Artigo Revisado por pares

Amelioration of Experimental Autoimmune Encephalomyelitis by the Quinoline-3-Carboxamide Paquinimod

2013; Elsevier BV; Volume: 182; Issue: 5 Linguagem: Inglês

10.1016/j.ajpath.2013.01.032

ISSN

1525-2191

Autores

Sofia Helmersson, Anette Sundstedt, Adnan Deronic, Tomas Leanderson, Fredrik Ivars,

Tópico(s)

Immunotherapy and Immune Responses

Resumo

Quinoline-3-carboxamide compounds (Q compounds) have demonstrated efficacy in treating autoimmune disease in both humans and mice. However, the mode of action of these compounds is poorly understood. Here, we show that preventive treatment with the Q compound paquinimod (ABR-215757) during the first 5 days after induction of experimental autoimmune encephalomyelitis is sufficient to significantly ameliorate disease symptoms. Parallel cell-depletion experiments demonstrated that Ly6Chi inflammatory monocytes play an essential role in this phase. The paquinimod-induced amelioration correlated with reduced priming of antigen-specific CD4+ T cells and reduced frequency of IFN-γ– and IL-17–producing cells in draining lymph nodes. Importantly, the treatment did not inhibit T-cell division per se. In mice with established experimental autoimmune encephalomyelitis, the numbers of Ly6Chi CD115+ inflammatory monocytes and CD11b+CD11c+ dendritic cells (DCs) were reduced in spleen, but not in bone marrow or draining lymph nodes of treated mice. Inflammatory monocyte–derived DCs and CD4+ T cells were also reduced in the brain. In contrast, there was no decrease in DC subsets previously shown to be critical for effector CD4+ T-cell development in lymph nodes. Taken together, these data indicate that preventive treatment with paquinimod ameliorates experimental autoimmune encephalomyelitis by reducing effector T-cell priming and, on prolonged treatment, displays a selective effect by decreasing distinct subpopulations of splenic CD11b+ myeloid cells. Quinoline-3-carboxamide compounds (Q compounds) have demonstrated efficacy in treating autoimmune disease in both humans and mice. However, the mode of action of these compounds is poorly understood. Here, we show that preventive treatment with the Q compound paquinimod (ABR-215757) during the first 5 days after induction of experimental autoimmune encephalomyelitis is sufficient to significantly ameliorate disease symptoms. Parallel cell-depletion experiments demonstrated that Ly6Chi inflammatory monocytes play an essential role in this phase. The paquinimod-induced amelioration correlated with reduced priming of antigen-specific CD4+ T cells and reduced frequency of IFN-γ– and IL-17–producing cells in draining lymph nodes. Importantly, the treatment did not inhibit T-cell division per se. In mice with established experimental autoimmune encephalomyelitis, the numbers of Ly6Chi CD115+ inflammatory monocytes and CD11b+CD11c+ dendritic cells (DCs) were reduced in spleen, but not in bone marrow or draining lymph nodes of treated mice. Inflammatory monocyte–derived DCs and CD4+ T cells were also reduced in the brain. In contrast, there was no decrease in DC subsets previously shown to be critical for effector CD4+ T-cell development in lymph nodes. Taken together, these data indicate that preventive treatment with paquinimod ameliorates experimental autoimmune encephalomyelitis by reducing effector T-cell priming and, on prolonged treatment, displays a selective effect by decreasing distinct subpopulations of splenic CD11b+ myeloid cells. Quinoline-3-carboxamides (Q compounds) are currently in clinical development for the treatment of multiple sclerosis,1Comi G. Abramsky O. Arbizu T. Boyko A. Gold R. Havrdová E. Komoly S. Selmaj K. Sharrack B. Filippi M. LAQ/5063 Study GroupOral laquinimod in patients with relapsing-remitting multiple sclerosis: 36-week double-blind active extension of the multi-centre, randomized, double-blind, parallel-group placebo-controlled study.Mult Scler. 2010; 16: 1360-1366Crossref PubMed Scopus (53) Google Scholar, 2Comi G. Pulizzi A. Rovaris M. Abramsky O. Arbizu T. Boiko A. Gold R. Havrdova E. Komoly S. Selmaj K. Sharrack B. Filippi M. LAQ/5062 Study GroupEffect of laquinimod on MRI-monitored disease activity in patients with relapsing-remitting multiple sclerosis: a multicentre, randomised, double-blind, placebo-controlled phase IIb study.Lancet. 2008; 371: 2085-2092Abstract Full Text Full Text PDF PubMed Scopus (252) Google Scholar, 3Polman C. Barkhof F. Sandberg-Wollheim M. Linde A. Nordle O. Nederman T. Laquinimod in Relapsing MS Study GroupTreatment with laquinimod reduces development of active MRI lesions in relapsing MS.Neurology. 2005; 64: 987-991Crossref PubMed Scopus (217) Google Scholar prostate cancer,4Bratt O. Häggman M. Ahlgren G. Nordle O. Björk A. Damber J.E. Open-label, clinical phase I studies of tasquinimod in patients with castration-resistant prostate cancer.Br J Cancer. 2009; 101: 1233-1240Crossref PubMed Scopus (63) Google Scholar systemic lupus erythematosus,5Bengtsson A.A. Sturfelt G. Lood C. Rönnblom L. van Vollenhoven R.F. Axelsson B. Sparre B. Tuvesson H. Ohman M.W. Leanderson T. Pharmacokinetics, tolerability, and preliminary efficacy of paquinimod (ABR-215757), a new quinoline-3-carboxamide derivative: studies in lupus-prone mice and a multicenter, randomized, double-blind, placebo-controlled, repeat-dose, dose-ranging study in patients with systemic lupus erythematosus.Arthritis Rheum. 2012; 64: 1579-1588Crossref PubMed Scopus (87) Google Scholar and systemic sclerosis. These compounds have also shown efficacy in various experimental models of human autoimmune disease,6Björk J. Kleinau S. Paradoxical effects of LS-2616 (linomide) treatment `type II collagen arthritis model in mice.Agents Actions. 1989; 27: 319-321Crossref PubMed Scopus (56) Google Scholar, 7Brunmark C. Runström A. Ohlsson L. Sparre B. Brodin T. Aström M. Hedlund G. The new orally active immunoregulator laquinimod (ABR-215062) effectively inhibits development and relapses of experimental autoimmune encephalomyelitis.J Neuroimmunol. 2002; 130: 163-172Abstract Full Text Full Text PDF PubMed Scopus (162) Google Scholar, 8Gross D.J. Weiss L. Reibstein I. Hedlund G. Dahlén E. Rapoport M.J. Slavin S. The immunomodulator linomide: role in treatment and prevention of autoimmune diabetes mellitus.Int Immunopharmacol. 2001; 1: 1131-1139Crossref PubMed Scopus (22) Google Scholar, 9Runström A. Leanderson T. Ohlsson L. Axelsson B. Inhibition of the development of chronic experimental autoimmune encephalomyelitis by laquinimod (ABR-215062) in IFN-beta k.o. and wild type mice.J Neuroimmunol. 2006; 173: 69-78Abstract Full Text Full Text PDF PubMed Scopus (61) Google Scholar, 10Wegner C. Stadelmann C. Pförtner R. Raymond E. Feigelson S. Alon R. Timan B. Hayardeny L. Brück W. Laquinimod interferes with migratory capacity of T cells and reduces IL-17 levels, inflammatory demyelination and acute axonal damage in mice with experimental autoimmune encephalomyelitis.J Neuroimmunol. 2010; 227: 133-143Abstract Full Text Full Text PDF PubMed Scopus (111) Google Scholar but are unique in that they do not induce immunosuppression.7Brunmark C. Runström A. Ohlsson L. Sparre B. Brodin T. Aström M. Hedlund G. The new orally active immunoregulator laquinimod (ABR-215062) effectively inhibits development and relapses of experimental autoimmune encephalomyelitis.J Neuroimmunol. 2002; 130: 163-172Abstract Full Text Full Text PDF PubMed Scopus (162) Google Scholar, 11Yang J.S. Xu L.Y. Xiao B.G. Hedlund G. Link H. Laquinimod (ABR-215062) suppresses the development of experimental autoimmune encephalomyelitis, modulates the Th1/Th2 balance and induces the Th3 cytokine TGF-beta in Lewis rats.J Neuroimmunol. 2004; 156: 3-9Abstract Full Text Full Text PDF PubMed Scopus (165) Google Scholar We have recently identified the human S100-A9 protein as one molecular target for the Q compound paquinimod (ABR-215757).12Björk P. Björk A. Vogl T. Stenström M. Liberg D. Olsson A. Roth J. Ivars F. Leanderson T. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides.PLoS Biol. 2009; 7: e97Crossref PubMed Scopus (258) Google Scholar The S100-A9 protein was shown to bind both to Toll-like receptor 4 (TLR4) and to the advanced glycosylation end product-specific receptor (RAGE), and paquinimod competitively inhibited both bindings.12Björk P. Björk A. Vogl T. Stenström M. Liberg D. Olsson A. Roth J. Ivars F. Leanderson T. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides.PLoS Biol. 2009; 7: e97Crossref PubMed Scopus (258) Google Scholar The binding of S100-A9 to TLR4 was shown to induce NF-κB activation and cytokine secretion in myeloid cells.13Riva M. Källberg E. Björk P. Hancz D. Vogl T. Roth J. Ivars F. Leanderson T. Induction of nuclear factor-kappaB responses by the S100A9 protein is Toll-like receptor-4-dependent.Immunology. 2012; 137: 172-182Crossref PubMed Scopus (106) Google Scholar Thus, one mode of action of paquinimod in inflammatory settings may be to interfere with S100-A9–induced responses. Experimental autoimmune encephalomyelitis (EAE) is a model of human multiple sclerosis. Disease development in this model is T-cell–dependent and involves the activity of both Th1 and Th17 T cells.14Kroenke M.A. Carlson T.J. Andjelkovic A.V. Segal B.M. IL-12- and IL-23-modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition.J Exp Med. 2008; 205: 1535-1541Crossref PubMed Scopus (493) Google Scholar, 15Lees J.R. Golumbek P.T. Sim J. Dorsey D. Russell J.H. Regional CNS responses to IFN-gamma determine lesion localization patterns during EAE pathogenesis.J Exp Med. 2008; 205: 2633-2642Crossref PubMed Scopus (145) Google Scholar, 16Luger D. Silver P.B. Tang J. Cua D. Chen Z. Iwakura Y. Bowman E.P. Sgambellone N.M. Chan C.C. Caspi R.R. Either a Th17 or a Th1 effector response can drive autoimmunity: conditions of disease induction affect dominant effector category.J Exp Med. 2008; 205: 799-810Crossref PubMed Scopus (614) Google Scholar, 17Stromnes I.M. Cerretti L.M. Liggitt D. Harris R.A. Goverman J.M. Differential regulation of central nervous system autoimmunity by T(H)1 and T(H)17 cells.Nat Med. 2008; 14: 337-342Crossref PubMed Scopus (496) Google Scholar The disease is most often induced by the immunization of animals with an emulsion of neural antigens in complete Freund's adjuvant (CFA). The induction protocol usually also involves injection of pertussis toxin, which is thought to be a TLR4 agonist18Kerfoot S.M. Long E.M. Hickey M.J. Andonegui G. Lapointe B.M. Zanardo R.C. Bonder C. James W.G. Robbins S.M. Kubes P. TLR4 contributes to disease-inducing mechanisms resulting in central nervous system autoimmune disease.J Immunol. 2004; 173: 7070-7077PubMed Google Scholar possibly promoting generation of Th17 cells19Chen X. Howard O.M. Oppenheim J.J. Pertussis toxin by inducing IL-6 promotes the generation of IL-17-producing CD4 cells.J Immunol. 2007; 178: 6123-6129PubMed Google Scholar, 20Linthicum D.S. Munoz J.J. Blaskett A. Acute experimental autoimmune encephalomyelitis in mice. I. Adjuvant action of Bordetella pertussis is due to vasoactive amine sensitization and increased vascular permeability of the central nervous system.Cell Immunol. 1982; 73: 299-310Crossref PubMed Scopus (181) Google Scholar and which is believed to affect the permeability of the blood–brain barrier.20Linthicum D.S. Munoz J.J. Blaskett A. Acute experimental autoimmune encephalomyelitis in mice. I. Adjuvant action of Bordetella pertussis is due to vasoactive amine sensitization and increased vascular permeability of the central nervous system.Cell Immunol. 1982; 73: 299-310Crossref PubMed Scopus (181) Google Scholar Although Q compounds have shown efficacy in several EAE models,7Brunmark C. Runström A. Ohlsson L. Sparre B. Brodin T. Aström M. Hedlund G. The new orally active immunoregulator laquinimod (ABR-215062) effectively inhibits development and relapses of experimental autoimmune encephalomyelitis.J Neuroimmunol. 2002; 130: 163-172Abstract Full Text Full Text PDF PubMed Scopus (162) Google Scholar, 9Runström A. Leanderson T. Ohlsson L. Axelsson B. Inhibition of the development of chronic experimental autoimmune encephalomyelitis by laquinimod (ABR-215062) in IFN-beta k.o. and wild type mice.J Neuroimmunol. 2006; 173: 69-78Abstract Full Text Full Text PDF PubMed Scopus (61) Google Scholar, 10Wegner C. Stadelmann C. Pförtner R. Raymond E. Feigelson S. Alon R. Timan B. Hayardeny L. Brück W. Laquinimod interferes with migratory capacity of T cells and reduces IL-17 levels, inflammatory demyelination and acute axonal damage in mice with experimental autoimmune encephalomyelitis.J Neuroimmunol. 2010; 227: 133-143Abstract Full Text Full Text PDF PubMed Scopus (111) Google Scholar, 11Yang J.S. Xu L.Y. Xiao B.G. Hedlund G. Link H. Laquinimod (ABR-215062) suppresses the development of experimental autoimmune encephalomyelitis, modulates the Th1/Th2 balance and induces the Th3 cytokine TGF-beta in Lewis rats.J Neuroimmunol. 2004; 156: 3-9Abstract Full Text Full Text PDF PubMed Scopus (165) Google Scholar, 12Björk P. Björk A. Vogl T. Stenström M. Liberg D. Olsson A. Roth J. Ivars F. Leanderson T. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides.PLoS Biol. 2009; 7: e97Crossref PubMed Scopus (258) Google Scholar, 21Schulze-Topphoff U. Shetty A. Varrin-Doyer M. Molnarfi N. Sagan S.A. Sobel R.A. Nelson P.A. Zamvil S.S. Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity.PLoS One. 2012; 7: e33797Crossref PubMed Scopus (83) Google Scholar, 22Thöne J. Ellrichmann G. Seubert S. Peruga I. Lee D.H. Conrad R. Hayardeny L. Comi G. Wiese S. Linker R.A. Gold R. Modulation of autoimmune demyelination by laquinimod via induction of brain-derived neurotrophic factor.Am J Pathol. 2012; 180: 267-274Abstract Full Text Full Text PDF PubMed Scopus (125) Google Scholar their exact mode of action in the amelioration of disease is poorly understood. Previous reports have suggested that Q compounds may alter the Th1/Th2 balance in experimental neural diseases.11Yang J.S. Xu L.Y. Xiao B.G. Hedlund G. Link H. Laquinimod (ABR-215062) suppresses the development of experimental autoimmune encephalomyelitis, modulates the Th1/Th2 balance and induces the Th3 cytokine TGF-beta in Lewis rats.J Neuroimmunol. 2004; 156: 3-9Abstract Full Text Full Text PDF PubMed Scopus (165) Google Scholar, 23Zou L.P. Abbas N. Volkmann I. Nennesmo I. Levi M. Wahren B. Winblad B. Hedlund G. Zhu J. Suppression of experimental autoimmune neuritis by ABR-215062 is associated with altered Th1/Th2 balance and inhibited migration of inflammatory cells into the peripheral nerve tissue.Neuropharmacology. 2002; 42: 731-739Crossref PubMed Scopus (94) Google Scholar Recent reports have also indicated interference of these compounds with T-cell migration, cytokine production, antigen presentation, and monocyte kinetics.10Wegner C. Stadelmann C. Pförtner R. Raymond E. Feigelson S. Alon R. Timan B. Hayardeny L. Brück W. Laquinimod interferes with migratory capacity of T cells and reduces IL-17 levels, inflammatory demyelination and acute axonal damage in mice with experimental autoimmune encephalomyelitis.J Neuroimmunol. 2010; 227: 133-143Abstract Full Text Full Text PDF PubMed Scopus (111) Google Scholar, 21Schulze-Topphoff U. Shetty A. Varrin-Doyer M. Molnarfi N. Sagan S.A. Sobel R.A. Nelson P.A. Zamvil S.S. Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity.PLoS One. 2012; 7: e33797Crossref PubMed Scopus (83) Google Scholar, 22Thöne J. Ellrichmann G. Seubert S. Peruga I. Lee D.H. Conrad R. Hayardeny L. Comi G. Wiese S. Linker R.A. Gold R. Modulation of autoimmune demyelination by laquinimod via induction of brain-derived neurotrophic factor.Am J Pathol. 2012; 180: 267-274Abstract Full Text Full Text PDF PubMed Scopus (125) Google Scholar, 24Gurevich M. Gritzman T. Orbach R. Tuller T. Feldman A. Achiron A. Laquinimod suppress antigen presentation in relapsing-remitting multiple sclerosis: in-vitro high-throughput gene expression study.J Neuroimmunol. 2010; 221: 87-94Abstract Full Text Full Text PDF PubMed Scopus (63) Google Scholar, 25Mishra M.K. Wang J. Silva C. Mack M. Yong V.W. Kinetics of proinflammatory monocytes in a model of multiple sclerosis and its perturbation by laquinimod.Am J Pathol. 2012; 181: 642-651Abstract Full Text Full Text PDF PubMed Scopus (70) Google Scholar The Q compound paquinimod has shown efficacy in several other models of autoimmune disease, including a model of systemic lupus erythematosus,5Bengtsson A.A. Sturfelt G. Lood C. Rönnblom L. van Vollenhoven R.F. Axelsson B. Sparre B. Tuvesson H. Ohman M.W. Leanderson T. Pharmacokinetics, tolerability, and preliminary efficacy of paquinimod (ABR-215757), a new quinoline-3-carboxamide derivative: studies in lupus-prone mice and a multicenter, randomized, double-blind, placebo-controlled, repeat-dose, dose-ranging study in patients with systemic lupus erythematosus.Arthritis Rheum. 2012; 64: 1579-1588Crossref PubMed Scopus (87) Google Scholar as well as a diabetes model of nonobese diabetic (NOD) mice and a collagen-induced arthritis model (unpublished data). Thus, it is likely that paquinimod targets mechanisms common to all these models. In EAE, paquinimod could, in principle, interfere with disease development at several different stages. Because EAE is highly T-cell–dependent, in the present study we studied the effect of the Q compound paquinimod on T-cell activation and T-cell effector function. A large body of data also supports an important role of myeloid cells (in particular, Ly6Chi inflammatory monocytes) in EAE. These cells are recruited to the central nervous system (CNS) and play an important role in the pathogenesis of EAE.26Dogan R.N. Elhofy A. Karpus W.J. 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: 7376-7384PubMed Google Scholar, 27King I.L. Dickendesher T.L. Segal B.M. Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease.Blood. 2009; 113: 3190-3197Crossref PubMed Scopus (336) Google Scholar, 28Mildner A. Mack M. Schmidt H. Brück W. Djukic M. Zabel M.D. Hille A. Priller J. Prinz M. CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system.Brain. 2009; 132: 2487-2500Crossref PubMed Scopus (353) Google Scholar, 29Zhu B. Kennedy J.K. Wang Y. Sandoval-Garcia C. Cao L. Xiao S. Wu C. Elyaman W. Khoury S.J. Plasticity of Ly-6C(hi) myeloid cells in T cell regulation.J Immunol. 2011; 187: 2418-2432Crossref PubMed Scopus (53) Google Scholar Interestingly, in various models of infection and inflammation, the inflammatory monocytes have been shown to differentiate into dendritic cells (DCs).30Auffray C. Fogg D.K. Narni-Mancinelli E. Senechal B. Trouillet C. Saederup N. Leemput J. Bigot K. Campisi L. Abitbol M. Molina T. Charo I. Hume D.A. Cumano A. Lauvau G. Geissmann F. CX3CR1+ CD115+ CD135+ common macrophage/DC precursors and the role of CX3CR1 in their response to inflammation.J Exp Med. 2009; 206: 595-606Crossref PubMed Scopus (321) Google Scholar, 31Cheong C. Matos I. Choi J.H. Dandamudi D.B. Shrestha E. Longhi M.P. Jeffrey K.L. Anthony R.M. Kluger C. Nchinda G. Koh H. Rodriguez A. Idoyaga J. Pack M. Velinzon K. Park C.G. Steinman R.M. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas.Cell. 2010; 143: 416-429Abstract Full Text Full Text PDF PubMed Scopus (473) Google Scholar, 32Flores-Langarica A. Marshall J.L. Bobat S. Mohr E. Hitchcock J. Ross E.A. Coughlan R.E. Khan M. Van Rooijen N. Henderson I.R. Maclennan I.C. Cunningham A.F. T-zone localized monocyte-derived dendritic cells promote Th1 priming to Salmonella [Erratum appeared in Eur J Immunol 2011, 41:3085].Eur J Immunol. 2011; 41: 2654-2665Crossref PubMed Scopus (33) Google Scholar, 33León B. López-Bravo M. Ardavín C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania.Immunity. 2007; 26: 519-531Abstract Full Text Full Text PDF PubMed Scopus (526) Google Scholar, 34Nakano H. Lin K.L. Yanagita M. Charbonneau C. Cook D.N. Kakiuchi T. Gunn M.D. Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses.Nat Immunol. 2009; 10: 394-402Crossref PubMed Scopus (276) Google Scholar, 35Serbina N.V. Salazar-Mather T.P. Biron C.A. Kuziel W.A. Pamer E.G. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection.Immunity. 2003; 19: 59-70Abstract Full Text Full Text PDF PubMed Scopus (942) Google Scholar Both in EAE and during infection, these cells induce IFN-γ production in CD4+ T cells,29Zhu B. Kennedy J.K. Wang Y. Sandoval-Garcia C. Cao L. Xiao S. Wu C. Elyaman W. Khoury S.J. Plasticity of Ly-6C(hi) myeloid cells in T cell regulation.J Immunol. 2011; 187: 2418-2432Crossref PubMed Scopus (53) Google Scholar, 31Cheong C. Matos I. Choi J.H. Dandamudi D.B. Shrestha E. Longhi M.P. Jeffrey K.L. Anthony R.M. Kluger C. Nchinda G. Koh H. Rodriguez A. Idoyaga J. Pack M. Velinzon K. Park C.G. Steinman R.M. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas.Cell. 2010; 143: 416-429Abstract Full Text Full Text PDF PubMed Scopus (473) Google Scholar, 32Flores-Langarica A. Marshall J.L. Bobat S. Mohr E. Hitchcock J. Ross E.A. Coughlan R.E. Khan M. Van Rooijen N. Henderson I.R. Maclennan I.C. Cunningham A.F. T-zone localized monocyte-derived dendritic cells promote Th1 priming to Salmonella [Erratum appeared in Eur J Immunol 2011, 41:3085].Eur J Immunol. 2011; 41: 2654-2665Crossref PubMed Scopus (33) Google Scholar, 33León B. López-Bravo M. Ardavín C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania.Immunity. 2007; 26: 519-531Abstract Full Text Full Text PDF PubMed Scopus (526) Google Scholar, 34Nakano H. Lin K.L. Yanagita M. Charbonneau C. Cook D.N. Kakiuchi T. Gunn M.D. Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses.Nat Immunol. 2009; 10: 394-402Crossref PubMed Scopus (276) Google Scholar indicating their possible role also in initiating the encephalitogenic T-cell response. It has also been suggested that dermal langerin+CD103+ DCs may be important for the induction of encephalitogenic T cells.36King I.L. Kroenke M.A. Segal B.M. GM-CSF-dependent, CD103+ dermal dendritic cells play a critical role in Th effector cell differentiation after subcutaneous immunization.J Exp Med. 2010; 207: 953-961Crossref PubMed Scopus (159) Google Scholar This issue, however, is controversial.37Edelson B.T. Bradstreet T.R. Wumesh K.C. Hildner K. Herzog J.W. Sim J. Russell J.H. Murphy T.L. Unanue E.R. Murphy K.M. Batf3-dependent CD11b(low/-) peripheral dendritic cells are GM-CSF-independent and are not required for Th cell priming after subcutaneous immunization.PLoS One. 2011; 6: e25660Crossref PubMed Scopus (97) Google Scholar We have previously reported that paquinimod treatment selectively reduces the number of splenic CD4+ DCs under steady-state conditions.38Stenström M. Anderson P. Eroukhmanoff L. Leanderson T. Ivars F. Selective depletion of splenic CD4 dendritic cells in mice treated with immunomodulatory quinoline-3-carboxamide ABR-215757.Int Immunopharmacol. 2010; 10: 837-842Crossref PubMed Scopus (5) Google Scholar Furthermore, and more importantly, we have shown that paquinimod reduces the number of splenic Gr-1+ CD11b+ cells under inflammatory conditions.39Helmersson S. Stenström M. Leanderson T. Ivars F. Specific effect of immunomodulatory quinoline-3-carboxamide ABR-215757 in GM-CSF stimulated bone marrow cell cultures: block of initiation of proliferation of Gr-1+ cells.Int Immunopharmacol. 2011; 11: 1045-1051Crossref PubMed Scopus (4) Google Scholar Given that Ly6Chi inflammatory monocytes are included in the Gr-1+CD11b+ population, our previous data suggested a possible target cell type for the ameliorating effect of Q compounds in autoimmune disease. In the present study, we used the myelin oligodendrocyte glycoprotein (MOG)–induced EAE model to study the possible effects of paquinimod treatment on cells involved in the induction phase of the disease, in particular CD4+ T cells, various antigen-presenting cells (APCs), and monocytes. C57BL/6 (B6) mice were purchased from Taconic Farms (Ejby, Denmark). (TCR)-Vβ3 transgenic mice were originally obtained from Mark M. Davis (Stanford University), and ovalbumin-specific TCR (OT-II) transgenic mice were bred at the BMC animal facility at Lund University. The mice were treated with the Q compound paquinimod (a gift from Active Biotech AB, Lund, Sweden) dissolved in drinking water available ad libitum. Except as otherwise stated, the compound was used at a dosage of 25 mg/kg per day, starting on the day before immunization. The experiments involving use of animals were performed in accordance with approved local ethical guidelines. Mice were immunized subcutaneously at the base of the tail with 50 μg MOG35-55 peptide (Schafer-N, Copenhagen, Denmark) in PBS emulsified in CFA. In some experiments, mice were immunized with PBS/CFA emulsion without added protein antigen. To induce development of EAE, the MOG-peptide–immunized B6 mice were also injected twice (on the day of immunization and 2 days later) with 200 ng pertussis toxin (List Biological Laboratories, Campbell, CA) in PBS. Mice were injected intravenously with 500 μg of either anti–Gr-1 antibody (RB6-8C5), anti-Ly6G antibody (1A8), or isotype control (MPC-11) (all from Bio X Cell, West Lebanon, NH) on the day before EAE induction. Mice were scored daily for development of EAE, starting at day 8 or 9 after immunization. Neurological defects were scored as 0 (no disease), 1 (weakness of the tail), 2 (flaccid tail), 3 (hind limb weakness), 4 (paralysis in one hind limb), 5 (total paralysis of hind limbs), 6 (forelimb weakness), 7 (forelimb paralysis), or 8 (dead). Lymph nodes or spleens were dissected, and single-cell suspensions were prepared. For isolation of splenic or lymph node DCs, the organs were cut into small pieces and treated with collagenase IV/DNase before preparation of single-cells suspensions. The single-cell suspensions were passed through 70-μm cell strainers before and after a wash. DCs and CD4+ T cells were enriched using magnetic columns and CD11c or CD4-conjugated paramagnetic beads, respectively, according to the manufacturer's protocols (Miltenyi Biotec, Bergisch Gladbach, Germany; Auburn, CA). In some experiments, lymph node cells were depleted of CD19+ and CD4+ cells by using CD4- and CD19-conjugated paramagnetic beads (Miltenyi Biotec) before analysis. To prepare cells from the CNS, brain and spinal cord were dissected after intracardial perfusion with PBS. The brain and spinal cord were squeezed through a stainless steel mesh and then treated with collagenase/DNase for 40 minutes at 37°C. The single-cell suspension was passed through a 100-μm cell strainer before washing. Leukocytes were isolated by centrifugation (400 × g for 20 minutes) on a 30% to 70% Percoll (GE Healthcare Bio-Sciences, Uppsala, Sweden) gradient. Leukocytes were collected from the 30% to 70% interphase and were washed once before further analysis by flow cytometry. Cells were stained with antibodies using standard protocols. The following antibodies were used: CD11b-Alexa700 (clone M1/70), CD11c-APC/Cy7 (clone N418), CD11c-phycoerythrin (PE)/Cy7 (clone N418), Ly6G-fluorescein isothiocyanate (FITC) (clone 1A8), B220-PerCP-Cy5.5 (clone RA3-6B2), CD45.2-APC/Cy7 (clone 104), CD4-PacificBlue (clone RM4-5), MHCII-PacificBlue (clone M5/114.15.2), CD62L-PE/Cy7 (clone MEL-14), F4/80-APC/Cy7 (clone BM8), CD40-PE (clone 1C10), CD64-PE (clone X54-5171.1), and CD3-PerCP-Cy5.5 (clone 145-2C11) from BioLegend (San Diego, CA); Ly6C-biotin (clone AL-21), SiglecF-PE (clone E50-2440), TCRβ-APC (clone H57-597), CD115-APC (clone AFS98), CD80-FITC (clone 1610-A1), and CD86-APC (clone GL1) from eBioscience (San Diego, CA); and streptavidin-BD HorizonV500, CD44-PE (clone IM7), CD103-bio (clone M290), CD103-PE (clone M290), and Ly6C-bio (clone AL-21) from BD Biosciences (San Jose, CA). CD8α-FITC (clone 53.6.72) was prepared in-house. Dead cells were excluded from analysis by staining the cells with either propidium iodide or fixable red fluorescent reactive dye (Life Technologies–Invitrogen, Carlsbad, CA). To study in vivo T-cell proliferation, immunized mice were injected with 2 mg/day 5-bromo-2′-deoxyuridine (BrdU; Sigma-Aldrich, St. Louis, MO) i.p. for 2 to 4 days. BrdU incorporation was studied using a FITC BrdU flow kit (BD Biosciences). Alternatively, to study in vivo T-cell proliferation, splenocytes were labeled with carboxyfluorescein diacetate succinimidyl ester (CFSE), using standard protocols. The number of divisions was expressed as the proliferation index, which was determined using ModFit LT software version 2.0 (Verity Software House, Topsham, ME). Cells were analyzed using an LSR II flow cytometer (BD Biosciences) with FlowJo software version 9.0 (Tree Star, Ashland, OR). Inguinal draining lymph nodes (dLNs) and spleen were dissected, and single-cell suspensions were prepared. Total cells were cultured with 50 ng/mL phorbol 12-myristate 13-acetate, 500 ng/mL ionomycin, and 10 μg/mL brefeldin A (Sigma-Aldrich) for 4 hours. Cells were then washed and stained with antibodies for intracellular cytokines using fixation/permeabilization diluent, fixation/permeabilization buffer, and permeabilization buffer (eBioscience) according to stand

Referência(s)