Abstract
Background
Sepsis, a complex disorder characterized by a dysregulated immune response to an inciting infection, affects over one million Americans annually. Dysglycemia during sepsis hospitalization confers increased risk of organ dysfunction and death, and novel targets for the treatment of sepsis and maintenance of glucose homeostasis are needed. Incretin hormones are secreted by enteroendocrine cells in response to enteral nutrients and potentiate insulin release from pancreatic β cells in a glucose-dependent manner, thereby reducing the risk of insulin-induced hypoglycemia. Incretin hormones also reduce systemic inflammation in preclinical studies, but studies of incretins in the setting of sepsis are limited.
Methods
In this bench-to-bedside mini-review, we detail the evidence to support incretin hormones as a therapeutic target in patients with sepsis. We performed a PubMed search using the medical subject headings “incretins,” “glucagon-like peptide-1,” “gastric inhibitory peptide,” “inflammation,” and “sepsis.”
Results
Incretin-based therapies decrease immune cell activation, inhibit proinflammatory cytokine release, and reduce organ dysfunction and mortality in preclinical models of sepsis. Several small clinical trials in critically ill patients have suggested potential benefit in glycemic control using exogenous incretin infusions, but these studies had limited power and were performed in mixed populations. Further clinical studies examining incretins specifically in septic populations are needed.
Conclusions
Targeting the incretin hormone axis in sepsis may provide a means of not only promoting euglycemia in sepsis but also attenuating the proinflammatory response and improving clinical outcomes.
This manuscript provides a bench-to-bedside review of the evidence supporting a potential therapeutic role for endogenous incretin hormones and exogenous incretin-derived medications in sepsis.
Sepsis is a life-threatening illness characterized by overwhelming systemic inflammation in response to an infectious insult (1, 2). Organ dysfunction in sepsis results from a combination of direct injury to host tissues from the invading pathogen and subsequent release of proinflammatory cytokines, including IL-1β, IL-6, and TNF-α (3, 4). No sepsis-specific therapies have proven successful to date (5, 6); rather, the treatments demonstrating the greatest benefit have been timely administrative of appropriate antimicrobial agents and supportive care (7, 8). Novel approaches to minimize tissue injury and organ dysfunction in the acute phase of sepsis are needed.
Dysglycemia in sepsis
Hyperglycemia is common in patients with sepsis, both with and without diabetes, with several contributing factors (9–12). Proinflammatory cytokines contribute to the development of peripheral insulin resistance and hyperglycemia in sepsis by inhibiting the expression and membrane translocation of glucose transporter type 4 in skeletal muscle and adipose tissues and impairing signaling through the insulin-receptor substrate-1 and protein kinase B pathways (13–15). The acute effects of proinflammatory cytokines on pancreatic insulin release are unclear, with some studies demonstrating augmentation of insulin secretion and others demonstrating suppression (16–19). Regardless, prolonged exposure to cytokines is injurious and triggers apoptosis in pancreatic β cells (20–22). Endogenous glucocorticoids, released in the physiologic response to stress, increase blood glucose levels by stimulating gluconeogenesis and glycogenolysis (23). Additionally, vasopressors and exogenous glucocorticoids, typically initiated to treat sepsis-induced hypotension and dysregulation of the hypothalamic–pituitary adrenal axis, decrease insulin sensitivity and worsen glycemic control in septic animal models and human studies (24–26). Thus, the factors contributing to hyperglycemia in sepsis are complex.
Once present, hyperglycemia may further impair the host immune response and potentially increase the risk for further organ injury (27, 28). High glucose concentrations promote proinflammatory cytokine release and adversely affect several types of innate immune cells (29, 30). For example, hyperglycemia decreases granule release in neutrophils, reduces neutrophil extracellular trap formation, and prevents effective chemotaxis resulting in reduced capacity to combat invading pathogens (31). Additionally, blood glucose can complex with bacterial components and complement proteins, thereby preventing effective opsonization and phagocytosis (32, 33). Thus, the development of hyperglycemia has direct pathologic implications in sepsis.
Dysglycemia in sepsis does not comprise only hyperglycemia. Hypoglycemia is an independent predictor of increased in-hospital and 1-year mortality in critically ill patients with sepsis and often occurs secondary to exogenous insulin administration (34–36). Acute hypoglycemia in healthy individuals (both with and without diabetes mellitus) has been shown to increase oxidative stress, platelet aggregation, proinflammatory cytokine production, and vascular adhesion molecule expression (37–39). Additionally, glucose variability is of prognostic importance because increased glycemic lability in patients with sepsis is associated with higher in-hospital mortality (40). Maintenance of euglycemia remains the goal in hospitalized patients with sepsis but has proved difficult to achieve.
Clinical trials of glycemic control in patients with sepsis
The adverse impact of dysglycemia prompted several large, randomized, controlled clinical studies in critically ill patients. A landmark study published by van den Berghe et al. (41) in 2001 demonstrated a substantial mortality benefit with the use of exogenous intravenous intensive insulin therapy to maintain tight glycemic control (blood glucose maintained between 80 and 110 mg/dL) in a mixed population of mostly surgical critically ill patients. Subsequent studies of tight glycemic control, however, yielded variable and potentially contradictory results (42–45). A meta-analysis of 12 randomized controlled trials specifically in patients with sepsis determined that, compared with conventional glucose management, intensive insulin therapy did not reduce mortality or length of intensive care unit (ICU) stay in patients with sepsis but did increase incidence of hypoglycemia (46). Thus, an ideal strategy of glycemic control in sepsis that prevents hyperglycemia while minimizing the risk of hypoglycemia remains elusive (and has not been accomplished with approaches that rely on exogenous insulin alone) but can potentially be achieved through modulation of the incretin axis.
The Incretin Hormone Axis
Physiology of incretin hormones
Incretin hormones are intestine-derived peptides released primarily in response to enteral nutrients and promote insulin secretion from pancreatic β cells in a glucose-dependent manner. Glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide-1 (GLP-1) are the best characterized incretin hormones to date. GIP is released in response to enteral administration of nutrients (including carbohydrates and fatty acids) by enteroendocrine cells (K cells) located mostly in the proximal small intestine (47, 48). GIP is rapidly degraded by the protease dipeptidyl-peptidase 4 (DPP-4, also known as CD26) with a half-life of 7 to 8 minutes (49). The receptor for GIP is a seven-transmembrane G-protein coupled receptor that increases cAMP and intracellular calcium in response to ligand binding. In pancreatic β cells, GIP receptor signaling induces insulin exocytosis (47, 50, 51). Additionally, GIP promotes glucagon secretion under conditions of hypoglycemia (52). Thus, under physiologic conditions, GIP is important in the maintenance of euglycemia.
Although the incretin hormones GIP and GLP-1 are similar in their ability to promote insulin secretion in a glucose-dependent manner, there are some notable differences. GLP-1 is derived from the preproglucagon peptide encoded by the GCG gene (which also encodes glucagon) and is released from enteroendocrine cells (L cells) located predominantly in the distal small bowel and colon (48, 53). GLP-1 is similarly degraded rapidly by DPP-4, but with a shorter half-life compared with GIP of only 2 to 5 minutes (54, 55). Whereas GIP release is stimulated by direct interaction of enteroendocrine cells with enteral nutrients, release of GLP-1 following a meal is more complex. An initial peak of GLP-1 (approximately 10 to 15 minutes postprandial) is likely promoted by gastric distention or vagal stimulation, and a later peak (approximately 30 to 60 minutes postprandial) is likely promoted by direct interaction with enteral nutrients (47, 56). GLP-1 promotes glucose-dependent insulin secretion from pancreatic β cells through binding of a G-protein–coupled receptor that results in increased insulin gene expression, improved mRNA stability, and increased insulin exocytosis. Furthermore, GLP-1 suppresses secretion of glucagon from pancreatic α cells under conditions of euglycemia and hyperglycemia (47, 57, 58). Extrapancreatic effects demonstrated by GLP-1 agonists include the promotion of satiety and the promotion of weight loss (59–61). Uncovering roles of increased incretin activity in extrapancreatic tissues is an area of ongoing research.
Incretin release in response to inflammatory stimuli
Although GIP and GLP-1 are primarily secreted in response to enteral nutrients, recent studies have suggested that incretins are also released in response to inflammatory stimuli. Lebrun et al. (62) demonstrated that GLP-1 is released from L cells in response to the intestinal insults dextran sodium sulfate and ischemia reperfusion. Similarly, Ellingsgaard et al. (16) demonstrated that IL-6 stimulates GLP-1 production in immortalized enteroendocrine cells in a dose-dependent manner mediated by increases in Pcsk1 (encoding the prohormone convertase 1/3, which processes preproglucagon to GLP-1) and Gcg gene expression. Systemic IL-6 administration to mice in the Ellingsgaard study increased circulating GLP-1 (16). Subsequent studies in mice confirmed the ability of not only IL-6 but also of IL-1β and of the bacterial cell wall component lipopolysaccharide (LPS) to increase GLP-1 levels (17, 63). LPS has also been shown to increase GIP through an IL-1β–dependent mechanism (64). GLP-1 promotes local mucosal healing; thus, release of incretins following an insult may serve to limit intestinal injury under conditions of systemic inflammation (65).
Observational cohort studies have similarly demonstrated associations between circulating GLP-1 levels and systemic inflammation (17, 66, 67). In a cohort study examining critically ill patients (of whom 66% presented with sepsis), GLP-1 levels were positively correlated with inflammatory markers, including c-reactive protein and procalcitonin, as well as markers of organ dysfunction, including cystatin C and coagulation factors. Interestingly, increased endogenous GLP-1 levels in this study predicted ICU mortality (67). However, the elevated GLP-1 levels should not be interpreted to indicate that incretins are harmful in sepsis; rather, the findings suggest that GLP-1 may have a role in countering the adverse effects of systemic inflammation. The anti-inflammatory mediators IL-1 receptor antagonist and the soluble receptor for TNF-α are similarly released in response to overwhelming inflammation and are also positively correlated with organ failure and mortality in septic populations despite not being innately harmful (68, 69). Furthermore, findings from preclinical and clinical studies of incretin-based therapies in sepsis demonstrate benefit as opposed to harm.
Therapeutic Effects of Incretin-Based Medications
Current incretin-based medications
Two main categories of incretin-based therapies are currently in use for the management of diabetes mellitus: incretin mimetics and DPP-4 inhibitors. Incretin mimetics, including exenatide, liraglutide, and semaglutide, are structurally similar to GLP-1 but have been modified to resist degradation. DPP-4 inhibitors, including sitagliptin and linagliptin, increase incretin levels by blocking protease degradation by DPP-4, thus increasing endogenous levels of GIP and GLP-1 (59, 70, 71). Incretin mimetics and DPP-4 inhibitors improve glycemic control in patients with diabetes mellitus type 2, and recent clinical trials have highlighted a cardioprotective effect with incretin mimetic therapy (72–74).
Therapeutic effects of incretin mimetics on activated immune cells
Incretin mimetics demonstrate anti-inflammatory effects in preclinical studies, and several studies report the detection of incretin receptors in different immune cells, including macrophages, monocytes, and B- and T-cell lymphocytes (75–77). Yusta et al. (78) demonstrated that exendin-4, a GLP-1 receptor agonist, reduced proinflammatory cytokine expression in activated intestinal intraepithelial lymphocytes. In the same study, exendin-4 attenuated injury in a dextran sodium sulfate model of colitis, whereas GLP-1R knockout mice sustained more severe intestinal injury compared with wild-type controls. Restoration of GLP-1 signaling in lymphocytes in GLP-1R knockout chimera mice rescued intestinal integrity. In studies of cultured macrophages, treatment with exendin-4 attenuated the inflammatory response to LPS through a protein kinase A–cAMP-dependent reduction in nuclear translocation of NF-κB and consequent decrease in IL-6, IL-1β, and TNF-α production (79, 80). Exendin-4 also increases macrophage production of IL-10 and arginase (79), suggesting GLP-1R stimulation can influence macrophage polarization shifting from the M1 classically activated proinflammatory to the M2 anti-inflammatory phenotype (81). Thus, incretin mimetic therapies demonstrate the ability to directly modify the activity of immune cells.
Therapeutic effects of incretin-based therapies in nonimmune cells and tissues
Anti-inflammatory effects of incretin-based therapies have been demonstrated in several nonimmune cell and tissue types (Fig. 1). For example, in human aortic or vascular endothelial cells, treatment with liraglutide decreases expression of vascular adhesion molecules and reduces adhesion of monocytes induced by LPS (82) or TNF-α (83). Additional studies in human vascular endothelial cells exposed to LPS confirm that incretin mimetics decrease vascular permeability and reduce proinflammatory cytokine levels (84, 85). In cardiomyoblasts, treatment with liraglutide in a two-hit TNF-α and hypoxia model decreases NLRP3 inflammasome activation and is associated with improvements in cell viability (86). Incretin infusions with either GIP or GLP-1 in mouse models significantly reduce vascular inflammation and decrease macrophage infiltration into the endothelium, consistent with findings that chronic use of incretin-based therapies attenuate atherosclerotic plaque development (87–91).
Figure 1.
Extra-pancreatic effects of incretin hormones released in response to enteral nutrients or proinflammatory stimuli, endogenous incretin hormones, and incretin-based therapies demonstrate anti-inflammatory and protective effects across multiple organ systems.
Anti-inflammatory effects of incretin-based therapies are also evident in adipose tissues (92, 93), pancreatic islets (94–96), hepatocytes (97, 98), and renal glomerular cells (99–101) in preclinical studies. Some studies suggest GIP may worsen inflammation from adipose tissues in obese individuals (102, 103). However, recent preclinical studies with novel GIP analogs suggest an anti-inflammatory effect in adipose tissue (104), comparable to studies with DPP-4 inhibitors that raise levels of both GIP and GLP-1 (94, 105). Interestingly, GIP and GLP-1 analogs are being explored as therapeutic agents to reduce neuroinflammation in murine models of Alzheimer’s disease and Parkinson disease, with findings of decreased cytokine production, inhibition of microglial cell activation, improved neuronal insulin sensitivity, and improved cognitive performance (106–109).
Effects of incretin-based therapies on systemic inflammation
The anti-inflammatory effects of incretin-based therapies have been confirmed in clinical studies. An acute 3-hour infusion of exogenous GLP-1 reduced circulating IL-6 levels in a small study of obese individuals with diabetes mellitus type 2; a trend toward reduced inflammation with GIP infusion was also observed (110). Chronic incretin mimetic use in patients with diabetes decreases circulating proinflammatory cytokines, including IL-6, TNF-α, IL-1β, and monocyte chemoattractant protein-1 after 8 to 12 weeks on therapy, with persistent anti-inflammatory effects reported 6 to 12 months after initiating treatment (111–113). Chronic DPP-4 use similarly decreases proinflammatory cytokines and vascular adhesion modules and increases anti-inflammatory mediators, including IL-10 (114, 115). Given that sepsis is characterized by overwhelming systemic inflammation and multiorgan dysfunction, the consistent preclinical and clinical anti-inflammatory effects of incretins support further investigation of the incretin hormone axis in sepsis.
Incretin-based therapies in preclinical models of sepsis
Several studies have examined the use of incretin-based therapies in mouse models of sepsis. In an endotoxemic rat study by Yanay et al. (116), the GLP-1 analog exendin-4 decreased circulating IL-6, TNF-α, IL-1β, and interferon-γ levels and prevented LPS-induced hypoglycemia. A subsequent study by Steven et al. (117) demonstrated that both the incretin mimetic liraglutide and the DPP-4 inhibitor linagliptin decreased mortality, improved hypotension, suppressed inflammation, and reduced oxidative stress in a severe endotoxemic rat model. In a separate study in endotoxemic mice, linagliptin and liraglutide reduced oxidative stress, suppressed inflammatory mediator production, and prevented thrombocytopenia and microvascular thrombosis in the pulmonary vasculature (118). Protection against LPS-induced thrombocytopenia was lost in mice deficient for the GLP-1 receptor, highlighting the importance of intact incretin signaling. Interestingly, infusion of exogenous GLP-1 after intravenous LPS exposure reduces permeability in mesenteric vasculature, highlighting additional protective effects against intestinal injury in settings of systemic inflammation (119). Importantly, incretin-based therapies demonstrate favorable effects not only in sterile models of inflammation but also in preclinical bacteremic models.
Lee et al. (84) demonstrated benefit with incretin mimetics in a cecal-ligation and puncture model of sepsis, whereby high-dose exendin-4 administered twice daily starting 16 hours after septic insult improved survival, associated with decreases in the proinflammatory protein high mobility group box protein-1. In a separate study, a long-lasting polyethylene-glycol–conjugated exendin-4 molecule reduced lung injury; lowered IL-6, IL-1β, TNF-α, and monocyte chemoattractant protein-1 levels; decreased peritoneal leukocyte counts; and reduced mortality after cecal-ligation and puncture (85). Thus, exogenous incretin-based therapies consistently improve outcomes in preclinical septic models, and we hypothesize that physiologic activation of the incretin hormone pathway may induce similar beneficial effects.
Recently, our laboratory demonstrated that infusion of low-level enteral dextrose in a murine endotoxemia model increased glucose disposal, insulin secretion, and insulin sensitivity; improved mean arterial blood pressure; and decreased systemic inflammation in a GIP-dependent fashion (120). When GIP signaling was inhibited with pharmacologic blockade, the beneficial effects of enteral dextrose were lost, and infusion of exogenous GIP improved glucose disposal and reduced IL-6 levels. Our study demonstrated that activation of endogenous incretin hormones in the acute phase of sepsis may be achieved through provision of enteral nutrients and that therapeutic benefits of incretins may be realized in the absence of exogenous administration (120). Taken together, the improved outcomes with endogenous incretin stimulation and with exogenous incretin-based therapy support the notion that increased incretin activity is of potential clinical benefit to patients presenting with sepsis.
Clinical Studies of Incretin-Based Therapies in Critically Ill Patients
Incretin-based therapies have not been tested in clinical trials of sepsis. However, several small clinical trials have been conducted testing the effect of exogenous incretins in the broader context of critical illness (121, 122). In cardiac and in mixed medical and surgical ICU populations, exogenous GLP-1 is effective in reducing blood glucose and glycemic variability compared with placebo without an increased risk of hypoglycemia (123–129). Infusion of the GLP-1 analog exenatide may similarly improve glycemic control but has been associated with gastrointestinal side effects (130). Despite being well tolerated, infusions of exogenous GIP have not demonstrated benefit in reducing blood sugar in critically ill patients, but in these studies the effects of GIP were tested in the fasted state, where glucose-stimulated insulin secretion may be blunted, and in response to a liquid meal bolus, which stimulates endogenous incretin hormone production and may limit the host’s ability to respond to additional exogenous GIP (131, 132). Existing clinical trials were performed mostly in mixed ICU populations with few patients with sepsis enrolled (Table 1). Further clinical trials are needed to better characterize the incretin hormone axis during sepsis and to determine a potential therapeutic role. A prospective single-center, double-blind, placebo-controlled randomized clinical trial from our laboratory is currently recruiting patients to define the effects of a low-level enteral dextrose infusion on inflammation and on endogenous incretin hormone production in critically ill patients with sepsis (ClinicalTrials.gov no. NCT03454087).
Table 1.
Clinical Trials of Exogenous Incretion Therapies in Critically Ill Populations
| Study | Year | Design | Population | Number of Participants | Participants With Sepsis, n (%) | Treatment | Primary Results | Adverse Effects |
|---|---|---|---|---|---|---|---|---|
| Meier et al. (133) | 2004 | Double-blind, placebo-controlled, randomized crossover study | Postsurgical patients with diabetes | 8 | Not reported | GLP-1 infusion | Reduced blood glucose, increased endogenous insulin secretion | None |
| Sokos et al. (124) | 2007 | Double-blind, placebo-controlled, randomized clinical trial | Cardiac patients undergoing coronary artery bypass grafting | 20 | Not reported | GLP-1 infusion | Reduced blood glucose, insulin requirements, and cardiac arrhythmias | Hypoglycemia in 1 patient in the GLP-1 group and 2 patients in the placebo group |
| Deane et al. (125) | 2009 | Double-blind, placebo-controlled, randomized crossover study | Mixed medical-surgical patients without diabetes | 7 | Not reported | GLP-1 infusion | Reduced blood glucose | None |
| Deane et al. (126) | 2010 | Double-blind, placebo-controlled, randomized crossover study | Mixed medical-surgical patients without diabetes | 25 | 4 (16%) | GLP-1 infusion | Reduced blood glucose and small intestine glucose uptake | Hyperglycemia in 1 patient in the GLP-1 group and 2 in the placebo group |
| Deane et al. (127) | 2011 | Double-blind, placebo-controlled, randomized crossover study | Mixed medical-surgical patients with diabetes | 11 | 5 (45%) | GLP-1 infusion | Reduced in blood glucose | None |
| Abuannadi et al. (130) | 2012 | Single-center feasibility study; all patients received intervention | Cardiac patients in the ICU | 40 | Not reported | Exenatide infusion | Blood glucose control similar to historical controls receiving insulin infusion protocols targeting blood glucose 100–140 mg/dL | Hyperglycemia in 3 patients, hypoglycemia in 4, nausea in 16, emesis in two |
| Lee et al. (132) | 2013 | Double-blind, placebo-controlled, randomized crossover study | Mixed medical-surgical patients without diabetes | 20 | 9 (45%) | GIP infusion in patients already receiving GLP-1 infusion | No additional effect on blood glucose | None |
| Galiatsatos et al. (128) | 2014 | Double-blind, placebo-controlled, randomized clinical trial | Surgical and burn patients in the ICU | 18 | Not reported | GLP-1 infusion | Reduced blood glucose and glycemic variability | Hypoglycemia in 1 patient in the GLP-1 group and 3 in the placebo group |
| Kar et al. (131) | 2015 | Double-blind, placebo-controlled, randomized crossover study | Mixed medical-surgical patients without diabetes | 20 | 2 (10%) | GIP infusion | No effect on blood glucose or gastric emptying | None |
| Miller et al. (129) | 2017 | Double-blind, placebo-controlled, randomized crossover study | Mixed medical-surgical patients without diabetes | 12 | 1 (8%) | GLP-1 infusion | Reduced blood glucose and small intestine glucose uptake | None |
Conclusion
Incretin-based therapies have revolutionized the chronic management of patients with diabetes by providing a means to improve glycemic control, mitigate the risk of hypoglycemia, increase weight loss, improve insulin sensitivity, and reduce the risk of cardiovascular disease. Therapies targeting increased incretin activity may provide similar benefit in the care of critically ill patients with sepsis by promoting the maintenance of euglycemia and modulating the host inflammatory response. Preclinical studies of incretin-based therapies demonstrate anti-inflammatory effects in various tissues, including vascular endothelium, pancreatic β cells, and hepatocytes, and improved mortality in sterile inflammation and infectious animal models of sepsis. Initial clinical studies of exogenous incretins in acutely ill patients demonstrate that GIP and GLP-1 infusions are well tolerated in mixed critically ill populations and may improve glycemic control without the risk of hypoglycemia. Additional clinical studies are needed to explore the role of incretin-based therapies as a novel target to optimize glycemic control, reduce systemic inflammation, and improve outcomes in patients with sepsis.
Acknowledgments
We acknowledge the Health Science Library System at the University of Pittsburgh for their assistance in optimizing the PubMed search for this mini-review. We thank Kathryn Nauman, Vascular Medicine Institute at the University of Pittsburgh, for her assistance with Figure 1 in this manuscript.
Financial Support: This work was supported by the National Institutes of Health Grants 5K23GM122069 (to F.A.S.), 1R01DK114012 and 1R01DK119627 (to M.J.J.), and 5P01HL114453 (to B.J.M.).
Clinical Trial Information: ClinicalTrials.gov no. NCT03454087 (registered 5 March 2018).
Author Contributions: F.A.S., C.P.O., and B.J.M. contributed to the conception and design of the mini-review. F.A.S. performed the literature review and drafted the manuscript. All authors critically revised the manuscript, agreed to be fully accountable for ensuring the integrity and accuracy of the report, and read and approved the final manuscript.
Additional Information
Disclosure Summary: The authors have nothing to disclose.
Data Availability: Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Glossary
Abbreviations:
- DPP-4
dipeptidyl-peptidase 4
- GIP
glucose-dependent insulinotropic peptide
- GLP-1
glucagon-like peptide-1
- ICU
intensive care unit
- LPS
lipopolysaccharide
References and Notes
- 1. Seymour CW, Liu VX, Iwashyna TJ, Brunkhorst FM, Rea TD, Scherag A, Rubenfeld G, Kahn JM, Shankar-Hari M, Singer M, Deutschman CS, Escobar GJ, Angus DC. Assessment of clinical criteria for sepsis: for the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):762–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, Angus DC, Rubenfeld GD, Singer M; Sepsis Definitions Task Force. Developing a new definition and assessing new clinical criteria for septic shock: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):775–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Wiersinga WJ, Leopold SJ, Cranendonk DR, van der Poll T. Host innate immune responses to sepsis. Virulence. 2014;5(1):36–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348(2):138–150. [DOI] [PubMed] [Google Scholar]
- 5. Martí-Carvajal AJ, Solà I, Lathyris D, Cardona AF. Human recombinant activated protein C for severe sepsis. Cochrane Database Syst Rev. 2012; (3):CD004388. [DOI] [PubMed] [Google Scholar]
- 6. Rice TW, Wheeler AP, Bernard GR, Vincent JL, Angus DC, Aikawa N, Demeyer I, Sainati S, Amlot N, Cao C, Ii M, Matsuda H, Mouri K, Cohen J. A randomized, double-blind, placebo-controlled trial of TAK-242 for the treatment of severe sepsis. Crit Care Med. 2010;38(8):1685–1694. [DOI] [PubMed] [Google Scholar]
- 7. Liu VX, Fielding-Singh V, Greene JD, Baker JM, Iwashyna TJ, Bhattacharya J, Escobar GJ. The timing of early antibiotics and hospital mortality in sepsis. Am J Respir Crit Care Med. 2017;196(7):856–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Seymour CW, Gesten F, Prescott HC, Friedrich ME, Iwashyna TJ, Phillips GS, Lemeshow S, Osborn T, Terry KM, Levy MM. Time to treatment and mortality during mandated emergency care for sepsis. N Engl J Med. 2017;376(23):2235–2244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Krinsley JS. Association between hyperglycemia and increased hospital mortality in a heterogeneous population of critically ill patients. Mayo Clin Proc. 2003;78(12):1471–1478. [DOI] [PubMed] [Google Scholar]
- 10. van Vught LA, Wiewel MA, Klein Klouwenberg PM, Hoogendijk AJ, Scicluna BP, Ong DS, Cremer OL, Horn J, Bonten MM, Schultz MJ, van der Poll T, Molecular D; Molecular Diagnosis and Risk Stratification of Sepsis Consortium. Admission hyperglycemia in critically ill sepsis patients: association with outcome and host response. Crit Care Med. 2016;44(7):1338–1346. [DOI] [PubMed] [Google Scholar]
- 11. Preechasuk L, Suwansaksri N, Ipichart N, Vannasaeng S, Permpikul C, Sriwijitkamol A. Hyperglycemia and glycemic variability are associated with the severity of sepsis in nondiabetic subjects. J Crit Care. 2017;38:319–323. [DOI] [PubMed] [Google Scholar]
- 12. Marik PE, Raghavan M. Stress-hyperglycemia, insulin and immunomodulation in sepsis. Intensive Care Med. 2004;30(5):748–756. [DOI] [PubMed] [Google Scholar]
- 13. Borst SE. The role of TNF-alpha in insulin resistance. Endocrine. 2004;23(2-3):177–182. [DOI] [PubMed] [Google Scholar]
- 14. Jager J, Grémeaux T, Cormont M, Le Marchand-Brustel Y, Tanti JF. Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression. Endocrinology. 2007;148(1):241–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Fan J, Li YH, Wojnar MM, Lang CH. Endotoxin-induced alterations in insulin-stimulated phosphorylation of insulin receptor, IRS-1, and MAP kinase in skeletal muscle. Shock. 1996;6(3):164–170. [PubMed] [Google Scholar]
- 16. Ellingsgaard H, Hauselmann I, Schuler B, Habib AM, Baggio LL, Meier DT, Eppler E, Bouzakri K, Wueest S, Muller YD, Hansen AM, Reinecke M, Konrad D, Gassmann M, Reimann F, Halban PA, Gromada J, Drucker DJ, Gribble FM, Ehses JA, Donath MY. Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nat Med. 2011;17(11):1481–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Kahles F, Meyer C, Möllmann J, Diebold S, Findeisen HM, Lebherz C, Trautwein C, Koch A, Tacke F, Marx N, Lehrke M. GLP-1 secretion is increased by inflammatory stimuli in an IL-6-dependent manner, leading to hyperinsulinemia and blood glucose lowering. Diabetes. 2014;63(10):3221–3229. [DOI] [PubMed] [Google Scholar]
- 18. Park C, Kim JR, Shim JK, Kang BS, Park YG, Nam KS, Lee YC, Kim CH. Inhibitory effects of streptozotocin, tumor necrosis factor-alpha, and interleukin-1beta on glucokinase activity in pancreatic islets and gene expression of GLUT2 and glucokinase. Arch Biochem Biophys. 1999;362(2):217–224. [DOI] [PubMed] [Google Scholar]
- 19. Woodske ME, Yokoe T, Zou B, Romano LC, Rosa TC, Garcia-Ocana A, Alonso LC, O’Donnell CP, McVerry BJ. Hyperinsulinemia predicts survival in a hyperglycemic mouse model of critical illness. Crit Care Med. 2009;37(9):2596–2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Ehses JA, Böni-Schnetzler M, Faulenbach M, Donath MY. Macrophages, cytokines and beta-cell death in Type 2 diabetes. Biochem Soc Trans. 2008;36(3):340–342. [DOI] [PubMed] [Google Scholar]
- 21. Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-Jemelka HI, Spinas GA, Kaiser N, Halban PA, Donath MY. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest. 2002;110(6):851–860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Mandrup-Poulsen T, Bendtzen K, Nerup J, Dinarello CA, Svenson M, Nielsen JH. Affinity-purified human interleukin I is cytotoxic to isolated islets of Langerhans. Diabetologia. 1986;29(1):63–67. [DOI] [PubMed] [Google Scholar]
- 23. Sapolsky RM, Romero LM, Munck AU. How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions. Endocr Rev. 2000;21(1):55–89. [DOI] [PubMed] [Google Scholar]
- 24. Gelfand RA, Matthews DE, Bier DM, Sherwin RS. Role of counterregulatory hormones in the catabolic response to stress. J Clin Invest. 1984;74(6):2238–2248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Hunt DG, Ivy JL. Epinephrine inhibits insulin-stimulated muscle glucose transport. J Appl Physiol. 2002;93(5):1638–1643. [DOI] [PubMed] [Google Scholar]
- 26. Keh D, Trips E, Marx G, Wirtz SP, Abduljawwad E, Bercker S, Bogatsch H, Briegel J, Engel C, Gerlach H, Goldmann A, Kuhn SO, Hüter L, Meier-Hellmann A, Nierhaus A, Kluge S, Lehmke J, Loeffler M, Oppert M, Resener K, Schädler D, Schuerholz T, Simon P, Weiler N, Weyland A, Reinhart K, Brunkhorst FM; SepNet–Critical Care Trials Group. Effect of hydrocortisone on development of shock among patients with severe sepsis: the HYPRESS randomized clinical trial. JAMA. 2016;316(17):1775–1785. [DOI] [PubMed] [Google Scholar]
- 27. Yu WK, Li WQ, Li N, Li JS. Influence of acute hyperglycemia in human sepsis on inflammatory cytokine and counterregulatory hormone concentrations. World J Gastroenterol. 2003;9(8):1824–1827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Leonidou L, Mouzaki A, Michalaki M, DeLastic AL, Kyriazopoulou V, Bassaris HP, Gogos CA. Cytokine production and hospital mortality in patients with sepsis-induced stress hyperglycemia. J Infect. 2007;55(4):340–346. [DOI] [PubMed] [Google Scholar]
- 29. Shanmugam N, Reddy MA, Guha M, Natarajan R. High glucose-induced expression of proinflammatory cytokine and chemokine genes in monocytic cells. Diabetes. 2003;52(5):1256–1264. [DOI] [PubMed] [Google Scholar]
- 30. Kanter JE, Kramer F, Barnhart S, Averill MM, Vivekanandan-Giri A, Vickery T, Li LO, Becker L, Yuan W, Chait A, Braun KR, Potter-Perigo S, Sanda S, Wight TN, Pennathur S, Serhan CN, Heinecke JW, Coleman RA, Bornfeldt KE. Diabetes promotes an inflammatory macrophage phenotype and atherosclerosis through acyl-CoA synthetase 1. Proc Natl Acad Sci USA. 2012;109(12):E715–E724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Jafar N, Edriss H, Nugent K. The effect of short-term hyperglycemia on the innate immune system. Am J Med Sci. 2016;351(2):201–211. [DOI] [PubMed] [Google Scholar]
- 32. van Oss CJ, Border JR. Influence of intermittent hyperglycemic glucose levels on the phagocytosis of microorganisms by human granulocytes in vitro. Immunol Commun. 1978;7(6):669–676. [DOI] [PubMed] [Google Scholar]
- 33. Lin JC, Siu LK, Fung CP, Tsou HH, Wang JJ, Chen CT, Wang SC, Chang FY. Impaired phagocytosis of capsular serotypes K1 or K2 Klebsiella pneumoniae in type 2 diabetes mellitus patients with poor glycemic control. J Clin Endocrinol Metab. 2006;91(8):3084–3087. [DOI] [PubMed] [Google Scholar]
- 34. Park S, Kim DG, Suh GY, Kang JG, Ju YS, Lee YJ, Park JY, Lee SW, Jung KS. Mild hypoglycemia is independently associated with increased risk of mortality in patients with sepsis: a 3-year retrospective observational study. Crit Care. 2012;16(5):R189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Waeschle RM, Moerer O, Hilgers R, Herrmann P, Neumann P, Quintel M. The impact of the severity of sepsis on the risk of hypoglycaemia and glycaemic variability. Crit Care. 2008;12(5):R129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Krinsley JS, Grover A. Severe hypoglycemia in critically ill patients: risk factors and outcomes. Crit Care Med. 2007;35(10):2262–2267. [DOI] [PubMed] [Google Scholar]
- 37. Wright RJ, Newby DE, Stirling D, Ludlam CA, Macdonald IA, Frier BM. Effects of acute insulin-induced hypoglycemia on indices of inflammation: putative mechanism for aggravating vascular disease in diabetes. Diabetes Care. 2010;33(7):1591–1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Gogitidze Joy N, Hedrington MS, Briscoe VJ, Tate DB, Ertl AC, Davis SN. Effects of acute hypoglycemia on inflammatory and pro-atherothrombotic biomarkers in individuals with type 1 diabetes and healthy individuals [published correction appears in Diabetes Care. 2010;33(9):2129]. Diabetes Care. 2010;33(7):1529–1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Wang J, Alexanian A, Ying R, Kizhakekuttu TJ, Dharmashankar K, Vasquez-Vivar J, Gutterman DD, Widlansky ME. Acute exposure to low glucose rapidly induces endothelial dysfunction and mitochondrial oxidative stress: role for AMP kinase. Arterioscler Thromb Vasc Biol. 2012;32(3):712–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Ali NA, O’Brien JM Jr, Dungan K, Phillips G, Marsh CB, Lemeshow S, Connors AF Jr, Preiser JC. Glucose variability and mortality in patients with sepsis. Crit Care Med. 2008;36(8):2316–2321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. 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 critically ill patients. N Engl J Med. 2001;345(19):1359–1367. [DOI] [PubMed] [Google Scholar]
- 42. Arabi YM, Dabbagh OC, Tamim HM, Al-Shimemeri AA, Memish ZA, Haddad SH, Syed SJ, Giridhar HR, Rishu AH, Al-Daker MO, Kahoul SH, Britts RJ, Sakkijha MH. Intensive versus conventional insulin therapy: a randomized controlled trial in medical and surgical critically ill patients. Crit Care Med. 2008;36(12):3190–3197. [DOI] [PubMed] [Google Scholar]
- 43. Finfer S, Heritier S, Committee NSM, Committee SSE; NICE Study Management Committee and SUGAR Study Executive Committee. The NICE-SUGAR (Normoglycaemia in Intensive Care Evaluation and Survival Using Glucose Algorithm Regulation) Study: statistical analysis plan. Crit Care Resusc. 2009;11(1):46–57. [PubMed] [Google Scholar]
- 44. Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U, John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M, Hartog C, Natanson C, Loeffler M, Reinhart K; German Competence Network Sepsis (SepNet). Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med. 2008;358(2):125–139. [DOI] [PubMed] [Google Scholar]
- 45. Preiser JC, Devos P, Ruiz-Santana S, Mélot C, Annane D, Groeneveld J, Iapichino G, Leverve X, Nitenberg G, Singer P, Wernerman J, Joannidis M, Stecher A, Chioléro R. A prospective randomised multi-centre controlled trial on tight glucose control by intensive insulin therapy in adult intensive care units: the Glucontrol study. Intensive Care Med. 2009;35(10):1738–1748. [DOI] [PubMed] [Google Scholar]
- 46. Song F, Zhong LJ, Han L, Xie GH, Xiao C, Zhao B, Hu YQ, Wang SY, Qin CJ, Zhang Y, Lai DM, Cui P, Fang XM. Intensive insulin therapy for septic patients: a meta-analysis of randomized controlled trials. BioMed Res Int. 2014;2014:698265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132(6):2131–2157. [DOI] [PubMed] [Google Scholar]
- 48. Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013;17(6):819–837. [DOI] [PubMed] [Google Scholar]
- 49. Kieffer TJ, McIntosh CH, Pederson RA. Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology. 1995;136(8):3585–3596. [DOI] [PubMed] [Google Scholar]
- 50. Ding WG, Gromada J. Protein kinase A-dependent stimulation of exocytosis in mouse pancreatic beta-cells by glucose-dependent insulinotropic polypeptide. Diabetes. 1997;46(4):615–621. [DOI] [PubMed] [Google Scholar]
- 51. McIntosh CH, Widenmaier S, Kim SJ. Glucose-dependent insulinotropic polypeptide signaling in pancreatic β-cells and adipocytes. J Diabetes Investig. 2012;3(2):96–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Christensen M, Vedtofte L, Holst JJ, Vilsbøll T, Knop FK. Glucose-dependent insulinotropic polypeptide: a bifunctional glucose-dependent regulator of glucagon and insulin secretion in humans. Diabetes. 2011;60(12):3103–3109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368(9548):1696–1705. [DOI] [PubMed] [Google Scholar]
- 54. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes. 1995;44(9):1126–1131. [DOI] [PubMed] [Google Scholar]
- 55. Mentlein R, Gallwitz B, Schmidt WE. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7-36)amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem. 1993;214(3):829–835. [DOI] [PubMed] [Google Scholar]
- 56. Herrmann C, Göke R, Richter G, Fehmann HC, Arnold R, Göke B. Glucagon-like peptide-1 and glucose-dependent insulin-releasing polypeptide plasma levels in response to nutrients. Digestion. 1995;56(2):117–126. [DOI] [PubMed] [Google Scholar]
- 57. Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 2018;27(4):740–756. [DOI] [PubMed] [Google Scholar]
- 58. MacDonald PE, El-Kholy W, Riedel MJ, Salapatek AM, Light PE, Wheeler MB. The multiple actions of GLP-1 on the process of glucose-stimulated insulin secretion. Diabetes. 2002;51(Suppl 3):S434–S442. [DOI] [PubMed] [Google Scholar]
- 59. Nauck M. Incretin therapies: highlighting common features and differences in the modes of action of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Diabetes Obes Metab. 2016;18(3):203–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Pratley R, Nauck M, Bailey T, Montanya E, Cuddihy R, Filetti S, Garber A, Thomsen AB, Hartvig H, Davies M; 1860-LIRA-DPP-4 Study Group. One year of liraglutide treatment offers sustained and more effective glycaemic control and weight reduction compared with sitagliptin, both in combination with metformin, in patients with type 2 diabetes: a randomised, parallel-group, open-label trial. Int J Clin Pract. 2011;65(4):397–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Turton MD, O’Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, Choi SJ, Taylor GM, Heath MM, Lambert PD, Wilding JP, Smith DM, Ghatei MA, Herbert J, Bloom SR. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379(6560):69–72. [DOI] [PubMed] [Google Scholar]
- 62. Lebrun LJ, Lenaerts K, Kiers D, Pais de Barros JP, Le Guern N, Plesnik J, Thomas C, Bourgeois T, Dejong CHC, Kox M, Hundscheid IHR, Khan NA, Mandard S, Deckert V, Pickkers P, Drucker DJ, Lagrost L, Grober J, Enteroendocrine L. Enteroendocrine L cells sense LPS after gut barrier injury to enhance GLP-1 secretion. Cell Reports. 2017;21(5):1160–1168. [DOI] [PubMed] [Google Scholar]
- 63. Nguyen AT, Mandard S, Dray C, Deckert V, Valet P, Besnard P, Drucker DJ, Lagrost L, Grober J. Lipopolysaccharides-mediated increase in glucose-stimulated insulin secretion: involvement of the GLP-1 pathway. Diabetes. 2014;63(2):471–482. [DOI] [PubMed] [Google Scholar]
- 64. Kahles F, Meyer C, Diebold S, Foldenauer AC, Stöhr R, Möllmann J, Lebherz C, Findeisen HM, Marx N, Lehrke M. Glucose-dependent insulinotropic peptide secretion is induced by inflammatory stimuli in an interleukin-1-dependent manner in mice. Diabetes Obes Metab. 2016;18(11):1147–1151. [DOI] [PubMed] [Google Scholar]
- 65. Hytting-Andreasen R, Balk-Møller E, Hartmann B, Pedersen J, Windeløv JA, Holst JJ, Kissow H. Endogenous glucagon-like peptide- 1 and 2 are essential for regeneration after acute intestinal injury in mice. PLoS One. 2018;13(6):e0198046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Lebherz C, Kahles F, Piotrowski K, Vogeser M, Foldenauer AC, Nassau K, Kilger E, Marx N, Parhofer KG, Lehrke M. Interleukin-6 predicts inflammation-induced increase of Glucagon-like peptide-1 in humans in response to cardiac surgery with association to parameters of glucose metabolism. Cardiovasc Diabetol. 2016;15(1):21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Lebherz C, Schlieper G, Mollmann J, Kahles F, Schwarz M, Brunsing J, Dimkovic N, Koch A, Trautwein C, Floge J, Marx N, Tacke F, Lehrke M.. GLP-1 levels predict mortality in patients with critical illness as well as end-stage renal disease.Am J Med. 2017;130(7):833–841 e833. [DOI] [PubMed] [Google Scholar]
- 68. Endo S, Inada K, Yamada Y, Kasai T, Takakuwa T, Nakae H, Kamei Y, Shimamura T, Suzuki T, Taniguchi S, Yoshida M. Plasma levels of interleukin-1 receptor antagonist (IL-1ra) and severity of illness in patients with burns. J Med. 1996;27(1-2):57–71. [PubMed] [Google Scholar]
- 69. Iglesias J, Marik PE, Levine JS, Norasept IISI; Norasept II Study Investigators. Elevated serum levels of the type I and type II receptors for tumor necrosis factor-alpha as predictive factors for ARF in patients with septic shock. Am J Kidney Dis. 2003;41(1):62–75. [DOI] [PubMed] [Google Scholar]
- 70. Garber AJ. Long-acting glucagon-like peptide 1 receptor agonists: a review of their efficacy and tolerability. Diabetes Care. 2011;34(Suppl 2):S279–S284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Nauck MA. Incretin-based therapies for type 2 diabetes mellitus: properties, functions, and clinical implications. Am J Med. 2011;124(1, Suppl):S3–S18. [DOI] [PubMed] [Google Scholar]
- 72. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsbøll T, Investigators S; SUSTAIN-6 Investigators. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375(19):1834–1844. [DOI] [PubMed] [Google Scholar]
- 73. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB, LEADER Steering Committee, LEADER Trial Investigators. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Zheng SL, Roddick AJ, Aghar-Jaffar R, Shun-Shin MJ, Francis D, Oliver N, Meeran K. Association between use of sodium-glucose cotransporter 2 inhibitors, glucagon-like peptide 1 agonists, and dipeptidyl peptidase 4 inhibitors with all-cause mortality in patients with type 2 diabetes: a systematic review and meta-analysis. JAMA. 2018;319(15):1580–1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Hadjiyanni I, Siminovitch KA, Danska JS, Drucker DJ. Glucagon-like peptide-1 receptor signalling selectively regulates murine lymphocyte proliferation and maintenance of peripheral regulatory T cells. Diabetologia. 2010;53(4):730–740. [DOI] [PubMed] [Google Scholar]
- 76. Lee YS, Jun HS. Anti-inflammatory effects of GLP-1-based therapies beyond glucose control. Mediators Inflamm. 2016;2016:3094642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Panjwani N, Mulvihill EE, Longuet C, Yusta B, Campbell JE, Brown TJ, Streutker C, Holland D, Cao X, Baggio LL, Drucker DJ. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(-/-) mice. Endocrinology. 2013;154(1):127–139. [DOI] [PubMed] [Google Scholar]
- 78. Yusta B, Baggio LL, Koehler J, Holland D, Cao X, Pinnell LJ, Johnson-Henry KC, Yeung W, Surette MG, Bang KW, Sherman PM, Drucker DJ. GLP-1R agonists modulate enteric immune responses through the intestinal intraepithelial lymphocyte GLP-1R. Diabetes. 2015;64(7):2537–2549. [DOI] [PubMed] [Google Scholar]
- 79. Bułdak Ł, Machnik G, Bułdak RJ, Łabuzek K, Bołdys A, Belowski D, Basiak M, Okopień B. Exenatide (a GLP-1 agonist) expresses anti-inflammatory properties in cultured human monocytes/macrophages in a protein kinase A and B/Akt manner [published correction appears in Pharmacol Rep. 2017;69(2):376]. Pharmacol Rep. 2016;68(2):329–337. [DOI] [PubMed] [Google Scholar]
- 80. Guo C, Huang T, Chen A, Chen X, Wang L, Shen F, Gu X. Glucagon-like peptide 1 improves insulin resistance in vitro through anti-inflammation of macrophages. Braz J Med Biol Res. 2016;49(12):e5826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117(1):175–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Krasner NM, Ido Y, Ruderman NB, Cacicedo JM. Glucagon-like peptide-1 (GLP-1) analog liraglutide inhibits endothelial cell inflammation through a calcium and AMPK dependent mechanism. PLoS One. 2014;9(5):e97554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Liu H, Dear AE, Knudsen LB, Simpson RW. A long-acting glucagon-like peptide-1 analogue attenuates induction of plasminogen activator inhibitor type-1 and vascular adhesion molecules. J Endocrinol. 2009;201(1):59–66. [DOI] [PubMed] [Google Scholar]
- 84. Lee W, Ku SK, Park EJ, Na DH, Kim KM, Bae JS. Exendin-4 inhibits HMGB1-induced inflammatory responses in HUVECs and in murine polymicrobial sepsis. Inflammation. 2014;37(5):1876–1888. [DOI] [PubMed] [Google Scholar]
- 85. Lee W, Park EJ, Kwak S, Lee KC, Na DH, Bae JS. Trimeric PEG-conjugated exendin-4 for the treatment of sepsis. Biomacromolecules. 2016;17(3):1160–1169. [DOI] [PubMed] [Google Scholar]
- 86. Chen A, Chen Z, Xia Y, Lu D, Yang X, Sun A, Zou Y, Qian J, Ge J. Liraglutide attenuates NLRP3 inflammasome-dependent pyroptosis via regulating SIRT1/NOX4/ROS pathway in H9c2 cells. Biochem Biophys Res Commun. 2018;499(2):267–272. [DOI] [PubMed] [Google Scholar]
- 87. Jojima T, Uchida K, Akimoto K, Tomotsune T, Yanagi K, Iijima T, Suzuki K, Kasai K, Aso Y. Liraglutide, a GLP-1 receptor agonist, inhibits vascular smooth muscle cell proliferation by enhancing AMP-activated protein kinase and cell cycle regulation, and delays atherosclerosis in ApoE deficient mice. Atherosclerosis. 2017;261:44–51. [DOI] [PubMed] [Google Scholar]
- 88. Zeng Y, Li C, Guan M, Zheng Z, Li J, Xu W, Wang L, He F, Xue Y. The DPP-4 inhibitor sitagliptin attenuates the progress of atherosclerosis in apolipoprotein-E-knockout mice via AMPK- and MAPK-dependent mechanisms. Cardiovasc Diabetol. 2014;13(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Nagashima M, Watanabe T, Terasaki M, Tomoyasu M, Nohtomi K, Kim-Kaneyama J, Miyazaki A, Hirano T. Native incretins prevent the development of atherosclerotic lesions in apolipoprotein E knockout mice. Diabetologia. 2011;54(10):2649–2659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Nogi Y, Nagashima M, Terasaki M, Nohtomi K, Watanabe T, Hirano T. Glucose-dependent insulinotropic polypeptide prevents the progression of macrophage-driven atherosclerosis in diabetic apolipoprotein E-null mice. PLoS One. 2012;7(4):e35683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Hirano T, Mori Y. Anti-atherogenic and anti-inflammatory properties of glucagon-like peptide-1, glucose-dependent insulinotropic polypepide, and dipeptidyl peptidase-4 inhibitors in experimental animals. J Diabetes Investig. 2016;7(Suppl 1):80–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Lee YS, Park MS, Choung JS, Kim SS, Oh HH, Choi CS, Ha SY, Kang Y, Kim Y, Jun HS. Glucagon-like peptide-1 inhibits adipose tissue macrophage infiltration and inflammation in an obese mouse model of diabetes. Diabetologia. 2012;55(9):2456–2468. [DOI] [PubMed] [Google Scholar]
- 93. Pastel E, Joshi S, Knight B, Liversedge N, Ward R, Kos K. Effects of Exendin-4 on human adipose tissue inflammation and ECM remodelling. Nutr Diabetes. 2016;6(12):e235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Dobrian AD, Ma Q, Lindsay JW, Leone KA, Ma K, Coben J, Galkina EV, Nadler JL. Dipeptidyl peptidase IV inhibitor sitagliptin reduces local inflammation in adipose tissue and in pancreatic islets of obese mice. Am J Physiol Endocrinol Metab. 2011;300(2):E410–E421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Langlois A, Dal S, Vivot K, Mura C, Seyfritz E, Bietiger W, Dollinger C, Peronet C, Maillard E, Pinget M, Jeandidier N, Sigrist S. Improvement of islet graft function using liraglutide is correlated with its anti-inflammatory properties. Br J Pharmacol. 2016;173(24):3443–3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Li L, El-Kholy W, Rhodes CJ, Brubaker PL. Glucagon-like peptide-1 protects beta cells from cytokine-induced apoptosis and necrosis: role of protein kinase B. Diabetologia. 2005;48(7):1339–1349. [DOI] [PubMed] [Google Scholar]
- 97. Gao H, Zeng Z, Zhang H, Zhou X, Guan L, Deng W, Xu L. The glucagon-like peptide-1 analogue liraglutide inhibits oxidative stress and inflammatory response in the liver of rats with diet-induced non-alcoholic fatty liver disease. Biol Pharm Bull. 2015;38(5):694–702. [DOI] [PubMed] [Google Scholar]
- 98. Wang Y, Parlevliet ET, Geerling JJ, van der Tuin SJ, Zhang H, Bieghs V, Jawad AH, Shiri-Sverdlov R, Bot I, de Jager SC, Havekes LM, Romijn JA, Willems van Dijk K, Rensen PC. Exendin-4 decreases liver inflammation and atherosclerosis development simultaneously by reducing macrophage infiltration. Br J Pharmacol. 2014;171(3):723–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Birnbaum Y, Bajaj M, Qian J, Ye Y. Dipeptidyl peptidase-4 inhibition by Saxagliptin prevents inflammation and renal injury by targeting the Nlrp3/ASC inflammasome. BMJ Open Diabetes Res Care. 2016;4(1):e000227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Kodera R, Shikata K, Kataoka HU, Takatsuka T, Miyamoto S, Sasaki M, Kajitani N, Nishishita S, Sarai K, Hirota D, Sato C, Ogawa D, Makino H. Glucagon-like peptide-1 receptor agonist ameliorates renal injury through its anti-inflammatory action without lowering blood glucose level in a rat model of type 1 diabetes. Diabetologia. 2011;54(4):965–978. [DOI] [PubMed] [Google Scholar]
- 101. Sancar-Bas S, Gezginci-Oktayoglu S, Bolkent S. Exendin-4 attenuates renal tubular injury by decreasing oxidative stress and inflammation in streptozotocin-induced diabetic mice. Growth Factors. 2015;33(5-6):419–429. [DOI] [PubMed] [Google Scholar]
- 102. Chen S, Okahara F, Osaki N, Shimotoyodome A. Increased GIP signaling induces adipose inflammation via a HIF-1α-dependent pathway and impairs insulin sensitivity in mice. Am J Physiol Endocrinol Metab. 2015;308(5):E414–E425. [DOI] [PubMed] [Google Scholar]
- 103. Gögebakan Ö, Osterhoff MA, Schüler R, Pivovarova O, Kruse M, Seltmann AC, Mosig AS, Rudovich N, Nauck M, Pfeiffer AF. GIP increases adipose tissue expression and blood levels of MCP-1 in humans and links high energy diets to inflammation: a randomised trial. Diabetologia. 2015;58(8):1759–1768. [DOI] [PubMed] [Google Scholar]
- 104. Varol C, Zvibel I, Spektor L, Mantelmacher FD, Vugman M, Thurm T, Khatib M, Elmaliah E, Halpern Z, Fishman S. Long-acting glucose-dependent insulinotropic polypeptide ameliorates obesity-induced adipose tissue inflammation. J Immunol. 2014;193(8):4002–4009. [DOI] [PubMed] [Google Scholar]
- 105. Zhuge F, Ni Y, Nagashimada M, Nagata N, Xu L, Mukaida N, Kaneko S, Ota T. DPP-4 inhibition by linagliptin attenuates obesity-related inflammation and insulin resistance by regulating M1/M2 macrophage polarization. Diabetes. 2016;65(10):2966–2979. [DOI] [PubMed] [Google Scholar]
- 106. Bertilsson G, Patrone C, Zachrisson O, Andersson A, Dannaeus K, Heidrich J, Kortesmaa J, Mercer A, Nielsen E, Rönnholm H, Wikström L. Peptide hormone exendin-4 stimulates subventricular zone neurogenesis in the adult rodent brain and induces recovery in an animal model of Parkinson’s disease. J Neurosci Res. 2008;86(2):326–338. [DOI] [PubMed] [Google Scholar]
- 107. Bomfim TR, Forny-Germano L, Sathler LB, Brito-Moreira J, Houzel JC, Decker H, Silverman MA, Kazi H, Melo HM, McClean PL, Holscher C, Arnold SE, Talbot K, Klein WL, Munoz DP, Ferreira ST, De Felice FG. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease-associated Aβ oligomers. J Clin Invest. 2012;122(4):1339–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Iwai T, Ito S, Tanimitsu K, Udagawa S, Oka J. Glucagon-like peptide-1 inhibits LPS-induced IL-1beta production in cultured rat astrocytes. Neurosci Res. 2006;55(4):352–360. [DOI] [PubMed] [Google Scholar]
- 109. Iwai T, Sawabe T, Tanimitsu K, Suzuki M, Sasaki-Hamada S, Oka J. Glucagon-like peptide-1 protects synaptic and learning functions from neuroinflammation in rodents. J Neurosci Res. 2014;92(4):446–454. [DOI] [PubMed] [Google Scholar]
- 110. Daousi C, Pinkney JH, Cleator J, Wilding JP, Ranganath LR. Acute peripheral administration of synthetic human GLP-1 (7-36 amide) decreases circulating IL-6 in obese patients with type 2 diabetes mellitus: a potential role for GLP-1 in modulation of the diabetic pro-inflammatory state? Regul Pept. 2013;183:54–61. [DOI] [PubMed] [Google Scholar]
- 111. Chaudhuri A, Ghanim H, Vora M, Sia CL, Korzeniewski K, Dhindsa S, Makdissi A, Dandona P. Exenatide exerts a potent antiinflammatory effect. J Clin Endocrinol Metab. 2012;97(1):198–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Derosa G, Franzetti IG, Querci F, Carbone A, Ciccarelli L, Piccinni MN, Fogari E, Maffioli P. Variation in inflammatory markers and glycemic parameters after 12 months of exenatide plus metformin treatment compared with metformin alone: a randomized placebo-controlled trial. Pharmacotherapy. 2013;33(8):817–826. [DOI] [PubMed] [Google Scholar]
- 113. Hogan AE, Gaoatswe G, Lynch L, Corrigan MA, Woods C, O’Connell J, O’Shea D. Glucagon-like peptide 1 analogue therapy directly modulates innate immune-mediated inflammation in individuals with type 2 diabetes mellitus. Diabetologia. 2014;57(4):781–784. [DOI] [PubMed] [Google Scholar]
- 114. Satoh-Asahara N, Sasaki Y, Wada H, Tochiya M, Iguchi A, Nakagawachi R, Odori S, Kono S, Hasegawa K, Shimatsu A. A dipeptidyl peptidase-4 inhibitor, sitagliptin, exerts anti-inflammatory effects in type 2 diabetic patients. Metabolism. 2013;62(3):347–351. [DOI] [PubMed] [Google Scholar]
- 115. Tremblay AJ, Lamarche B, Deacon CF, Weisnagel SJ, Couture P. Effects of sitagliptin therapy on markers of low-grade inflammation and cell adhesion molecules in patients with type 2 diabetes. Metabolism. 2014;63(9):1141–1148. [DOI] [PubMed] [Google Scholar]
- 116. Yanay O, Bailey AL, Kernan K, Zimmerman JJ, Osborne WR. Effects of exendin-4, a glucagon like peptide-1 receptor agonist, on neutrophil count and inflammatory cytokines in a rat model of endotoxemia. J Inflamm Res. 2015;8:129–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Steven S, Hausding M, Kröller-Schön S, Mader M, Mikhed Y, Stamm P, Zinßius E, Pfeffer A, Welschof P, Agdauletova S, Sudowe S, Li H, Oelze M, Schulz E, Klein T, Münzel T, Daiber A. Gliptin and GLP-1 analog treatment improves survival and vascular inflammation/dysfunction in animals with lipopolysaccharide-induced endotoxemia. Basic Res Cardiol. 2015;110(2):6. [DOI] [PubMed] [Google Scholar]
- 118. Steven S, Jurk K, Kopp M, Kröller-Schön S, Mikhed Y, Schwierczek K, Roohani S, Kashani F, Oelze M, Klein T, Tokalov S, Danckwardt S, Strand S, Wenzel P, Münzel T, Daiber A. Glucagon-like peptide-1 receptor signalling reduces microvascular thrombosis, nitro-oxidative stress and platelet activation in endotoxaemic mice. Br J Pharmacol. 2017;174(12):1620–1632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Dozier KC, Cureton EL, Kwan RO, Curran B, Sadjadi J, Victorino GP. Glucagon-like peptide-1 protects mesenteric endothelium from injury during inflammation. Peptides. 2009;30(9):1735–1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Shah FA, Singamsetty S, Guo L, Chuan BW, McDonald S, Cooper BA, O’Donnell BJ, Stefanovski D, Wice B, Zhang Y, O’Donnell CP, McVerry BJ. Stimulation of the endogenous incretin glucose-dependent insulinotropic peptide by enteral dextrose improves glucose homeostasis and inflammation in murine endotoxemia. Transl Res. 2018;193:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Hulst AH, Plummer MP, Hollmann MW, DeVries JH, Preckel B, Deane AM, Hermanides J. Systematic review of incretin therapy during peri-operative and intensive care. Crit Care. 2018;22(1):299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Plummer MP, Hermanides J, Deane AM. Incretin physiology and pharmacology in the intensive care unit. Crit Care Clin. 2019;35(2):341–355. [DOI] [PubMed] [Google Scholar]
- 123. Meier JJ, Gallwitz B, Kask B, Deacon CF, Holst JJ, Schmidt WE, Nauck MA. Stimulation of insulin secretion by intravenous bolus injection and continuous infusion of gastric inhibitory polypeptide in patients with type 2 diabetes and healthy control subjects. Diabetes. 2004;53(Suppl 3):S220–S224. [DOI] [PubMed] [Google Scholar]
- 124. Sokos GG, Bolukoglu H, German J, Hentosz T, Magovern GJ Jr, Maher TD, Dean DA, Bailey SH, Marrone G, Benckart DH, Elahi D, Shannon RP. Effect of glucagon-like peptide-1 (GLP-1) on glycemic control and left ventricular function in patients undergoing coronary artery bypass grafting. Am J Cardiol. 2007;100(5):824–829. [DOI] [PubMed] [Google Scholar]
- 125. Deane AM, Chapman MJ, Fraser RJ, Burgstad CM, Besanko LK, Horowitz M. The effect of exogenous glucagon-like peptide-1 on the glycaemic response to small intestinal nutrient in the critically ill: a randomised double-blind placebo-controlled cross over study. Crit Care. 2009;13(3):R67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Deane AM, Chapman MJ, Fraser RJ, Summers MJ, Zaknic AV, Storey JP, Jones KL, Rayner CK, Horowitz M. Effects of exogenous glucagon-like peptide-1 on gastric emptying and glucose absorption in the critically ill: relationship to glycemia. Crit Care Med. 2010;38(5):1261–1269. [DOI] [PubMed] [Google Scholar]
- 127. Deane AM, Summers MJ, Zaknic AV, Chapman MJ, Fraser RJ, Di Bartolomeo AE, Wishart JM, Horowitz M. Exogenous glucagon-like peptide-1 attenuates the glycaemic response to postpyloric nutrient infusion in critically ill patients with type-2 diabetes. Crit Care. 2011;15(1):R35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Galiatsatos P, Gibson BR, Rabiee A, Carlson O, Egan JM, Shannon RP, Andersen DK, Elahi D. The glucoregulatory benefits of glucagon-like peptide-1 (7-36) amide infusion during intensive insulin therapy in critically ill surgical patients: a pilot study. Crit Care Med. 2014;42(3):638–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Miller A, Deane AM, Plummer MP, Cousins CE, Chapple LS, Horowitz M, Chapman MJ. Exogenous glucagon-like peptide-1 attenuates glucose absorption and reduces blood glucose concentration after small intestinal glucose delivery in critical illness. Crit Care Resusc. 2017;19(1):37–42. [PubMed] [Google Scholar]
- 130. Abuannadi M, Kosiborod M, Riggs L, House JA, Hamburg MS, Kennedy KF, Marso SP. Management of hyperglycemia with the administration of intravenous exenatide to patients in the cardiac intensive care unit. Endocr Pract. 2013;19(1):81–90. [DOI] [PubMed] [Google Scholar]
- 131. Kar P, Cousins CE, Annink CE, Jones KL, Chapman MJ, Meier JJ, Nauck MA, Horowitz M, Deane AM. Effects of glucose-dependent insulinotropic polypeptide on gastric emptying, glycaemia and insulinaemia during critical illness: a prospective, double blind, randomised, crossover study. Crit Care. 2015;19(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Lee MY, Fraser JD, Chapman MJ, Sundararajan K, Umapathysivam MM, Summers MJ, Zaknic AV, Rayner CK, Meier JJ, Horowitz M, Deane AM. The effect of exogenous glucose-dependent insulinotropic polypeptide in combination with glucagon-like peptide-1 on glycemia in the critically ill. Diabetes Care. 2013;36(10):3333–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Meier JJ, Weyhe D, Michaely M, Senkal M, Zumtobel V, Nauck MA, Holst JJ, Schmidt WE, Gallwitz B. Intravenous glucagon-like peptide 1 normalizes blood glucose after major surgery in patients with type 2 diabetes. Crit Care Med. 2004;32(3):848–851. [DOI] [PubMed] [Google Scholar]

