Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2022

Open Access 01-12-2022 | Septicemia | Review

Sepsis-associated brain injury: underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments

Authors: Nobufumi Sekino, Magdy Selim, Amjad Shehadah

Published in: Journal of Neuroinflammation | Issue 1/2022

Login to get access

Abstract

Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis causes cerebral dysfunction in the short and long term and induces disruption of the blood–brain barrier (BBB), neuroinflammation, hypoperfusion, and accumulation of amyloid β (Aβ) and tau protein in the brain. White matter changes and brain atrophy can be detected using brain imaging, but unfortunately, there is no specific treatment that directly addresses the underlying mechanisms of cognitive impairments in sepsis. Here, we review the underlying mechanisms of sepsis-associated brain injury, with a focus on BBB dysfunction and Aβ and tau protein accumulation in the brain. We also describe the neurological manifestations and imaging findings of sepsis-associated brain injury, and finally, we propose potential therapeutic strategies for acute and long-term cognitive impairments associated with sepsis. In the acute phase of sepsis, we suggest using antibiotics (such as rifampicin), targeting proinflammatory cytokines, and preventing ischemic injuries and hypoperfusion. In the late phase of sepsis, we suggest targeting neuroinflammation, BBB dysfunction, Aβ and tau protein phosphorylation, glycogen synthase kinase-3 beta (GSK3β), and the receptor for advanced glycation end products (RAGE). These proposed strategies are meant to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating acute and long-term cognitive impairments in patients with sepsis.
Literature
1.
go back to reference Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801–10.CrossRefPubMedPubMedCentral Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801–10.CrossRefPubMedPubMedCentral
2.
go back to reference Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the Global Burden of Disease Study. Lancet. 2020;395(10219):200–11.PubMedPubMedCentralCrossRef Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the Global Burden of Disease Study. Lancet. 2020;395(10219):200–11.PubMedPubMedCentralCrossRef
4.
go back to reference Barichello T, Sayana P, Giridharan VV, Arumanayagam AS, Narendran B, Della Giustina A, Petronilho F, Quevedo J, Dal-Pizzol F. Long-term cognitive outcomes after sepsis: a translational systematic review. Mol Neurobiol. 2019;56(1):186–251.PubMedCrossRef Barichello T, Sayana P, Giridharan VV, Arumanayagam AS, Narendran B, Della Giustina A, Petronilho F, Quevedo J, Dal-Pizzol F. Long-term cognitive outcomes after sepsis: a translational systematic review. Mol Neurobiol. 2019;56(1):186–251.PubMedCrossRef
5.
go back to reference Nwafor DC, Brichacek AL, Mohammad AS, Griffith J, Lucke-Wold BP, Benkovic SA, Geldenhuys WJ, Lockman PR, Brown CM. Targeting the blood-brain barrier to prevent sepsis-associated cognitive impairment. J Cent Nerv Syst Dis. 2019;11:1179573519840652.PubMedPubMedCentralCrossRef Nwafor DC, Brichacek AL, Mohammad AS, Griffith J, Lucke-Wold BP, Benkovic SA, Geldenhuys WJ, Lockman PR, Brown CM. Targeting the blood-brain barrier to prevent sepsis-associated cognitive impairment. J Cent Nerv Syst Dis. 2019;11:1179573519840652.PubMedPubMedCentralCrossRef
6.
go back to reference Pandharipande PP, Girard TD, Jackson JC, Morandi A, Thompson JL, Pun BT, Brummel NE, Hughes CG, Vasilevskis EE, Shintani AK, et al. Long-term cognitive impairment after critical illness. N Engl J Med. 2013;369(14):1306–16.PubMedPubMedCentralCrossRef Pandharipande PP, Girard TD, Jackson JC, Morandi A, Thompson JL, Pun BT, Brummel NE, Hughes CG, Vasilevskis EE, Shintani AK, et al. Long-term cognitive impairment after critical illness. N Engl J Med. 2013;369(14):1306–16.PubMedPubMedCentralCrossRef
7.
go back to reference Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, Kumar A, Sevransky JE, Sprung CL, Nunnally ME, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Crit Care Med. 2017;45(3):486–552.PubMedCrossRef Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, Kumar A, Sevransky JE, Sprung CL, Nunnally ME, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Crit Care Med. 2017;45(3):486–552.PubMedCrossRef
8.
go back to reference Ruiz S, Vardon-Bounes F, Merlet-Dupuy V, Conil JM, Buleon M, Fourcade O, Tack I, Minville V. Sepsis modeling in mice: ligation length is a major severity factor in cecal ligation and puncture. Intensive Care Med Exp. 2016;4(1):22.PubMedPubMedCentralCrossRef Ruiz S, Vardon-Bounes F, Merlet-Dupuy V, Conil JM, Buleon M, Fourcade O, Tack I, Minville V. Sepsis modeling in mice: ligation length is a major severity factor in cecal ligation and puncture. Intensive Care Med Exp. 2016;4(1):22.PubMedPubMedCentralCrossRef
9.
go back to reference Buras JA, Holzmann B, Sitkovsky M. Animal models of sepsis: setting the stage. Nat Rev Drug Discov. 2005;4(10):854–65.PubMedCrossRef Buras JA, Holzmann B, Sitkovsky M. Animal models of sepsis: setting the stage. Nat Rev Drug Discov. 2005;4(10):854–65.PubMedCrossRef
11.
go back to reference Gabarin RS, Li M, Zimmel PA, Marshall JC, Li Y, Zhang H. Intracellular and extracellular lipopolysaccharide signaling in sepsis: avenues for novel therapeutic strategies. J Innate Immun. 2021;13(6):323–32.PubMedPubMedCentralCrossRef Gabarin RS, Li M, Zimmel PA, Marshall JC, Li Y, Zhang H. Intracellular and extracellular lipopolysaccharide signaling in sepsis: avenues for novel therapeutic strategies. J Innate Immun. 2021;13(6):323–32.PubMedPubMedCentralCrossRef
12.
go back to reference Remick DG, Newcomb DE, Bolgos GL, Call DR. Comparison of the mortality and inflammatory response of two models of sepsis: lipopolysaccharide vs. cecal ligation and puncture. Shock. 2000;13(2):110–6.PubMedCrossRef Remick DG, Newcomb DE, Bolgos GL, Call DR. Comparison of the mortality and inflammatory response of two models of sepsis: lipopolysaccharide vs. cecal ligation and puncture. Shock. 2000;13(2):110–6.PubMedCrossRef
13.
go back to reference Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11(5):373–84.PubMedCrossRef Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11(5):373–84.PubMedCrossRef
14.
go back to reference Raymond SL, Holden DC, Mira JC, Stortz JA, Loftus TJ, Mohr AM, Moldawer LL, Moore FA, Larson SD, Efron PA. Microbial recognition and danger signals in sepsis and trauma. Biochim Biophys Acta Mol Basis Dis. 2017;1863(10 Pt B):2564–73.PubMedCrossRef Raymond SL, Holden DC, Mira JC, Stortz JA, Loftus TJ, Mohr AM, Moldawer LL, Moore FA, Larson SD, Efron PA. Microbial recognition and danger signals in sepsis and trauma. Biochim Biophys Acta Mol Basis Dis. 2017;1863(10 Pt B):2564–73.PubMedCrossRef
15.
16.
go back to reference Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999;18(49):6853–66.PubMedCrossRef Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999;18(49):6853–66.PubMedCrossRef
18.
go back to reference Maciel M, Benedet SR, Lunardelli EB, Delziovo H, Domingues RL, Vuolo F, Tomasi CD, Walz R, Ritter C, Dal-Pizzol F. Predicting long-term cognitive dysfunction in survivors of critical illness with plasma inflammatory markers: a retrospective cohort study. Mol Neurobiol. 2019;56(1):763–7.PubMedCrossRef Maciel M, Benedet SR, Lunardelli EB, Delziovo H, Domingues RL, Vuolo F, Tomasi CD, Walz R, Ritter C, Dal-Pizzol F. Predicting long-term cognitive dysfunction in survivors of critical illness with plasma inflammatory markers: a retrospective cohort study. Mol Neurobiol. 2019;56(1):763–7.PubMedCrossRef
19.
go back to reference Laflamme N, Echchannaoui H, Landmann R, Rivest S. Cooperation between toll-like receptor 2 and 4 in the brain of mice challenged with cell wall components derived from gram-negative and gram-positive bacteria. Eur J Immunol. 2003;33(4):1127–38.PubMedCrossRef Laflamme N, Echchannaoui H, Landmann R, Rivest S. Cooperation between toll-like receptor 2 and 4 in the brain of mice challenged with cell wall components derived from gram-negative and gram-positive bacteria. Eur J Immunol. 2003;33(4):1127–38.PubMedCrossRef
20.
go back to reference Ericsson A, Liu C, Hart RP, Sawchenko PE. Type 1 interleukin-1 receptor in the rat brain: distribution, regulation, and relationship to sites of IL-1-induced cellular activation. J Comp Neurol. 1995;361(4):681–98.PubMedCrossRef Ericsson A, Liu C, Hart RP, Sawchenko PE. Type 1 interleukin-1 receptor in the rat brain: distribution, regulation, and relationship to sites of IL-1-induced cellular activation. J Comp Neurol. 1995;361(4):681–98.PubMedCrossRef
21.
go back to reference Vallieres L, Rivest S. Regulation of the genes encoding interleukin-6, its receptor, and gp130 in the rat brain in response to the immune activator lipopolysaccharide and the proinflammatory cytokine interleukin-1beta. J Neurochem. 1997;69(4):1668–83.PubMedCrossRef Vallieres L, Rivest S. Regulation of the genes encoding interleukin-6, its receptor, and gp130 in the rat brain in response to the immune activator lipopolysaccharide and the proinflammatory cytokine interleukin-1beta. J Neurochem. 1997;69(4):1668–83.PubMedCrossRef
22.
go back to reference Nadeau S, Rivest S. Regulation of the gene encoding tumor necrosis factor alpha (TNF-alpha) in the rat brain and pituitary in response in different models of systemic immune challenge. J Neuropathol Exp Neurol. 1999;58(1):61–77.PubMedCrossRef Nadeau S, Rivest S. Regulation of the gene encoding tumor necrosis factor alpha (TNF-alpha) in the rat brain and pituitary in response in different models of systemic immune challenge. J Neuropathol Exp Neurol. 1999;58(1):61–77.PubMedCrossRef
23.
go back to reference Skelly DT, Hennessy E, Dansereau MA, Cunningham C. A systematic analysis of the peripheral and CNS effects of systemic LPS, IL-1beta, [corrected] TNF-alpha and IL-6 challenges in C57BL/6 mice. PLoS ONE. 2013;8(7):e69123.PubMedPubMedCentralCrossRef Skelly DT, Hennessy E, Dansereau MA, Cunningham C. A systematic analysis of the peripheral and CNS effects of systemic LPS, IL-1beta, [corrected] TNF-alpha and IL-6 challenges in C57BL/6 mice. PLoS ONE. 2013;8(7):e69123.PubMedPubMedCentralCrossRef
24.
go back to reference Liu X, Nemeth DP, McKim DB, Zhu L, DiSabato DJ, Berdysz O, Gorantla G, Oliver B, Witcher KG, Wang Y, et al. Cell-type-specific interleukin 1 receptor 1 signaling in the brain regulates distinct neuroimmune activities. Immunity. 2019;50(3):764–6.PubMedCrossRef Liu X, Nemeth DP, McKim DB, Zhu L, DiSabato DJ, Berdysz O, Gorantla G, Oliver B, Witcher KG, Wang Y, et al. Cell-type-specific interleukin 1 receptor 1 signaling in the brain regulates distinct neuroimmune activities. Immunity. 2019;50(3):764–6.PubMedCrossRef
25.
go back to reference Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol. 1993;47(2):169–76.PubMedCrossRef Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol. 1993;47(2):169–76.PubMedCrossRef
26.
go back to reference van Gool WA, van de Beek D, Eikelenboom P. Systemic infection and delirium: when cytokines and acetylcholine collide. Lancet. 2010;375(9716):773–5.PubMedCrossRef van Gool WA, van de Beek D, Eikelenboom P. Systemic infection and delirium: when cytokines and acetylcholine collide. Lancet. 2010;375(9716):773–5.PubMedCrossRef
27.
go back to reference Hoogland IC, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D. Systemic inflammation and microglial activation: systematic review of animal experiments. J Neuroinflamm. 2015;12:114.CrossRef Hoogland IC, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D. Systemic inflammation and microglial activation: systematic review of animal experiments. J Neuroinflamm. 2015;12:114.CrossRef
28.
go back to reference Zrzavy T, Hoftberger R, Berger T, Rauschka H, Butovsky O, Weiner H, Lassmann H. Pro-inflammatory activation of microglia in the brain of patients with sepsis. Neuropathol Appl Neurobiol. 2019;45(3):278–90.PubMedCrossRef Zrzavy T, Hoftberger R, Berger T, Rauschka H, Butovsky O, Weiner H, Lassmann H. Pro-inflammatory activation of microglia in the brain of patients with sepsis. Neuropathol Appl Neurobiol. 2019;45(3):278–90.PubMedCrossRef
29.
go back to reference Mazeraud A, Righy C, Bouchereau E, Benghanem S, Bozza FA, Sharshar T. Septic-associated encephalopathy: a comprehensive review. Neurotherapeutics. 2020;17(2):392–403.PubMedPubMedCentralCrossRef Mazeraud A, Righy C, Bouchereau E, Benghanem S, Bozza FA, Sharshar T. Septic-associated encephalopathy: a comprehensive review. Neurotherapeutics. 2020;17(2):392–403.PubMedPubMedCentralCrossRef
30.
go back to reference Lemstra AW, Groen in’t Woud JC, Hoozemans JJ, van Haastert ES, Rozemuller AJ, Eikelenboom P, van Gool WA. Microglia activation in sepsis: a case-control study. J Neuroinflamm. 2007;4:4.CrossRef Lemstra AW, Groen in’t Woud JC, Hoozemans JJ, van Haastert ES, Rozemuller AJ, Eikelenboom P, van Gool WA. Microglia activation in sepsis: a case-control study. J Neuroinflamm. 2007;4:4.CrossRef
31.
go back to reference Moraes CA, Santos G, de Sampaio e Spohr TC, D’Avila JC, Lima FR, Benjamim CF, Bozza FA, Gomes FC. Activated microglia-induced deficits in excitatory synapses through IL-1beta: implications for cognitive impairment in sepsis. Mol Neurobiol. 2015;52(1):653–63.PubMedCrossRef Moraes CA, Santos G, de Sampaio e Spohr TC, D’Avila JC, Lima FR, Benjamim CF, Bozza FA, Gomes FC. Activated microglia-induced deficits in excitatory synapses through IL-1beta: implications for cognitive impairment in sepsis. Mol Neurobiol. 2015;52(1):653–63.PubMedCrossRef
33.
go back to reference Hughes CG, Patel MB, Pandharipande PP. Pathophysiology of acute brain dysfunction: what’s the cause of all this confusion? Curr Opin Crit Care. 2012;18(5):518–26.PubMedCrossRef Hughes CG, Patel MB, Pandharipande PP. Pathophysiology of acute brain dysfunction: what’s the cause of all this confusion? Curr Opin Crit Care. 2012;18(5):518–26.PubMedCrossRef
34.
go back to reference Haley MJ, Lawrence CB. The blood-brain barrier after stroke: structural studies and the role of transcytotic vesicles. J Cereb Blood Flow Metab. 2017;37(2):456–70.PubMedCrossRef Haley MJ, Lawrence CB. The blood-brain barrier after stroke: structural studies and the role of transcytotic vesicles. J Cereb Blood Flow Metab. 2017;37(2):456–70.PubMedCrossRef
35.
go back to reference Bernardo-Castro S, Sousa JA, Bras A, Cecilia C, Rodrigues B, Almendra L, Machado C, Santo G, Silva F, Ferreira L, et al. Pathophysiology of blood-brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery. Front Neurol. 2020;11:594672.PubMedPubMedCentralCrossRef Bernardo-Castro S, Sousa JA, Bras A, Cecilia C, Rodrigues B, Almendra L, Machado C, Santo G, Silva F, Ferreira L, et al. Pathophysiology of blood-brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery. Front Neurol. 2020;11:594672.PubMedPubMedCentralCrossRef
36.
go back to reference Stokum JA, Gerzanich V, Simard JM. Molecular pathophysiology of cerebral edema. J Cereb Blood Flow Metab. 2016;36(3):513–38.PubMedCrossRef Stokum JA, Gerzanich V, Simard JM. Molecular pathophysiology of cerebral edema. J Cereb Blood Flow Metab. 2016;36(3):513–38.PubMedCrossRef
37.
go back to reference Paris L, Tonutti L, Vannini C, Bazzoni G. Structural organization of the tight junctions. Biochim Biophys Acta. 2008;1778(3):646–59.PubMedCrossRef Paris L, Tonutti L, Vannini C, Bazzoni G. Structural organization of the tight junctions. Biochim Biophys Acta. 2008;1778(3):646–59.PubMedCrossRef
38.
go back to reference Towner RA, Saunders D, Smith N, Towler W, Cruz M, Do S, Maher JE, Whitaker K, Lerner M, Morton KA. Assessing long-term neuroinflammatory responses to encephalopathy using MRI approaches in a rat endotoxemia model. Geroscience. 2018;40(1):49–60.PubMedPubMedCentralCrossRef Towner RA, Saunders D, Smith N, Towler W, Cruz M, Do S, Maher JE, Whitaker K, Lerner M, Morton KA. Assessing long-term neuroinflammatory responses to encephalopathy using MRI approaches in a rat endotoxemia model. Geroscience. 2018;40(1):49–60.PubMedPubMedCentralCrossRef
39.
go back to reference Stubbs DJ, Yamamoto AK, Menon DK. Imaging in sepsis-associated encephalopathy–insights and opportunities. Nat Rev Neurol. 2013;9(10):551–61.PubMedCrossRef Stubbs DJ, Yamamoto AK, Menon DK. Imaging in sepsis-associated encephalopathy–insights and opportunities. Nat Rev Neurol. 2013;9(10):551–61.PubMedCrossRef
40.
go back to reference Ehler J, Barrett LK, Taylor V, Groves M, Scaravilli F, Wittstock M, Kolbaske S, Grossmann A, Henschel J, Gloger M, et al. Translational evidence for two distinct patterns of neuroaxonal injury in sepsis: a longitudinal, prospective translational study. Crit Care. 2017;21(1):262.PubMedPubMedCentralCrossRef Ehler J, Barrett LK, Taylor V, Groves M, Scaravilli F, Wittstock M, Kolbaske S, Grossmann A, Henschel J, Gloger M, et al. Translational evidence for two distinct patterns of neuroaxonal injury in sepsis: a longitudinal, prospective translational study. Crit Care. 2017;21(1):262.PubMedPubMedCentralCrossRef
41.
go back to reference Dal-Pizzol F, Rojas HA, dos Santos EM, Vuolo F, Constantino L, Feier G, Pasquali M, Comim CM, Petronilho F, Gelain DP, et al. Matrix metalloproteinase-2 and metalloproteinase-9 activities are associated with blood-brain barrier dysfunction in an animal model of severe sepsis. Mol Neurobiol. 2013;48(1):62–70.PubMedCrossRef Dal-Pizzol F, Rojas HA, dos Santos EM, Vuolo F, Constantino L, Feier G, Pasquali M, Comim CM, Petronilho F, Gelain DP, et al. Matrix metalloproteinase-2 and metalloproteinase-9 activities are associated with blood-brain barrier dysfunction in an animal model of severe sepsis. Mol Neurobiol. 2013;48(1):62–70.PubMedCrossRef
42.
go back to reference Erikson K, Tuominen H, Vakkala M, Liisanantti JH, Karttunen T, Syrjala H, Ala-Kokko TI. Brain tight junction protein expression in sepsis in an autopsy series. Crit Care. 2020;24(1):385.PubMedPubMedCentralCrossRef Erikson K, Tuominen H, Vakkala M, Liisanantti JH, Karttunen T, Syrjala H, Ala-Kokko TI. Brain tight junction protein expression in sepsis in an autopsy series. Crit Care. 2020;24(1):385.PubMedPubMedCentralCrossRef
43.
go back to reference Yanagida K, Liu CH, Faraco G, Galvani S, Smith HK, Burg N, Anrather J, Sanchez T, Iadecola C, Hla T. Size-selective opening of the blood-brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci USA. 2017;114(17):4531–6.PubMedPubMedCentralCrossRef Yanagida K, Liu CH, Faraco G, Galvani S, Smith HK, Burg N, Anrather J, Sanchez T, Iadecola C, Hla T. Size-selective opening of the blood-brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci USA. 2017;114(17):4531–6.PubMedPubMedCentralCrossRef
44.
go back to reference Winkler MS, Nierhaus A, Holzmann M, Mudersbach E, Bauer A, Robbe L, Zahrte C, Geffken M, Peine S, Schwedhelm E, et al. Decreased serum concentrations of sphingosine-1-phosphate in sepsis. Crit Care. 2015;19:372.PubMedPubMedCentralCrossRef Winkler MS, Nierhaus A, Holzmann M, Mudersbach E, Bauer A, Robbe L, Zahrte C, Geffken M, Peine S, Schwedhelm E, et al. Decreased serum concentrations of sphingosine-1-phosphate in sepsis. Crit Care. 2015;19:372.PubMedPubMedCentralCrossRef
45.
go back to reference Lopes Pinheiro MA, Kroon J, Hoogenboezem M, Geerts D, van Het Hof B, van der Pol SM, van Buul JD, de Vries HE. Acid sphingomyelinase-derived ceramide regulates ICAM-1 function during T cell transmigration across brain endothelial cells. J Immunol. 2016;196(1):72–9.PubMedCrossRef Lopes Pinheiro MA, Kroon J, Hoogenboezem M, Geerts D, van Het Hof B, van der Pol SM, van Buul JD, de Vries HE. Acid sphingomyelinase-derived ceramide regulates ICAM-1 function during T cell transmigration across brain endothelial cells. J Immunol. 2016;196(1):72–9.PubMedCrossRef
46.
go back to reference Wu H, Deng R, Chen X, Wong WC, Chen H, Gao L, Nie Y, Wu W, Shen J. Caveolin-1 is critical for lymphocyte trafficking into central nervous system during experimental autoimmune encephalomyelitis. J Neurosci. 2016;36(19):5193–9.PubMedPubMedCentralCrossRef Wu H, Deng R, Chen X, Wong WC, Chen H, Gao L, Nie Y, Wu W, Shen J. Caveolin-1 is critical for lymphocyte trafficking into central nervous system during experimental autoimmune encephalomyelitis. J Neurosci. 2016;36(19):5193–9.PubMedPubMedCentralCrossRef
47.
go back to reference Goldblum SE, Ding X, Campbell-Washington J. TNF-alpha induces endothelial cell F-actin depolymerization, new actin synthesis, and barrier dysfunction. Am J Physiol. 1993;264(4 Pt 1):C894-905.PubMedCrossRef Goldblum SE, Ding X, Campbell-Washington J. TNF-alpha induces endothelial cell F-actin depolymerization, new actin synthesis, and barrier dysfunction. Am J Physiol. 1993;264(4 Pt 1):C894-905.PubMedCrossRef
48.
go back to reference O’Carroll SJ, Kho DT, Wiltshire R, Nelson V, Rotimi O, Johnson R, Angel CE, Graham ES. Pro-inflammatory TNFalpha and IL-1beta differentially regulate the inflammatory phenotype of brain microvascular endothelial cells. J Neuroinflamm. 2015;12:131.CrossRef O’Carroll SJ, Kho DT, Wiltshire R, Nelson V, Rotimi O, Johnson R, Angel CE, Graham ES. Pro-inflammatory TNFalpha and IL-1beta differentially regulate the inflammatory phenotype of brain microvascular endothelial cells. J Neuroinflamm. 2015;12:131.CrossRef
49.
go back to reference Haines RJ, Beard RS Jr, Wu MH. Protein tyrosine kinase 6 mediates TNFalpha-induced endothelial barrier dysfunction. Biochem Biophys Res Commun. 2015;456(1):190–6.PubMedCrossRef Haines RJ, Beard RS Jr, Wu MH. Protein tyrosine kinase 6 mediates TNFalpha-induced endothelial barrier dysfunction. Biochem Biophys Res Commun. 2015;456(1):190–6.PubMedCrossRef
50.
go back to reference Lv S, Song HL, Zhou Y, Li LX, Cui W, Wang W, Liu P. Tumour necrosis factor-alpha affects blood-brain barrier permeability and tight junction-associated occludin in acute liver failure. Liver Int. 2010;30(8):1198–210.PubMedCrossRef Lv S, Song HL, Zhou Y, Li LX, Cui W, Wang W, Liu P. Tumour necrosis factor-alpha affects blood-brain barrier permeability and tight junction-associated occludin in acute liver failure. Liver Int. 2010;30(8):1198–210.PubMedCrossRef
51.
go back to reference Janz DR, Abel TW, Jackson JC, Gunther ML, Heckers S, Ely EW. Brain autopsy findings in intensive care unit patients previously suffering from delirium: a pilot study. J Crit Care. 2010;25(3):538.e537-512.CrossRef Janz DR, Abel TW, Jackson JC, Gunther ML, Heckers S, Ely EW. Brain autopsy findings in intensive care unit patients previously suffering from delirium: a pilot study. J Crit Care. 2010;25(3):538.e537-512.CrossRef
52.
go back to reference Pfister D, Siegemund M, Dell-Kuster S, Smielewski P, Rüegg S, Strebel SP, Marsch SC, Pargger H, Steiner LA. Cerebral perfusion in sepsis-associated delirium. Crit Care. 2008;12(3):R63.PubMedPubMedCentralCrossRef Pfister D, Siegemund M, Dell-Kuster S, Smielewski P, Rüegg S, Strebel SP, Marsch SC, Pargger H, Steiner LA. Cerebral perfusion in sepsis-associated delirium. Crit Care. 2008;12(3):R63.PubMedPubMedCentralCrossRef
53.
go back to reference Taccone FS, Castanares-Zapatero D, Peres-Bota D, Vincent JL, Berre J, Melot C. Cerebral autoregulation is influenced by carbon dioxide levels in patients with septic shock. Neurocrit Care. 2010;12(1):35–42.PubMedCrossRef Taccone FS, Castanares-Zapatero D, Peres-Bota D, Vincent JL, Berre J, Melot C. Cerebral autoregulation is influenced by carbon dioxide levels in patients with septic shock. Neurocrit Care. 2010;12(1):35–42.PubMedCrossRef
54.
go back to reference Bermejo-Martin JF, Martin-Fernandez M, Lopez-Mestanza C, Duque P, Almansa R. Shared features of endothelial dysfunction between sepsis and its preceding risk factors (aging and chronic disease). J Clin Med. 2018;7(11):400.PubMedCentralCrossRef Bermejo-Martin JF, Martin-Fernandez M, Lopez-Mestanza C, Duque P, Almansa R. Shared features of endothelial dysfunction between sepsis and its preceding risk factors (aging and chronic disease). J Clin Med. 2018;7(11):400.PubMedCentralCrossRef
55.
go back to reference Nael K, Trouard TP, Lafleur SR, Krupinski EA, Salamon N, Kidwell CS. White matter ischemic changes in hyperacute ischemic stroke: voxel-based analysis using diffusion tensor imaging and MR perfusion. Stroke. 2015;46(2):413–8.PubMedPubMedCentralCrossRef Nael K, Trouard TP, Lafleur SR, Krupinski EA, Salamon N, Kidwell CS. White matter ischemic changes in hyperacute ischemic stroke: voxel-based analysis using diffusion tensor imaging and MR perfusion. Stroke. 2015;46(2):413–8.PubMedPubMedCentralCrossRef
57.
go back to reference Wen Y, Yang S, Liu R, Simpkins JW. Transient cerebral ischemia induces site-specific hyperphosphorylation of tau protein. Brain Res. 2004;1022(1–2):30–8.PubMedCrossRef Wen Y, Yang S, Liu R, Simpkins JW. Transient cerebral ischemia induces site-specific hyperphosphorylation of tau protein. Brain Res. 2004;1022(1–2):30–8.PubMedCrossRef
58.
go back to reference Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer’s disease. Lancet. 2021;397:1577–90.PubMedPubMedCentralCrossRef Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer’s disease. Lancet. 2021;397:1577–90.PubMedPubMedCentralCrossRef
59.
go back to reference Brown GC. The endotoxin hypothesis of neurodegeneration. J Neuroinflamm. 2019;16(1):180.CrossRef Brown GC. The endotoxin hypothesis of neurodegeneration. J Neuroinflamm. 2019;16(1):180.CrossRef
60.
go back to reference Gasparotto J, Girardi CS, Somensi N, Ribeiro CT, Moreira JCF, Michels M, Sonai B, Rocha M, Steckert AV, Barichello T, et al. Receptor for advanced glycation end products mediates sepsis-triggered amyloid-β accumulation, Tau phosphorylation, and cognitive impairment. J Biol Chem. 2018;293(1):226–44.PubMedCrossRef Gasparotto J, Girardi CS, Somensi N, Ribeiro CT, Moreira JCF, Michels M, Sonai B, Rocha M, Steckert AV, Barichello T, et al. Receptor for advanced glycation end products mediates sepsis-triggered amyloid-β accumulation, Tau phosphorylation, and cognitive impairment. J Biol Chem. 2018;293(1):226–44.PubMedCrossRef
61.
go back to reference Kirk RA, Kesner RP, Wang LM, Wu Q, Towner RA, Hoffman JM, Morton KA. Lipopolysaccharide exposure in a rat sepsis model results in hippocampal amyloid-β plaque and phosphorylated tau deposition and corresponding behavioral deficits. Geroscience. 2019;41(4):467–81.PubMedPubMedCentralCrossRef Kirk RA, Kesner RP, Wang LM, Wu Q, Towner RA, Hoffman JM, Morton KA. Lipopolysaccharide exposure in a rat sepsis model results in hippocampal amyloid-β plaque and phosphorylated tau deposition and corresponding behavioral deficits. Geroscience. 2019;41(4):467–81.PubMedPubMedCentralCrossRef
62.
go back to reference Milioli MVM, Burger H, Olivieri R, Michels M, Ávila P, Abatti M, Indalécio A, Ritter C, Dal-Pizzol F. The impact of age on long-term behavioral and neurochemical parameters in an animal model of severe sepsis. Neurosci Lett. 2019;708:134339.PubMedCrossRef Milioli MVM, Burger H, Olivieri R, Michels M, Ávila P, Abatti M, Indalécio A, Ritter C, Dal-Pizzol F. The impact of age on long-term behavioral and neurochemical parameters in an animal model of severe sepsis. Neurosci Lett. 2019;708:134339.PubMedCrossRef
63.
go back to reference Gasparotto J, Girardi CS, Somensi N, Ribeiro CT, Moreira JCF, Michels M, Sonai B, Rocha M, Steckert AV, Barichello T, et al. Receptor for advanced glycation end products mediates sepsis-triggered amyloid-beta accumulation, Tau phosphorylation, and cognitive impairment. J Biol Chem. 2018;293(1):226–44.PubMedCrossRef Gasparotto J, Girardi CS, Somensi N, Ribeiro CT, Moreira JCF, Michels M, Sonai B, Rocha M, Steckert AV, Barichello T, et al. Receptor for advanced glycation end products mediates sepsis-triggered amyloid-beta accumulation, Tau phosphorylation, and cognitive impairment. J Biol Chem. 2018;293(1):226–44.PubMedCrossRef
64.
go back to reference Zhao T, Xia Y, Wang D, Pang L. Association between elevated serum tau protein level and sepsis-associated encephalopathy in patients with severe sepsis. Can J Infect Dis Med Microbiol. 2019;2019:1876174.PubMedPubMedCentralCrossRef Zhao T, Xia Y, Wang D, Pang L. Association between elevated serum tau protein level and sepsis-associated encephalopathy in patients with severe sepsis. Can J Infect Dis Med Microbiol. 2019;2019:1876174.PubMedPubMedCentralCrossRef
65.
go back to reference Orhun G, Esen F, Ozcan PE, Sencer S, Bilgic B, Ulusoy C, Noyan H, Kucukerden M, Ali A, Barburoglu M, et al. Neuroimaging findings in sepsis-induced brain dysfunction: association with clinical and laboratory findings. Neurocrit Care. 2019;30(1):106–17.PubMedCrossRef Orhun G, Esen F, Ozcan PE, Sencer S, Bilgic B, Ulusoy C, Noyan H, Kucukerden M, Ali A, Barburoglu M, et al. Neuroimaging findings in sepsis-induced brain dysfunction: association with clinical and laboratory findings. Neurocrit Care. 2019;30(1):106–17.PubMedCrossRef
66.
go back to reference Rogne AG, Muller EG, Udnaes E, Sigurdardottir S, Raudeberg R, Connelly JP, Revheim ME, Hassel B, Dahlberg D. beta-Amyloid may accumulate in the human brain after focal bacterial infection: an (18) F-flutemetamol positron emission tomography study. Eur J Neurol. 2021;28(3):877–83.PubMedCrossRef Rogne AG, Muller EG, Udnaes E, Sigurdardottir S, Raudeberg R, Connelly JP, Revheim ME, Hassel B, Dahlberg D. beta-Amyloid may accumulate in the human brain after focal bacterial infection: an (18) F-flutemetamol positron emission tomography study. Eur J Neurol. 2021;28(3):877–83.PubMedCrossRef
67.
go back to reference Kamer AR, Pirraglia E, Tsui W, Rusinek H, Vallabhajosula S, Mosconi L, Yi L, McHugh P, Craig RG, Svetcov S, et al. Periodontal disease associates with higher brain amyloid load in normal elderly. Neurobiol Aging. 2015;36(2):627–33.PubMedCrossRef Kamer AR, Pirraglia E, Tsui W, Rusinek H, Vallabhajosula S, Mosconi L, Yi L, McHugh P, Craig RG, Svetcov S, et al. Periodontal disease associates with higher brain amyloid load in normal elderly. Neurobiol Aging. 2015;36(2):627–33.PubMedCrossRef
68.
go back to reference Kantonen J, Mahzabin S, Mayranpaa MI, Tynninen O, Paetau A, Andersson N, Sajantila A, Vapalahti O, Carpen O, Kekalainen E, et al. Neuropathologic features of four autopsied COVID-19 patients. Brain Pathol. 2020;30(6):1012–6.PubMedPubMedCentralCrossRef Kantonen J, Mahzabin S, Mayranpaa MI, Tynninen O, Paetau A, Andersson N, Sajantila A, Vapalahti O, Carpen O, Kekalainen E, et al. Neuropathologic features of four autopsied COVID-19 patients. Brain Pathol. 2020;30(6):1012–6.PubMedPubMedCentralCrossRef
69.
go back to reference Rhodes CH, Priemer DS, Esma K, Perl DP, James G. Β-Amyloid deposits in young COVID patients. Preprints with THE LANCET. 2022. Rhodes CH, Priemer DS, Esma K, Perl DP, James G. Β-Amyloid deposits in young COVID patients. Preprints with THE LANCET. 2022.
70.
go back to reference Wang JZ, Xia YY, Grundke-Iqbal I, Iqbal K. Abnormal hyperphosphorylation of tau: sites, regulation, and molecular mechanism of neurofibrillary degeneration. J Alzheimers Dis. 2013;33(Suppl 1):S123-139.PubMed Wang JZ, Xia YY, Grundke-Iqbal I, Iqbal K. Abnormal hyperphosphorylation of tau: sites, regulation, and molecular mechanism of neurofibrillary degeneration. J Alzheimers Dis. 2013;33(Suppl 1):S123-139.PubMed
71.
go back to reference Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A. Glycogen synthase kinase-3 beta (GSK-3β) signaling: Implications for Parkinson’s disease. Pharmacol Res. 2015;97:16–26.PubMedCrossRef Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A. Glycogen synthase kinase-3 beta (GSK-3β) signaling: Implications for Parkinson’s disease. Pharmacol Res. 2015;97:16–26.PubMedCrossRef
72.
go back to reference Gong R, Ge Y, Chen S, Liang E, Esparza A, Sabo E, Yango A, Gohh R, Rifai A, Dworkin LD. Glycogen synthase kinase 3beta: a novel marker and modulator of inflammatory injury in chronic renal allograft disease. Am J Transplant. 2008;8(9):1852–63.PubMedCrossRef Gong R, Ge Y, Chen S, Liang E, Esparza A, Sabo E, Yango A, Gohh R, Rifai A, Dworkin LD. Glycogen synthase kinase 3beta: a novel marker and modulator of inflammatory injury in chronic renal allograft disease. Am J Transplant. 2008;8(9):1852–63.PubMedCrossRef
73.
go back to reference Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol. 2005;6(8):777–84.PubMedPubMedCentralCrossRef Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol. 2005;6(8):777–84.PubMedPubMedCentralCrossRef
74.
go back to reference Hoeflich KP, Luo J, Rubie EA, Tsao MS, Jin O, Woodgett JR. Requirement for glycogen synthase kinase-3beta in cell survival and NF-kappaB activation. Nature. 2000;406(6791):86–90.PubMedCrossRef Hoeflich KP, Luo J, Rubie EA, Tsao MS, Jin O, Woodgett JR. Requirement for glycogen synthase kinase-3beta in cell survival and NF-kappaB activation. Nature. 2000;406(6791):86–90.PubMedCrossRef
76.
go back to reference Wang H, Brown J, Gu Z, Garcia CA, Liang R, Alard P, Beurel E, Jope RS, Greenway T, Martin M. Convergence of the mammalian target of rapamycin complex 1- and glycogen synthase kinase 3-beta-signaling pathways regulates the innate inflammatory response. J Immunol. 2011;186(9):5217–26.PubMedCrossRef Wang H, Brown J, Gu Z, Garcia CA, Liang R, Alard P, Beurel E, Jope RS, Greenway T, Martin M. Convergence of the mammalian target of rapamycin complex 1- and glycogen synthase kinase 3-beta-signaling pathways regulates the innate inflammatory response. J Immunol. 2011;186(9):5217–26.PubMedCrossRef
78.
go back to reference Yu Y, Ye RD. Microglial Abeta receptors in Alzheimer’s disease. Cell Mol Neurobiol. 2015;35(1):71–83.PubMedCrossRef Yu Y, Ye RD. Microglial Abeta receptors in Alzheimer’s disease. Cell Mol Neurobiol. 2015;35(1):71–83.PubMedCrossRef
79.
go back to reference Cai Z, Liu N, Wang C, Qin B, Zhou Y, Xiao M, Chang L, Yan LJ, Zhao B. Role of RAGE in Alzheimer’s disease. Cell Mol Neurobiol. 2016;36(4):483–95.PubMedCrossRef Cai Z, Liu N, Wang C, Qin B, Zhou Y, Xiao M, Chang L, Yan LJ, Zhao B. Role of RAGE in Alzheimer’s disease. Cell Mol Neurobiol. 2016;36(4):483–95.PubMedCrossRef
80.
go back to reference Wautier MP, Tessier FJ, Wautier JL. Advanced glycation end products: a risk factor for human health. Ann Pharm Fr. 2014;72(6):400–8.PubMedCrossRef Wautier MP, Tessier FJ, Wautier JL. Advanced glycation end products: a risk factor for human health. Ann Pharm Fr. 2014;72(6):400–8.PubMedCrossRef
81.
go back to reference Paudel YN, Angelopoulou E, Piperi C, Othman I, Aamir K, Shaikh MF. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s disease (AD): from risk factors to therapeutic targeting. Cells. 2020;9(2):383.PubMedCentralCrossRef Paudel YN, Angelopoulou E, Piperi C, Othman I, Aamir K, Shaikh MF. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s disease (AD): from risk factors to therapeutic targeting. Cells. 2020;9(2):383.PubMedCentralCrossRef
82.
go back to reference Leclerc E, Fritz G, Vetter SW, Heizmann CW. Binding of S100 proteins to RAGE: an update. Biochim Biophys Acta. 2009;1793(6):993–1007.PubMedCrossRef Leclerc E, Fritz G, Vetter SW, Heizmann CW. Binding of S100 proteins to RAGE: an update. Biochim Biophys Acta. 2009;1793(6):993–1007.PubMedCrossRef
83.
go back to reference Sirois CM, Jin T, Miller AL, Bertheloot D, Nakamura H, Horvath GL, Mian A, Jiang J, Schrum J, Bossaller L, et al. RAGE is a nucleic acid receptor that promotes inflammatory responses to DNA. J Exp Med. 2013;210(11):2447–63.PubMedPubMedCentralCrossRef Sirois CM, Jin T, Miller AL, Bertheloot D, Nakamura H, Horvath GL, Mian A, Jiang J, Schrum J, Bossaller L, et al. RAGE is a nucleic acid receptor that promotes inflammatory responses to DNA. J Exp Med. 2013;210(11):2447–63.PubMedPubMedCentralCrossRef
84.
go back to reference Li K, Dai D, Zhao B, Yao L, Yao S, Wang B, Yang Z. Association between the RAGE G82S polymorphism and Alzheimer’s disease. J Neural Transm (Vienna). 2010;117(1):97–104.CrossRef Li K, Dai D, Zhao B, Yao L, Yao S, Wang B, Yang Z. Association between the RAGE G82S polymorphism and Alzheimer’s disease. J Neural Transm (Vienna). 2010;117(1):97–104.CrossRef
85.
go back to reference Lue LF, Walker DG, Brachova L, Beach TG, Rogers J, Schmidt AM, Stern DM, Yan SD. Involvement of microglial receptor for advanced glycation endproducts (RAGE) in Alzheimer’s disease: identification of a cellular activation mechanism. Exp Neurol. 2001;171(1):29–45.PubMedCrossRef Lue LF, Walker DG, Brachova L, Beach TG, Rogers J, Schmidt AM, Stern DM, Yan SD. Involvement of microglial receptor for advanced glycation endproducts (RAGE) in Alzheimer’s disease: identification of a cellular activation mechanism. Exp Neurol. 2001;171(1):29–45.PubMedCrossRef
86.
go back to reference Arancio O, Zhang HP, Chen X, Lin C, Trinchese F, Puzzo D, Liu S, Hegde A, Yan SF, Stern A, et al. RAGE potentiates Abeta-induced perturbation of neuronal function in transgenic mice. EMBO J. 2004;23(20):4096–105.PubMedPubMedCentralCrossRef Arancio O, Zhang HP, Chen X, Lin C, Trinchese F, Puzzo D, Liu S, Hegde A, Yan SF, Stern A, et al. RAGE potentiates Abeta-induced perturbation of neuronal function in transgenic mice. EMBO J. 2004;23(20):4096–105.PubMedPubMedCentralCrossRef
87.
go back to reference Fang F, Lue LF, Yan S, Xu H, Luddy JS, Chen D, Walker DG, Stern DM, Yan S, Schmidt AM, et al. RAGE-dependent signaling in microglia contributes to neuroinflammation, Abeta accumulation, and impaired learning/memory in a mouse model of Alzheimer’s disease. FASEB J. 2010;24(4):1043–55.PubMedPubMedCentralCrossRef Fang F, Lue LF, Yan S, Xu H, Luddy JS, Chen D, Walker DG, Stern DM, Yan S, Schmidt AM, et al. RAGE-dependent signaling in microglia contributes to neuroinflammation, Abeta accumulation, and impaired learning/memory in a mouse model of Alzheimer’s disease. FASEB J. 2010;24(4):1043–55.PubMedPubMedCentralCrossRef
88.
go back to reference Fang F, Yu Q, Arancio O, Chen D, Gore SS, Yan SS, Yan SF. RAGE mediates Abeta accumulation in a mouse model of Alzheimer’s disease via modulation of beta- and gamma-secretase activity. Hum Mol Genet. 2018;27(6):1002–14.PubMedPubMedCentralCrossRef Fang F, Yu Q, Arancio O, Chen D, Gore SS, Yan SS, Yan SF. RAGE mediates Abeta accumulation in a mouse model of Alzheimer’s disease via modulation of beta- and gamma-secretase activity. Hum Mol Genet. 2018;27(6):1002–14.PubMedPubMedCentralCrossRef
89.
go back to reference Sharshar T, Annane D, de la Grandmaison GL, Brouland JP, Hopkinson NS, Francoise G. The neuropathology of septic shock. Brain Pathol. 2004;14(1):21–33.PubMedCrossRef Sharshar T, Annane D, de la Grandmaison GL, Brouland JP, Hopkinson NS, Francoise G. The neuropathology of septic shock. Brain Pathol. 2004;14(1):21–33.PubMedCrossRef
90.
go back to reference Valderrama EV, Humbert K, Lord A, Frontera J, Yaghi S. Severe acute respiratory syndrome coronavirus 2 infection and ischemic stroke. Stroke. 2020;51(7):e124–7.PubMedCrossRef Valderrama EV, Humbert K, Lord A, Frontera J, Yaghi S. Severe acute respiratory syndrome coronavirus 2 infection and ischemic stroke. Stroke. 2020;51(7):e124–7.PubMedCrossRef
91.
93.
go back to reference Czempik PF, Pluta MP, Krzych ŁJ. Sepsis-associated brain dysfunction: a review of current literature. Int J Environ Res Public Health. 2020;17(16):5852.PubMedCentralCrossRef Czempik PF, Pluta MP, Krzych ŁJ. Sepsis-associated brain dysfunction: a review of current literature. Int J Environ Res Public Health. 2020;17(16):5852.PubMedCentralCrossRef
96.
go back to reference Sonneville R, de Montmollin E, Poujade J, Garrouste-Orgeas M, Souweine B, Darmon M, Mariotte E, Argaud L, Barbier F, Goldgran-Toledano D, et al. Potentially modifiable factors contributing to sepsis-associated encephalopathy. Intensive Care Med. 2017;43(8):1075–84.PubMedCrossRef Sonneville R, de Montmollin E, Poujade J, Garrouste-Orgeas M, Souweine B, Darmon M, Mariotte E, Argaud L, Barbier F, Goldgran-Toledano D, et al. Potentially modifiable factors contributing to sepsis-associated encephalopathy. Intensive Care Med. 2017;43(8):1075–84.PubMedCrossRef
97.
go back to reference Chung HY, Wickel J, Brunkhorst FM, Geis C. Sepsis-associated encephalopathy: from delirium to dementia? J Clin Med. 2020;9(3):703.PubMedCentralCrossRef Chung HY, Wickel J, Brunkhorst FM, Geis C. Sepsis-associated encephalopathy: from delirium to dementia? J Clin Med. 2020;9(3):703.PubMedCentralCrossRef
98.
go back to reference Zhang H, Yuan J, Chen Q, Cao Y, Wang Z, Lu W, Bao J. Development and validation of a predictive score for ICU delirium in critically ill patients. BMC Anesthesiol. 2021;21(1):37.PubMedPubMedCentralCrossRef Zhang H, Yuan J, Chen Q, Cao Y, Wang Z, Lu W, Bao J. Development and validation of a predictive score for ICU delirium in critically ill patients. BMC Anesthesiol. 2021;21(1):37.PubMedPubMedCentralCrossRef
99.
go back to reference Atterton B, Paulino MC, Povoa P, Martin-Loeches I. Sepsis associated delirium. Medicina (Kaunas). 2020;56(5):240.CrossRef Atterton B, Paulino MC, Povoa P, Martin-Loeches I. Sepsis associated delirium. Medicina (Kaunas). 2020;56(5):240.CrossRef
100.
go back to reference Girard TD, Thompson JL, Pandharipande PP, Brummel NE, Jackson JC, Patel MB, Hughes CG, Chandrasekhar R, Pun BT, Boehm LM, et al. Clinical phenotypes of delirium during critical illness and severity of subsequent long-term cognitive impairment: a prospective cohort study. Lancet Respir Med. 2018;6(3):213–22.PubMedPubMedCentralCrossRef Girard TD, Thompson JL, Pandharipande PP, Brummel NE, Jackson JC, Patel MB, Hughes CG, Chandrasekhar R, Pun BT, Boehm LM, et al. Clinical phenotypes of delirium during critical illness and severity of subsequent long-term cognitive impairment: a prospective cohort study. Lancet Respir Med. 2018;6(3):213–22.PubMedPubMedCentralCrossRef
101.
go back to reference Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav Immun. 2007;21(2):153–60.PubMedCrossRef Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav Immun. 2007;21(2):153–60.PubMedCrossRef
102.
go back to reference Kelley KW, Bluthé RM, Dantzer R, Zhou JH, Shen WH, Johnson RW, Broussard SR. Cytokine-induced sickness behavior. Brain Behav Immun. 2003;17(Suppl 1):S112-118.PubMedCrossRef Kelley KW, Bluthé RM, Dantzer R, Zhou JH, Shen WH, Johnson RW, Broussard SR. Cytokine-induced sickness behavior. Brain Behav Immun. 2003;17(Suppl 1):S112-118.PubMedCrossRef
103.
go back to reference Shattuck EC, Muehlenbein MP. Towards an integrative picture of human sickness behavior. Brain Behav Immun. 2016;57:255–62.PubMedCrossRef Shattuck EC, Muehlenbein MP. Towards an integrative picture of human sickness behavior. Brain Behav Immun. 2016;57:255–62.PubMedCrossRef
104.
go back to reference Pereira de Souza Goldim M, Della Giustina A, Mathias K, de Oliveira Junior A, Fileti ME, De Carli R, Zarbato G, Garbossa L, da Rosa N, Oliveira J, et al. Sickness behavior score is associated with neuroinflammation and late behavioral changes in polymicrobial sepsis animal model. Inflammation. 2020;43(3):1019–34.PubMedCrossRef Pereira de Souza Goldim M, Della Giustina A, Mathias K, de Oliveira Junior A, Fileti ME, De Carli R, Zarbato G, Garbossa L, da Rosa N, Oliveira J, et al. Sickness behavior score is associated with neuroinflammation and late behavioral changes in polymicrobial sepsis animal model. Inflammation. 2020;43(3):1019–34.PubMedCrossRef
105.
go back to reference Honarmand K, Lalli RS, Priestap F, Chen JL, McIntyre CW, Owen AM, Slessarev M. Natural history of cognitive impairment in critical illness survivors. A systematic review. Am J Respir Crit Care Med. 2020;202(2):193–201.PubMedPubMedCentralCrossRef Honarmand K, Lalli RS, Priestap F, Chen JL, McIntyre CW, Owen AM, Slessarev M. Natural history of cognitive impairment in critical illness survivors. A systematic review. Am J Respir Crit Care Med. 2020;202(2):193–201.PubMedPubMedCentralCrossRef
107.
go back to reference Mart MF, Williams Roberson S, Salas B, Pandharipande PP, Ely EW. Prevention and management of delirium in the intensive care unit. Semin Respir Crit Care Med. 2021;42(1):112–26.PubMedCrossRef Mart MF, Williams Roberson S, Salas B, Pandharipande PP, Ely EW. Prevention and management of delirium in the intensive care unit. Semin Respir Crit Care Med. 2021;42(1):112–26.PubMedCrossRef
108.
go back to reference Bennett IJ, Madden DJ. Disconnected aging: cerebral white matter integrity and age-related differences in cognition. Neuroscience. 2014;276:187–205.PubMedCrossRef Bennett IJ, Madden DJ. Disconnected aging: cerebral white matter integrity and age-related differences in cognition. Neuroscience. 2014;276:187–205.PubMedCrossRef
109.
go back to reference Hase Y, Horsburgh K, Ihara M, Kalaria RN. White matter degeneration in vascular and other ageing-related dementias. J Neurochem. 2018;144(5):617–33.PubMedCrossRef Hase Y, Horsburgh K, Ihara M, Kalaria RN. White matter degeneration in vascular and other ageing-related dementias. J Neurochem. 2018;144(5):617–33.PubMedCrossRef
110.
go back to reference Orhun G, Tuzun E, Bilgic B, Ergin Ozcan P, Sencer S, Barburoglu M, Esen F. Brain volume changes in patients with acute brain dysfunction due to sepsis. Neurocrit Care. 2020;32(2):459–68.PubMedCrossRef Orhun G, Tuzun E, Bilgic B, Ergin Ozcan P, Sencer S, Barburoglu M, Esen F. Brain volume changes in patients with acute brain dysfunction due to sepsis. Neurocrit Care. 2020;32(2):459–68.PubMedCrossRef
111.
go back to reference Luitse MJ, van Asch CJ, Klijn CJ. Deep coma and diffuse white matter abnormalities caused by sepsis-associated encephalopathy. Lancet. 2013;381(9884):2222.PubMedCrossRef Luitse MJ, van Asch CJ, Klijn CJ. Deep coma and diffuse white matter abnormalities caused by sepsis-associated encephalopathy. Lancet. 2013;381(9884):2222.PubMedCrossRef
112.
go back to reference Sharshar T, Carlier R, Bernard F, Guidoux C, Brouland JP, Nardi O, de la Grandmaison GL, Aboab J, Gray F, Menon D, et al. Brain lesions in septic shock: a magnetic resonance imaging study. Intensive Care Med. 2007;33(5):798–806.PubMedCrossRef Sharshar T, Carlier R, Bernard F, Guidoux C, Brouland JP, Nardi O, de la Grandmaison GL, Aboab J, Gray F, Menon D, et al. Brain lesions in septic shock: a magnetic resonance imaging study. Intensive Care Med. 2007;33(5):798–806.PubMedCrossRef
113.
go back to reference Seidel G, Gaser C, Götz T, Günther A, Hamzei F. Accelerated brain ageing in sepsis survivors with cognitive long-term impairment. Eur J Neurosci. 2020;52(10):4395–402.PubMedCrossRef Seidel G, Gaser C, Götz T, Günther A, Hamzei F. Accelerated brain ageing in sepsis survivors with cognitive long-term impairment. Eur J Neurosci. 2020;52(10):4395–402.PubMedCrossRef
114.
go back to reference Gunther ML, Morandi A, Krauskopf E, Pandharipande P, Girard TD, Jackson JC, Thompson J, Shintani AK, Geevarghese S, Miller RR 3rd, et al. The association between brain volumes, delirium duration, and cognitive outcomes in intensive care unit survivors: the VISIONS cohort magnetic resonance imaging study*. Crit Care Med. 2012;40(7):2022–32.PubMedPubMedCentralCrossRef Gunther ML, Morandi A, Krauskopf E, Pandharipande P, Girard TD, Jackson JC, Thompson J, Shintani AK, Geevarghese S, Miller RR 3rd, et al. The association between brain volumes, delirium duration, and cognitive outcomes in intensive care unit survivors: the VISIONS cohort magnetic resonance imaging study*. Crit Care Med. 2012;40(7):2022–32.PubMedPubMedCentralCrossRef
115.
go back to reference Garofalo AM, Lorente-Ros M, Goncalvez G, Carriedo D, Ballén-Barragán A, Villar-Fernández A, Peñuelas Ó, Herrero R, Granados-Carreño R, Lorente JA. Histopathological changes of organ dysfunction in sepsis. Intensive Care Med Exp. 2019;7(Suppl 1):45.PubMedPubMedCentralCrossRef Garofalo AM, Lorente-Ros M, Goncalvez G, Carriedo D, Ballén-Barragán A, Villar-Fernández A, Peñuelas Ó, Herrero R, Granados-Carreño R, Lorente JA. Histopathological changes of organ dysfunction in sepsis. Intensive Care Med Exp. 2019;7(Suppl 1):45.PubMedPubMedCentralCrossRef
116.
go back to reference Sharshar T, Gray F, Lorin de la Grandmaison G, Hopkinson NS, Ross E, Dorandeu A, Orlikowski D, Raphael JC, Gajdos P, Annane D. Apoptosis of neurons in cardiovascular autonomic centres triggered by inducible nitric oxide synthase after death from septic shock. Lancet. 2003;362(9398):1799–805.PubMedCrossRef Sharshar T, Gray F, Lorin de la Grandmaison G, Hopkinson NS, Ross E, Dorandeu A, Orlikowski D, Raphael JC, Gajdos P, Annane D. Apoptosis of neurons in cardiovascular autonomic centres triggered by inducible nitric oxide synthase after death from septic shock. Lancet. 2003;362(9398):1799–805.PubMedCrossRef
117.
go back to reference Bozza FA, Garteiser P, Oliveira MF, Doblas S, Cranford R, Saunders D, Jones I, Towner RA, Castro-Faria-Neto HC. Sepsis-associated encephalopathy: a magnetic resonance imaging and spectroscopy study. J Cereb Blood Flow Metab. 2010;30(2):440–8.PubMedCrossRef Bozza FA, Garteiser P, Oliveira MF, Doblas S, Cranford R, Saunders D, Jones I, Towner RA, Castro-Faria-Neto HC. Sepsis-associated encephalopathy: a magnetic resonance imaging and spectroscopy study. J Cereb Blood Flow Metab. 2010;30(2):440–8.PubMedCrossRef
118.
go back to reference Tomiyama T, Shoji A, Kataoka K, Suwa Y, Asano S, Kaneko H, Endo N. Inhibition of amyloid beta protein aggregation and neurotoxicity by rifampicin. Its possible function as a hydroxyl radical scavenger. J Biol Chem. 1996;271(12):6839–44.PubMedCrossRef Tomiyama T, Shoji A, Kataoka K, Suwa Y, Asano S, Kaneko H, Endo N. Inhibition of amyloid beta protein aggregation and neurotoxicity by rifampicin. Its possible function as a hydroxyl radical scavenger. J Biol Chem. 1996;271(12):6839–44.PubMedCrossRef
119.
go back to reference Zhu L, Yuan Q, Zeng Z, Zhou R, Luo R, Zhang J, Tsang CK, Bi W. Rifampicin suppresses amyloid-β accumulation through enhancing autophagy in the hippocampus of a lipopolysaccharide-induced mouse model of cognitive decline. J Alzheimers Dis. 2021;79(3):1171–84.PubMedCrossRef Zhu L, Yuan Q, Zeng Z, Zhou R, Luo R, Zhang J, Tsang CK, Bi W. Rifampicin suppresses amyloid-β accumulation through enhancing autophagy in the hippocampus of a lipopolysaccharide-induced mouse model of cognitive decline. J Alzheimers Dis. 2021;79(3):1171–84.PubMedCrossRef
120.
go back to reference Iizuka T, Morimoto K, Sasaki Y, Kameyama M, Kurashima A, Hayasaka K, Ogata H, Goto H. Preventive effect of rifampicin on alzheimer disease needs at least 450 mg daily for 1 year: an FDG-PET follow-up study. Dement Geriatr Cogn Dis Extra. 2017;7(2):204–14.PubMedPubMedCentralCrossRef Iizuka T, Morimoto K, Sasaki Y, Kameyama M, Kurashima A, Hayasaka K, Ogata H, Goto H. Preventive effect of rifampicin on alzheimer disease needs at least 450 mg daily for 1 year: an FDG-PET follow-up study. Dement Geriatr Cogn Dis Extra. 2017;7(2):204–14.PubMedPubMedCentralCrossRef
121.
go back to reference Lee CY, Huang CH, Lu PL, Ko WC, Chen YH, Hsueh PR. Role of rifampin for the treatment of bacterial infections other than mycobacteriosis. J Infect. 2017;75(5):395–408.PubMedCrossRef Lee CY, Huang CH, Lu PL, Ko WC, Chen YH, Hsueh PR. Role of rifampin for the treatment of bacterial infections other than mycobacteriosis. J Infect. 2017;75(5):395–408.PubMedCrossRef
123.
go back to reference Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, Sanvito F, Ponzoni M, Doglioni C, Cristofori P, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24(6):739–48.PubMedCrossRef Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, Sanvito F, Ponzoni M, Doglioni C, Cristofori P, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24(6):739–48.PubMedCrossRef
124.
go back to reference Shakoory B, Carcillo JA, Chatham WW, Amdur RL, Zhao H, Dinarello CA, Cron RQ, Opal SM. Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior phase III trial. Crit Care Med. 2016;44(2):275–81.PubMedPubMedCentralCrossRef Shakoory B, Carcillo JA, Chatham WW, Amdur RL, Zhao H, Dinarello CA, Cron RQ, Opal SM. Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior phase III trial. Crit Care Med. 2016;44(2):275–81.PubMedPubMedCentralCrossRef
125.
go back to reference Abraham E, Wunderink R, Silverman H, Perl TM, Nasraway S, Levy H, Bone R, Wenzel RP, Balk R, Allred R, et al. Efficacy and safety of monoclonal antibody to human tumor necrosis factor alpha in patients with sepsis syndrome. A randomized, controlled, double-blind, multicenter clinical trial. TNF-alpha MAb Sepsis Study Group. JAMA. 1995;273(12):934–41.PubMedCrossRef Abraham E, Wunderink R, Silverman H, Perl TM, Nasraway S, Levy H, Bone R, Wenzel RP, Balk R, Allred R, et al. Efficacy and safety of monoclonal antibody to human tumor necrosis factor alpha in patients with sepsis syndrome. A randomized, controlled, double-blind, multicenter clinical trial. TNF-alpha MAb Sepsis Study Group. JAMA. 1995;273(12):934–41.PubMedCrossRef
128.
go back to reference Myburgh JA, Finfer S, Bellomo R, Billot L, Cass A, Gattas D, Glass P, Lipman J, Liu B, McArthur C, et al. Hydroxyethyl starch or saline for fluid resuscitation in intensive care. N Engl J Med. 2012;367(20):1901–11.PubMedCrossRef Myburgh JA, Finfer S, Bellomo R, Billot L, Cass A, Gattas D, Glass P, Lipman J, Liu B, McArthur C, et al. Hydroxyethyl starch or saline for fluid resuscitation in intensive care. N Engl J Med. 2012;367(20):1901–11.PubMedCrossRef
129.
go back to reference Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, et al. Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med. 2008;358(2):125–39.PubMedCrossRef Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, et al. Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med. 2008;358(2):125–39.PubMedCrossRef
130.
go back to reference Perner A, Haase N, Guttormsen AB, Tenhunen J, Klemenzson G, Åneman A, Madsen KR, Møller MH, Elkjær JM, Poulsen LM, et al. Hydroxyethyl starch 130/0.42 versus Ringer’s acetate in severe sepsis. N Engl J Med. 2012;367(2):124–34.PubMedCrossRef Perner A, Haase N, Guttormsen AB, Tenhunen J, Klemenzson G, Åneman A, Madsen KR, Møller MH, Elkjær JM, Poulsen LM, et al. Hydroxyethyl starch 130/0.42 versus Ringer’s acetate in severe sepsis. N Engl J Med. 2012;367(2):124–34.PubMedCrossRef
131.
go back to reference Tymianski M. Combining neuroprotection with endovascular treatment of acute stroke: is there hope? Stroke. 2017;48(6):1700–5.PubMedCrossRef Tymianski M. Combining neuroprotection with endovascular treatment of acute stroke: is there hope? Stroke. 2017;48(6):1700–5.PubMedCrossRef
132.
go back to reference Hill MD, Goyal M, Menon BK, Nogueira RG, McTaggart RA, Demchuk AM, Poppe AY, Buck BH, Field TS, Dowlatshahi D, et al. Efficacy and safety of nerinetide for the treatment of acute ischaemic stroke (ESCAPE-NA1): a multicentre, double-blind, randomised controlled trial. Lancet. 2020;395(10227):878–87.PubMedCrossRef Hill MD, Goyal M, Menon BK, Nogueira RG, McTaggart RA, Demchuk AM, Poppe AY, Buck BH, Field TS, Dowlatshahi D, et al. Efficacy and safety of nerinetide for the treatment of acute ischaemic stroke (ESCAPE-NA1): a multicentre, double-blind, randomised controlled trial. Lancet. 2020;395(10227):878–87.PubMedCrossRef
133.
go back to reference Olivieri R, Michels M, Pescador B, Avila P, Abatti M, Cucker L, Burger H, Dominguini D, Quevedo J, Dal-Pizzol F. The additive effect of aging on sepsis-induced cognitive impairment and neuroinflammation. J Neuroimmunol. 2018;314:1–7.PubMedCrossRef Olivieri R, Michels M, Pescador B, Avila P, Abatti M, Cucker L, Burger H, Dominguini D, Quevedo J, Dal-Pizzol F. The additive effect of aging on sepsis-induced cognitive impairment and neuroinflammation. J Neuroimmunol. 2018;314:1–7.PubMedCrossRef
134.
go back to reference Waisman A, Hauptmann J, Regen T. The role of IL-17 in CNS diseases. Acta Neuropathol. 2015;129(5):625–37.PubMedCrossRef Waisman A, Hauptmann J, Regen T. The role of IL-17 in CNS diseases. Acta Neuropathol. 2015;129(5):625–37.PubMedCrossRef
135.
go back to reference Kawanokuchi J, Shimizu K, Nitta A, Yamada K, Mizuno T, Takeuchi H, Suzumura A. Production and functions of IL-17 in microglia. J Neuroimmunol. 2008;194(1–2):54–61.PubMedCrossRef Kawanokuchi J, Shimizu K, Nitta A, Yamada K, Mizuno T, Takeuchi H, Suzumura A. Production and functions of IL-17 in microglia. J Neuroimmunol. 2008;194(1–2):54–61.PubMedCrossRef
136.
go back to reference Ye B, Tao T, Zhao A, Wen L, He X, Liu Y, Fu Q, Mi W, Lou J. Blockade of IL-17A/IL-17R pathway protected mice from sepsis-associated encephalopathy by inhibition of microglia activation. Mediators Inflamm. 2019;2019:8461725.PubMedPubMedCentral Ye B, Tao T, Zhao A, Wen L, He X, Liu Y, Fu Q, Mi W, Lou J. Blockade of IL-17A/IL-17R pathway protected mice from sepsis-associated encephalopathy by inhibition of microglia activation. Mediators Inflamm. 2019;2019:8461725.PubMedPubMedCentral
137.
139.
140.
go back to reference Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG, et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med. 2019;25(2):270–6.PubMedPubMedCentralCrossRef Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG, et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med. 2019;25(2):270–6.PubMedPubMedCentralCrossRef
141.
go back to reference Zhang SQ, Xiao J, Chen M, Zhou LQ, Shang K, Qin C, Tian DS. Sphingosine-1-phosphate signaling in ischemic stroke: from bench to bedside and beyond. Front Cell Neurosci. 2021;15:781098.PubMedPubMedCentralCrossRef Zhang SQ, Xiao J, Chen M, Zhou LQ, Shang K, Qin C, Tian DS. Sphingosine-1-phosphate signaling in ischemic stroke: from bench to bedside and beyond. Front Cell Neurosci. 2021;15:781098.PubMedPubMedCentralCrossRef
142.
go back to reference Vutukuri R, Brunkhorst R, Kestner RI, Hansen L, Bouzas NF, Pfeilschifter J, Devraj K, Pfeilschifter W. Alteration of sphingolipid metabolism as a putative mechanism underlying LPS-induced BBB disruption. J Neurochem. 2018;144(2):172–85.PubMedCrossRef Vutukuri R, Brunkhorst R, Kestner RI, Hansen L, Bouzas NF, Pfeilschifter J, Devraj K, Pfeilschifter W. Alteration of sphingolipid metabolism as a putative mechanism underlying LPS-induced BBB disruption. J Neurochem. 2018;144(2):172–85.PubMedCrossRef
143.
144.
go back to reference Aytan N, Choi JK, Carreras I, Brinkmann V, Kowall NW, Jenkins BG, Dedeoglu A. Fingolimod modulates multiple neuroinflammatory markers in a mouse model of Alzheimer’s disease. Sci Rep. 2016;6:24939.PubMedPubMedCentralCrossRef Aytan N, Choi JK, Carreras I, Brinkmann V, Kowall NW, Jenkins BG, Dedeoglu A. Fingolimod modulates multiple neuroinflammatory markers in a mouse model of Alzheimer’s disease. Sci Rep. 2016;6:24939.PubMedPubMedCentralCrossRef
145.
go back to reference Gaire BP, Lee CH, Sapkota A, Lee SY, Chun J, Cho HJ, Nam TG, Choi JW. Identification of sphingosine 1-phosphate receptor subtype 1 (S1P1) as a pathogenic factor in transient focal cerebral ischemia. Mol Neurobiol. 2018;55(3):2320–32.PubMedCrossRef Gaire BP, Lee CH, Sapkota A, Lee SY, Chun J, Cho HJ, Nam TG, Choi JW. Identification of sphingosine 1-phosphate receptor subtype 1 (S1P1) as a pathogenic factor in transient focal cerebral ischemia. Mol Neurobiol. 2018;55(3):2320–32.PubMedCrossRef
146.
go back to reference Nitzsche A, Poittevin M, Benarab A, Bonnin P, Faraco G, Uchida H, Favre J, Garcia-Bonilla L, Garcia MCL, Leger PL, et al. Endothelial S1P1 signaling counteracts infarct expansion in ischemic stroke. Circ Res. 2021;128(3):363–82.PubMedCrossRef Nitzsche A, Poittevin M, Benarab A, Bonnin P, Faraco G, Uchida H, Favre J, Garcia-Bonilla L, Garcia MCL, Leger PL, et al. Endothelial S1P1 signaling counteracts infarct expansion in ischemic stroke. Circ Res. 2021;128(3):363–82.PubMedCrossRef
147.
go back to reference Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–6.PubMedCrossRef Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–6.PubMedCrossRef
148.
go back to reference Hampel H, Lista S, Mango D, Nisticò R, Perry G, Avila J, Hernandez F, Geerts H, Vergallo A. Lithium as a treatment for Alzheimer’s disease: the systems pharmacology perspective. J Alzheimers Dis. 2019;69(3):615–29.PubMedCrossRef Hampel H, Lista S, Mango D, Nisticò R, Perry G, Avila J, Hernandez F, Geerts H, Vergallo A. Lithium as a treatment for Alzheimer’s disease: the systems pharmacology perspective. J Alzheimers Dis. 2019;69(3):615–29.PubMedCrossRef
149.
go back to reference Matsunaga S, Kishi T, Annas P, Basun H, Hampel H, Iwata N. Lithium as a treatment for Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2015;48(2):403–10.PubMedCrossRef Matsunaga S, Kishi T, Annas P, Basun H, Hampel H, Iwata N. Lithium as a treatment for Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2015;48(2):403–10.PubMedCrossRef
151.
go back to reference Angus DC, Yang L, Kong L, Kellum JA, Delude RL, Tracey KJ, Weissfeld L, Gen IMSI. Circulating high-mobility group box 1 (HMGB1) concentrations are elevated in both uncomplicated pneumonia and pneumonia with severe sepsis. Crit Care Med. 2007;35(4):1061–7.PubMedCrossRef Angus DC, Yang L, Kong L, Kellum JA, Delude RL, Tracey KJ, Weissfeld L, Gen IMSI. Circulating high-mobility group box 1 (HMGB1) concentrations are elevated in both uncomplicated pneumonia and pneumonia with severe sepsis. Crit Care Med. 2007;35(4):1061–7.PubMedCrossRef
152.
go back to reference Chavan SS, Huerta PT, Robbiati S, Valdes-Ferrer SI, Ochani M, Dancho M, Frankfurt M, Volpe BT, Tracey KJ, Diamond B. HMGB1 mediates cognitive impairment in sepsis survivors. Mol Med. 2012;18:930–7.PubMedPubMedCentralCrossRef Chavan SS, Huerta PT, Robbiati S, Valdes-Ferrer SI, Ochani M, Dancho M, Frankfurt M, Volpe BT, Tracey KJ, Diamond B. HMGB1 mediates cognitive impairment in sepsis survivors. Mol Med. 2012;18:930–7.PubMedPubMedCentralCrossRef
153.
go back to reference Jarczak D, Kluge S, Nierhaus A. Sepsis-pathophysiology and therapeutic concepts. Front Med (Lausanne). 2021;8:628302.CrossRef Jarczak D, Kluge S, Nierhaus A. Sepsis-pathophysiology and therapeutic concepts. Front Med (Lausanne). 2021;8:628302.CrossRef
154.
go back to reference van der Slikke EC, An AY, Hancock REW, Bouma HR. Exploring the pathophysiology of post-sepsis syndrome to identify therapeutic opportunities. EBioMedicine. 2020;61:103044.PubMedPubMedCentralCrossRef van der Slikke EC, An AY, Hancock REW, Bouma HR. Exploring the pathophysiology of post-sepsis syndrome to identify therapeutic opportunities. EBioMedicine. 2020;61:103044.PubMedPubMedCentralCrossRef
Metadata
Title
Sepsis-associated brain injury: underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments
Authors
Nobufumi Sekino
Magdy Selim
Amjad Shehadah
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2022
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02464-4

Other articles of this Issue 1/2022

Journal of Neuroinflammation 1/2022 Go to the issue