Skip to main content
Springer logoLink to Springer
. 2011 Jul 30;31(4):379–388. doi: 10.1007/s10096-011-1337-4

The impact of diabetes on the pathogenesis of sepsis

G C K W Koh 1,2,3,4,, S J Peacock 1,4, T van der Poll 2, W J Wiersinga 2
PMCID: PMC3303037  PMID: 21805196

Abstract

Diabetes is associated with an increased susceptibility to infection and sepsis. Conflicting data exist on whether the mortality of patients with sepsis is influenced by the presence of diabetes, fuelling the ongoing debate on the benefit of tight glucose regulation in patients with sepsis. The main reason for which diabetes predisposes to infection appears to be abnormalities of the host response, particularly in neutrophil chemotaxis, adhesion and intracellular killing, defects that have been attributed to the effect of hyperglycaemia. There is also evidence for defects in humoral immunity, and this may play a larger role than previously recognised. We review the literature on the immune response in diabetes and its potential contribution to the pathogenesis of sepsis. In addition, the effect of diabetes treatment on the immune response is discussed, with specific reference to insulin, metformin, sulphonylureas and thiazolidinediones.

Introduction

Patients with diabetes mellitus have an increased risk of developing infections and sepsis [1, 2], and constitute 20.1–22.7% of all sepsis patients [3, 4]. This association was first observed a thousand years ago by Avicenna (980–1027), who noted that diabetes was frequently complicated by tuberculosis [5]. In the pre-insulin era, Joslin noted, in a series of 1,000 cases, that diabetic coma was usually precipitated by infection [6], and infection remains an important cause of death in diabetics [7]. Much of the literature does not distinguish between types of diabetes and regards all complications as secondary to hyperglycaemia and independent of diabetes aetiology.

We review the pathogenesis of infection in the diabetic patient and the altered host response, focusing on data from human studies.

Risk of infection and clinical considerations

A small number of conditions are strongly associated with diabetes, including malignant otitis externa [810], emphysematous pyelonephritis [1114], emphysematous cholecystitis [15, 16], Klebsiella liver abscesses [17], rhinocerebral mucormycosis [18, 19] and melioidosis [20]. However, these are rare, and most infections in diabetics are those that occur also in the general population. Two population-based studies have proved pivotal to our understanding of the susceptibilities of patients with diabetes [1, 2]: a study of 523,749 Canadians with diabetes and an equal number of matched controls [2] found that diabetes increased the risk for cystitis (risk ratio 1.39–1.43), pneumonia (1.46–1.48), cellulitis (1.81–1.85) and tuberculosis (1.12–1.21). A study of 7,417 Dutch patients with diabetes found a higher incidence of lower respiratory tract infection (adjusted odds ratios [ORs] 1.42 for type 1 diabetes and 1.32 for type 2), urinary tract infection (1.96 and 1.24), and skin and mucous membrane infection (1.59 and 1.33) [1]. The association between diabetes and tuberculosis was re-confirmed by a recent meta-analysis [21].

Although diabetes mellitus is implicated in susceptibility to infection, its influence on the subsequent clinical course and outcome is less clear. Some studies have shown an association with increased mortality [2225], others found no effect [4, 2634], while still others found improved survival [15, 16, 35]. The largest of these (12.5 million sepsis cases) [15] found that diabetics were less likely to develop acute respiratory failure and linked this to two previous studies which found that diabetics seem to be protected from acute lung injury [36, 37]. The largest single study to show an adverse effect of diabetes on mortality in sepsis was conducted in 29,900 Danish patients with community-acquired pneumonia and found that patients with diabetes had a higher risk of mortality (OR 1.2) [24].

The reasons for the different outcomes between these studies are unclear, but may relate to differences in the study population, varying outcome measures and differences in statistical analysis and in diabetes drug prescription habits between countries [38]. Population-based studies are less prone to selection bias compared to hospital-based studies, but more detailed clinical information is usually available in hospital-based studies. In terms of outcome measures, studies with outcomes at longer time points (e.g. 6 months versus 28-day mortality) are more likely to find informative differences, but are much more difficult to conduct [39]. Observational studies often make use of multi-variable regression techniques to correct for confounders (a common, but incorrect, approach to model-building is to include all measured parameters and then remove parameters on the basis of their p-value). Over-adjustment or unnecessary adjustment for variables that are not confounders can produce biased or spurious results [40]. Patients with diabetes also have multiple co-morbidities that may worsen outcomes: it is debatable whether these co-morbidities should be adjusted for, since many are caused by diabetes and, therefore, cannot be regarded as confounders [41]. Nevertheless, a number of studies have attempted to adjust for these comorbidities [23, 24, 26]. The possible influences of drug treatment are described below.

Diabetes and the immune system

In 1904, Lassar suggested that high levels of glucose may drive infection by serving as a nutrient source for bacteria [42], but in 1911, Handmann showed that glucose supplementation did not enhance bacterial growth [43], and proposed, instead, a defect in immune function. Da Costa and Beardsley first demonstrated the existence of an immune defect in 1907 [44]. The subsequent literature on this topic is complicated by the fact that different techniques have been used over the years, and gaps of a decade or more may separate experiments, making it difficult to compare results. Most studies have shown defects in neutrophil function, with good evidence for abnormalities in adhesion, chemotaxis and intracellular killing, but evidence for a phagocytosis defect are contradictory. The evidence that neutrophil defects are solely responsible for the increases in the susceptibility of diabetics to infection is equivocal [45]; there is good evidence that humoral responses in diabetics are poorer and may play a larger role than previously recognised.

General markers of inflammation

Diabetes is associated with elevations in C-reactive protein (CRP) [46], tumour necrosis factor alpha (TNF-α) [47], interleukin (IL)-6 [46] and IL-8 [48], but no differences are seen in circulating cell surface markers or coagulation markers between patients with and without diabetes in the context of sepsis. In a cohort of 1,799 patients with community-acquired pneumonia (CAP) [49], concentrations of pro-inflammatory cytokines (TNF-α, IL-6 and IL-10), coagulation (anti-thrombin, Factor IX and thrombin–anti-thrombin complexes) and fibrinolysis (PAI-1 and D-dimer) biomarkers were similar in subjects with and without diabetes at presentation and in the first week of hospitalisation [49]. In addition, monocyte expression of CD120a, CD120b, HLA-DR, TLR4 and TLR2 on monocytes was not different between the groups [49]. These results are consistent with a cohort study of 830 sepsis patients, in whom plasma concentrations of IL-6 and TNF-α were elevated to the same extent in patients with and without diabetes, both at admission and at follow-up [4]. In this second study, diabetes was not found to exacerbate the known pro-coagulant response seen in sepsis [4]. Since sepsis and diabetes both induce a pro-inflammatory and pro-coagulant state, and since both interfere with the host response, the lack of a strong influence of diabetes on the pro-inflammatory and coagulation pathways during sepsis is remarkable. Preclinical studies in healthy volunteers have shown that acute hyperglycaemia and insulin resistance may both directly influence inflammation and coagulation [50, 51], but these changes may not be detectable on the background of the much larger abnormalities attributable to sepsis. There is also evidence that local responses may be impaired in diabetes, e.g. levels of urinary IL-6 and IL-8 are lower in diabetic women with bacteriuria [52]. Endothelial activation has been implicated in the pathogenesis of sepsis [53] and diabetes is itself known to activate endothelium. A recent study of 207 sepsis patients (of whom 30% had diabetes) showed that markers of endothelial cell activation (plasma E-selectin and soluble fms-like tyrosine kinase-1 [sFLT-1]) were higher in diabetes [54].

Neutrophils

Adhesion

The recruitment of neutrophils to a site of inflammation requires endothelial adhesion followed by transmigration and exit from the circulation, a process requiring the expression by neutrophils of integrins (e.g. CD11a/CD18 and CD11b/CB18) [55, 56], which then bind to endothelial cell adhesion molecules (e.g. ICAM-1 [5759]). A study in which neutrophils were harvested from 26 patients with diabetes and an equal number of controls demonstrated that adhesion to bovine aortic endothelium was increased for neutrophils from diabetics, but only if the endothelium was also incubated with plasma from patients with diabetes [60]. Increased adhesion appears to be due to both an increase in the expression of integrins by diabetic neutrophils and of adhesion molecules by endothelium. Diabetic neutrophils have increased the expression of CD11b and CD11c [61], and glucose itself appears to be able to stimulate the expression of ICAM-1 by endothelial cells [5759, 6264], possibly via an osmotic effect [63, 64].

Chemotaxis

Chemotaxis is the ability of neutrophils to detect and move towards a chemical inflammatory stimulus. Studies may be divided by technique: those using the two-chamber Boyden technique [65] have produced conflicting results [66, 67], but those using the subagarose technique [68] (which includes a negative control, which Boyden’s technique lacks) have reproducibly shown a defect in diabetes [61, 69].

Phagocytosis

Phagocytosis is the engulfment and ingestion of foreign bodies by a cell, allowing neutrophils to remove and destroy pathogens. The evidence for a defect in phagocytosis in diabetes is contradictory, with some reporting a defect [7073], but others not [61, 74]. These inconsistencies may be attributed to differences in methodology: neutrophils will not phagocytose unopsonised particles, so bacteria and cells need first to be incubated with serum containing C3b or IgG. Many studies have used autologous serum [7073], but those that have used a standard serum or opsonin have found no defect [61, 74]. In 1976, Bagdade found that phagocytosis of Streptococcus pneumonia was reduced in neutrophils recovered from eight patients with poorly controlled diabetes, but this defect improved with diabetes treatment [70]. Notably, control neutrophils incubated with serum taken from patients with diabetes also demonstrated a defect in phagocytosis, implying that the defect was, in fact, due to defective opsonisation and not to a deficit in neutrophil function per se: in other words, the defect is humoral. In 1984, Davidson et al. studied the ingestion of Candida guilliermondii by neutrophils from 11 patients with diabetes and found that phagocytosis was reduced. However, if pre-opsonised yeast cells were used, then phagocytosis was no different from controls, again suggesting that a humoral defect must exist [72]. Delamaire et al. used a single control serum for all samples to remove the possibility of a difference in opsonisation [61], convincingly demonstrating that no phagocytosis defect exists.

Killing

Neutrophils have two distinct mechanisms for killing bacteria, intracellular and extracellular. Phagocytosed bacteria are killed by superoxide anions and other oxygen-derived species. Culture-based methods have demonstrated a defect in the intracellular killing of Staphylococcus aureus [69, 75, 76], Streptococcus pneumoniae [71, 77] and Candida albicans [78]. More recent studies have confirmed this finding used chemiluminescence methods [7982], a superior method compared to culture, because it separates the effect of phagocytosis from that of intracellular killing. The killing defect cannot be corrected by incubation with normal serum [76], suggesting that it is cellular in origin, but improves with glycaemic control [81].

Neutrophils are also able to kill bacteria extracellularly by expelling chromatin, which combines with granule proteins to form neutrophil extracellular traps (NETs) [83]. Interestingly, β-hydroxybutyrate (a ketone body present in diabetic ketoacidosis) has been shown to inhibit the formation of NETs [84], but the relevance of this finding to patients remains to be demonstrated.

Monocytes

Monocytes in diabetes have been less well studied than neutrophils, but also appear to have defects of chemotaxis [85] and phagocytosis [86, 87]. Adhesion to endothelium is also enhanced [88, 89]. In contrast to neutrophils, intracellular killing seems to be enhanced [90]. Monocytes obtained from 24 diabetic patients produced similar amounts of TNF-α when compared to healthy controls when stimulated with lipopolysaccharide (LPS), but the IL-6 levels were higher in patients with type 1 diabetes [91].

Lymphocytes

Few studies have investigated the effect of diabetes on lymphocyte function. One measure of lymphocyte function is transformation in response to a mitogen or bacterial antigen. Studies containing acidotic patients appear to find that responses are diminished [92, 93] and that correction of the acidosis leads to prompt resolution of the defect [93], but more recent studies have found deficient proliferative T-cell responses, even in treated patients [82, 94]. Diabetic T-cells express higher levels of CD152, a downregulator of the immune response [95]. Three other studies failed to find a defect [67, 96, 97].

Humoral defects

In 1907, Da Costa and Beardsley [44, 98] found that sera from diabetes patients were less able to opsonise S. aureus compared to sera from controls. In 1973, Farid and Anderson surveyed 46 patients and found that IgG levels were lower in insulin-treated diabetics, but not patients on oral treatments or diet alone. More recently, a study of 66 patients with type 1 diabetes demonstrated that total IgG levels were lower in uncontrolled diabetics as measured by HbA1c [99]. Also, the apparent defect in neutrophil phagocytosis appear to be humoral and not cellular in origin (see above).

The best evidence for a humoral defect in diabetic patients comes from vaccine studies. It was described as early as 1930 that deficient agglutinin responses are seen in the diabetic patients after subcutaneous typhoid vaccination [100, 101]. Multiple studies have shown that patients with diabetes are less likely to mount a protective antibody response to hepatitis B vaccination [102105], leading some authorities to recommend routinely adding a booster dose to the standard regimen for patients with diabetes [102, 106]. The literature on influenza vaccination is more mixed (reviewed by Brydak and Machala [107]). Pozzilli et al. looked at 52 diabetic patients and found fewer activated lymphocytes in patients with type 2 diabetes following influenza vaccination, but no differences in antibody responses [108]. Muszkat et al., studying a more elderly population, found lower antibody responses in patients with type 2 diabetes [109]. Diabetes is also associated with a waning in the duration of protection afforded by tetanus vaccination, although the initial response appears to be normal [110, 111]. Diabetics appear to respond well to pneumococcal polysaccharide vaccine [112], although there are no studies studying the duration of protection in diabetic patients. There are no studies specifically linking humoral responses in sepsis to diabetes.

Complement abnormalities

Inherited deficiencies of component 4 (C4) have been implicated in the pathogenesis of type 1 diabetes [113115], but whether this contributes to susceptibility to infection in type 1 diabetics is not known. By contrast, obesity and elevated insulin levels (as which occurs in type 2 diabetes) appear to be associated with elevations in C3 [116]. Karlsson et al., looking for biomarkers for maturity-onset diabetes of the young (MODY), found that complement C5 and C8 are both elevated in diabetes, regardless of aetiology [117], a possible mechanism for these abnormalities being that complement activation can be driven by glycated immunoglobulins [118]. One explanation for why diabetic sera are less able to opsonise bacteria may be that glucose attacks the thioester bond of complement C3 and prevents it from binding to the bacterial surface [119].

The role of hyperglycaemia

Warren noted in 1930 that the risk of infection in diabetic patients was inversely proportional to the degree of diabetes control [120], a finding replicated in 1982 by Rayfield [121]. The strongest evidence for the role of glycaemic control in preventing infection comes from the surgical literature. In a single-centre study of 8,910 cardiac surgery patients, glycaemic control in the immediate post-operative period was associated with a reduction in the risk of deep wound infection. In a multi-centre observational study of 55,408 diabetic post-surgical patients, the risk of post-operative infection was increased if serum glucose concentrations exceeded 8.3 mM [122]. Not all studies from the last 10 years have been able to replicate this finding: most notably, a carefully designed Australian study of 68 patients in the community failed to find a relationship between glycaemic control and infection risk [123], but the median HbA1c in that study was only 7.4%.

Diabetes medications and the immune response during sepsis

Insulin

Stegenga et al. dissected out the separate roles of insulin and glucose in infectious disease pathogenesis by studying healthy volunteers in whom insulin and glucose levels were maintained at preset levels for 6–8 h using tightly controlled infusions of insulin, glucose, somatostatin and glucagon, and intravenous E. coli LPS given to simulate sepsis [124]. Hyperglycaemia reduced neutrophil degranulation following LPS administration (independent of insulin concentration). Neither hyperinsulinaemia nor hyperglycaemia affected plasma cytokine levels (TNF-α, IL-6, IL-8 or IL-10) [125]. A second study using a similar design found intranuclear NF-κB downregulation following insulin infusion [126]. In an intensive care unit (ICU) setting, high-dose insulin therapy was associated with the more rapid resolution of CRP levels and white blood cell counts, suggesting that an anti-inflammatory effect of insulin might be beneficial in sepsis [127].

Diabetic leukocytes display a reduced rate of glycolysis in vitro [128], which can be corrected by insulin supplementation [129]. The energy required for chemotaxis is supplied almost entirely by glycolysis [130], as their mitochondria are metabolically inactive [131] and insulin supplementation is able to reverse the chemotaxis defect seen in diabetes [66].

Clinical evidence for a benefit of intensive insulin therapy in sepsis [3, 132, 133] is contradictory. A single-centre study demonstrated a reduction in cardiothoracic ICU mortality with intensive intravenous insulin [132], a second single-centre study at the same centre, but on the medical ICU, found no effect on mortality [133], and a subsequent multi-centre trial concluded that intensive insulin therapy increased mortality [3]. A recent meta-analysis concluded that, in critically ill patients, tight glucose control does not reduce mortality, but does increase the risk of severe hypoglycaemia [134].

Metformin

Metformin is prescribed as the first line treatment in Europe because it is associated with a 36% reduction in the all-cause mortality compared with diet alone [135]. There is little evidence for an immunomodulatory effect of metformin, although one study reported an association with reduced pro-inflammatory cytokine macrophage migration inhibitory factor (MIF) levels in obesity [136]. The main complication of metformin treatment in the context of sepsis is the risk of lactic acidosis [137], due to the metformin-mediated inhibition of pyruvate dehydrogenase promoting anaerobic respiration. This has prompted some authorities to recommend withdrawing metformin in sepsis [138].

Sulphonylureas

The best studied sulphonylurea in the context of sepsis is glibenclamide (= glyburide, United States adopted name [USAN]). Glibenclamide inhibits monocyte IL-1 secretion and has been used for over ten years in the laboratory specifically for that purpose [139]. The mechanism for this is the inhibition of inflammasome assembly [140], although the exact protein target has not been identified. Other sulphonylureas may not share this property [140]. Glibenclamide was associated with reduced inflammation and a halving in mortality in melioidosis, an infection strongly associated with diabetes [38]. Glibenclamide also has a direct pressor effect on vascular smooth muscle in vitro [141], and it has been proposed that glibenclamide therapy might find use as a vasopressor in septic shock [142]. Two small clinical studies in septic shock failed to find any effect on blood pressure [143, 144], although neither study was designed to look for an effect on mortality.

Thiazolidinediones

Observational studies of diabetic patients on the thiazolidinediones have demonstrated the suppression of nuclear factor-κB [145, 146]. Rosiglitazone reduced renal injury [147] and improved other markers of end-organ damage [148] in murine sepsis models, while ciglitazone reduced bacterial burdens and local inflammation in a murine model of pneumococcal pneumonia [149], suggesting that thiazolidinediones may find use as an adjunctive treatment for sepsis [150].

Conclusions

Infection remains an important cause of morbidity and mortality in diabetics, probably due to abnormalities of the host response, particularly in neutrophil chemotaxis, adhesion and intracellular killing. Humoral defects exist (both in antibody responses and complement opsonisation) and may explain earlier reports of a defect in phagocytosis, but are poorly studied in the pathogenesis of sepsis. Very little is known about the molecular mechanisms by which diabetes produces these effects, but the functional modification of host proteins and osmotic effects have both been proposed. For newly recognised phenomena such as neutrophil extracellular traps (NETs), we know almost nothing of the effect of diabetes, although preliminary evidence is that they may be important. Epidemiological studies of diabetes have produced conflicting results, and some of this difference may be explained by differences in the study design and epidemiological techniques used. Many studies have, so far, ignored the effects of drugs on the host response, and this omission may also explain the conflicting results in the literature.

Acknowledgements

G.C.K.W.K. is funded by the Wellcome Trust of Great Britain (086532/Z/08/Z). Sharon Peacock receives funding from the Wellcome Trust of Great Britain, the Health Protection Agency (UK), the European Union and the UK Clinical Research Collaboration. Tom van der Poll receives funding from BEGETU, the European Union, de Landsteiner Stichting voor Bloedtransfusie Research, CTMM and the Netherlands Asthma Foundation. W.J.W. is funded by a VENI grant from the Netherlands Organization for Scientific Research. We wish to thank Andrew King at the Wellcome Trust Sanger Institute for helping to locate the relevant literature.

Conflict of interest

The authors have no conflicts of interest to declare.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Abbreviations

E. coli

Escherichia coli

C3

Complement component 3

C5

Complement component 5

C8

Complement component 8

CAP

Community-acquired pneumonia

CD

Cluster of differentiation, a systematic classification of cell surface antigens

CR3

Complement receptor 3, also known as CD11b/CD18

CRP

C-reactive protein

HbA1c

Glycated hemoglobin, a measure of glycaemic control in diabetes

ICU

Intensive care unit

IL

Interleukin

LPS

Lipopolysaccharide

Mac-1

Macrophage 1 antigen, also known as CD11b/CD18

MODY

Maturity-onset diabetes of the young

NET

Neutrophil extracellular traps

OR

Odds ratio

S. aureus

Staphylococcus aureus

S. epidermidis

Staphylococcus epidermidis

sFLT-1

Soluble fms-like tyrosine kinase-1

TNF-α

Tumour necrosis factor alpha

USAN

United States adopted name

WHO

World Health Organization, Geneva, Switzerland

References

  • 1.Muller LMAJ, Gorter KJ, Hak E, Goudzwaard WL, Schellevis FG, Hoepelman AIM, Rutten GEHM. Increased risk of common infections in patients with type 1 and type 2 diabetes mellitus. Clin Infect Dis. 2005;41:281–288. doi: 10.1086/431587. [DOI] [PubMed] [Google Scholar]
  • 2.Shah BR, Hux JE. Quantifying the risk of infectious diseases for people with diabetes. Diabetes Care. 2003;26:510–513. doi: 10.2337/diacare.26.2.510. [DOI] [PubMed] [Google Scholar]
  • 3.Finfer S, Chittock DR, Su SY-S, Blair D, Foster D, Dhingra V, Bellomo R, Cook D, Dodek P, Hébert WR, Heritier S, Heyland DK, McArthur C, McDonald E, Mitchell I, Myburgh JA, Norton R, Potter J, Robinson BG, Ronco JJ. Intensive versus conventional glucose control in critically ill patients. N Engl J Med. 2009;360:1283–1297. doi: 10.1056/NEJMoa0810625. [DOI] [PubMed] [Google Scholar]
  • 4.Stegenga ME, Vincent J-L, Vail GM, Xie J, Haney DJ, Williams MD, Bernard GR, van der Poll T. Diabetes does not alter mortality or hemostatic and inflammatory responses in patients with severe sepsis. Crit Care Med. 2010;38:539–545. doi: 10.1097/CCM.0b013e3181c02726. [DOI] [PubMed] [Google Scholar]
  • 5.Restrepo BI. Convergence of the tuberculosis and diabetes epidemics: renewal of old acquaintances. Clin Infect Dis. 2007;45:436–438. doi: 10.1086/519939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Joslin EP. The treatment of diabetes mellitus: with observations upon the disease based upon one thousand cases. Philadelphia: Lea & Febiger; 1916. [Google Scholar]
  • 7.Bertoni AG, Saydah S, Brancati FL. Diabetes and the risk of infection-related mortality in the U.S. Diabetes Care. 2001;24:1044–1049. doi: 10.2337/diacare.24.6.1044. [DOI] [PubMed] [Google Scholar]
  • 8.Chandler JR. Malignant external otitis. Laryngoscope. 1968;78:1257–1294. [Google Scholar]
  • 9.Ali T, Meade K, Anari S, ElBadawey MR, Zammit-Maempel I. Malignant otitis externa: case series. J Laryngol Otol. 2010;124:846–851. doi: 10.1017/S0022215110000691. [DOI] [PubMed] [Google Scholar]
  • 10.Carfrae MJ, Kesser BW. Malignant otitis externa. Otolaryngol Clin North Am. 2008;41:537–549. doi: 10.1016/j.otc.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 11.Khaira A, Gupta A, Rana DS, Gupta A, Bhalla A, Khullar D. Retrospective analysis of clinical profile prognostic factors and outcomes of 19 patients of emphysematous pyelonephritis. Int Urol Nephrol. 2009;41:959–966. doi: 10.1007/s11255-009-9552-y. [DOI] [PubMed] [Google Scholar]
  • 12.Michaeli J, Mogle P, Perlberg S, Heiman S, Caine M. Emphysematous pyelonephritis. J Urol. 1984;131:203–208. doi: 10.1016/s0022-5347(17)50309-2. [DOI] [PubMed] [Google Scholar]
  • 13.Ahlering TE, Boyd SD, Hamilton CL, Bragin SD, Chandrasoma PT, Lieskovsky G, Skinner DG. Emphysematous pyelonephritis: a 5-year experience with 13 patients. J Urol. 1985;134:1086–1088. doi: 10.1016/s0022-5347(17)47635-x. [DOI] [PubMed] [Google Scholar]
  • 14.Cook DJ, Achong MR, Dobranowski J. Emphysematous pyelonephritis. Complicated urinary tract infection in diabetes. Diabetes Care. 1989;12:229–232. doi: 10.2337/diacare.12.3.229. [DOI] [PubMed] [Google Scholar]
  • 15.Esper AM, Moss M, Martin GS. The effect of diabetes mellitus on organ dysfunction with sepsis: an epidemiological study. Crit Care. 2009;13:R18. doi: 10.1186/cc7717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Thomsen RW, Hundborg HH, Lervang H-H, Johnsen SP, Sørensen HT, Schønheyder HC. Diabetes and outcome of community-acquired pneumococcal bacteremia: a 10-year population-based cohort study. Diabetes Care. 2004;27:70–76. doi: 10.2337/diacare.27.1.70. [DOI] [PubMed] [Google Scholar]
  • 17.Tsai F-C, Huang Y-T, Chang L-Y, Wang J-T. Pyogenic liver abscess as endemic disease, Taiwan. Emerg Infect Dis. 2008;14:1592–1600. doi: 10.3201/eid1410.071254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gregory JE, Golden A, Haymaker W. Mucormycosis of the central nervous system: a report of three cases. Bull Johns Hopkins Hosp. 1943;73:405–419. [Google Scholar]
  • 19.Mallis A, Mastronikolis SN, Naxakis SS, Papadas AT. Rhinocerebral mucormycosis: an update. Eur Rev Med Pharmacol Sci. 2010;14:987–992. [PubMed] [Google Scholar]
  • 20.Cheng AC, Currie BJ. Melioidosis: epidemiology, pathophysiology, and management. Clin Microbiol Rev. 2005;18:383–416. doi: 10.1128/CMR.18.2.383-416.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dooley KE, Chaisson RE. Tuberculosis and diabetes mellitus: convergence of two epidemics. Lancet Infect Dis. 2009;9:737–746. doi: 10.1016/S1473-3099(09)70282-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fine MJ, Smith MA, Carson CA, Mutha SS, Sankey SS, Weissfeld LA, Kapoor WN. Prognosis and outcomes of patients with community-acquired pneumonia. A meta-analysis. JAMA. 1996;275:134–141. [PubMed] [Google Scholar]
  • 23.Thomsen RW, Hundborg HH, Lervang H-H, Johnsen SP, Schønheyder HC, Sørensen HT. Diabetes mellitus as a risk and prognostic factor for community-acquired bacteremia due to enterobacteria: a 10-year, population-based study among adults. Clin Infect Dis. 2005;40:628–631. doi: 10.1086/427699. [DOI] [PubMed] [Google Scholar]
  • 24.Kornum JB, Thomsen RW, Riis A, Lervang H-H, Schønheyder HC, Sørensen HT. Type 2 diabetes and pneumonia outcomes: a population-based cohort study. Diabetes Care. 2007;30:2251–2257. doi: 10.2337/dc06-2417. [DOI] [PubMed] [Google Scholar]
  • 25.Falagas ME, Alexiou VG, Giannopoulou KP, Siempos II. Risk factors for mortality in patients with emphysematous pyelonephritis: a meta-analysis. J Urol. 2007;178:880–885. doi: 10.1016/j.juro.2007.05.017. [DOI] [PubMed] [Google Scholar]
  • 26.Vincent J-L, Preiser J-C, Sprung CL, Moreno R, Sakr Y. Insulin-treated diabetes is not associated with increased mortality in critically ill patients. Crit Care. 2010;14:R12. doi: 10.1186/cc8866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Michalia M, Kompoti M, Koutsikou A, Paridou A, Giannopoulou P, Trikka-Graphakos E, Clouva-Molyvdas P. Diabetes mellitus is an independent risk factor for ICU-acquired bloodstream infections. Intensive Care Med. 2009;35:448–454. doi: 10.1007/s00134-008-1288-0. [DOI] [PubMed] [Google Scholar]
  • 28.Tsai CL, Lee CC, Ma MH, Fang CC, Chen SY, Chen WJ, Chang SC, Mehta SH. Impact of diabetes on mortality among patients with community-acquired bacteremia. J Infect. 2007;55:27–33. doi: 10.1016/j.jinf.2007.01.004. [DOI] [PubMed] [Google Scholar]
  • 29.McAlister FA, Majumdar SR, Blitz S, Rowe BH, Romney J, Marrie TJ. The relation between hyperglycemia and outcomes in 2,471 patients admitted to the hospital with community-acquired pneumonia. Diabetes Care. 2005;28:810–815. doi: 10.2337/diacare.28.4.810. [DOI] [PubMed] [Google Scholar]
  • 30.Kaplan V, Angus DC, Griffin MF, Clermont G, Scott Watson R, Linde-Zwirble WT. Hospitalized community-acquired pneumonia in the elderly: age- and sex-related patterns of care and outcome in the United States. Am J Respir Crit Care Med. 2002;165:766–772. doi: 10.1164/ajrccm.165.6.2103038. [DOI] [PubMed] [Google Scholar]
  • 31.Cooper G, Platt R. Staphylococcus aureus bacteremia in diabetic patients. Endocarditis and mortality. Am J Med. 1982;73:658–662. doi: 10.1016/0002-9343(82)90407-7. [DOI] [PubMed] [Google Scholar]
  • 32.Libert M, Elkholti M, Massaut J, Karmali R, Mascart G, Cherifi S. Risk factors for meticillin resistance and outcome of Staphylococcus aureus bloodstream infection in a Belgian university hospital. J Hosp Infect. 2008;68:17–24. doi: 10.1016/j.jhin.2007.08.021. [DOI] [PubMed] [Google Scholar]
  • 33.Carton JA, Maradona JA, Nuño FJ, Fernandez-Alvarez R, Pérez-Gonzalez F, Asensi V. Diabetes mellitus and bacteraemia: a comparative study between diabetic and non-diabetic patients. Eur J Med. 1992;1:281–287. [PubMed] [Google Scholar]
  • 34.Vardakas KZ, Siempos II, Falagas ME. Diabetes mellitus as a risk factor for nosocomial pneumonia and associated mortality. Diabet Med. 2007;24:1168–1171. doi: 10.1111/j.1464-5491.2007.02234.x. [DOI] [PubMed] [Google Scholar]
  • 35.Graham BB, Keniston A, Gajic O, Trillo Alvarez CA, Medvedev S, Douglas IS. Diabetes mellitus does not adversely affect outcomes from a critical illness. Crit Care Med. 2010;38:16–24. doi: 10.1097/CCM.0b013e3181b9eaa5. [DOI] [PubMed] [Google Scholar]
  • 36.Gong MN, Thompson BT, Williams P, Pothier L, Boyce PD, Christiani DC. Clinical predictors of and mortality in acute respiratory distress syndrome: potential role of red cell transfusion. Crit Care Med. 2005;33:1191–1198. doi: 10.1097/01.ccm.0000165566.82925.14. [DOI] [PubMed] [Google Scholar]
  • 37.Moss M, Guidot DM, Steinberg KP, Duhon GF, Treece P, Wolken R, Hudson LD, Parsons PE. Diabetic patients have a decreased incidence of acute respiratory distress syndrome. Crit Care Med. 2000;28:2187–2192. doi: 10.1097/00003246-200007000-00001. [DOI] [PubMed] [Google Scholar]
  • 38.Koh GCKW, Maude RR, Schreiber MF, Limmathurotsakul D, Wiersinga WJ, Wuthiekanun V, Lee SJ, Mahavanakul W, Chaowagul W, Chierakul W, White NJ, van der Poll T, Day NP, Dougan G, Peacock SJ. Glyburide is anti-inflammatory and associated with reduced mortality in melioidosis. Clin Infect Dis. 2011;52:717–725. doi: 10.1093/cid/ciq192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Watson RS, Angus DC. Assessing outcomes in critical care. J Intensive Care Med. 2002;17:103–111. [Google Scholar]
  • 40.Schisterman EF, Cole SR, Platt RW. Overadjustment bias and unnecessary adjustment in epidemiologic studies. Epidemiology. 2009;20:488–495. doi: 10.1097/EDE.0b013e3181a819a1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hennekens CH, Buring JE. Epidemiology in medicine. Philadelphia: Lippincott Williams & Wilkins; 1987. p. 287. [Google Scholar]
  • 42.Lassar O. Ernährungstherapie bei Hautkrankheiten. Dermatologische Zeitschrift. 1904;11:189–209. [Google Scholar]
  • 43.Handmann E. Über die Ursache der verminderten Resistenz des Diabetikers gegen Infektionen. Deutsches Archiv Für Klinische Medizin. 1911;102:1–14. [Google Scholar]
  • 44.Da Costa JC., Jr The opsonic index in diabetes mellitus: a preliminary record of the findings in 22 cases of glycosuria, with remarks on the technique of the opsonin test and on its clinical utility. Am J Med Sci. 1907;134:57–70. [Google Scholar]
  • 45.Balasoiu D, van Kessel KC, van Kats-Renaud HJ, Collet TJ, Hoepelman AI. Granulocyte function in women with diabetes and asymptomatic bacteriuria. Diabetes Care. 1997;20:392–395. doi: 10.2337/diacare.20.3.392. [DOI] [PubMed] [Google Scholar]
  • 46.Pickup JC, Mattock MB, Chusney GD, Burt D. NIDDM as a disease of the innate immune system: association of acute-phase reactants and interleukin-6 with metabolic syndrome X. Diabetologia. 1997;40:1286–1292. doi: 10.1007/s001250050822. [DOI] [PubMed] [Google Scholar]
  • 47.Myśliwska J, Zorena K, Bakowska A, Skuratowicz-Kubica A, Myśliwski A. Significance of tumor necrosis factor alpha in patients with long-standing type-I diabetes mellitus. Horm Metab Res. 1998;30:158–161. doi: 10.1055/s-2007-978857. [DOI] [PubMed] [Google Scholar]
  • 48.Zozuliñska D, Majchrzak A, Sobieska M, Wiktorowicz K, Wierusz-Wysocka B. Serum interleukin-8 level is increased in diabetic patients. Diabetologia. 1999;42:117–118. doi: 10.1007/s001250051124. [DOI] [PubMed] [Google Scholar]
  • 49.Yende S, van der Poll T, Lee M, Huang DT, Newman AB, Kong L, Kellum JA, Harris TB, Bauer D, Satterfield S, Angus DC. The influence of pre-existing diabetes mellitus on the host immune response and outcome of pneumonia: analysis of two multicentre cohort studies. Thorax. 2010;65:870–877. doi: 10.1136/thx.2010.136317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Rao AK, Chouhan V, Chen X, Sun L, Boden G. Activation of the tissue factor pathway of blood coagulation during prolonged hyperglycemia in young healthy men. Diabetes. 1999;48:1156–1161. doi: 10.2337/diabetes.48.5.1156. [DOI] [PubMed] [Google Scholar]
  • 51.Stegenga ME, van der Crabben SN, Levi M, de Vos AF, Tanck MW, Sauerwein HP, van der Poll T. Hyperglycemia stimulates coagulation, whereas hyperinsulinemia impairs fibrinolysis in healthy humans. Diabetes. 2006;55:1807–1812. doi: 10.2337/db05-1543. [DOI] [PubMed] [Google Scholar]
  • 52.Geerlings SE, Brouwer EC, Van Kessel KC, Gaastra W, Stolk RP, Hoepelman AI. Cytokine secretion is impaired in women with diabetes mellitus. Eur J Clin Invest. 2000;30:995–1001. doi: 10.1046/j.1365-2362.2000.00745.x. [DOI] [PubMed] [Google Scholar]
  • 53.Aird WC. The role of the endothelium in severe sepsis and multiple organ dysfunction syndrome. Blood. 2003;101:3765–3777. doi: 10.1182/blood-2002-06-1887. [DOI] [PubMed] [Google Scholar]
  • 54.Schuetz P, Yano K, Sorasaki M, Ngo L, St Hilaire M, Lucas JM, Aird W, Shapiro NI. Influence of diabetes on endothelial cell response during sepsis. Diabetologia. 2011;54:996–1003. doi: 10.1007/s00125-011-2059-y. [DOI] [PubMed] [Google Scholar]
  • 55.Neelamegham S, Taylor AD, Burns AR, Smith CW, Simon SI. Hydrodynamic shear shows distinct roles for LFA-1 and Mac-1 in neutrophil adhesion to intercellular adhesion molecule-1. Blood. 1998;92:1626–1638. [PubMed] [Google Scholar]
  • 56.Hentzen ER, Neelamegham S, Kansas GS, Benanti JA, McIntire LV, Smith CW, Simon SI. Sequential binding of CD11a/CD18 and CD11b/CD18 defines neutrophil capture and stable adhesion to intercellular adhesion molecule-1. Blood. 2000;95:911–920. [PubMed] [Google Scholar]
  • 57.Li J, Jin H-B, Sun Y-M, Su Y, Wang L-F. KB-R7943 inhibits high glucose-induced endothelial ICAM-1 expression and monocyte-endothelial adhesion. Biochem Biophys Res Commun. 2010;392:516–519. doi: 10.1016/j.bbrc.2009.12.183. [DOI] [PubMed] [Google Scholar]
  • 58.Takami S, Yamashita S, Kihara S, Kameda-Takemura K, Matsuzawa Y. High concentration of glucose induces the expression of intercellular adhesion molecule-1 in human umbilical vein endothelial cells. Atherosclerosis. 1998;138:35–41. doi: 10.1016/s0021-9150(97)00286-4. [DOI] [PubMed] [Google Scholar]
  • 59.Altannavch TS, Roubalová K, Kucera P, Andel M. Effect of high glucose concentrations on expression of ELAM-1, VCAM-1 and ICAM-1 in HUVEC with and without cytokine activation. Physiol Res. 2004;53:77–82. [PubMed] [Google Scholar]
  • 60.Andersen B, Goldsmith GH, Spagnuolo PJ. Neutrophil adhesive dysfunction in diabetes mellitus; the role of cellular and plasma factors. J Lab Clin Med. 1988;111:275–285. [PubMed] [Google Scholar]
  • 61.Delamaire M, Maugendre D, Moreno M, Le Goff MC, Allannic H, Genetet B. Impaired leucocyte functions in diabetic patients. Diabet Med. 1997;14:29–34. doi: 10.1002/(SICI)1096-9136(199701)14:1<29::AID-DIA300>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  • 62.Morigi M, Angioletti S, Imberti B, Donadelli R, Micheletti G, Figliuzzi M, Remuzzi A, Zoja C, Remuzzi G. Leukocyte-endothelial interaction is augmented by high glucose concentrations and hyperglycemia in a NF-κB-dependent fashion. J Clin Invest. 1998;101:1905–1915. doi: 10.1172/JCI656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Taki H, Kashiwagi A, Tanaka Y, Horiike K. Expression of intercellular adhesion molecules 1 (ICAM-1) via an osmotic effect in human umbilical vein endothelial cells exposed to high glucose medium. Life Sci. 1996;58:1713–1721. doi: 10.1016/0024-3205(96)00151-8. [DOI] [PubMed] [Google Scholar]
  • 64.Park CW, Kim JH, Lee JH, Kim YS, Ahn HJ, Shin YS, Kim SY, Choi EJ, Chang YS, Bang BK. High glucose-induced intercellular adhesion molecule-1 (ICAM-1) expression through an osmotic effect in rat mesangial cells is PKC-NF-κB-dependent. Diabetologia. 2000;43:1544–1553. doi: 10.1007/s001250051567. [DOI] [PubMed] [Google Scholar]
  • 65.Boyden S. The chemotactic effect of mixtures of antibody and antigen on polymorphonuclear leucocytes. J Exp Med. 1962;115:453–466. doi: 10.1084/jem.115.3.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mowat A, Baum J. Chemotaxis of polymorphonuclear leukocytes from patients with diabetes mellitus. N Engl J Med. 1971;284:621–627. doi: 10.1056/NEJM197103252841201. [DOI] [PubMed] [Google Scholar]
  • 67.Valerius NH, Eff C, Hansen NE, Karle H, Nerup J, Søeberg B, Sørensen SF. Neutrophil and lymphocyte function in patients with diabetes mellitus. Acta Med Scand. 1982;211:463–467. doi: 10.1111/j.0954-6820.1982.tb01983.x. [DOI] [PubMed] [Google Scholar]
  • 68.Nelson RD, Quie PG, Simmons RL. Chemotaxis under agarose: a new and simple method for measuring chemotaxis and spontaneous migration of human polymorphonuclear leukocytes and monocytes. J Immunol. 1975;115:1650–1656. [PubMed] [Google Scholar]
  • 69.Tater D, Tepaut B, Bercovici JP, Youinou P. Polymorphonuclear cell derangements in type I diabetes. Horm Metab Res. 1987;19:642–647. doi: 10.1055/s-2007-1011899. [DOI] [PubMed] [Google Scholar]
  • 70.Bybee JD, Rogers DE. The phagocytic activity of polymorphonuclear leukocytes obtained from patients with diabetes mellitus. J Lab Clin Med. 1964;64:1–13. [PubMed] [Google Scholar]
  • 71.Bagdade JD, Root RK, Bulger RJ. Impaired leukocyte function in patients with poorly controlled diabetes. Diabetes. 1974;23:9–15. doi: 10.2337/diab.23.1.9. [DOI] [PubMed] [Google Scholar]
  • 72.Bagdade JD. Phagocytic and microbicidal function in diabetes mellitus. Acta Endocrinol Suppl (Copenh) 1976;205:27–34. [PubMed] [Google Scholar]
  • 73.Alexiewicz JM, Kumar D, Smogorzewski M, Klin M, Massry SG. Polymorphonuclear leukocytes in non-insulin-dependent diabetes mellitus: abnormalities in metabolism and function. Ann Intern Med. 1995;123:919–924. doi: 10.7326/0003-4819-123-12-199512150-00004. [DOI] [PubMed] [Google Scholar]
  • 74.Davidson NJ, Sowden JM, Fletcher J. Defective phagocytosis in insulin controlled diabetics: evidence for a reaction between glucose and opsonising proteins. J Clin Pathol. 1984;37:783–786. doi: 10.1136/jcp.37.7.783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Dziatkowiak H, Kowalska M, Denys A. Phagocytic and bactericidal activity of granulocytes in diabetic children. Diabetes. 1982;31:1041–1043. doi: 10.2337/diacare.31.12.1041. [DOI] [PubMed] [Google Scholar]
  • 76.Tan JS, Anderson JL, Watanakunakorn C, Phair JP. Neutrophil dysfunction in diabetes mellitus. J Lab Clin Med. 1975;85(1):26–33. [PubMed] [Google Scholar]
  • 77.Bagdade JD, Nielson KL, Bulger RJ. Reversible abnormalities in phagocytic function in poorly controlled diabetic patients. Am J Med Sci. 1972;263:451–456. doi: 10.1097/00000441-197206000-00005. [DOI] [PubMed] [Google Scholar]
  • 78.Wilson RM, Reeves WG. Neutrophil phagocytosis and killing in insulin-dependent diabetes. Clin Exp Immunol. 1986;63:478–484. [PMC free article] [PubMed] [Google Scholar]
  • 79.Shah SV, Wallin JD, Eilen SD. Chemiluminescence and superoxide anion production by leukocytes from diabetic patients. J Clin Endocrinol Metab. 1983;57:402–409. doi: 10.1210/jcem-57-2-402. [DOI] [PubMed] [Google Scholar]
  • 80.Wykretowicz A, Wierusz-Wysocka B, Wysocki J, Szczepanik A, Wysocki H. Impairment of the oxygen-dependent microbicidal mechanisms of polymorphonuclear neutrophils in patients with type 2 diabetes is not associated with increased susceptibility to infection. Diabetes Res Clin Pract. 1993;19:195–201. doi: 10.1016/0168-8227(93)90114-k. [DOI] [PubMed] [Google Scholar]
  • 81.Sato N, Kashima K, Tanaka Y, Shimizu H, Mori M. Effect of granulocyte-colony stimulating factor on generation of oxygen-derived free radicals and myeloperoxidase activity in neutrophils from poorly controlled NIDDM patients. Diabetes. 1997;46:133–137. doi: 10.2337/diab.46.1.133. [DOI] [PubMed] [Google Scholar]
  • 82.Daoud AK, Tayyar MA, Fouda IM, Harfeil NA. Effects of diabetes mellitus vs. in vitro hyperglycemia on select immune cell functions. J Immunotoxicol. 2009;6:36–41. doi: 10.1080/15476910802604564. [DOI] [PubMed] [Google Scholar]
  • 83.Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535. doi: 10.1126/science.1092385. [DOI] [PubMed] [Google Scholar]
  • 84.Grinberg N, Elazar S, Rosenshine I, Shpigel NY. β-Hydroxybutyrate abrogates formation of bovine neutrophil extracellular traps and bactericidal activity against mammary pathogenic Escherichia coli. Infect Immun. 2008;76:2802–2807. doi: 10.1128/IAI.00051-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hill HR, Augustine NH, Rallison ML, Santos JI. Defective monocyte chemotactic responses in diabetes mellitus. J Clin Immunol. 1983;3:70–77. doi: 10.1007/BF00919141. [DOI] [PubMed] [Google Scholar]
  • 86.Geisler C, Almdal T, Bennedsen J, Rhodes JM, Kølendorf K. Monocyte functions in diabetes mellitus. Acta Pathol Microbiol Immunol Scand C. 1982;90:33–37. doi: 10.1111/j.1699-0463.1982.tb01414.x. [DOI] [PubMed] [Google Scholar]
  • 87.Katz S, Klein B, Elian I, Fishman P, Djaldetti M. Phagocytotic activity of monocytes from diabetic patients. Diabetes Care. 1983;6:479–482. doi: 10.2337/diacare.6.5.479. [DOI] [PubMed] [Google Scholar]
  • 88.Nandy D, Janardhanan R, Mukhopadhyay D, Basu A. Effect of hyperglycemia on human monocyte activation. J Investig Med. 2011;59:661–667. doi: 10.231/JIM.0b013e31820ee432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Rattan V, Shen Y, Sultana C, Kumar D, Kalra VK. Glucose-induced transmigration of monocytes is linked to phosphorylation of PECAM-1 in cultured endothelial cells. Am J Physiol. 1996;271:E711–E717. doi: 10.1152/ajpendo.1996.271.4.E711. [DOI] [PubMed] [Google Scholar]
  • 90.Kitahara M, Eyre HJ, Lynch RE, Rallison ML, Hill HR. Metabolic activity of diabetic monocytes. Diabetes. 1980;29:251–256. doi: 10.2337/diab.29.4.251. [DOI] [PubMed] [Google Scholar]
  • 91.Foss-Freitas MC, Foss NT, Donadi EA, Foss MC. In vitro TNF-alpha and IL-6 production by adherent peripheral blood mononuclear cells obtained from type 1 and type 2 diabetic patients evaluated according to the metabolic control. Ann N Y Acad Sci. 2006;1079:177–180. doi: 10.1196/annals.1375.027. [DOI] [PubMed] [Google Scholar]
  • 92.MacCuish AC, Urbaniak SJ, Campbell CJ, Duncan LJ, Irvine WJ. Phytohemagglutinin transformation and circulating lymphocyte subpopulations in insulin-dependent diabetic patients. Diabetes. 1974;23:708–712. doi: 10.2337/diab.23.8.708. [DOI] [PubMed] [Google Scholar]
  • 93.Speert DP, Silva J., Jr Abnormalities of in vitro lymphocyte response to mitogens in diabetic children during acute ketoacidosis. Am J Dis Child. 1978;132:1014–1017. doi: 10.1001/archpedi.1978.02120350078016. [DOI] [PubMed] [Google Scholar]
  • 94.Eibl N, Spatz M, Fischer GF, Mayr WR, Samstag A, Wolf HM, Schernthaner G, Eibl MM. Impaired primary immune response in type-1 diabetes: results from a controlled vaccination study. Clin Immunol. 2002;103:249–259. doi: 10.1006/clim.2002.5220. [DOI] [PubMed] [Google Scholar]
  • 95.Spatz M, Eibl N, Hink S, Wolf HM, Fischer GF, Mayr WR, Schernthaner G, Eibl MM. Impaired primary immune response in type-1 diabetes. Functional impairment at the level of APCs and T-cells. Cell Immunol. 2003;221:15–26. doi: 10.1016/s0008-8749(03)00043-1. [DOI] [PubMed] [Google Scholar]
  • 96.Casey J, Sturm C., Jr Impaired response of lymphocytes from non-insulin-dependent diabetics to staphage lysate and tetanus antigen. J Clin Microbiol. 1982;15:109–114. doi: 10.1128/jcm.15.1.109-114.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Casey JI, Heeter BJ, Klyshevich KA. Impaired response of lymphocytes of diabetic subjects to antigen of Staphylococcus aureus. J Infect Dis. 1977;136:495–501. doi: 10.1093/infdis/136.4.495. [DOI] [PubMed] [Google Scholar]
  • 98.Da Costa JC, Jr, Beardsley EJG. The resistance of diabetics to bacterial infection: a study of the opsonophagocytic properties of the blood in 74 cases of diabetes mellitus and related conditions. Am J Med Sci. 1908;136:361–373. [Google Scholar]
  • 99.Liberatore RDR, Jr, Barbosa SFC, Das Graças Alkimin M, Bellinati-Pires R, Florido MPC, Isaac L, Kirschfink M, Grumach AS. Is immunity in diabetic patients influencing the susceptibility to infections? Immunoglobulins, complement and phagocytic function in children and adolescents with type 1 diabetes mellitus. Pediatr Diabetes. 2005;6:206–212. doi: 10.1111/j.1399-543X.2005.00136.x. [DOI] [PubMed] [Google Scholar]
  • 100.Moen JK, Reimann HA. Immune reactions in diabetes. Arch Intern Med. 1933;51:789–795. [Google Scholar]
  • 101.Richardson R. Immunity in diabetes: influence of diabetes on the development of antibacterial properties in the blood. J Clin Invest. 1933;12:1143–1149. doi: 10.1172/JCI100565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Fiçicioğlu C, Mikla S, Midilli K, Aydin A, Cam H, Erğin S. Reduced immune response to hepatitis B vaccine in children with insulin dependent diabetes. Acta Paediatr Jpn. 1995;37:687–690. doi: 10.1111/j.1442-200x.1995.tb03404.x. [DOI] [PubMed] [Google Scholar]
  • 103.Pozzilli P, Arduini P, Visalli N, Sutherland J, Pezzella M, Galli C, Corradini SG, Biasio L, Gale EA, Andreani D. Reduced protection against hepatitis B virus following vaccination in patients with type 1 (insulin-dependent) diabetes. Diabetologia. 1987;30:817–819. doi: 10.1007/BF00275749. [DOI] [PubMed] [Google Scholar]
  • 104.Alavian S-M, Tabatabaei SV. The effect of diabetes mellitus on immunological response to hepatitis B virus vaccine in individuals with chronic kidney disease: a meta-analysis of current literature. Vaccine. 2010;28:3773–3777. doi: 10.1016/j.vaccine.2010.03.038. [DOI] [PubMed] [Google Scholar]
  • 105.Bouter KP, Diepersloot RJ, Wismans PJ, Gmelig Meyling FH, Hoekstra JB, Heijtink RA, van Hattum J. Humoral immune response to a yeast-derived hepatitis B vaccine in patients with type 1 diabetes mellitus. Diabet Med. 1992;9:66–69. doi: 10.1111/j.1464-5491.1992.tb01717.x. [DOI] [PubMed] [Google Scholar]
  • 106.Douvin C, Simon D, Charles MA, Deforges L, Bierling P, Lehner V, Budkowska A, Dhumeaux D. Hepatitis B vaccination in diabetic patients. Randomized trial comparing recombinant vaccines containing and not containing pre-S2 antigen. Diabetes Care. 1997;20:148–151. doi: 10.2337/diacare.20.2.148. [DOI] [PubMed] [Google Scholar]
  • 107.Brydak LB, Machala M. Humoral immune response to influenza vaccination in patients from high risk groups. Drugs. 2000;60:35–53. doi: 10.2165/00003495-200060010-00004. [DOI] [PubMed] [Google Scholar]
  • 108.Pozzilli P, Gale EA, Visalli N, Baroni M, Crovari P, Frighi V, Cavallo MG, Andreani D. The immune response to influenza vaccination in diabetic patients. Diabetologia. 1986;29:850–854. doi: 10.1007/BF00870139. [DOI] [PubMed] [Google Scholar]
  • 109.Muszkat M, Friedman G, Dannenberg HD, Greenbaum E, Lipo M, Heymann Y, Zakay-Rones Z, Ben-Yehuda A. Response to influenza vaccination in community and in nursing home residing elderly: relation to clinical factors. Exp Gerontol. 2003;38:1199–1203. doi: 10.1016/j.exger.2003.07.004. [DOI] [PubMed] [Google Scholar]
  • 110.Tamer A, Karabay O, Ekerbicer H, Tahtaci M, Selam B, Celebi H. Impaired immunity against tetanus in type 2 diabetes. Med Sci Monit. 2005;11:CR580–CR584. [PubMed] [Google Scholar]
  • 111.Kiliç D, Kaygusuz S, Saygun M, Cakmak A, Uzer H, Doğanci L. Seroprevalence of tetanus immunity among noninsulin-dependent diabetes mellitus patients. J Diabetes Complications. 2003;17:258–263. doi: 10.1016/s1056-8727(02)00255-6. [DOI] [PubMed] [Google Scholar]
  • 112.Beam TR, Jr, Crigler ED, Goldman JK, Schiffman G. Antibody response to polyvalent pneumococcal polysaccharide vaccine in diabetics. JAMA. 1980;244:2621–2624. [PubMed] [Google Scholar]
  • 113.Mijovic CH, Fletcher JA, Bradwell AR, Barnett AH. Low C4 levels in type 1 (insulin-dependent) diabetes. Diabetologia. 1987;30:824. doi: 10.1007/BF00275752. [DOI] [PubMed] [Google Scholar]
  • 114.Vergani D, Johnston C, B-Abdullah N, Barnett AH. Low serum C4 concentrations: an inherited predisposition to insulin dependent diabetes? Br Med J (Clin Res Ed) 1983;286:926–928. doi: 10.1136/bmj.286.6369.926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Jenhani F, Bardi R, Gorgi Y, Ayed K, Jeddi M. C4 polymorphism in multiplex families with insulin dependent diabetes in the Tunisian population: standard C4 typing methods and RFLP analysis. J Autoimmun. 1992;5:149–160. doi: 10.1016/0896-8411(92)90196-w. [DOI] [PubMed] [Google Scholar]
  • 116.Hernández-Mijares A, Jarabo-Bueno MM, López-Ruiz A, Solá-Izquierdo E, Morillas-Ariño C, Martínez-Triguero ML. Levels of C3 in patients with severe, morbid and extreme obesity: its relationship to insulin resistance and different cardiovascular risk factors. Int J Obes (Lond) 2007;31:927–932. doi: 10.1038/sj.ijo.0803543. [DOI] [PubMed] [Google Scholar]
  • 117.Karlsson E, Shaat N, Groop L. Can complement factors 5 and 8 and transthyretin be used as biomarkers for MODY 1 (HNF4A-MODY) and MODY 3 (HNF1A-MODY)? Diabet Med. 2008;25:788–791. doi: 10.1111/j.1464-5491.2008.02467.x. [DOI] [PubMed] [Google Scholar]
  • 118.Davin JC, Bouts AH, Krediet RT, van der Weel M, Weening RS, Groothoff J, Out TA. IgG glycation and function during continuous ambulatory peritoneal dialysis. Nephrol Dial Transplant. 1997;12:310–314. doi: 10.1093/ndt/12.2.310. [DOI] [PubMed] [Google Scholar]
  • 119.Hostetter MK. Handicaps to host defense. Effects of hyperglycemia on C3 and Candida albicans. Diabetes. 1990;39:271–275. doi: 10.2337/diab.39.3.271. [DOI] [PubMed] [Google Scholar]
  • 120.Warren S. Pathology of diabetes mellitus. Philadelphia: Lea & Febiger; 1930. [Google Scholar]
  • 121.Rayfield EJ, Ault MJ, Keusch GT, Brothers MJ, Nechemias C, Smith H. Infection and diabetes: the case for glucose control. Am J Med. 1982;72:439–450. doi: 10.1016/0002-9343(82)90511-3. [DOI] [PubMed] [Google Scholar]
  • 122.King JT, Jr, Goulet JL, Perkal MF, Rosenthal RA. Glycemic control and infections in patients with diabetes undergoing noncardiac surgery. Ann Surg. 2011;253:158–165. doi: 10.1097/SLA.0b013e3181f9bb3a. [DOI] [PubMed] [Google Scholar]
  • 123.Davis TME, Weerarathne T, Foong Y, Mason C, Davis WA. Community-acquired infections in type 2 diabetic patients and their nondiabetic partners. The Fremantle Diabetes Study. J Diabetes Complications. 2005;19:259–263. doi: 10.1016/j.jdiacomp.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 124.Stegenga ME, van der Crabben SN, Blümer RME, Levi M, Meijers JCM, Serlie MJ, Tanck MWT, Sauerwein HP, van der Poll T. Hyperglycemia enhances coagulation and reduces neutrophil degranulation, whereas hyperinsulinemia inhibits fibrinolysis during human endotoxemia. Blood. 2008;112:82–89. doi: 10.1182/blood-2007-11-121723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Stegenga ME, van der Crabben SN, Dessing MC, Pater JM, van den Pangaart PS, de Vos AF, Tanck MW, Roos D, Sauerwein HP, van der Poll T. Effect of acute hyperglycaemia and/or hyperinsulinaemia on proinflammatory gene expression, cytokine production and neutrophil function in humans. Diabet Med. 2008;25:157–164. doi: 10.1111/j.1464-5491.2007.02348.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Dandona P, Aljada A, Mohanty P, Ghanim H, Hamouda W, Assian E, Ahmad S. Insulin inhibits intranuclear nuclear factor κB and stimulates IκB in mononuclear cells in obese subjects: evidence for an anti-inflammatory effect? J Clin Endocrinol Metab. 2001;86:3257–3265. doi: 10.1210/jcem.86.7.7623. [DOI] [PubMed] [Google Scholar]
  • 127.Hansen TK, Thiel S, Wouters PJ, Christiansen JS, Van den Berghe G. Intensive insulin therapy exerts antiinflammatory effects in critically ill patients and counteracts the adverse effect of low mannose-binding lectin levels. J Clin Endocrinol Metab. 2003;88:1082–1088. doi: 10.1210/jc.2002-021478. [DOI] [PubMed] [Google Scholar]
  • 128.Esmann V. The diabetic leukocyte. Enzyme. 1972;13:32–55. [PubMed] [Google Scholar]
  • 129.Leroux JP, Marchand JC, Hong Tuan Ha R, Cartier P. The influence of insulin on glucose permeability and metabolism of human granulocytes. Eur J Biochem. 1975;58:367–373. doi: 10.1111/j.1432-1033.1975.tb02383.x. [DOI] [PubMed] [Google Scholar]
  • 130.Sbarra AJ, Karnovsky ML. The biochemical basis of phagocytosis. I. Metabolic changes during the ingestion of particles by polymorphonuclear leukocytes. J Biol Chem. 1959;234:1355–1362. [PubMed] [Google Scholar]
  • 131.Maianski NA, Geissler J, Srinivasula SM, Alnemri ES, Roos D, Kuijpers TW. Functional characterization of mitochondria in neutrophils: a role restricted to apoptosis. Cell Death Differ. 2004;11:143–153. doi: 10.1038/sj.cdd.4401320. [DOI] [PubMed] [Google Scholar]
  • 132.van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R. Intensive insulin therapy in the critically ill patients. N Engl J Med. 2001;345:1359–1367. doi: 10.1056/NEJMoa011300. [DOI] [PubMed] [Google Scholar]
  • 133.van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, Van Wijngaerden E, Bobbaers H, Bouillon R. Intensive insulin therapy in the medical ICU. N Engl J Med. 2006;354:449–461. doi: 10.1056/NEJMoa052521. [DOI] [PubMed] [Google Scholar]
  • 134.Griesdale DEG, de Souza RJ, van Dam RM, Heyland DK, Cook DJ, Malhotra A, Dhaliwal R, Henderson WR, Chittock DR, Finfer S, Talmor D. Intensive insulin therapy and mortality among critically ill patients: a meta-analysis including NICE-SUGAR study data. CMAJ. 2009;180:821–827. doi: 10.1503/cmaj.090206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.UK Prospective Diabetes Study (UKPDS) Group Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34) Lancet. 1998;352:854–865. [PubMed] [Google Scholar]
  • 136.Dandona P, Aljada A, Ghanim H, Mohanty P, Tripathy C, Hofmeyer D, Chaudhuri A. Increased plasma concentration of macrophage migration inhibitory factor (MIF) and MIF mRNA in mononuclear cells in the obese and the suppressive action of metformin. J Clin Endocrinol Metab. 2004;89:5043–5047. doi: 10.1210/jc.2004-0436. [DOI] [PubMed] [Google Scholar]
  • 137.Fitzgerald E, Mathieu S, Ball A. Metformin associated lactic acidosis. BMJ. 2009;339:b3660. doi: 10.1136/bmj.b3660. [DOI] [PubMed] [Google Scholar]
  • 138.Jones GC, Macklin JP, Alexander WD. Contraindications to the use of metformin. BMJ. 2003;326:4–5. doi: 10.1136/bmj.326.7379.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Deakin AM, Payne AN, Blackwell GJ. Role of potassium channels in the regulation of cytokine release from THP-1 cells. Inflamm Res. 1994;41:C188–C190. [Google Scholar]
  • 140.Lamkanfi M, Mueller JL, Vitari AC, Misaghi S, Fedorova A, Deshayes K, Lee WP, Hoffman HM, Dixit VM. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J Cell Biol. 2009;187:61–70. doi: 10.1083/jcb.200903124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Standen NB, Quayle JM, Davies NW, Brayden JE, Huang Y, Nelson MT. Hyperpolarizing vasodilators activate ATP-sensitive K+ channels in arterial smooth muscle. Science. 1989;245:177–180. doi: 10.1126/science.2501869. [DOI] [PubMed] [Google Scholar]
  • 142.Landry DW, Oliver JA. The ATP-sensitive K+ channel mediates hypotension in endotoxemia and hypoxic lactic acidosis in dog. J Clin Invest. 1992;89:2071–2074. doi: 10.1172/JCI115820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Warrillow S, Egi M, Bellomo R. Randomized, double-blind, placebo-controlled crossover pilot study of a potassium channel blocker in patients with septic shock. Crit Care Med. 2006;34:980–985. doi: 10.1097/01.CCM.0000206114.19707.7C. [DOI] [PubMed] [Google Scholar]
  • 144.Morelli A, Lange M, Ertmer C, Broeking K, Van Aken H, Orecchioni A, Rocco M, Bachetoni A, Traber DL, Landoni G, Pietropaoli P, Westphal M. Glibenclamide dose response in patients with septic shock: effects on norepinephrine requirements, cardiopulmonary performance, and global oxygen transport. Shock. 2007;28:530–535. doi: 10.1097/shk.0b013e3180556a3c. [DOI] [PubMed] [Google Scholar]
  • 145.Mohanty P, Aljada A, Ghanim H, Hofmeyer D, Tripathy D, Syed T, Al-Haddad W, Dhindsa S, Dandona P. Evidence for a potent antiinflammatory effect of rosiglitazone. J Clin Endocrinol Metab. 2004;89:2728–2735. doi: 10.1210/jc.2003-032103. [DOI] [PubMed] [Google Scholar]
  • 146.Ghanim H, Garg R, Aljada A, Mohanty P, Kumbkarni Y, Assian E, Hamouda W, Dandona P. Suppression of nuclear factor-kappaB and stimulation of inhibitor kappaB by troglitazone: evidence for an anti-inflammatory effect and a potential antiatherosclerotic effect in the obese. J Clin Endocrinol Metab. 2001;86:1306–1312. doi: 10.1210/jcem.86.3.7309. [DOI] [PubMed] [Google Scholar]
  • 147.Lee S, Kim W, Kang KP, Moon S-O, Sung MJ, Kim DH, Kim HJ, Park SK. Agonist of peroxisome proliferator-activated receptor-gamma, rosiglitazone, reduces renal injury and dysfunction in a murine sepsis model. Nephrol Dial Transplant. 2005;20:1057–1065. doi: 10.1093/ndt/gfh705. [DOI] [PubMed] [Google Scholar]
  • 148.Wu W-T, Lee C-C, Lee C-J, Subeq Y-M, Lee R-P, Hsu B-G. Rosiglitazone ameliorates endotoxin-induced organ damage in conscious rats. Biol Res Nurs. 2011;13:38–43. doi: 10.1177/1099800409353358. [DOI] [PubMed] [Google Scholar]
  • 149.Stegenga ME, Florquin S, de Vos AF, van der Poll T. The thiazolidinedione ciglitazone reduces bacterial outgrowth and early inflammation during Streptococcus pneumoniae pneumonia in mice. Crit Care Med. 2009;37:614–618. doi: 10.1097/CCM.0b013e31819599b6. [DOI] [PubMed] [Google Scholar]
  • 150.Zingarelli B, Cook JA. Peroxisome proliferator-activated receptor-gamma is a new therapeutic target in sepsis and inflammation. Shock. 2005;23:393–399. doi: 10.1097/01.shk.0000160521.91363.88. [DOI] [PubMed] [Google Scholar]

Articles from European Journal of Clinical Microbiology & Infectious Diseases are provided here courtesy of Springer

RESOURCES