young DI, Kaprelyants AS, Kell DB, Young M. A family of autocrine growth factors in Mycobacterium tuberculosis. Mol Microbiol (2002) 46:623–35. 10.1046/j.1365-2958.2002.03184.x [DOI] [PubMed] [Google Scholar]
  • 180.Mukamolova GV, Murzin AG, Salina EG, Demina GR, Kell DB, Kaprelyants AS, et al. Muralytic activity of Micrococcus luteus Rpf and its relationship to physiological activity in promoting bacterial growth and resuscitation. Mol Microbiol (2006) 59:84–98. 10.1111/j.1365-2958.2005.04930.x [DOI] [PubMed] [Google Scholar]
  • 181.Mukamolova GV, Turapov O, Malkin J, Woltmann G, Barer MR. Resuscitation-promoting factors reveal an occult population of tubercle bacilli in sputum. Am J Respir Crit Care Med (2010) 181:174–80. 10.1164/rccm.200905-0661OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Yeremeev VV, Kondratieva TK, Rubakova EI, Petrovskaya SN, Kazarian KA, Telkov MV, et al. Proteins of the Rpf family: immune cell reactivity and vaccination efficacy against tuberculosis in mice. Infect Immun (2003) 71:4789–94. 10.1128/IAI.71.8.4789-4794.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Zvi A, Ariel N, Fulkerson J, Sadoff JC, Shafferman A. Whole genome identification of Mycobacterium tuberculosis vaccine candidates by comprehensive data mining and bioinformatic analyses. BMC Med Genomics (2008) 1:18. 10.1186/1755-8794-1-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Dye C, Scheele S, Dolin P, Pathania V, Raviglione RC. Global burden of tuberculosis – estimated incidence, prevalence, and mortality by country. JAMA (1999) 282:677–86. 10.1001/jama.282.7.677 [DOI] [PubMed] [Google Scholar]
  • 185.Flynn JL, Chan J. Tuberculosis: latency and reactivation. Infect Immun (2001) 69:4195–201. 10.1128/IAI.69.7.4195-4201.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Wayne LG, Sohaskey CD. Nonreplicating persistence of Mycobacterium tuberculosis. Annu Rev Microbiol (2001) 55:139–63. 10.1146/annurev.micro.55.1.139 [DOI] [PubMed] [Google Scholar]
  • 187.Gomez JE, McKinney JD. M. tuberculosis persistence, latency, and drug tolerance. Tuberculosis (Edinb) (2004) 84:29–44. 10.1016/j.tube.2003.08.003 [DOI] [PubMed] [Google Scholar]
  • 188.Amieva MR, El-Omar EM. Host-bacterial interactions in Helicobacter pylori infection. Gastroenterology (2008) 134:306–23. 10.1053/j.gastro.2007.11.009 [DOI] [PubMed] [Google Scholar]
  • 189.Cover TL, Blaser MJ. Helicobacter pylori in health and disease. Gastroenterology (2009) 136:1863–73. 10.1053/j.gastro.2009.01.073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Domingue GJ, Woody HB. Bacterial persistence and expression of disease. Clin Microbiol Rev (1997) 10:320–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Young D, Stark J, Kirschner D. Systems biology of persistent infection: tuberculosis as a case study. Nat Rev Microbiol (2008) 6:520–8. 10.1038/nrmicro1919 [DOI] [PubMed] [Google Scholar]
  • 192.Lewis K. Persister cells. Annu Rev Microbiol (2010) 64:357–72. 10.1146/annurev.micro.112408.134306 [DOI] [PubMed] [Google Scholar]
  • 193.Amato SM, Fazen CH, Henry TC, Mok WWK, Orman MA, Sandvik EL, et al. The role of metabolism in bacterial persistence. Front Microbiol (2014) 5:70. 10.3389/Fmicb.2014.00070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Conlon BP. Staphylococcus aureus chronic and relapsing infections: evidence of a role for persister cells: an investigation of persister cells, their formation and their role in S. aureus disease. Bioessays (2014) 36:991–6. 10.1002/bies.201400080 [DOI] [PubMed] [Google Scholar]
  • 195.Kester JC, Fortune SM. Persisters and beyond: mechanisms of phenotypic drug resistance and drug tolerance in bacteria. Crit Rev Biochem Mol Biol (2014) 49:91–101. 10.3109/10409238.2013.869543 [DOI] [PubMed] [Google Scholar]
  • 196.Levin BR, Concepción-Acevedo J, Udekwu KI. Persistence: a copacetic and parsimonious hypothesis for the existence of non-inherited resistance to antibiotics. Curr Opin Microbiol (2014) 21:18–21. 10.1016/j.mib.2014.06.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Maisonneuve E, Gerdes K. Molecular mechanisms underlying bacterial persisters. Cell (2014) 157:539–48. 10.1016/j.cell.2014.02.050 [DOI] [PubMed] [Google Scholar]
  • 198.Rank RG, Yeruva L. Hidden in plain sight: chlamydial gastrointestinal infection and its relevance to persistence in human genital infection. Infect Immun (2014) 82:1362–71. 10.1128/IAI.01244-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Holden DW. Persisters unmasked. Science (2015) 347:30–2. 10.1126/science.1262033 [DOI] [PubMed] [Google Scholar]
  • 200.Orman MA, Mok WWK, Brynildsen MP. Aminoglycoside-enabled elucidation of bacterial persister metabolism. Curr Protoc Microbiol (2015) 36:1–14. 10.1002/9780471729259.mc1709s36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Stepanyan K, Wenseleers T, Duéñez-Guzmán EA, Muratori F, Van Den Bergh B, Verstraeten N, et al. Fitness trade-offs explain low levels of persister cells in the opportunistic pathogen Pseudomonas aeruginosa. Mol Ecol (2015) 24:1572–83. 10.1111/mec.13127 [DOI] [PubMed] [Google Scholar]
  • 202.Gerdes K, Semsey S. Pumping persisters. Nature (2016) 534:41–2. 10.1038/nature18442 [DOI] [PubMed] [Google Scholar]
  • 203.Pu Y, Zhao Z, Li Y, Zou J, Ma Q, Zhao Y, et al. Enhanced efflux activity facilitates drug tolerance in dormant bacterial cells. Mol Cell (2016) 62:284–94. 10.1016/j.molcel.2016.03.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Dehio C, Berry C, Bartenschlager R. Persistent intracellular pathogens. FEMS Microbiol Rev (2012) 36:513. 10.1111/j.1574-6976.2012.00336.x [DOI] [PubMed] [Google Scholar]
  • 205.Prideaux B, Via LE, Zimmerman MD, Eum S, Sarathy J, O’Brien P, et al. The association between sterilizing activity and drug distribution into tuberculosis lesions. Nat Med (2015) 21:1223–7. 10.1038/nm.3937 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Dobson PD, Kell DB. Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule? Nat Rev Drug Discov (2008) 7:205–20. 10.1038/nrd2438 [DOI] [PubMed] [Google Scholar]
  • 207.Kell DB, Dobson PD, Oliver SG. Pharmaceutical drug transport: the issues and the implications that it is essentially carrier-mediated only. Drug Discov Today (2011) 16:704–14. 10.1016/j.drudis.2011.05.010 [DOI] [PubMed] [Google Scholar]
  • 208.Kell DB, Dobson PD, Bilsland E, Oliver SG. The promiscuous binding of pharmaceutical drugs and their transporter-mediated uptake into cells: what we (need to) know and how we can do so. Drug Discov Today (2013) 18:218–39. 10.1016/j.drudis.2012.11.008 [DOI] [PubMed] [Google Scholar]
  • 209.Kell DB, Oliver SG. How drugs get into cells: tested and testable predictions to help discriminate between transporter-mediated uptake and lipoidal bilayer diffusion. Front Pharmacol (2014) 5:231. 10.3389/fphar.2014.00231 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Thwaites GE, Gant V. Are bloodstream leukocytes Trojan horses for the metastasis of Staphylococcus aureus? Nat Rev Microbiol (2011) 9:215–22. 10.1038/nrmicro2508 [DOI] [PubMed] [Google Scholar]
  • 211.Seubert A, Schulein R, Dehio C. Bacterial persistence within erythrocytes: a unique pathogenic strategy of Bartonella spp. Int J Med Microbiol (2002) 291:555–60. 10.1078/1438-4221-00167 [DOI] [PubMed] [Google Scholar]
  • 212.Mysorekar IU, Hultgren SJ. Mechanisms of uropathogenic Escherichia coli persistence and eradication from the urinary tract. Proc Natl Acad Sci U S A (2006) 103:14170–5. 10.1073/pnas.0602136103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Blango MG, Mulvey MA. Persistence of uropathogenic Escherichia coli in the face of multiple antibiotics. Antimicrob Agents Chemother (2010) 54:1855–63. 10.1128/AAC.00014-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Chen SL, Wu M, Henderson JP, Hooton TM, Hibbing ME, Hultgren SJ, et al. Genomic diversity and fitness of E. coli strains recovered from the intestinal and urinary tracts of women with recurrent urinary tract infection. Sci Transl Med (2013) 5:184ra160. 10.1126/scitranslmed.3005497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Goneau LW, Hannan TJ, Macphee RA, Schwartz DJ, Macklaim JM, Gloor GB, et al. Subinhibitory antibiotic therapy alters recurrent urinary tract infection pathogenesis through modulation of bacterial virulence and host immunity. MBio (2015) 6:e00356–15. 10.1128/mBio.00356-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Sullivan WJ, Jr, Jeffers V. Mechanisms of Toxoplasma gondii persistence and latency. FEMS Microbiol Rev (2012) 36:717–33. 10.1111/j.1574-6976.2011.00305.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Posey JE, Gherardini FC. Lack of a role for iron in the Lyme disease pathogen. Science (2000) 288:1651–3. 10.1126/science.288.5471.1651 [DOI] [PubMed] [Google Scholar]
  • 218.Aguirre JD, Clark HM, Mcilvin M, Vazquez C, Palmere SL, Grab DJ, et al. A manganese-rich environment supports superoxide dismutase activity in a Lyme disease pathogen, Borrelia burgdorferi. J Biol Chem (2013) 288:8468–78. 10.1074/jbc.M112.433540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Weinberg ED. Nutritional immunity. Host’s attempt to withold iron from microbial invaders. JAMA (1975) 231:39–41. 10.1001/jama.1975.03240130021018 [DOI] [PubMed] [Google Scholar]
  • 220.Weinberg ED. Iron withholding: a defense against infection and neoplasia. Physiol Rev (1984) 64:65–102. [DOI] [PubMed] [Google Scholar]
  • 221.Marx JJM. Iron and infection: competition between host and microbes for a precious element. Best Pract Res Clin Haematol (2002) 15:411–26. 10.1053/beha.2002.0001 [DOI] [PubMed] [Google Scholar]
  • 222.Fischbach MA, Lin HN, Liu DR, Walsh CT. How pathogenic bacteria evade mammalian sabotage in the battle for iron. Nat Chem Biol (2006) 2:132–8. 10.1038/nchembio771 [DOI] [PubMed] [Google Scholar]
  • 223.Weinberg ED, Miklossy J. Iron withholding: a defense against disease. J Alzheimers Dis (2008) 13:451–63. [DOI] [PubMed] [Google Scholar]
  • 224.Reid DW, Anderson GJ, Lamont IL. Role of lung iron in determining the bacterial and host struggle in cystic fibrosis. Am J Physiol Lung Cell Mol Physiol (2009) 297:L795–802. 10.1152/ajplung.00132.2009 [DOI] [PubMed] [Google Scholar]
  • 225.Weinberg ED. Iron availability and infection. Biochim Biophys Acta (2009) 1790:600–5. 10.1016/j.bbagen.2008.07.002 [DOI] [PubMed] [Google Scholar]
  • 226.Chu BC, Garcia-Herrero A, Johanson TH, Krewulak KD, Lau CK, Peacock RS, et al. Siderophore uptake in bacteria and the battle for iron with the host; a bird’s eye view. Biometals (2010) 23:601–11. 10.1007/s10534-010-9361-x [DOI] [PubMed] [Google Scholar]
  • 227.Nairz M, Schroll A, Sonnweber T, Weiss G. The struggle for iron – a metal at the host-pathogen interface. Cell Microbiol (2010) 12:1691–702. 10.1111/j.1462-5822.2010.01529.x [DOI] [PubMed] [Google Scholar]
  • 228.Skaar EP. The battle for iron between bacterial pathogens and their vertebrate hosts. PLoS Pathog (2010) 6:e1000949. 10.1371/journal.ppat.1000949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Hood MI, Skaar EP. Nutritional immunity: transition metals at the pathogen-host interface. Nat Rev Microbiol (2012) 10:525–37. 10.1038/nrmicro2836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Cassat JE, Skaar EP. Iron in infection and immunity. Cell Host Microbe (2013) 13:509–19. 10.1016/j.chom.2013.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Deriu E, Liu JZ, Pezeshki M, Edwards RA, Ochoa RJ, Contreras H, et al. Probiotic bacteria reduce Salmonella typhimurium intestinal colonization by competing for iron. Cell Host Microbe (2013) 14:26–37. 10.1016/j.chom.2013.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Leal SM, Jr, Roy S, Vareechon C, Carrion S, Clark H, Lopez-Berges MS, et al. Targeting iron acquisition blocks infection with the fungal pathogens Aspergillus fumigatus and Fusarium oxysporum. PLoS Pathog (2013) 9:e1003436. 10.1371/journal.ppat.1003436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Silva-Gomes S, Vale-Costa S, Appelberg R, Gomes MS. Iron in intracellular infection: to provide or to deprive? Front Cell Infect Microbiol (2013) 3:96. 10.3389/fcimb.2013.00096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Armitage AE, Drakesmith H. The battle for iron. Science (2014) 346:1299–300. 10.1126/science.aaa2468 [DOI] [PubMed] [Google Scholar]
  • 235.Becker KW, Skaar EP. Metal limitation and toxicity at the interface between host and pathogen. FEMS Microbiol Rev (2014) 38:1235–49. 10.1111/1574-6976.12087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Diaz-Ochoa VE, Jellbauer S, Klaus S, Raffatellu M. Transition metal ions at the crossroads of mucosal immunity and microbial pathogenesis. Front Cell Infect Microbiol (2014) 4:2. 10.3389/fcimb.2014.00002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Potrykus J, Ballou ER, Childers DS, Brown AJP. Conflicting interests in the pathogen-host tug of war: fungal micronutrient scavenging versus mammalian nutritional immunity. PLoS Pathog (2014) 10:e1003910. 10.1371/journal.ppat.1003910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Barber MF, Elde NC. Buried treasure: evolutionary perspectives on microbial iron piracy. Trends Genet (2015) 31:627–36. 10.1016/j.tig.2015.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Nairz M, Ferring-Appel D, Casarrubea D, Sonnweber T, Viatte L, Schroll A, et al. Iron regulatory proteins mediate host resistance to Salmonella infection. Cell Host Microbe (2015) 18:254–61. 10.1016/j.chom.2015.06.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Raymond KN, Allred BE, Sia AK. Coordination chemistry of microbial iron transport. Acc Chem Res (2015) 48:2496–505. 10.1021/acs.accounts.5b00301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Subashchandrabose S, Mobley HLT. Back to the metal age: battle for metals at the host-pathogen interface during urinary tract infection. Metallomics (2015) 7:935–42. 10.1039/c4mt00329b [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Wessling-Resnick M. Nramp1 and other transporters involved in metal withholding during infection. J Biol Chem (2015) 290:18984–90. 10.1074/jbc.R115.643973 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Xia Y, Farah N, Maxan A, Zhou J, Lehmann C. Therapeutic iron restriction in sepsis. Med Hypotheses (2016) 89:37–9. 10.1016/j.mehy.2016.01.018 [DOI] [PubMed] [Google Scholar]
  • 244.Entman SS, Richardson LD, Killam AP. Elevated serum ferritin in the altered ferrokinetics of toxemia of pregnancy. Am J Obstet Gynecol (1982) 144:418–22. 10.1016/0002-9378(82)90247-2 [DOI] [PubMed] [Google Scholar]
  • 245.Samuels P, Main EK, Mennuti MT, Gabbe SG. The origin of increased serum iron in pregnancy-induced hypertension. Am J Obstet Gynecol (1987) 157:721–5. 10.1016/S0002-9378(87)80037-6 [DOI] [PubMed] [Google Scholar]
  • 246.Raman L, Pawashe AB, Yasodhara P. Hyperferritinemia in pregnancy induced hypertension and eclampsia. J Postgrad Med (1992) 38:65–7. [PubMed] [Google Scholar]
  • 247.Hubel CA, Kozlov AV, Kagan VE, Evans RW, Davidge ST, McLaughlin MK, et al. Decreased transferrin and increased transferrin saturation in sera of women with preeclampsia: implications for oxidative stress. Am J Obstet Gynecol (1996) 175:692–700. 10.1053/ob.1996.v175.a74252 [DOI] [PubMed] [Google Scholar]
  • 248.Lao TT, Tam KF, Chan LY. Third trimester iron status and pregnancy outcome in non-anaemic women; pregnancy unfavourably affected by maternal iron excess. Hum Reprod (2000) 15:1843–8. 10.1093/humrep/15.8.1843 [DOI] [PubMed] [Google Scholar]
  • 249.Rayman MP, Barlis J, Evans RW, Redman CW, King LJ. Abnormal iron parameters in the pregnancy syndrome preeclampsia. Am J Obstet Gynecol (2002) 187:412–8. 10.1067/mob.2002.123895 [DOI] [PubMed] [Google Scholar]
  • 250.Serdar Z, Gür E, Develioğlu O. Serum iron and copper status and oxidative stress in severe and mild preeclampsia. Cell Biochem Funct (2006) 24:209–15. 10.1002/cbf.1235 [DOI] [PubMed] [Google Scholar]
  • 251.Smith TG, Robbins PA. Iron, pre-eclampsia and hypoxia-inducible factor. BJOG (2007) 114:1581–2. 10.1111/j.1471-0528.2007.01490.x [DOI] [PubMed] [Google Scholar]
  • 252.Bhatla N, Kaul N, Lal N, Kriplani A, Agarwal N, Saxena R, et al. Comparison of effect of daily versus weekly iron supplementation during pregnancy on lipid peroxidation. J Obstet Gynaecol Res (2009) 35:438–45. 10.1111/j.1447-0756.2008.00972.x [DOI] [PubMed] [Google Scholar]
  • 253.Siddiqui IA, Jaleel A, Kadri HM, Saeed WA, Tamimi W. Iron status parameters in preeclamptic women. Arch Gynecol Obstet (2011) 284:587–91. 10.1007/s00404-010-1728-2 [DOI] [PubMed] [Google Scholar]
  • 254.Fatima N, Islam F, Noor L, Das SR, Zeba D, Zesmin F. Serum ferritin in preeclampsia and eclampsia: a case control study. Faridpur Med Coll J (2013) 8:18–21. 10.3329/fmcj.v8i1.16892 [DOI] [Google Scholar]
  • 255.Kandi S, Sudhakar T, Ramadevi C, Venugopal B, Rajkumar, Rafi M, et al. Pre eclampsia and iron status: a review. Am J Med Biol Res (2014) 2:121–3. 10.12691/ajmbr-2-6-1 [DOI] [Google Scholar]
  • 256.Negi R, Pande D, Karki K, Kumar A, Khanna RS, Khanna HD. Association of oxidative DNA damage, protein oxidation and antioxidant function with oxidative stress induced cellular injury in pre-eclamptic/eclamptic mothers during fetal circulation. Chem Biol Interact (2014) 208:77–83. 10.1016/j.cbi.2013.11.010 [DOI] [PubMed] [Google Scholar]
  • 257.Bester J, Soma P, Kell DB, Pretorius E. Viscoelastic and ultrastructural characteristics of whole blood and plasma in Alzheimer-type dementia, and the possible role of bacterial lipopolysaccharides (LPS). Oncotarget (2015) 6:35284–303. 10.18632/oncotarget.6074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Pretorius E, Mbotwe S, Bester J, Robinson C, Kell DB. Acute induction of anomalous blood clotting by highly substoichiometric levels of bacterial lipopolysaccharide (LPS). bioRxiv (2016) 053538. 10.1101/053538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Pretorius E, Mbotwe S, Bester J, Robinson CJ, Kell DB. Acute induction of anomalous and amyloidogenic blood clotting by molecular amplification of highly substoichiometric levels of bacterial lipopolysaccharide. J R Soc Interface (2016) 123:20160539. 10.1098/rsif.2016.0539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Lin IH, Miller DS, Bertics PJ, Murphy CJ, De Pablo JJ, Abbott NL. Endotoxin-induced structural transformations in liquid crystalline droplets. Science (2011) 332:1297–300. 10.1126/science.1195639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Miller DS, Abbott NL. Influence of droplet size, pH and ionic strength on endotoxin-triggered ordering transitions in liquid crystalline droplets. Soft Matter (2013) 9:374–82. 10.1039/C2SM26811F [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Rahman MM, Abe SK, Rahman MS, Kanda M, Narita S, Bilano V, et al. Maternal anemia and risk of adverse birth and health outcomes in low- and middle-income countries: systematic review and meta-analysis. Am J Clin Nutr (2016) 103:495–504. 10.3945/ajcn.115.107896 [DOI] [PubMed] [Google Scholar]
  • 263.Harris CM, Kell DB. The estimation of microbial biomass. Biosensors (1985) 1:17–84. 10.1016/0265-928X(85)85005-7 [DOI] [PubMed] [Google Scholar]
  • 264.Kaprelyants AS, Kell DB. Do bacteria need to communicate with each other for growth? Trends Microbiol (1996) 4:237–42. 10.1016/0966-842X(96)10035-4 [DOI] [PubMed] [Google Scholar]
  • 265.Domingue GJ. Electron dense cytoplasmic particles and chronic infection – a bacterial pleomorphy hypothesis. Endocytobiosis Cell Res (1995) 11:19–40. [Google Scholar]
  • 266.Domingue GJ, Sr. Cryptic bacterial infection in chronic prostatitis: diagnostic and therapeutic implications. Curr Opin Urol (1998) 8:45–9. 10.1097/00042307-199801000-00009 [DOI] [PubMed] [Google Scholar]
  • 267.Mattman L. Cell Wall Deficient Forms: Stealth Pathogens. 3rd ed Boca Raton: CRC Press; (2001). [Google Scholar]
  • 268.Domingue GJ. Demystifying pleomorphic forms in persistence and expression of disease: are they bacteria, and is peptidoglycan the solution? Discov Med (2010) 10:234–46. [PubMed] [Google Scholar]
  • 269.Davey HM, Kell DB. Flow cytometry and cell sorting of heterogeneous microbial populations: the importance of single-cell analysis. Microbiol Rev (1996) 60:641–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Kaprelyants AS, Kell DB. Rapid assessment of bacterial viability and vitality using rhodamine 123 and flow cytometry. J Appl Bacteriol (1992) 72:410–22. 10.1111/j.1365-2672.1992.tb01854.x [DOI] [Google Scholar]
  • 271.Gant VA, Warnes G, Phillips I, Savidge GF. The application of flow cytometry to the study of bacterial responses to antibiotics. J Med Microbiol (1993) 39:147–54. 10.1099/00222615-39-2-147 [DOI] [PubMed] [Google Scholar]
  • 272.Mason DJ, Shanmuganathan S, Mortimer FC, Gant VA. A fluorescent Gram stain for flow cytometry and epifluorescence microscopy. Appl Environ Microbiol (1998) 64:2681–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Nebe-Von-Caron G, Stephens PJ, Hewitt CJ, Powell JR, Badley RA. Analysis of bacterial function by multi-colour fluorescence flow cytometry and single cell sorting. J Microbiol Methods (2000) 42:97–114. 10.1016/S0167-7012(00)00181-0 [DOI] [PubMed] [Google Scholar]
  • 274.Shapiro HM, Nebe-Von-Caron G. Multiparameter flow cytometry of bacteria. Methods Mol Biol (2004) 263:33–44. 10.1385/1-59259-773-4:033 [DOI] [PubMed] [Google Scholar]
  • 275.Müller S, Nebe-Von-Caron G. Functional single-cell analyses: flow cytometry and cell sorting of microbial populations and communities. FEMS Microbiol Rev (2010) 34:554–87. 10.1111/j.1574-6976.2010.00214.x [DOI] [PubMed] [Google Scholar]
  • 276.Davey HM. Life, death, and in-between: meanings and methods in microbiology. Appl Environ Microbiol (2011) 77:5571–6. 10.1128/AEM.00744-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Davey HM, Hexley P. Red but not dead? Membranes of stressed Saccharomyces cerevisiae are permeable to propidium iodide. Environ Microbiol (2011) 13:163–71. 10.1111/j.1462-2920.2010.02317.x [DOI] [PubMed] [Google Scholar]
  • 278.Kell DB, Markx GH, Davey CL, Todd RW. Real-time monitoring of cellular biomass: methods and applications. Trends Anal Chem (1990) 9:190–4. 10.1016/0165-9936(90)87042-K [DOI] [Google Scholar]
  • 279.Kell DB, Sonnleitner B. GMP – good modelling practice: an essential component of good manufacturing practice. Trends Biotechnol (1995) 13:481–92. 10.1016/S0167-7799(00)89006-X [DOI] [Google Scholar]
  • 280.Firstenberg-Eden R, Eden G. Impedance Microbiology. Letchworth: Research Studies Press; (1984). [Google Scholar]
  • 281.Harris CM, Todd RW, Bungard SJ, Lovitt RW, Morris JG, Kell DB. The dielectric permittivity of microbial suspensions at radio frequencies: a novel method for the estimation of microbial biomass. Enzyme Microbiol Technol (1987) 9:181–6. 10.1016/0141-0229(87)90075-5 [DOI] [Google Scholar]
  • 282.Kell DB, Davey CL. Conductimetric and impedimetric devices. In: Cass AEG, editor. Biosensors: A Practical Approach. Oxford: IRL Press; (1990). p. 125–54. [Google Scholar]
  • 283.Woese CR, Fox GE. Phylogenetic structure of the prokaryotic domain: the primary kingdoms. Proc Natl Acad Sci U S A (1977) 74:5088–90. 10.1073/pnas.74.11.5088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Petti CA. Detection and identification of microorganisms by gene amplification and sequencing. Clin Infect Dis (2007) 44:1108–14. 10.1086/512818 [DOI] [PubMed] [Google Scholar]
  • 285.Klouche M, Schröder U. Rapid methods for diagnosis of bloodstream infections. Clin Chem Lab Med (2008) 46:888–908. 10.1515/CCLM.2008.157 [DOI] [PubMed] [Google Scholar]
  • 286.Woo PCY, Lau SKP, Teng JLL, Tse H, Yuen KY. Then and now: use of 16S rDNA gene sequencing for bacterial identification and discovery of novel bacteria in clinical microbiology laboratories. Clin Microbiol Infect (2008) 14:908–34. 10.1111/j.1469-0691.2008.02070.x [DOI] [PubMed] [Google Scholar]
  • 287.Cherkaoui A, Emonet S, Ceroni D, Candolfi B, Hibbs J, Francois P, et al. Development and validation of a modified broad-range 16S rDNA PCR for diagnostic purposes in clinical microbiology. J Microbiol Methods (2009) 79:227–31. 10.1016/j.mimet.2009.09.014 [DOI] [PubMed] [Google Scholar]
  • 288.Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci U S A (2011) 108(Suppl 1):4516–22. 10.1073/pnas.1000080107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Kramski M, Gaeguta AJ, Lichtfuss GF, Rajasuriar R, Crowe SM, French MA, et al. Novel sensitive real-time PCR for quantification of bacterial 16S rRNA genes in plasma of HIV-infected patients as a marker for microbial translocation. J Clin Microbiol (2011) 49:3691–3. 10.1128/JCM.01018-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Kämpfer P. Systematics of prokaryotes: the state of the art. Antonie Van Leeuwenhoek (2012) 101:3–11. 10.1007/s10482-011-9660-4 [DOI] [PubMed] [Google Scholar]
  • 291.Ohlin A, Bäckman A, Ewald U, Schollin J, Björkqvist M. Diagnosis of neonatal sepsis by broad-range 16S real-time polymerase chain reaction. Neonatology (2012) 101:241–6. 10.1159/000334655 [DOI] [PubMed] [Google Scholar]
  • 292.Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol (2013) 31:814–21. 10.1038/nbt.2676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Mizrahi-Man O, Davenport ER, Gilad Y. Taxonomic classification of bacterial 16S rRNA genes using short sequencing reads: evaluation of effective study designs. PLoS One (2013) 8:e53608. 10.1371/journal.pone.0053608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Valencia-Shelton F, Loeffelholz M. Nonculture techniques for the detection of bacteremia and fungemia. Future Microbiol (2014) 9:543–59. 10.2217/fmb.14.8 [DOI] [PubMed] [Google Scholar]
  • 295.Yarza P, Yilmaz P, Pruesse E, Glöckner FO, Ludwig W, Schleifer KH, et al. Uniting the classification of cultured and uncultured bacteria and archaea using 16S rRNA gene sequences. Nat Rev Microbiol (2014) 12:635–45. 10.1038/nrmicro3330 [DOI] [PubMed] [Google Scholar]
  • 296.Zumla A, Al-Tawfiq JA, Enne VI, Kidd M, Drosten C, Breuer J, et al. Rapid point of care diagnostic tests for viral and bacterial respiratory tract infections – needs, advances, and future prospects. Lancet Infect Dis (2014) 14:1123–35. 10.1016/S1473-3099(14)70827-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.D’Amore R, Ijaz UZ, Schirmer M, Kenny JG, Gregory R, Darby AC, et al. A comprehensive benchmarking study of protocols and sequencing platforms for 16S rRNA community profiling. BMC Genomics (2016) 17:55. 10.1186/s12864-015-2194-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Versalovic J, Carroll KC, Funke G, Jorgensen JH, Landry ML, Warnock DW. Manual of Clinical Microbiology. 10th ed Washington: American Society of Microbiology; (2011). [Google Scholar]
  • 299.Nikkari S, McLaughlin IJ, Bi W, Dodge DE, Relman DA. Does blood of healthy subjects contain bacterial ribosomal DNA? J Clin Microbiol (2001) 39:1956–9. 10.1128/JCM.39.5.1956-1959.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.McLaughlin RW, Vali H, Lau PC, Palfree RGE, De Ciccio A, Sirois M, et al. Are there naturally occurring pleomorphic bacteria in the blood of healthy humans? J Clin Microbiol (2002) 40:4771–5. 10.1128/JCM.40.12.4771-4775.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Moriyama K, Ando C, Tashiro K, Kuhara S, Okamura S, Nakano S, et al. Polymerase chain reaction detection of bacterial 16S rRNA gene in human blood. Microbiol Immunol (2008) 52:375–82. 10.1111/j.1348-0421.2008.00048.x [DOI] [PubMed] [Google Scholar]
  • 302.Amar J, Serino M, Lange C, Chabo C, Iacovoni J, Mondot S, et al. Involvement of tissue bacteria in the onset of diabetes in humans: evidence for a concept. Diabetologia (2011) 54:3055–61. 10.1007/s00125-011-2329-8 [DOI] [PubMed] [Google Scholar]
  • 303.Gaibani P, Mariconti M, Bua G, Bonora S, Sassera D, Landini MP, et al. Development of a broad-range 23S rDNA real-time PCR assay for the detection and quantification of pathogenic bacteria in human whole blood and plasma specimens. Biomed Res Int (2013) 2013:264651. 10.1155/2013/264651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Dinakaran V, Rathinavel A, Pushpanathan M, Sivakumar R, Gunasekaran P, Rajendhran J. Elevated levels of circulating DNA in cardiovascular disease patients: metagenomic profiling of microbiome in the circulation. PLoS One (2014) 9:e105221. 10.1371/journal.pone.0105221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Sato J, Kanazawa A, Ikeda F, Yoshihara T, Goto H, Abe H, et al. Gut dysbiosis and detection of “live gut bacteria” in blood of Japanese patients with type 2 diabetes. Diabetes Care (2014) 37:2343–50. 10.2337/dc13-2817 [DOI] [PubMed] [Google Scholar]
  • 306.Damgaard C, Magnussen K, Enevold C, Nilsson M, Tolker-Nielsen T, Holmstrup P, et al. Viable bacteria associated with red blood cells and plasma in freshly drawn blood donations. PLoS One (2015) 10:e0120826. 10.1371/journal.pone.0120826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Gyarmati P, Kjellander C, Aust C, Kalin M, Öhrmalm L, Giske CG. Bacterial landscape of bloodstream infections in neutropenic patients via high throughput sequencing. PLoS One (2015) 10:e0135756. 10.1371/journal.pone.0135756 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Gyarmati P, Kjellander C, Aust C, Song Y, Öhrmalm L, Giske CG. Metagenomic analysis of bloodstream infections in patients with acute leukemia and therapy-induced neutropenia. Sci Rep (2016) 6:23532. 10.1038/srep23532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Païssé S, Valle C, Servant F, Courtney M, Burcelin R, Amar J, et al. Comprehensive description of blood microbiome from healthy donors assessed by 16S targeted metagenomic sequencing. Transfusion (2016) 56:1138–47. 10.1111/trf.13477 [DOI] [PubMed] [Google Scholar]
  • 310.Domingue GJ, Schlegel JU. Novel bacterial structures in human blood: cultural isolation. Infect Immun (1977) 15:621–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Belstrøm D, Holmstrup P, Damgaard C, Borch TS, Skjødt MO, Bendtzen K, et al. The atherogenic bacterium Porphyromonas gingivalis evades circulating phagocytes by adhering to erythrocytes. Infect Immun (2011) 79:1559–65. 10.1128/IAI.01036-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Billings F. Focal Infection. New York: Appleton; (1915). [Google Scholar]
  • 313.Price WA. Dental Infections Oral and Systemic, Being a Contribution to the Pathology of Dental Infections, Focal Infections and the Degenerative Diseases, Parts I and II. Cleveland: Penton Press; (1923). [Google Scholar]
  • 314.Miklossy J, Kis A, Radenovic A, Miller L, Forro L, Martins R, et al. Beta-amyloid deposition and Alzheimer’s type changes induced by Borrelia spirochetes. Neurobiol Aging (2006) 27:228–36. 10.1016/j.neurobiolaging.2005.01.018 [DOI] [PubMed] [Google Scholar]
  • 315.Miklossy J. Chronic inflammation and amyloidogenesis in Alzheimer’s disease – role of spirochetes. J Alzheimers Dis (2008) 13:381–91. [DOI] [PubMed] [Google Scholar]
  • 316.Woolard MD, Frelinger JA. Outsmarting the host: bacteria modulating the immune response. Immunol Res (2008) 41:188–202. 10.1007/s12026-008-8021-5 [DOI] [PubMed] [Google Scholar]
  • 317.Nicolson GL, Haier J. Role of chronic bacterial and viral infections in neurodegenerative, neurobehavioural, psychiatric, autoimmune and fatiguing illnesses: part 1. Br J Med Pract (2009) 2:20–8. [Google Scholar]
  • 318.Proal AD, Albert PJ, Marshall T. Autoimmune disease in the era of the metagenome. Autoimmun Rev (2009) 8:677–81. 10.1016/j.autrev.2009.02.016 [DOI] [PubMed] [Google Scholar]
  • 319.Nicolson GL, Haier J. Role of chronic bacterial and viral infections in neurodegenerative, neurobehavioural, psychiatric, autoimmune and fatiguing illnesses: part 2. Br J Med Pract (2010) 3:301–10. [Google Scholar]
  • 320.Miklossy J. Alzheimer’s disease – a neurospirochetosis. Analysis of the evidence following Koch’s and Hill’s criteria. J Neuroinflammation (2011) 8:90. 10.1186/1742-2094-8-90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Proal AD, Albert PJ, Blaney GP, Lindseth IA, Benediktsson C, Marshall TG. Immunostimulation in the era of the metagenome. Cell Mol Immunol (2011) 8:213–25. 10.1038/cmi.2010.77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Brecher ME, Hay SN. Bacterial contamination of blood components. Clin Microbiol Rev (2005) 18:195–204. 10.1128/CMR.18.1.195-204.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Halliwell B, Gutteridge JMC. Free Radicals in Biology and Medicine. 4th ed Oxford: Oxford University Press; (2006). [Google Scholar]
  • 324.Kaeberlein T, Lewis K, Epstein SS. Isolating “uncultivable” microorganisms in pure culture in a simulated natural environment. Science (2002) 296:1127–9. 10.1126/science.1070633 [DOI] [PubMed] [Google Scholar]
  • 325.Bollmann A, Lewis K, Epstein SS. Incubation of environmental samples in a diffusion chamber increases the diversity of recovered isolates. Appl Environ Microbiol (2007) 73:6386–90. 10.1128/AEM.01309-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.D’Onofrio A, Crawford JM, Stewart EJ, Witt K, Gavrish E, Epstein S, et al. Siderophores from neighboring organisms promote the growth of uncultured bacteria. Chem Biol (2010) 17:254–64. 10.1016/j.chembiol.2010.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Lewis K, Epstein S, D’Onofrio A, Ling LL. Uncultured microorganisms as a source of secondary metabolites. J Antibiot (Tokyo) (2010) 63:468–76. 10.1038/ja.2010.87 [DOI] [PubMed] [Google Scholar]
  • 328.Ling LL, Schneider T, Peoples AJ, Spoering AL, Engels I, Conlon BP, et al. A new antibiotic kills pathogens without detectable resistance. Nature (2015) 517:455–9. 10.1038/nature14098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Deschner J, Eick S, Damanaki A, Nokhbehsaim M. The role of adipokines in periodontal infection and healing. Mol Oral Microbiol (2014) 29:258–69. 10.1111/omi.12070 [DOI] [PubMed] [Google Scholar]
  • 330.Herrera JA, Chaudhuri G, López-Jaramillo P. Is infection a major risk factor for preeclampsia? Med Hypotheses (2001) 57:393–7. 10.1054/mehy.2001.1378 [DOI] [PubMed] [Google Scholar]
  • 331.López-Jaramillo P, Casas JP, Serrano N. Preeclampsia: from epidemiological observations to molecular mechanisms. Braz J Med Biol Res (2001) 34:1227–35. 10.1590/S0100-879X2001001000001 [DOI] [PubMed] [Google Scholar]
  • 332.Von Dadelszen P, Magee LA. Could an infectious trigger explain the differential maternal response to the shared placental pathology of preeclampsia and normotensive intrauterine growth restriction? Acta Obstet Gynecol Scand (2002) 81:642–8. 10.1034/j.1600-0412.2002.810710.x [DOI] [PubMed] [Google Scholar]
  • 333.Marshall B. Helicobacter pylori: 20 years on. Clin Med (2002) 2:147–52. 10.1007/s102380200021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Suzuki H, Marshall BJ, Hibi T. Overview: Helicobacter pylori and extragastric disease. Int J Hematol (2006) 84:291–300. 10.1532/IJH97.06180 [DOI] [PubMed] [Google Scholar]
  • 335.Prelipcean CC, Mihai C, Gogalniceanu P, Mitrica D, Drug VL, Stanciu C. Extragastric manifestations of Helicobacter pylori infection. Rev Med Chir Soc Med Nat Iasi (2007) 111:575–83. [PubMed] [Google Scholar]
  • 336.Pellicano R, Franceschi F, Saracco G, Fagoonee S, Roccarina D, Gasbarrini A. Helicobacters and extragastric diseases. Helicobacter (2009) 14(Suppl 1):58–68. 10.1111/j.1523-5378.2009.00699.x [DOI] [PubMed] [Google Scholar]
  • 337.Figura N, Franceschi F, Santucci A, Bernardini G, Gasbarrini G, Gasbarrini A. Extragastric manifestations of Helicobacter pylori infection. Helicobacter (2010) 15(Suppl 1):60–8. 10.1111/j.1523-5378.2010.00778.x [DOI] [PubMed] [Google Scholar]
  • 338.Banić M, Franceschi F, Babić Z, Gasbarrini A. Extragastric manifestations of Helicobacter pylori infection. Helicobacter (2012) 17(Suppl 1):49–55. 10.1111/j.1523-5378.2012.00983.x [DOI] [PubMed] [Google Scholar]
  • 339.Roubaud Baudron C, Franceschi F, Salles N, Gasbarrini A. Extragastric diseases and Helicobacter pylori. Helicobacter (2013) 18(Suppl 1):44–51. 10.1111/hel.12077 [DOI] [PubMed] [Google Scholar]
  • 340.Franceschi F, Tortora A, Gasbarrini G, Gasbarrini A. Helicobacter pylori and extragastric diseases. Helicobacter (2014) 19(Suppl 1):52–8. 10.1111/hel.12159 [DOI] [PubMed] [Google Scholar]
  • 341.Franceschi F, Zuccala G, Roccarina D, Gasbarrini A. Clinical effects of Helicobacter pylori outside the stomach. Nat Rev Gastroenterol Hepatol (2014) 11:234–42. 10.1038/nrgastro.2013.243 [DOI] [PubMed] [Google Scholar]
  • 342.Testerman TL, Morris J. Beyond the stomach: an updated view of Helicobacter pylori pathogenesis, diagnosis, and treatment. World J Gastroenterol (2014) 20:12781–808. 10.3748/wjg.v20.i36.12781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Ponzetto A, Cardaropoli S, Piccoli E, Rolfo A, Gennero L, Kanduc D, et al. Pre-eclampsia is associated with Helicobacter pylori seropositivity in Italy. J Hypertens (2006) 24:2445–9. 10.1097/HJH.0b013e3280109e8c [DOI] [PubMed] [Google Scholar]
  • 344.Franceschi F, Di Simone N, D’Ippolito S, Castellani R, Di Nicuolo F, Gasbarrini G, et al. Antibodies anti-CagA cross-react with trophoblast cells: a risk factor for pre-eclampsia? Helicobacter (2012) 17:426–34. 10.1111/j.1523-5378.2012.00966.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Shimoda A, Ueda K, Nishiumi S, Murata-Kamiya N, Mukai SA, Sawada S, et al. Exosomes as nanocarriers for systemic delivery of the Helicobacter pylori virulence factor CagA. Sci Rep (2016) 6:18346. 10.1038/srep18346 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Raghupathy R. Cytokines as key players in the pathophysiology of preeclampsia. Med Princ Pract (2013) 22:8–19. 10.1159/000354200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Wurdinger T, Gatson NN, Balaj L, Kaur B, Breakefield XO, Pegtel DM. Extracellular vesicles and their convergence with viral pathways. Adv Virol (2012) 2012:767694. 10.1155/2012/767694 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.van Dongen HM, Masoumi N, Witwer KW, Pegtel DM. Extracellular vesicles exploit viral entry routes for cargo delivery. Microbiol Mol Biol Rev (2016) 80:369–86. 10.1128/MMBR.00063-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Xie F, Hu Y, Magee LA, Money DM, Patrick DM, Brunham RM, et al. Chlamydia pneumoniae infection in preeclampsia. Hypertens Pregnancy (2010) 29:468–77. 10.3109/10641950903242642 [DOI] [PubMed] [Google Scholar]
  • 350.Heine RP, Ness RB, Roberts JM. Seroprevalence of antibodies to Chlamydia pneumoniae in women with preeclampsia. Obstet Gynecol (2003) 101:221–6. 10.1016/S0029-7844(02)02591-7 [DOI] [PubMed] [Google Scholar]
  • 351.El-Shourbagy MAA, El-Refaie TA, Sayed KKA, Wahba KAH, El-Din ASS, Fathy MM. Impact of seroconversion and antichlamydial treatment on the rate of pre-eclampsia among Egyptian primigravidae. Int J Gynaecol Obstet (2011) 113:137–40. 10.1016/j.ijgo.2010.11.014 [DOI] [PubMed] [Google Scholar]
  • 352.Haggerty CL, Klebanoff MA, Panum I, Uldum SA, Bass DC, Olsen J, et al. Prenatal Chlamydia trachomatis infection increases the risk of preeclampsia. Pregnancy Hypertens (2013) 3:151–4. 10.1016/j.preghy.2013.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Haggerty CL, Panum I, Uldum SA, Bass DC, Olsen J, Darville T, et al. Chlamydia trachomatis infection may increase the risk of preeclampsia. Pregnancy Hypertens (2013) 3:28–33. 10.1016/j.preghy.2012.09.002 [DOI] [PubMed] [Google Scholar]
  • 354.Xie F, Hu Y, Magee LA, Money DM, Patrick DM, Krajden M, et al. An association between cytomegalovirus infection and pre-eclampsia: a case-control study and data synthesis. Acta Obstet Gynecol Scand (2010) 89:1162–7. 10.3109/00016349.2010.499449 [DOI] [PubMed] [Google Scholar]
  • 355.Xie F, Von Dadelszen P, Nadeau J. CMV infection, TLR-2 and -4 expression, and cytokine profiles in early-onset preeclampsia with HELLP syndrome. Am J Reprod Immunol (2014) 71:379–86. 10.1111/aji.12199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Panarelli M, Sattar N. Pre-eclampsia associated with Helicobacter pylori seropositivity. J Hypertens (2006) 24:2353–4. 10.1097/HJH.0b013e3280113638 [DOI] [PubMed] [Google Scholar]
  • 357.Mosbah A, Nabiel Y. Helicobacter pylori, Chlamydiae pneumoniae and trachomatis as probable etiological agents of preeclampsia. J Matern Fetal Neonatal Med (2016) 29:1607–12. 10.3109/14767058.2015.1056146 [DOI] [PubMed] [Google Scholar]
  • 358.Tersigni C, Franceschi F, Todros T, Cardaropoli S, Scambia G, Di Simone N. Insights into the role of Helicobacter pylori infection in preeclampsia: from the bench to the bedside. Front Immunol (2014) 5:484. 10.3389/fimmu.2014.00484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Üstün Y, Engin-Üstün Y, Ozkaplan E, Otlu B, Sait Tekerekoğlu M. Association of Helicobacter pylori infection with systemic inflammation in preeclampsia. J Matern Fetal Neonatal Med (2010) 23:311–4. 10.3109/14767050903121456 [DOI] [PubMed] [Google Scholar]
  • 360.Aksoy H, Ozkan A, Aktas F, Borekci B. Helicobacter pylori seropositivity and its relationship with serum malondialdehyde and lipid profile in preeclampsia. J Clin Lab Anal (2009) 23:219–22. 10.1002/jcla.20330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.Cardaropoli S, Rolfo A, Todros T. Helicobacter pylori and pregnancy-related disorders. World J Gastroenterol (2014) 20:654–64. 10.3748/wjg.v20.i3.654 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Pugliese A, Beltramo T, Todros T, Cardaropoli S, Ponzetto A. Interleukin-18 and gestosis: correlation with Helicobacter pylori seropositivity. Cell Biochem Funct (2008) 26:817–9. 10.1002/cbf.1503 [DOI] [PubMed] [Google Scholar]
  • 363.Cardaropoli S, Giuffrida D, Piazzese A, Todros T. Helicobacter pylori seropositivity and pregnancy-related diseases: a prospective cohort study. J Reprod Immunol (2015) 109:41–7. 10.1016/j.jri.2015.02.004 [DOI] [PubMed] [Google Scholar]
  • 364.Cardaropoli S, Rolfo A, Piazzese A, Ponzetto A, Todros T. Helicobacter pylori’s virulence and infection persistence define pre-eclampsia complicated by fetal growth retardation. World J Gastroenterol (2011) 17:5156–65. 10.3748/wjg.v17.i47.5156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.McDonnold M, Dunn H, Hester A, Pacheco LD, Hankins GD, Saade GR, et al. High risk human papillomavirus at entry to prenatal care and risk of preeclampsia. Am J Obstet Gynecol (2014) 210(138):138.e1–5. 10.1016/j.ajog.2013.09.040 [DOI] [PubMed] [Google Scholar]
  • 366.Hill JA, Devoe LD, Bryans CI, Jr. Frequency of asymptomatic bacteriuria in preeclampsia. Obstet Gynecol (1986) 67:529–32. [PubMed] [Google Scholar]
  • 367.Hsu CD, Witter FR. Urogenital infection in preeclampsia. Int J Gynaecol Obstet (1995) 49:271–5. 10.1016/0020-7292(95)02373-K [DOI] [PubMed] [Google Scholar]
  • 368.Mittendorf R, Lain KY, Williams MA, Walker CK. Preeclampsia. A nested, case-control study of risk factors and their interactions. J Reprod Med (1996) 41:491–6. [PubMed] [Google Scholar]
  • 369.Easter SR, Cantonwine DE, Zera CA, Lim KH, Parry SI, Mcelrath TF. Urinary tract infection during pregnancy, angiogenic factor profiles, and risk of preeclampsia. Am J Obstet Gynecol (2016) 214:387.e1–7. 10.1016/j.ajog.2015.09.101 [DOI] [PubMed] [Google Scholar]
  • 370.Mazor-Dray E, Levy A, Schlaeffer F, Sheiner E. Maternal urinary tract infection: is it independently associated with adverse pregnancy outcome? J Matern Fetal Neonatal Med (2009) 22:124–8. 10.1080/14767050802488246 [DOI] [PubMed] [Google Scholar]
  • 371.Minassian C, Thomas SL, Williams DJ, Campbell O, Smeeth L. Acute maternal infection and risk of pre-eclampsia: a population-based case-control study. PLoS One (2013) 8:e73047. 10.1371/journal.pone.0073047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Rezavand N, Veisi F, Zangane M, Amini R, Almasi A. Association between asymptomatic bacteriuria and pre-eclampsia. Glob J Health Sci (2016) 8:235–9. 10.5539/gjhs.v8n7p235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 373.Karmon A, Sheiner E. The relationship between urinary tract infection during pregnancy and preeclampsia: causal, confounded or spurious? Arch Gynecol Obstet (2008) 277:479–81. 10.1007/s00404-008-0643-2 [DOI] [PubMed] [Google Scholar]
  • 374.Villar J, Carroli G, Wojdyla D, Abalos E, Giordano D, Ba’aqeel H, et al. Preeclampsia, gestational hypertension and intrauterine growth restriction, related or independent conditions? Am J Obstet Gynecol (2006) 194:921–31. 10.1016/j.ajog.2005.10.813 [DOI] [PubMed] [Google Scholar]
  • 375.Bánhidy F, Ács N, Puhó EH, Czeizel AE. Pregnancy complications and birth outcomes of pregnant women with urinary tract infections and related drug treatments. Scand J Infect Dis (2007) 39:390–7. 10.1080/00365540601087566 [DOI] [PubMed] [Google Scholar]
  • 376.López-Jaramillo P, Herrera JA, Arenas-Mantilla M, Jauregui IE, Mendoza MA. Subclinical infection as a cause of inflammation in preeclampsia. Am J Ther (2008) 15:373–6. 10.1097/MJT.0b013e318164c149 [DOI] [PubMed] [Google Scholar]
  • 377.Ide M, Papapanou PN. Epidemiology of association between maternal periodontal disease and adverse pregnancy outcomes – systematic review. J Periodontol (2013) 84:S181–94. 10.1902/jop.2013.134009 [DOI] [PubMed] [Google Scholar]
  • 378.Dunlop AL, Mulle JG, Ferranti EP, Edwards S, Dunn AB, Corwin EJ. Maternal microbiome and pregnancy outcomes that impact infant health: a review. Adv Neonatal Care (2015) 15:377–85. 10.1097/ANC.0000000000000218 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.Hlimi T. Association of anemia, pre-eclampsia and eclampsia with seasonality: a realist systematic review. Health Place (2015) 31:180–92. 10.1016/j.healthplace.2014.12.003 [DOI] [PubMed] [Google Scholar]
  • 380.Brabin BJ, Johnson PM. Placental malaria and pre-eclampsia through the looking glass backwards? J Reprod Immunol (2005) 65:1–15. 10.1016/j.jri.2004.09.006 [DOI] [PubMed] [Google Scholar]
  • 381.Anya SE. Seasonal variation in the risk and causes of maternal death in the Gambia: malaria appears to be an important factor. Am J Trop Med Hyg (2004) 70:510–3. [PubMed] [Google Scholar]
  • 382.Sartelet H, Rogier C, Milko-Sartelet I, Angel G, Michel G. Malaria associated pre-eclampsia in Senegal. Lancet (1996) 347:1121. 10.1016/S0140-6736(96)90321-9 [DOI] [PubMed] [Google Scholar]
  • 383.Romero R, Mazor M, Wu YK, Sirtori M, Oyarzun E, Mitchell MD, et al. Infection in the pathogenesis of preterm labor. Semin Perinatol (1988) 12:262–79. [PubMed] [Google Scholar]
  • 384.Toth M, Witkin SS, Ledger W, Thaler H. The role of infection in the etiology of preterm birth. Obstet Gynecol (1988) 71:723–6. [PubMed] [Google Scholar]
  • 385.Cassell GH, Waites KB, Watson HL, Crouse DT, Harasawa R. Ureaplasma urealyticum intrauterine infection: role in prematurity and disease in newborns. Clin Microbiol Rev (1993) 6:69–87. 10.1128/CMR.6.1.69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 386.McGregor JA, French JI, Jones W, Milligan K, Mckinney PJ, Patterson E, et al. Bacterial vaginosis is associated with prematurity and vaginal fluid mucinase and sialidase: results of a controlled trial of topical clindamycin cream. Am J Obstet Gynecol (1994) 170:1048–59; discussion 1059–60. 10.1016/S0002-9378(94)70098-2 [DOI] [PubMed] [Google Scholar]
  • 387.Goldenberg RL, Hauth JC, Andrews WW. Intrauterine infection and preterm delivery. N Engl J Med (2000) 342:1500–7. 10.1056/NEJM200005183422007 [DOI] [PubMed] [Google Scholar]
  • 388.Gonçalves LF, Chaiworapongsa T, Romero R. Intrauterine infection and prematurity. Ment Retard Dev Disabil Res Rev (2002) 8:3–13. 10.1002/mrdd.10008 [DOI] [PubMed] [Google Scholar]
  • 389.Gerber S, Vial Y, Hohlfeld P, Witkin SS. Detection of Ureaplasma urealyticum in second-trimester amniotic fluid by polymerase chain reaction correlates with subsequent preterm labor and delivery. J Infect Dis (2003) 187:518–21. 10.1086/368205 [DOI] [PubMed] [Google Scholar]
  • 390.Gardella C, Riley DE, Hitti J, Agnew K, Krieger JN, Eschenbach D. Identification and sequencing of bacterial rDNAs in culture-negative amniotic fluid from women in premature labor. Am J Perinatol (2004) 21:319–23. 10.1055/s-2004-831884 [DOI] [PubMed] [Google Scholar]
  • 391.Espinoza J, Erez O, Romero R. Preconceptional antibiotic treatment to prevent preterm birth in women with a previous preterm delivery. Am J Obstet Gynecol (2006) 194:630–7. 10.1016/j.ajog.2005.11.050 [DOI] [PubMed] [Google Scholar]
  • 392.Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes of preterm birth. Lancet (2008) 371:75–84. 10.1016/S0140-6736(08)60074-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.Check JH. A practical approach to the prevention of miscarriage. Part 4 – role of infection. Clin Exp Obstet Gynecol (2010) 37:252–5. [PubMed] [Google Scholar]
  • 394.Bastek JA, Gómez LM, Elovitz MA. The role of inflammation and infection in preterm birth. Clin Perinatol (2011) 38:385–406. 10.1016/j.clp.2011.06.003 [DOI] [PubMed] [Google Scholar]
  • 395.Johnson HL, Ghanem KG, Zenilman JM, Erbelding EJ. Sexually transmitted infections and adverse pregnancy outcomes among women attending inner city public sexually transmitted diseases clinics. Sex Transm Dis (2011) 38:167–71. 10.1097/OLQ.0b013e3181f2e85f [DOI] [PubMed] [Google Scholar]
  • 396.Rours GIJG, Duijts L, Moll HA, Arends LR, De Groot R, Jaddoe VW, et al. Chlamydia trachomatis infection during pregnancy associated with preterm delivery: a population-based prospective cohort study. Eur J Epidemiol (2011) 26:493–502. 10.1007/s10654-011-9586-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Jefferson KK. The bacterial etiology of preterm birth. Adv Appl Microbiol (2012) 80:1–22. 10.1016/B978-0-12-394381-1.00001-5 [DOI] [PubMed] [Google Scholar]
  • 398.Lee SYR, Leung CW. Histological chorioamnionitis – implication for bacterial colonization, laboratory markers of infection, and early onset sepsis in very-low-birth-weight neonates. J Matern Fetal Neonatal Med (2012) 25:364–8. 10.3109/14767058.2011.579208 [DOI] [PubMed] [Google Scholar]
  • 399.Subramaniam A, Abramovici A, Andrews WW, Tita AT. Antimicrobials for preterm birth prevention: an overview. Infect Dis Obstet Gynecol (2012) 2012:157159. 10.1155/2012/157159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400.Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med (2014) 6:237ra265. 10.1126/scitranslmed.3008599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Joergensen JS, Kjaer Weile LK, Lamont RF. The early use of appropriate prophylactic antibiotics in susceptible women for the prevention of preterm birth of infectious etiology. Expert Opin Pharmacother (2014) 15:2173–91. 10.1517/14656566.2014.950225 [DOI] [PubMed] [Google Scholar]
  • 402.Allen-Daniels MJ, Serrano MG, Pflugner LP, Fettweis JM, Prestosa MA, Koparde VN, et al. Identification of a gene in Mycoplasma hominis associated with preterm birth and microbial burden in intraamniotic infection. Am J Obstet Gynecol (2015) 212:779.e1–13. 10.1016/j.ajog.2015.01.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.de Andrade Ramos B, Kanninen TT, Sisti G, Witkin SS. Microorganisms in the female genital tract during pregnancy: tolerance versus pathogenesis. Am J Reprod Immunol (2015) 73:383–9. 10.1111/aji.12326 [DOI] [PubMed] [Google Scholar]
  • 404.Kacerovsky M, Vrbacky F, Kutova R, Pliskova L, Andrys C, Musilova I, et al. Cervical microbiota in women with preterm prelabor rupture of membranes. PLoS One (2015) 10:e0126884. 10.1371/journal.pone.0126884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 405.Lamont RF. Advances in the prevention of infection-related preterm birth. Front Immunol (2015) 6:566. 10.3389/fimmu.2015.00566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406.Ueno T, Niimi H, Yoneda N, Yoneda S, Mori M, Tabata H, et al. Eukaryote-made thermostable DNA polymerase enables rapid PCR-based detection of mycoplasma, ureaplasma and other bacteria in the amniotic fluid of preterm labor cases. PLoS One (2015) 10:e0129032. 10.1371/journal.pone.0129032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407.Frey HA, Klebanoff MA. The epidemiology, etiology, and costs of preterm birth. Semin Fetal Neonatal Med (2016) 21:68–73. 10.1016/j.siny.2015.12.011 [DOI] [PubMed] [Google Scholar]
  • 408.Nadeau HCG, Subramaniam A, Andrews WW. Infection and preterm birth. Semin Fetal Neonatal Med (2016) 21:100–5. 10.1016/j.siny.2015.12.008 [DOI] [PubMed] [Google Scholar]
  • 409.Vinturache AE, Gyamfi-Bannerman C, Hwang J, Mysorekar IU, Jacobsson B, Preterm Birth International Collaborative (Prebic) Maternal microbiome – a pathway to preterm birth. Semin Fetal Neonatal Med (2016) 21:94–9. 10.1016/j.siny.2016.02.004 [DOI] [PubMed] [Google Scholar]
  • 410.Yoneda S, Shiozaki A, Yoneda N, Ito M, Shima T, Fukuda K, et al. Antibiotic therapy increases the risk of preterm birth in preterm labor without intra-amniotic microbes, but may prolong the gestation period in preterm labor with microbes, evaluated by rapid and high-sensitive PCR system. Am J Reprod Immunol (2016) 75:440–50. 10.1111/aji.12484 [DOI] [PubMed] [Google Scholar]
  • 411.Oliver RS, Lamont RF. Infection and antibiotics in the aetiology, prediction and prevention of preterm birth. J Obstet Gynaecol (2013) 33:768–75. 10.3109/01443615.2013.842963 [DOI] [PubMed] [Google Scholar]
  • 412.Kacerovsky M, Lenco J, Musilova I, Tambor V, Lamont R, Torloni MR, et al. Proteomic biomarkers for spontaneous preterm birth: a systematic review of the literature. Reprod Sci (2014) 21:283–95. 10.1177/1933719113503415 [DOI] [PubMed] [Google Scholar]
  • 413.McClure EM, Goldenberg RL. Infection and stillbirth. Semin Fetal Neonatal Med (2009) 14:182–9. 10.1016/j.siny.2009.02.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 414.Menezes EV, Yakoob MY, Soomro T, Haws RA, Darmstadt GL, Bhutta ZA. Reducing stillbirths: prevention and management of medical disorders and infections during pregnancy. BMC Pregnancy Childbirth (2009) 9(Suppl 1):S4. 10.1186/1471-2393-9-S1-S4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.Nigro G, Mazzocco M, Mattia E, Di Renzo GC, Carta G, Anceschi MM. Role of the infections in recurrent spontaneous abortion. J Matern Fetal Neonatal Med (2011) 24:983–9. 10.3109/14767058.2010.547963 [DOI] [PubMed] [Google Scholar]
  • 416.Giakoumelou S, Wheelhouse N, Cuschieri K, Entrican G, Howie SEM, Horne AW. The role of infection in miscarriage. Hum Reprod Update (2015) 22:116–33. 10.1093/humupd/dmv041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.Toth A, Lesser ML, Brooks-Toth CW, Feiner C. Outcome of subsequent pregnancies following antibiotic therapy after primary or multiple spontaneous abortions. Surg Gynecol Obstet (1986) 163:243–50. [PubMed] [Google Scholar]
  • 418.Lauder AP, Roche AM, Sherrill-Mix S, Bailey A, Laughlin AL, Bittinger K, et al. Comparison of placenta samples with contamination controls does not provide evidence for a distinct placenta microbiota. Microbiome (2016) 4:29. 10.1186/s40168-016-0172-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419.Onderdonk AB, Delaney ML, Dubois AM, Allred EN, Leviton A, Extremely Low Gestational Age Newborns Study Investigators . Detection of bacteria in placental tissues obtained from extremely low gestational age neonates. Am J Obstet Gynecol (2008) 198(110):e111–7. 10.1016/j.ajog.2007.05.044 [DOI] [PubMed] [Google Scholar]
  • 420.Satokari R, Gronroos T, Laitinen K, Salminen S, Isolauri E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol (2009) 48:8–12. 10.1111/j.1472-765X.2008.02475.x [DOI] [PubMed] [Google Scholar]
  • 421.Stout MJ, Conlon B, Landeau M, Lee I, Bower C, Zhao Q, et al. Identification of intracellular bacteria in the basal plate of the human placenta in term and preterm gestations. Am J Obstet Gynecol (2013) 208(226):e221–7. 10.1016/j.ajog.2013.01.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 422.Cao B, Mysorekar IU. Intracellular bacteria in placental basal plate localize to extravillous trophoblasts. Placenta (2014) 35:139–42. 10.1016/j.placenta.2013.12.007 [DOI] [PubMed] [Google Scholar]
  • 423.Cao B, Stout MJ, Lee I, Mysorekar IU. Placental microbiome and its role in preterm birth. Neoreviews (2014) 15:e537–45. 10.1542/neo.15-12-e537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 424.Doyle RM, Alber DG, Jones HE, Harris K, Fitzgerald F, Peebles D, et al. Term and preterm labour are associated with distinct microbial community structures in placental membranes which are independent of mode of delivery. Placenta (2014) 35:1099–101. 10.1016/j.placenta.2014.10.007 [DOI] [PubMed] [Google Scholar]
  • 425.Mysorekar IU, Cao B. Microbiome in parturition and preterm birth. Semin Reprod Med (2014) 32:50–5. 10.1055/s-0033-1361830 [DOI] [PubMed] [Google Scholar]
  • 426.Abrahamsson TR, Wu RY, Jenmalm MC. Gut microbiota and allergy: the importance of the pregnancy period. Pediatr Res (2015) 77:214–9. 10.1038/pr.2014.165 [DOI] [PubMed] [Google Scholar]
  • 427.Antony KM, Ma J, Mitchell KB, Racusin DA, Versalovic J, Aagaard K. The preterm placental microbiome varies in association with excess maternal gestational weight gain. Am J Obstet Gynecol (2015) 212: 653.e651–616. 10.1016/j.ajog.2014.12.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 428.Garmi G, Okopnik M, Keness Y, Zafran N, Berkowitz E, Salim R. Correlation between clinical, placental histology and microbiological findings in spontaneous preterm births. Fetal Diagn Ther (2015) 40:141–9. 10.1159/000441518 [DOI] [PubMed] [Google Scholar]
  • 429.Zheng J, Xiao X, Zhang Q, Mao L, Yu M, Xu J. The placental microbiome varies in association with low birth weight in full-term neonates. Nutrients (2015) 7:6924–37. 10.3390/nu7085315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 430.Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep (2016) 6:23129. 10.1038/srep23129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 431.Kuperman AA, Koren O. Antibiotic use during pregnancy: how bad is it? BMC Med (2016) 14:91. 10.1186/s12916-016-0636-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432.Prince AL, Ma J, Kannan PS, Alvarez M, Gisslen T, Harris RA, et al. The placental microbiome is altered among subjects with spontaneous preterm birth with and without chorioamnionitis. Am J Obstet Gynecol (2016) 214:627.e1–16. 10.1016/j.ajog.2016.01.193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 433.Viniker DA. Hypothesis on the role of sub-clinical bacteria of the endometrium (bacteria endometrialis) in gynaecological and obstetric enigmas. Hum Reprod Update (1999) 5:373–85. 10.1093/humupd/5.4.373 [DOI] [PubMed] [Google Scholar]
  • 434.Svare JA, Schmidt H, Hansen BB, Lose G. Bacterial vaginosis in a cohort of Danish pregnant women: prevalence and relationship with preterm delivery, low birthweight and perinatal infections. BJOG (2006) 113:1419–25. 10.1111/j.1471-0528.2006.01087.x [DOI] [PubMed] [Google Scholar]
  • 435.Sheldon IM, Bromfield JJ. Innate immunity in the human endometrium and ovary. Am J Reprod Immunol (2011) 66(Suppl 1):63–71. 10.1111/j.1600-0897.2011.01034.x [DOI] [PubMed] [Google Scholar]
  • 436.Aagaard K, Riehle K, Ma J, Segata N, Mistretta TA, Coarfa C, et al. A metagenomic approach to characterization of the vaginal microbiome signature in pregnancy. PLoS One (2012) 7:e36466. 10.1371/journal.pone.0036466 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 437.Walther-António MR, Jeraldo P, Berg Miller ME, Yeoman CJ, Nelson KE, Wilson BA, et al. Pregnancy’s stronghold on the vaginal microbiome. PLoS One (2014) 9:e98514. 10.1371/journal.pone.0098514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 438.Huang YE, Wang Y, He Y, Ji Y, Wang LP, Sheng HF, et al. Homogeneity of the vaginal microbiome at the cervix, posterior fornix, and vaginal canal in pregnant Chinese women. Microb Ecol (2015) 69:407–14. 10.1007/s00248-014-0487-1 [DOI] [PubMed] [Google Scholar]
  • 439.Witkin SS. The vaginal microbiome, vaginal anti-microbial defence mechanisms and the clinical challenge of reducing infection-related preterm birth. BJOG (2015):213–8. 10.1111/1471-0528.13115 [DOI] [PubMed] [Google Scholar]
  • 440.Nelson DB, Rockwell LC, Prioleau MD, Goetzl L. The role of the bacterial microbiota on reproductive and pregnancy health. Anaerobe (2016) 42:67–73. 10.1016/j.anaerobe.2016.09.001 [DOI] [PubMed] [Google Scholar]
  • 441.Payne MS, Bayatibojakhi S. Exploring preterm birth as a polymicrobial disease: an overview of the uterine microbiome. Front Immunol (2014) 5:595. 10.3389/fimmu.2014.00595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 442.Verstraelen H, Vilchez-Vargas R, Desimpel F, Jauregui R, Vankeirsbilck N, Weyers S, et al. Characterisation of the human uterine microbiome in non-pregnant women through deep sequencing of the V1-2 region of the 16S rRNA gene. PeerJ (2016) 4:e1602. 10.7717/peerj.1602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 443.Dong Y, St Clair PJ, Ramzy I, Kagan-Hallet KS, Gibbs RS. A microbiologic and clinical study of placental inflammation at term. Obstet Gynecol (1987) 70:175–82. [PubMed] [Google Scholar]
  • 444.Bearfield C, Davenport ES, Sivapathasundaram V, Allaker RP. Possible association between amniotic fluid micro-organism infection and microflora in the mouth. BJOG (2002) 109:527–33. 10.1111/j.1471-0528.2002.01349.x [DOI] [PubMed] [Google Scholar]
  • 445.Combs CA, Gravett M, Garite TJ, Hickok DE, Lapidus J, Porreco R, et al. Amniotic fluid infection, inflammation, and colonization in preterm labor with intact membranes. Am J Obstet Gynecol (2014) 210:125.e1–15. 10.1016/j.ajog.2013.11.032 [DOI] [PubMed] [Google Scholar]
  • 446.Wassenaar TM, Panigrahi P. Is a foetus developing in a sterile environment? Lett Appl Microbiol (2014) 59:572–9. 10.1111/lam.12334 [DOI] [PubMed] [Google Scholar]
  • 447.Combs CA, Garite TJ, Lapidus JA, Lapointe JP, Gravett M, Rael J, et al. Detection of microbial invasion of the amniotic cavity by analysis of cervicovaginal proteins in women with preterm labor and intact membranes. Am J Obstet Gynecol (2015) 212:482.e481–e412. 10.1016/j.ajog.2015.02.007 [DOI] [PubMed] [Google Scholar]
  • 448.Koleva PT, Kim JS, Scott JA, Kozyrskyj AL. Microbial programming of health and disease starts during fetal life. Birth Defects Res C Embryo Today (2015) 105:265–77. 10.1002/bdrc.21117 [DOI] [PubMed] [Google Scholar]
  • 449.Pelzer ES, Allan JA, Cunningham K, Mengersen K, Allan JM, Launchbury T, et al. Microbial colonization of follicular fluid: alterations in cytokine expression and adverse assisted reproduction technology outcomes. Hum Reprod (2011) 26:1799–812. 10.1093/humrep/der108 [DOI] [PubMed] [Google Scholar]
  • 450.Pelzer ES, Allan JA, Waterhouse MA, Ross T, Beagley KW, Knox CL. Microorganisms within human follicular fluid: effects on IVF. PLoS One (2013) 8:e59062. 10.1371/journal.pone.0059062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 451.Barak S, Oettinger-Barak O, Machtei EE, Sprecher H, Ohel G. Evidence of periopathogenic microorganisms in placentas of women with preeclampsia. J Periodontol (2007) 78:670–6. 10.1902/jop.2007.060362 [DOI] [PubMed] [Google Scholar]
  • 452.McDonagh S, Maidji E, Ma W, Chang HT, Fisher S, Pereira L. Viral and bacterial pathogens at the maternal-fetal interface. J Infect Dis (2004) 190:826–34. 10.1086/422330 [DOI] [PubMed] [Google Scholar]
  • 453.Amarasekara R, Jayasekara RW, Senanayake H, Dissanayake VH. Microbiome of the placenta in pre-eclampsia supports the role of bacteria in the multifactorial cause of pre-eclampsia. J Obstet Gynaecol Res (2015) 41:662–9. 10.1111/jog.12619 [DOI] [PubMed] [Google Scholar]
  • 454.Muehlenbachs A, Mutabingwa TK, Edmonds S, Fried M, Duffy PE. Hypertension and maternal-fetal conflict during placental malaria. PLoS Med (2006) 3:e446. 10.1371/journal.pmed.0030446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 455.Duffy PE. Plasmodium in the placenta: parasites, parity, protection, prevention and possibly preeclampsia. Parasitology (2007) 134:1877–81. 10.1017/S0031182007000170 [DOI] [PubMed] [Google Scholar]
  • 456.Vanterpool SF, Been JV, Houben ML, Nikkels PG, De Krijger RR, Zimmermann LJ, et al. Porphyromonas gingivalis within placental villous mesenchyme and umbilical cord stroma is associated with adverse pregnancy outcome. PLoS One (2016) 11:e0146157. 10.1371/journal.pone.0146157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 457.Chaparro A, Blanlot C, Ramirez V, Sanz A, Quintero A, Inostroza C, et al. Porphyromonas gingivalis, Treponema denticola and toll-like receptor 2 are associated with hypertensive disorders in placental tissue: a case-control study. J Periodontal Res (2013) 48:802–9. 10.1111/jre.12074 [DOI] [PubMed] [Google Scholar]
  • 458.Ebringer A, Rashid T, Wilson C. Rheumatoid arthritis, proteus, anti-CCP antibodies and Karl Popper. Autoimmun Rev (2010) 9:216–23. 10.1016/j.autrev.2009.10.006 [DOI] [PubMed] [Google Scholar]
  • 459.Ebringer A. Rheumatoid Arthritis and Proteus. London: Springer; (2012). [Google Scholar]
  • 460.Ebringer A, Rashid T. Rheumatoid arthritis is caused by a Proteus urinary tract infection. APMIS (2014) 122:363–8. 10.1111/apm.12154 [DOI] [PubMed] [Google Scholar]
  • 461.Vatanen T, Kostic AD, D’Hennezel E, Siljander H, Franzosa EA, Yassour M, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell (2016) 165:842–53. 10.1016/j.cell.2016.04.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 462.Ferrier L, Mazelin L, Cenac N, Desreumaux P, Janin A, Emilie D, et al. Stress-induced disruption of colonic epithelial barrier: role of interferon-gamma and myosin light chain kinase in mice. Gastroenterology (2003) 125:795–804. 10.1016/S0016-5085(03)01057-6 [DOI] [PubMed] [Google Scholar]
  • 463.Honda K, Littman DR. The microbiome in infectious disease and inflammation. Annu Rev Immunol (2012) 30:759–95. 10.1146/annurev-immunol-020711-074937 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 464.Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends Microbiol (2013) 21:6–13. 10.1016/j.tim.2012.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 465.Amar J, Burcelin R, Ruidavets JB, Cani PD, Fauvel J, Alessi MC, et al. Energy intake is associated with endotoxemia in apparently healthy men. Am J Clin Nutr (2008) 87:1219–23. [DOI] [PubMed] [Google Scholar]
  • 466.Lassenius MI, Pietiläinen KH, Kaartinen K, Pussinen PJ, Syrjänen J, Forsblom C, et al. Bacterial endotoxin activity in human serum is associated with dyslipidemia, insulin resistance, obesity, and chronic inflammation. Diabetes Care (2011) 34:1809–15. 10.2337/dc10-2197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 467.Vors C, Pineau G, Drai J, Meugnier E, Pesenti S, Laville M, et al. Postprandial endotoxemia linked with chylomicrons and lipopolysaccharides handling in obese versus lean men: a lipid dose-effect trial. J Clin Endocrinol Metab (2015) 100:3427–35. 10.1210/JC.2015-2518 [DOI] [PubMed] [Google Scholar]
  • 468.Sawchuck DJ, Wittmann BK. Pre-eclampsia renamed and reframed: intra-abdominal hypertension in pregnancy. Med Hypotheses (2014) 83:619–32. 10.1016/j.mehy.2014.08.001 [DOI] [PubMed] [Google Scholar]
  • 469.Perez PF, Dore J, Leclerc M, Levenez F, Benyacoub J, Serrant P, et al. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics (2007) 119:e724–32. 10.1542/peds.2006-1649 [DOI] [PubMed] [Google Scholar]
  • 470.Gensollen T, Iyer SS, Kasper DL, Blumberg RS. How colonization by microbiota in early life shapes the immune system. Science (2016) 352:539–44. 10.1126/science.aad9378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 471.Newnham JP, Newnham IA, Ball CM, Wright M, Pennell CE, Swain J, et al. Treatment of periodontal disease during pregnancy: a randomized controlled trial. Obstet Gynecol (2009) 114:1239–48. 10.1097/AOG.0b013e3181c15b40 [DOI] [PubMed] [Google Scholar]
  • 472.Huang X, Wang J, Liu J, Hua L, Zhang D, Hu T, et al. Maternal periodontal disease and risk of preeclampsia: a meta-analysis. J Huazhong Univ Sci Technolog Med Sci (2014) 34:729–35. 10.1007/s11596-014-1343-8 [DOI] [PubMed] [Google Scholar]
  • 473.Gilbert GL, Garland SM, Fairley KF, Mcdowall DM. Bacteriuria due to ureaplasmas and other fastidious organisms during pregnancy: prevalence and significance. Pediatr Infect Dis (1986) 5:S239–43. 10.1097/00006454-198611010-00007 [DOI] [PubMed] [Google Scholar]
  • 474.Ha JE, Jun JK, Ko HJ, Paik DI, Bae KH. Association between periodontitis and preeclampsia in never-smokers: a prospective study. J Clin Periodontol (2014) 41:869–74. 10.1111/jcpe.12281 [DOI] [PubMed] [Google Scholar]
  • 475.Sgolastra F, Petrucci A, Severino M, Gatto R, Monaco A. Relationship between periodontitis and pre-eclampsia: a meta-analysis. PLoS One (2013) 8:e71387. 10.1371/journal.pone.0071387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 476.Boggess KA, Edelstein BL. Oral health in women during preconception and pregnancy: implications for birth outcomes and infant oral health. Matern Child Health J (2006) 10:S169–74. 10.1007/s10995-006-0095-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 477.Boggess KA, Berggren EK, Koskenoja V, Urlaub D, Lorenz C. Severe preeclampsia and maternal self-report of oral health, hygiene, and dental care. J Periodontol (2013) 84:143–51. 10.1902/jop.2012.120079 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 478.Armitage GC. Bi-directional relationship between pregnancy and periodontal disease. Periodontol 2000 (2013) 61:160–76. 10.1111/j.1600-0757.2011.00396.x [DOI] [PubMed] [Google Scholar]
  • 479.Moura da Silva G, Coutinho SB, Piscoya MDBV, Ximenes RAA, Jamelli SR. Periodontitis as a risk factor for preeclampsia. J Periodontol (2012) 83:1388–96. 10.1902/jop.2012.110256 [DOI] [PubMed] [Google Scholar]
  • 480.Jahromi BN, Adibi R, Adibi S, Salarian L. Periodontal disease as a risk factor for preeclampsia. Womens Health Bull (2014) 1:e18908. 10.17795/whb-18908 [DOI] [Google Scholar]
  • 481.Lachat MF, Solnik AL, Nana AD, Citron TL. Periodontal disease in pregnancy: review of the evidence and prevention strategies. J Perinat Neonatal Nurs (2011) 25:312–9. 10.1097/JPN.0b013e31821072e4 [DOI] [PubMed] [Google Scholar]
  • 482.Politano GT, Passini R, Nomura ML, Velloso L, Morari J, Couto E. Correlation between periodontal disease, inflammatory alterations and pre-eclampsia. J Periodontal Res (2011) 46:505–11. 10.1111/j.1600-0765.2011.01368.x [DOI] [PubMed] [Google Scholar]
  • 483.Nabet C, Lelong N, Colombier ML, Sixou M, Musset AM, Goffinet F, et al. Maternal periodontitis and the causes of preterm birth: the case-control Epipap study. J Clin Periodontol (2010) 37:37–45. 10.1111/j.1600-051X.2009.01503.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 484.Madianos PN, Bobetsis YA, Offenbacher S. Adverse pregnancy outcomes (APOs) and periodontal disease: pathogenic mechanisms. J Periodontol (2013) 84:S170–80. 10.1902/jop.2013.1340015 [DOI] [PubMed] [Google Scholar]
  • 485.Bobetsis YA, Barros SP, Offenbacher S. Exploring the relationship between periodontal disease and pregnancy complications. J Am Dent Assoc (2006) 137(Suppl):7S–13S. 10.14219/jada.archive.2006.0403 [DOI] [PubMed] [Google Scholar]
  • 486.Herrera JA, Parra B, Herrera E, Botero JE, Arce RM, Contreras A, et al. Periodontal disease severity is related to high levels of C-reactive protein in pre-eclampsia. J Hypertens (2007) 25:1459–64. 10.1097/HJH.0b013e3281139ea9 [DOI] [PubMed] [Google Scholar]
  • 487.Ruma M, Boggess K, Moss K, Jared H, Murtha A, Beck J, et al. Maternal periodontal disease, systemic inflammation, and risk for preeclampsia. Am J Obstet Gynecol (2008) 198:389.e1–5. 10.1016/j.ajog.2007.12.002 [DOI] [PubMed] [Google Scholar]
  • 488.Pralhad S, Thomas B, Kushtagi P. Periodontal disease and pregnancy hypertension: a clinical correlation. J Periodontol (2013) 84:1118–25. 10.1902/jop.2012.120264 [DOI] [PubMed] [Google Scholar]
  • 489.Zi MY, Longo PL, Bueno-Silva B, Mayer MP. Mechanisms involved in the association between periodontitis and complications in pregnancy. Front Public Health (2014) 2:290. 10.3389/fpubh.2014.00290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 490.Swati P, Thomas B, Vahab SA, Kapaettu S, Kushtagi P. Simultaneous detection of periodontal pathogens in subgingival plaque and placenta of women with hypertension in pregnancy. Arch Gynecol Obstet (2012) 285:613–9. 10.1007/s00404-011-2012-9 [DOI] [PubMed] [Google Scholar]
  • 491.Shub A, Swain JR, Newnham JP. Periodontal disease and adverse pregnancy outcomes. J Matern Fetal Neonatal Med (2006) 19:521–8. 10.1080/14767050600797749 [DOI] [PubMed] [Google Scholar]
  • 492.Xiong X, Buekens P, Fraser WD, Beck J, Offenbacher S. Periodontal disease and adverse pregnancy outcomes: a systematic review. BJOG (2006) 113:135–43. 10.1111/j.1471-0528.2005.00827.x [DOI] [PubMed] [Google Scholar]
  • 493.Ha JE, Oh KJ, Yang HJ, Jun JK, Jin BH, Paik DI, et al. Oral health behaviors, periodontal disease, and pathogens in preeclampsia: a case-control study in Korea. J Periodontol (2011) 82:1685–92. 10.1902/jop.2011.110035 [DOI] [PubMed] [Google Scholar]
  • 494.Komine-Aizawa S, Hirohata N, Aizawa S, Abiko Y, Hayakawa S. Porphyromonas gingivalis lipopolysaccharide inhibits trophoblast invasion in the presence of nicotine. Placenta (2015) 36:27–33. 10.1016/j.placenta.2014.10.015 [DOI] [PubMed] [Google Scholar]
  • 495.Contreras A, Herrera JA, Soto JE, Arce RM, Jaramillo A, Botero JE. Periodontitis is associated with preeclampsia in pregnant women. J Periodontol (2006) 77:182–8. 10.1902/jop.2006.050020 [DOI] [PubMed] [Google Scholar]
  • 496.Oettinger-Barak O, Barak S, Ohel G, Oettinger M, Kreutzer H, Peled M, et al. Severe pregnancy complication (preeclampsia) is associated with greater periodontal destruction. J Periodontol (2005) 76:134–7. 10.1902/jop.2005.76.1.134 [DOI] [PubMed] [Google Scholar]
  • 497.Foxman B. Recurring urinary tract infection: incidence and risk factors. Am J Public Health (1990) 80:331–3. 10.2105/AJPH.80.3.331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 498.Foxman B. Epidemiology of urinary tract infections: incidence, morbidity, and economic costs. Am J Med (2002) 113(Suppl 1A):5S–13S. 10.1016/S0002-9343(02)01054-9 [DOI] [PubMed] [Google Scholar]
  • 499.Marrs CF, Zhang L, Foxman B. Escherichia coli mediated urinary tract infections: are there distinct uropathogenic E. coli (UPEC) pathotypes? FEMS Microbiol Lett (2005) 252:183–90. 10.1016/j.femsle.2005.08.028 [DOI] [PubMed] [Google Scholar]
  • 500.Hannan TJ, Mysorekar IU, Hung CS, Isaacson-Schmid ML, Hultgren SJ. Early severe inflammatory responses to uropathogenic E. coli predispose to chronic and recurrent urinary tract infection. PLoS Pathog (2010) 6:e1001042. 10.1371/journal.ppat.1001042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 501.Hunstad DA, Justice SS. Intracellular lifestyles and immune evasion strategies of uropathogenic Escherichia coli. Annu Rev Microbiol (2010) 64:203–21. 10.1146/annurev.micro.112408.134258 [DOI] [PubMed] [Google Scholar]
  • 502.Ejrnaes K. Bacterial characteristics of importance for recurrent urinary tract infections caused by Escherichia coli. Dan Med Bull (2011) 58:B4187. [PubMed] [Google Scholar]
  • 503.Hannan TJ, Totsika M, Mansfield KJ, Moore KH, Schembri MA, Hultgren SJ. Host-pathogen checkpoints and population bottlenecks in persistent and intracellular uropathogenic Escherichia coli bladder infection. FEMS Microbiol Rev (2012) 36:616–48. 10.1111/j.1574-6976.2012.00339.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 504.Foxman B. Urinary tract infection syndromes: occurrence, recurrence, bacteriology, risk factors, and disease burden. Infect Dis Clin North Am (2014) 28:1–13. 10.1016/j.idc.2013.09.003 [DOI] [PubMed] [Google Scholar]
  • 505.Justice SS, Hung C, Theriot JA, Fletcher DA, Anderson GG, Footer MJ, et al. Differentiation and developmental pathways of uropathogenic Escherichia coli in urinary tract pathogenesis. Proc Natl Acad Sci U S A (2004) 101:1333–8. 10.1073/pnas.0308125100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 506.Bower JM, Eto DS, Mulvey MA. Covert operations of uropathogenic Escherichia coli within the urinary tract. Traffic (2005) 6:18–31. 10.1111/j.1600-0854.2004.00251.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 507.Rosen DA, Hooton TM, Stamm WE, Humphrey PA, Hultgren SJ. Detection of intracellular bacterial communities in human urinary tract infection. PLoS Med (2007) 4:e329. 10.1371/journal.pmed.0040329 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 508.Dhakal BK, Kulesus RR, Mulvey MA. Mechanisms and consequences of bladder cell invasion by uropathogenic Escherichia coli. Eur J Clin Invest (2008) 38(Suppl 2):2–11. 10.1111/j.1365-2362.2008.01986.x [DOI] [PubMed] [Google Scholar]
  • 509.Schwartz DJ, Chen SL, Hultgren SJ, Seed PC. Population dynamics and niche distribution of uropathogenic Escherichia coli during acute and chronic urinary tract infection. Infect Immun (2011) 79:4250–9. 10.1128/IAI.05339-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 510.Brauner A, Jacobson SH, Kuhn I. Urinary Escherichia coli causing recurrent infections – a prospective follow-up of biochemical phenotypes. Clin Nephrol (1992) 38:318–23. [PubMed] [Google Scholar]
  • 511.Russo TA, Stapleton A, Wenderoth S, Hooton TM, Stamm WE. Chromosomal restriction fragment length polymorphism analysis of Escherichia coli strains causing recurrent urinary tract infections in young women. J Infect Dis (1995) 172:440–5. 10.1093/infdis/172.2.440 [DOI] [PubMed] [Google Scholar]
  • 512.Ikäheimo R, Siitonen A, Heiskanen T, Kärkkäinen U, Kuosmanen P, Lipponen P, et al. Recurrence of urinary tract infection in a primary care setting: analysis of a 1-year follow-up of 179 women. Clin Infect Dis (1996) 22:91–9. 10.1093/clinids/22.1.91 [DOI] [PubMed] [Google Scholar]
  • 513.Rosen DA, Pinkner JS, Jones JM, Walker JN, Clegg S, Hultgren SJ. Utilization of an intracellular bacterial community pathway in Klebsiella pneumoniae urinary tract infection and the effects of FimK on type 1 pilus expression. Infect Immun (2008) 76:3337–45. 10.1128/IAI.00090-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 514.Luo Y, Ma Y, Zhao Q, Wang L, Guo L, Ye L, et al. Similarity and divergence of phylogenies, antimicrobial susceptibilities, and virulence factor profiles of Escherichia coli isolates causing recurrent urinary tract infections that persist or result from reinfection. J Clin Microbiol (2012) 50:4002–7. 10.1128/JCM.02086-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 515.Özlü T, Alçelik A, Çalışkan B, Dönmez ME. Preeclampsia: is it because of the asymptomatic, unrecognized renal scars caused by urinary tract infections in childhood that become symptomatic with pregnancy? Med Hypotheses (2012) 79:653–5. 10.1016/j.mehy.2012.08.002 [DOI] [PubMed] [Google Scholar]
  • 516.Kincaid-Smith P, Bullen M. Bacteriuria in pregnancy. Lancet (1965) 1:395–9. 10.1016/S0140-6736(65)90001-2 [DOI] [PubMed] [Google Scholar]
  • 517.Loh K, Sivalingam N. Urinary tract infections in pregnancy. Malays Fam Physician (2007) 2:54–7. [PMC free article] [PubMed] [Google Scholar]
  • 518.Macejko AM, Schaeffer AJ. Asymptomatic bacteriuria and symptomatic urinary tract infections during pregnancy. Urol Clin North Am (2007) 34:35–42. 10.1016/j.ucl.2006.10.010 [DOI] [PubMed] [Google Scholar]
  • 519.Schnarr J, Smaill F. Asymptomatic bacteriuria and symptomatic urinary tract infections in pregnancy. Eur J Clin Invest (2008) 38(Suppl 2):50–7. 10.1111/j.1365-2362.2008.02009.x [DOI] [PubMed] [Google Scholar]
  • 520.Imade PE, Izekor PE, Eghafona NO, Enabulele OI, Ophori E. Asymptomatic bacteriuria among pregnant women. N Am J Med Sci (2010) 2:263–6. 10.4297/najms.2010.2263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 521.Gilbert NM, O’Brien VP, Hultgren S, Macones G, Lewis WG, Lewis AL. Urinary tract infection as a preventable cause of pregnancy complications: opportunities, challenges, and a global call to action. Glob Adv Health Med (2013) 2:59–69. 10.7453/gahmj.2013.061 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 522.Glaser AP, Schaeffer AJ. Urinary tract infection and bacteriuria in pregnancy. Urol Clin North Am (2015) 42:547–60. 10.1016/j.ucl.2015.05.004 [DOI] [PubMed] [Google Scholar]
  • 523.Germain SJ, Sacks GP, Sooranna SR, Sargent IL, Redman CWG. Systemic inflammatory priming in normal pregnancy and preeclampsia: the role of circulating syncytiotrophoblast microparticles. J Immunol (2007) 178:5949–56. 10.4049/jimmunol.178.9.5949 [DOI] [PubMed] [Google Scholar]
  • 524.Redman CWG, Sargent IL. Circulating microparticles in normal pregnancy and pre-eclampsia. Placenta (2008) 29(Suppl A):S73–7. 10.1016/j.placenta.2007.11.016 [DOI] [PubMed] [Google Scholar]
  • 525.Hahn S, Rusterholz C, Hösli I, Lapaire O. Cell-free nucleic acids as potential markers for preeclampsia. Placenta (2011) 32(Suppl):S17–20. 10.1016/j.placenta.2010.06.018 [DOI] [PubMed] [Google Scholar]
  • 526.Rusterholz C, Messerli M, Hoesli I, Hahn S. Placental microparticles, DNA, and RNA in preeclampsia. Hypertens Pregnancy (2011) 30:364–75. 10.3109/10641951003599571 [DOI] [PubMed] [Google Scholar]
  • 527.Redman CWG, Tannetta DS, Dragovic RA, Gardiner C, Southcombe JH, Collett GP, et al. Review: does size matter? Placental debris and the pathophysiology of pre-eclampsia. Placenta (2012) 33(Suppl):S48–54. 10.1016/j.placenta.2011.12.006 [DOI] [PubMed] [Google Scholar]
  • 528.Goulopoulou S, Davidge ST. Molecular mechanisms of maternal vascular dysfunction in preeclampsia. Trends Mol Med (2015) 21:88–97. 10.1016/j.molmed.2014.11.009 [DOI] [PubMed] [Google Scholar]
  • 529.Niccolai E, Emmi G, Squatrito D, Silvestri E, Emmi L, Amedei A, et al. Microparticles: bridging the gap between autoimmunity and thrombosis. Semin Thromb Hemost (2015) 41:413–22. 10.1055/s-0035-1549850 [DOI] [PubMed] [Google Scholar]
  • 530.Souza ACP, Yuen PST, Star RA. Microparticles: markers and mediators of sepsis-induced microvascular dysfunction, immunosuppression, and AKI. Kidney Int (2015) 87:1100–8. 10.1038/ki.2015.26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 531.Mitchell MD, Peiris HN, Kobayashi M, Koh YQ, Duncombe G, Illanes SE, et al. Placental exosomes in normal and complicated pregnancy. Am J Obstet Gynecol (2015) 213:S173–81. 10.1016/j.ajog.2015.07.001 [DOI] [PubMed] [Google Scholar]
  • 532.de Jonge L, Bos HJ, Van Langen IM, De Jong-Van Den Berg LTW, Bakker MK. Antibiotics prescribed before, during and after pregnancy in the Netherlands: a drug utilization study. Pharmacoepidemiol Drug Saf (2014) 23:60–8. 10.1002/pds.3492 [DOI] [PubMed] [Google Scholar]
  • 533.Bookstaver PB, Bland CM, Griffin B, Stover KR, Eiland LS, McLaughlin M. A review of antibiotic use in pregnancy. Pharmacotherapy (2015) 35:1052–62. 10.1002/phar.1649 [DOI] [PubMed] [Google Scholar]
  • 534.Palmsten K, Hernández-Díaz S, Chambers CD, Mogun H, Lai S, Gilmer TP, et al. The most commonly dispensed prescription medications among pregnant women enrolled in the U.S. Medicaid program. Obstet Gynecol (2015) 126:465–73. 10.1097/AOG.0000000000000982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 535.Mawson AR. Effects of antiretroviral therapy on occurrence of pre-eclampsia. Lancet (2003) 361:347–8. 10.1016/S0140-6736(03)12359-8 [DOI] [PubMed] [Google Scholar]
  • 536.Suy A, Martínez E, Coll O, Lonca M, Palacio M, De Lazzari E, et al. Increased risk of pre-eclampsia and fetal death in HIV-infected pregnant women receiving highly active antiretroviral therapy. AIDS (2006) 20:59–66. 10.1097/01.aids.0000198090.70325.bd [DOI] [PubMed] [Google Scholar]
  • 537.Wimalasundera RC, Larbalestier N, Smith JH, De Ruiter A, Mc GTSA, Hughes AD, et al. Pre-eclampsia, antiretroviral therapy, and immune reconstitution. Lancet (2002) 360:1152–4. 10.1016/S0140-6736(02)11195-0 [DOI] [PubMed] [Google Scholar]
  • 538.Hall DR. Is pre-eclampsia less common in patients with HIV/AIDS? J Reprod Immunol (2007) 76:75–7. 10.1016/j.jri.2007.04.005 [DOI] [PubMed] [Google Scholar]
  • 539.Adams JW, Watts DH, Phelps BR. A systematic review of the effect of HIV infection and antiretroviral therapy on the risk of pre-eclampsia. Int J Gynaecol Obstet (2016) 133:17–21. 10.1016/j.ijgo.2015.08.007 [DOI] [PubMed] [Google Scholar]
  • 540.Todros T, Verdiglione P, Oggè G, Paladini D, Vergani P, Cardaropoli S. Low incidence of hypertensive disorders of pregnancy in women treated with spiramycin for Toxoplasma infection. Br J Clin Pharmacol (2006) 61:336–40. 10.1111/j.1365-2125.2005.02572.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 541.Alvarado-Esquivel C, Vázquez-Alaníz F, Sandoval-Carrillo AA, Salas-Pacheco JM, Hernández-Tinoco J, Sanchez-Anguiano LF, et al. Lack of association between Toxoplasma gondii infection and hypertensive disorders in pregnancy: a case-control study in a Northern Mexican population. Parasit Vectors (2014) 7:167. 10.1186/1756-3305-7-167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 542.Hall D, Gebhardt S, Theron G, Grové D. Pre-eclampsia and gestational hypertension are less common in HIV infected women. Pregnancy Hypertens (2014) 4:91–6. 10.1016/j.preghy.2013.11.008 [DOI] [PubMed] [Google Scholar]
  • 543.McCarthy FP, Kingdom JC, Kenny LC, Walsh SK. Animal models of preeclampsia; uses and limitations. Placenta (2011) 32:413–9. 10.1016/j.placenta.2011.03.010 [DOI] [PubMed] [Google Scholar]
  • 544.Aubuchon M, Schulz LC, Schust DJ. Preeclampsia: animal models for a human cure. Proc Natl Acad Sci U S A (2011) 108:1197–8. 10.1073/pnas.1018164108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 545.Kumasawa K, Ikawa M, Kidoya H, Hasuwa H, Saito-Fujita T, Morioka Y, et al. Pravastatin induces placental growth factor (PGF) and ameliorates preeclampsia in a mouse model. Proc Natl Acad Sci U S A (2011) 108:1451–5. 10.1073/pnas.1011293108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 546.Faas MM, Schuiling GA, Baller JF, Visscher CA, Bakker WW. A new animal model for human preeclampsia: ultra-low-dose endotoxin infusion in pregnant rats. Am J Obstet Gynecol (1994) 171:158–64. 10.1016/0002-9378(94)90463-4 [DOI] [PubMed] [Google Scholar]
  • 547.Faas MM, Schuiling GA, Linton EA, Sargent IL, Redman CWG. Activation of peripheral leukocytes in rat pregnancy and experimental preeclampsia. Am J Obstet Gynecol (2000) 182:351–7. 10.1016/S0002-9378(00)70223-7 [DOI] [PubMed] [Google Scholar]
  • 548.Sakawi Y, Tarpey M, Chen YF, Calhoun DA, Connor MG, Chestnut DH, et al. Evaluation of low-dose endotoxin administration during pregnancy as a model of preeclampsia. Anesthesiology (2000) 93:1446–55. 10.1097/00000542-200012000-00017 [DOI] [PubMed] [Google Scholar]
  • 549.Lin F, Zeng P, Xu ZY, Ye DY, Yu XF, Wang N, et al. Treatment of lipoxin A4 and its analogue on low-dose endotoxin induced preeclampsia in rat and possible mechanisms. Reprod Toxicol (2012) 34:677–85. 10.1016/j.reprotox.2012.09.009 [DOI] [PubMed] [Google Scholar]
  • 550.Cotechini T, Komisarenko M, Sperou A, MacDonald-Goodfellow S, Adams MA, Graham CH. Inflammation in rat pregnancy inhibits spiral artery remodeling leading to fetal growth restriction and features of preeclampsia. J Exp Med (2014) 211:165–79. 10.1084/jem.20130295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 551.Xue PP, Zheng MM, Gong P, Lin CM, Zhou JJ, Li YJ, et al. Single administration of ultra-low-dose lipopolysaccharide in rat early pregnancy induces TLR4 activation in the placenta contributing to preeclampsia. PLoS One (2015) 10:e0124001. 10.1371/journal.pone.0124001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 552.Kalkunte S, Boij R, Norris W, Friedman J, Lai Z, Kurtis J, et al. Sera from preeclampsia patients elicit symptoms of human disease in mice and provide a basis for an in vitro predictive assay. Am J Pathol (2010) 177:2387–98. 10.2353/ajpath.2010.100475 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 553.Nymark M, Pussinen PJ, Tuomainen AM, Forsblom C, Groop PH, Lehto M, et al. Serum lipopolysaccharide activity is associated with the progression of kidney disease in finnish patients with type 1 diabetes. Diabetes Care (2009) 32:1689–93. 10.2337/dc09-0467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 554.Al-Attas OS, Al-Daghri NM, Al-Rubeaan K, Da Silva NF, Sabico SL, Kumar S, et al. Changes in endotoxin levels in T2DM subjects on anti-diabetic therapies. Cardiovasc Diabetol (2009) 8:20. 10.1186/1475-2840-8-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 555.Hawkesworth S, Moore SE, Fulford AJC, Barclay GR, Darboe AA, Mark H, et al. Evidence for metabolic endotoxemia in obese and diabetic Gambian women. Nutr Diabetes (2013) 3:e83. 10.1038/nutd.2013.24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 556.Zaman GS, Zaman F. Relationship between postprandial endotoxemia in nonobese postmenopausal women and diabetic nonobese postmenopausal women. J Nat Sci Biol Med (2015) 6:89–93. 10.4103/0976-9668.149098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 557.Rusterholz C, Hahn S, Holzgreve W. Role of placentally produced inflammatory and regulatory cytokines in pregnancy and the etiology of preeclampsia. Semin Immunopathol (2007) 29:151–62. 10.1007/s00281-007-0071-6 [DOI] [PubMed] [Google Scholar]
  • 558.Xie F, Turvey SE, Williams MA, Mor G, Von Dadelszen P. Toll-like receptor signaling and pre-eclampsia. Am J Reprod Immunol (2010) 63:7–16. 10.1111/j.1600-0897.2009.00745.x [DOI] [PubMed] [Google Scholar]
  • 559.Anton L, Brown AG, Parry S, Elovitz MA. Lipopolysaccharide induces cytokine production and decreases extravillous trophoblast invasion through a mitogen-activated protein kinase-mediated pathway: possible mechanisms of first trimester placental dysfunction. Hum Reprod (2012) 27:61–72. 10.1093/humrep/der362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 560.Ahn H, Park J, Gilman-Sachs A, Kwak-Kim J. Immunologic characteristics of preeclampsia, a comprehensive review. Am J Reprod Immunol (2011) 65:377–94. 10.1111/j.1600-0897.2010.00913.x [DOI] [PubMed] [Google Scholar]
  • 561.Pineda A, Verdin-Terán SL, Camacho A, Moreno-Fierros L. Expression of toll-like receptor TLR-2, TLR-3, TLR-4 and TLR-9 is increased in placentas from patients with preeclampsia. Arch Med Res (2011) 42:382–91. 10.1016/j.arcmed.2011.08.003 [DOI] [PubMed] [Google Scholar]
  • 562.Chatterjee P, Weaver LE, Doersch KM, Kopriva SE, Chiasson VL, Allen SJ, et al. Placental toll-like receptor 3 and toll-like receptor 7/8 activation contributes to preeclampsia in humans and mice. PLoS One (2012) 7:e41884. 10.1371/journal.pone.0041884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 563.Gallo PM, Rapsinski GJ, Wilson RP, Oppong GO, Sriram U, Goulian M, et al. Amyloid-DNA composites of bacterial biofilms stimulate autoimmunity. Immunity (2015) 42:1171–84. 10.1016/j.immuni.2015.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 564.Rapsinski GJ, Wynosky-Dolfi MA, Oppong GO, Tursi SA, Wilson RP, Brodsky IE, et al. Toll-like receptor 2 and NLRP3 cooperate to recognize a functional bacterial amyloid, curli. Infect Immun (2015) 83:693–701. 10.1128/IAI.02370-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 565.Spaulding CN, Dodson KW, Chapman MR, Hultgren SJ. Fueling the fire with fibers: bacterial amyloids promote inflammatory disorders. Cell Host Microbe (2015) 18:1–2. 10.1016/j.chom.2015.06.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 566.Wucherpfennig KW. Mechanisms for the induction of autoimmunity by infectious agents. J Clin Invest (2001) 108:1097–104. 10.1172/JCI14235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 567.Wucherpfennig KW. Structural basis of molecular mimicry. J Autoimmun (2001) 16:293–302. 10.1006/jaut.2000.0499 [DOI] [PubMed] [Google Scholar]
  • 568.Kohm AP, Fuller KG, Miller SD. Mimicking the way to autoimmunity: an evolving theory of sequence and structural homology. Trends Microbiol (2003) 11:101–5. 10.1016/S0966-842X(03)00006-4 [DOI] [PubMed] [Google Scholar]
  • 569.Morris JA, Broughton SJ, Wessels Q. Microbes, molecular mimicry and molecules of mood and motivation. Med Hypotheses (2016) 87:40–3. 10.1016/j.mehy.2015.12.011 [DOI] [PubMed] [Google Scholar]
  • 570.Jain D, Kaur KJ, Goel M, Salunke DM. Structural basis of functional mimicry between carbohydrate and peptide ligands of con A. Biochem Biophys Res Commun (2000) 272:843–9. 10.1006/bbrc.2000.2871 [DOI] [PubMed] [Google Scholar]
  • 571.Goel M, Krishnan L, Kaur S, Kaur KJ, Salunke DM. Plasticity within the antigen-combining site may manifest as molecular mimicry in the humoral immune response. J Immunol (2004) 173:7358–67. 10.4049/jimmunol.173.12.7358 [DOI] [PubMed] [Google Scholar]
  • 572.Uh A, Nicholson RC, Gonzalez GV, Simmons CF, Gombart A, Smith R, et al. Lipopolysaccharide stimulation of trophoblasts induces corticotropin-releasing hormone expression through MyD88. Am J Obstet Gynecol (2008) 199(317):e311–6. 10.1016/j.ajog.2008.06.091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 573.Chen Q, Viall C, Kang Y, Liu B, Stone P, Chamley L. Anti-phospholipid antibodies increase non-apoptotic trophoblast shedding: a contribution to the pathogenesis of pre-eclampsia in affected women? Placenta (2009) 30:767–73. 10.1016/j.placenta.2009.06.008 [DOI] [PubMed] [Google Scholar]
  • 574.Chen Q, Guo F, Hensby-Bennett S, Stone P, Chamley L. Antiphospholipid antibodies prolong the activation of endothelial cells induced by necrotic trophoblastic debris: implications for the pathogenesis of preeclampsia. Placenta (2012) 33:810–5. 10.1016/j.placenta.2012.07.019 [DOI] [PubMed] [Google Scholar]
  • 575.Pantham P, Rosario R, Chen Q, Print CG, Chamley LW. Transcriptomic analysis of placenta affected by antiphospholipid antibodies: following the TRAIL of trophoblast death. J Reprod Immunol (2012) 94:151–4. 10.1016/j.jri.2012.03.487 [DOI] [PubMed] [Google Scholar]
  • 576.Tong M, Viall CA, Chamley LW. Antiphospholipid antibodies and the placenta: a systematic review of their in vitro effects and modulation by treatment. Hum Reprod Update (2015) 21:97–118. 10.1093/humupd/dmu049 [DOI] [PubMed] [Google Scholar]
  • 577.Dechend R, Homuth V, Wallukat G, Kreuzer J, Park JK, Theuer J, et al. AT(1) receptor agonistic antibodies from preeclamptic patients cause vascular cells to express tissue factor. Circulation (2000) 101:2382–7. 10.1161/01.CIR.101.20.2382 [DOI] [PubMed] [Google Scholar]
  • 578.Roberts JM. Angiotensin-1 receptor autoantibodies: a role in the pathogenesis of preeclampsia? Circulation (2000) 101:2335–7. 10.1161/01.CIR.101.20.2335 [DOI] [PubMed] [Google Scholar]
  • 579.Wallukat G, Neichel D, Nissen E, Homuth V, Luft FC. Agonistic autoantibodies directed against the angiotensin II AT1 receptor in patients with preeclampsia. Can J Physiol Pharmacol (2003) 81:79–83. 10.1139/y02-160 [DOI] [PubMed] [Google Scholar]
  • 580.Dechend R, Muller DN, Wallukat G, Homuth V, Krause M, Dudenhausen J, et al. AT1 receptor agonistic antibodies, hypertension, and preeclampsia. Semin Nephrol (2004) 24:571–9. 10.1016/j.semnephrol.2004.07.006 [DOI] [PubMed] [Google Scholar]
  • 581.Dechend R, Homuth V, Wallukat G, Muller DN, Krause M, Dudenhausen J, et al. Agonistic antibodies directed at the angiotensin II, AT1 receptor in preeclampsia. J Soc Gynecol Investig (2006) 13:79–86. 10.1016/j.jsgi.2005.11.006 [DOI] [PubMed] [Google Scholar]
  • 582.Hubel CA, Wallukat G, Wolf M, Herse F, Rajakumar A, Roberts JM, et al. Agonistic angiotensin II type 1 receptor autoantibodies in postpartum women with a history of preeclampsia. Hypertension (2007) 49:612–7. 10.1161/01.HYP.0000256565.20983.d4 [DOI] [PubMed] [Google Scholar]
  • 583.Dechend R, Luft FC. Are we getting closer to a nobel prize for unraveling preeclampsia? Curr Cardiol Rep (2008) 10:440–7. 10.1007/s11886-008-0070-9 [DOI] [PubMed] [Google Scholar]
  • 584.Herse F, Staff AC, Hering L, Müller DN, Luft FC, Dechend R. AT1-receptor autoantibodies and uteroplacental RAS in pregnancy and pre-eclampsia. J Mol Med (Berl) (2008) 86:697–703. 10.1007/s00109-008-0332-4 [DOI] [PubMed] [Google Scholar]
  • 585.Zhou CC, Zhang Y, Irani RA, Zhang H, Mi T, Popek EJ, et al. Angiotensin receptor agonistic autoantibodies induce pre-eclampsia in pregnant mice. Nat Med (2008) 14:855–62. 10.1038/nm.1856 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 586.Lorquet S, Pequeux C, Munaut C, Foidart JM. Aetiology and physiopathology of preeclampsia and related forms. Acta Clin Belg (2010) 65:237–41. 10.1179/acb.2010.051 [DOI] [PubMed] [Google Scholar]
  • 587.Parrish MR, Murphy SR, Rutland S, Wallace K, Wenzel K, Wallukat G, et al. The effect of immune factors, tumor necrosis factor-alpha, and agonistic autoantibodies to the angiotensin II type I receptor on soluble fms-like tyrosine-1 and soluble endoglin production in response to hypertension during pregnancy. Am J Hypertens (2010) 23:911–6. 10.1038/ajh.2010.70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 588.Carbillon L. AT1-receptor autoantibody: a true causal factor of pre-eclampsia or only a marker of poor placentation? Am J Hypertens (2011) 24:375; author reply 376. 10.1038/ajh.2010.249 [DOI] [PubMed] [Google Scholar]
  • 589.Herse F, LaMarca B. Angiotensin II type 1 receptor autoantibody (AT1-AA)-mediated pregnancy hypertension. Am J Reprod Immunol (2013) 69:413–8. 10.1111/aji.12072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 590.Siddiqui AH, Irani RA, Zhang W, Wang W, Blackwell SC, Kellems RE, et al. Angiotensin receptor agonistic autoantibody-mediated soluble fms-like tyrosine kinase-1 induction contributes to impaired adrenal vasculature and decreased aldosterone production in preeclampsia. Hypertension (2013) 61:472–9. 10.1161/HYPERTENSIONAHA.111.00157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 591.Pietarinen I, Kivinen S, Ylostalo P, Makitalo R, Laakso L. Smooth muscle antibodies in pre-eclampsia of pregnancy. Gynecol Obstet Invest (1982) 13:142–9. 10.1159/000299502 [DOI] [PubMed] [Google Scholar]
  • 592.Alanen A. Serum IgE and smooth muscle antibodies in pre-eclampsia. Acta Obstet Gynecol Scand (1984) 63:581–2. 10.3109/00016348409155540 [DOI] [PubMed] [Google Scholar]
  • 593.Fliser D, Buchholz K, Haller H, Olmesartan EUTO, Pravastatin In I, Atherosclerosis I. Antiinflammatory effects of angiotensin II subtype 1 receptor blockade in hypertensive patients with microinflammation. Circulation (2004) 110:1103–7. 10.1161/01.CIR.0000140265.21608.8E [DOI] [PubMed] [Google Scholar]
  • 594.Dagenais NJ, Jamali F. Protective effects of angiotensin II interruption: evidence for antiinflammatory actions. Pharmacotherapy (2005) 25:1213–29. 10.1592/phco.2005.25.9.1213 [DOI] [PubMed] [Google Scholar]
  • 595.Platten M, Youssef S, Hur EM, Ho PP, Han MH, Lanz TV, et al. Blocking angiotensin-converting enzyme induces potent regulatory T cells and modulates TH1- and TH17-mediated autoimmunity. Proc Natl Acad Sci U S A (2009) 106:14948–53. 10.1073/pnas.0903958106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 596.Ma G, Li Y, Zhang J, Liu H, Hou D, Zhu L, et al. Association between the presence of autoantibodies against adrenoreceptors and severe pre-eclampsia: a pilot study. PLoS One (2013) 8:e57983. 10.1371/journal.pone.0057983 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 597.Li Y, Ma G, Zhang Z, Yue Y, Yuan Y, Wang Y, et al. Association of autoantibodies against the M2-muscarinic receptor with perinatal outcomes in women with severe preeclampsia. J Transl Med (2013) 11:285. 10.1186/1479-5876-11-285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 598.Cornelius DC, Lamarca B. TH17- and IL-17-mediated autoantibodies and placental oxidative stress play a role in the pathophysiology of pre-eclampsia. Minerva Ginecol (2014) 66:243–9. [PMC free article] [PubMed] [Google Scholar]
  • 599.Darmochwał-Kolarz D, Kludka-Sternik M, Tabarkiewicz J, Kolarz B, Rolinski J, Leszczynska-Gorzelak B, et al. The predominance of Th17 lymphocytes and decreased number and function of Treg cells in preeclampsia. J Reprod Immunol (2012) 93:75–81. 10.1016/j.jri.2012.01.006 [DOI] [PubMed] [Google Scholar]
  • 600.Wenzel K, Rajakumar A, Haase H, Geusens N, Hubner N, Schulz H, et al. Angiotensin II type 1 receptor antibodies and increased angiotensin II sensitivity in pregnant rats. Hypertension (2011) 58:77–84. 10.1161/HYPERTENSIONAHA.111.171348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 601.Stepan H, Wallukat G, Schultheiss HP, Faber R, Walther T. Is parvovirus B19 the cause for autoimmunity against the angiotensin II type receptor? J Reprod Immunol (2007) 73:130–4. 10.1016/j.jri.2006.08.084 [DOI] [PubMed] [Google Scholar]
  • 602.Irani RA, Zhang Y, Zhou CC, Blackwell SC, Hicks MJ, Ramin SM, et al. Autoantibody-mediated angiotensin receptor activation contributes to preeclampsia through tumor necrosis factor-alpha signaling. Hypertension (2010) 55:1246–53. 10.1161/HYPERTENSIONAHA.110.150540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 603.Irani RA, Xia Y. Renin angiotensin signaling in normal pregnancy and preeclampsia. Semin Nephrol (2011) 31:47–58. 10.1016/j.semnephrol.2010.10.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 604.Mistry HD, Kurlak LO, Broughton Pipkin F. The placental renin-angiotensin system and oxidative stress in pre-eclampsia. Placenta (2013) 34:182–6. 10.1016/j.placenta.2012.11.027 [DOI] [PubMed] [Google Scholar]
  • 605.Verdonk K, Visser W, Van Den Meiracker AH, Danser AH. The renin-angiotensin-aldosterone system in pre-eclampsia: the delicate balance between good and bad. Clin Sci (Lond) (2014) 126:537–44. 10.1042/CS20130455 [DOI] [PubMed] [Google Scholar]
  • 606.Diveu C, Mcgeachy MJ, Cua DJ. Cytokines that regulate autoimmunity. Curr Opin Immunol (2008) 20:663–8. 10.1016/j.coi.2008.09.003 [DOI] [PubMed] [Google Scholar]
  • 607.Dong C. Regulation and pro-inflammatory function of interleukin-17 family cytokines. Immunol Rev (2008) 226:80–6. 10.1111/j.1600-065X.2008.00709.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 608.Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med (2009) 361:888–98. 10.1056/NEJMra0707449 [DOI] [PubMed] [Google Scholar]
  • 609.Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat Rev Drug Discov (2012) 11:763–76. 10.1038/nrd3794 [DOI] [PubMed] [Google Scholar]
  • 610.Benedetti G, Miossec P. Interleukin 17 contributes to the chronicity of inflammatory diseases such as rheumatoid arthritis. Eur J Immunol (2014) 44:339–47. 10.1002/eji.201344184 [DOI] [PubMed] [Google Scholar]
  • 611.Aujla SJ, Dubin PJ, Kolls JK. Th17 cells and mucosal host defense. Semin Immunol (2007) 19:377–82. 10.1016/j.smim.2007.10.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 612.Ivanov II, Zhou L, Littman DR. Transcriptional regulation of Th17 cell differentiation. Semin Immunol (2007) 19:409–17. 10.1016/j.smim.2007.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 613.Stockinger B, Veldhoen M, Martin B. Th17 T cells: linking innate and adaptive immunity. Semin Immunol (2007) 19:353–61. 10.1016/j.smim.2007.10.008 [DOI] [PubMed] [Google Scholar]
  • 614.Annunziato F, Cosmi L, Liotta F, Maggi E, Romagnani S. The phenotype of human Th17 cells and their precursors, the cytokines that mediate their differentiation and the role of Th17 cells in inflammation. Int Immunol (2008) 20:1361–8. 10.1093/intimm/dxn106 [DOI] [PubMed] [Google Scholar]
  • 615.Ouyang W, Kolls JK, Zheng Y. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity (2008) 28:454–67. 10.1016/j.immuni.2008.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 616.Curtis MM, Way SS. Interleukin-17 in host defence against bacterial, mycobacterial and fungal pathogens. Immunology (2009) 126:177–85. 10.1111/j.1365-2567.2008.03017.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 617.Eyerich K, Pennino D, Scarponi C, Foerster S, Nasorri F, Behrendt H, et al. IL-17 in atopic eczema: linking allergen-specific adaptive and microbial-triggered innate immune response. J Allergy Clin Immunol (2009) 123:59–66.e4. 10.1016/j.jaci.2008.10.031 [DOI] [PubMed] [Google Scholar]
  • 618.Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal Immunol (2009) 2:403–11. 10.1038/mi.2009.100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 619.Cypowyj S, Picard C, Marodi L, Casanova JL, Puel A. Immunity to infection in IL-17-deficient mice and humans. Eur J Immunol (2012) 42:2246–54. 10.1002/eji.201242605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 620.Rubino SJ, Geddes K, Girardin SE. Innate IL-17 and IL-22 responses to enteric bacterial pathogens. Trends Immunol (2012) 33:112–8. 10.1016/j.it.2012.01.003 [DOI] [PubMed] [Google Scholar]
  • 621.Weber A, Zimmermann C, Kieseier BC, Hartung HP, Hofstetter HH. Bacteria and their cell wall components uniformly co-activate interleukin-17-producing thymocytes. Clin Exp Immunol (2014) 178:504–15. 10.1111/cei.12414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 622.Beringer A, Noack M, Miossec P. IL-17 in chronic inflammation: from discovery to targeting. Trends Mol Med (2016) 22:230–41. 10.1016/j.molmed.2016.01.001 [DOI] [PubMed] [Google Scholar]
  • 623.Kumar P, Monin L, Castillo P, Elsegeiny W, Horne W, Eddens T, et al. Intestinal interleukin-17 receptor signaling mediates reciprocal control of the gut microbiota and autoimmune inflammation. Immunity (2016) 44:659–71. 10.1016/j.immuni.2016.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 624.Saito S, Nakashima A, Ito M, Shima T. Clinical implication of recent advances in our understanding of IL-17 and reproductive immunology. Expert Rev Clin Immunol (2011) 7:649–57. 10.1586/eci.11.49 [DOI] [PubMed] [Google Scholar]
  • 625.Fu B, Tian Z, Wei H. TH17 cells in human recurrent pregnancy loss and pre-eclampsia. Cell Mol Immunol (2014) 11:564–70. 10.1038/cmi.2014.54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 626.Ozkan ZS, Simsek M, Ilhan F, Deveci D, Godekmerdan A, Sapmaz E. Plasma IL-17, IL-35, interferon-gamma, SOCS3 and TGF-beta levels in pregnant women with preeclampsia, and their relation with severity of disease. J Matern Fetal Neonatal Med (2014) 27:1513–7. 10.3109/14767058.2013.861415 [DOI] [PubMed] [Google Scholar]
  • 627.Santner-Nanan B, Peek MJ, Khanam R, Richarts L, Zhu E, Fazekas De St Groth B, et al. Systemic increase in the ratio between Foxp3+ and IL-17-producing CD4+ T cells in healthy pregnancy but not in preeclampsia. J Immunol (2009) 183:7023–30. 10.4049/jimmunol.0901154 [DOI] [PubMed] [Google Scholar]
  • 628.Jianjun Z, Yali H, Zhiqun W, Mingming Z, Xia Z. Imbalance of T-cell transcription factors contributes to the Th1 type immunity predominant in pre-eclampsia. Am J Reprod Immunol (2010) 63:38–45. 10.1111/j.1600-0897.2009.00763.x [DOI] [PubMed] [Google Scholar]
  • 629.Saito S. Th17 cells and regulatory T cells: new light on pathophysiology of preeclampsia. Immunol Cell Biol (2010) 88:615–7. 10.1038/icb.2010.68 [DOI] [PubMed] [Google Scholar]
  • 630.Toldi G, Rigó J, Stenczer B, Vásárhelyi B, Molvarec A. Increased prevalence of IL-17-producing peripheral blood lymphocytes in pre-eclampsia. Am J Reprod Immunol (2011) 66:223–9. 10.1111/j.1600-0897.2011.00987.x [DOI] [PubMed] [Google Scholar]
  • 631.Cornelius DC, Hogg JP, Scott J, Wallace K, Herse F, Moseley J, et al. Administration of interleukin-17 soluble receptor C suppresses TH17 cells, oxidative stress, and hypertension in response to placental ischemia during pregnancy. Hypertension (2013) 62:1068–73. 10.1161/HYPERTENSIONAHA.113.01514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 632.Laresgoiti-Servitje E. A leading role for the immune system in the pathophysiology of preeclampsia. J Leukoc Biol (2013) 94:247–57. 10.1189/jlb.1112603 [DOI] [PubMed] [Google Scholar]
  • 633.Perez-Sepulveda A, Torres MJ, Khoury M, Illanes SE. Innate immune system and preeclampsia. Front Immunol (2014) 5:244. 10.3389/fimmu.2014.00244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 634.Cao W, Wang X, Chen T, Zhu H, Xu W, Zhao S, et al. The expression of notch/notch ligand, IL-35, IL-17, and Th17/Treg in preeclampsia. Dis Markers (2015) 2015:316182. 10.1155/2015/316182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 635.Molvarec A, Czegle I, Szijártó J, Rigó J. Increased circulating interleukin-17 levels in preeclampsia. J Reprod Immunol (2015) 112:53–7. 10.1016/j.jri.2015.05.007 [DOI] [PubMed] [Google Scholar]
  • 636.Wang H, Guo M, Liu F, Wang J, Zhou Z, Ji J, et al. Role of IL-17 variants in preeclampsia in Chinese Han women. PLoS One (2015) 10:e0140118. 10.1371/journal.pone.0140118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 637.Vargas-Rojas MI, Solleiro-Villavicencio H, Soto-Vega E. Th1, Th2, Th17 and Treg levels in umbilical cord blood in preeclampsia. J Matern Fetal Neonatal Med (2016) 29:1642–5. 10.3109/14767058.2015.1057811 [DOI] [PubMed] [Google Scholar]
  • 638.Darmochwał-Kolarz D, Oleszczuk J. The critical role of Th17 cells, Treg cells and co-stimulatory molecules in the development of pre-eclampsia. Dev Period Med (2014) 18:141–7. [PubMed] [Google Scholar]
  • 639.Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med (2006) 355:1018–28. 10.1056/NEJMoa063842 [DOI] [PubMed] [Google Scholar]
  • 640.Wang H, Ma S. The cytokine storm and factors determining the sequence and severity of organ dysfunction in multiple organ dysfunction syndrome. Am J Emerg Med (2008) 26:711–5. 10.1016/j.ajem.2007.10.031 [DOI] [PubMed] [Google Scholar]
  • 641.Tscherne DM, García-Sastre A. Virulence determinants of pandemic influenza viruses. J Clin Invest (2011) 121:6–13. 10.1172/JCI44947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 642.D’Elia RV, Harrison K, Oyston PC, Lukaszewski RA, Clark GC. Targeting the “cytokine storm” for therapeutic benefit. Clin Vaccine Immunol (2013) 20:319–27. 10.1128/CVI.00636-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 643.Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA (2016) 315:801–10. 10.1001/jama.2016.0287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 644.Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, Finfer S, et al. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med (2012) 366:2055–64. 10.1056/NEJMoa1202290 [DOI] [PubMed] [Google Scholar]
  • 645.Lai PS, Matteau A, Iddriss A, Hawes JC, Ranieri V, Thompson BT. An updated meta-analysis to understand the variable efficacy of drotrecogin alfa (activated) in severe sepsis and septic shock. Minerva Anestesiol (2013) 79:33–43. [PMC free article] [PubMed] [Google Scholar]
  • 646.Vulcano M, Meiss RP, Isturiz MA. Deferoxamine reduces tissue injury and lethality in LPS-treated mice. Int J Immunopharmacol (2000) 22:635–44. 10.1016/S0192-0561(00)00026-6 [DOI] [PubMed] [Google Scholar]
  • 647.Messaris E, Antonakis PT, Memos N, Chatzigianni E, Leandros E, Konstadoulakis MM. Deferoxamine administration in septic animals: improved survival and altered apoptotic gene expression. Int Immunopharmacol (2004) 4:455–9. 10.1016/j.intimp.2004.01.012 [DOI] [PubMed] [Google Scholar]
  • 648.Islam S, Jarosch S, Zhou J, Del Carmen Parquet C, Toguri JT, Colp P, et al. Anti-inflammatory and anti-bacterial effects of iron chelation in experimental sepsis. J Surg Res (2016) 200:266–73. 10.1016/j.jss.2015.07.001 [DOI] [PubMed] [Google Scholar]
  • 649.Doron MW, Makhlouf RA, Katz VL, Lawson EE, Stiles AD. Increased incidence of sepsis at birth in neutropenic infants of mothers with preeclampsia. J Pediatr (1994) 125:452–8. 10.1016/S0022-3476(05)83294-9 [DOI] [PubMed] [Google Scholar]
  • 650.Kocherlakota P, La Gamma EF. Preliminary report: rhG-CSF may reduce the incidence of neonatal sepsis in prolonged preeclampsia-associated neutropenia. Pediatrics (1998) 102:1107–11. 10.1542/peds.102.5.1107 [DOI] [PubMed] [Google Scholar]
  • 651.Bhaumik S, Ghosh S, Haldar KK, Mitra PK, Manna B. Risk of early onset neonatal septicemia in babies born to mothers with pre-eclampsia. Indian Pediatr (2000) 37:775–9. [PubMed] [Google Scholar]
  • 652.Procianoy RS, Silveira RC, Mussi-Pinhata MM, Rugolo LMSS, Leone CR, Lopes JMD, et al. Sepsis and neutropenia in very low birth weight infants delivered of mothers with preeclampsia. J Pediatr (2010) 157:434–U118. 10.1016/j.jpeds.2010.02.066 [DOI] [PubMed] [Google Scholar]
  • 653.Namdev S, Bhat V, Adhisivam B, Zachariah B. Oxidative stress and antioxidant status among neonates born to mothers with pre-eclampsia and their early outcome. J Matern Fetal Neonatal Med (2014) 27:1481–4. 10.3109/14767058.2013.860521 [DOI] [PubMed] [Google Scholar]
  • 654.Kenny LC, Dunn WB, Ellis DI, Myers J, Baker PN, The Gopec Consortium et al. Novel biomarkers for pre-eclampsia detected using metabolomics and machine learning. Metabolomics (2005) 1:227–34. 10.1007/s11306-005-0003-1 [DOI] [Google Scholar]
  • 655.Kenny LC, Broadhurst D, Brown M, Dunn WB, Redman CWG, Kell DB, et al. Detection and identification of novel metabolomic biomarkers in preeclampsia. Reprod Sci (2008) 15:591–7. 10.1177/1933719108316908 [DOI] [PubMed] [Google Scholar]
  • 656.Odibo AO, Goetzinger KR, Odibo L, Cahill AG, Macones GA, Nelson DM, et al. First-trimester prediction of preeclampsia using metabolomic biomarkers: a discovery phase study. Prenat Diagn (2011) 31:990–4. 10.1002/pd.2822 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 657.Bahado-Singh RO, Akolekar R, Mandal R, Dong E, Xia J, Kruger M, et al. Metabolomics and first-trimester prediction of early-onset preeclampsia. J Matern Fetal Neonatal Med (2012) 25:1840–7. 10.3109/14767058.2012.680254 [DOI] [PubMed] [Google Scholar]
  • 658.Dunn WB, Brown M, Worton SA, Davies K, Jones RL, Kell DB, et al. The metabolome of human placental tissue: investigation of first trimester tissue and changes related to preeclampsia in late pregnancy. Metabolomics (2012) 8:579–97. 10.1007/s11306-011-0348-6 [DOI] [Google Scholar]
  • 659.Woodham PC, O’Connell T, Grimes J, Haeri S, Eichelberger K, Baker A, et al. Metabolomics to predict severe preeclampsia in early pregnancy. Am J Obstet Gynecol (2012) 206:S348–348. 10.1016/j.ajog.2011.10.809 [DOI] [Google Scholar]
  • 660.Kuc S, Koster MPH, Pennings JLA, Hankemeier T, Berger R, Harms AC, et al. Metabolomics profiling for identification of novel potential markers in early prediction of preeclampsia. PLoS One (2014) 9:e98540. 10.1371/journal.pone.0098540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 661.Koster MPH, Vreeken RJ, Harms AC, Dane AD, Kuc S, Schielen PCJI, et al. First-trimester serum acylcarnitine levels to predict preeclampsia: a metabolomics approach. Dis Markers (2015) 2015:857108. 10.1155/2015/857108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 662.Myers JE, Hart S, Armstrong S, Mires GJ, Beynon R, Gaskell SJ, et al. Evidence for multiple circulating factors in preeclampsia. Am J Obstet Gynecol (2007) 196(266):e261–6. 10.1016/j.ajog.2006.10.875 [DOI] [PubMed] [Google Scholar]
  • 663.Kolla V, Jenö P, Moes S, Lapaire O, Hoesli I, Hahn S. Quantitative proteomic (iTRAQ) analysis of 1st trimester maternal plasma samples in pregnancies at risk for preeclampsia. J Biomed Biotechnol (2012) 2012:305964. 10.1155/2012/305964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 664.Myers JE, Kenny LC, Mccowan LM, Chan EH, Dekker GA, Poston L, et al. Angiogenic factors combined with clinical risk factors to predict preterm pre-eclampsia in nulliparous women: a predictive test accuracy study. BJOG (2013) 120:1215–23. 10.1111/1471-0528.12195 [DOI] [PubMed] [Google Scholar]
  • 665.Myers JE, Tuytten R, Thomas G, Laroy W, Kas K, Vanpoucke G, et al. Integrated proteomics pipeline yields novel biomarkers for predicting preeclampsia. Hypertension (2013) 61:1281–8. 10.1161/HYPERTENSIONAHA.113.01168 [DOI] [PubMed] [Google Scholar]
  • 666.Polsani S, Phipps E, Jim B. Emerging new biomarkers of preeclampsia. Adv Chronic Kidney Dis (2013) 20:271–9. 10.1053/j.ackd.2013.01.001 [DOI] [PubMed] [Google Scholar]
  • 667.Hahn S, Lapaire O, Than NG. Biomarker development for presymptomatic molecular diagnosis of preeclampsia: feasible, useful or even unnecessary? Expert Rev Mol Diagn (2015) 15:617–29. 10.1586/14737159.2015.1025757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 668.Khan GH, Galazis N, Docheva N, Layfield R, Atiomo W. Overlap of proteomics biomarkers between women with pre-eclampsia and PCOS: a systematic review and biomarker database integration. Hum Reprod (2015) 30:133–48. 10.1093/humrep/deu268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 669.Paulus P, Jennewein C, Zacharowski K. Biomarkers of endothelial dysfunction: can they help us deciphering systemic inflammation and sepsis? Biomarkers (2011) 16(Suppl 1):S11–21. 10.3109/1354750X.2011.587893 [DOI] [PubMed] [Google Scholar]
  • 670.Netea MG, Joosten LAB, Latz E, Mills KHG, Natoli G, Stunnenberg HG, et al. Trained immunity: a program of innate immune memory in health and disease. Science (2016) 352:aaf1098. 10.1126/science.aaf1098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 671.Savvidou MD, Lees CC, Parra M, Hingorani AD, Nicolaides KH. Levels of C-reactive protein in pregnant women who subsequently develop pre-eclampsia. BJOG (2002) 109:297–301. 10.1111/j.1471-0528.2002.01130.x [DOI] [PubMed] [Google Scholar]
  • 672.Kashanian M, Aghbali F, Mahali N. Evaluation of the diagnostic value of the first-trimester maternal serum high-sensitivity C-reactive protein level for prediction of pre-eclampsia. J Obstet Gynaecol Res (2013) 39:1549–54. 10.1111/jog.12105 [DOI] [PubMed] [Google Scholar]
  • 673.Saito S, Shiozaki A, Nakashima A, Sakai M, Sasaki Y. The role of the immune system in preeclampsia. Mol Aspects Med (2007) 28:192–209. 10.1016/j.mam.2007.02.006 [DOI] [PubMed] [Google Scholar]
  • 674.Rinehart BK, Terrone DA, Lagoo-Deenadayalan S, Barber WH, Hale EA, Martin JN, Jr, et al. Expression of the placental cytokines tumor necrosis factor alpha, interleukin 1beta, and interleukin 10 is increased in preeclampsia. Am J Obstet Gynecol (1999) 181:915–20. 10.1016/S0002-9378(99)70325-X [DOI] [PubMed] [Google Scholar]
  • 675.Serin ÝS, Özçelik B, Bapbuð M, Kýlýç H, Okur D, Erez R. Predictive value of tumor necrosis factor alpha (TNF-alpha) in preeclampsia. Eur J Obstet Gynecol Reprod Biol (2002):143–5. 10.1016/S0301-2115(01)00484-5 [DOI] [PubMed] [Google Scholar]
  • 676.Todros T, Bontempo S, Piccoli E, Ietta F, Romagnoli R, Biolcati M, et al. Increased levels of macrophage migration inhibitory factor (MIF) in preeclampsia. Eur J Obstet Gynecol Reprod Biol (2005) 123:162–6. 10.1016/j.ejogrb.2005.03.014 [DOI] [PubMed] [Google Scholar]
  • 677.Mehr S, Doyle LW. Cytokines as markers of bacterial sepsis in newborn infants: a review. Pediatr Infect Dis J (2000) 19:879–87. 10.1097/00006454-200009000-00014 [DOI] [PubMed] [Google Scholar]
  • 678.Roger T, Glauser MP, Calandra T. Macrophage migration inhibitory factor (MIF) modulates innate immune responses induced by endotoxin and Gram-negative bacteria. J Endotoxin Res (2001) 7:456–60. 10.1179/096805101101533089 [DOI] [PubMed] [Google Scholar]
  • 679.Takala A, Nupponen I, Kylanpää-Bäck ML, Repo H. Markers of inflammation in sepsis. Ann Med (2002) 34:614–23. 10.1080/078538902321117841 [DOI] [PubMed] [Google Scholar]
  • 680.Briassoulis G, Galani A. Prognostic markers of pediatric meningococcal sepsis. Expert Rev Anti Infect Ther (2014) 12:1017–20. 10.1586/14787210.2014.945431 [DOI] [PubMed] [Google Scholar]
  • 681.Su H, Chang SS, Han CM, Wu KY, Li MC, Huang CY, et al. Inflammatory markers in cord blood or maternal serum for early detection of neonatal sepsis-a systemic review and meta-analysis. J Perinatol (2014) 34:268–74. 10.1038/jp.2013.186 [DOI] [PubMed] [Google Scholar]
  • 682.Kell DB, Knowles JD. The role of modeling in systems biology. In: Szallasi Z, Stelling J, Periwal V, editors. System Modeling in Cellular Biology: From Concepts to Nuts and Bolts. Cambridge: MIT Press; (2006). p. 3–18. [Google Scholar]
  • 683.Palsson BØ. Systems Biology: Properties of Reconstructed Networks. Cambridge: Cambridge University Press; (2006). [Google Scholar]
  • 684.Palsson BØ. Systems Biology: Constraint-Based Reconstruction and Analysis. Cambridge: Cambridge University Press; (2015). [Google Scholar]
  • 685.Hucka M, Finney A, Sauro HM, Bolouri H, Doyle JC, Kitano H, et al. The systems biology markup language (SBML): a medium for representation and exchange of biochemical network models. Bioinformatics (2003) 19:524–31. 10.1093/bioinformatics/btg015 [DOI] [PubMed] [Google Scholar]
  • 686.Funahashi A, Matsuoka Y, Jouraku A, Morohashi M, Kikuchi N, Kitano H. CellDesigner 3.5: a versatile modeling tool for biochemical networks. Proc IEEE (2008) 96:1254–65. 10.1109/JPROC.2008.925458 [DOI] [Google Scholar]
  • 687.Hoops S, Sahle S, Gauges R, Lee C, Pahle J, Simus N, et al. COPASI: a complex pathway simulator. Bioinformatics (2006) 22:3067–74. 10.1093/bioinformatics/btl485 [DOI] [PubMed] [Google Scholar]
  • 688.Smoot ME, Ono K, Ruscheinski J, Wang PL, Ideker T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics (2011) 27:431–2. 10.1093/bioinformatics/btq675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 689.Madazli R, Kuseyrioglu B, Uzun H, Uludag S, Ocak V. Prediction of preeclampsia with maternal mid-trimester placental growth factor, activin A, fibronectin and uterine artery Doppler velocimetry. Int J Gynaecol Obstet (2005) 89:251–7. 10.1016/j.ijgo.2005.02.008 [DOI] [PubMed] [Google Scholar]
  • 690.Diesch CH, Holzgreve W, Hahn S, Zhong XY. Comparison of activin A and cell-free fetal DNA levels in maternal plasma from patients at high risk for preeclampsia. Prenat Diagn (2006) 26:1267–70. 10.1002/pd.1606 [DOI] [PubMed] [Google Scholar]
  • 691.Reddy A, Suri S, Sargent IL, Redman CW, Muttukrishna S. Maternal circulating levels of activin A, inhibin A, sFlt-1 and endoglin at parturition in normal pregnancy and pre-eclampsia. PLoS One (2009) 4:e4453. 10.1371/journal.pone.0004453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 692.Phillips DJ, Jones KL, Scheerlinck JY, Hedger MP, De Kretser DM. Evidence for activin A and follistatin involvement in the systemic inflammatory response. Mol Cell Endocrinol (2001) 180:155–62. 10.1016/S0303-7207(01)00516-0 [DOI] [PubMed] [Google Scholar]
  • 693.Hodges R, Salvador L, D’Antona D, Georgiou HM, Wallace EM. Activin A as a marker of intrauterine infection in women with preterm prelabour rupture of membranes. J Perinatol (2010) 30:22–6. 10.1038/jp.2009.109 [DOI] [PubMed] [Google Scholar]
  • 694.Rosenberg VA, Buhimschi IA, Dulay AT, Abdel-Razeq SS, Oliver EA, Duzyj CM, et al. Modulation of amniotic fluid activin-A and inhibin-A in women with preterm premature rupture of the membranes and infection-induced preterm birth. Am J Reprod Immunol (2012) 67:122–31. 10.1111/j.1600-0897.2011.01074.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 695.Petrakou E, Fotopoulos S, Anagnostakou M, Anatolitou F, Samitas K, Semitekolou M, et al. Activin-A exerts a crucial anti-inflammatory role in neonatal infections. Pediatr Res (2013) 74:675–81. 10.1038/pr.2013.159 [DOI] [PubMed] [Google Scholar]
  • 696.Shu C, Liu Z, Cui L, Wei C, Wang S, Tang JJ, et al. Protein profiling of preeclampsia placental tissues. PLoS One (2014) 9:e112890. 10.1371/journal.pone.0112890 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 697.Lugli A, Forster Y, Haas P, Nocito A, Bucher C, Bissig H, et al. Calretinin expression in human normal and neoplastic tissues: a tissue microarray analysis on 5233 tissue samples. Hum Pathol (2003) 34:994–1000. 10.1053/S0046-8177(03)00339-3 [DOI] [PubMed] [Google Scholar]
  • 698.Rizzo A, Carratelli CR, De Filippis A, Bevilacqua N, Tufano MA, Buommino E. Transforming activities of Chlamydia pneumoniae in human mesothelial cells. Int Microbiol (2014) 17:185–93. 10.2436/20.1501.01.221 [DOI] [PubMed] [Google Scholar]
  • 699.Xu QL, Zhu M, Jin Y, Wang N, Xu HX, Quan LM, et al. The predictive value of the first-trimester maternal serum chemerin level for pre-eclampsia. Peptides (2014) 62:150–4. 10.1016/j.peptides.2014.10.002 [DOI] [PubMed] [Google Scholar]
  • 700.Stepan H, Philipp A, Roth I, Kralisch S, Jank A, Schaarschmidt W, et al. Serum levels of the adipokine chemerin are increased in preeclampsia during and 6 months after pregnancy. Regul Pept (2011) 168:69–72. 10.1016/j.regpep.2011.03.005 [DOI] [PubMed] [Google Scholar]
  • 701.AL-Refai AA. Evaluation of serum levels of the adipokines chemerin and resistin in preeclampsia. Life Sci J (2012) 9:5143–51. [Google Scholar]
  • 702.Duan DM, Niu JM, Lei Q, Lin XH, Chen X. Serum levels of the adipokine chemerin in preeclampsia. J Perinat Med (2012) 40:121–7. 10.1515/Jpm.2011.127 [DOI] [PubMed] [Google Scholar]
  • 703.Wang LQ, Yang TL, Ding YL, Zhong Y, Yu L, Peng M. Chemerin plays a protective role by regulating human umbilical vein endothelial cell-induced nitric oxide signaling in preeclampsia. Endocrine (2015) 48:299–308. 10.1007/s12020-014-0286-y [DOI] [PubMed] [Google Scholar]
  • 704.Broadhurst D, Kell DB. Statistical strategies for avoiding false discoveries in metabolomics and related experiments. Metabolomics (2006) 2:171–96. 10.1007/s11306-006-0037-z [DOI] [Google Scholar]
  • 705.Kukla M, Zwirska-Korczala K, Gabriel A, Waluga M, Warakomska I, Szczygiel B, et al. Chemerin, vaspin and insulin resistance in chronic hepatitis C. J Viral Hepat (2010) 17:661–7. 10.1111/j.1365-2893.2009.01224.x [DOI] [PubMed] [Google Scholar]
  • 706.Kukla M, Mazur W, Buldak RJ, Żwirska-Korczala K. Potential role of leptin, adiponectin and three novel adipokines-visfatin, chemerin and vaspin-in chronic hepatitis. Mol Med (2011) 17:1397–410. 10.2119/molmed.2010.00105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 707.Kulig P, Kantyka T, Zabel BA, Banas M, Chyra A, Stefanska A, et al. Regulation of chemerin chemoattractant and antibacterial activity by human cysteine cathepsins. J Immunol (2011) 187:1403–10. 10.4049/jimmunol.1002352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 708.Banas M, Zabieglo K, Kasetty G, Kapinska-Mrowiecka M, Borowczyk J, Drukala J, et al. Chemerin is an antimicrobial agent in human epidermis. PLoS One (2013) 8:e58709. 10.1371/journal.pone.0058709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 709.Zabel BA, Kwitniewski M, Banas M, Zabieglo K, Murzyn K, Cichy J. Chemerin regulation and role in host defense. Am J Clin Exp Immunol (2014) 3:1–19. [PMC free article] [PubMed] [Google Scholar]
  • 710.Banas M, Zegar A, Kwitniewski M, Zabieglo K, Marczynska J, Kapinska-Mrowiecka M, et al. The expression and regulation of chemerin in the epidermis. PLoS One (2015) 10:e0117830. 10.1371/journal.pone.0117830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 711.Horn P, Metzing UB, Steidl R, Romeike B, Rauchfuß F, Sponholz C, et al. Chemerin in peritoneal sepsis and its associations with glucose metabolism and prognosis: a translational cross-sectional study. Crit Care (2016) 20:39. 10.1186/s13054-016-12090-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 712.Cornelius DC. Copeptin: a new biomarker that is specific for preeclampsia? Hypertension (2014) 64:1189–91. 10.1161/HYPERTENSIONAHA.114.04255 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 713.Birdir C, Janssen K, Stanescu AD, Enekwe A, Kasimir-Bauer S, Gellhaus A, et al. Maternal serum copeptin, MR-proANP and procalcitonin levels at 11-13 weeks gestation in the prediction of preeclampsia. Arch Gynecol Obstet (2015) 292:1033–42. 10.1007/s00404-015-3745-7 [DOI] [PubMed] [Google Scholar]
  • 714.Jochberger S, Morgenthaler NG, Mayr VD, Luckner G, Wenzel V, Ulmer H, et al. Copeptin and arginine vasopressin concentrations in critically ill patients. J Clin Endocrinol Metab (2006) 91:4381–6. 10.1210/jc.2005-2830 [DOI] [PubMed] [Google Scholar]
  • 715.Morgenthaler NG, Struck J, Jochberger S, Dünser MW. Copeptin: clinical use of a new biomarker. Trends Endocrinol Metab (2008) 19:43–9. 10.1016/j.tem.2007.11.001 [DOI] [PubMed] [Google Scholar]
  • 716.Katan M, Christ-Crain M. The stress hormone copeptin: a new prognostic biomarker in acute illness. Swiss Med Wkly (2010) 140:w13101. 10.4414/smw.2010.13101 [DOI] [PubMed] [Google Scholar]
  • 717.Nickel CH, Bingisser R, Morgenthaler NG. The role of copeptin as a diagnostic and prognostic biomarker for risk stratification in the emergency department. BMC Med (2012) 10:7. 10.1186/1741-7015-10-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 718.Dobša L, Edozien KC. Copeptin and its potential role in diagnosis and prognosis of various diseases. Biochem Med (Zagreb) (2013) 23:172–90. 10.11613/BM.2013.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 719.Odermatt J, Bolliger R, Hersberger L, Ottiger M, Christ-Crain M, Briel M, et al. Copeptin predicts 10-year all-cause mortality in community patients: a 10-year prospective cohort study. Clin Chem Lab Med (2016) 54:1681–90. 10.1515/cclm-2016-0151 [DOI] [PubMed] [Google Scholar]
  • 720.Jochberger S, Luckner G, Mayr VD, Wenzel V, Morgenthaler NG, Friesenecker BE, et al. Course of vasopressin and copeptin plasma concentrations in a patient with severe septic shock. Anaesth Intensive Care (2006) 34:498–500. [DOI] [PubMed] [Google Scholar]
  • 721.Müller B, Morgenthaler N, Stolz D, Schuetz P, Muller C, Bingisser R, et al. Circulating levels of copeptin, a novel biomarker, in lower respiratory tract infections. Eur J Clin Invest (2007) 37:145–52. 10.1111/j.1365-2362.2007.01762.x [DOI] [PubMed] [Google Scholar]
  • 722.Jochberger S, Dörler J, Luckner G, Mayr VD, Wenzel V, Ulmer H, et al. The vasopressin and copeptin response to infection, severe sepsis, and septic shock. Crit Care Med (2009) 37:476–82. 10.1097/CCM.0b013e3181957532 [DOI] [PubMed] [Google Scholar]
  • 723.Krüger S, Ewig S, Kunde J, Hanschmann A, Marre R, Suttorp N, et al. C-terminal provasopressin (copeptin) in patients with community-acquired pneumonia – influence of antibiotic pre-treatment: results from the German competence network CAPNETZ. J Antimicrob Chemother (2009) 64:159–62. 10.1093/jac/dkp148 [DOI] [PubMed] [Google Scholar]
  • 724.Limper M, Goeijenbier M, Wagenaar JF, Gasem MH, Isbandrio B, Kunde J, et al. Copeptin as a predictor of disease severity and survival in leptospirosis. J Infect (2010) 61:92–4. 10.1016/j.jinf.2010.03.029 [DOI] [PubMed] [Google Scholar]
  • 725.Fluri F, Morgenthaler NG, Mueller B, Christ-Crain M, Katan M. Copeptin, procalcitonin and routine inflammatory markers-predictors of infection after stroke. PLoS One (2012) 7:e48309. 10.1371/journal.pone.0048309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 726.Kolditz M, Halank M, Schulte-Hubbert B, Bergmann S, Albrecht S, Höffken G. Copeptin predicts clinical deterioration and persistent instability in community-acquired pneumonia. Respir Med (2012) 106:1320–8. 10.1016/j.rmed.2012.06.008 [DOI] [PubMed] [Google Scholar]
  • 727.Du JM, Sang G, Jiang CM, He XJ, Han Y. Relationship between plasma copeptin levels and complications of community-acquired pneumonia in preschool children. Peptides (2013) 45:61–5. 10.1016/j.peptides.2013.04.015 [DOI] [PubMed] [Google Scholar]
  • 728.Masajtis-Zagajewska A, Kurnatowska I, Wajdlich M, Nowicki M. Utility of copeptin and standard inflammatory markers in the diagnostics of upper and lower urinary tract infections. BMC Urol (2015) 15:67. 10.1186/s12894-015-0061-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 729.Wrotek A, Jackowska T, Pawlik K. Sodium and copeptin levels in children with community acquired pneumonia. Adv Exp Med Biol (2015) 835:31–6. 10.1007/5584_2014_41 [DOI] [PubMed] [Google Scholar]
  • 730.Strevens H, Wide-Swensson D, Grubb A, Hansen A, Horn T, Ingemarsson I, et al. Serum cystatin C reflects glomerular endotheliosis in normal, hypertensive and pre-eclamptic pregnancies. BJOG (2003) 110:825–30. 10.1111/j.1471-0528.2003.02051.x [DOI] [PubMed] [Google Scholar]
  • 731.Yang X, Wang H, Wang Z, Dong M. Alteration and significance of serum cardiac troponin I and cystatin C in preeclampsia. Clin Chim Acta (2006) 374:168–9. 10.1016/j.cca.2006.07.006 [DOI] [PubMed] [Google Scholar]
  • 732.Kristensen K, Wide-Swensson D, Schmidt C, Blirup-Jensen S, Lindström V, Strevens H, et al. Cystatin C, beta-2-microglobulin and beta-trace protein in pre-eclampsia. Acta Obstet Gynecol Scand (2007) 86:921–6. 10.1080/00016340701318133 [DOI] [PubMed] [Google Scholar]
  • 733.Guo HX, Wang CH, Li ZQ, Gong SP, Zhou ZQ, Leng LZ, et al. The application of serum cystatin C in estimating the renal function in women with preeclampsia. Reprod Sci (2012) 19:712–7. 10.1177/1933719111431001 [DOI] [PubMed] [Google Scholar]
  • 734.Novakov Mikic A, Cabarkapa V, Nikolic A, Maric D, Brkic S, Mitic G, et al. Cystatin C in pre-eclampsia. J Matern Fetal Neonatal Med (2012) 25:961–5. 10.3109/14767058.2011.601366 [DOI] [PubMed] [Google Scholar]
  • 735.Xiao J, Niu J, Ye X, Yu Q, Gu Y. Combined biomarkers evaluation for diagnosing kidney injury in preeclampsia. Hypertens Pregnancy (2013) 32:439–49. 10.3109/10641955.2013.827203 [DOI] [PubMed] [Google Scholar]
  • 736.Yalcin S, Ulas T, Eren MA, Aydogan H, Camuzcuoglu A, Kucuk A, et al. Relationship between oxidative stress parameters and cystatin C levels in patients with severe preeclampsia. Medicina (Kaunas) (2013) 49:118–23. [PubMed] [Google Scholar]
  • 737.Odden MC, Scherzer R, Bacchetti P, Szczech LA, Sidney S, Grunfeld C, et al. Cystatin C level as a marker of kidney function in human immunodeficiency virus infection: the FRAM study. Arch Intern Med (2007) 167:2213–9. 10.1001/archinte.167.20.2213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 738.Randers E, Kornerup K, Erlandsen EJ, Hasling C, Danielsen H. Cystatin C levels in sera of patients with acute infectious diseases with high C-reactive protein levels. Scand J Clin Lab Invest (2001) 61:333–5. 10.1080/00365510118007 [DOI] [PubMed] [Google Scholar]
  • 739.Gupta SK, Kitch D, Tierney C, Melbourne K, Ha B, Mccomsey GA, et al. Markers of renal disease and function are associated with systemic inflammation in HIV infection. HIV Med (2015) 16:591–8. 10.1111/hiv.12268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 740.Walker JB, Nesheim ME. The molecular weights, mass distribution, chain composition, and structure of soluble fibrin degradation products released from a fibrin clot perfused with plasmin. J Biol Chem (1999) 274:5201–12. 10.1074/jbc.274.8.5201 [DOI] [PubMed] [Google Scholar]
  • 741.Bellart J, Gilabert R, Anglès A, Piera V, Miralles RM, Monasterio J, et al. Tissue factor levels and high ratio of fibrinopeptide A:d-dimer as a measure of endothelial procoagulant disorder in pre-eclampsia. Br J Obstet Gynaecol (1999) 106:594–7. 10.1111/j.1471-0528.1999.tb08330.x [DOI] [PubMed] [Google Scholar]
  • 742.Gulec UK, Ozgunen FT, Guzel AB, Buyukkurt S, Seydaoglu G, Urunsak IF, et al. An analysis of C-reactive protein, procalcitonin, and d-dimer in pre-eclamptic patients. Am J Reprod Immunol (2012) 68:331–7. 10.1111/j.1600-0897.2012.01171.x [DOI] [PubMed] [Google Scholar]
  • 743.Pinheiro MDB, Junqueira DRG, Coelho FF, Freitas LG, Carvalho MG, Gomes KB, et al. d-dimer in preeclampsia: systematic review and meta-analysis. Clin Chim Acta (2012) 414:166–70. 10.1016/j.cca.2012.08.003 [DOI] [PubMed] [Google Scholar]
  • 744.Bozkurt M, Yumru AE, Sahin L, Salman S. Troponin I and d-dimer levels in preeclampsia and eclampsia: prospective study. Clin Exp Obstet Gynecol (2015) 42:26–31. [PubMed] [Google Scholar]
  • 745.Rahman R, Begum K, Khondker L, Majumder NI, Nahar K, Sultana R, et al. Role of d-dimer in determining coagulability status in pre-eclamptic and normotensive pregnant women. Mymensingh Med J (2015) 24:115–20. [PubMed] [Google Scholar]
  • 746.Di Castelnuovo A, De Curtis A, Costanzo S, Persichillo M, Olivieri M, Zito F, et al. Association of d-dimer levels with all-cause mortality in a healthy adult population: findings from the MOLI-SANI study. Haematologica (2013) 98:1476–80. 10.3324/haematol.2012.083410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 747.Jennings I, Woods TAL, Kitchen DP, Kitchen S, Walker ID. Laboratory d-dimer measurement: improved agreement between methods through calibration. Thromb Haemost (2007) 98:1127–35. 10.1160/TH07-05-0377 [DOI] [PubMed] [Google Scholar]
  • 748.Khalafallah AA, Morse M, Al-Barzan AM, Adams M, Dennis A, Bates G, et al. d-dimer levels at different stages of pregnancy in Australian women: a single centre study using two different immunoturbidimetric assays. Thromb Res (2012) 130:e171–7. 10.1016/j.thromres.2012.05.022 [DOI] [PubMed] [Google Scholar]
  • 749.Rodelo JR, De La Rosa G, Valencia ML, Ospina S, Arango CM, Gómez CI, et al. d-dimer is a significant prognostic factor in patients with suspected infection and sepsis. Am J Emerg Med (2012) 30:1991–9. 10.1016/j.ajem.2012.04.033 [DOI] [PubMed] [Google Scholar]
  • 750.Khalafallah A, Jarvis C, Morse M, Albarzan AM, Stewart P, Bates G, et al. Evaluation of the innovance d-dimer assay for the diagnosis of disseminated intravascular coagulopathy in different clinical settings. Clin Appl Thromb Hemost (2014) 20:91–7. 10.1177/1076029612454936 [DOI] [PubMed] [Google Scholar]
  • 751.Hentschke MR, Lucas LS, Mistry HD, Pinheiro Da Costa BE, Poli-De-Figueiredo CE. Endocan-1 concentrations in maternal and fetal plasma and placentae in pre-eclampsia in the third trimester of pregnancy. Cytokine (2015) 74:152–6. 10.1016/j.cyto.2015.04.013 [DOI] [PubMed] [Google Scholar]
  • 752.Cakmak M, Yilmaz H, Bağlar E, Darcin T, Inan O, Aktas A, et al. Serum levels of endocan correlate with the presence and severity of pre-eclampsia. Clin Exp Hypertens (2016) 38:137–42. 10.3109/10641963.2015.1060993 [DOI] [PubMed] [Google Scholar]
  • 753.Mosevoll KA, Lindås R, Wendelbo Ø, Bruserud Ø, Reikvam H. Systemic levels of the endothelium-derived soluble adhesion molecules endocan and E-selectin in patients with suspected deep vein thrombosis. Springerplus (2014) 3:571. 10.1186/2193-1801-3-571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 754.Yuksel MA, Tuten A, Oncul M, Acikgoz AS, Temel Yuksel I, Toprak MS, et al. Serum endocan concentration in women with pre-eclampsia. Arch Gynecol Obstet (2015) 292:69–73. 10.1007/s00404-014-3605-x [DOI] [PubMed] [Google Scholar]
  • 755.Balta S, Mikhailidis DP, Demirkol S, Ozturk C, Celik T, Iyisoy A. Endocan: a novel inflammatory indicator in cardiovascular disease? Atherosclerosis (2015) 243:339–43. 10.1016/j.atherosclerosis.2015.09.030 [DOI] [PubMed] [Google Scholar]
  • 756.Pawlak K, Mysliwiec M, Pawlak D. Endocan – the new endothelial activation marker independently associated with soluble endothelial adhesion molecules in uraemic patients with cardiovascular disease. Clin Biochem (2015) 48:425–30. 10.1016/j.clinbiochem.2015.01.006 [DOI] [PubMed] [Google Scholar]
  • 757.Filep JG. Endocan or endothelial cell-specific molecule-1: a novel prognostic marker of sepsis? Crit Care Med (2006) 34:574–5. 10.1097/01.CCM.0000199054.13823.A7 [DOI] [PubMed] [Google Scholar]
  • 758.Scherpereel A, Depontieu F, Grigoriu B, Cavestri B, Tsicopoulos A, Gentina T, et al. Endocan, a new endothelial marker in human sepsis. Crit Care Med (2006) 34:532–7. 10.1097/01.CCM.0000198525.82124.74 [DOI] [PubMed] [Google Scholar]
  • 759.Mihajlovic DM, Lendak DF, Brkic SV, Draskovic BG, Mitic GP, Novakov Mikic AS, et al. Endocan is useful biomarker of survival and severity in sepsis. Microvasc Res (2014) 93:92–7. 10.1016/j.mvr.2014.04.004 [DOI] [PubMed] [Google Scholar]
  • 760.Pauly D, Hamed S, Behnes M, Lepiorz D, Lang S, Akin I, et al. Endothelial cell-specific molecule-1/endocan: diagnostic and prognostic value in patients suffering from severe sepsis and septic shock. J Crit Care (2016) 31:68–75. 10.1016/j.jcrc.2015.09.019 [DOI] [PubMed] [Google Scholar]
  • 761.Shovlin CL, Hughes JMB, Scott J, Seidman CE, Seidman JG. Characterization of endoglin and identification of novel mutations in hereditary hemorrhagic telangiectasia. Am J Hum Genet (1997) 61:68–79. 10.1086/513906 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 762.Shovlin CL. Hereditary haemorrhagic telangiectasia: pathophysiology, diagnosis and treatment. Blood Rev (2010) 24:203–19. 10.1016/j.blre.2010.07.001 [DOI] [PubMed] [Google Scholar]
  • 763.Gregory AL, Xu G, Sotov V, Letarte M. Review: the enigmatic role of endoglin in the placenta. Placenta (2014) 35(Suppl):S93–9. 10.1016/j.placenta.2013.10.020 [DOI] [PubMed] [Google Scholar]
  • 764.Bell MJ, Roberts JM, Founds SA, Jeyabalan A, Terhorst L, Conley YP. Variation in endoglin pathway genes is associated with preeclampsia: a case-control candidate gene association study. BMC Pregnancy Childbirth (2013) 13:82. 10.1186/1471-2393-13-82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 765.Luft FC. Soluble endoglin (sEng) joins the soluble fms-like tyrosine kinase (sFlt) receptor as a pre-eclampsia molecule. Nephrol Dial Transplant (2006) 21:3052–4. 10.1093/ndt/gfl439 [DOI] [PubMed] [Google Scholar]
  • 766.Venkatesha S, Toporsian M, Lam C, Hanai J, Mammoto T, Kim YM, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med (2006) 12:642–9. 10.1038/nm1429 [DOI] [PubMed] [Google Scholar]
  • 767.Govender N, Moodley J, Gathiram P, Naicker T. Soluble fms-like tyrosine kinase-1 in HIV infected pre-eclamptic South African Black women. Placenta (2014) 35:618–24. 10.1016/j.placenta.2014.04.013 [DOI] [PubMed] [Google Scholar]
  • 768.Pratt A, Da Silva Costa F, Borg AJ, Kalionis B, Keogh R, Murthi P. Placenta-derived angiogenic proteins and their contribution to the pathogenesis of preeclampsia. Angiogenesis (2015) 18:115–23. 10.1007/s10456-014-9452-3 [DOI] [PubMed] [Google Scholar]
  • 769.Kleinrouweler CE, Wiegerinck MMJ, Ris-Stalpers C, Bossuyt PMM, van der Post JAM, Von Dadelszen P, et al. Accuracy of circulating placental growth factor, vascular endothelial growth factor, soluble fms-like tyrosine kinase 1 and soluble endoglin in the prediction of pre-eclampsia: a systematic review and meta-analysis. BJOG (2012) 119:778–87. 10.1111/j.1471-0528.2012.03311.x [DOI] [PubMed] [Google Scholar]
  • 770.Muenzner P, Rohde M, Kneitz S, Hauck CR. CEACAM engagement by human pathogens enhances cell adhesion and counteracts bacteria-induced detachment of epithelial cells. J Cell Biol (2005) 170:825–36. 10.1083/jcb.200412151 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 771.Clemente M, Núñez O, Lorente R, Rincón D, Matilla A, Salcedo M, et al. Increased intrahepatic and circulating levels of endoglin, a TGF-beta 1 co-receptor, in patients with chronic hepatitis C virus infection: relationship to histological and serum markers of hepatic fibrosis. J Viral Hepat (2006) 13:625–32. 10.1111/j.1365-2893.2006.00733.x [DOI] [PubMed] [Google Scholar]
  • 772.Silver KL, Conroy AL, Leke RG, Leke RJ, Gwanmesia P, Molyneux ME, et al. Circulating soluble endoglin levels in pregnant women in Cameroon and Malawi – associations with placental malaria and fetal growth restriction. PLoS One (2011) 6:e24985. 10.1371/journal.pone.0024985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 773.Sasmito SD, Ulfiati A, Wardana A, Nugraheni F, Pradiptasari NF, Zulaifa Z, et al. Endoglin expression and the level of tgf-β are increased in the placental tissue and correlated with low fetal weight in malaria infected mice. J Trop Life Sci (2015) 5:1–7. 10.11594/jtls.05.01.01 [DOI] [Google Scholar]
  • 774.Jansen P, Mumme T, Randau T, Gravius S, Hermanns-Sachweh B. Endoglin (CD105) expression differentiates between aseptic loosening and periprosthetic joint infection after total joint arthroplasty. Springerplus (2014) 3:561. 10.1186/2193-1801-3-561 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 775.Valerio LG. Mammalian iron metabolism. Toxicol Mech Methods (2007) 17:497–517. 10.1080/15376510701556690 [DOI] [PubMed] [Google Scholar]
  • 776.Andrews NC. Forging a field: the golden age of iron biology. Blood (2008) 112:219–30. 10.1182/blood-2007-12-077388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 777.Hower V, Mendes P, Torti FM, Laubenbacher R, Akman S, Shulaev V, et al. A general map of iron metabolism and tissue-specific subnetworks. Mol Biosyst (2009) 5:422–43. 10.1039/b816714c [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 778.Chifman J, Kniss A, Neupane P, Williams I, Leung B, Deng Z, et al. The core control system of intracellular iron homeostasis: a mathematical model. J Theor Biol (2012) 300:91–9. 10.1016/j.jtbi.2012.01.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 779.Mitchell S, Mendes P. A computational model of liver iron metabolism. PLoS Comput Biol (2013) 9:e1003299. 10.1371/journal.pcbi.1003299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 780.Oliveira F, Rocha S, Fernandes R. Iron metabolism: from health to disease. J Clin Lab Anal (2014) 28:210–8. 10.1002/jcla.21668 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 781.Kell DB, Pretorius E. Interpreting raised serum ferritin levels. BMJ (2015) 351 10.1136/bmj.h3692 [DOI] [PubMed] [Google Scholar]
  • 782.Nielsen P, Günther U, Dürken M, Fischer R, Düllmann J. Serum ferritin iron in iron overload and liver damage: correlation to body iron stores and diagnostic relevance. J Lab Clin Med (2000) 135:413–8. 10.1067/mlc.2000.106456 [DOI] [PubMed] [Google Scholar]
  • 783.Watanabe K, Yamashita Y, Ohgawara K, Sekiguchi M, Satake N, Orino K, et al. Iron content of rat serum ferritin. J Vet Med Sci (2001) 63:587–9. 10.1292/Jvms.63.587 [DOI] [PubMed] [Google Scholar]
  • 784.Yamanishi H, Iyama S, Yamaguchi Y, Kanakura Y, Iwatani Y. Relation between iron content of serum ferritin and clinical status factors extracted by factor analysis in patients with hyperferritinemia. Clin Biochem (2002) 35:523–9. 10.1016/S0009-9120(02)00380-6 [DOI] [PubMed] [Google Scholar]
  • 785.Konz T, Añôn Alvarez E, Montes-Bayon M, Sanz-Medel A. Antibody labeling and elemental mass spectrometry (inductively coupled plasma-mass spectrometry) using isotope dilution for highly sensitive ferritin determination and iron-ferritin ratio measurements. Anal Chem (2013) 85:8334–40. 10.1021/ac401692k [DOI] [PubMed] [Google Scholar]
  • 786.Entman SS, Richardson LD, Killam AP. Altered ferrokinetics in toxemia of pregnancy – a possible indicator of decreased red cell survival. Clin Exp Hypertens B (1983) 2:171–8. 10.3109/10641958309023469 [DOI] [PubMed] [Google Scholar]
  • 787.Hubel CA, Bodnar LM, Many A, Harger G, Ness RB, Roberts JM. Nonglycosylated ferritin predominates in the circulation of women with preeclampsia but not intrauterine growth restriction. Clin Chem (2004) 50:948–51. 10.1373/clinchem.2003.030932 [DOI] [PubMed] [Google Scholar]
  • 788.Visser A, van de Vyver A. Severe hyperferritinemia in Mycobacteria tuberculosis infection. Clin Infect Dis (2011) 52:273–4. 10.1093/cid/ciq126 [DOI] [PubMed] [Google Scholar]
  • 789.Ishida JH, Johansen KL. Iron and infection in hemodialysis patients. Semin Dial (2014) 27:26–36. 10.1111/sdi.12168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 790.Esteller M. Non-coding RNAs in human disease. Nat Rev Genet (2011) 12:861–74. 10.1038/nrg3074 [DOI] [PubMed] [Google Scholar]
  • 791.Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res (2014) 42:D68–73. 10.1093/nar/gkt1181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 792.Goulart LF, Bettella F, Sønderby IE, Schork AJ, Thompson WK, Mattingsdal M, et al. microRNAs enrichment in GWAS of complex human phenotypes. BMC Genomics (2015) 16:304. 10.1186/s12864-015-1513-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 793.Londin E, Loher P, Telonis AG, Quann K, Clark P, Jing Y, et al. Analysis of 13 cell types reveals evidence for the expression of numerous novel primate- and tissue-specific microRNAs. Proc Natl Acad Sci U S A (2015) 112:E1106–15. 10.1073/pnas.1420955112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 794.Luo SS, Ishibashi O, Ishikawa G, Ishikawa T, Katayama A, Mishima T, et al. Human villous trophoblasts express and secrete placenta-specific microRNAs into maternal circulation via exosomes. Biol Reprod (2009) 81:717–29. 10.1095/biolreprod.108.075481 [DOI] [PubMed] [Google Scholar]
  • 795.Ouyang Y, Mouillet JF, Coyne CB, Sadovsky Y. Review: placenta-specific microRNAs in exosomes – good things come in nano-packages. Placenta (2014) 35(Suppl):S69–73. 10.1016/j.placenta.2013.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 796.Record M. Intercellular communication by exosomes in placenta: a possible role in cell fusion? Placenta (2014) 35:297–302. 10.1016/j.placenta.2014.02.009 [DOI] [PubMed] [Google Scholar]
  • 797.Miura K, Miura S, Yamasaki K, Higashijima A, Kinoshita A, Yoshiura K, et al. Identification of pregnancy-associated microRNAs in maternal plasma. Clin Chem (2010) 56:1767–71. 10.1373/clinchem.2010.147660 [DOI] [PubMed] [Google Scholar]
  • 798.Morales-Prieto DM, Markert UR. microRNAs in pregnancy. J Reprod Immunol (2011) 88:106–11. 10.1016/j.jri.2011.01.004 [DOI] [PubMed] [Google Scholar]
  • 799.Mouillet JF, Ouyang Y, Coyne CB, Sadovsky Y. microRNAs in placental health and disease. Am J Obstet Gynecol (2015) 213:S163–72. 10.1016/j.ajog.2015.05.057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 800.Jairajpuri DS, Almawi WY. microRNA expression pattern in pre-eclampsia (review). Mol Med Rep (2016) 13:2351–8. 10.3892/mmr.2016.4846 [DOI] [PubMed] [Google Scholar]
  • 801.Kotlabova K, Doucha J, Hromadnikova I. Placental-specific microRNA in maternal circulation –identification of appropriate pregnancy-associated microRNAs with diagnostic potential. J Reprod Immunol (2011) 89:185–91. 10.1016/j.jri.2011.02.006 [DOI] [PubMed] [Google Scholar]
  • 802.Zhao Z, Moley KH, Gronowski AM. Diagnostic potential for miRNAs as biomarkers for pregnancy-specific diseases. Clin Biochem (2013) 46:953–60. 10.1016/j.clinbiochem.2013.01.026 [DOI] [PubMed] [Google Scholar]
  • 803.Tsochandaridis M, Nasca L, Toga C, Levy-Mozziconacci A. Circulating microRNAs as clinical biomarkers in the predictions of pregnancy complications. Biomed Res Int (2015) 2015:294954. 10.1155/2015/294954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 804.Zhu XM, Han T, Sargent IL, Yin GW, Yao YQ. Differential expression profile of microRNAs in human placentas from preeclamptic pregnancies vs normal pregnancies. Am J Obstet Gynecol (2009) 200:661.e661–7. 10.1016/j.ajog.2008.12.045 [DOI] [PubMed] [Google Scholar]
  • 805.Wang D, Song W, Na Q. The emerging roles of placenta-specific microRNAs in regulating trophoblast proliferation during the first trimester. Aust N Z J Obstet Gynaecol (2012) 52:565–70. 10.1111/j.1479-828X.2012.01481.x [DOI] [PubMed] [Google Scholar]
  • 806.Chen DB, Wang W. Human placental microRNAs and preeclampsia. Biol Reprod (2013) 88:130. 10.1095/biolreprod.113.107805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 807.Dong F, Zhang Y, Xia F, Yang Y, Xiong S, Jin L, et al. Genome-wide miRNA profiling of villus and decidua of recurrent spontaneous abortion patients. Reproduction (2014) 148:33–41. 10.1530/REP-14-0095 [DOI] [PubMed] [Google Scholar]
  • 808.Hromadnikova I, Kotlabova K, Hympanova L, Doucha J, Krofta L. First trimester screening of circulating C19MC microRNAs can predict subsequent onset of gestational hypertension. PLoS One (2014) 9:e113735. 10.1371/journal.pone.0113735 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 809.Anton L, Olarerin-George AO, Hogenesch JB, Elovitz MA. Placental expression of miR-517a/b and miR-517c contributes to trophoblast dysfunction and preeclampsia. PLoS One (2015) 10:e0122707. 10.1371/journal.pone.0122707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 810.Yang S, Li H, Ge Q, Guo L, Chen F. Deregulated microRNA species in the plasma and placenta of patients with preeclampsia. Mol Med Rep (2015) 12:527–34. 10.3892/mmr.2015.3414 [DOI] [PubMed] [Google Scholar]
  • 811.Ura B, Feriotto G, Monasta L, Bilel S, Zweyer M, Celeghini C. Potential role of circulating microRNAs as early markers of preeclampsia. Taiwan J Obstet Gynecol (2014) 53:232–4. 10.1016/j.tjog.2014.03.001 [DOI] [PubMed] [Google Scholar]
  • 812.Bortolin-Cavaillé ML, Dance M, Weber M, Cavaillé J. C19MC microRNAs are processed from introns of large Pol-II, non-protein-coding transcripts. Nucleic Acids Res (2009) 37:3464–73. 10.1093/nar/gkp205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 813.Morales-Prieto DM, Ospina-Prieto S, Chaiwangyen W, Schoenleben M, Markert UR. Pregnancy-associated miRNA-clusters. J Reprod Immunol (2013) 97:51–61. 10.1016/j.jri.2012.11.001 [DOI] [PubMed] [Google Scholar]
  • 814.Delorme-Axford E, Donker RB, Mouillet JF, Chu T, Bayer A, Ouyang Y, et al. Human placental trophoblasts confer viral resistance to recipient cells. Proc Natl Acad Sci U S A (2013) 110:12048–53. 10.1073/pnas.1304718110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 815.Bayer A, Delorme-Axford E, Sleigher C, Frey TK, Trobaugh DW, Klimstra WB, et al. Human trophoblasts confer resistance to viruses implicated in perinatal infection. Am J Obstet Gynecol (2015) 212(71):e71–8. 10.1016/j.ajog.2014.07.060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 816.Olarerin-George AO, Anton L, Hwang YC, Elovitz MA, Hogenesch JB. A functional genomics screen for microRNA regulators of NF-kappaB signaling. BMC Biol (2013) 11:19. 10.1186/1741-7007-11-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 817.Song GY, Song WW, Han Y, Wang D, Na Q. Characterization of the role of microRNA-517a expression in low birth weight infants. J Dev Orig Health Dis (2013) 4:522–6. 10.1017/S204017441300024X [DOI] [PubMed] [Google Scholar]
  • 818.Mouillet JF, Ouyang Y, Bayer A, Coyne CB, Sadovsky Y. The role of trophoblastic microRNAs in placental viral infection. Int J Dev Biol (2014) 58:281–9. 10.1387/ijdb.130349ys [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 819.Moro L, Bardaji A, Macete E, Barrios D, Morales-Prieto DM, Espana C, et al. Placental microparticles and microRNAs in pregnant women with Plasmodium falciparum or HIV infection. PLoS One (2016) 11:e0146361. 10.1371/journal.pone.0146361 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 820.Khatun S, Kanayama N, Belayet HM, Bhuiyan AB, Jahan S, Begum A, et al. Increased concentrations of plasma neuropeptide Y in patients with eclampsia and preeclampsia. Am J Obstet Gynecol (2000) 182:896–900. 10.1016/S0002-9378(00)70342-5 [DOI] [PubMed] [Google Scholar]
  • 821.Hauser GJ, Dayao EK, Zukowska-Grojec Z. Effect of neuropeptide Y on endotoxin-induced suppression of the response to various agonists in conscious rats. Life Sci (1995) 57:235–44. 10.1016/0024-3205(95)00266-9 [DOI] [PubMed] [Google Scholar]
  • 822.Kuncová J, Sýkora R, Chvojka J, Švíglerová J, Štengl M, Kroužecký A, et al. Plasma and tissue levels of neuropeptide y in experimental septic shock: relation to hemodynamics, inflammation, oxidative stress, and hemofiltration. Artif Organs (2011) 35:625–33. 10.1111/j.1525-1594.2010.01154.x [DOI] [PubMed] [Google Scholar]
  • 823.Goetz DH, Holmes MA, Borregaard N, Bluhm ME, Raymond KN, Strong RK. The neutrophil lipocalin NGAL is a bacteriostatic agent that interferes with siderophore-mediated iron acquisition. Mol Cell (2002) 10:1033–43. 10.1016/S1097-2765(02)00708-6 [DOI] [PubMed] [Google Scholar]
  • 824.Bachman MA, Lenio S, Schmidt L, Oyler JE, Weiser JN. Interaction of lipocalin 2, transferrin, and siderophores determines the replicative niche of Klebsiella pneumoniae during pneumonia. mBio (2012) 3:e224–211. 10.1128/mBio.00224-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 825.Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature (2004) 432:917–21. 10.1038/nature03104 [DOI] [PubMed] [Google Scholar]
  • 826.Chakraborty S, Kaur S, Guha S, Batra SK. The multifaceted roles of neutrophil gelatinase associated lipocalin (NGAL) in inflammation and cancer. Biochim Biophys Acta (2012) 1826:129–69. 10.1016/j.bbcan.2012.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 827.Cemgil Arikan D, Ozkaya M, Adali E, Kilinc M, Coskun A, Ozer A, et al. Plasma lipocalin-2 levels in pregnant women with pre-eclampsia, and their relation with severity of disease. J Matern Fetal Neonatal Med (2010) 24:291–6 10.3109/14767058.2010.487138 [DOI] [PubMed] [Google Scholar]
  • 828.D’Anna R, Baviera G, Giordano D, Todarello G, Corrado F, Buemi M. Second trimester neutrophil gelatinase-associated lipocalin as a potential prediagnostic marker of preeclampsia. Acta Obstet Gynecol Scand (2008) 87:1370–3. 10.1080/00016340802464463 [DOI] [PubMed] [Google Scholar]
  • 829.D’Anna R, Baviera G, Giordano D, Russo S, Dugo N, Santamaria A, et al. First trimester serum PAPP-A and NGAL in the prediction of late-onset pre-eclampsia. Prenat Diagn (2009) 29:1066–8. 10.1002/pd.2339 [DOI] [PubMed] [Google Scholar]
  • 830.D’Anna R, Baviera G, Giordano D, Todarello G, Russo S, Recupero S, et al. Neutrophil gelatinase-associated lipocalin serum evaluation through normal pregnancy and in pregnancies complicated by preeclampsia. Acta Obstet Gynecol Scand (2010) 89:275–8. 10.3109/00016340903443676 [DOI] [PubMed] [Google Scholar]
  • 831.Stepan H, Philipp A, Reiche M, Klostermann K, Schrey S, Reisenbuchler C, et al. Serum levels of the adipokine lipocalin-2 are increased in preeclampsia. J Endocrinol Invest (2010) 33:629–32. 10.3275/6884 [DOI] [PubMed] [Google Scholar]
  • 832.Kim SM, Park JS, Norwitz ER, Jung HJ, Kim BJ, Park CW, et al. Circulating levels of neutrophil gelatinase-associated lipocalin (NGAL) correlate with the presence and severity of preeclampsia. Reprod Sci (2013) 20:1083–9. 10.1177/1933719113477480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 833.Scazzochio E, Munmany M, Garcia L, Meler E, Crispi F, Gratacos E, et al. Prognostic role of maternal neutrophil gelatinase-associated lipocalin in women with severe early-onset preeclampsia. Fetal Diagn Ther (2013) 35:127–32. 10.1159/000356499 [DOI] [PubMed] [Google Scholar]
  • 834.Karampas G, Eleftheriades M, Panoulis K, Rizou M, Haliassos A, Hassiakos D, et al. Maternal serum levels of neutrophil gelatinase-associated lipocalin (NGAL), matrix metalloproteinase-9 (MMP-9) and their complex MMP-9/NGAL in pregnancies with preeclampsia and those with a small for gestational age neonate. A longitudinal study. Prenat Diagn (2014) 34:726–33. 10.1002/pd.4337 [DOI] [PubMed] [Google Scholar]
  • 835.Sachan R, Patel M, Gaurav A, Gangwar R, Sachan P. Correlation of serum neutrophil gelatinase associated lipocalin with disease severity in hypertensive disorders of pregnancy. Adv Biomed Res (2014) 3:223. 10.4103/2277-9175.145690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 836.Simonazzi G, Capelli I, Curti A, Comai G, Rizzo N, La Manna G. Serum and urinary neutrophil gelatinase-associated lipocalin monitoring in normal pregnancy versus pregnancies complicated by pre-eclampsia. In Vivo (2015) 29:117–21. [PubMed] [Google Scholar]
  • 837.Karampas GA, Eleftheriades MI, Panoulis KC, Rizou MD, Haliassos AD, Metallinou DK, et al. Prediction of pre-eclampsia combining NGAL and other biochemical markers with Doppler in the first and/or second trimester of pregnancy. A pilot study. Eur J Obstet Gynecol Reprod Biol (2016) 205:153–7. 10.1016/j.ejogrb.2016.08.034 [DOI] [PubMed] [Google Scholar]
  • 838.Mishra J, Dent C, Tarabishi R, Mitsnefes MM, Ma Q, Kelly C, et al. Neutrophil gelatinase-associated lipocalin (NGAL) as a biomarker for acute renal injury after cardiac surgery. Lancet (2005) 365:1231–8. 10.1016/S0140-6736(05)74811-X [DOI] [PubMed] [Google Scholar]
  • 839.Soni SS, Cruz D, Bobek I, Chionh CY, Nalesso F, Lentini P, et al. NGAL: a biomarker of acute kidney injury and other systemic conditions. Int Urol Nephrol (2010) 42:141–50. 10.1007/s11255-009-9608-z [DOI] [PubMed] [Google Scholar]
  • 840.Clerico A, Galli C, Fortunato A, Ronco C. Neutrophil gelatinase-associated lipocalin (NGAL) as biomarker of acute kidney injury: a review of the laboratory characteristics and clinical evidences. Clin Chem Lab Med (2012) 50:1505–17. 10.1515/cclm-2011-0814 [DOI] [PubMed] [Google Scholar]
  • 841.Patel M, Sachan R, Gangwar R, Sachan P, Natu S. Correlation of serum neutrophil gelatinase-associated lipocalin with acute kidney injury in hypertensive disorders of pregnancy. Int J Nephrol Renovasc Dis (2013) 6:181–6. 10.2147/IJNRD.S45523 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 842.Aydoğdu M, Gürsel G, Sancak B, Yeni S, Sari G, Taşyürek S, et al. The use of plasma and urine neutrophil gelatinase associated lipocalin (NGAL) and cystatin C in early diagnosis of septic acute kidney injury in critically ill patients. Dis Markers (2013) 34:237–46. 10.3233/DMA-130966 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 843.Mårtensson J, Bell M, Xu S, Bottai M, Ravn B, Venge P, et al. Association of plasma neutrophil gelatinase-associated lipocalin (NGAL) with sepsis and acute kidney dysfunction. Biomarkers (2013) 18:349–56. 10.3109/1354750X.2013.787460 [DOI] [PubMed] [Google Scholar]
  • 844.Otto GP, Busch M, Sossdorf M, Claus RA. Impact of sepsis-associated cytokine storm on plasma NGAL during acute kidney injury in a model of polymicrobial sepsis. Crit Care (2013) 17:419. 10.1186/cc12540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 845.Zughaier SM, Tangpricha V, Leong T, Stecenko AA, Mccarty NA. Peripheral monocytes derived from patients with cystic fibrosis and healthy donors secrete NGAL in response to Pseudomonas aeruginosa infection. J Investig Med (2013) 61:1018–25. 10.231/JIM.0b013e31829cbd14 [DOI] [PubMed] [Google Scholar]
  • 846.MacDonald SPJ, Stone SF, Neil CL, Van Eeden PE, Fatovich DM, Arendts G, et al. Sustained elevation of resistin, NGAL and IL-8 are associated with severe sepsis/septic shock in the emergency department. PLoS One (2014) 9:e110678. 10.1371/journal.pone.0110678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 847.Smertka M, Wroblewska J, Suchojad A, Majcherczyk M, Jadamus-Niebroj D, Owsianka-Podlesny T, et al. Serum and urinary NGAL in septic newborns. Biomed Res Int (2014) 2014:717318. 10.1155/2014/717318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 848.Hjortrup PB, Haase N, Treschow F, Møller MH, Perner A. Predictive value of NGAL for use of renal replacement therapy in patients with severe sepsis. Acta Anaesthesiol Scand (2015) 59:25–34. 10.1111/aas.12427 [DOI] [PubMed] [Google Scholar]
  • 849.Nga HS, Medeiros P, Menezes P, Bridi R, Balbi A, Ponce D. Sepsis and AKI in clinical emergency room patients: the role of urinary NGAL. Biomed Res Int (2015) 2015:413751. 10.1155/2015/413751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 850.Suchojad A, Tarko A, Smertka M, Majcherczyk M, Brzozowska A, Wroblewska J, et al. Factors limiting usefulness of serum and urinary NGAL as a marker of acute kidney injury in preterm newborns. Ren Fail (2015) 37:439–45. 10.3109/0886022X.2014.996109 [DOI] [PubMed] [Google Scholar]
  • 851.Kanda J, Mori K, Kawabata H, Kuwabara T, Mori KP, Imamaki H, et al. An AKI biomarker lipocalin 2 in the blood derives from the kidney in renal injury but from neutrophils in normal and infected conditions. Clin Exp Nephrol (2014) 19:99–106. 10.1007/s10157-014-0952-7 [DOI] [PubMed] [Google Scholar]
  • 852.Mårtensson J, Bellomo R. The rise and fall of NGAL in acute kidney injury. Blood Purif (2014) 37:304–10. 10.1159/000364937 [DOI] [PubMed] [Google Scholar]
  • 853.Srisawat N, Praditpornsilpa K, Patarakul K, Techapornrung M, Daraswang T, Sukmark T, et al. Neutrophil gelatinase associated lipocalin (NGAL) in leptospirosis acute kidney injury: a multicenter study in Thailand. PLoS One (2015) 10:e0143367. 10.1371/journal.pone.0143367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 854.Leelahavanichkul A, Somparn P, Issara-Amphorn J, Eiam-Ong S, Avihingsanon Y, Hirankarn N, et al. Serum neutrophil gelatinase associated lipocalin (NGAL) outperforms serum creatinine in detecting sepsis-induced acute kidney injury, experiments on bilateral nephrectomy and bilateral ureter obstruction mouse models. Shock (2016) 45:570–6. 10.1097/SHK.0000000000000530 [DOI] [PubMed] [Google Scholar]
  • 855.Dewerchin M, Carmeliet P. PlGF: a multitasking cytokine with disease-restricted activity. Cold Spring Harb Perspect Med (2012) 2:a011056. 10.1101/cshperspect.a011056 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 856.De Vivo A, Baviera G, Giordano D, Todarello G, Corrado F, D’Anna R. Endoglin, PlGF and sFlt-1 as markers for predicting pre-eclampsia. Acta Obstet Gynecol Scand (2008) 87:837–42. 10.1080/00016340802253759 [DOI] [PubMed] [Google Scholar]
  • 857.Kaufmann I, Rusterholz C, Hosli I, Hahn S, Lapaire O. Can detection of late-onset PE at triage by sflt-1 or PlGF be improved by the use of additional biomarkers? Prenat Diagn (2012) 32:1288–94. 10.1002/pd.3995 [DOI] [PubMed] [Google Scholar]
  • 858.Gómez-Arriaga PI, Herraiz I, López-Jiménez EA, Gómez-Montes E, Denk B, Galindo A. Uterine artery Doppler and sFlt-1/PlGF ratio: usefulness in diagnosis of pre-eclampsia. Ultrasound Obstet Gynecol (2013) 41:530–7. 10.1002/uog.12400 [DOI] [PubMed] [Google Scholar]
  • 859.Ohkuchi A, Hirashima C, Takahashi K, Suzuki H, Matsubara S, Suzuki M. Onset threshold of the plasma levels of soluble fms-like tyrosine kinase 1/placental growth factor ratio for predicting the imminent onset of preeclampsia within 4 weeks after blood sampling at 19-31 weeks of gestation. Hypertens Res (2013) 36:1073–80. 10.1038/hr.2013.95 [DOI] [PubMed] [Google Scholar]
  • 860.Hirashima C, Ohkuchi A, Takahashi K, Suzuki H, Matsubara S, Suzuki M. A novel three-step approach for predicting the imminent onset of preeclampsia within 4 weeks after blood sampling at 19-31 weeks of gestation. Hypertens Res (2014) 37:519–25. 10.1038/hr.2014.31 [DOI] [PubMed] [Google Scholar]
  • 861.Lai J, Garcia-Tizon Larroca S, Peeva G, Poon LC, Wright D, Nicolaides KH. Competing risks model in screening for preeclampsia by serum placental growth factor and soluble fms-like tyrosine kinase-1 at 30-33 weeks’ gestation. Fetal Diagn Ther (2014) 35:240–8. 10.1159/000359968 [DOI] [PubMed] [Google Scholar]
  • 862.Masoura S, Kalogiannidis I, Makedou K, Theodoridis T, Koiou K, Gerou S, et al. Biomarkers of endothelial dysfunction in preeclampsia and neonatal morbidity: a case-control study. Eur J Obstet Gynecol Reprod Biol (2014) 175:119–23. 10.1016/j.ejogrb.2014.01.012 [DOI] [PubMed] [Google Scholar]
  • 863.Ohkuchi A, Hirashima C, Takahashi K, Shirasuna K, Suzuki H, Ariga H, et al. A trio of risk factors for the onset of preeclampsia in the second and early third trimesters. Pregnancy Hypertens (2014) 4:224–30. 10.1016/j.preghy.2014.04.024 [DOI] [PubMed] [Google Scholar]
  • 864.Dröge L, Herraìz I, Zeisler H, Schlembach D, Stepan H, Küssel L, et al. Maternal serum sFlt-1/PlGF ratio in twin pregnancies with and without pre-eclampsia in comparison with singleton pregnancies. Ultrasound Obstet Gynecol (2015) 45:286–93. 10.1002/uog.14760 [DOI] [PubMed] [Google Scholar]
  • 865.Rolfo A, Attini R, Tavassoli E, Neve FV, Nigra M, Cicilano M, et al. Is it possible to differentiate chronic kidney disease and preeclampsia by means of new and old biomarkers? A prospective study. Dis Markers (2015) 2015:127083. 10.1155/2015/127083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 866.Stepan H, Herraiz I, Schlembach D, Verlohren S, Brennecke S, Chantraine F, et al. Implementation of the sFlt-1/PlGF ratio for prediction and diagnosis of pre-eclampsia in singleton pregnancy: implications for clinical practice. Ultrasound Obstet Gynecol (2015) 45:241–6. 10.1002/uog.14799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 867.Khalil A, Maiz N, Garcia-Mandujano R, Penco JM, Nicolaides KH. Longitudinal changes in maternal serum placental growth factor and soluble fms-like tyrosine kinase-1 in women at increased risk of pre-eclampsia. Ultrasound Obstet Gynecol (2016) 47:324–31. 10.1002/uog.15750 [DOI] [PubMed] [Google Scholar]
  • 868.Kim MY, Buyon JP, Guerra MM, Rana S, Zhang D, Laskin CA, et al. Angiogenic factor imbalance early in pregnancy predicts adverse outcomes in patients with lupus and antiphospholipid antibodies: results of the PROMISSE study. Am J Obstet Gynecol (2016) 214: 108.e101–e114. 10.1016/j.ajog.2015.09.066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 869.Zeisler H, Llurba E, Chantraine F, Vatish M, Staff AC, Sennstrom M, et al. Predictive value of the sFlt-1:PlGF ratio in women with suspected preeclampsia. N Engl J Med (2016) 374:13–22. 10.1056/NEJMoa1414838 [DOI] [PubMed] [Google Scholar]
  • 870.Levine RJ, Thadhani R, Qian C, Lam C, Lim KH, Yu KF, et al. Urinary placental growth factor and risk of preeclampsia. JAMA (2005) 293:77–85. 10.1001/jama.293.1.77 [DOI] [PubMed] [Google Scholar]
  • 871.Odibo AO, Patel KR, Spitalnik A, Odibo L, Huettner P. Placental pathology, first-trimester biomarkers and adverse pregnancy outcomes. J Perinatol (2014) 34:186–91. 10.1038/Jp.2013.176 [DOI] [PubMed] [Google Scholar]
  • 872.Bramham K, Seed PT, Lightstone L, Nelson-Piercy C, Gill C, Webster P, et al. Diagnostic and predictive biomarkers for pre-eclampsia in patients with established hypertension and chronic kidney disease. Kidney Int (2016) 89:874–85. 10.1016/j.kint.2015.10.012 [DOI] [PubMed] [Google Scholar]
  • 873.Duckworth S, Griffin M, Seed PT, North R, Myers J, Mackillop L, et al. Diagnostic biomarkers in women with suspected preeclampsia in a prospective multicenter study. Obstet Gynecol (2016) 128:245–52. 10.1097/AOG.0000000000001508 [DOI] [PubMed] [Google Scholar]
  • 874.Yano K, Liaw PC, Mullington JM, Shih SC, Okada H, Bodyak N, et al. Vascular endothelial growth factor is an important determinant of sepsis morbidity and mortality. J Exp Med (2006) 203:1447–58. 10.1084/jem.20060375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 875.Yano K, Okada Y, Beldi G, Shih SC, Bodyak N, Okada H, et al. Elevated levels of placental growth factor represent an adaptive host response in sepsis. J Exp Med (2008) 205:2623–31. 10.1084/jem.20080398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 876.Foidart JM, Schaaps JP, Chantraine F, Munaut C, Lorquet S. Dysregulation of anti-angiogenic agents (sFlt-1, PLGF, and sEndoglin) in preeclampsia – a step forward but not the definitive answer. J Reprod Immunol (2009) 82:106–11. 10.1016/j.jri.2009.09.001 [DOI] [PubMed] [Google Scholar]
  • 877.Furuya M, Kurasawa K, Nagahama K, Kawachi K, Nozawa A, Takahashi T, et al. Disrupted balance of angiogenic and antiangiogenic signalings in preeclampsia. J Pregnancy (2011) 2011:123717. 10.1155/2011/123717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 878.De Oliveira L, Sass N, Boute T, Moron AF. sFlt-1 and PlGF levels in a patient with mirror syndrome related to cytomegalovirus infection. Eur J Obstet Gynecol Reprod Biol (2011) 158:366–7. 10.1016/j.ejogrb.2011.04.049 [DOI] [PubMed] [Google Scholar]
  • 879.Benton SJ, Hu Y, Xie F, Kupfer K, Lee SW, Magee LA, et al. Can placental growth factor in maternal circulation identify fetuses with placental intrauterine growth restriction? Am J Obstet Gynecol (2012) 206(163):e161–7. 10.1016/j.ajog.2011.09.019 [DOI] [PubMed] [Google Scholar]
  • 880.Acharya A. Promising biomarkers for superimposed pre-eclampsia in pregnant women with established hypertension and chronic kidney disease. Kidney Int (2016) 89:743–6. 10.1016/j.kint.2016.01.008 [DOI] [PubMed] [Google Scholar]
  • 881.Montagnana M, Lippi G, Albiero A, Scevarolli S, Salvagno GL, Franchi M, et al. Procalcitonin values in preeclamptic women are related to severity of disease. Clin Chem Lab Med (2008) 46:1050–1. 10.1515/CCLM.2008.199 [DOI] [PubMed] [Google Scholar]
  • 882.Can M, Sancar E, Harma M, Guven B, Mungan G, Acikgoz S. Inflammatory markers in preeclamptic patients. Clin Chem Lab Med (2011) 49:1469–72. 10.1515/CCLM.2011.232 [DOI] [PubMed] [Google Scholar]
  • 883.Simon L, Gauvin F, Amre DK, Saint-Louis P, Lacroix J. Serum procalcitonin and C-reactive protein levels as markers of bacterial infection: a systematic review and meta-analysis. Clin Infect Dis (2004) 39:206–17. 10.1086/421997 [DOI] [PubMed] [Google Scholar]
  • 884.Ucar B, Yildiz B, Aksit MA, Yarar C, Colak O, Akbay Y, et al. Serum amyloid A, procalcitonin, tumor necrosis factor-alpha, and interleukin-1beta levels in neonatal late-onset sepsis. Mediators Inflamm (2008) 2008:737141. 10.1155/2008/737141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 885.Riedel S, Melendez JH, An AT, Rosenbaum JE, Zenilman JM. Procalcitonin as a marker for the detection of bacteremia and sepsis in the emergency department. Am J Clin Pathol (2011) 135:182–9. 10.1309/AJCP1MFYINQLECV2 [DOI] [PubMed] [Google Scholar]
  • 886.Riedel S. Procalcitonin and the role of biomarkers in the diagnosis and management of sepsis. Diagn Microbiol Infect Dis (2012) 73:221–7. 10.1016/j.diagmicrobio.2012.05.002 [DOI] [PubMed] [Google Scholar]
  • 887.Schuetz P, Muller B, Christ-Crain M, Stolz D, Tamm M, Bouadma L, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev (2012) 9:CD007498. 10.1002/14651858.CD007498.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 888.Ulla M, Pizzolato E, Lucchiari M, Loiacono M, Soardo F, Forno D, et al. Diagnostic and prognostic value of presepsin in the management of sepsis in the emergency department: a multicenter prospective study. Crit Care (2013) 17:R168. 10.1186/cc12847 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 889.Henriquez-Camacho C, Losa J. Biomarkers for sepsis. Biomed Res Int (2014) 2014:547818. 10.1155/2014/547818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 890.Tian G, Pan SY, Ma G, Liao W, Su QG, Gu BC, et al. Serum levels of procalcitonin as a biomarker for differentiating between sepsis and systemic inflammatory response syndrome in the neurological intensive care unit. J  Clin Neurosci (2014) 21:1153–8. 10.1016/j.jocn.2013.09.021 [DOI] [PubMed] [Google Scholar]
  • 891.Westwood M, Ramaekers B, Whiting P, Tomini F, Joore M, Armstrong N, et al. Procalcitonin testing to guide antibiotic therapy for the treatment of sepsis in intensive care settings and for suspected bacterial infection in emergency department settings: a systematic review and cost-effectiveness analysis. Health Technol Assess (2015) 19:v–xxv, 1–236. 10.3310/hta19960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 892.Engin-Üstün Y, Üstün Y, Karabulut AB, Özkaplan E, Meydanli MM, Kafkaslı A. Serum amyloid A levels are increased in pre-eclampsia. Gynecol Obstet Invest (2007) 64:117–20. 10.1159/000100329 [DOI] [PubMed] [Google Scholar]
  • 893.Kristensen K, Wide-Swensson D, Lindström V, Schmidt C, Grubb A, Strevens H. Serum amyloid A protein and C-reactive protein in normal pregnancy and preeclampsia. Gynecol Obstet Invest (2009) 67:275–80. 10.1159/000214081 [DOI] [PubMed] [Google Scholar]
  • 894.Malle E, De Beer FC. Human serum amyloid A (SAA) protein: a prominent acute-phase reactant for clinical practice. Eur J Clin Invest (1996) 26:427–35. 10.1046/j.1365-2362.1996.159291.x [DOI] [PubMed] [Google Scholar]
  • 895.Pizzini C, Mussap M, Plebani M, Fanos V. C-reactive protein and serum amyloid A protein in neonatal infections. Scand J Infect Dis (2000) 32:229–35. 10.1080/00365540050165848 [DOI] [PubMed] [Google Scholar]
  • 896.Sipe JD. Serum amyloid A: from fibril to function. Current status. Amyloid (2000) 7:10–2. 10.3109/13506120009146815 [DOI] [PubMed] [Google Scholar]
  • 897.Urieli-Shoval S, Linke RP, Matzner Y. Expression and function of serum amyloid A, a major acute-phase protein, in normal and disease states. Curr Opin Hematol (2000) 7:64–9. 10.1097/00062752-200001000-00012 [DOI] [PubMed] [Google Scholar]
  • 898.Falsey AR, Walsh EE, Francis CW, Looney RJ, Kolassa JE, Hall WJ, et al. Response of C-reactive protein and serum amyloid A to influenza A infection in older adults. J Infect Dis (2001) 183:995–9. 10.1086/319275 [DOI] [PubMed] [Google Scholar]
  • 899.Arnon S, Litmanovitz I, Regev RH, Bauer S, Shainkin-Kestenbaum R, Dolfin T. Serum amyloid A: an early and accurate marker of neonatal early-onset sepsis. J Perinatol (2007) 27:297–302. 10.1038/sj.jp.7211682 [DOI] [PubMed] [Google Scholar]
  • 900.Bozinovski S, Hutchinson A, Thompson M, MacGregor L, Black J, Giannakis E, et al. Serum amyloid a is a biomarker of acute exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med (2008) 177:269–78. 10.1164/rccm.200705-678OC [DOI] [PubMed] [Google Scholar]
  • 901.Cicarelli DD, Vieira JE, Bensenor FE. Comparison of C-reactive protein and serum amyloid a protein in septic shock patients. Mediators Inflamm (2008) 2008:631414. 10.1155/2008/631414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 902.Ebert EC, Nagar M. Gastrointestinal manifestations of amyloidosis. Am J  Gastroenterol (2008) 103:776–87. 10.1111/j.1572-0241.2007.01669.x [DOI] [PubMed] [Google Scholar]
  • 903.Lannergård A, Larsson A, Friman G, Ewald U. Human serum amyloid A (SAA) and high sensitive C-reactive protein (hsCRP) in preterm newborn infants with nosocomial infections. Acta Paediatr (2008) 97:1061–5. 10.1111/j.1651-2227.2008.00814.x [DOI] [PubMed] [Google Scholar]
  • 904.Çetinkaya M, Özkan H, Köksal N, Çelebi S, Hacımustafaoğlu M. Comparison of serum amyloid A concentrations with those of C-reactive protein and procalcitonin in diagnosis and follow-up of neonatal sepsis in premature infants. J Perinatol (2009) 29:225–31. 10.1038/jp.2008.207 [DOI] [PubMed] [Google Scholar]
  • 905.Lannergård A, Viberg A, Cars O, Karlsson MO, Sandström M, Larsson A. The time course of body temperature, serum amyloid A protein, C-reactive protein and interleukin-6 in patients with bacterial infection during the initial 3 days of antibiotic therapy. Scand J Infect Dis (2009) 41:663–71. 10.1080/00365540903127417 [DOI] [PubMed] [Google Scholar]
  • 906.Yuan H, Huang J, Lv B, Yan W, Hu G, Wang J, et al. Diagnosis value of the serum amyloid A test in neonatal sepsis: a meta-analysis. Biomed Res Int (2013) 2013:520294. 10.1155/2013/520294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 907.Derebe MG, Zlatkov CM, Gattu S, Ruhn KA, Vaishnava S, Diehl GE, et al. Serum amyloid A is a retinol binding protein that transports retinol during bacterial infection. Elife (2014) 3:e03206. 10.7554/eLife.03206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 908.Buhimschi IA, Nayeri UA, Zhao G, Shook LL, Pensalfini A, Funai EF, et al. Protein misfolding, congophilia, oligomerization, and defective amyloid processing in preeclampsia. Sci Transl Med (2014) 6:245ra292. 10.1126/scitranslmed.3008808 [DOI] [PubMed] [Google Scholar]
  • 909.Little CS, Hammond CJ, Macintyre A, Balin BJ, Appelt DM. Chlamydia pneumoniae induces Alzheimer-like amyloid plaques in brains of BALB/c mice. Neurobiol Aging (2004) 25:419–29. 10.1016/S0197-4580(03)00127-1 [DOI] [PubMed] [Google Scholar]
  • 910.Wozniak MA, Itzhaki RF, Shipley SJ, Dobson CB. Herpes simplex virus infection causes cellular beta-amyloid accumulation and secretase upregulation. Neurosci Lett (2007) 429:95–100. 10.1016/j.neulet.2007.09.077 [DOI] [PubMed] [Google Scholar]
  • 911.Veas CJ, Aguilera VC, Munoz IJ, Gallardo VI, Miguel PL, Gonzalez MA, et al. Fetal endothelium dysfunction is associated with circulating maternal levels of sE-selectin, sVCAM1, and sFlt-1 during pre-eclampsia. J Matern Fetal Neonatal Med (2011) 24:1371–7. 10.3109/14767058.2011.556204 [DOI] [PubMed] [Google Scholar]
  • 912.Whitehead CL, Palmer KR, Nilsson U, Gao Y, Saglam B, Lappas M, et al. Placental expression of a novel primate-specific splice variant of sFlt-1 is upregulated in pregnancies complicated by severe early onset pre-eclampsia. BJOG (2011):1268–71. 10.1111/j.1471-0528.2011.02962.x [DOI] [PubMed] [Google Scholar]
  • 913.Higgins LE, Rey De Castro N, Addo N, Wareing M, Greenwood SL, Jones RL, et al. Placental features of late-onset adverse pregnancy outcome. PLoS One (2015) 10:e0129117. 10.1371/journal.pone.0129117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 914.Hod T, Cerdeira AS, Karumanchi SA. Molecular mechanisms of preeclampsia. Cold Spring Harb Perspect Med (2015) 5:a023473. 10.1101/cshperspect.a023473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 915.Thadhani R, Kisner T, Hagmann H, Bossung V, Noack S, Schaarschmidt W, et al. Pilot study of extracorporeal removal of soluble fms-like tyrosine kinase 1 in preeclampsia. Circulation (2011) 124:940–50. 10.1161/CIRCULATIONAHA.111.034793 [DOI] [PubMed] [Google Scholar]
  • 916.Li F, Hagaman JR, Kim HS, Maeda N, Jennette JC, Faber JE, et al. eNOS deficiency acts through endothelin to aggravate sFlt-1-induced pre-eclampsia-like phenotype. J Am Soc Nephrol (2012) 23:652–60. 10.1681/ASN.2011040369 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 917.Carney EF. Hypertension: sFlt-1 removal seems to be beneficial in women with pre-eclampsia. Nat Rev Nephrol (2015) 11:690. 10.1038/nrneph.2015.168 [DOI] [PubMed] [Google Scholar]
  • 918.Thadhani R, Hagmann H, Schaarschmidt W, Roth B, Cingoez T, Karumanchi SA, et al. Removal of soluble fms-like tyrosine kinase-1 by dextran sulfate apheresis in preeclampsia. J Am Soc Nephrol (2016) 27:903–13. 10.1681/ASN.2015020157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 919.Shapiro NI, Yano K, Okada H, Fischer C, Howell M, Spokes KC, et al. A prospective, observational study of soluble FLT-1 and vascular endothelial growth factor in sepsis. Shock (2008) 29:452–7. 10.1097/SHK.0b013e31815072c1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 920.Schuetz P, Jones AE, Aird WC, Shapiro NI. Endothelial cell activation in emergency department patients with sepsis-related and non-sepsis-related hypotension. Shock (2011) 36:104–8. 10.1097/SHK.0b013e31821e4e04 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 921.Shapiro NI, Schuetz P, Yano K, Sorasaki M, Parikh SM, Jones AE, et al. The association of endothelial cell signaling, severity of illness, and organ dysfunction in sepsis. Crit Care (2010) 14:R182. 10.1186/cc9290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 922.Alves BE, Montalvao SA, Aranha FJ, Lorand-Metze I, De Souza CA, Annichino-Bizzacchi JM, et al. Time-course of sFlt-1 and VEGF-A release in neutropenic patients with sepsis and septic shock: a prospective study. J Transl Med (2011) 9:23. 10.1186/1479-5876-9-23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 923.Xing K, Murthy S, Liles WC, Singh JM. Clinical utility of biomarkers of endothelial activation in sepsis – a systematic review. Crit Care (2012) 16:R7. 10.1186/cc11145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 924.Hsu CD, Iriye B, Johnson TR, Witter FR, Hong SF, Chan DW. Elevated circulating thrombomodulin in severe preeclampsia. Am J Obstet Gynecol (1993) 169:148–9. 10.1016/0002-9378(93)90151-8 [DOI] [PubMed] [Google Scholar]
  • 925.Minakami H, Takahashi T, Izumi A, Tamada T. Increased levels of plasma thrombomodulin in preeclampsia. Gynecol Obstet Invest (1993) 36:208–10. 10.1159/000292631 [DOI] [PubMed] [Google Scholar]
  • 926.Bontis J, Vavilis D, Agorastos T, Zournatzi V, Konstantinidis T, Tagou K. Maternal plasma level of thrombomodulin is increased in mild preeclampsia. Eur J Obstet Gynecol Reprod Biol (1995) 60:139–41. 10.1016/0028-2243(95)02093-8 [DOI] [PubMed] [Google Scholar]
  • 927.Hsu CD, Copel JA, Hong SF, Chan DW. Thrombomodulin levels in preeclampsia, gestational hypertension, and chronic hypertension. Obstet Gynecol (1995) 86:897–9. 10.1016/0029-7844(95)00326-M [DOI] [PubMed] [Google Scholar]
  • 928.Shaarawy M, Didy HE. Thrombomodulin, plasminogen activator inhibitor type 1 (PAI-1) and fibronectin as biomarkers of endothelial damage in preeclampsia and eclampsia. Int J Gynaecol Obstet (1996) 55:135–9. 10.1016/S0020-7292(96)02755-5 [DOI] [PubMed] [Google Scholar]
  • 929.Boffa MC, Valsecchi L, Fausto A, Gozin D, Vigano’ D’Angelo S, Safa O, et al. Predictive value of plasma thrombomodulin in preeclampsia and gestational hypertension. Thromb Haemost (1998) 79:1092–5. [PubMed] [Google Scholar]
  • 930.Kobayashi H, Sadakata H, Suzuki K, She MY, Shibata S, Terao T. Thrombomodulin release from umbilical endothelial cells initiated by preeclampsia plasma-induced neutrophil activation. Obstet Gynecol (1998) 92:425–30. 10.1016/S0029-7844(98)00217-8 [DOI] [PubMed] [Google Scholar]
  • 931.Brenner B. Haemostatic changes in pregnancy. Thromb Res (2004) 114:409–14. 10.1016/j.thromres.2004.08.004 [DOI] [PubMed] [Google Scholar]
  • 932.Bosco C, Parra M, Barja P, Rodrigo R, Fernández V, Suarez M, et al. Increased immunohistochemical expression of thrombomodulin at placental perivascular myofibroblast in severe preeclampsia (PE). Histol Histopathol (2005) 20:1045–55. [DOI] [PubMed] [Google Scholar]
  • 933.Wiwanitkit V. Correlation between thrombomodulin and severe preeclampsia: a summary. Clin Appl Thromb Hemost (2008) 14:99–101. 10.1177/1076029607303965 [DOI] [PubMed] [Google Scholar]
  • 934.Turner RJ, Bloemenkamp KWM, Bruijn JA, Baelde HJ. Loss of thrombomodulin in placental dysfunction in preeclampsia. Arterioscler Thromb Vasc Biol (2016) 36:728–35. 10.1161/ATVBAHA.306780 [DOI] [PubMed] [Google Scholar]
  • 935.Saito H, Maruyama I, Shimazaki S, Yamamoto Y, Aikawa N, Ohno R, et al. Efficacy and safety of recombinant human soluble thrombomodulin (ART-123) in disseminated intravascular coagulation: results of a phase III, randomized, double-blind clinical trial. J Thromb Haemost (2007) 5:31–41. 10.1111/j.1538-7836.2006.02267.x [DOI] [PubMed] [Google Scholar]
  • 936.Levi M, van der Poll T. Thrombomodulin in sepsis. Minerva Anestesiol (2013) 79:294–8. [PubMed] [Google Scholar]
  • 937.Mimuro J, Takahashi H, Kitajima I, Tsuji H, Eguchi Y, Matsushita T, et al. Impact of recombinant soluble thrombomodulin (thrombomodulin alfa) on disseminated intravascular coagulation. Thromb Res (2013) 131:436–43. 10.1016/j.thromres.2013.03.008 [DOI] [PubMed] [Google Scholar]
  • 938.Vincent JL, Ramesh MK, Ernest D, Larosa SP, Pachl J, Aikawa N, et al. A randomized, double-blind, placebo-controlled, phase 2b study to evaluate the safety and efficacy of recombinant human soluble thrombomodulin, ART-123, in patients with sepsis and suspected disseminated intravascular coagulation. Crit Care Med (2013) 41:2069–79. 10.1097/CCM.0b013e31828e9b03 [DOI] [PubMed] [Google Scholar]
  • 939.Shirahata A, Mimuro J, Takahashi H, Kitajima I, Tsuji H, Eguchi Y, et al. Recombinant soluble human thrombomodulin (thrombomodulin alfa) in the treatment of neonatal disseminated intravascular coagulation. Eur J Pediatr (2014) 173:303–11. 10.1007/s00431-013-2155-8 [DOI] [PubMed] [Google Scholar]
  • 940.Levi M. Recombinant soluble thrombomodulin: coagulation takes another chance to reduce sepsis mortality. J Thromb Haemost (2015) 13:505–7. 10.1111/jth.12868 [DOI] [PubMed] [Google Scholar]
  • 941.Yamakawa K, Aihara M, Ogura H, Yuhara H, Hamasaki T, Shimazu T. Recombinant human soluble thrombomodulin in severe sepsis: a systematic review and meta-analysis. J Thromb Haemost (2015) 13:508–19. 10.1111/jth.12841 [DOI] [PubMed] [Google Scholar]
  • 942.Yoshimura J, Yamakawa K, Ogura H, Umemura Y, Takahashi H, Morikawa M, et al. Benefit profile of recombinant human soluble thrombomodulin in sepsis-induced disseminated intravascular coagulation: a multicenter propensity score analysis. Crit Care (2015) 19:78. 10.1186/s13054-015-0810-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 943.Hayakawa M, Yamakawa K, Saito S, Uchino S, Kudo D, Iizuka Y, et al. Recombinant human soluble thrombomodulin and mortality in sepsis-induced disseminated intravascular coagulation. A multicentre retrospective study. Thromb Haemost (2016) 115:1157–66. 10.1160/TH15-12-0987 [DOI] [PubMed] [Google Scholar]
  • 944.Kim YM, Romero R, Oh SY, Kim CJ, Kilburn BA, Armant DR, et al. Toll-like receptor 4: a potential link between “danger signals,” the innate immune system and preeclampsia? Am J Obstet Gynecol (2005) 193:921–7. 10.1016/j.ajog.2005.06.053 [DOI] [PubMed] [Google Scholar]
  • 945.Bonney EA. Preeclampsia: a view through the danger model. J Reprod Immunol (2007) 76:68–74. 10.1016/j.jri.2007.03.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 946.Fasshauer M, Waldeyer T, Seeger J, Schrey S, Ebert T, Kratzsch J, et al. Serum levels of the adipokine visfatin are increased in pre-eclampsia. Clin Endocrinol (Oxf) (2008) 69:69–73. 10.1111/j.1365-2265.2007.03147.x [DOI] [PubMed] [Google Scholar]
  • 947.Adali E, Yildizhan R, Kolusari A, Kurdoglu M, Bugdayci G, Sahin HG, et al. Increased visfatin and leptin in pregnancies complicated by pre-eclampsia. J Matern Fetal Neonatal Med (2009) 22:873–9. 10.1080/14767050902994622 [DOI] [PubMed] [Google Scholar]
  • 948.Mazaki-Tovi S, Vaisbuch E, Romero R, Kusanovic JP, Chaiworapongsa T, Kim SK, et al. Maternal and neonatal circulating visfatin concentrations in patients with pre-eclampsia and a small-for-gestational age neonate. J Matern Fetal Neonatal Med (2010) 23:1119–28. 10.3109/14767050903572190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 949.Hu W, Wang Z, Wang H, Huang H, Dong M. Serum visfatin levels in late pregnancy and pre-eclampsia. Acta Obstet Gynecol Scand (2008) 87:413–8. 10.1080/00016340801976012 [DOI] [PubMed] [Google Scholar]
  • 950.Zonneveld R, Martinelli R, Shapiro NI, Kuijpers TW, Plotz FB, Carman CV. Soluble adhesion molecules as markers for sepsis and the potential pathophysiological discrepancy in neonates, children and adults. Crit Care (2014) 18:204. 10.1186/cc13733 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 951.de Pablo R, Monserrat J, Reyes E, Díaz D, Rodríguez-Zapata M, De La Hera A, et al. Circulating sICAM-1 and sE-selectin as biomarker of infection and prognosis in patients with systemic inflammatory response syndrome. Eur J Intern Med (2013) 24:132–8. 10.1016/j.ejim.2012.10.009 [DOI] [PubMed] [Google Scholar]
  • 952.Mussap M, Noto A, Fravega M, Fanos V. Soluble CD14 subtype presepsin (sCD14-ST) and lipopolysaccharide binding protein (LBP) in neonatal sepsis: new clinical and analytical perspectives for two old biomarkers. J Matern Fetal Neonatal Med (2011) 24(Suppl 2):12–4. 10.3109/14767058.2011.601923 [DOI] [PubMed] [Google Scholar]
  • 953.Okamura Y, Yokoi H. Development of a point-of-care assay system for measurement of presepsin (sCD14-ST). Clin Chim Acta (2011) 412:2157–61. 10.1016/j.cca.2011.07.024 [DOI] [PubMed] [Google Scholar]
  • 954.Endo S, Suzuki Y, Takahashi G, Shozushima T, Ishikura H, Murai A, et al. Usefulness of presepsin in the diagnosis of sepsis in a multicenter prospective study. J Infect Chemother (2012) 18:891–7. 10.1007/s10156-012-0435-2 [DOI] [PubMed] [Google Scholar]
  • 955.Endo S, Suzuki Y, Takahashi G, Shozushima T, Ishikura H, Murai A, et al. Presepsin as a powerful monitoring tool for the prognosis and treatment of sepsis: a multicenter prospective study. J Infect Chemother (2014) 20:30–4. 10.1016/j.jiac.2013.07.005 [DOI] [PubMed] [Google Scholar]
  • 956.Nakamura Y, Ishikura H, Nishida T, Kawano Y, Yuge R, Ichiki R, et al. Usefulness of presepsin in the diagnosis of sepsis in patients with or without acute kidney injury. BMC Anesthesiol (2014) 14:88. 10.1186/1471-2253-14-88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 957.Wu J, Hu L, Zhang G, Wu F, He T. Accuracy of presepsin in sepsis diagnosis: a systematic review and meta-analysis. PLoS One (2015) 10:e0133057. 10.1371/journal.pone.0133057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 958.Ali FT, Ali MA, Elnakeeb MM, Bendary HN. Presepsin is an early monitoring biomarker for predicting clinical outcome in patients with sepsis. Clin Chim Acta (2016) 460:93–101. 10.1016/j.cca.2016.06.030 [DOI] [PubMed] [Google Scholar]
  • 959.Klouche K, Cristol JP, Devin J, Gilles V, Kuster N, Larcher R, et al. Diagnostic and prognostic value of soluble CD14 subtype (presepsin) for sepsis and community-acquired pneumonia in ICU patients. Ann Intensive Care (2016) 6:59. 10.1186/s13613-016-0160-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 960.Song X, Song Y, Yuan Y, Zhang P, Zhang X. Prognostic value of presepsin for outcomes and complications in enterocutaneous fistula complicated by abdominal sepsis. Int J Surg (2016) 33(Pt A):96–101. 10.1016/j.ijsu.2016.07.070 [DOI] [PubMed] [Google Scholar]
  • 961.Takahashi G, Shibata S, Fukui Y, Okamura Y, Inoue Y. Diagnostic accuracy of procalcitonin and presepsin for infectious disease in patients with acute kidney injury. Diagn Microbiol Infect Dis (2016) 86:205–10. 10.1016/j.diagmicrobio.2016.07.015 [DOI] [PubMed] [Google Scholar]
  • 962.Kell DB, Westerhoff HV. Metabolic control theory: its role in microbiology and biotechnology. FEMS Microbiol Rev (1986) 39:305–20. 10.1111/j.1574-6968.1986.tb01863.x [DOI] [Google Scholar]
  • 963.Kell DB. Forces, fluxes and the control of microbial growth and metabolism. The twelfth Fleming lecture. J Gen Microbiol (1987) 133:1651–65. [Google Scholar]
  • 964.Fell DA. Metabolic control analysis – a survey of its theoretical and experimental development. Biochem J (1992) 286:313–30. 10.1042/bj2860313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 965.Heinrich R, Schuster S. The Regulation of Cellular Systems. New York: Chapman & Hall; (1996). [Google Scholar]
  • 966.Kell DB, Oliver SG. The metabolome 18 years on: a concept comes of age. Metabolomics (2016) 12:148. 10.1007/s11306-016-1108-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 967.Oliver SG, Winson MK, Kell DB, Baganz F. Systematic functional analysis of the yeast genome. Trends Biotechnol (1998) 16:373–8. 10.1016/S0167-7799(98)01214-1 [DOI] [PubMed] [Google Scholar]
  • 968.Cornish-Bowden A, Cárdenas ML. Silent genes given voice. Nature (2001) 409:571–2. 10.1038/35054646 [DOI] [PubMed] [Google Scholar]
  • 969.Raamsdonk LM, Teusink B, Broadhurst D, Zhang N, Hayes A, Walsh M, et al. A functional genomics strategy that uses metabolome data to reveal the phenotype of silent mutations. Nat Biotechnol (2001) 19:45–50. 10.1038/83496 [DOI] [PubMed] [Google Scholar]
  • 970.Goodacre R, Vaidyanathan S, Dunn WB, Harrigan GG, Kell DB. Metabolomics by numbers: acquiring and understanding global metabolite data. Trends Biotechnol (2004) 22:245–52. 10.1016/j.tibtech.2004.03.007 [DOI] [PubMed] [Google Scholar]
  • 971.Kell DB. Metabolomics and systems biology: making sense of the soup. Curr Opin Microbiol (2004) 7:296–307. 10.1016/j.mib.2004.04.012 [DOI] [PubMed] [Google Scholar]
  • 972.Kitano H. Biological robustness. Nat Rev Genet (2004) 5:826–37. 10.1038/nrg1471 [DOI] [PubMed] [Google Scholar]
  • 973.Kitano H, Oda K, Kimura T, Matsuoka Y, Csete M, Doyle J, et al. Metabolic syndrome and robustness tradeoffs. Diabetes (2004) 53(Suppl 3):S6–15. 10.2337/diabetes.53.suppl_3.S6 [DOI] [PubMed] [Google Scholar]
  • 974.Wilhelm T, Behre J, Schuster S. Analysis of structural robustness of metabolic networks. Syst Biol (Stevenage) (2004) 1:114–20. 10.1049/sb:20045004 [DOI] [PubMed] [Google Scholar]
  • 975.Grimbs S, Selbig J, Bulik S, Holzhutter HG, Steuer R. The stability and robustness of metabolic states: identifying stabilizing sites in metabolic networks. Mol Syst Biol (2007) 3:146. 10.1038/msb4100186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 976.Kell DB. Finding novel pharmaceuticals in the systems biology era using multiple effective drug targets, phenotypic screening, and knowledge of transporters: where drug discovery went wrong and how to fix it. FEBS J (2013) 280:5957–80. 10.1111/febs.12268 [DOI] [PubMed] [Google Scholar]
  • 977.Quinton-Tulloch MJ, Bruggeman FJ, Snoep JL, Westerhoff HV. Trade-off of dynamic fragility but not of robustness in metabolic pathways in silico. FEBS J (2013) 280:160–73. 10.1111/febs.12057 [DOI] [PubMed] [Google Scholar]
  • 978.Kell DB, Goodacre R. Metabolomics and systems pharmacology: why and how to model the human metabolic network for drug discovery. Drug Discov Today (2014) 19:171–82. 10.1016/j.drudis.2013.07.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 979.Begley P, Francis-Mcintyre S, Dunn WB, Broadhurst DI, Halsall A, Tseng A, et al. Development and performance of a gas chromatography-time-of-flight mass spectrometry analysis for large-scale non-targeted metabolomic studies of human serum. Anal Chem (2009) 81:7038–46. 10.1021/ac9011599 [DOI] [PubMed] [Google Scholar]
  • 980.Zelena E, Dunn WB, Broadhurst D, Francis-Mcintyre S, Carroll KM, Begley P, et al. Development of a robust and repeatable UPLC-MS method for the long-term metabolomic study of human serum. Anal Chem (2009) 81:1357–64. 10.1021/ac8019366 [DOI] [PubMed] [Google Scholar]
  • 981.Dunn WB, Broadhurst D, Begley P, Zelena E, Francis-Mcintyre S, Anderson N, et al. Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat Protoc (2011) 6:1060–83. 10.1038/nprot.2011.335 [DOI] [PubMed] [Google Scholar]
  • 982.Dunn WB, Lin W, Broadhurst D, Begley P, Brown M, Zelena E, et al. Molecular phenotyping of a UK population: defining the human serum metabolome. Metabolomics (2015) 11:9–26. 10.1007/s11306-014-0707-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 983.Swainston N, Mendes P, Kell DB. An analysis of a ‘community-driven’ reconstruction of the human metabolic network. Metabolomics (2013) 9:757–64. 10.1007/s11306-013-0564-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 984.Thiele I, Swainston N, Fleming RMT, Hoppe A, Sahoo S, Aurich MK, et al. A community-driven global reconstruction of human metabolism. Nat Biotechnol (2013) 31:419–25. 10.1038/nbt.2488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 985.Swainston N, Smallbone K, Hefzi H, Dobson PD, Brewer J, Hanscho M, et al. Recon 2.2: from reconstruction to model of human metabolism. Metabolomics (2016) 12:109. 10.1007/s11306-016-1051-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 986.Herrgård MJ, Swainston N, Dobson P, Dunn WB, Arga KY, Arvas M, et al. A consensus yeast metabolic network obtained from a community approach to systems biology. Nat Biotechnol (2008) 26:1155–60. 10.1038/nbt1492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 987.Bolte AC, Van Geijn HP, Dekker GA. Pathophysiology of preeclampsia and the role of serotonin. Eur J Obstet Gynecol Reprod Biol (2001) 95:12–21. 10.1016/S0301-2115(00)00367-5 [DOI] [PubMed] [Google Scholar]
  • 988.Kell DB, Kaprelyants AS, Grafen A. On pheromones, social behaviour and the functions of secondary metabolism in bacteria. Trends Ecol Evol (1995) 10:126–9. 10.1016/S0169-5347(00)89013-8 [DOI] [PubMed] [Google Scholar]
  • 989.Takano E. Gamma-butyrolactones: Streptomyces signalling molecules regulating antibiotic production and differentiation. Curr Opin Microbiol (2006) 9:287–94. 10.1016/j.mib.2006.04.003 [DOI] [PubMed] [Google Scholar]
  • 990.Hyppönen E, Cavadino A, Williams D, Fraser A, Vereczkey A, Fraser WD, et al. Vitamin D and pre-eclampsia: original data, systematic review and meta-analysis. Ann Nutr Metab (2013) 63:331–40. 10.1159/000358338 [DOI] [PubMed] [Google Scholar]
  • 991.Tabesh M, Salehi-Abargouei A, Tabesh M, Esmaillzadeh A. Maternal vitamin D status and risk of pre-eclampsia: a systematic review and meta-analysis. J Clin Endocrinol Metab (2013) 98:3165–73. 10.1210/jc.2013-1257 [DOI] [PubMed] [Google Scholar]
  • 992.Bakacak M, Serin S, Ercan O, Kostu B, Avci F, Kilinc M, et al. Comparison of vitamin D levels in cases with preeclampsia, eclampsia and healthy pregnant women. Int J Clin Exp Med (2015) 8:16280–6. [PMC free article] [PubMed] [Google Scholar]
  • 993.Barrera D, Diaz L, Noyola-Martinez N, Halhali A. Vitamin D and inflammatory cytokines in healthy and preeclamptic pregnancies. Nutrients (2015) 7:6465–90. 10.3390/nu7085293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 994.Gargari BP, Tabrizi FPF, Sadien B, Jafarabadi MA, Farzadi L. Vitamin D status is related to oxidative stress but not high-sensitive C-reactive protein in women with pre-eclampsia. Gynecol Obstet Invest (2015) 81:308–14. 10.1159/000441781 [DOI] [PubMed] [Google Scholar]
  • 995.Harvey NC, Holroyd C, Ntani G, Javaid K, Cooper P, Moon R, et al. Vitamin D supplementation in pregnancy: a systematic review. Health Technol Assess (2014) 18:1–190. 10.3310/hta18450 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 996.Kiely ME, Zhang JY, Kinsella M, Khashan AS, Kenny LC. Vitamin D status is associated with utero-placental dysfunction in a large prospective pregnancy cohort with low 25(OH)D3 and ubiquitous 3-epi-25(OH)D3 and 25(OH)D2. Am J Clin Nutr (2016) 102:354–61. 10.3945/ajcn.116.130419 [DOI] [PubMed] [Google Scholar]
  • 997.van Weert B, Van Den Berg D, Hrudey EJ, Oostvogels AJJM, De Miranda E, Vrijkotte TGM. Is first trimester vitamin D status in nulliparous women associated with pregnancy related hypertensive disorders? Midwifery (2016) 34:–22. 10.1016/j.midw.2015.12.007 [DOI] [PubMed] [Google Scholar]
  • 998.Palacios C, De-Regil LM, Lombardo LK, Peña-Rosas JP. Vitamin D supplementation during pregnancy: updated meta-analysis on maternal outcomes. J Steroid Biochem Mol Biol (2016) 164:148–55. 10.1016/j.jsbmb.2016.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 999.Bahado-Singh RO, Syngelaki A, Akolekar R, Mandal R, Bjondahl TC, Han B, et al. Validation of metabolomic models for prediction of early-onset preeclampsia. Am J Obstet Gynecol (2015) 213:530.e1–10. 10.1016/j.ajog.2015.06.044 [DOI] [PubMed] [Google Scholar]
  • 1000.Melland-Smith M, Ermini L, Chauvin S, Craig-Barnes H, Tagliaferro A, Todros T, et al. Disruption of sphingolipid metabolism augments ceramide-induced autophagy in preeclampsia. Autophagy (2015) 11:653–69. 10.1080/15548627.2015.1034414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1001.Funke C, Schneider SA, Berg D, Kell DB. Genetics and iron in the systems biology of Parkinson’s disease and some related disorders. Neurochem Int (2013) 62:637–52. 10.1016/j.neuint.2012.11.015 [DOI] [PubMed] [Google Scholar]
  • 1002.Manyonda IT, Slater DM, Fenske C, Hole D, Choy MY, Wilson C. A role for noradrenaline in pre-eclampsia: towards a unifying hypothesis for the pathophysiology. Br J Obstet Gynaecol (1998) 105:641–8. 10.1111/j.1471-0528.1998.tb10179.x [DOI] [PubMed] [Google Scholar]
  • 1003.Ferreira-de-Almeida JA, Amenta F, Cardoso F, Polonia JJ. Association of circulating endothelium and noradrenaline with increased calcium-channel binding sites in the placental bed in pre-eclampsia. Br J Obstet Gynaecol (1998) 105:1104–12. 10.1111/j.1471-0528.1998.tb09944.x [DOI] [PubMed] [Google Scholar]
  • 1004.Lyte M, Ernst S. Catecholamine-induced growth of Gram-negative bacteria. Life Sci (1992) 50:203–12. 10.1016/0024-3205(92)90273-R [DOI] [PubMed] [Google Scholar]
  • 1005.Freestone PP, Haigh RD, Williams PH, Lyte M. Stimulation of bacterial growth by heat-stable, norepinephrine-induced autoinducers. FEMS Microbiol Lett (1999) 172:53–60. 10.1111/j.1574-6968.1999.tb13449.x [DOI] [PubMed] [Google Scholar]
  • 1006.Reissbrodt R, Rienaecker I, Romanova JM, Freestone PPE, Haigh RD, Lyte M, et al. Resuscitation of Salmonella enterica serovar typhimurium and enterohemorrhagic Escherichia coli from the viable but nonculturable state by heat-stable enterobacterial autoinducer. Appl Environ Microbiol (2002) 68:4788–94. 10.1128/AEM.68.10.4788-4794.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1007.Lyte M. Microbial endocrinology and infectious disease in the 21st century. Trends Microbiol (2004) 12:14–20. 10.1016/j.tim.2003.11.004 [DOI] [PubMed] [Google Scholar]
  • 1008.Lyte M. The effect of stress on microbial growth. Anim Health Res Rev (2014) 15:172–4. 10.1017/S146625231400019X [DOI] [PubMed] [Google Scholar]
  • 1009.Freestone PP, Lyte M, Neal CP, Maggs AF, Haigh RD, Williams PH. The mammalian neuroendocrine hormone norepinephrine supplies iron for bacterial growth in the presence of transferrin or lactoferrin. J Bacteriol (2000) 182:6091–8. 10.1128/JB.182.21.6091-6098.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1010.Freestone PPE, Haigh RD, Williams PH, Lyte M. Involvement of enterobactin in norepinephrine-mediated iron supply from transferrin to enterohaemorrhagic Escherichia coli. FEMS Microbiol Lett (2003) 222:39–43. 10.1016/S0378-1097(03)00243-X [DOI] [PubMed] [Google Scholar]
  • 1011.O’Donnell PM, Aviles H, Lyte M, Sonnenfeld G. Enhancement of in vitro growth of pathogenic bacteria by norepinephrine: importance of inoculum density and role of transferrin. Appl Environ Microbiol (2006) 72:5097–9. 10.1128/AEM.00075-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1012.Kvetnansky R, Sabban EL, Palkovits M. Catecholaminergic systems in stress: structural and molecular genetic approaches. Physiol Rev (2009) 89:535–606. 10.1152/physrev.00042.2006 [DOI] [PubMed] [Google Scholar]
  • 1013.Ribas Ripoll V, Romay E, Brunelli L, Pastorelli R, Goma G, Navas A, et al. Metabolite analysis in sepsis through conditional independence maps. Conf Proc IEEE Eng Med Biol Soc (2015) 2015:6477–80. 10.1109/EMBC.2015.7319876 [DOI] [PubMed] [Google Scholar]
  • 1014.Su L, Huang Y, Zhu Y, Xia L, Wang R, Xiao K, et al. Discrimination of sepsis stage metabolic profiles with an LC/MS-MS-based metabolomics approach. BMJ Open Respir Res (2014) 1:e000056. 10.1136/bmjresp-2014-000056 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1015.Eggers AE. A suggestion about the cause of inflammation in acute atherosis complicating poor placentation in preeclampsia. Med Hypotheses (2015) 85:718–9. 10.1016/j.mehy.2015.10.022 [DOI] [PubMed] [Google Scholar]
  • 1016.D’Anna R, Baviera G, Scilipoti A, Leonardi I, Leo R. The clinical utility of serum uric acid measurements in pre-eclampsia and transient hypertension in pregnancy. Panminerva Med (2000) 42:101–3. [PubMed] [Google Scholar]
  • 1017.Williams KP, Galerneau F. The role of serum uric acid as a prognostic indicator of the severity of maternal and fetal complications in hypertensive pregnancies. J Obstet Gynaecol Can (2002) 24:628–32. 10.1016/S1701-2163(16)30193-1 [DOI] [PubMed] [Google Scholar]
  • 1018.Roberts JM, Bodnar LM, Lain KY, Hubel CA, Markovic N, Ness RB, et al. Uric acid is as important as proteinuria in identifying fetal risk in women with gestational hypertension. Hypertension (2005) 46:1263–9. 10.1161/01.HYP.0000188703.27002.14 [DOI] [PubMed] [Google Scholar]
  • 1019.Bainbridge SA, Roberts JM. Uric acid as a pathogenic factor in preeclampsia. Placenta (2008) 29:S67–72. 10.1016/j.placenta.2007.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1020.Koopmans CM, Van Pampus MG, Groen H, Aarnoudse JG, Van Den Berg PP, Mol BWJ. Accuracy of serum uric acid as a predictive test for maternal complications in pre-eclampsia: bivariate meta-analysis and decision analysis. Eur J Obstet Gynecol Reprod Biol (2009) 146:8–14. 10.1016/j.ejogrb.2009.05.014 [DOI] [PubMed] [Google Scholar]
  • 1021.Agarwal V, Gupta BK, Vishnu A, Mamtatyagi, Shiprasolanki, Kiran J. Association of lipid profile and uric acid with pre-eclampsia of third trimester in nullipara women. J Clin Diagn Res (2014) 8:CC04–07. 10.7860/JCDR/2014/7901.4533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1022.Pereira KN, Knoppka CK, Da Silva JE. Association between uric acid and severity of pre-eclampsia. Clin Lab (2014) 60:309–14. [DOI] [PubMed] [Google Scholar]
  • 1023.Lim KH, Friedman SA, Ecker JL, Kao L, Kilpatrick SJ. The clinical utility of serum uric acid measurements in hypertensive diseases of pregnancy. Am J Obstet Gynecol (1998) 178:1067–71. 10.1016/S0002-9378(98)70549-6 [DOI] [PubMed] [Google Scholar]
  • 1024.Cnossen JS, De Ruyter-Hanhijarvi H, van der Post JA, Mol BW, Khan KS, Ter Riet G. Accuracy of serum uric acid determination in predicting pre-eclampsia: a systematic review. Acta Obstet Gynecol Scand (2006) 85:519–25. 10.1080/00016340500342037 [DOI] [PubMed] [Google Scholar]
  • 1025.Powers RW, Bodnar LM, Ness RB, Cooper KM, Gallaher MJ, Frank MP, et al. Uric acid concentrations in early pregnancy among preeclamptic women with gestational hyperuricemia at delivery. Am J Obstet Gynecol (2006) 194:160–6. 10.1016/j.ajog.2005.06.066 [DOI] [PubMed] [Google Scholar]
  • 1026.Thangaratinam S, Ismail KM, Sharp S, Coomarasamy A, Khan KS, Tests in Prediction of Pre-Eclampsia Severity Review, Group . Accuracy of serum uric acid in predicting complications of pre-eclampsia: a systematic review. BJOG (2006) 113:369–78. 10.1111/j.1471-0528.2006.00908.x [DOI] [PubMed] [Google Scholar]
  • 1027.Chen Q, Lau S, Tong M, Wei J, Shen F, Zhao J, et al. Serum uric acid may not be involved in the development of preeclampsia. J Hum Hypertens (2016) 30:136–40. 10.1038/jhh.2015.47 [DOI] [PubMed] [Google Scholar]
  • 1028.Eltzschig HK, Sitkovsky MV, Robson SC. Purinergic signaling during inflammation. N Engl J Med (2012) 367:2322–33. 10.1056/NEJMra1205750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1029.Kaddurah-Daouk R, Zhu H, Sharma S, Bogdanov M, Rozen SG, Matson W, et al. Alterations in metabolic pathways and networks in Alzheimer’s disease. Transl Psychiatry (2013) 3:e244. 10.1038/tp.2013.18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1030.McFarland NR, Burdett T, Desjardins CA, Frosch MP, Schwarzschild MA. Postmortem brain levels of urate and precursors in Parkinson’s disease and related disorders. Neurodegener Dis (2013) 12:189–98. 10.1159/000346370 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1031.Esteve C, Jones EA, Kell DB, Boutin H, Mcdonnell LA. Mass spectral imaging shows major derangements in neurogranin and in purine metabolism in the triple-knockout 3xTg Alzheimer-like mouse model. Front Neurosci (Forthcoming 2016). [DOI] [PubMed] [Google Scholar]
  • 1032.Martinon F. Mechanisms of uric acid crystal-mediated autoinflammation. Immunol Rev (2010) 233:218–32. 10.1111/j.0105-2896.2009.00860.x [DOI] [PubMed] [Google Scholar]
  • 1033.Lyngdoh T, Vuistiner P, Marques-Vidal P, Rousson V, Waeber G, Vollenweider P, et al. Serum uric acid and adiposity: deciphering causality using a bidirectional Mendelian randomization approach. PLoS One (2012) 7:e39321. 10.1371/journal.pone.0039321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1034.Orengo JM, Leliwa-Sytek A, Evans JE, Evans B, van de Hoef D, Nyako M, et al. Uric acid is a mediator of the Plasmodium falciparum-induced inflammatory response. PLoS One (2009) 4:e5194. 10.1371/Journal.Pone.0005194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1035.Kool M, Soullie T, Van Nimwegen M, Willart MA, Muskens F, Jung S, et al. Alum adjuvant boosts adaptive immunity by inducing uric acid and activating inflammatory dendritic cells. J Exp Med (2008) 205:869–82. 10.1084/jem.20071087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1036.Chalcraft KR. Comprehensive Metabolomics Analysis of Peanut Allergy and Peanut-Induced Anaphylaxis [Ph.D. Thesis] Open Access Dissertations and Theses. Paper 7733. Hamilton, ON: McMaster University; (2013). [Google Scholar]
  • 1037.Chalcraft KR, Kong J, Waserman S, Jordana M, Mccarry BE. Comprehensive metabolomic analysis of peanut-induced anaphylaxis in a murine model. Metabolomics (2014) 10:452–60. 10.1007/s11306-013-0589-7 [DOI] [Google Scholar]
  • 1038.Kong J, Chalcraft K, Mandur TS, Jimenez-Saiz R, Walker TD, Goncharova S, et al. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy (2015) 70:495–505. 10.1111/all.12579 [DOI] [PubMed] [Google Scholar]
  • 1039.van Deventer SJH, Büller HR, Ten Cate JW, Aarden LA, Hack CE, Sturk A. Experimental endotoxemia in humans: analysis of cytokine release and coagulation, fibrinolytic, and complement pathways. Blood (1990) 76:2520–6. [PubMed] [Google Scholar]
  • 1040.Johnson K, Aarden L, Choi Y, De Groot E, Creasey A. The proinflammatory cytokine response to coagulation and endotoxin in whole blood. Blood (1996) 87:5051–60. [PubMed] [Google Scholar]
  • 1041.American College of Chest Physicians. From the bench to the bedside: the future of sepsis research. Executive summary of an American College of Chest Physicians, National Institute of Allergy and Infectious Disease, and National Heart, Lung, and Blood Institute Workshop. Chest (1997) 111:744–53. [PubMed] [Google Scholar]
  • 1042.Pernerstorfer T, Stohlawetz P, Hollenstein U, Dzirlo L, Eichler HG, Kapiotis S, et al. Endotoxin-induced activation of the coagulation cascade in humans: effect of acetylsalicylic acid and acetaminophen. Arterioscler Thromb Vasc Biol (1999) 19:2517–23. 10.1161/01.ATV.19.10.2517 [DOI] [PubMed] [Google Scholar]
  • 1043.Pernerstorfer T, Hollenstein U, Hansen JB, Stohlawetz P, Eichler HG, Handler S, et al. Lepirudin blunts endotoxin-induced coagulation activation. Blood (2000) 95:1729–34. [PubMed] [Google Scholar]
  • 1044.Dellinger RP. Inflammation and coagulation: implications for the septic patient. Clin Infect Dis (2003) 36:1259–65. 10.1086/374835 [DOI] [PubMed] [Google Scholar]
  • 1045.Schouten M, Wiersinga WJ, Levi M, van der Poll T. Inflammation, endothelium, and coagulation in sepsis. J Leukoc Biol (2008) 83:536–45. 10.1189/jlb.0607373 [DOI] [PubMed] [Google Scholar]
  • 1046.Anas AA, Wiersinga WJ, De Vos AF, van der Poll T. Recent insights into the pathogenesis of bacterial sepsis. Neth J Med (2010) 68:147–52. [PubMed] [Google Scholar]
  • 1047.Saracco P, Vitale P, Scolfaro C, Pollio B, Pagliarino M, Timeus F. The coagulopathy in sepsis: significance and implications for treatment. Pediatr Rep (2011) 3:e30. 10.4081/pr.2011.e30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1048.Tsao CM, Ho ST, Wu CC. Coagulation abnormalities in sepsis. Acta Anaesthesiol Taiwan (2015) 53:16–22. 10.1016/j.aat.2014.11.002 [DOI] [PubMed] [Google Scholar]
  • 1049.Armstrong MT, Rickles FR, Armstrong PB. Capture of lipopolysaccharide (endotoxin) by the blood clot: a comparative study. PLoS One (2013) 8:e80192. 10.1371/journal.pone.0080192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1050.Al-ofi E, Coffelt SB, Anumba DO. Fibrinogen, an endogenous ligand of toll-like receptor 4, activates monocytes in pre-eclamptic patients. J Reprod Immunol (2014) 103:23–8. 10.1016/j.jri.2014.02.004 [DOI] [PubMed] [Google Scholar]
  • 1051.Chatterjee T, Maitra D, Chakravarty T, Datta AG. Studies on plasma fibrinogen level in pre-eclampsia and eclampsia. Experientia (1978) 34:562–3. 10.1007/BF01936958 [DOI] [PubMed] [Google Scholar]
  • 1052.Manten GTR, Sikkema JM, Franx A, Hameeteman TM, Visser GHA, De Groot PG, et al. Increased high molecular weight fibrinogen in pre-eclampsia. Thromb Res (2003) 111:143–7. 10.1016/j.thromres.2003.08.025 [DOI] [PubMed] [Google Scholar]
  • 1053.Üstün Y, Engin-Üstün Y, Kamacı M. Association of fibrinogen and C-reactive protein with severity of preeclampsia. Eur J Obstet Gynecol Reprod Biol (2005) 121:154–8. 10.1016/j.ejogrb.2004.12.009 [DOI] [PubMed] [Google Scholar]
  • 1054.Sersam LW. Inflammatory markers in pre-eclampsia and related conditions. Int J Gynaecol Obstet (2009):69–70. 10.1016/j.ijgo.2008.11.016 [DOI] [PubMed] [Google Scholar]
  • 1055.Sogani S, Sarakar PD. Evaluation of plasma fibrinogen and plasma fibrin degradation product (FDP) in preeclampsia. J Clin Biomed Sci (2013) 3:201–3. [Google Scholar]
  • 1056.Perry KG, Martin JN. Abnormal hemostasis and coagulopathy in preeclampsia and eclampsia. Clin Obstet Gynecol (1992) 35:338–50. 10.1097/00003081-199235020-00015 [DOI] [PubMed] [Google Scholar]
  • 1057.Metz J, Cincotta R, Francis M, Derosa L, Balloch A. Screening for consumptive coagulopathy in preeclampsia. Int J Gynecol Obstet (1994) 46:3–9. 10.1016/0020-7292(94)90301-8 [DOI] [PubMed] [Google Scholar]
  • 1058.Gilabert J, Estellés A, Grancha S, España F, Aznar J. Fibrinolytic system and reproductive process with special reference to fibrinolytic failure in pre-eclampsia. Hum Reprod (1995) 10(Suppl 2):121–31. 10.1093/humrep/10.suppl_2.121 [DOI] [PubMed] [Google Scholar]
  • 1059.He S, Bremme K, Blomback M. Acquired deficiency of antithrombin in association with a hypercoagulable state and impaired function of liver and/or kidney in preeclampsia. Blood Coagul Firbinolysis (1997) 8:232–8. 10.1097/00001721-199706000-00004 [DOI] [PubMed] [Google Scholar]
  • 1060.Hladunewich M, Karumanchi SA, Lafayette R. Pathophysiology of the clinical manifestations of preeclampsia. Clin J Am Soc Nephrol (2007) 2:543–9. 10.2215/CJN.03761106 [DOI] [PubMed] [Google Scholar]
  • 1061.Kher A, Bauersachs R, Nielsen JD. The management of thrombosis in pregnancy: role of low-molecular-weight heparin. Thromb Haemost (2007) 97:505–13. 10.1160/TH06-10-0606 [DOI] [PubMed] [Google Scholar]
  • 1062.Jahromi BN, Rafiee SH. Coagulation factors in severe preeclampsia. Iran Red Crescent Med J (2009) 11:321–4. [Google Scholar]
  • 1063.Ducloy-Bouthors AS. Clotting disorders and preeclampsia (in French). Ann Fr Anesth Reanim (2010) 29:e121–34. 10.1016/j.annfar.2010.03.012 [DOI] [PubMed] [Google Scholar]
  • 1064.Martínez-Zamora MA, Tassies D, Carmona F, Espinosa G, Cervera R, Reverter JC, et al. Clot lysis time and thrombin activatable fibrinolysis inhibitor in severe preeclampsia with or without associated antiphospholipid antibodies. J Reprod Immunol (2010) 86:133–40. 10.1016/j.jri.2010.05.002 [DOI] [PubMed] [Google Scholar]
  • 1065.Dusse LM, Rios DRA, Pinheiro MB, Cooper AJ, Lwaleed BA. Pre-eclampsia: relationship between coagulation, fibrinolysis and inflammation. Clin Chim Acta (2011) 412:17–21. 10.1016/j.cca.2010.09.030 [DOI] [PubMed] [Google Scholar]
  • 1066.Godoi LC, Gomes KB, Alpoim PN, Carvalho MDG, Lwaleed BA, Sant’ana Dusse LM. Preeclampsia: the role of tissue factor and tissue factor pathway inhibitor. J Thromb Thrombolysis (2012) 34:1–6. 10.1007/s11239-012-0708-8 [DOI] [PubMed] [Google Scholar]
  • 1067.Battinelli EM, Marshall A, Connors JM. The role of thrombophilia in pregnancy. Thrombosis (2013) 2013:516420. 10.1155/2013/516420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1068.Han L, Liu X, Li H, Zou J, Yang Z, Han J, et al. Blood coagulation parameters and platelet indices: changes in normal and preeclamptic pregnancies and predictive values for preeclampsia. PLoS One (2014) 9:e114488. 10.1371/journal.pone.0114488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1069.Koh SCL, Anandakumar C, Montan S, Ratnam SS. Plasminogen activators, plasminogen activator inhibitors and markers of intravascular coagulation in pre-eclampsia. Gynecol Obstet Invest (1993) 35:214–21. 10.1159/000292703 [DOI] [PubMed] [Google Scholar]
  • 1070.Kaneko T, Wada H. Diagnostic criteria and laboratory tests for disseminated intravascular coagulation. J Clin Exp Hematop (2011) 51:67–76. 10.3960/jslrt.51.67 [DOI] [PubMed] [Google Scholar]
  • 1071.Levi M, van der Poll T. Disseminated intravascular coagulation: a review for the internist. Int Emerg Med (2013) 8:23–32. 10.1007/s11739-012-0859-9 [DOI] [PubMed] [Google Scholar]
  • 1072.Asakura H. Classifying types of disseminated intravascular coagulation: clinical and animal models. J Intensive Care (2014) 2:20. 10.1186/2052-0492-2-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1073.Wada H, Matsumoto T, Yamashita Y, Hatada T. Disseminated intravascular coagulation: testing and diagnosis. Clin Chim Acta (2014) 436:130–4. 10.1016/j.cca.2014.04.020 [DOI] [PubMed] [Google Scholar]
  • 1074.Zeerleder S, Hack CE, Wuillemin WA. Disseminated intravascular coagulation in sepsis. Chest (2005) 128:2864–75. 10.1378/chest.128.4.2864 [DOI] [PubMed] [Google Scholar]
  • 1075.Xu M, Dong MQ, Cao FL, Liu ML, Wang YX, Dong HY, et al. Tanshinone IIA reduces lethality and acute lung injury in LPS-treated mice by inhibition of PLA2 activity. Eur J Pharmacol (2009) 607:194–200. 10.1016/j.ejphar.2009.02.003 [DOI] [PubMed] [Google Scholar]
  • 1076.Semeraro N, Ammollo CT, Semeraro F, Colucci M. Sepsis-associated disseminated intravascular coagulation and thromboembolic disease. Mediterr J Hematol Infect Dis (2010) 2:e2010024. 10.4084/MJHID.2010.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1077.Acikgoz S, Akduman D, Eskici ZM, Can M, Mungan G, Guven B, et al. Thrombocyte and erythrocyte indices in sepsis and disseminated intravascular coagulation. J Med Biochem (2012) 31:60–4. 10.2478/v10011-011-0043-1 [DOI] [Google Scholar]
  • 1078.Wu LC, Lin X, Sun H. Tanshinone IIA protects rabbits against LPS-induced disseminated intravascular coagulation (DIC). Acta Pharmacol Sin (2012) 33:1254–9. 10.1038/aps.2012.84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1079.Iba T, Ito T, Maruyama I, Jilma B, Brenner T, Muller MC, et al. Potential diagnostic markers for disseminated intravascular coagulation of sepsis. Blood Rev (2016) 30:149–55. 10.1016/j.blre.2015.10.002 [DOI] [PubMed] [Google Scholar]
  • 1080.Okamoto K, Tamura T, Sawatsubashi Y. Sepsis and disseminated intravascular coagulation. J Intensive Care (2016) 4:23. 10.1186/s40560-016-0149-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1081.D’Elia AV, Fabbro D, Driul L, Barillari G, Marchesoni D, Damante G. Plasminogen activator inhibitor-1 gene polymorphisms in pre-eclampsia. Semin Thromb Hemost (2011) 37:97–105. 10.1055/s-0030-1270334 [DOI] [PubMed] [Google Scholar]
  • 1082.Buurma AJ, Turner RJ, Driessen JH, Mooyaart AL, Schoones JW, Bruijn JA, et al. Genetic variants in pre-eclampsia: a meta-analysis. Hum Reprod Update (2013) 19:289–303. 10.1093/humupd/dms060 [DOI] [PubMed] [Google Scholar]
  • 1083.Morgan JA, Bombell S, Mcguire W. Association of plasminogen activator inhibitor-type 1 (-675 4G/5G) polymorphism with pre-eclampsia: systematic review. PLoS One (2013) 8:e56907. 10.1371/journal.pone.0056907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1084.Kell DB, Pretorius E. Proteins behaving badly. Substoichiometric molecular control and amplification of the initiation and nature of amyloid fibril formation: lessons from and for blood clotting. Progr Biophys Mol Biol (2016). 10.1016/j.pbiomolbio.2016.08.006 [DOI] [PubMed] [Google Scholar]
  • 1085.Kell DB, Pretorius E. Substoichiometric molecular control and amplification of the initiation and nature of amyloid fibril formation: lessons from and for blood clotting. bioRxiv preprint. bioRxiv (2016) 054734. 10.1101/054734 [DOI] [PubMed] [Google Scholar]
  • 1086.Akassoglou K, Adams RA, Bauer J, Mercado P, Tseveleki V, Lassmann H, et al. Fibrin depletion decreases inflammation and delays the onset of demyelination in a tumor necrosis factor transgenic mouse model for multiple sclerosis. Proc Natl Acad Sci U S A (2004) 101:6698–703. 10.1073/pnas.0303859101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1087.Levi M, van der Poll T, Buller HR. Bidirectional relation between inflammation and coagulation. Circulation (2004) 109:2698–704. 10.1161/01.CIR.0000131660.51520.9A [DOI] [PubMed] [Google Scholar]
  • 1088.Flick MJ, Lajeunesse CM, Talmage KE, Witte DP, Palumbo JS, Pinkerton MD, et al. Fibrin(ogen) exacerbates inflammatory joint disease through a mechanism linked to the integrin alphaMbeta2 binding motif. J Clin Invest (2007) 117:3224–35. 10.1172/JCI30134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1089.Jennewein C, Paulus P, Zacharowski K. Linking inflammation and coagulation: novel drug targets to treat organ ischemia. Curr Opin Anaesthesiol (2011) 24:375–80. 10.1097/ACO.0b013e3283489ac0 [DOI] [PubMed] [Google Scholar]
  • 1090.Jennewein C, Tran N, Paulus P, Ellinghaus P, Eble JA, Zacharowski K. Novel aspects of fibrin(ogen) fragments during inflammation. Mol Med (2011) 17:568–73. 10.2119/molmed.2010.00146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1091.Schuliga M. The inflammatory actions of coagulant and fibrinolytic proteases in disease. Mediators Inflamm (2015) 2015:437695. 10.1155/2015/437695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1092.Miranda S, Opazo C, Larrondo LF, Munoz FJ, Ruiz F, Leighton F, et al. The role of oxidative stress in the toxicity induced by amyloid beta-peptide in Alzheimer’s disease. Progr Neurobiol (2000) 62:633–48. 10.1016/S0301-0082(00)00015-0 [DOI] [PubMed] [Google Scholar]
  • 1093.Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia-Alloza M, De Calignon A, Rozkalne A, et al. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer’s disease. Nature (2008) 451:720–4. 10.1038/nature06616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1094.Rival T, Page RM, Chandraratna DS, Sendall TJ, Ryder E, Liu B, et al. Fenton chemistry and oxidative stress mediate the toxicity of the beta-amyloid peptide in a Drosophila model of Alzheimer’s disease. Eur J Neurosci (2009) 29:1335–47. 10.1111/j.1460-9568.2009.06701.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1095.Liu B, Moloney A, Meehan S, Morris K, Thomas SE, Serpell LC, et al. Iron promotes the toxicity of amyloid beta peptide by impeding its ordered aggregation. J Biol Chem (2011) 286:4248–56. 10.1074/jbc.M110.158980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1096.Minter MR, Taylor JM, Crack PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J Neurochem (2016) 136:457–74. 10.1111/jnc.13411 [DOI] [PubMed] [Google Scholar]
  • 1097.Özcan F, Turak O, Durak A, Işleyen A, Uçar F, Giniş Z, et al. Red cell distribution width and inflammation in patients with non-dipper hypertension. Blood Press (2013) 22:80–5. 10.3109/08037051.2012.707336 [DOI] [PubMed] [Google Scholar]
  • 1098.Heilmann L, Mattheck C, Kurz E. Changes in blood rheology and their influence on oxygen diffusion in normal and pathological pregnancies. Arch Gynäkol (1977) 223:283–98. 10.1007/Bf00667368 [DOI] [PubMed] [Google Scholar]
  • 1099.Stoeff S, Dikov I, Vretenarska M, Jovtchev S, Trifonova N, Penev M. Quantitative utility assessment of some biophysical and rheological tests used in hypertension research. 1. Relevance analyses of the test parameters. C R Acad Bulg Sci (2003) 56:95–100. [Google Scholar]
  • 1100.Kurt RK, Aras Z, Silfeler DB, Kunt C, Islimye M, Kosar O. Relationship of red cell distribution width with the presence and severity of preeclampsia. Clin Appl Thromb Hemost (2015) 21:128–31. 10.1177/1076029613490827 [DOI] [PubMed] [Google Scholar]
  • 1101.Abdullahi H, Osman A, Rayis DA, Gasim GI, Imam AM, Adam I. Red blood cell distribution width is not correlated with preeclampsia among pregnant Sudanese women. Diagn Pathol (2014) 9:29. 10.1186/1746-1596-9-29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1102.McMahon CJ, Hopkins S, Vail A, King AT, Smith D, Illingworth KJ, et al. Inflammation as a predictor for delayed cerebral ischemia after aneurysmal subarachnoid haemorrhage. J Neurointerv Surg (2013) 5:512–7. 10.1136/neurintsurg-2012-010386 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1103.Swanepoel AC, Pretorius E. Scanning electron microscopy analysis of erythrocytes in thromboembolic ischemic stroke. Int J Lab Hematol (2012) 34:185–91. 10.1111/j.1751-553X.2011.01379.x [DOI] [PubMed] [Google Scholar]
  • 1104.Buys AV, Van Rooy MJ, Soma P, Van Papendorp D, Lipinski B, Pretorius E. Changes in red blood cell membrane structure in type 2 diabetes: a scanning electron and atomic force microscopy study. Cardiovasc Diabetol (2013) 12:25. 10.1186/1475-2840-12-25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1105.Pretorius E, Lipinski B. Iron alters red blood cell morphology. Blood (2013) 121:9. 10.1182/blood-2012-09-454793 [DOI] [PubMed] [Google Scholar]
  • 1106.Pretorius E, Lipinski B. Thromboembolic ischemic stroke changes red blood cell morphology. Cardiovasc Pathol (2013) 22:241–2. 10.1016/j.carpath.2012.11.005 [DOI] [PubMed] [Google Scholar]
  • 1107.Lang F, Lang KS, Lang PA, Huber SM, Wieder T. Mechanisms and significance of eryptosis. Antioxid Redox Signal (2006) 8:1183–92. 10.1089/ars.2006.8.1183 [DOI] [PubMed] [Google Scholar]
  • 1108.Lang F, Gulbins E, Lerche H, Huber SM, Kempe DS, Föller M. Eryptosis, a window to systemic disease. Cell Physiol Biochem (2008) 22:373–80. 10.1159/000185448 [DOI] [PubMed] [Google Scholar]
  • 1109.Lang F, Lang E, Foller M. Physiology and pathophysiology of eryptosis. Transfus Med Hemother (2012) 39:308–14. 10.1159/000342534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1110.Lang F, Qadri SM. Mechanisms and significance of eryptosis, the suicidal death of erythrocytes. Blood Purif (2012) 33:125–30. 10.1159/000334163 [DOI] [PubMed] [Google Scholar]
  • 1111.Lang E, Lang F. Triggers, inhibitors, mechanisms, and significance of eryptosis: the suicidal erythrocyte death. Biomed Res Int (2015) 2015:513518. 10.1155/2015/513518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1112.Lang E, Zelenak C, Eberhard M, Bissinger R, Rotte A, Ghashghaeinia M, et al. Impact of cyclin-dependent kinase CDK4 inhibition on eryptosis. Cell Physiol Biochem (2015) 37:1178–86. 10.1159/000430241 [DOI] [PubMed] [Google Scholar]
  • 1113.Qadri SM, Donkor DA, Bhakta V, Eltringham-Smith LJ, Dwivedi DJ, Moore JC, et al. Phosphatidylserine externalization and procoagulant activation of erythrocytes induced by Pseudomonas aeruginosa virulence factor pyocyanin. J Cell Mol Med (2016) 20:710–20. 10.1111/jcmm.12778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1114.Lang E, Bissinger R, Gulbins E, Lang F. Ceramide in the regulation of eryptosis, the suicidal erythrocyte death. Apoptosis (2015) 20:758–67. 10.1007/s10495-015-1094-4 [DOI] [PubMed] [Google Scholar]
  • 1115.Hansson SR, Nääv Å, Erlandsson L. Oxidative stress in preeclampsia and the role of free fetal hemoglobin. Front Physiol (2014) 5:516. 10.3389/fphys.2014.00516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1116.Askie LM, Duley L, Henderson-Smart DJ, Stewart LA, Paris Collaborative Group . Antiplatelet agents for prevention of pre-eclampsia: a meta-analysis of individual patient data. Lancet (2007) 369:1791–8. 10.1016/S0140-6736(07)60712-0 [DOI] [PubMed] [Google Scholar]
  • 1117.Roberts JM, Catov JM. Aspirin for pre-eclampsia: compelling data on benefit and risk. Lancet (2007) 369:1765–6. 10.1016/S0140-6736(07)60713-2 [DOI] [PubMed] [Google Scholar]
  • 1118.Rossi AC, Mullin PM. Prevention of pre-eclampsia with low-dose aspirin or vitamins C and E in women at high or low risk: a systematic review with meta-analysis. Eur J Obstet Gynecol Reprod Biol (2011) 158:9–16. 10.1016/j.ejogrb.2011.04.010 [DOI] [PubMed] [Google Scholar]
  • 1119.Villa PM, Kajantie E, Räikkönen K, Pesonen AK, Hämäläinen E, Vainio M, et al. Aspirin in the prevention of pre-eclampsia in high-risk women: a randomised placebo-controlled PREDO trial and a meta-analysis of randomised trials. BJOG (2013) 120:64–74. 10.1111/j.1471-0528.2012.03493.x [DOI] [PubMed] [Google Scholar]
  • 1120.Bujold E, Morency AM, Roberge S, Lacasse Y, Forest JC, Giguère Y. Acetylsalicylic acid for the prevention of preeclampsia and intra-uterine growth restriction in women with abnormal uterine artery Doppler: a systematic review and meta-analysis. J Obstet Gynaecol Can (2009) 31:818–26. 10.1016/S1701-2163(16)34300-6 [DOI] [PubMed] [Google Scholar]
  • 1121.Roberge S, Giguere Y, Villa P, Nicolaides K, Vainio M, Forest JC, et al. Early administration of low-dose aspirin for the prevention of severe and mild preeclampsia: a systematic review and meta-analysis. Am J Perinatol (2012) 29:551–6. 10.1055/s-0032-1310527 [DOI] [PubMed] [Google Scholar]
  • 1122.Roberge S, Demers S, Bujold E. Initiation of aspirin in early gestation for the prevention of pre-eclampsia. BJOG (2013) 120:773–4. 10.1111/1471-0528.12170 [DOI] [PubMed] [Google Scholar]
  • 1123.Bergeron TS, Roberge S, Carpentier C, Sibai B, Mccaw-Binns A, Bujold E. Prevention of preeclampsia with aspirin in multiple gestations: a systematic review and meta-analysis. Am J Perinatol (2016) 33:605–10. 10.1055/s-0035-1570381 [DOI] [PubMed] [Google Scholar]
  • 1124.Roberge S, Sibai B, McCaw-Binns A, Bujold E. Low-dose aspirin in early gestation for prevention of preeclampsia and small-for-gestational-age neonates: meta-analysis of large randomized trials. Am J Perinatol (2016) 33:781–5. 10.1055/s-0036-1572495 [DOI] [PubMed] [Google Scholar]
  • 1125.Hofmeyr GJ, Belizán JM, Von Dadelszen P, Calcium Pre-Eclampsia Study Group . Low-dose calcium supplementation for preventing pre-eclampsia: a systematic review and commentary. BJOG (2014) 121:951–7. 10.1111/1471-0528.12613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1126.Leslie K, Thilaganathan B, Papageorghiou A. Early prediction and prevention of pre-eclampsia. Best Pract Res Clin Obstet Gynaecol (2011) 25:343–54. 10.1016/j.bpobgyn.2011.01.002 [DOI] [PubMed] [Google Scholar]
  • 1127.Thangaratinam S, Langenveld J, Mol BW, Khan KS. Prediction and primary prevention of pre-eclampsia. Best Pract Res Clin Obstet Gynaecol (2011) 25:419–33. 10.1016/j.bpobgyn.2011.02.008 [DOI] [PubMed] [Google Scholar]
  • 1128.Gillon TE, Pels A, Von Dadelszen P, Macdonell K, Magee LA. Hypertensive disorders of pregnancy: a systematic review of international clinical practice guidelines. PLoS One (2014) 9:e113715. 10.1371/journal.pone.0113715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1129.Cottrell EC, Sibley CP. From pre-clinical studies to clinical trials: generation of novel therapies for pregnancy complications. Int J Mol Sci (2015) 16:12907–24. 10.3390/ijms160612907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1130.Altman D, Carroli G, Duley L, Farrell B, Moodley J, Neilson J, et al. Do women with pre-eclampsia, and their babies, benefit from magnesium sulphate? The magpie trial: a randomised placebo-controlled trial. Lancet (2002) 359:1877–90. 10.1016/S0140-6736(02)08778-0 [DOI] [PubMed] [Google Scholar]
  • 1131.Duley L. Evidence and practice: the magnesium sulphate story. Best Pract Res Clin Obstet Gynaecol (2005) 19:57–74. 10.1016/j.bpobgyn.2004.10.010 [DOI] [PubMed] [Google Scholar]
  • 1132.Pratt JJ, Niedle PS, Vogel JP, Oladapo OT, Bohren M, Tunçalp Ö, et al. Alternative regimens of magnesium sulfate for treatment of preeclampsia and eclampsia: a systematic review of non-randomized studies. Acta Obstet Gynecol Scand (2016) 95:144–56. 10.1111/aogs.12807 [DOI] [PubMed] [Google Scholar]
  • 1133.Williamson RD, McCarthy C, Kenny LC, O’Keeffe GW. Magnesium sulphate prevents lipopolysaccharide-induced cell death in an in vitro model of the human placenta. Pregnancy Hypertens (2016). 10.1016/j.preghy.2016.08.237 [DOI] [PubMed] [Google Scholar]
  • 1134.Koren G, Pastuszak A, Ito S. Drugs in pregnancy. N Engl J Med (1998) 338:1128–37. 10.1056/NEJM199804163381607 [DOI] [PubMed] [Google Scholar]
  • 1135.Everett TR, Wilkinson IB, Lees CC. Drug development in preeclampsia: a ‘no go’ area? J Matern Fetal Neonatal Med (2012) 25:50–2. 10.3109/14767058.2011.557791 [DOI] [PubMed] [Google Scholar]
  • 1136.Martinez de Tejada B. Antibiotic use and misuse during pregnancy and delivery: benefits and risks. Int J Environ Res Public Health (2014) 11:7993–8009. 10.3390/ijerph110807993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1137.Thinkhamrop J, Hofmeyr GJ, Adetoro O, Lumbiganon P, Ota E. Antibiotic prophylaxis during the second and third trimester to reduce adverse pregnancy outcomes and morbidity. Cochrane Database Syst Rev (2015) 1:CD002250. 10.1002/14651858.CD002250.pub2 [DOI] [PubMed] [Google Scholar]
  • 1138.Vazquez JC, Abalos E. Treatments for symptomatic urinary tract infections during pregnancy. Cochrane Database Syst Rev (2011) CD002256. 10.1002/14651858.CD002256.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1139.Mathew D, Khan K, Thornton JG, Todros T. Antibiotics for preventing hypertensive diseases in pregnancy (protocol). Cochrane Database Syst Rev (2007) CD006841. 10.1002/14651858.CD006841 [DOI] [Google Scholar]
  • 1140.Pound MW, May DB. Proposed mechanisms and preventative options of Jarisch-Herxheimer reactions. J Clin Pharm Ther (2005) 30:291–5. 10.1111/j.1365-2710.2005.00631.x [DOI] [PubMed] [Google Scholar]
  • 1141.See S, Scott EK, Levin MW. Penicillin-induced Jarisch-Herxheimer reaction. Ann Pharmacother (2005) 39:2128–30. 10.1345/aph.1G308 [DOI] [PubMed] [Google Scholar]
  • 1142.Belum GR, Belum VR, Chaitanya Arudra SK, Reddy BS. The Jarisch-Herxheimer reaction: revisited. Travel Med Infect Dis (2013) 11:231–7. 10.1016/j.tmaid.2013.04.001 [DOI] [PubMed] [Google Scholar]
  • 1143.Guerrier G, D’Ortenzio E. The Jarisch-Herxheimer reaction in leptospirosis: a systematic review. PLoS One (2013) 8:e59266. 10.1371/journal.pone.0059266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1144.Prins JM, van Deventer SJH, Kuijper EJ, Speelman P. Clinical relevance of antibiotic-induced endotoxin release. Antimicrob Agents Chemother (1994) 38:1211–8. 10.1128/AAC.38.6.1211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1145.Kirikae T, Nakano M, Morrison DC. Antibiotic-induced endotoxin release from bacteria and its clinical significance. Microbiol Immunol (1997) 41:285–94. 10.1111/j.1348-0421.1997.tb01203.x [DOI] [PubMed] [Google Scholar]
  • 1146.Holzheimer RG. Antibiotic induced endotoxin release and clinical sepsis: a review. J Chemother (2001) 13(Spec No 1):159–72. 10.1179/joc.2001.13.Supplement-2.159 [DOI] [PubMed] [Google Scholar]
  • 1147.Lepper PM, Held TK, Schneider EM, Bölke E, Gerlach H, Trautmann M. Clinical implications of antibiotic-induced endotoxin release in septic shock. Intensive Care Med (2002) 28:824–33. 10.1007/s00134-002-1330-6 [DOI] [PubMed] [Google Scholar]
  • 1148.Andersson JA, Fitts EC, Kirtley ML, Ponnusamy D, Peniche AG, Dann SM, et al. New role for FDA-approved drugs in combating antibiotic-resistant bacteria. Antimicrob Agents Chemother (2016) 60:3717–29. 10.1128/AAC.00326-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1149.Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH. Vitamin C and vitamin E in pregnant women at risk for pre-eclampsia (VIP trial): randomised placebo-controlled trial. Lancet (2006) 367:1145–54. 10.1016/S0140-6736(06)68433-X [DOI] [PubMed] [Google Scholar]
  • 1150.Ly C, Yockell-Lelievre J, Ferraro ZM, Arnason JT, Ferrier J, Gruslin A. The effects of dietary polyphenols on reproductive health and early development. Hum Reprod Update (2015) 21:228–48. 10.1093/humupd/dmu058 [DOI] [PubMed] [Google Scholar]
  • 1151.Liang ST, Wong VC, So WW, Ma HK, Chan V, Todd D. Homozygous alpha-thalassaemia: clinical presentation, diagnosis and management. A review of 46 cases. Br J Obstet Gynaecol (1985) 92:680–4. 10.1111/j.1471-0528.1985.tb01447.x [DOI] [PubMed] [Google Scholar]
  • 1152.Tungwiwat W, Fucharoen S, Fucharoen G, Ratanasiri T, Sanchaisuriya K. Development and application of a real-time quantitative PCR for prenatal detection of fetal alpha(0)-thalassemia from maternal plasma. Ann N Y Acad Sci (2006) 1075:103–7. 10.1196/annals.1368.013 [DOI] [PubMed] [Google Scholar]
  • 1153.Senden IP, De Groot CJ, Steegers EA, Bertina RM, Swinkels DW. Preeclampsia and the C282Y mutation in the hemochromatosis (HFE) gene. Clin Chem (2004) 50:973–4. 10.1373/clinchem.2004.031591 [DOI] [PubMed] [Google Scholar]
  • 1154.Perron NR, Brumaghim JL. A review of the antioxidant mechanisms of polyphenol compounds related to iron binding. Cell Biochem Biophys (2009) 53:75–100. 10.1007/s12013-009-9043-x [DOI] [PubMed] [Google Scholar]
  • 1155.Dodd JM, Mcleod A, Windrim RC, Kingdom J. Antithrombotic therapy for improving maternal or infant health outcomes in women considered at risk of placental dysfunction. Cochrane Database Syst Rev (2010) CD006780. 10.1002/14651858.CD006780.pub2 [DOI] [PubMed] [Google Scholar]
  • 1156.Li Y, Wu Y, Gong X, Shi X, Qiao F, Liu H. Low molecular weight heparin decreases the permeability of glomerular endothelial cells when exposed to pre-eclampsia serum in vitro. Nephrology (Carlton) (2012) 17:754–9. 10.1111/j.1440-1797.2012.01657.x [DOI] [PubMed] [Google Scholar]
  • 1157.Dodd JM, Mcleod A, Windrim RC, Kingdom J. Antithrombotic therapy for improving maternal or infant health outcomes in women considered at risk of placental dysfunction. Cochrane Database Syst Rev (2013) 7:CD006780. 10.1002/14651858.CD006780.pub3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1158.Zhang Y, Liu F, Chen S, Zhong M. Low-molecular-weight heparin protects kidney through an anti-apoptotic mechanism in a rat pre-eclamptic model. Eur J Obstet Gynecol Reprod Biol (2015) 188:51–5. 10.1016/j.ejogrb.2015.02.001 [DOI] [PubMed] [Google Scholar]
  • 1159.Darmochwał-Kolarz D, Kolarz B, Korzeniewski M, Kimber-Trojnar Z, Patro-Malysza J, Mierzynski R, et al. A prevention of pre-eclampsia with the use of acetylsalicylic acid and low-molecular weight heparin – molecular mechanisms. Curr Pharm Biotechnol (2016) 17:624–8. 10.2174/1389201017666160301103312 [DOI] [PubMed] [Google Scholar]
  • 1160.Roberge S, Demers S, Nicolaides KH, Bureau M, Côté S, Bujold E. Prevention of pre-eclampsia by low-molecular-weight heparin in addition to aspirin: a meta-analysis. Ultrasound Obstet Gynecol (2016) 47:548–53. 10.1002/uog.15789 [DOI] [PubMed] [Google Scholar]
  • 1161.Eggers AE. A hypothesis about how to achieve anticoagulation without bleeding. Med Hypotheses (2015) 85:720–2. 10.1016/j.mehy.2015.10.021 [DOI] [PubMed] [Google Scholar]
  • 1162.Kell DB. How drugs pass through biological cell membranes – a paradigm shift in our understanding? Beilstein Mag (2016) 2 Available from: http://www.beilstein-institut.de/download/628/609_kell.pdf [Google Scholar]
  • 1163.Park K, Lee S, Ahn HS, Kim D. Predicting the multi-modal binding propensity of small molecules: towards an understanding of drug promiscuity. Mol Biosyst (2009) 5:844–53. 10.1039/b901356c [DOI] [PubMed] [Google Scholar]
  • 1164.Pérez-Nueno VI, Venkatraman V, Mavridis L, Ritchie DW. Detecting drug promiscuity using Gaussian ensemble screening. J Chem Inf Model (2012) 52:1948–61. 10.1021/ci3000979 [DOI] [PubMed] [Google Scholar]
  • 1165.Tarcsay A, Keserü GM. Contributions of molecular properties to drug promiscuity. J Med Chem (2013) 56:1789–95. 10.1021/jm301514n [DOI] [PubMed] [Google Scholar]
  • 1166.Arrowsmith CH, Audia JE, Austin C, Baell J, Bennett J, Blagg J, et al. The promise and peril of chemical probes. Nat Chem Biol (2015) 11:536–41. 10.1038/nchembio.1867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1167.Niphakis MJ, Lum KM, Cognetta AB, Correia BE, Ichu TA, Olucha J, et al. A global map of lipid-binding proteins and their ligandability in cells. Cell (2015) 161:1668–80. 10.1016/j.cell.2015.05.045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1168.Mestres J, Gregori-Puigjané E, Valverde S, Solé RV. The topology of drug-target interaction networks: implicit dependence on drug properties and target families. Mol Biosyst (2009) 5:1051–7. 10.1039/b905821b [DOI] [PubMed] [Google Scholar]
  • 1169.Hopkins AL. Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol (2008) 4:682–90. 10.1038/nchembio.118 [DOI] [PubMed] [Google Scholar]
  • 1170.Berger SI, Iyengar R. Network analyses in systems pharmacology. Bioinformatics (2009) 25:2466–72. 10.1093/bioinformatics/btp465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1171.van der Graaf PH, Benson N. Systems pharmacology: bridging systems biology and pharmacokinetics-pharmacodynamics (PKPD) in drug discovery and development. Pharm Res (2011) 28:1460–4. 10.1007/s11095-011-0467-9 [DOI] [PubMed] [Google Scholar]
  • 1172.Cucurull-Sanchez L, Spink KG, Moschos SA. Relevance of systems pharmacology in drug discovery. Drug Discov Today (2012) 17:665–70. 10.1016/j.drudis.2012.01.015 [DOI] [PubMed] [Google Scholar]
  • 1173.Prentice RL, Langer RD, Stefanick ML, Howard BV, Pettinger M, Anderson GL, et al. Combined analysis of women’s health initiative observational and clinical trial data on postmenopausal hormone treatment and cardiovascular disease. Am J Epidemiol (2006) 163:589–99. 10.1093/aje/kwj079 [DOI] [PubMed] [Google Scholar]
  • 1174.Couzin J. Cholesterol veers off script. Science (2008) 322:220–3. 10.1126/science.322.5899.220 [DOI] [PubMed] [Google Scholar]
  • 1175.Peterson RT. Chemical biology and the limits of reductionism. Nat Chem Biol (2008) 4:635–8. 10.1038/nchembio1108-635 [DOI] [PubMed] [Google Scholar]
  • 1176.Robinson JG. Models for describing relations among the various statin drugs, low-density lipoprotein cholesterol lowering, pleiotropic effects, and cardiovascular risk. Am J Cardiol (2008) 101:1009–15. 10.1016/j.amjcard.2007.11.060 [DOI] [PubMed] [Google Scholar]
  • 1177.Endo A. A historical perspective on the discovery of statins. Proc Jpn Acad Ser B Phys Biol Sci (2010) 86:484–93. 10.2183/pjab.86.484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1178.Wagner BK, Kitami T, Gilbert TJ, Peck D, Ramanathan A, Schreiber SL, et al. Large-scale chemical dissection of mitochondrial function. Nat Biotechnol (2008) 26:343–51. 10.1038/nbt1387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1179.Libby P, Aikawa M. Stabilization of atherosclerotic plaques: new mechanisms and clinical targets. Nat Med (2002) 8:1257–62. 10.1038/nm1102-1257 [DOI] [PubMed] [Google Scholar]
  • 1180.Liao JK. Beyond lipid lowering: the role of statins in vascular protection. Int J Cardiol (2002) 86:5–18. 10.1016/S0167-5273(02)00195-X [DOI] [PubMed] [Google Scholar]
  • 1181.Undas A, Brozek J, Musial J. Anti-inflammatory and antithrombotic effects of statins in the management of coronary artery disease. Clin Lab (2002) 48:287–96. [PubMed] [Google Scholar]
  • 1182.Weitz-Schmidt G. Statins as anti-inflammatory agents. Trends Pharmacol Sci (2002) 23:482–6. 10.1016/S0165-6147(02)02077-1 [DOI] [PubMed] [Google Scholar]
  • 1183.Blanco-Colio LM, Tuñon J, Martin-Ventura JL, Egido J. Anti-inflammatory and immunomodulatory effects of statins. Kidney Int (2003) 63:12–23. 10.1046/j.1523-1755.2003.00744.x [DOI] [PubMed] [Google Scholar]
  • 1184.Kwak BR, Mulhaupt F, Mach F. Atherosclerosis: anti-inflammatory and immunomodulatory activities of statins. Autoimmun Rev (2003) 2:332–8. 10.1016/S1568-9972(03)00049-1 [DOI] [PubMed] [Google Scholar]
  • 1185.Steffens S, Mach F. Anti-inflammatory properties of statins. Semin Vasc Med (2004) 4:417–22. 10.1055/s-2004-869599 [DOI] [PubMed] [Google Scholar]
  • 1186.Jain MK, Ridker PM. Anti-inflammatory effects of statins: clinical evidence and basic mechanisms. Nat Rev Drug Discov (2005) 4:977–87. 10.1038/nrd1901 [DOI] [PubMed] [Google Scholar]
  • 1187.Abeles AM, Pillinger MH. Statins as antiinflammatory and immunomodulatory agents: a future in rheumatologic therapy? Arthritis Rheum (2006) 54:393–407. 10.1002/art.21521 [DOI] [PubMed] [Google Scholar]
  • 1188.Endres M. Statins: potential new indications in inflammatory conditions. Atheroscler Suppl (2006) 7:31–5. 10.1016/j.atherosclerosissup.2006.01.005 [DOI] [PubMed] [Google Scholar]
  • 1189.Li JJ, Zheng X, Li J. Statins may be beneficial for patients with slow coronary flow syndrome due to its anti-inflammatory property. Med Hypotheses (2007) 69:333–7. 10.1016/j.mehy.2006.09.070 [DOI] [PubMed] [Google Scholar]
  • 1190.Mira E, Manes S. Immunomodulatory and anti-inflammatory activities of statins. Endocr Metab Immune Disord Drug Targets (2009) 9:237–47. 10.2174/187153009789044383 [DOI] [PubMed] [Google Scholar]
  • 1191.Dinarello CA. Anti-inflammatory agents: present and future. Cell (2010) 140:935–50. 10.1016/j.cell.2010.02.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1192.Bu DX, Griffin G, Lichtman AH. Mechanisms for the anti-inflammatory effects of statins. Curr Opin Lipidol (2011) 22:165–70. 10.1097/MOL.0b013e3283453e41 [DOI] [PubMed] [Google Scholar]
  • 1193.Antonopoulos AS, Margaritis M, Lee R, Channon K, Antoniades C. Statins as anti-inflammatory agents in atherogenesis: molecular mechanisms and lessons from the recent clinical trials. Curr Pharm Des (2012) 18:1519–30. 10.2174/138161212799504803 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1194.Liappis AP, Kan VL, Rochester CG, Simon GL. The effect of statins on mortality in patients with bacteremia. Clin Infect Dis (2001) 33:1352–7. 10.1086/323334 [DOI] [PubMed] [Google Scholar]
  • 1195.Terblanche M, Almog Y, Rosenson RS, Smith TS, Hackam DG. Statins: panacea for sepsis? Lancet Infect Dis (2006) 6:242–8. 10.1016/S1473-3099(06)70439-X [DOI] [PubMed] [Google Scholar]
  • 1196.Falagas ME, Makris GC, Matthaiou DK, Rafailidis PI. Statins for infection and sepsis: a systematic review of the clinical evidence. J Antimicrob Chemother (2008) 61:774–85. 10.1093/jac/dkn019 [DOI] [PubMed] [Google Scholar]
  • 1197.Sun HY, Singh N. Antimicrobial and immunomodulatory attributes of statins: relevance in solid-organ transplant recipients. Clin Infect Dis (2009) 48:745–55. 10.1086/597039 [DOI] [PubMed] [Google Scholar]
  • 1198.Kozarov E, Padro T, Badimon L. View of statins as antimicrobials in cardiovascular risk modification. Cardiovasc Res (2014) 102:362–74. 10.1093/cvr/cvu058 [DOI] [PubMed] [Google Scholar]
  • 1199.Sandek A, Utchill S, Rauchhaus M. The endotoxin-lipoprotein hypothesis – an update. Arch Med Sci (2007) 3:S81–90. [Google Scholar]
  • 1200.Ukinc K, Ersoz HO, Erem C, Hacihasanoglu AB, Karti SS. Effects of one year simvastatin and atorvastatin treatments on acute phase reactants in uncontrolled type 2 diabetic patients. Endocrine (2009) 35:380–8. 10.1007/s12020-009-9157-3 [DOI] [PubMed] [Google Scholar]
  • 1201.Mascitelli L, Goldstein MR. Might the beneficial effects of statin drugs be related to their action on iron metabolism? QJM (2012) 105:1225–9. 10.1093/qjmed/hcs204 [DOI] [PubMed] [Google Scholar]
  • 1202.Kanugula AK, Gollavilli PN, Vasamsetti SB, Karnewar S, Gopoju R, Ummanni R, et al. Statin-induced inhibition of breast cancer proliferation and invasion involves attenuation of iron transport: intermediacy of nitric oxide and antioxidant defence mechanisms. FEBS J (2014) 281:3719–38. 10.1111/febs.12893 [DOI] [PubMed] [Google Scholar]
  • 1203.Lecarpentier E, Morel O, Fournier T, Elefant E, Chavatte-Palmer P, Tsatsaris V. Statins and pregnancy: between supposed risks and theoretical benefits. Drugs (2012) 72:773–88. 10.2165/11632010-000000000-00000 [DOI] [PubMed] [Google Scholar]
  • 1204.Costantine MM, Cleary K, Eunice Kennedy Shriver National Institute of Child, Health, Human Development Obstetric – Fetal Pharmacology Research Units Network . Pravastatin for the prevention of preeclampsia in high-risk pregnant women. Obstet Gynecol (2013) 121:349–53. 10.1097/AOG.0b013e31827d8ad5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1205.Downing JW, Baysinger CL, Johnson RF, Paschall RL. Review: potential druggable targets for the treatment of early onset preeclampsia. Pregnancy Hypertens (2013) 3:203–10. 10.1016/j.preghy.2013.04.120 [DOI] [PubMed] [Google Scholar]
  • 1206.Carver AR, Tamayo E, Perez-Polo JR, Saade GR, Hankins GDV, Costantine MM. The effect of maternal pravastatin therapy on adverse sensorimotor outcomes of the offspring in a murine model of preeclampsia. Int J Dev Neurosci (2014) 33:33–40. 10.1016/j.ijdevneu.2013.11.004 [DOI] [PubMed] [Google Scholar]
  • 1207.Cindrova-Davies T. The therapeutic potential of antioxidants, ER chaperones, NO and H2S donors, and statins for treatment of preeclampsia. Front Pharmacol (2014) 5:119. 10.3389/fphar.2014.00119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1208.Girardi G. Can statins prevent pregnancy complications? J Reprod Immunol (2014) 10(1–102):161–7. 10.1016/j.jri.2013.07.005 [DOI] [PubMed] [Google Scholar]
  • 1209.McDonnold M, Tamayo E, Kechichian T, Gamble P, Longo M, Hankins GDV, et al. The effect of prenatal pravastatin treatment on altered fetal programming of postnatal growth and metabolic function in a preeclampsia-like murine model. Am J Obstet Gynecol (2014) 210(542):e541–7. 10.1016/j.ajog.2014.01.010 [DOI] [PubMed] [Google Scholar]
  • 1210.Ramma W, Ahmed A. Therapeutic potential of statins and the induction of heme oxygenase-1 in preeclampsia. J Reprod Immunol (2014) 10(1–102):153–60. 10.1016/j.jri.2013.12.120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1211.Saad AF, Kechichian T, Yin H, Sbrana E, Longo M, Wen M, et al. Effects of pravastatin on angiogenic and placental hypoxic imbalance in a mouse model of preeclampsia. Reprod Sci (2014) 21:138–45. 10.1177/1933719113492207 [DOI] [PubMed] [Google Scholar]
  • 1212.Staff AC, Johnsen GM, Dechend R, Redman CWG. Preeclampsia and uteroplacental acute atherosis: immune and inflammatory factors. J Reprod Immunol (2014) 10(1–102):120–6. 10.1016/j.jri.2013.09.001 [DOI] [PubMed] [Google Scholar]
  • 1213.Ahmed A, Ramma W. Unravelling the theories of pre-eclampsia: are the protective pathways the new paradigm? Br J Pharmacol (2015) 172:1574–86. 10.1111/bph.12977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1214.Bauer AJ, Banek CT, Needham K, Gillham H, Capoccia S, Regal JF, et al. Pravastatin attenuates hypertension, oxidative stress, and angiogenic imbalance in rat model of placental ischemia-induced hypertension. Hypertension (2013) 61:1103–10. 10.1161/HYPERTENSIONAHA.111.00226 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1215.Carver AR, Andrikopoulou M, Lei J, Tamayo E, Gamble P, Hou Z, et al. Maternal pravastatin prevents altered fetal brain development in a preeclamptic CD-1 mouse model. PLoS One (2014) 9:e100873. 10.1371/journal.pone.0100873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1216.Lefkou E, Mamopoulos A, Fragakis N, Dagklis T, Vosnakis C, Nounopoulos E, et al. Clinical improvement and successful pregnancy in a preeclamptic patient with antiphospholipid syndrome treated with pravastatin. Hypertension (2014) 63:e118–9. 10.1161/HYPERTENSIONAHA.114.03115 [DOI] [PubMed] [Google Scholar]
  • 1217.Brownfoot FC, Tong S, Hannan NJ, Binder NK, Walker SP, Cannon P, et al. Effects of pravastatin on human placenta, endothelium, and women with severe preeclampsia. Hypertension (2015) 66:687–97; discussion 445. 10.1161/HYPERTENSIONAHA.115.05445 [DOI] [PubMed] [Google Scholar]
  • 1218.Chaiworapongsa T, Romero R, Korzeniewski SJ, Chaemsaithong P, Hernandez-Andrade E, Segars JH, et al. Pravastatin to prevent recurrent fetal death in massive perivillous fibrin deposition of the placenta (MPFD). J Matern Fetal Neonatal Med (2016) 29:855–62. 10.3109/14767058.2015.1022864 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1219.Downing J. Sildenafil for the treatment of preeclampsia. Hypertens Pregnancy (2010) 29:248–50; author reply 251–2. 10.3109/10641951003634766 [DOI] [PubMed] [Google Scholar]
  • 1220.Kakigano A, Tomimatsu T, Mimura K, Kanayama T, Fujita S, Minato K, et al. Drug repositioning for preeclampsia therapeutics by in vitro screening: phosphodiesterase-5 inhibitor vardenafil restores endothelial dysfunction via induction of placental growth factor. Reprod Sci (2015) 22:1272–80. 10.1177/1933719115574340 [DOI] [PubMed] [Google Scholar]
  • 1221.Ramesar SV, Mackraj I, Gathiram P, Moodley J. Sildenafil citrate decreases sFlt-1 and sEng in pregnant l-NAME treated Sprague-Dawley rats. Eur J Obstet Gynecol Reprod Biol (2011) 157:136–40. 10.1016/j.ejogrb.2011.03.005 [DOI] [PubMed] [Google Scholar]
  • 1222.Herraiz S, Pellicer B, Serra V, Cauli O, Cortijo J, Felipo V, et al. Sildenafil citrate improves perinatal outcome in fetuses from pre-eclamptic rats. BJOG (2012) 119:1394–402. 10.1111/j.1471-0528.2012.03430.x [DOI] [PubMed] [Google Scholar]
  • 1223.Nassar AH, Masrouha KZ, Itani H, Nader KA, Usta IM. Effects of sildenafil in Nomega-nitro-L-arginine methyl ester-induced intrauterine growth restriction in a rat model. Am J Perinatol (2012) 29:429–34. 10.1055/s-0032-1304823 [DOI] [PubMed] [Google Scholar]
  • 1224.Stanley JL, Andersson IJ, Poudel R, Rueda-Clausen CF, Sibley CP, Davidge ST, et al. Sildenafil citrate rescues fetal growth in the catechol-O-methyl transferase knockout mouse model. Hypertension (2012) 59:1021–8. 10.1161/HYPERTENSIONAHA.111.186270 [DOI] [PubMed] [Google Scholar]
  • 1225.George EM, Palei AC, Dent EA, Granger JP. Sildenafil attenuates placental ischemia-induced hypertension. Am J Physiol Regul Integr Comp Physiol (2013) 305:R397–403. 10.1152/ajpregu.00216.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1226.Stanley JL, Sulek K, Andersson IJ, Davidge ST, Kenny LC, Sibley CP, et al. Sildenafil therapy normalizes the aberrant metabolomic profile in the comt(-/-) mouse model of preeclampsia/fetal growth restriction. Sci Rep (2015) 5:18241. 10.1038/srep18241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1227.Karasu E, Kayacan N, Sadan G, Dinc B. Different effects of different phosphodiesterase type-5 inhibitors in pre-eclampsia. Pregnancy Hypertens (2011) 1:231–7. 10.1016/j.preghy.2011.04.002 [DOI] [PubMed] [Google Scholar]
  • 1228.Karasu E, Kayacan N, Sadan G, Dinc B. Endothelial dysfunction in the human umbilical artery due to preeclampsia can be prevented by sildenafil. Clin Exp Hypertens (2012) 34:79–85. 10.3109/10641963.2011.628730 [DOI] [PubMed] [Google Scholar]
  • 1229.Ganzevoort W, Alfirevic Z, Von Dadelszen P, Kenny L, Papageorghiou A, Van Wassenaer-Leemhuis A, et al. STRIDER: sildenafil therapy in dismal prognosis early-onset intrauterine growth restriction – a protocol for a systematic review with individual participant data and aggregate data meta-analysis and trial sequential analysis. Syst Rev (2014) 3:23. 10.1186/2046-4053-3-23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1230.Abalos E, Duley L, Steyn DW. Antihypertensive drug therapy for mild to moderate hypertension during pregnancy. Cochrane Database Syst Rev (2014) 2:CD002252. 10.1002/14651858.CD002252.pub3 [DOI] [PubMed] [Google Scholar]
  • 1231.Mahmud H, Föller M, Lang F. Stimulation of erythrocyte cell membrane scrambling by methyldopa. Kidney Blood Press Res (2008) 31:299–306. 10.1159/000153250 [DOI] [PubMed] [Google Scholar]
  • 1232.Firoz T, Magee LA, Macdonell K, Payne BA, Gordon R, Vidler M, et al. Oral antihypertensive therapy for severe hypertension in pregnancy and postpartum: a systematic review. BJOG (2014) 121:1210–8; discussion 1220. 10.1111/1471-0528.12737 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1233.Magee LA, Namouz-Haddad S, Cao V, Koren G, Von Dadelszen P. Labetalol for hypertension in pregnancy. Expert Opin Drug Saf (2015) 14:453–61. 10.1517/14740338.2015.998197 [DOI] [PubMed] [Google Scholar]
  • 1234.Ishimaru T, Ishida J, Nakamura S, Hashimoto M, Matsukura T, Nakamura A, et al. Short-term suppression of the renin-angiotensin system in mice associated with hypertension during pregnancy. Mol Med Rep (2012) 6:28–32. 10.3892/mmr.2012.886 [DOI] [PubMed] [Google Scholar]
  • 1235.Marshall TG, Lee RE, Marshall FE. Common angiotensin receptor blockers may directly modulate the immune system via VDR, PPAR and CCR2b. Theor Biol Med Model (2006) 3:1. 10.1186/1742-4682-3-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1236.Ziegler EJ, Fisher CJ, Jr, Sprung CL, Straube RC, Sadoff JC, Foulke GE, et al. Treatment of Gram-negative bacteremia and septic shock with HA-1A human monoclonal antibody against endotoxin. A randomized, double-blind, placebo-controlled trial. The HA-1A Sepsis Study Group. N Engl J Med (1991) 324:429–36. 10.1056/NEJM199102143240701 [DOI] [PubMed] [Google Scholar]
  • 1237.Baumgartner JD, Glauser MP. Immunotherapy of endotoxemia and septicemia. Immunobiology (1993) 187:464–77. 10.1016/S0171-2985(11)80357-8 [DOI] [PubMed] [Google Scholar]
  • 1238.Derkx B, Wittes J, Mccloskey R. Randomized, placebo-controlled trial of HA-1A, a human monoclonal antibody to endotoxin, in children with meningococcal septic shock. European Pediatric Meningococcal Septic Shock Trial Study Group. Clin Infect Dis (1999) 28:770–7. 10.1086/515184 [DOI] [PubMed] [Google Scholar]
  • 1239.Yentis SM, Soni N, Riches PG. In vitro effects of HA-1A (centoxin) on cytokine production in whole blood from intensive care unit patients. Br J Anaesth (1994) 73:805–11. 10.1093/bja/73.6.805 [DOI] [PubMed] [Google Scholar]
  • 1240.Marks L. The birth pangs of monoclonal antibody therapeutics: the failure and legacy of centoxin. MAbs (2012) 4:403–12. 10.4161/mabs.19909 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1241.Helmerhorst EJ, Maaskant JJ, Appelmelk BJ. Anti-lipid A monoclonal antibody centoxin (HA-1A) binds to a wide variety of hydrophobic ligands. Infect Immun (1998) 66:870–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1242.Tan NS, Ho B, Ding JL. High-affinity LPS binding domain(s) in recombinant factor C of a horseshoe crab neutralizes LPS-induced lethality. FASEB J (2000) 14:859–70. 10.1096/fj.1530-6860 [DOI] [PubMed] [Google Scholar]
  • 1243.Tan NS, Ng ML, Yau YH, Chong PKW, Ho B, Ding JL. Definition of endotoxin binding sites in horseshoe crab factor C recombinant sushi proteins and neutralization of endotoxin by sushi peptides. FASEB J (2000) 14:1801–13. 10.1096/fj.99-0866com [DOI] [PubMed] [Google Scholar]
  • 1244.Yau YH, Ho B, Tan NS, Ng ML, Ding JL. High therapeutic index of factor C sushi peptides: potent antimicrobials against Pseudomonas aeruginosa. Antimicrob Agents Chemother (2001) 45:2820–5. 10.1128/AAC.45.10.2820-2825.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1245.Li P, Sun M, Wohland T, Ho B, Ding JL. The molecular mechanism of interaction between sushi peptide and Pseudomonas endotoxin. Cell Mol Immunol (2006) 3:21–8. [PubMed] [Google Scholar]
  • 1246.Li P, Sun M, Wohland T, Yang D, Ho B, Ding JL. Molecular mechanisms that govern the specificity of sushi peptides for Gram-negative bacterial membrane lipids. Biochemistry (2006) 45:10554–62. 10.1021/bi0602765 [DOI] [PubMed] [Google Scholar]
  • 1247.Ding JL, Li P, Ho B. The sushi peptides: structural characterization and mode of action against Gram-negative bacteria. Cell Mol Life Sci (2008) 65:1202–19. 10.1007/s00018-008-7456-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1248.Leptihn S, Har JY, Chen J, Ho B, Wohland T, Ding JL. Single molecule resolution of the antimicrobial action of quantum dot-labeled sushi peptide on live bacteria. BMC Biol (2009) 7:22. 10.1186/1741-7007-7-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1249.Leptihn S, Guo L, Frecer V, Ho B, Ding JL, Wohland T. One step at a time: action mechanism of sushi 1 antimicrobial peptide and derived molecules. Virulence (2010) 1:42–4. 10.4161/viru.1.1.10229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1250.Frecer V, Ho B, Ding JL. De novo design of potent antimicrobial peptides. Antimicrob Agents Chemother (2004) 48:3349–57. 10.1128/AAC.48.9.3349-3357.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1251.Bhattacharjya S. De novo designed lipopolysaccharide binding peptides: structure based development of antiendotoxic and antimicrobial drugs. Curr Med Chem (2010) 17:3080–93. 10.2174/092986710791959756 [DOI] [PubMed] [Google Scholar]
  • 1252.Brantsaeter AL, Myhre R, Haugen M, Myking S, Sengpiel V, Magnus P, et al. Intake of probiotic food and risk of preeclampsia in primiparous women: the Norwegian Mother and Child Cohort Study. Am J Epidemiol (2011) 174:807–15. 10.1093/aje/kwr168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1253.Gluckman P, Beedle A, Hanson M. Principles of Evolutionary Medicine. Oxford: Oxford University Press; (2009). [Google Scholar]
  • 1254.Hemmings DG. Signal transduction underlying the vascular effects of sphingosine 1-phosphate and sphingosylphosphorylcholine. Naunyn Schmiedebergs Arch Pharmacol (2006) 373:18–29. 10.1007/s00210-006-0046-5 [DOI] [PubMed] [Google Scholar]
  • 1255.Hemmings DG, Hudson NK, Halliday D, O’Hara M, Baker PN, Davidge ST, et al. Sphingosine-1-phosphate acts via rho-associated kinase and nitric oxide to regulate human placental vascular tone. Biol Reprod (2006) 74:88–94. 10.1095/biolreprod.105.043034 [DOI] [PubMed] [Google Scholar]
  • 1256.Teijaro JR, Walsh KB, Cahalan S, Fremgen DM, Roberts E, Scott F, et al. Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell (2011) 146:980–91. 10.1016/j.cell.2011.08.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1257.Oldstone MBA, Teijaro JR, Walsh KB, Rosen H. Dissecting influenza virus pathogenesis uncovers a novel chemical approach to combat the infection. Virology (2013) 435:92–101. 10.1016/j.virol.2012.09.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1258.Maceyka M, Spiegel S. Sphingolipid metabolites in inflammatory disease. Nature (2014) 510:58–67. 10.1038/nature13475 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1259.Oldstone MBA, Rosen H. Cytokine storm plays a direct role in the morbidity and mortality from influenza virus infection and is chemically treatable with a single sphingosine-1-phosphate agonist molecule. Curr Top Microbiol Immunol (2014) 378:129–47. 10.1007/978-3-319-05879-5_6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1260.Walsh KB, Teijaro JR, Brock LG, Fremgen DM, Collins PL, Rosen H, et al. Animal model of respiratory syncytial virus: CD8+ T cells cause a cytokine storm that is chemically tractable by sphingosine-1-phosphate 1 receptor agonist therapy. J Virol (2014) 88:6281–93. 10.1128/JVI.00464-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1261.Arish M, Husein A, Kashif M, Saleem M, Akhter Y, Rub A. Sphingosine-1-phosphate signaling: unraveling its role as a drug target against infectious diseases. Drug Discov Today (2016) 21:133–42. 10.1016/j.drudis.2015.09.013 [DOI] [PubMed] [Google Scholar]
  • 1262.Zhang J, Dunk CE, Lye SJ. Sphingosine signalling regulates decidual NK cell angiogenic phenotype and trophoblast migration. Hum Reprod (2013) 28:3026–37. 10.1093/humrep/det339 [DOI] [PubMed] [Google Scholar]
  • 1263.Nagamatsu T, Iwasawa-Kawai Y, Ichikawa M, Kawana K, Yamashita T, Osuga Y, et al. Emerging roles for lysophospholipid mediators in pregnancy. Am J Reprod Immunol (2014) 72:182–91. 10.1111/aji.12239 [DOI] [PubMed] [Google Scholar]
  • 1264.Romanowicz L, Bańkowski E. Altered sphingolipid composition in Wharton’s jelly of pre-eclamptic newborns. Pathobiology (2010) 77:78–87. 10.1159/000278289 [DOI] [PubMed] [Google Scholar]
  • 1265.Romanowicz L, Bańkowski E. Lipid compounds of human Wharton’s jelly and their alterations in preeclampsia. Int J Exp Pathol (2010) 91:1–9. 10.1111/j.1365-2613.2009.00675.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1266.Romanowicz L, Bańkowski E. Sphingolipids of human umbilical cord vein and their alteration in preeclampsia. Mol Cell Biochem (2010) 340:81–9. 10.1007/s11010-010-0403-z [DOI] [PubMed] [Google Scholar]
  • 1267.Baig S, Lim JY, Fernandis AZ, Wenk MR, Kale A, Su LL, et al. Lipidomic analysis of human placental syncytiotrophoblast microvesicles in adverse pregnancy outcomes. Placenta (2013) 34:436–42. 10.1016/j.placenta.2013.02.004 [DOI] [PubMed] [Google Scholar]
  • 1268.Li Q, Pan Z, Wang X, Gao Z, Ren C, Yang W. miR-125b-1-3p inhibits trophoblast cell invasion by targeting sphingosine-1-phosphate receptor 1 in preeclampsia. Biochem Biophys Res Commun (2014) 453:57–63. 10.1016/j.bbrc.2014.09.059 [DOI] [PubMed] [Google Scholar]
  • 1269.Carrillo-Vico A, Lardone PJ, Naji L, Fernández-Santos JM, Martín-Lacave I, Guerrero JM, et al. Beneficial pleiotropic actions of melatonin in an experimental model of septic shock in mice: regulation of pro-/anti-inflammatory cytokine network, protection against oxidative damage and anti-apoptotic effects. J Pineal Res (2005) 39:400–8. 10.1111/j.1600-079X.2005.00265.x [DOI] [PubMed] [Google Scholar]
  • 1270.Gitto E, Pellegrino S, Gitto P, Barberi I, Reiter RJ. Oxidative stress of the newborn in the pre- and postnatal period and the clinical utility of melatonin. J Pineal Res (2009) 46:128–39. 10.1111/j.1600-079X.2008.00649.x [DOI] [PubMed] [Google Scholar]
  • 1271.Hobson SR, Lim R, Gardiner EE, Alers NO, Wallace EM. Phase I pilot clinical trial of antenatal maternally administered melatonin to decrease the level of oxidative stress in human pregnancies affected by pre-eclampsia (PAMPR): study protocol. BMJ Open (2013) 3:e003788. 10.1136/bmjopen-2013-003788 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1272.Westh H, Lisby G, Breysse F, Boddinghaus B, Chomarat M, Gant V, et al. Multiplex real-time PCR and blood culture for identification of bloodstream pathogens in patients with suspected sepsis. Clin Microbiol Infect (2009) 15:544–51. 10.1111/j.1469-0691.2009.02736.x [DOI] [PubMed] [Google Scholar]
  • 1273.Yang B, Wang Y, Qian PY. Sensitivity and correlation of hypervariable regions in 16S rRNA genes in phylogenetic analysis. BMC Bioinformatics (2016) 17:135. 10.1186/s12859-016-0992-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1274.Khamis A, Raoult D, La Scola B. Comparison between rpoB and 16S rRNA gene sequencing for molecular identification of 168 clinical isolates of Corynebacterium. J Clin Microbiol (2005) 43:1934–6. 10.1128/JCM.43.4.1934-1936.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1275.Adékambi T, Drancourt M, Raoult D. The rpoB gene as a tool for clinical microbiologists. Trends Microbiol (2009) 17:37–45. 10.1016/j.tim.2008.09.008 [DOI] [PubMed] [Google Scholar]
  • 1276.Roux S, Enault F, Bronner G, Debroas D. Comparison of 16S rRNA and protein-coding genes as molecular markers for assessing microbial diversity (bacteria and archaea) in ecosystems. FEMS Microbiol Ecol (2011) 78:617–28. 10.1111/j.1574-6941.2011.01190.x [DOI] [PubMed] [Google Scholar]
  • 1277.Liu W, Li L, Khan MA, Zhu F. Popular molecular markers in bacteria. Mol Gen Microbiol Virol (2012) 27(3):103–7. 10.3103/S0891416812030056 [DOI] [PubMed] [Google Scholar]
  • 1278.Větrovský T, Baldrian P. The variability of the 16S rRNA gene in bacterial genomes and its consequences for bacterial community analyses. PLoS One (2013) 8:e57923. 10.1371/journal.pone.0057923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1279.Das S, Dash HR, Mangwani N, Chakraborty J, Kumari S. Understanding molecular identification and polyphasic taxonomic approaches for genetic relatedness and phylogenetic relationships of microorganisms. J Microbiol Methods (2014) 103:80–100. 10.1016/j.mimet.2014.05.013 [DOI] [PubMed] [Google Scholar]
  • 1280.Cody AJ, McCarthy ND, Jansen Van Rensburg M, Isinkaye T, Bentley SD, Parkhill J, et al. Real-time genomic epidemiological evaluation of human Campylobacter isolates by use of whole-genome multilocus sequence typing. J Clin Microbiol (2013) 51:2526–34. 10.1128/JCM.00066-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1281.Lazar L, Rigó J, Jr, Nagy B, Balogh K, Makó V, Cervenak L, et al. Relationship of circulating cell-free DNA levels to cell-free fetal DNA levels, clinical characteristics and laboratory parameters in preeclampsia. BMC Med Genet (2009) 10:120. 10.1186/1471-2350-10-120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1282.Galbiati S, Causarano V, Pinzani P, Francesca S, Orlando C, Smid M, et al. Evaluation of a panel of circulating DNA, RNA and protein potential markers for pathologies of pregnancy. Clin Chem Lab Med (2010) 48:791–4. 10.1515/CCLM.2010.160 [DOI] [PubMed] [Google Scholar]
  • 1283.Miranda ML, Macher HC, Munoz-Hernandez R, Vallejo-Vaz A, Moreno-Luna R, Villar J, et al. Role of circulating cell-free DNA levels in patients with severe preeclampsia and HELLP syndrome. Am J Hypertens (2013) 26:1377–80. 10.1093/ajh/hpt187 [DOI] [PubMed] [Google Scholar]
  • 1284.Oudejans CBM. Maternal plasma RNA sequencing. Clin Biochem (2015) 48:942–7. 10.1016/j.clinbiochem.2015.03.004 [DOI] [PubMed] [Google Scholar]
  • 1285.Hudecova I. Digital PCR analysis of circulating nucleic acids. Clin Biochem (2015) 48:948–56. 10.1016/j.clinbiochem.2015.03.015 [DOI] [PubMed] [Google Scholar]
  • 1286.Coates ARM, Hu Y. New strategies for antibacterial drug design: targeting non-multiplying latent bacteria. Drugs R D (2006) 7:133–51. 10.2165/00126839-200607030-00001 [DOI] [PubMed] [Google Scholar]
  • 1287.Hu Y, Shamaei-Tousi A, Liu Y, Coates A. A new approach for the discovery of antibiotics by targeting non-multiplying bacteria: a novel topical antibiotic for staphylococcal infections. PLoS One (2010) 5:e11818. 10.1371/journal.pone.0011818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1288.Coates AR, Halls G, Hu Y. Novel classes of antibiotics or more of the same? Br J Pharmacol (2011) 163:184–94. 10.1111/j.1476-5381.2011.01250.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1289.Laxminarayan R, Duse A, Wattal C, Zaidi AK, Wertheim HF, Sumpradit N, et al. Antibiotic resistance – the need for global solutions. Lancet Infect Dis (2013) 13:1057–98. 10.1016/S1473-3099(13)70318-9 [DOI] [PubMed] [Google Scholar]
  • 1290.Hu Y, Liu A, Ortega-Muro F, Alameda-Martin L, Mitchison D, Coates A. High-dose rifampicin kills persisters, shortens treatment duration, and reduces relapse rate in vitro and in vivo. Front Microbiol (2015) 6:641. 10.3389/fmicb.2015.00641 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1291.Barber M, Tait RC, Scott J, Rumley A, Lowe GD, Stott DJ. Dementia in subjects with atrial fibrillation: hemostatic function and the role of anticoagulation. J Thromb Haemost (2004) 2:1873–8. 10.1111/j.1538-7836.2004.00993.x [DOI] [PubMed] [Google Scholar]
  • 1292.Murthy SB, Jawaid A, Qureshi SU, Schulz PE, Schulz PE. The apolipoprotein 2 allele in Alzheimer’s disease: suggestions for a judicious use of antiplatelet and anticoagulant medications. J Am Geriatr Soc (2009) 57:1124–5. 10.1111/j.1532-5415.2009.02284.x [DOI] [PubMed] [Google Scholar]
  • 1293.Ahn HJ, Glickman JF, Poon KL, Zamolodchikov D, Jno-Charles OC, Norris EH, et al. A novel abeta-fibrinogen interaction inhibitor rescues altered thrombosis and cognitive decline in Alzheimer’s disease mice. J Exp Med (2014) 211:1049–62. 10.1084/jem.20131751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1294.Krishnan R, Tsubery H, Proschitsky MY, Asp E, Lulu M, Gilead S, et al. A bacteriophage capsid protein provides a general amyloid interaction motif (GAIM) that binds and remodels misfolded protein assemblies. J Mol Biol (2014) 426:2500–19. 10.1016/j.jmb.2014.04.015 [DOI] [PubMed] [Google Scholar]
  • 1295.Ankarcrona M, Winblad B, Monteiro C, Fearns C, Powers ET, Johansson J, et al. Current and future treatment of amyloid diseases. J Intern Med (2016) 280:177–202. 10.1111/joim.12506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 1296.Marshall BJ. Helicobacter pylori. Am J Gastroenterol (1994) 89:S116–28. [PubMed] [Google Scholar]

Articles from Frontiers in Medicine are provided here courtesy of Frontiers Media SA

RESOURCES