Mini review

Cholesterol dependent cytolysins and the brain: Revealing a potential therapeutic avenue for bacterial meningitis

  • Received: 18 May 2023 Revised: 24 July 2023 Accepted: 14 August 2023 Published: 21 August 2023
  • Bacterial meningitis is a catastrophic nervous system disorder with high mortality and wide range of morbidities. Some of the meningitis-causing bacteria occupy cholesterol dependent cytolysins (CDCs) to increase their pathogenicity and arrange immune-evasion strategy. Studies have observed that the relationship between CDCs and pathogenicity in these meningitides is complex and involves interactions between CDC, blood-brain barrier (BBB), glial cells and neurons. In BBB, these CDCs acts on capillary endothelium, tight junction (TJ) proteins and neurovascular unit (NVU). CDCs also observed to elicit intriguing effects on brain inflammation which involves microglia and astrocyte activations, along with neuronal damage as the end-point of pathological pathways in bacterial meningitis. As some studies mentioned potential advantage of CDC-targeted therapeutic mechanisms to combat CNS infections, it might be a fruitful avenue to deepen our understanding of CDC as a candidate for adjuvant therapy to combat bacterial meningitis.

    Citation: Tjokorda Istri Pramitasuri, Ni Made Susilawathi, Ni Made Adi Tarini, AA Raka Sudewi, Matthew C Evans. Cholesterol dependent cytolysins and the brain: Revealing a potential therapeutic avenue for bacterial meningitis[J]. AIMS Microbiology, 2023, 9(4): 647-667. doi: 10.3934/microbiol.2023033

    Related Papers:

  • Bacterial meningitis is a catastrophic nervous system disorder with high mortality and wide range of morbidities. Some of the meningitis-causing bacteria occupy cholesterol dependent cytolysins (CDCs) to increase their pathogenicity and arrange immune-evasion strategy. Studies have observed that the relationship between CDCs and pathogenicity in these meningitides is complex and involves interactions between CDC, blood-brain barrier (BBB), glial cells and neurons. In BBB, these CDCs acts on capillary endothelium, tight junction (TJ) proteins and neurovascular unit (NVU). CDCs also observed to elicit intriguing effects on brain inflammation which involves microglia and astrocyte activations, along with neuronal damage as the end-point of pathological pathways in bacterial meningitis. As some studies mentioned potential advantage of CDC-targeted therapeutic mechanisms to combat CNS infections, it might be a fruitful avenue to deepen our understanding of CDC as a candidate for adjuvant therapy to combat bacterial meningitis.



    加载中

    Acknowledgments



    TIP received scholarship which supports all of her academic works, including the publication of this manuscript from the Directorate of Resources, Directorate General of Higher Education, Ministry of Education, Culture, Research, and Technology, the Republic of Indonesia through Pendidikan Magister Menuju Doktor untuk Sarjana Unggul (PMDSU) scheme. The author thanked Professor Andrew S.C. Rice, MB BS, MD, FRCP, FRCA, FFPMRCA for overall supervision in the process of constructing this manuscript.

    Conflict of interest



    The authors declare that there were no financial or commercial ties that might be viewed as potential conflicts of interest during the conduct of this manuscript.

    Author contributions



    TIP, MCE, NMS, NMAT and AARS were involved in overall structure and elaboration of concepts for review. Manuscript writing by TIP and MCE. All authors contributed to editing of manuscript.

    [1] Dzupova O, Rozsypal H, Prochazka B, et al. (2009) Acute bacterial meningitis in adults: predictors of outcome. Scand J Infect Dis 41: 348-354. https://doi.org/10.1080/00365540902849391
    [2] Roed C, Omland LH, Engsig FN, et al. (2010) Long-term mortality in patients diagnosed with meningococcal disease: a Danish nationwide cohort study. PLoS One 5: e9662. https://doi.org/10.1371/journal.pone.0009662
    [3] van de Beek D, Cabellos C, Dzupova O, et al. (2016) ESCMID guideline: diagnosis and treatment of acute bacterial meningitis. Clin Microbiol Infect 22 Suppl 3: S37-62. https://doi.org/10.1016/j.cmi.2016.01.007
    [4] Brouwer MC, van de Beek D (2018) Epidemiology of community-acquired bacterial meningitis. Curr Opin Infect Dis 31: 78-84. https://doi.org/10.1097/QCO.0000000000000417
    [5] Castelblanco RL, Lee M, Hasbun R (2014) Epidemiology of bacterial meningitis in the USA from 1997 to 2010: a population-based observational study. Lancet Infect Dis 14: 813-819. https://doi.org/10.1016/S1473-3099(14)70805-9
    [6] Paradowska-Stankiewicz I, Piotrowska A (2017) Meningitis and encephalitis in Poland in 2015. Przegl Epidemiol 71: 493-500.
    [7] van de Beek D (2012) Progress and challenges in bacterial meningitis. Lancet 380: 1623-1624. https://doi.org/10.1016/S0140-6736(12)61808-X
    [8] Luksic I, Mulic R, Falconer R, et al. (2013) Estimating global and regional morbidity from acute bacterial meningitis in children: assessment of the evidence. Croat Med J 54: 510-518. https://doi.org/10.3325/cmj.2013.54.510
    [9] Snoek L, Goncalves BP, Horvath-Puho E, et al. (2022) Short-term and long-term risk of mortality and neurodevelopmental impairments after bacterial meningitis during infancy in children in Denmark and the Netherlands: a nationwide matched cohort study. Lancet Child Adolesc Health 6: 633-642. https://doi.org/10.1016/S2352-4642(22)00155-9
    [10] Kerdsin A, Segura M, Fittipaldi N, et al. (2022) Sociocultural factors influencing human Streptococcus suis disease in Southeast Asia. Foods 11. https://doi.org/10.3390/foods11091190
    [11] McGill F, Heyderman RS, Panagiotou S, et al. (2016) Acute bacterial meningitis in adults. Lancet 388: 3036-3047. https://doi.org/10.1016/S0140-6736(16)30654-7
    [12] Iacovache I, Bischofberger M, van der Goot FG (2010) Structure and assembly of pore-forming proteins. Curr Opin Struct Biol 20: 241-246. https://doi.org/10.1016/j.sbi.2010.01.013
    [13] Lucas R, Czikora I, Sridhar S, et al. (2013) Mini-review: novel therapeutic strategies to blunt actions of pneumolysin in the lungs. Toxins (Basel) 5: 1244-1260. https://doi.org/10.3390/toxins5071244
    [14] Braun JS, Hoffmann O, Schickhaus M, et al. (2007) Pneumolysin causes neuronal cell death through mitochondrial damage. Infect Immun 75: 4245-4254. https://doi.org/10.1128/IAI.00031-07
    [15] Hupp S, Grandgirard D, Mitchell TJ, et al. (2019) Pneumolysin and the bacterial capsule of Streptococcus pneumoniae cooperatively inhibit taxis and motility of microglia. J Neuroinflammation 16: 105. https://doi.org/10.1186/s12974-019-1491-7
    [16] Chen S, Xie W, Wu K, et al. (2016) Suilysin stimulates the release of heparin binding protein from neutrophils and increases vascular permeability in mice. Front Microbiol 7: 1338. https://doi.org/10.3389/fmicb.2016.01338
    [17] Doran KS, Liu GY, Nizet V (2003) Group B streptococcal beta-hemolysin/cytolysin activates neutrophil signaling pathways in brain endothelium and contributes to development of meningitis. J Clin Invest 112: 736-744. https://doi.org/10.1172/JCI200317335
    [18] Pinkney M, Kapur V, Smith J, et al. (1995) Different forms of streptolysin O produced by Streptococcus pyogenes and by Escherichia coli expressing recombinant toxin: cleavage by streptococcal cysteine protease. Infect Immun 63: 2776-2779. https://doi.org/10.1128/iai.63.7.2776-2779.1995
    [19] Chen C, Nguyen BN, Mitchell G, et al. (2018) The listeriolysin O PEST-like sequence co-opts AP-2-mediated endocytosis to prevent plasma membrane damage during Listeria lnfection. Cell Host Microbe 23: 786-795 e785. https://doi.org/10.1016/j.chom.2018.05.006
    [20] Almutairi MM, Gong C, Xu YG, et al. (2016) Factors controlling permeability of the blood-brain barrier. Cell Mol Life Sci 73: 57-77. https://doi.org/10.1007/s00018-015-2050-8
    [21] Bello C, Smail Y, Sainte-Rose V, et al. (2020) Role of astroglial Connexin 43 in pneumolysin cytotoxicity and during pneumococcal meningitis. PLoS Pathog 16: e1009152. https://doi.org/10.1371/journal.ppat.1009152
    [22] Bischofberger M, Gonzalez MR, van der Goot FG (2009) Membrane injury by pore-forming proteins. Curr Opin Cell Biol 21: 589-595. https://doi.org/10.1016/j.ceb.2009.04.003
    [23] Block ML, Hong JS (2005) Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol 76: 77-98. https://doi.org/10.1016/j.pneurobio.2005.06.004
    [24] Kim JY, Paton JC, Briles DE, et al. (2015) Streptococcus pneumoniae induces pyroptosis through the regulation of autophagy in murine microglia. Oncotarget 6: 44161-44178. https://doi.org/10.18632/oncotarget.6592
    [25] Fortsch C, Hupp S, Ma J, et al. (2011) Changes in astrocyte shape induced by sublytic concentrations of the cholesterol-dependent cytolysin pneumolysin still require pore-forming capacity. Toxins (Basel) 3: 43-62. https://doi.org/10.3390/toxins3010043
    [26] Hupp S, Fortsch C, Graber F, et al. (2022) Pneumolysin boosts the neuroinflammatory response to Streptococcus pneumoniae through enhanced endocytosis. Nat Commun 13: 5032. https://doi.org/10.1038/s41467-022-32624-2
    [27] Hupp S, Heimeroth V, Wippel C, et al. (2012) Astrocytic tissue remodeling by the meningitis neurotoxin pneumolysin facilitates pathogen tissue penetration and produces interstitial brain edema. Glia 60: 137-146. https://doi.org/10.1002/glia.21256
    [28] Hupp S, Ribes S, Seele J, et al. (2017) Magnesium therapy improves outcome in Streptococcus pneumoniae meningitis by altering pneumolysin pore formation. Br J Pharmacol 174: 4295-4307. https://doi.org/10.1111/bph.14027
    [29] Iliev AI, Djannatian JR, Nau R, et al. (2007) Cholesterol-dependent actin remodeling via RhoA and Rac1 activation by the Streptococcus pneumoniae toxin pneumolysin. Proc Natl Acad Sci U S A 104: 2897-2902. https://doi.org/10.1073/pnas.0608213104
    [30] Muller A, Lekhuleni C, Hupp S, et al. (2022) Meningitis-associated pneumococcal serotype 8, ST 53, strain is hypervirulent in a rat model and has non-haemolytic pneumolysin which can be attenuated by liposomes. Front Cell Infect Microbiol 12: 1106063. https://doi.org/10.3389/fcimb.2022.1106063
    [31] Reiss A, Braun JS, Jager K, et al. (2011) Bacterial pore-forming cytolysins induce neuronal damage in a rat model of neonatal meningitis. J Infect Dis 203: 393-400. https://doi.org/10.1093/infdis/jiq047
    [32] Wippel C, Fortsch C, Hupp S, et al. (2011) Extracellular calcium reduction strongly increases the lytic capacity of pneumolysin from streptococcus pneumoniae in brain tissue. J Infect Dis 204: 930-936. https://doi.org/10.1093/infdis/jir434
    [33] Wippel C, Maurer J, Fortsch C, et al. (2013) Bacterial cytolysin during meningitis disrupts the regulation of glutamate in the brain, leading to synaptic damage. PLoS Pathog 9: e1003380. https://doi.org/10.1371/journal.ppat.1003380
    [34] Tabusi M, Thorsdottir S, Lysandrou M, et al. (2021) Neuronal death in pneumococcal meningitis is triggered by pneumolysin and RrgA interactions with beta-actin. PLoS Pathog 17: e1009432. https://doi.org/10.1371/journal.ppat.1009432
    [35] Zysk G, Schneider-Wald BK, Hwang JH, et al. (2001) Pneumolysin is the main inducer of cytotoxicity to brain microvascular endothelial cells caused by Streptococcus pneumoniae. Infect Immun 69: 845-852. https://doi.org/10.1128/IAI.69.2.845-852.2001
    [36] Braun JS, Sublett JE, Freyer D, et al. (2002) Pneumococcal pneumolysin and H(2)O(2) mediate brain cell apoptosis during meningitis. J Clin Invest 109: 19-27. https://doi.org/10.1172/JCI12035
    [37] Charland N, Nizet V, Rubens CE, et al. (2000) Streptococcus suis serotype 2 interactions with human brain microvascular endothelial cells. Infect Immun 68: 637-643. https://doi.org/10.1128/IAI.68.2.637-643.2000
    [38] Lv Q, Hao H, Bi L, et al. (2014) Suilysin remodels the cytoskeletons of human brain microvascular endothelial cells by activating RhoA and Rac1 GTPase. Protein Cell 5: 261-264. https://doi.org/10.1007/s13238-014-0037-0
    [39] Tenenbaum T, Adam R, Eggelnpohler I, et al. (2005) Strain-dependent disruption of blood-cerebrospinal fluid barrier by Streptoccocus suis in vitro. FEMS Immunol Med Microbiol 44: 25-34. https://doi.org/10.1016/j.femsim.2004.12.006
    [40] Vanier G, Segura M, Friedl P, et al. (2004) Invasion of porcine brain microvascular endothelial cells by Streptococcus suis serotype 2. Infect Immun 72: 1441-1449. https://doi.org/10.1128/IAI.72.3.1441-1449.2004
    [41] Zheng H, Punaro MC, Segura M, et al. (2011) Toll-like receptor 2 is partially involved in the activation of murine astrocytes by Streptococcus suis, an important zoonotic agent of meningitis. J Neuroimmunol 234: 71-83. https://doi.org/10.1016/j.jneuroim.2011.02.005
    [42] Sui Y, Chen Y, Lv Q, et al. (2022) Suilyin disrupts the blood-brain barrier by activating group III secretory phospholipase A2. Life (Basel) 12. https://doi.org/10.3390/life12060919
    [43] Seele J, Tauber SC, Bunkowski S, et al. (2018) The inflammatory response and neuronal injury in Streptococcus suis meningitis. BMC Infect Dis 18: 297. https://doi.org/10.1186/s12879-018-3206-6
    [44] Nizet V, Kim KS, Stins M, et al. (1997) Invasion of brain microvascular endothelial cells by group B streptococci. Infect Immun 65: 5074-5081. https://doi.org/10.1128/iai.65.12.5074-5081.1997
    [45] Liu YH, Wu PH, Kang CC, et al. (2019) Group A streptococcus subcutaneous infection-induced central nervous system inflammation is attenuated by blocking peripheral TNF. Front Microbiol 10: 265. https://doi.org/10.3389/fmicb.2019.00265
    [46] Goldmann O, Sastalla I, Wos-Oxley M, et al. (2009) Streptococcus pyogenes induces oncosis in macrophages through the activation of an inflammatory programmed cell death pathway. Cell Microbiol 11: 138-155. https://doi.org/10.1111/j.1462-5822.2008.01245.x
    [47] Hugo F, Reichwein J, Arvand M, et al. (1986) Use of a monoclonal antibody to determine the mode of transmembrane pore formation by streptolysin O. Infect Immun 54: 641-645. https://doi.org/10.1128/iai.54.3.641-645.1986
    [48] Zhang M, Gillaspy AF, Gipson JR, et al. (2018) Neuroinvasive Listeria monocytogenes infection triggers IFN-activation of microglia and upregulates microglial miR-155. Front Immunol 9: 2751. https://doi.org/10.3389/fimmu.2018.02751
    [49] Bukrinsky MI, Mukhamedova N, Sviridov D (2020) Lipid rafts and pathogens: the art of deception and exploitation. J Lipid Res 61: 601-610. https://doi.org/10.1194/jlr.TR119000391
    [50] Farrand AJ, LaChapelle S, Hotze EM, et al. (2010) Only two amino acids are essential for cytolytic toxin recognition of cholesterol at the membrane surface. Proc Natl Acad Sci U S A 107: 4341-4346. https://doi.org/10.1073/pnas.0911581107
    [51] Giddings KS, Zhao J, Sims PJ, et al. (2004) Human CD59 is a receptor for the cholesterol-dependent cytolysin intermedilysin. Nat Struct Mol Biol 11: 1173-1178. https://doi.org/10.1038/nsmb862
    [52] Pulzova L, Bhide MR, Andrej K (2009) Pathogen translocation across the blood-brain barrier. FEMS Immunol Med Microbiol 57: 203-213. https://doi.org/10.1111/j.1574-695X.2009.00594.x
    [53] Zhao Y, Gan L, Ren L, et al. (2022) Factors influencing the blood-brain barrier permeability. Brain Res 1788: 147937. https://doi.org/10.1016/j.brainres.2022.147937
    [54] Anil A, Banerjee A (2020) Pneumococcal encounter with the blood-brain barrier endothelium. Front Cell Infect Microbiol 10: 590682. https://doi.org/10.3389/fcimb.2020.590682
    [55] Giovannoni F, Quintana FJ (2020) The role of astrocytes in CNS inflammation. Trends Immunol 41: 805-819. https://doi.org/10.1016/j.it.2020.07.007
    [56] Patil V, Bohara R, Winter C, et al. (2023) An insight into new glycotherapeutics in glial inflammation: Understanding the role of glycosylation in mitochondrial function and acute to the chronic phases of inflammation. CNS Neurosci Ther 29: 429-444. https://doi.org/10.1111/cns.14016
    [57] van Sorge NM, Doran KS (2012) Defense at the border: the blood-brain barrier versus bacterial foreigners. Future Microbiol 7: 383-394. https://doi.org/10.2217/fmb.12.1
    [58] Wolburg H, Lippoldt A (2002) Tight junctions of the blood-brain barrier: development, composition and regulation. Vascul Pharmacol 38: 323-337. https://doi.org/10.1016/S1537-1891(02)00200-8
    [59] Fassbender K, Schminke U, Ries S, et al. (1997) Endothelial-derived adhesion molecules in bacterial meningitis: association to cytokine release and intrathecal leukocyte-recruitment. J Neuroimmunol 74: 130-134. https://doi.org/10.1016/S0165-5728(96)00214-7
    [60] Al-Numani D, Segura M, Dore M, et al. (2003) Up-regulation of ICAM-1, CD11a/CD18 and CD11c/CD18 on human THP-1 monocytes stimulated by Streptococcus suis serotype 2. Clin Exp Immunol 133: 67-77. https://doi.org/10.1046/j.1365-2249.2003.02189.x
    [61] Springer TA (1994) Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell 76: 301-314. https://doi.org/10.1016/0092-8674(94)90337-9
    [62] Weber JR, Angstwurm K, Burger W, et al. (1995) Anti ICAM-1 (CD 54) monoclonal antibody reduces inflammatory changes in experimental bacterial meningitis. J Neuroimmunol 63: 63-68. https://doi.org/10.1016/0165-5728(95)00131-X
    [63] Twaddell SH, Baines KJ, Grainge C, et al. (2019) The emerging role of neutrophil extracellular traps in respiratory disease. Chest 156: 774-782. https://doi.org/10.1016/j.chest.2019.06.012
    [64] Pramitasuri TI, Laksmidewi A, Putra IBK, et al. (2021) Neutrophil extracellular traps in coronavirus disease-19-associated ischemic stroke: a novel avenue in neuroscience. Exp Neurobiol 30: 1-12. https://doi.org/10.5607/en20048
    [65] Nel JG, Theron AJ, Durandt C, et al. (2016) Pneumolysin activates neutrophil extracellular trap formation. Clin Exp Immunol 184: 358-367. https://doi.org/10.1111/cei.12766
    [66] Bonilla MC, Quiros ON, Wendt M, et al. (2022) New insights into neutrophil extracellular trap (NETs) formation from porcine neutrophils in response to bacterial infections. Int J Mol Sci 23. https://doi.org/10.3390/ijms23168953
    [67] Caudrillier A, Kessenbrock K, Gilliss BM, et al. (2012) Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J Clin Invest 122: 2661-2671. https://doi.org/10.1172/JCI61303
    [68] Elaskalani O, Abdol Razak NB, Metharom P (2018) Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones. Cell Commun Signal 16: 24. https://doi.org/10.1186/s12964-018-0235-0
    [69] Letsiou E, Teixeira Alves LG, Felten M, et al. (2021) Neutrophil-derived extracellular vesicles activate platelets after pneumolysin exposure. Cells 10. https://doi.org/10.3390/cells10123581
    [70] Hawkins RA, O'Kane RL, Simpson IA, et al. (2006) Structure of the blood-brain barrier and its role in the transport of amino acids. J Nutr 136: 218S-226S. https://doi.org/10.1093/jn/136.1.218S
    [71] Malley R, Henneke P, Morse SC, et al. (2003) Recognition of pneumolysin by Toll-like receptor 4 confers resistance to pneumococcal infection. Proc Natl Acad Sci U S A 100: 1966-1971. https://doi.org/10.1073/pnas.0435928100
    [72] Yoo IH, Shin HS, Kim YJ, et al. (2010) Role of pneumococcal pneumolysin in the induction of an inflammatory response in human epithelial cells. FEMS Immunol Med Microbiol 60: 28-35. https://doi.org/10.1111/j.1574-695X.2010.00699.x
    [73] Hackett SP, Stevens DL (1992) Streptococcal toxic shock syndrome: synthesis of tumor necrosis factor and interleukin-1 by monocytes stimulated with pyrogenic exotoxin A and streptolysin O. J Infect Dis 165: 879-885. https://doi.org/10.1093/infdis/165.5.879
    [74] Stassen M, Muller C, Richter C, et al. (2003) The streptococcal exotoxin streptolysin O activates mast cells to produce tumor necrosis factor alpha by p38 mitogen-activated protein kinase- and protein kinase C-dependent pathways. Infect Immun 71: 6171-6177. https://doi.org/10.1128/IAI.71.11.6171-6177.2003
    [75] Engelhardt S, Al-Ahmad AJ, Gassmann M, et al. (2014) Hypoxia selectively disrupts brain microvascular endothelial tight junction complexes through a hypoxia-inducible factor-1 (HIF-1) dependent mechanism. J Cell Physiol 229: 1096-1105. https://doi.org/10.1002/jcp.24544
    [76] Kim KW, Lee SJ, Kim JC (2017) TNF-alpha upregulates HIF-1alpha expression in pterygium fibroblasts and enhances their susceptibility to VEGF independent of hypoxia. Exp Eye Res 164: 74-81. https://doi.org/10.1016/j.exer.2017.08.008
    [77] Miao Z, Dong Y, Fang W, et al. (2014) VEGF increases paracellular permeability in brain endothelial cells via upregulation of EphA2. Anat Rec (Hoboken) 297: 964-972. https://doi.org/10.1002/ar.22878
    [78] Page S, Munsell A, Al-Ahmad AJ (2016) Cerebral hypoxia/ischemia selectively disrupts tight junctions complexes in stem cell-derived human brain microvascular endothelial cells. Fluids Barriers CNS 13: 16. https://doi.org/10.1186/s12987-016-0042-1
    [79] Zhang Y, Ding X, Miao C, et al. (2019) Propofol attenuated TNF-alpha-modulated occludin expression by inhibiting Hif-1alpha/ VEGF/ VEGFR-2/ ERK signaling pathway in hCMEC/D3 cells. BMC Anesthesiol 19: 127. https://doi.org/10.1186/s12871-019-0788-5
    [80] Kago T, Takagi N, Date I, et al. (2006) Cerebral ischemia enhances tyrosine phosphorylation of occludin in brain capillaries. Biochem Biophys Res Commun 339: 1197-1203. https://doi.org/10.1016/j.bbrc.2005.11.133
    [81] Kim KA, Kim D, Kim JH, et al. (2020) Autophagy-mediated occludin degradation contributes to blood-brain barrier disruption during ischemia in bEnd.3 brain endothelial cells and rat ischemic stroke models. Fluids Barriers CNS 17: 21. https://doi.org/10.1186/s12987-020-00182-8
    [82] Yuan S, Liu KJ, Qi Z (2020) Occludin regulation of blood-brain barrier and potential therapeutic target in ischemic stroke. Brain Circ 6: 152-162. https://doi.org/10.4103/bc.bc_29_20.
    [83] Hawkins BT, Davis TP (2005) The blood-brain barrier/neurovascular unit in health and disease. Pharmacol Rev 57: 173-185. https://doi.org/10.1124/pr.57.2.4
    [84] Maekawa M, Ishizaki T, Boku S, et al. (1999) Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science 285: 895-898. https://doi.org/10.1126/science.285.5429.895
    [85] Ohashi K, Nagata K, Maekawa M, et al. (2000) Rho-associated kinase ROCK activates LIM-kinase 1 by phosphorylation at threonine 508 within the activation loop. J Biol Chem 275: 3577-3582. https://doi.org/10.1074/jbc.275.5.3577
    [86] Madaule P, Eda M, Watanabe N, et al. (1998) Role of citron kinase as a target of the small GTPase Rho in cytokinesis. Nature 394: 491-494. https://doi.org/10.1038/28873
    [87] Chesarone MA, DuPage AG, Goode BL (2010) Unleashing formins to remodel the actin and microtubule cytoskeletons. Nat Rev Mol Cell Biol 11: 62-74. https://doi.org/10.1038/nrm2816
    [88] Chesarone MA, Goode BL (2009) Actin nucleation and elongation factors: mechanisms and interplay. Curr Opin Cell Biol 21: 28-37. https://doi.org/10.1016/j.ceb.2008.12.001
    [89] Gil E, Wall E, Noursadeghi M, et al. (2022) Streptococcus pneumoniae meningitis and the CNS barriers. Front Cell Infect Microbiol 12: 1106596. https://doi.org/10.3389/fcimb.2022.1106596
    [90] Glass CK, Saijo K, Winner B, et al. (2010) Mechanisms underlying inflammation in neurodegeneration. Cell 140: 918-934. https://doi.org/10.1016/j.cell.2010.02.016
    [91] Karin M (2005) Inflammation-activated protein kinases as targets for drug development. Proc Am Thorac Soc 2: 386-390; discussion 394-385. https://doi.org/10.1513/pats.200504-034SR
    [92] Michels M, Vieira AS, Vuolo F, et al. (2015) The role of microglia activation in the development of sepsis-induced long-term cognitive impairment. Brain, Behavior, and Immunity 43: 54-59. https://doi.org/10.1016/j.bbi.2014.07.002
    [93] Kinsner A, Pilotto V, Deininger S, et al. (2005) Inflammatory neurodegeneration induced by lipoteichoic acid from Staphylococcus aureus is mediated by glia activation, nitrosative and oxidative stress, and caspase activation. J Neurochem 95: 1132-1143. https://doi.org/10.1111/j.1471-4159.2005.03422.x
    [94] Dessing MC, Hirst RA, de Vos AF, et al. (2009) Role of Toll-like receptors 2 and 4 in pulmonary inflammation and injury induced by pneumolysin in mice. PLoS One 4: e7993. https://doi.org/10.1371/journal.pone.0007993
    [95] Ebert S, Gerber J, Bader S, et al. (2005) Dose-dependent activation of microglial cells by Toll-like receptor agonists alone and in combination. J Neuroimmunol 159: 87-96. https://doi.org/10.1016/j.jneuroim.2004.10.005
    [96] Bi L, Pian Y, Chen S, et al. (2015) Toll-like receptor 4 confers inflammatory response to Suilysin. Front Microbiol 6: 644. https://doi.org/10.3389/fmicb.2015.00644
    [97] Zhang S, Zheng Y, Chen S, et al. (2016) Suilysin-induced platelet-neutrophil complexes formation is triggered by pore formation-dependent calcium influx. Sci Rep 6: 36787. https://doi.org/10.1038/srep36787
    [98] Park JM, Ng VH, Maeda S, et al. (2004) Anthrolysin O and other gram-positive cytolysins are toll-like receptor 4 agonists. J Exp Med 200: 1647-1655. https://doi.org/10.1084/jem.20041215
    [99] Cui W, Sun C, Ma Y, et al. (2020) Inhibition of TLR4 induces M2 microglial polarization and provides neuroprotection via the NLRP3 inflammasome in Alzheimer's disease. Front Neurosci 14: 444. https://doi.org/10.3389/fnins.2020.00444
    [100] Muhammad T, Ikram M, Ullah R, et al. (2019) Hesperetin, a citrus flavonoid, attenuates LPS-induced neuroinflammation, apoptosis and memory impairments by modulating TLR4/NF-kappaB signaling. Nutrients 11. https://doi.org/10.3390/nu11030648
    [101] Tao L, Zhang L, Gao R, et al. (2018) Andrographolide alleviates acute brain injury in a rat model of traumatic brain injury: possible involvement of inflammatory signaling. Front Neurosci 12: 657. https://doi.org/10.3389/fnins.2018.00657
    [102] Wachholz S, Esslinger M, Plumper J, et al. (2016) Microglia activation is associated with IFN-alpha induced depressive-like behavior. Brain Behav Immun 55: 105-113. https://doi.org/10.1016/j.bbi.2015.09.016
    [103] Shewell LK, Day CJ, Jen FE, et al. (2020) All major cholesterol-dependent cytolysins use glycans as cellular receptors. Sci Adv 6: eaaz4926. https://doi.org/10.1126/sciadv.aaz4926
    [104] Brambilla R, Bracchi-Ricard V, Hu WH, et al. (2005) Inhibition of astroglial nuclear factor kappaB reduces inflammation and improves functional recovery after spinal cord injury. J Exp Med 202: 145-156. https://doi.org/10.1084/jem.20041918
    [105] Brambilla R, Persaud T, Hu X, et al. (2009) Transgenic inhibition of astroglial NF-kappa B improves functional outcome in experimental autoimmune encephalomyelitis by suppressing chronic central nervous system inflammation. J Immunol 182: 2628-2640. https://doi.org/10.4049/jimmunol.0802954
    [106] Carbone DL, Popichak KA, Moreno JA, et al. (2009) Suppression of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced nitric-oxide synthase 2 expression in astrocytes by a novel diindolylmethane analog protects striatal neurons against apoptosis. Mol Pharmacol 75: 35-43. https://doi.org/10.1124/mol.108.050781
    [107] Hamby ME, Hewett JA, Hewett SJ (2006) TGF-beta1 potentiates astrocytic nitric oxide production by expanding the population of astrocytes that express NOS-2. Glia 54: 566-577. https://doi.org/10.1002/glia.20411
    [108] Liu X, Sullivan KA, Madl JE, et al. (2006) Manganese-induced neurotoxicity: the role of astroglial-derived nitric oxide in striatal interneuron degeneration. Toxicol Sci 91: 521-531. https://doi.org/10.1093/toxsci/kfj150
    [109] Takano T, Oberheim N, Cotrina ML, et al. (2009) Astrocytes and ischemic injury. Stroke 40: S8-12. https://doi.org/10.1161/STROKEAHA.108.533166
    [110] Parpura V, Heneka MT, Montana V, et al. (2012) Glial cells in (patho)physiology. J Neurochem 121: 4-27. https://doi.org/10.1111/j.1471-4159.2012.07664.x
    [111] Silver J, Miller JH (2004) Regeneration beyond the glial scar. Nat Rev Neurosci 5: 146-156. https://doi.org/10.1038/nrn1326
    [112] Hirsch EC, Hunot S (2009) Neuroinflammation in Parkinson's disease: a target for neuroprotection?. Lancet Neurol 8: 382-397. https://doi.org/10.1016/S1474-4422(09)70062-6
    [113] Olney JW (1969) Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate. Science 164: 719-721. https://doi.org/10.1126/science.164.3880.719
    [114] Farmen K, Tofino-Vian M, Iovino F (2021) Neuronal damage and neuroinflammation, a bridge between bacterial meningitis and neurodegenerative diseases. Front Cell Neurosci 15: 680858. https://doi.org/10.3389/fncel.2021.680858
    [115] Braun JS, Novak R, Murray PJ, et al. (2001) Apoptosis-inducing factor mediates microglial and neuronal apoptosis caused by pneumococcus. J Infect Dis 184: 1300-1309. https://doi.org/10.1086/324013
    [116] Azzone GF, Azzi A (1965) Volume changes induced by inorganic phosphate in liver mitochondria. Biochem J 94: 10C-11C. https://doi.org/10.1042/bj0940010C
    [117] Celsi F, Pizzo P, Brini M, et al. (2009) Mitochondria, calcium and cell death: a deadly triad in neurodegeneration. Biochim Biophys Acta 1787: 335-344. https://doi.org/10.1016/j.bbabio.2009.02.021
    [118] Szalai G, Krishnamurthy R, Hajnoczky G (1999) Apoptosis driven by IP(3)-linked mitochondrial calcium signals. EMBO J 18: 6349-6361. https://doi.org/10.1093/emboj/18.22.6349
    [119] Takeuchi H, Mizuno T, Zhang G, et al. (2005) Neuritic beading induced by activated microglia is an early feature of neuronal dysfunction toward neuronal death by inhibition of mitochondrial respiration and axonal transport. J Biol Chem 280: 10444-10454. https://doi.org/10.1074/jbc.M413863200
    [120] Krantic S, Mechawar N, Reix S, et al. (2007) Apoptosis-inducing factor: a matter of neuron life and death. Prog Neurobiol 81: 179-196. https://doi.org/10.1016/j.pneurobio.2006.12.002
    [121] Zhu C, Wang X, Huang Z, et al. (2007) Apoptosis-inducing factor is a major contributor to neuronal loss induced by neonatal cerebral hypoxia-ischemia. Cell Death Differ 14: 775-784. https://doi.org/10.1038/sj.cdd.4402053
    [122] Salha D, Szeto J, Myers L, et al. (2012) Neutralizing antibodies elicited by a novel detoxified pneumolysin derivative, PlyD1, provide protection against both pneumococcal infection and lung injury. Infect Immun 80: 2212-2220. https://doi.org/10.1128/IAI.06348-11
    [123] Allen AG, Bolitho S, Lindsay H, et al. (2001) Generation and characterization of a defined mutant of Streptococcus suis lacking suilysin. Infect Immun 69: 2732-2735. https://doi.org/10.1128/IAI.69.4.2732-2735.2001
    [124] Asano K, Sashinami H, Osanai A, et al. (2016) Passive immunization with anti-ActA and anti-listeriolysin O antibodies protects against Listeria monocytogenes infection in mice. Sci Rep 6: 39628. https://doi.org/10.1038/srep39628
    [125] Nato F, Reich K, Lhopital S, et al. (1991) Production and characterization of neutralizing and nonneutralizing monoclonal antibodies against listeriolysin O. Infect Immun 59: 4641-4646. https://doi.org/10.1128/iai.59.12.4641-4646.1991
    [126] Phelps CC, Vadia S, Boyaka PN, et al. (2020) A listeriolysin O subunit vaccine is protective against Listeria monocytogenes. Vaccine 38: 5803-5813. https://doi.org/10.1016/j.vaccine.2020.06.049
    [127] Besancon H, Babiychuk V, Larpin Y, et al. (2021) Tailored liposomal nanotraps for the treatment of Streptococcal infections. J Nanobiotechnology 19: 46. https://doi.org/10.1186/s12951-021-00775-x
    [128] Subramanian K, Iovino F, Tsikourkitoudi V, et al. (2020) Mannose receptor-derived peptides neutralize pore-forming toxins and reduce inflammation and development of pneumococcal disease. EMBO Mol Med 12: e12695. https://doi.org/10.15252/emmm.202012695
    [129] Li G, Wang G, Si X, et al. (2019) Inhibition of suilysin activity and inflammation by myricetin attenuates Streptococcus suis virulence. Life Sci 223: 62-68. https://doi.org/10.1016/j.lfs.2019.03.024
    [130] Li G, Lu G, Qi Z, et al. (2017) Morin attenuates Streptococcus suis pathogenicity in Mice by neutralizing suilysin activity. Front Microbiol 8: 460. https://doi.org/10.3389/fmicb.2017.00460
    [131] Henry BD, Neill DR, Becker KA, et al. (2015) Engineered liposomes sequester bacterial exotoxins and protect from severe invasive infections in mice. Nat Biotechnol 33: 81-88. https://doi.org/10.1038/nbt.3037
  • Reader Comments
  • © 2023 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1422) PDF downloads(219) Cited by(0)

Article outline

Figures and Tables

Figures(2)  /  Tables(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog