HEMOKINI I HEMOKINSKI RECEPTORI U PATOGENEZI MULTIPLE SKLEROZE

  • Ljiljana Stojković Institut za nuklearne nauke “Vinča”, Laboratorija za radiobiologiju i molekularnu genetiku

Sažetak


Migracija leukocita u tkivo centralnog nervnog sistema (CNS) je jedan od procesa koji karakteriše patogenezu multiple skleroze (MS), a stimulisana je od strane hemokina. Kao konstitutivno eksprimirani u CNS, hemokini CXCL12 i CX3CL1 i njihovi receptori posreduju u uspostavljanju i održavanju tkivne homeostaze, dok su inducibilne promene njihove ekspresije u CNS povezane sa patogenezom MS. Hemokin CCL2, sintetisan od strane aktiviranih glijalnih i endotelnih ćelija u nervnom tkivu zahvaćenom inflamacijom, stimuliše infiltraciju CCR2+ monocita u CNS. Glijalni hemokini regulišu regrutovanje funkcionalno različitih subpopulacija T-limfocita tokom inflamacije u CNS, s obzirom da aktivirane glijalne ćelije unutar plakova sintetišu ligande za receptore koje eksprimiraju proinflamatorni Th1 limfociti (CXCR3 i CCR5) i antiinflamatorni Th2 limfociti (CCR4 i CCR8). Hemokin CCL20, koga produkuju Th17 limfociti i epitel horoidnog pleksusa, indukuje CCR6-posredovanu migraciju limfocita u CNS, što može imati i patogenu i protektivnu ulogu u MS, budući da CCR6 eksprimiraju proinflamatorni Th17 kao i antiinflamatorni regulatorni TREG limfociti. Zreli B-limfociti eksprimiraju receptor za hemokin CXCL13 (CXCR5), te je nivo CXCL13 u likvoru indikativan za intratekalni humoralni imunski odgovor u CNS tokom patogeneze MS. Lekovi koji se primenjuju u terapiji MS mogu da ostvaruju imunomodulatorne efekte preko uticaja na ekspresiju/aktivnost hemokina i hemokinskih receptora. Među aktuelnim terapeuticima teško je izdvojiti pojedinačni koji trajno efikasno suprimira bolest. Jedan od razloga za to leži upravo u kompleksnosti mreže hemokina i hemokinskih receptora, u pogledu njihove brojnosti i višestrukih funkcija koje imaju.

Reference

Compston A, Coles A. Multiple sclerosis. Lancet 2008; 372(9648): 1502-17.

Confavreux C, Vukusic S. Natural history of multiple sclerosis: implications for counselling and therapy. Curr Opin Neurol 2002; 15(3): 257-66.

Hauser SL, Goodin DS. Multiple sclerosis and other demyelinating diseases. In: Braunwald E, Fauci AS, Kasper DL, Hauser SL, Longo DL, Jameson JL. Harrison’s Principles of Internal Medicine. 15th ed. New York City: McGraw-Hill Professional Publishing, 2001: 2452-61.

Feuerstein GZ, Liu T, Barone FC. Cytokines, inflammation, and brain injury: role of tumor necrosis factor-alpha. Cerebrovasc Brain Metab Rev 1994; 6(4): 341-60.

Frohman EM, Racke MK, Raine CS. Multiple sclerosis - the plaque and its pathogenesis. N Engl J Med 2006; 354: 942-55.

Olsson T, Zhi WW, Höjeberg B, et al. Autoreactive T lymphocytes in multiple sclerosis determined by antigen-induced secretion of interferon-gamma. J Clin Invest 1990; 86: 981-5.

Navikas V, Link H. Review: cytokines and the pathogenesis of multiple sclerosis. J Neurosci Res 1996; 45: 322-33.

O’Connor RA, Prendergast CT, Sabatos CA, et al. Cutting edge: Th1 cells facilitate the entry of Th17 cells to the central nervous system during experimental autoimmune encephalomyelitis. J Immunol 2008; 181: 3750-4.

Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 2005; 6(11): 1123-32.

Komiyama Y, Nakae S, Matsuki T, et al. IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol 2006; 177: 566-73.

Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol 1989; 7: 145-73.

McGeachy MJ, Stephens LA, Anderton SM. Natural recovery and protection from autoimmune encephalomyelitis: contribution of CD4+CD25+ regulatory cells within the central nervous system. J Immunol 2005; 175: 3025-32.

Haas J, Hug A, Viehöver A, et al. Reduced suppressive effect of CD4+CD25 high regulatory T cells on the T cell immune response against myelin oligodendrocyte glycoprotein in patients with multiple sclerosis. Eur J Immunol 2005; 35: 3343-52.

Lucchinetti C, Brück W, Parisi J, Scheithauer B, Rodriguez M, Lassmann H. Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol 2000; 47: 707-17.

Qin Y, Duquette P, Zhang Y, Talbot P, Poole R, Antel J. Clonal expansion and somatic hypermutation of V (H) genes of B cells from cerebrospinal fluid in multiple sclerosis. J Clin Invest 1998; 102: 1045-50.

Abbas AK, Lichtman AH. Basic immunology: functions and disorders of the immune system. 2nd ed. Philadelphia: Elsevier Saunders, 2006.

Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol 2000; 18: 217-42.

IUIS/WHO Subcommittee on Chemokine Nomenclature. Chemokine/chemokine receptor nomenclature. J Interferon Cytokine Res 2002; 22(10): 1067-8.

Mantovani A. The chemokine system: redundancy for robust outputs. Immunol Today 1999; 20: 254-7.

Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med 2006; 354: 610-21.

Gerard C, Rollins BJ. Chemokines and disease. Nat Immunol 2001; 2: 108-15.

Ransohoff RM, Hamilton TA, Tani M, et al. Astrocyte expression of mRNA encoding cytokines IP-10 and JE/MCP-1 in experimental autoimmune encephalomyelitis. FASEB J 1993; 7: 592-600.

Réaux-Le Goazigo A, Van Steenwinckel J, Rostène W, Mélik Parsadaniantz S. Current status of chemokines in the adult CNS. Prog Neurobiol 2013; 104: 67-92.

Ambrosini E, Aloisi F. Chemokines and glial cells: a complex network in the central nervous system. Neurochem Res 2004; 29(5): 1017-38.

Holman DW, Klein RS, Ransohoff RM. The blood-brain barrier, chemokines and multiple sclerosis. Biochim Biophys Acta 2011; 1812: 220-30.

Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci 2006; 7: 41-53.

McCandless EE, Wang Q, Woerner BM, Harper JM, Klein RS. CXCL12 limits inflammation by localizing mononuclear infiltrates to the perivascular space during experimental autoimmune encephalomyelitis. J Immunol 2006; 177: 8053-64.

McCandless EE, Piccio L, Woerner BM, et al. Pathological expression of CXCL12 at the blood-brain barrier correlates with severity of multiple sclerosis. Am J Pathol 2008; 172: 799-808.

Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA. A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1). J Exp Med 1996; 184: 1101-9.

Krumbholz M, Theil D, Cepok S, et al. Chemokines in multiple sclerosis: CXCL12 and CXCL13 up-regulation is differentially linked to CNS immune cell recruitment. Brain 2006; 129: 200-11.

Van der Meer P, Ulrich AM, Gonzalez-Scarano F, Lavi E. Immunohistochemical analysis of CCR2, CCR3, CCR5, and CXCR4 in the human brain: potential mechanisms for HIV dementia. Exp Mol Pathol 2000; 69: 192-201.

McCandless EE, Budde M, Lees JR, Dorsey D, Lyng E, Klein RS. IL-1R signaling within the central nervous system regulates CXCL12 expression at the blood-brain barrier and disease severity during experimental autoimmune encephalomyelitis. J Immunol 2009; 183: 613-20.

Pashenkov M, Söderström M, Link H. Secondary lymphoid organ chemokines are elevated in the cerebrospinal fluid during central nervous system inflammation. J Neuroimmunol 2003; 135(1-2): 154-60.

Li M, Ransohoff RM. Multiple roles of chemokine CXCL12 in the central nervous system: a migration from immunology to neurobiology. Prog Neurobiol 2008; 84(2): 116-31.

Miljković D, Stanojević Z, Momcilović M, Odoardi F, Flügel A, Mostarica-Stojković M. CXCL12 expression within the CNS contributes to the resistance against experimental autoimmune encephalomyelitis in Albino Oxford rats. Immunobiology 2011; 216(9): 979-87.

Patel JR, McCandless EE, Dorsey D, Klein RS. CXCR4 promotes differentiation of oligodendrocyte progenitors and remyelination. Proc Natl Acad Sci U S A 2010; 107(24): 11062-7.

Banisadr G, Frederick TJ, Freitag C, et al. The role of CXCR4 signaling in the migration of transplanted oligodendrocyte progenitors into the cerebral white matter. Neurobiol Dis 2011; 44(1): 19-27.

Maciejewski-Lenoir D, Chen S, Feng L, Maki R, Bacon KB. Characterization of fractalkine in rat brain cells: migratory and activation signals for CX3CR1-expressing microglia. J Immunol 1999; 163: 1628-35.

Pan Y, Lloyd C, Zhou H, et al. Neurotactin, a membrane-anchored chemokine upregulated in brain inflammation. Nature 1997; 387(6633): 611-7.

Huang D, Shi FD, Jung S, et al. The neuronal chemokine CX3CL1/fractalkine selectively recruits NK cells that modify experimental autoimmune encephalomyelitis within the central nervous system. FASEB J 2006; 20(7): 896-905.

Kastenbauer S, Koedel U, Wick M, Kieseier BC, Hartung HP, Pfister HW. CSF and serum levels of soluble fractalkine (CX3CL1) in inflammatory diseases of the nervous system. J Neuroimmunol 2003; 137: 210-7.

Sunnemark D, Eltayeb S, Nilsson M, et al. CX3CL1 (fractalkine) and CX3CR1 expression in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis: kinetics and cellular origin. J Neuroinflammation 2005; 2: 17.

Stojković L, Djurić T, Stanković A, et al. The association of V249I and T280M fractalkine receptor haplotypes with disease course of multiple sclerosis. J Neuroimmunol 2012; 245(1-2): 87-92.

Mahad DJ, Ransohoff RM. The role of MCP-1 (CCL2) and CCR2 in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Semin Immunol 2003; 15(1): 23-32.

Karpus WJ, Kennedy KJ. MIP-1alpha and MCP-1 differentially regulate acute and relapsing autoimmune encephalomyelitis as well as Th1/Th2 lymphocyte differentiation. J Leukoc Biol 1997; 62(5): 681-7.

Hagman S, Raunio M, Rossi M, Dastidar P, Elovaara I. Disease-associated inflammatory biomarker profiles in blood in different subtypes of multiple sclerosis: prospective clinical and MRI follow-up study. J Neuroimmunol 2011; 234(1-2): 141-7.

Huang DR, Wang J, Kivisakk P, Rollins BJ, Ransohoff RM. Absence of monocyte chemoattractant protein 1 in mice leads to decreased local macrophage recruitment and antigen-specific T helper cell type 1 immune response in experimental autoimmune encephalomyelitis. J Exp Med 2001; 193(6): 713-26.

Izikson L, Klein RS, Charo IF, Weiner HL, Luster AD. Resistance to experimental autoimmune encephalomyelitis in mice lacking the CC chemokine receptor (CCR)2. J Exp Med 2000; 192(7): 1075-80.

Laborde E, Macsata RW, Meng F, et al. Discovery, optimization, and pharmacological characterization of novel heteroaroylphenylureas antagonists of C-C chemokine ligand 2 function. J Med Chem 2011; 54(6): 1667-81.

Dogan RN, Elhofy A, Karpus WJ. Production of CCL2 by central nervous system cells regulates development of murine experimental autoimmune encephalomyelitis through the recruitment of TNF- and iNOS-expressing macrophages and myeloid dendritic cells. J Immunol 2008; 180(11): 7376-84.

Nakajima H, Fukuda K, Doi Y, et al. Expression of TH1/TH2-related chemokine receptors on peripheral T cells and correlation with clinical disease activity in patients with multiple sclerosis. Eur Neurol 2004; 52(3): 162-8.

Krauthausen M, Saxe S, Zimmermann J, Emrich M, Heneka MT, Müller M. CXCR3 modulates glial accumulation and activation in cuprizone-induced demyelination of the central nervous system. J Neuroinflammation 2014; 11: 109.

Gu SM, Park MH, Yun HM, et al. CCR5 knockout suppresses experimental autoimmune encephalomyelitis in C57BL/6 mice. Oncotarget 2016; 7(13): 15382-93.

Murphy CA, Hoek RM, Wiekowski MT, Lira SA, Sedgwick JD. Interactions between hemopoietically derived TNF and central nervous system-resident glial chemokines underlie initiation of autoimmune inflammation in the brain. J Immunol 2002; 169: 7054-62.

Columba-Cabezas S, Serafini B, Ambrosini E, et al. Induction of macrophage-derived chemokine/CCL22 expression in experimental autoimmune encephalomyelitis and cultured microglia: implications for disease regulation. J Neuroimmunol 2002; 130: 10-21.

Maghazachi AA. G protein-coupled receptors in natural killer cells. J Leukoc Biol 2003; 74: 16-24.

Carlson T, Kroenke M, Rao P, Lane TE, Segal B. The Th17-ELR+ CXC chemokine pathway is essential for the development of central nervous system autoimmune disease. J Exp Med 2008; 205: 811-23.

Mildner A, Schmidt H, Nitsche M, et al. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci 2007; 10: 1544-53.

Shimizu Y, Ota K, Kubo S, et al. Association of Th1/Th2-related chemokine receptors in peripheral T cells with disease activity in patients with multiple sclerosis and neuromyelitis optica. Eur Neurol 2011; 66(2): 91-7.

Uzawa A, Mori M, Hayakawa S, Masuda S, Nomura F, Kuwabara S. Expression of chemokine receptors on peripheral blood lymphocytes in multiple sclerosis and neuromyelitis optica. BMC Neurol 2010; 10: 113.

Ni J, Zhu YN, Zhong XG, et al. The chemokine receptor antagonist, TAK-779, decreased experimental autoimmune encephalomyelitis by reducing inflammatory cell migration into the central nervous system, without affecting T cell function. Br J Pharmacol 2009; 158(8): 2046-56.

Imai T, Hieshima K, Haskell C, et al. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell 1997; 91: 521-30.

Tabata S, Kadowaki N, Kitawaki T, et al. Distribution and kinetics of SR-PSOX/CXCL16 and CXCR6 expression on human dendritic cell subsets and CD4+ T cells. J Leukoc Biol 2005; 77: 777-86.

Blauth K, Zhang X, Chopra M, Rogan S, Markovic-Plese S. The role of fractalkine (CX3CL1) in regulation of CD4(+) cell migration to the central nervous system in patients with relapsing-remitting multiple sclerosis. Clin Immunol 2015; 157(2): 121-32.

Broux B, Pannemans K, Zhang X, et al. CX(3)CR1 drives cytotoxic CD4(+)CD28(-) T cells into the brain of multiple sclerosis patients. J Autoimmun 2012; 38(1): 10-9.

Fukumoto N, Shimaoka T, Fujimura H, et al. Critical roles of CXC chemokine ligand 16/scavenger receptor that binds phosphatidylserine and oxidized lipoprotein in the pathogenesis of both acute and adoptive transfer experimental autoimmune encephalomyelitis. J Immunol 2004; 173(3): 1620-7.

Le Blanc LM, van Lieshout AW, Adema GJ, van Riel PL, Verbeek MM, Radstake TR. CXCL16 is elevated in the cerebrospinal fluid versus serum and in inflammatory conditions with suspected and proved central nervous system involvement. Neurosci Lett 2006; 397(1-2): 145-8.

Stojković L, Stanković A, Djurić T, Dinčić E, Alavantić D, Zivković M. The gender-specific association of CXCL16 A181V gene polymorphism with susceptibility to multiple sclerosis, and its effects on PBMC mRNA and plasma soluble CXCL16 levels: preliminary findings. J Neurol 2014; 261(8): 1544-51.

Sato W, Aranami T, Yamamura T. Cutting edge: Human Th17 cells are identified as bearing CCR2+CCR5− phenotype. J Immunol 2007; 178(12): 7525-9.

Yamazaki T, Yang XO, Chung Y, et al. CCR6 regulates the migration of inflammatory and regulatory T cells. J Immunol 2008; 181(12): 8391-401.

Reboldi A, Coisne C, Baumjohann D, et al. C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol 2009; 10(5): 514-23.

Ambrosini E, Remoli ME, Giacomini E, et al. Astrocytes produce dendritic cell-attracting chemokines in vitro and in multiple sclerosis lesions. J Neuropathol Exp Neurol 2005; 64(8): 706-15.

Liston A, Kohler RE, Townley S, et al. Inhibition of CCR6 function reduces the severity of experimental autoimmune encephalomyelitis via effects on the priming phase of the immune response. J Immunol 2009; 182(5): 3121-30.

Elhofy A, Depaolo RW, Lira SA, Lukacs NW, Karpus WJ. Mice deficient for CCR6 fail to control chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2009; 213(1-2): 91-9.

Alvarez E, Piccio L, Mikesell RJ, et al. CXCL13 is a biomarker of inflammation in multiple sclerosis, neuromyelitis optica, and other neurological conditions. Mult Scler 2013; 19(9): 1204-8.

Khademi M, Kockum I, Andersson ML, et al. Cerebrospinal fluid CXCL13 in multiple sclerosis: a suggestive prognostic marker for the disease course. Mult Scler 2011; 17(3): 335-43.

Kowarik MC, Cepok S, Sellner J, et al. CXCL13 is the major determinant for B cell recruitment to the CSF during neuroinflammation. J Neuroinflammation 2012; 9: 93.

Axelsson M, Mattsson N, Malmestrom C, Zetterberg H, Lycke J. The influence of disease duration, clinical course, and immunosuppressive therapy on the synthesis of intrathecal oligoclonal IgG bands in multiple sclerosis. J Neuroimmunol 2013; 264(1-2): 100-5.

Sloka JS, Stefanelli M. The mechanism of action of methylprednisolone in the treatment of multiple sclerosis. Mult Scler 2005; 11(4): 425-32.

Jalosinski M, Karolczak K, Mazurek A, Glabinski A. The effects of methylprednisolone and mitoxantrone on CCL5-induced migration of lymphocytes in multiple sclerosis. Acta Neurol Scand 2008; 118(2): 120-5.

Michałowska-Wender G, Losy J, Szczuciński A, Biernacka-Łukanty J, Wender M. Effect of methylprednisolone treatment on expression of sPECAM-1 and CXCL10 chemokine in serum of MS patients. Pharmacol Rep 2006; 58(6): 920-3.

Rudick RA, Goodkin DE, Jacobs LD, et al. Impact of interferon beta-1a on neurologic disability in relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG). Neurology 1997; 49(2): 358-63.

Zang YC, Halder JB, Samanta AK, Hong J, Rivera VM, Zhang JZ. Regulation of chemokine receptor CCR5 and production of RANTES and MIP-1alpha by interferon-beta. J Neuroimmunol 2001; 112(1-2): 174-80.

Dhib-Jalbut S, Sumandeep S, Valenzuela R, Ito K, Patel P, Rametta M. Immune response during interferon beta-1b treatment in patients with multiple sclerosis who experienced relapses and those who were relapse-free in the START study. J Neuroimmunol 2013; 254(1-2): 131-40.

Farina C, Weber MS, Meinl E, Wekerle H, Hohlfeld R. Glatiramer acetate in multiple sclerosis: update on potential mechanisms of action. Lancet Neurol 2005; 4(9): 567-75.

Høglund RA, Hestvik AL, Holmøy T, Maghazachi AA. Expression and functional activity of chemokine receptors in glatiramer acetate-specific T cells isolated from multiple sclerosis patient receiving the drug glatiramer acetate. Hum Immunol 2011; 72(2): 124-34.

Objavljeno
2017/03/21
Rubrika
Pregledni članak