The dysfunction of complement and coagulation in diseases: the implications for the therapeutic interventions

Honghong Jiang , Yiming Guo , Qihang Wang , Yiran Wang , Dingchuan Peng , Yigong Fang , Lei Yan , Zhuolin Ruan , Sheng Zhang , Yong Zhao , Wendan Zhang , Wei Shang , Zhichun Feng

MedComm ›› 2024, Vol. 5 ›› Issue (11) : e785

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (11) : e785 DOI: 10.1002/mco2.785
REVIEW

The dysfunction of complement and coagulation in diseases: the implications for the therapeutic interventions

Author information +
History +
PDF

Abstract

The complement system, comprising over 30 proteins, is integral to the immune system, and the coagulation system is critical for vascular homeostasis. The activation of the complement and coagulation systems involves an organized proteolytic cascade, and the overactivation of these systems is a central pathogenic mechanism in several diseases. This review describes the role of complement and coagulation system activation in critical illness, particularly sepsis. The complexities of sepsis reveal significant knowledge gaps that can be compared to a profound abyss, highlighting the urgent need for further investigation and exploration. It is well recognized that the inflammatory network, coagulation, and complement systems are integral mechanisms through which multiple factors contribute to increased susceptibility to infection and may result in a disordered immune response during septic events in patients. Given the overlapping pathogenic mechanisms in sepsis, immunomodulatory therapies currently under development may be particularly beneficial for patients with sepsis who have concurrent infections. Herein, we present recent findings regarding the molecular relationships between the coagulation and complement pathways in the advancement of sepsis, and propose potential intervention targets related to the crosstalk between coagulation and complement, aiming to provide more valuable treatment of sepsis.

Keywords

coagulation / complement system / immune response / inflammatory network / sepsis

Cite this article

Download citation ▾
Honghong Jiang, Yiming Guo, Qihang Wang, Yiran Wang, Dingchuan Peng, Yigong Fang, Lei Yan, Zhuolin Ruan, Sheng Zhang, Yong Zhao, Wendan Zhang, Wei Shang, Zhichun Feng. The dysfunction of complement and coagulation in diseases: the implications for the therapeutic interventions. MedComm, 2024, 5(11): e785 DOI:10.1002/mco2.785

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Nesargikar P, Spiller B, Chavez R. The complement system: history, pathways, cascade and inhibitors. Eur J Microbiol Immunol (Bp). 2012; 2(2): 103-111.

[2]

Lan R, Mark J, Alport JW, et al. Advances in Immunology. N Engl J Med. 2001; 344(14): 1058-1066.

[3]

Nunes M, Vlok M, Proal A, et al. Data-independent LC-MS/MS analysis of ME/CFS plasma reveals a dysregulated coagulation system, endothelial dysfunction, downregulation of complement machinery. Cardiovasc Diabetol. 2024; 23(1): 254.

[4]

Detmer SA, Chan DC. Complementation between mouse Mfn1 and Mfn2 protects mitochondrial fusion defects caused by CMT2A disease mutations. J Cell Biol. 2007; 176(4): 405-414.

[5]

Becker N, Franz N, Eguchi A, et al. Elevated extracellular particle concentration in plasma predicts in-hospital mortality after severe trauma. Front Immunol. 2024; 15: 1390380.

[6]

Zhang W, Jiang H, Huang P, et al. Dracorhodin targeting CMPK2 attenuates inflammation: a novel approach to sepsis therapy. Clin Transl Med. 2023; 13(10): e1449.

[7]

Borkowska S, Suszynska M, Mierzejewska K, et al. Novel evidence that crosstalk between the complement, coagulation and fibrinolysis proteolytic cascades is involved in mobilization of hematopoietic stem/progenitor cells (HSPCs). Leukemia. 2014; 28(11): 2148-2154.

[8]

Merle NS, Noe R, Halbwachs-Mecarelli L, et al. Complement system part II: role in immunity. Front Immunol. 2015; 6(May): 1-26.

[9]

Ward PA, Guo RF, Riedemann NC. Manipulation of the complement system for benefit in sepsis. Crit Care Res Pract. 2012; 2012: 427607.

[10]

Cedzyński M, Świerzko AS. Components of the lectin pathway of complement in solid tumour cancers. Cancers (Basel). 2022; 14(6): 1543.

[11]

Garred P, Tenner AJ, Mollnes TE. Therapeutic targeting of the complement system: from rare diseases to pandemics. Pharmacol Rev. 2021; 73(2): 792-827.

[12]

Huber-Lang M, Sarma JV, Zetoune FS, et al. Generation of C5a in the absence of C3: a new complement activation pathway. Nat Med. 2006; 12(6): 682-687.

[13]

Slim MA, van Mourik N, Bakkerus L, et al. Towards personalized medicine: a scoping review of immunotherapy in sepsis. Crit Care. 2024; 28(1): 183.

[14]

Kravitz MS, Kattouf N, Stewart IJ, et al. Plasma for prevention and treatment of glycocalyx degradation in trauma and sepsis. Crit Care. 2024; 28(1): 254.

[15]

Wei X, Tu Y, Bu S, et al. Unraveling the intricate web: complement activation shapes the pathogenesis of sepsis-induced coagulopathy. J Innate Immun. 2024; 31(1): 337-353.

[16]

Karasu E, Nilsson B, Köhl J, et al. Targeting complement pathways in polytrauma-and sepsis-induced multiple-organ dysfunction. Front Immunol. 2019; 10(Mar): 1-14.

[17]

Napier BA, Brubaker SW, Sweeney TE, et al. Complement pathway amplifies caspase-11-dependent cell death and endotoxin-induced sepsis severity. J Exp Med. 2016; 213(11): 2365-2382.

[18]

Slim MA, Turgman O, van Vught LA, et al. Non-conventional immunomodulation in the management of sepsis. Eur J Intern Med. 2024; 121: 9-16.

[19]

Tsao CM, Ho ST, Wu CC. Coagulation abnormalities in sepsis. Acta Anaesthesiol Taiwan. 2015; 53(1): 16-22.

[20]

Huang SSY, Toufiq M, Eghtesady P, et al. The molecular landscape of sepsis severity in infants: enhanced coagulation, innate immunity, and T cell repression. Front Immunol. 2024: 15:1281111.

[21]

Leite GGF, de Brabander J, Michels EHA, et al. Monocyte state 1 (MS1) cells in critically ill patients with sepsis or non-infectious conditions: association with disease course and host response. Crit Care. 2024; 28(1): 88.

[22]

Jackson SP, Darbousset R, Schoenwaelder SM. Thromboinflammation: Challenges of Therapeutically Targeting Coagulation and Other Host Defense Mechanisms. Blood. 2019; 133(9): 906-918. http://ashpublications.org/blood/article-pdf/133/9/906/1558226/blood882993.pdf

[23]

Iba T, Helms J, Levi M, et al. Thromboinflammation in acute injury: infections, heatstroke, and trauma. J Thromb Haemost. 2024; 22(1): 7-22.

[24]

Markiewski MM, Nilsson B, Nilsson Ekdahl K, et al. Complement and coagulation: strangers or partners in crime. Trends Immunol. 2007; 28(4): 184-192.

[25]

Chen CB, Hsu TH, Chung-Yee Hui R, et al. Disseminated intravascular coagulation in Stevens-Johnson syndrome and toxic epidermal necrolysis. J Am Acad Dermatol. 2021; 84(6): 1782-1791.

[26]

Ryan TAJ, Hooftman A, Rehill AM, et al. Dimethyl fumarate and 4-octyl itaconate are anticoagulants that suppress tissue factor in macrophages via inhibition of type I interferon. Nat Commun. 2023; 14(1): 3513.

[27]

Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci. 2024; 138(7): 435-487.

[28]

Hadler MD, Alle H, Geiger JRP. Parvalbumin interneuron cell-to-network plasticity: mechanisms and therapeutic avenues. Trends Pharmacol Sci. 2024; 45(7): 586-601.

[29]

Steinhagen F, Schmidt S, Schewe JC, et al. Immunotherapy in sepsis—brake or accelerate. Pharmacol Ther. 2020; 208: 107476.

[30]

Nicolaes GAF, Soehnlein O. Targeting extranuclear histones to alleviate acute and chronic inflammation. Trends Pharmacol Sci. 2024; 45(7): 651-662.

[31]

Haus M, Foltan M, Philipp A, et al. Neutrophil extracellular traps—a potential trigger for the development of thrombocytopenia during extracorporeal membrane oxygenation. Front Immunol. 2024; 15: 1339235.

[32]

Nicolaes GAF, Soehnlein O. Targeting extranuclear histones to alleviate acute and chronic inflammation. Trends Pharmacol Sci. 2024; 45(7): 651-662.

[33]

Iba T, Ito T, Maruyama I, et al. Potential diagnostic markers for disseminated intravascular coagulation of sepsis. Blood Rev. 2016; 30(2): 149-155.

[34]

Schneck E, Edinger F, Hecker M, et al. Blood levels of free-circulating mitochondrial DNA in septic shock and postsurgical systemic inflammation and its influence on coagulation: a secondary analysis of a prospective observational study. J Clin Med. 2020; 9(7): 1-14.

[35]

Liang H, Song H, Zhang X, et al. Metformin attenuated sepsis-related liver injury by modulating gut microbiota. Emerg Microbes Infect. 2022; 11(1): 815-828.

[36]

Martín-Fernández M, Heredia-Rodríguez M, González-Jiménez I, et al. Hyperoxemia in postsurgical sepsis/septic shock patients is associated with reduced mortality. Crit Care. 2022; 26(1): 1-9.

[37]

Schmidt C, Weißmüller S, Heinz CC. Multifaceted tissue-protective functions of polyvalent immunoglobulin preparations in severe infections—interactions with neutrophils, complement, and coagulation pathways. Biomedicines. 2023; 11(11): 3022.

[38]

Chang Z, Lu J, Zhang Q, et al. Clinical biomarker profiles reveals gender differences and mortality factors in sepsis. Front Immunol. 2024; 15: 1413729.

[39]

Hollenberg SM. Pathophysiology of sepsis-induced cardiomyopathy. Nat Rev Cardiol. 2021; 18(June): 424-434.

[40]

Giamarellos-Bourboulis EJ, Aschenbrenner AC, Bauer M, et al. The pathophysiology of sepsis and precision-medicine-based immunotherapy. Nat Immunol. 2024; 25(1): 19-28.

[41]

Jiang H, Zhang W, Yang J, et al. Miniaturized solid-phase extraction using a mesoporous molecular sieve SBA-15 as sorbent for the determination of triterpenoid saponins from Pulsatilla chinensis by ultrahigh-performance liquid chromatography-charged aerosol detection. J Pharm Biomed Anal. 2021; 194: 11381.

[42]

Shimizu J, Murao A, Lee Y, et al. Extracellular CIRP promotes Kupffer cell inflammatory polarization in sepsis. Front Immunol. 2024; 15: 1411930.

[43]

Zhang W-D, Jin MM, Jiang HH, et al. Study on the metabolites of betulinic acid in vivo and in vitro by ultra high performance liquid chromatography with time-of-flight mass spectrometry. J Sep Sci. 2019; 42(2): 628-635.

[44]

Zhang W, Jiang H, Jin M, et al. UHPLC-Q-TOF-MS/MS based screening and identification of the metabolites in vivo after oral administration of betulin. Fitoterapia. 2018; 127: 29-41.

[45]

Zhang H, Zhou X, McQuade T, et al. Functional complementation between FADD and RIP1 in embryos and lymphocytes. Nature. 2011; 471(7338): 373-377.

[46]

Zhang W, Jiang H, Wu G, et al. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing). 2023; 4(6): e418.

[47]

Zhang W, Jiang H, Yang J, et al. Safety assessment and antioxidant evaluation of betulin by LC-MS combined with free radical assays. Anal Biochem. 2019: 587:113460.

[48]

Gu X, Zhou F, Wang Y, et al. Respiratory viral sepsis: epidemiology, pathophysiology, diagnosis and treatment. Eur Respir Rev. 2020; 29(157): 1-12.

[49]

Cajander S, Kox M, Scicluna BP, et al. Profiling the dysregulated immune response in sepsis: overcoming challenges to achieve the goal of precision medicine. Lancet Respir Med. 2024; 12(4): 305-322.

[50]

Markiewski MM, Nilsson B, Nilsson Ekdahl K, et al. Complement and coagulation: strangers or partners in crime?. Trends Immunol. 2007; 28(4): 184-192.

[51]

Muhlfelder TW, Niemetz J, Kreutzer D, et al. C5 chemotactic fragment induces leukocyte production of tissue factor activity. A link between complement and coagulation. J Clin Invest. 1979; 63(1): 147-150.

[52]

Keshari RS, Silasi R, Popescu NI, et al. Inhibition of complement C5 protects against organ failure and reduces mortality in a baboon model of Escherichia coli sepsis. Proc Natl Acad Sci. 2017; 114(31): E6390-E6399.

[53]

Keshari RS, Popescu NI, Silasi R, et al. Complement C5 inhibition protects against hemolytic anemia and acute kidney injury in anthrax peptidoglycan-induced sepsis in baboons. Proc Natl Acad Sci U S A. 2021; 118(37): e2104347118.

[54]

Iba T, Levy JH. Sepsis-induced coagulopathy and disseminated intravascular coagulation. Anesthesiology. 2020(5): 1238-1245.

[55]

Prowle JR, Bagshaw SM, Forni LG. Tackling sepsis-associated AKI: are there any chances of REVIVAL with new approaches?. Intensive Care Med. 2024; 50(1): 131-133.

[56]

Cai M, Deng J, Wu S, et al. Alpha-1 antitrypsin targeted neutrophil elastase protects against sepsis-induced inflammation and coagulation in mice via inhibiting neutrophil extracellular trap formation. Life Sci. 2024; 353: 122923.

[57]

Fan X, McCullough RL, Huang E, et al. Diagnostic and prognostic significance of complement in patients with alcohol-associated hepatitis. Hepatology. 2021; 73(3): 983-997.

[58]

Macleod OJS, Bart JM, MacGregor P, et al. A receptor for the complement regulator factor H increases transmission of trypanosomes to tsetse flies. Nat Commun. 2020; 11(1): 1-12.

[59]

Muri L, Ispasanie E, Schubart A, et al. Alternative complement pathway inhibition abrogates pneumococcal opsonophagocytosis in vaccine-naïve, but not in vaccinated individuals. Front Immunol. 2021; 12(October): 1-12.

[60]

Lo MW, Kemper C, Woodruff TM. COVID-19: complement, coagulation, and collateral damage. J Immunol. 2020; 205(6): 1488-1495.

[61]

Papareddy P, Selle M, Partouche N, et al. Identifying biomarkers deciphering sepsis from trauma-induced sterile inflammation and trauma-induced sepsis. Front Immunol. 2023: 14:1310271.

[62]

Rittirsch D, Flierl MA, Ward PA. Harmful molecular mechanisms in sepsis. Nat Rev Immunol. 2008; 8(10): 776-787.

[63]

Zhang Y, Wang L, Kuang X, et al. Diagnostic and prognostic value of C1q in sepsis-induced coagulopathy. Clin Appl Thrombos/Hemostas. 2024; 30: 10760296241257517.

[64]

Gurevich VV, Gurevich EV. GPCR-dependent and -independent arrestin signaling. Trends Pharmacol Sci. 2019; 9(4): 217-230.

[65]

Bekassy Z, Lopatko Fagerström I, Bader M, et al. Crosstalk between the renin–angiotensin, complement and kallikrein–kinin systems in inflammation. Nat Rev Immunol. 2022; 22(7): 411-428.

[66]

Lupu F, Keshari RS, Lambris JD, et al. Crosstalk between the coagulation and complement systems in sepsis. Thromb Res. 2014; 133(suppl 1): S28-S31.

[67]

Perico L, Benigni A, Casiraghi F, et al. Immunity, endothelial injury and complement-induced coagulopathy in COVID-19. Nat Rev Nephrol. 2021; 17(1): 46-64.

[68]

Pryzdial ELG, Leatherdale A, Conway EM. Coagulation and complement: key innate defense participants in a seamless web. Front Immunol. 2022; 13: 918775.

[69]

Wang Y, Liu W, Xu Y, et al. Revealing the signaling of complement receptors C3aR and C5aR1 by anaphylatoxins. Nat Chem Biol. 2023; 19(11): 1351-1360.

[70]

Sprong T, Roos D, Weemaes C, et al. Deficient alternative complement pathway activation due to factor D deficiency by 2 novel mutations in the complement factor D gene in a family with meningococcal infections. Blood. 2006; 107(12): 4865-4870.

[71]

Park J, Lee SB, Lee S, et al. Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin. Nature. 2006; 441(7097): 1157-1161.

[72]

Rossi G, Meningococcaemia, complement system, and factor V Leiden. Lancet. 2004; 363(9415): 1166.

[73]

Peng M, Li Z, Niu D, et al. Complement factor B/C2 in molluscs regulates agglutination and illuminates evolution of the Bf/C2 family. FASEB J. 2019; 33(12): 13323-13333.

[74]

Maekawa T, Krauss JL, Abe T, et al. Porphyromonas gingivalis manipulates complement and TLR signaling to uncouple bacterial clearance from inflammation and promote dysbiosis. Cell Host Microbe. 2014; 15(6): 768-778.

[75]

Nahm MH, Yu J, Calix JJ, et al. Ficolin-2 lectin complement pathway mediates capsule-specific innate immunity against invasive pneumococcal disease. Front Immunol. 2022; 13(March): 1-10.

[76]

Ren J, Zhao Y, Yuan Y, et al. Complement depletion deteriorates clinical outcomes of severe abdominal sepsis: a conspirator of infection and coagulopathy in crime?. PLoS ONE. 2012; 7(10): 1-9.

[77]

Gilmore AC, Zhang Y, Cook HT, et al. Complement activity is regulated in C3 glomerulopathy by IgG-factor H fusion proteins with and without properdin targeting domains. Kidney Int. 2021; 99(2): 396-404.

[78]

Huber M, Fischer M, Misselwitz B, et al. Complement lysis activity in autologous plasma is associated with lower viral loads during the acute phase of HIV-1 infection. PLoS Med. 2006; 3(11): 2078-2093.

[79]

Molema G, Zijlstra JG, van Meurs M, et al. Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat Rev Nephrol. 2022; 18(2): 95-112.

[80]

Chousterman BG, Swirski FK, Weber GF. Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol. 2017; 39(5): 517-528.

[81]

Pawlinski R, Wang JG, Owens AP, et al. Hematopoietic and nonhematopoietic cell tissue factor activates the coagulation cascade in endotoxemic mice. Blood. 2010; 116(5): 806-814.

[82]

Iskander KN, Osuchowski MF, Stearns-Kurosawa DJ, et al. Sepsis: multiple abnormalities, heteroge-neous responses, and evolving understanding. Physiol Rev. 2013; 93: 1247-1288.

[83]

Yang X, Cheng X, Tang Y, et al. The role of type 1 interferons in coagulation induced by gram-negative bacteria. Blood. 2020; 135(14): 1087-1100.

[84]

Xing K, Murthy S, Liles WC, et al. Clinical utility of biomarkers of endothelial activation in sepsis–a systematic review. Crit Care. 2012; 16(1): R7.

[85]

Blasco RB, Patrucco E, Mota I, et al. Comment on “ALK” is a therapeutic target for lethal sepsis. Sci Transl Med. 2018; 10(471): 10-13.

[86]

Huang L, Chen J, Li X, et al. Polydatin improves sepsis-associated encephalopathy by activating Sirt1 and reducing p38 phosphorylation. J Surg Res. 2022; 276: 379-393.

[87]

Schouten M, Wiersinga WJ, Levi M, et al. Inflammation, endothelium, and coagulation in sepsis. J Leukoc Biol. 2008; 83(3): 536-545.

[88]

Chung HY, Wickel J, Hahn N, et al. Microglia mediate neurocognitive deficits by eliminating C1q-tagged synapses in sepsis-associated encephalopathy. Sci Adv. 2023; 9(21): eabq7806. https://www.science.org

[89]

Patil NK, Bohannon JK, Sherwood ER. Immunotherapy: a promising approach to reverse sepsis-induced immunosuppression. Pharmacol Res. 2016; 111: 688-702.

[90]

Langley RJ, Tsalik EL, Van Velkinburgh JC, et al. Sepsis: an integrated clinico-metabolomic model improves prediction of death in sepsis. Sci Transl Med. 2013; 5(195): 195ra95.

[91]

McDonald B, Davis RP, Kim SJ, et al. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood. 2017; 129(10): 1357-1367.

[92]

Luo L, Wu J, Qiao L, et al. Sestrin 2 attenuates sepsis-associated encephalopathy through the promotion of autophagy in hippocampal neurons. J Cell Mol Med. 2020; 24(12): 6634-6643.

[93]

Wu R, Wang N, Comish PB, et al. Inflammasome-dependent coagulation activation in sepsis. Front Immunol. 2021; 12(March): 1-8.

[94]

De Castro REV, Medeiros DNM, Prata-Barbosa A, et al. Surviving sepsis campaign international guidelines for the management of septic shock and sepsis-associated organ dysfunction in children. Pediatr Crit Care Med. 2020; 46: 924-925.

[95]

Thomas H. Sepsis: bile acids promote inflammation in cholestasis-associated sepsis. Nat Rev Gastroenterol Hepatol. 2017; 14(6): 324-325.

[96]

Ren C, Zhang H, Wu TT, et al. Autophagy: a potential therapeutic target for reversing sepsis-induced immunosuppression. Front Immunol. 2017; 8(Dec): 1-10.

[97]

Sureshbabu A, Patino E, Ma KC, et al. RIPK3 promotes sepsis-induced acute kidney injury via mitochondrial dysfunction. JCI Insight. 2018; 3(11): 1-17.

[98]

Asehnoune K, Moine P. Protease-activated receptor-1: key player in the sepsis coagulation-inflammation crosstalk. Crit Care. 2013; 17(1): 1-3.

[99]

McBride MA, Patil TK, Bohannon JK, et al. Immune checkpoints: novel therapeutic targets to attenuate sepsis-induced immunosuppression. Front Immunol. 2021; 11(February): 1-14.

[100]

Vincent JL, Francois B, Zabolotskikh I, et al. Effect of a recombinant human soluble thrombomodulin on mortality in patients with sepsis-associated coagulopathy: the SCARLET randomized clinical trial. JAMA. 2019; 321: 1993-2002.

[101]

Danese S, Vetrano S, Zhang L, et al. The protein C pathway in tissue inflammation and injury: pathogenic role and therapeutic implications the PC pathway: its anticoagulant and cytoprotective functions. Blood. 2010; 115: 1121-1130.

[102]

Wu R, Wang N, Comish PB, et al. Inflammasome-dependent coagulation activation in sepsis. Front Immunol. 2021: 12:641750.

[103]

Xue M, Sun Z, Shao M, et al. Diagnostic and prognostic utility of tissue factor for severe sepsis and sepsis-induced acute lung injury. J Transl Med. 2015; 13(1): 1-8.

[104]

Singer M. The role of mitochondrial dysfunction in sepsis-induced multi-organ failure. Virulence. 2014; 5(1): 66-72.

[105]

Souza ACP, Yuen PST, Star RA. Microparticles: markers and mediators of sepsis-induced microvascular dysfunction, immunosuppression, and AKI. Kidney Int. 2015; 87(6): 1100-1108.

[106]

Ho J, Yu J, Wong SH, et al. Autophagy in sepsis: degradation into exhaustion. Autophagy. 2016; 12(7): 1073-1082.

[107]

Liang X, Li T, Zhou Q, et al. Mesenchymal stem cells attenuate sepsis-induced liver injury via inhibiting M1 polarization of Kupffer cells. Mol Cell Biochem. 2019; 452(1–2): 187-197.

[108]

Gong S, Yan Z, Liu Z, et al. Intestinal microbiota mediates the susceptibility to polymicrobial sepsis-induced liver injury by granisetron generation in mice. Hepatology. 2019; 69(4): 1751-1767.

[109]

Zhang Z, Han N, Shen Y. S100A12 promotes inflammation and cell apoptosis in sepsis-induced ARDS via activation of NLRP3 inflammasome signaling. Mol Immunol. 2020; 122(March): 38-48.

[110]

Zhu Z, Chambers S, Zeng Y, et al. Gases in sepsis: novel mediators and therapeutic targets. Int J Mol Sci. 2022; 23(7): 3669.

[111]

Ma T, Han L, Gao Y, et al. The endoplasmic reticulum stress-mediated apoptosis signal pathway is involved in sepsis-induced abnormal lymphocyte apoptosis. Eur Surg Res. 2008; 41(2): 219-225.

[112]

Xia Y, Cao Y, Sun Y, et al. Calycosin alleviates sepsis-induced acute lung injury via the inhibition of mitochondrial ROS-mediated inflammasome activation. Front Pharmacol. 2021; 12(October): 1-14.

[113]

Ayala A, Elphick GF, Kim YS, et al. Sepsis-induced potentiation of peritoneal macrophage migration is mitigated by programmed cell death receptor-1 gene deficiency. J Innate Immun. 2014; 6(3): 325-338.

[114]

Dear JW, Leelahavanichkul A, Aponte A, et al. Liver proteomics for therapeutic drug discovery: inhibition of the cyclophilin receptor CD147 attenuates sepsis-induced acute renal failure. Crit Care Med. 2007; 35(10): 2319-2328.

[115]

Vignon P, Laterre PF, Daix T, et al. New agents in development for sepsis: any reason for hope. Drugs. 2020; 80(17): 1751-1761.

[116]

Takayama W, Endo A, Morishita K, et al. Dielectric blood coagulometry for the early detection of sepsis-induced disseminated intravascular coagulation: a prospective observational study. Crit Care Med. 2022; 50(1): E31-E39.

[117]

Zhang H, Zeng L, Xie M, et al. TMEM173 drives lethal coagulation in sepsis. Cell Host Microbe. 2020; 27(4): 556-570.

[118]

Cornelius DC, Travis OK, Tramel RW, et al. NLRP3 inflammasome inhibition attenuates sepsis-induced platelet activation and prevents multi-organ injury in cecal-ligation puncture. PLoS ONE. 2020; 15: 1-15.

[119]

Watanabe E, Nishida O, Kakihana Y, et al. Pharmacokinetics, pharmacodynamics, and safety of nivolumab in patients with sepsis-induced immunosuppression: a multicenter, open-label phase 1/2 study. Shock. 2020; 53(6): 686-694.

[120]

Schulman S, El-Darzi E, Florido MHC, et al. A coagulation defect arising from heterozygous premature termination of tissue factor. J Clin Invest. 2020; 130(10): 5302-5312.

[121]

Barichello T, Generoso JS, Singer M, et al. Biomarkers for sepsis: more than just fever and leukocytosis—a narrative review. Crit Care. 2022; 26: 14.

[122]

Li R, Hu X, Chen H, et al. Role of cholinergic anti-inflammatory pathway in protecting sepsis-induced acute lung injury through regulation of the conventional dendritic cells. Mediators Inflamm. 2022; 2022: 1474891.

[123]

Yang X, Cheng X, Tang Y, et al. Bacterial endotoxin activates the coagulation cascade through gasdermin D-dependent phosphatidylserine exposure. Immunity. 2019; 51(6): 983-996.

[124]

Jiang J, Huang K, Xu S, et al. Targeting NOX4 alleviates sepsis-induced acute lung injury via attenuation of redox-sensitive activation of CaMKII/ERK1/2/MLCK and endothelial cell barrier dysfunction. Redox Biol. 2020; 36: 101638.

[125]

Wang Y, Xu M, Yue P, et al. Novel insights into the potential mechanisms of N6-methyladenosine RNA modification on sepsis-induced cardiovascular dysfunction: an update summary on direct and indirect evidences. Front Cell Dev Biol. 2021; 9(November): 1-13.

[126]

Herrmann JB, Muenstermann M, Strobel L, et al. Complement C5a receptor 1 exacerbates the pathophysiology of N. meningitides sepsis and is a potential target for disease treatment. mBio. 2018; 9(1): e01755-17.

[127]

Gustavsen A, Nymo S, Landsem A, et al. Combined inhibition of complement and CD14 attenuates bacteria-induced inflammation in human whole blood more efficiently than antagonizing the toll-like receptor 4-MD2 complex. J Infect Dis. 2016; 214(1): 140-150.

[128]

Wu C, Lu W, Zhang Y, et al. Inflammasome activation triggers blood clotting and host death through pyroptosis. Immunity. 2019; 50(6): 1401-1411.

[129]

Zhang Y, Cui J, Zhang G, et al. Inflammasome activation promotes venous thrombosis through pyroptosis. Blood Adv. 2021; 5(12): 2619-2623.

[130]

Nielsen TB, Pantapalangkoor P, Yan J, et al. Diabetes exacerbates infection via hyperinflammation by signaling through TLR4 and RAGE. mBio. 2017; 8(4).

[131]

Samanta K, Setua S, Kumari S, et al. Gemcitabine combination nano therapies for pancreatic cancer. Pharmaceutics. 2019; 11(11): 1-25.

[132]

Garrido N, Griparic L, Jokitalo E, et al. Composition and dynamics of human mitochondrial nucleoids. Mol Biol Cell. 2003; 14(4): 1583-1596.

[133]

Karmakar M, Minns M, Greenberg EN, et al. N-GSDMD trafficking to neutrophil organelles facilitates IL-1β release independently of plasma membrane pores and pyroptosis. Nat Commun. 2020; 11(1): 1-14.

[134]

Afzali B, Noris M, Lambrecht BN, et al. The state of complement in COVID-19. Nat Rev Immunol. 2022; 22(2): 77-84.

[135]

Hu Q, Knight PH, Ren Y, et al. The emerging role of stimulator of interferons genes signaling in sepsis: inflammation. Autophagy Cell Death. 2019; 225(3): e13194.

[136]

Ward PA. The dark side of C5a in sepsis. Nat Rev Immunol. 2004; 4(2): 133-142.

[137]

Nakashima T, Miyamoto K, Shima N, et al. Dexmedetomidine improved renal function in patients with severe sepsis: an exploratory analysis of a randomized controlled trial. J Intensive Care. 2020; 8: 1.

[138]

Jackson WD, Gulino A, Fossati-Jimack L, et al. C3 drives inflammatory skin carcinogenesis independently of C5. J Invest Dermatol. 2021; 141(2): 404-414.

[139]

Vandewalle J, Vanderhaeghen T, Beyaert R, et al. Taking the STING out of sepsis. Cell Host Microbe. 2020; 27(4): 491-493.

[140]

Hoehlig K, Maasch C, Shushakova N, et al. A novel C5a-neutralizing mirror-image (l-)aptamer prevents organ failure and improves survival in experimental sepsis. Mol Ther. 2013; 21(12): 2236-2246.

[141]

Sprong T, Brandtzaeg P, Fung M, et al. Inhibition of C5a-induced inflammation with preserved C5b-9-mediated bactericidal activity in a human whole blood model of meningococcal sepsis. Blood. 2003; 102(10): 3702-3710.

[142]

Shao Z, Nishimura T, Leung LLK, et al. Carboxypeptidase B2 deficiency reveals opposite effects of complement C3a and C5a in a murine polymicrobial sepsis model. J Thromb Haemost. 2015; 13(6): 1090-1102.

[143]

Deng C, Zhao L, Yang Z, et al. Targeting HMGB1 for the treatment of sepsis and sepsis-induced organ injury. Acta Pharmacol Sin. 2022; 43(3): 520-528.

[144]

Venet F, Monneret G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol. 2018; 14(2): 121-137.

[145]

de Stoppelaar SF, van’t Veer C, van der Poll T. The role of platelets in sepsis. Thromb Haemost. 2014; 112(4): 666-677.

[146]

Lood C, Blanco LP, Purmalek MM, et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med. 2016; 22(2): 146-153.

[147]

Van Der Poll T, Van De Veerdonk FL, Scicluna BP, et al. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017; 17(7): 407-420.

[148]

Middleton EA, Rowley JW, Campbell RA, et al. Sepsis Alters the Transcriptional and Translational Landscape of Human and Murine Platelets. Blood. 2019; 8.

[149]

Tang D, Wang H, Billiar TR, et al. Emerging mechanisms of immunocoagulation in sepsis and septic shock. Trends Immunol. 2021; 42(6): 508-522.

[150]

Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015; 526(7575): 660-665.

[151]

Kayagaki N, Stowe IB, Lee BL, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015; 526(7575): 666-671.

[152]

Bergmann CB, Beckmann N, Salyer CE, et al. Potential targets to mitigate trauma-or sepsis-induced immune suppression. Front Immunol. 2021; 12(February): 1-19.

[153]

van der Meijden PEJ, Heemskerk JWM. Platelet biology and functions: new concepts and clinical perspectives. Nat Rev Cardiol. 2019; 16(3): 166-179.

[154]

Morrell CN, Aggrey AA, Chapman LM, et al. Emerging roles for platelets as immune and inflammatory cells. Blood. 2014; 123(18): 2759-2767.

[155]

Yang M, Jiang H, Ding C, et al. STING activation in platelets aggravates septic thrombosis by enhancing platelet activation and granule secretion. Immunity. 2023; 56(5): 1013-1026.

[156]

Fuchs TA, Brill A, Duerschmied D, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A. 2010; 107(36): 15880-15885.

[157]

Su M, Chen C, Li S, et al. Gasdermin D-dependent platelet pyroptosis exacerbates NET formation and inflammation in severe sepsis. Nat Cardiovasc Res. 2022; 1(8): 732-747.

[158]

Ngo ATP, Skidmore A, Oberg J, et al. Platelet factor 4 limits neutrophil extracellular trap-and cell-free DNA-induced thrombogenicity and endothelial injury. JCI Insight. 2023; 8(22): e171054.

[159]

Rathkey JK, Xiao TS, Abbott DW. Human polymorphisms in GSDMD alter the inflammatory response. J Biol Chem. 2020; 295(10): 3228-3238.

[160]

Ding X, Kambara H, Guo R, et al. Inflammasome-mediated GSDMD activation facilitates escape of Candida albicans from macrophages. Nat Commun. 2021; 12(1): 6699.

[161]

Ousingsawat J, Wanitchakool P, Schreiber R, et al. Contribution of TMEM16F to pyroptotic cell death. Cell Death Dis. 2018; 9(3): 625.

[162]

Wang G, Jin S, Huang W, et al. LPS-induced macrophage HMGB1-loaded extracellular vesicles trigger hepatocyte pyroptosis by activating the NLRP3 inflammasome. Cell Death Discov. 2021; 7(1): 1-11.

[163]

Deng M, Tang Y, Li W, et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity. 2018; 49(4): 740-753.

[164]

Kang R, Chen R, Zhang Q, et al. HMGB1 in health and disease. Mol Aspects Med. 2014; 40(July): 1-116.

[165]

Volchuk A, Ye A, Chi L, et al. Indirect regulation of HMGB1 release by gasdermin D. Nat Commun. 2020; 11(1): 1-11.

[166]

Zhao F, Fang Y, Deng S, et al. Glycyrrhizin protects rats from sepsis by blocking HMGB1 signaling. Biomed Res Int. 2017; 2017: 9719647.

[167]

Zhu X, Messer JS, Wang Y, et al. Cytosolic HMGB1 controls the cellular autophagy/apoptosis checkpoint during inflammation. J Clin Invest. 2015; 125(3): 1098-1110.

[168]

Kim HM, Kim YM. HMGB1: LPS delivery vehicle for caspase-11-mediated pyroptosis. Immunity. 2018; 49(4): 582-584.

[169]

Kang R, Livesey KM, Zeh HJ, et al. HMGB1: a novel Beclin 1-binding protein active in autophagy. Autophagy. 2010; 6(8): 1209-1211.

[170]

Tang D, Kang R, Livesey KM, et al. Endogenous HMGB1 regulates autophagy. J Cell Biol. 2010; 190(5): 881-892.

[171]

Liu L, Yang M, Kang R, et al. HMGB1-DNA complex-induced autophagy limits AIM2 inflammasome activation through RAGE. Biochem Biophys Res Commun. 2014; 450(1): 851-856.

[172]

Sun Z, Nyanzu M, Yang S, et al. VX765 attenuates pyroptosis and HMGB1/TLR4/NF-κB pathways to improve functional outcomes in TBI mice. Oxid Med Cell Longev. 2020: 2020:7879629.

[173]

Wang Q, Wu X, Tong X, et al. Xuebijing ameliorates sepsis-induced lung injury by downregulating HMGB1 and RAGE expressions in mice. Evid-Based Complement Altern Med. 2015; 2015: 860259.

[174]

Li Z, Fu WJ, Chen XQ, et al. Autophagy-based unconventional secretion of HMGB1 in glioblastoma promotes chemosensitivity to temozolomide through macrophage M1-like polarization. J Exp Clin Cancer Res. 2022; 41(1): 1-20.

[175]

Chen R, Huang Y, Quan J, et al. HMGB1 as a potential biomarker and therapeutic target for severe COVID-19. Heliyon. 2020; 6(12): e05672.

[176]

Steckert AV, Comim CM, Mina F, et al. Late brain alterations in sepsis-survivor rats. Synapse. 2013; 67(11): 786-793.

[177]

Schneidewind L. Reducing mortality in adults with sepsis, severe sepsis, or septic shock: effectiveness and safety of procalcitonin. Urologe. 2021; 60(5): 624-627.

[178]

Jiang S, qiang WangY, Tang Y, et al. Environmental enrichment protects against sepsis-associated encephalopathy-induced learning and memory deficits by enhancing the synthesis and release of vasopressin in the supraoptic nucleus. J Inflamm Res. 2022; 15(January): 363-379.

[179]

Haag SM, Gulen MF, Reymond L, et al. Targeting STING with covalent small-molecule inhibitors. Nature. 2018; 559(7713): 269-273.

[180]

Liu Y, Jesus AA, Marrero B, et al. Activated STING in a vascular and pulmonary syndrome. New Engl J Med. 2014; 371(6): 507-518.

[181]

Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008; 455(7213): 674-678.

[182]

Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015; 15(12): 760-770.

[183]

Ge W, Hu Q, Fang X, et al. LDK378 improves micro-and macro-circulation via alleviating STING-mediated inflammatory injury in a sepsis rat model induced by cecal ligation and puncture. J Inflamm (United Kingdom). 2019; 16(1): 1-10.

[184]

Liu N, Pang X, Zhang H, et al. The cGAS-STING pathway in bacterial infection and bacterial immunity. Front Immunol. 2022; 12(January): 1-18.

[185]

Long J, Yang C, Zheng Y, et al. Notch signaling protects CD4 T cells from STING-mediated apoptosis during acute systemic inflammation. Sci Adv. 2020; 6(39): 1-13.

[186]

Gaidt MM, Ebert TS, Chauhan D, et al. The DNA inflammasome in human myeloid cells is initiated by a STING-cell death program upstream of NLRP3. Cell. 2017; 171(5): 1110-1124.

[187]

Wu J, Chen YJ, Dobbs N, et al. STING-mediated disruption of calcium homeostasis chronically activates ER stress and primes T cell death. J Exp Med. 2019; 216(4): 867-883.

[188]

Cheng Y, Marion TN, Cao X, et al. A novel therapeutic target for macrophages in sepsis-induced immunosuppression. Front Immunol. 2018; 7(Nov): 1-8.

[189]

Gotts JE, Matthay MA. Sepsis: pathophysiology and clinical management. BMJ (Online). 2016; 353: i1585.

[190]

Heipertz EL, Harper J, Walker WE. STING and TRIF contribute to mouse sepsis, depending on severity of the disease model. Shock. 2017; 47(5): 621-631.

[191]

Prabakaran T, Bodda C, Krapp C, et al. Attenuation of c GAS—STING signaling is mediated by a p62/SQSTM 1-dependent autophagy pathway activated by TBK1. EMBO J. 2018; 37(8): e97858.

[192]

Rello J, Valenzuela-Sánchez F, Ruiz-Rodriguez M, et al. Sepsis: a review of advances in management. Adv Ther. 2017; 34(11): 2393-2411.

[193]

Silasi-Mansat R, Zhu H, Popescu NI, et al. Complement inhibition decreases the procoagulant response and confers organ protection in a baboon model of Escherichia coli sepsis. Blood. 2010; 116(6): 1002-1010.

[194]

Zhao L, Ohtaki Y, Yamaguchi K, et al. LPS-induced platelet response and rapid shock in mice: contribution of O-antigen region of LPS and involvement of the lectin pathway of the complement system. Blood. 2002; 100(9): 3233-3239.

[195]

Yang J, Liu Z, Wang C, et al. Mechanism of gasdermin D recognition by inflammatory caspases and their inhibition by a gasdermin D-derived peptide inhibitor. Proc Natl Acad Sci U S A. 2018; 115(26): 6792-6797.

[196]

Kang R, Zeng L, Zhu S, et al. Lipid peroxidation drives gasdermin D-mediated pyroptosis in lethal polymicrobial sepsis. Cell Host Microbe. 2018; 24(1): 97-108.

[197]

Zhou B, Liu J, Zeng L, et al. Extracellular SQSTM1 mediates bacterial septic death in mice through insulin receptor signalling. Nat Microbiol. 2020; 5(12): 1576-1587.

[198]

Jiang Y, He L, Green J, et al. Discovery of second-generation NLRP3 inflammasome inhibitors: design, synthesis, and biological characterization. J Med Chem. 2019; 62(21): 9718-9731.

[199]

Chen R, Zhu S, Zeng L, et al. AGER-mediated lipid peroxidation drives caspase-11 inflammasome activation in sepsis. Front Immunol. 2019; 10(Aug): 1904.

[200]

Hu JJ, Liu X, Xia S, et al. FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation. Nat Immunol. 2020; 21(7): 736-745.

[201]

Lu Y, Meng R, Wang X, et al. Caspase-11 signaling enhances graft-versus-host disease. Nat Commun. 2019; 10(1): 4044.

[202]

Yang X, Cheng X, Tang Y, et al. The Role of Type 1 Interferons in Coagulation Induced by Gram-Negative Bacteria. Blood. 2020; 135(14): 1084-1100.

[203]

Tang Y, Wang X, Li Z, et al. Heparin prevents caspase-11-dependent septic lethality independent of anticoagulant properties. Immunity. 2021; 54(3): 454-467.

[204]

Annane D, Buisson CB, Cariou A, et al. Design and conduct of the activated protein C and corticosteroids for human septic shock (APROCCHSS) trial. Ann Intensive Care. 2016; 6(1): 9.

[205]

Annane D, Renault A, Brun-Buisson C, et al. Hydrocortisone plus fludrocortisone for adults with septic shock. New Engl J Med. 2018; 378(9): 809-818.

[206]

Stern RM, Connell NT. Ravulizumab: a novel C5 inhibitor for the treatment of paroxysmal nocturnal hemoglobinuria. Ther Adv Hematol. 2019; 10: 204062071987472.

[207]

Hoy SM. Pegcetacoplan: first approval. Drugs. 2021; 81(12): 1423-1430.

[208]

Jayne DRW, Merkel PA, Schall TJ, et al. Avacopan for the treatment of ANCA-associated vasculitis. New Engl J Med. 2021; 384(7): 599-609.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

152

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/