The inflammatory response in sepsis

Markus Bosmann, Peter A Ward, Markus Bosmann, Peter A Ward

Abstract

The pathophysiology of sepsis and its accompanying systemic inflammatory response syndrome (SIRS) and the events that lead to multiorgan failure and death are poorly understood. It is known that, in septic humans and rodents, the development of SIRS is associated with a loss of the redox balance, but SIRS can also develop in noninfectious states. In addition, a hyperinflammatory state develops, together with impaired innate immune functions of phagocytes, immunosuppression, and complement activation, collectively leading to septic shock and lethality. Here, we discuss recent insights into the signaling pathways in immune and phagocytic cells that underlie sepsis and SIRS and consider how these might be targeted for therapeutic interventions to reverse or attenuate pathways that lead to lethality during sepsis.

Copyright © 2012 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Sepsis-induced defects in redox balance and anti-oxidant enzymes. In sepsis, an excessive oxidant state (presence of elevated levels of ROS, RNS) leads to reduced ubiquitination and nuclear translocation of the transcription factor, Nrf2, which in turn leads to reduced activation of genes that encode for anti-oxidant enzymes [19, 20, 22]. This imbalance intensifies the SIRS response and enhances development of MOF and lethality. Restoration of the redox balance by inducers of Nrf2 may reduce the intensity of SIRS and downstream events that would otherwise be lethal, at least based on studies in CLP mice. Dietary products, such as sulforaphane, cause of induction of Nrf2 and may be candidates for reversal of the redox imbalance in sepsis [18].
Figure 2
Figure 2
Sepsis-induced defects in redox system and depletion of cellular ATP. The nuclear hormone peroxisome proliferator activity receptors (PPARs) participate in responses to oxidative stresses, resulting in preservation of mitochondrial function and containment of inflammation [20, 26-30]. Sepsis causes reduced levels of PPARs accompanied by defective mitochondrial function as well as reductions in mitochondrial ATP, and reduced ability to restrain inflammatory responses such as SIRS. The two insets are transmission electron micrographs showing swollen mitochondria (in liver) during sepsis (X12,500). Several agonists for PPARs have been developed [27, 32, 33]. Sepsis and increased ROS and RNS also trigger DNA single strand breaks, which results in activation of poly ADP-ribose polymerase (PARP) for repair of DNA [17]. In the process, depletion of NAD+ levels can occur, accompanied by reductions in cellular and mitochondrial ATP. Inhibitors for PARP have been developed, which may preserve mitochondrial ATP during sepsis.
Figure 3
Figure 3
C5a and C5a receptors as targets in sepsis. In sepsis, C5a, and its receptors (C5aR, C5L2), are involved in a series of responses that promote development of SIRS, MOF, septic shock and lethality. Early in sepsis, robust activation of complement occurs, generating C5a which interacts with its receptors to promote development of ROS, RNS and onset of SIRS. C5aR and C5L2 are expressed in large amounts by neutrophils, at lower levels in macrophages and monocytes and non-myeloid cells (endothelial cells, alveolar epithelial cells, Kupffer cells, cardiomyocytes, etc.) [35, 36]. Development of SIRS and its complications (immunosuppression, defective innate and adaptive immune responses, consumptive coagulopathy) results in a sequence of events that is often lethal. In vivo neutralization of C5a with antibodies or blockade of C5a receptors (C5aR, C5L2) suppresses this cascade of adverse events and improves survival in CLP rodents. In vivo, blockade of C5a with neutralizing antibody in CLP rodents greatly improves survival, even if given up to 12 h after CLP [8].
Figure 4
Figure 4
Role of the parasympathetic and sympathetic nervous systems (PNS, SNS, respectively) in sepsis. a) The PNS, also referred to as the cholinergic system, acts via the vagus nerve to release acetylcholine from T cells, suppressing proinflammatory mediator release form activated macrophages and T cells [38]. b) Depending on which adrenergic receptor is in play, the SNS pathway may either intensify [45] or suppress the lung inflammatory response [46]. The cholinergic pathway can be activated by vagal nerve stimulation or use by chemical antagonists of the acetylcholine receptor, α7nAchR, resulting in suppressed release of proinflammatory mediators from macrophages. The SNS pathways can suppress the inflammatory response in the presence of β2AR agonists (formoterol, albuterol). Alternatively, agonists (epinephrine, norepinephrine) of β2ARs will intensify the lung inflammatory response [45]. Thus there are several strategies to target the PNS or SNS that may be used to suppress inflammatory responses and reduce ROS/RNS formation, the intensity of SIRS and other adverse events in sepsis.
Figure 5
Figure 5
Sepsis-induced immunosuppression and defective phagocytes. Sepsis leads to immunosuppression and defective phagocytosis, caused by depletion or functional deficiencies in macrophages and DCs, apoptosis of T and B cells, and tissue expression of inhibitory ligands and receptors that suppress immune responses, mostly by targeting T cells. Immunosuppression and defective phagocyte function, leads to failure to contain commensal and invading bacteria and fungi. The immunostimulant, IL-7, which promotes T and B cell proliferation and which has antiapoptotic effects, may be a candidate for clinical trials in humans to reverse the immunosuppression of sepsis [52].

References

    1. Martin GS, et al. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med. 2003;348:1546–1554.
    1. Coopersmith CM, et al. A comparison of critical care research funding and the financial burden of critical illness in the United States*. Crit Care Med. 2012;40:1072–1079.
    1. [Accessed November 18, 2011];Xigris [drotegogin alfa (activated)]: market withdrawal-failure survival benefit. 2011 . Posted October 25, 2011.
    1. Angus DC. The search for effective therapy for sepsis: back to the drawing board? JAMA. 2011;306:2614–2615.
    1. Erridge C. Endogenous ligands of TLR2 and TLR4: agonists or assistants? J Leukoc Biol. 2010;87:989–999.
    1. Clark JA, Coopersmith CM. Intestinal crosstalk: a new paradigm for understanding the gut as the “motor” of critical illness. Shock. 2007;28:384–393.
    1. Cai B, et al. Novel insights for systemic inflammation in sepsis and hemorrhage. Mediators Inflamm. 2010;2010:642462.
    1. Ward PA. The harmful role of C5a on innate immunity in sepsis. J Innate Immun. 2010;2:439–445.
    1. Andrades M, et al. Bench-to-bedside review: Sepsis - from the redox point of view. Critical Care. 2011;15:230.
    1. Berger MM, Chiolero RL. Antioxidant supplementation in sepsis and systemic inflammatory response syndrome. Crit Care Med. 2007;35:S584–590.
    1. Crimi E, et al. Role of oxidative stress in experimental sepsis and multisystem organ dysfunction. Free Radic Res. 2006;40:665–672.
    1. von Dessauer B, et al. Oxidative stress as a novel target in pediatric sepsis management. J Crit Care. 2011;26:103, e101–107.
    1. van Berlo D, et al. Neutrophil-derived ROS contribute to oxidative DNA damage induction by quartz particles. Free Radic Biol Med. 2010;49:1685–1693.
    1. Moore SF, MacKenzie AB. NADPH oxidase NOX2 mediates rapid cellular oxidation following ATP stimulation of endotoxin-primed macrophages. J Immunol. 2009;183:3302–3308.
    1. Lipinski B. Hydroxyl radical and its scavengers in health and disease. Oxid Med Cell Longev. 2011;2011:809696.
    1. Valko M, et al. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol. 2007;39:44–84.
    1. Martin LJ. DNA damage and repair: relevance to mechanisms of neurodegeneration. J Neuropathol Exp Neurol. 2008;67:377–387.
    1. Zakkar M, et al. Activation of Nrf2 in endothelial cells protects arteries from exhibiting a proinflammatory state. Arterioscler Thromb Vasc Biol. 2009;29:1851–1857.
    1. MacGarvey NC, et al. Activation of mitochondrial biogenesis by heme oxygenase-1-mediated NF-E2-related factor-2 induction rescues mice from lethal Staphylococcus aureus sepsis. Am J Respir Crit Care Med. 2012;185:851–861.
    1. Kong X, et al. Enhancing Nrf2 pathway by disruption of Keap1 in myeloid leukocytes protects against sepsis. Am J Respir Crit Care Med. 2011;184:928–938.
    1. Bell KF, et al. Activation of Nrf2-regulated glutathione pathway genes by ischemic preconditioning. Oxid Med Cell Longev. 2011;2011:689524.
    1. Kensler TW, Wakabayashi N. Nrf2: friend or foe for chemoprevention? Carcinogenesis. 2010;31:90–99.
    1. Sirgo G, et al. [Dendritic cells in sepsis: an approach to post-infectious immunosuppression] Med Intensiva. 2010;34:559–566.
    1. Scumpia PO, et al. CD11c+ dendritic cells are required for survival in murine polymicrobial sepsis. J Immunol. 2005;175:3282–3286.
    1. Delano MJ, et al. Sepsis induces early alterations in innate immunity that impact mortality to secondary infection. J Immunol. 2011;186:195–202.
    1. Polvani S, et al. PPARgamma and Oxidative Stress: Con(beta) Catenating NRF2 and FOXO. PPAR Res. 2012;2012:641087.
    1. Kaplan JM, Zingarelli B. Novel Therapeutic Agents in Pediatric Sepsis: Peroxisome Proliferator Receptor gamma (PPAR gamma) Agonists. Open Inflamm J. 2011;4:120–124.
    1. Barrera G, et al. The Role of PPAR Ligands in Controlling Growth-Related Gene Expression and their Interaction with Lipoperoxidation Products. PPAR Res. 2008;2008:524671.
    1. Choi JH, et al. Antidiabetic actions of a non-agonist PPARgamma ligand blocking Cdk5-mediated phosphorylation. Nature. 2011;477:477–481.
    1. Standage SW, et al. Reduced peroxisome proliferator-activated receptor alpha expression is associated with decreased survival and increased tissue bacterial load in sepsis. Shock. 2012;37:164–169.
    1. Vachharajani V, et al. Curcumin modulates leukocyte and platelet adhesion in murine sepsis. Microcirculation. 2010;17:407–416.
    1. Sun Y, Bennett A. Cannabinoids: a new group of agonists of PPARs. PPAR Res. 2007;2007:23513.
    1. Khazaei M, et al. Pan-PPAR Agonist, Bezafibrate, Restores Angiogenesis in Hindlimb Ischemia in Normal and Diabetic Rats. Int J Pept. 2012;2012:637212.
    1. Mathews MT, Berk BC. PARP-1 inhibition prevents oxidative and nitrosative stress-induced endothelial cell death via transactivation of the VEGF receptor 2. Arterioscler Thromb Vasc Biol. 2008;28:711–717.
    1. Bamberg CE, et al. The C5a receptor (C5aR) C5L2 is a modulator of C5aR-mediated signal transduction. J Biol Chem. 2010;285:7633–7644.
    1. Bosmann M, et al. Complement activation product C5a is a selective suppressor of TLR4-induced, but not TLR3-induced, production of IL-27(p28) from macrophages. J Immunol. 2012;188:5086–5093.
    1. Speyer CL, Ward PA. Role of endothelial chemokines and their receptors during inflammation. J Invest Surg. 2011;24:18–27.
    1. Andersson U, Tracey KJ. Reflex principles of immunological homeostasis. Annu Rev Immunol. 2012;30:313–335.
    1. Borovikova LV, et al. Role of vagus nerve signaling in CNI-1493-mediated suppression of acute inflammation. Auton Neurosci. 2000;85:141–147.
    1. Bonaz B. The cholinergic anti-inflammatory pathway and the gastrointestinal tract. Gastroenterology. 2007;133:1370–1373.
    1. Kamal A. Assessment of autonomic function in patients with rheumatoid arthritis using spectral analysis and approximate entropy method. Neurosciences (Riyadh) 2007;12:136–139.
    1. Stojanovich L, et al. Cardiovascular autonomic dysfunction in systemic lupus, rheumatoid arthritis, primary Sjogren syndrome and other autoimmune diseases. Lupus. 2007;16:181–185.
    1. Pierrakos C, et al. Acute respiratory distress syndrome: pathophysiology and therapeutic options. J Clin Med Res. 2012;4:7–16.
    1. Matthay MA, et al. The acute respiratory distress syndrome. J Clin Invest. 2012;122:2731–2740.
    1. Flierl MA, et al. Phagocyte-derived catecholamines enhance acute inflammatory injury. Nature. 2007;449:721–725.
    1. Bosmann M, et al. Anti-inflammatory effects of beta2 adrenergic receptor agonists in experimental acute lung injury. FASEB J. 2012;26:2137–2144.
    1. Boomer JS, et al. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA. 2011;306:2594–2605.
    1. Ward PA. Immunosuppression in sepsis. JAMA. 2011;306:2618–2619.
    1. Hotchkiss RS, et al. The sepsis seesaw: tilting toward immunosuppression. Nat Med. 2009;15:496–497.
    1. Takahashi HK, et al. Activation of survival and apoptotic signaling pathways in lymphocytes exposed to palmitic acid. J Cell Physiol. 2012;227:339–350.
    1. Warner EA, Moldawer LL. Using innate immunity to characterize the host response to microbial invasion in severe sepsis. Future Microbiol. 2008;3:177–189.
    1. Unsinger J, et al. IL-7 promotes T cell viability, trafficking, and functionality and improves survival in sepsis. J Immunol. 2010;184:3768–3779.
    1. Sereti I. Where have all the T cells gone? Blood. 2009;114:751–752.
    1. Inoue S, et al. IL-15 prevents apoptosis, reverses innate and adaptive immune dysfunction, and improves survival in sepsis. J Immunol. 2010;184:1401–1409.
    1. Dejager L, et al. Cecal ligation and puncture: the gold standard model for polymicrobial sepsis? Trends Microbiol. 2011;19:198–208.
    1. Rittirsch D, et al. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009;4:31–36.
    1. Lissauer ME, et al. Differential expression of toll-like receptor genes: sepsis compared with sterile inflammation 1 day before sepsis diagnosis. Shock. 2009;31:238–244.
    1. Remick DG, Ward PA. Evaluation of endotoxin models for the study of sepsis. Shock. 2005;24(Suppl 1):7–11.
    1. Schmidt WM, et al. In vivo profile of the human leukocyte microRNA response to endotoxemia. Biochem Biophys Res Commun. 2009;380:437–441.
    1. Zanotti-Cavazzoni SL, Goldfarb RD. Animal models of sepsis. Crit Care Clin. 2009;25:703–719. vii–viii.
    1. Fink MP. Animal models of sepsis and its complications. Kidney Int. 2008;74:991–993.
    1. Dyson A, Singer M. Animal models of sepsis: why does preclinical efficacy fail to translate to the clinical setting? Crit Care Med. 2009;37:S30–37.
    1. Doi K, et al. Animal models of sepsis and sepsis-induced kidney injury. J Clin Invest. 2009;119:2868–2878.
    1. Silva E, et al. Sepsis: from bench to bedside. Clinics (Sao Paulo) 2008;63:109–120.
    1. Cavassani KA, et al. The post sepsis-induced expansion and enhanced function of regulatory T cells create an environment to potentiate tumor growth. Blood. 2010;115:4403–4411.
    1. Angus DC. The lingering consequences of sepsis: a hidden public health disaster? JAMA. 2010;304:1833–1834.
    1. Lopes JA, et al. Long-term risk of mortality after acute kidney injury in patients with sepsis: a contemporary analysis. BMC Nephrol. 2010;11:9.

Source: PubMed

3
購読する