Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(6): 957-968, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38811815

RESUMEN

The adult central nervous system (CNS) possesses a limited capacity for self-repair. Severed CNS axons typically fail to regrow. There is an unmet need for treatments designed to enhance neuronal viability, facilitate axon regeneration and ultimately restore lost neurological functions to individuals affected by traumatic CNS injury, multiple sclerosis, stroke and other neurological disorders. Here we demonstrate that both mouse and human bone marrow neutrophils, when polarized with a combination of recombinant interleukin-4 (IL-4) and granulocyte colony-stimulating factor (G-CSF), upregulate alternative activation markers and produce an array of growth factors, thereby gaining the capacity to promote neurite outgrowth. Moreover, adoptive transfer of IL-4/G-CSF-polarized bone marrow neutrophils into experimental models of CNS injury triggered substantial axon regeneration within the optic nerve and spinal cord. These findings have far-reaching implications for the future development of autologous myeloid cell-based therapies that may bring us closer to effective solutions for reversing CNS damage.


Asunto(s)
Axones , Factor Estimulante de Colonias de Granulocitos , Interleucina-4 , Ratones Endogámicos C57BL , Regeneración Nerviosa , Neutrófilos , Animales , Neutrófilos/inmunología , Regeneración Nerviosa/inmunología , Ratones , Humanos , Axones/metabolismo , Axones/fisiología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos/farmacología , Interleucina-4/metabolismo , Activación Neutrófila , Traumatismos de la Médula Espinal/terapia , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/metabolismo , Traslado Adoptivo , Citocinas/metabolismo , Células Cultivadas
2.
Nat Immunol ; 21(12): 1496-1505, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33106668

RESUMEN

Transected axons typically fail to regenerate in the central nervous system (CNS), resulting in chronic neurological disability in individuals with traumatic brain or spinal cord injury, glaucoma and ischemia-reperfusion injury of the eye. Although neuroinflammation is often depicted as detrimental, there is growing evidence that alternatively activated, reparative leukocyte subsets and their products can be deployed to improve neurological outcomes. In the current study, we identify a unique granulocyte subset, with characteristics of an immature neutrophil, that had neuroprotective properties and drove CNS axon regeneration in vivo, in part via secretion of a cocktail of growth factors. This pro-regenerative neutrophil promoted repair in the optic nerve and spinal cord, demonstrating its relevance across CNS compartments and neuronal populations. Our findings could ultimately lead to the development of new immunotherapies that reverse CNS damage and restore lost neurological function across a spectrum of diseases.


Asunto(s)
Axones/metabolismo , Comunicación Celular , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Regeneración Nerviosa , Neuronas/metabolismo , Neutrófilos/metabolismo , Animales , Biomarcadores , Plasticidad de la Célula/inmunología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Sistema Nervioso Central/inmunología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Ratones , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Nervio Óptico/inmunología , Nervio Óptico/metabolismo , Receptores de Interleucina-8B/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Transcriptoma , Zimosan/metabolismo , Zimosan/farmacología
4.
Mult Scler ; 29(14): 1701-1708, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37877740

RESUMEN

Multiple sclerosis (MS) is most likely to adopt a progressive clinical course during middle age or beyond, and the number of older adults with MS is steadily increasing. Developing new strategies to manage progressive forms of MS, which do not respond to currently available disease-modifying therapies (DMTs), will require a deeper understanding of the mechanisms by which biological aging interacts with pathogenic pathways to propel disability accumulation. In experimental autoimmune encephalomyelitis (EAE), a widely used preclinical mouse model of MS, middle-aged animals experience a more severe and protracted clinical course than their younger counterparts. This exacerbated disease course is accompanied by persistent neuroinflammation. Clinical studies of age-related biomarkers, such as telomere length, senescence markers, and DNA methylation, suggest that biological aging is accelerated in people with MS compared with age- and sex-matched healthy controls. Furthermore, distinguishing biological age from chronological may afford more precision in determining aging effects in MS. Here we review the current literature on aging biology and its impact on MS pathogenesis. Future research on this topic may lead to the development of novel biomarkers and senotherapy agents that slow neurological decline in people with progressive MS by targeting relevant aging-related pathways.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple Crónica Progresiva , Esclerosis Múltiple , Persona de Mediana Edad , Humanos , Ratones , Animales , Anciano , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Envejecimiento , Progresión de la Enfermedad , Biomarcadores
5.
J Immunol ; 206(1): 3-10, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33443060

RESUMEN

Multiple sclerosis (MS), a neuroinflammatory disease that affects millions worldwide, is widely thought to be autoimmune in etiology. Historically, research into MS pathogenesis has focused on autoreactive CD4 T cells because of their critical role in the animal model, experimental autoimmune encephalomyelitis, and the association between MS susceptibility and single-nucleotide polymorphisms in the MHC class II region. However, recent studies have revealed prominent clonal expansions of CD8 T cells within the CNS during MS. In this paper, we review the literature on CD8 T cells in MS, with an emphasis on their potential effector and regulatory properties. We discuss the impact of disease modifying therapies, currently prescribed to reduce MS relapse rates, on CD8 T cell frequency and function. A deeper understanding of the role of CD8 T cells in MS may lead to the development of more effective and selective immunomodulatory drugs for particular subsets of patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Encefalitis/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Animales , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Ratones
6.
Proc Natl Acad Sci U S A ; 116(48): 24221-24230, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31699814

RESUMEN

The success of B cell depletion therapies and identification of leptomeningeal ectopic lymphoid tissue (ELT) in patients with multiple sclerosis (MS) has renewed interest in the antibody-independent pathogenic functions of B cells during neuroinflammation. The timing and location of B cell antigen presentation during MS and its animal model experimental autoimmune encephalomyelitis (EAE) remain undefined. Using a new EAE system that incorporates temporal regulation of MHCII expression by myelin-specific B cells, we observed the rapid formation of large B cell clusters in the spinal cord subarachnoid space. Neutrophils preceded the accumulation of meningeal B cell clusters, and inhibition of CXCR2-mediated granulocyte trafficking to the central nervous system reduced pathogenic B cell clusters and disease severity. Further, B cell-restricted very late antigen-4 (VLA-4) deficiency abrogated EAE dependent on B cell antigen presentation. Together, our findings demonstrate that neutrophils coordinate VLA-4-dependent B cell accumulation within the meninges during neuroinflammation, a key early step in the formation of ELT observed in MS.


Asunto(s)
Linfocitos B/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Integrina alfa4beta1/metabolismo , Meninges/inmunología , Esclerosis Múltiple/patología , Animales , Presentación de Antígeno , Linfocitos B/patología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Integrina alfa4beta1/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Masculino , Meninges/patología , Meningitis/inmunología , Meningitis/patología , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Células Mieloides/patología , Neutrófilos/inmunología , Neutrófilos/patología , Conejos , Receptores de Interleucina-8B/metabolismo , Espacio Subaracnoideo/patología
7.
Mult Scler ; 26(11): 1437-1440, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31237825

RESUMEN

BACKGROUND: Postoperative multiple sclerosis (MS) relapses are a concern among patients and providers. OBJECTIVE: To determine whether MS relapse risk is higher postoperatively. METHODS: Data were extracted from medical records of MS patients undergoing surgery at a tertiary center (2000-2016). Conditional logistic regression estimated within-patient unadjusted and age-adjusted odds of postoperative versus preoperative relapse. RESULTS: Among 281 patients and 609 surgeries, 12 postoperative relapses were identified. The odds of postoperative versus preoperative relapse in unadjusted (odds ratio (OR) = 0.56, 95% confidence interval (CI) = 0.18-1.79; p = 0.33) or age-adjusted models (OR = 0.66, 95% CI = 0.20-2.16; p = 0.49) were not increased. CONCLUSIONS: Surgery/anesthesia exposure did not increase postoperative relapse risk. These findings require confirmation in larger studies.


Asunto(s)
Anestesia , Esclerosis Múltiple , Anestesia/efectos adversos , Enfermedad Crónica , Humanos , Oportunidad Relativa , Recurrencia , Estudios Retrospectivos , Factores de Riesgo
8.
J Immunol ; 200(3): 966-973, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29288202

RESUMEN

GM-CSF has been portrayed as a critical cytokine in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and, ostensibly, in multiple sclerosis. C57BL/6 mice deficient in GM-CSF are resistant to EAE induced by immunization with myelin oligodendrocyte glycoprotein (MOG)35-55 The mechanism of action of GM-CSF in EAE is poorly understood. In this study, we show that GM-CSF augments the accumulation of MOG35-55-specific T cells in the skin draining lymph nodes of primed mice, but it is not required for the development of encephalitogenic T cells. Abrogation of GM-CSF receptor signaling in adoptive transfer recipients of MOG35-55-specific T cells did not alter the incidence of EAE or the trajectory of its initial clinical course, but it limited the extent of chronic CNS tissue damage and neurologic disability. The attenuated clinical course was associated with a relative dearth of MOG35-55-specific T cells, myeloid dendritic cells, and neutrophils, as well as an abundance of B cells, within CNS infiltrates. Our data indicate that GM-CSF drives chronic tissue damage and disability in EAE via pleiotropic pathways, but it is dispensable during early lesion formation and the onset of neurologic deficits.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Sistema Nervioso Central/citología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/trasplante , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Pulmón/patología , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Neutrófilos/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
9.
Curr Opin Rheumatol ; 31(3): 250-255, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30920972

RESUMEN

PURPOSE OF REVIEW: This review discusses concepts for diagnosing neuromyelitis optica spectrum disorders (NMOSD), distinguishing NMOSD from other inflammatory diseases of the central nervous system, and highlights recent and forthcoming data on acute and maintenance therapy of NMOSD. RECENT FINDINGS: The neurologic manifestations of NMOSD are heterogenous, extending beyond classic presentations of optic neuritis and longitudinally extensive transverse myelitis. NMOSD may be comorbid with rheumatologic diseases, such as systemic lupus erythematosus, but is recognized as a distinct entity. Recent studies of acute treatment of NMOSD support early use of plasmapheresis. Relapse prevention is essential, as relapses can be disabling and patients may have only partial recovery. Current practice generally recommends at least 5 years of maintenance treatment. Recent randomized data demonstrates superiority of rituximab over azathioprine. Phase 3 trials have recently been completed or are underway studying novel therapies employing B-cell depletion, complement inhibition, and cell-based mechanisms (among other mechanisms) for maintenance therapy of NMOSD. SUMMARY: NMOSD is a heterogeneous but well-defined clinical entity, distinct from other neurologic and systemic inflammatory diseases, and treatment is poised for expansion.


Asunto(s)
Azatioprina/uso terapéutico , Inmunosupresores/uso terapéutico , Neuromielitis Óptica/terapia , Rituximab/uso terapéutico , Comorbilidad , Diagnóstico Diferencial , Humanos , Depleción Linfocítica , Neuromielitis Óptica/diagnóstico , Neuromielitis Óptica/tratamiento farmacológico , Plasmaféresis , Recurrencia
10.
Ann Neurol ; 83(1): 131-141, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29283442

RESUMEN

OBJECTIVE: Myeloid cells, including macrophages and dendritic cells, are a prominent component of central nervous system (CNS) infiltrates during multiple sclerosis (MS) and the animal model experimental autoimmune encephalomyelitis (EAE). Although myeloid cells are generally thought to be proinflammatory, alternatively polarized subsets can serve noninflammatory and/or reparative functions. Here we investigate the heterogeneity and biological properties of myeloid cells during central nervous system autoimmunity. METHODS: Myeloid cell phenotypes in chronic active MS lesions were analyzed by immunohistochemistry. In addition, immune cells were isolated from the CNS during exacerbations and remissions of EAE and characterized by flow cytometric, genetic, and functional assays. RESULTS: Myeloid cells expressing inducible nitric oxide synthase (iNOS), indicative of a proinflammatory phenotype, were detected in the actively demyelinating rim of chronic active MS lesions, whereas macrophages expressing mannose receptor (CD206), a marker of alternatively polarized human myeloid cells, were enriched in the quiescent lesion core. During EAE, CNS-infiltrating myeloid cells, as well as microglia, shifted from expression of proinflammatory markers to expression of noninflammatory markers immediately prior to clinical remissions. Murine CNS myeloid cells expressing the alternative lineage marker arginase-1 (Arg1) were partially derived from iNOS+ precursors and were deficient in activating encephalitogenic T cells compared with their Arg1- counterparts. INTERPRETATION: These observations demonstrate the heterogeneity of CNS myeloid cells, their evolution during the course of autoimmune demyelinating disease, and their plasticity on the single cell level. Future therapeutic strategies for disease modification in individuals with MS may be focused on accelerating the transition of CNS myeloid cells from a proinflammatory to a noninflammatory phenotype. Ann Neurol 2018;83:131-141.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/patología , Plasticidad de la Célula , Células Mieloides/patología , Animales , Arginasa/genética , Células de la Médula Ósea/patología , Quimera , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Humanos , Inmunohistoquímica , Lectinas Tipo C/biosíntesis , Lectinas Tipo C/genética , Receptor de Manosa , Lectinas de Unión a Manosa/biosíntesis , Lectinas de Unión a Manosa/genética , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Fenotipo , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética
11.
Clin Immunol ; 189: 29-33, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-27664933

RESUMEN

Eotaxins are C-C motif chemokines first identified as potent eosinophil chemoattractants. They facilitate eosinophil recruitment to sites of inflammation in response to parasitic infections as well as allergic and autoimmune diseases such as asthma, atopic dermatitis, and inflammatory bowel disease. The eotaxin family currently includes three members: eotaxin-1 (CCL11), eotaxin-2 (CCL24), and eotaxin-3 (CCL26). Despite having only ~30% sequence homology to one another, each was identified based on its ability to bind the chemokine receptor, CCR3. Beyond their role in innate immunity, recent studies have shown that CCL11 and related molecules may directly contribute to degenerative processes in the central nervous system (CNS). CCL11 levels increase in the plasma and cerebrospinal fluid of both mice and humans as part of normal aging. In mice, these increases are associated with declining neurogenesis and impaired cognition and memory. In humans, elevated plasma levels of CCL11 have been observed in Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease, and secondary progressive multiple sclerosis when compared to age-matched, healthy controls. Since CCL11 is capable of crossing the blood-brain barrier of normal mice, it is plausible that eotaxins generated in the periphery may exert physiological and pathological actions in the CNS. Here, we briefly review known functions of eotaxin family members during innate immunity, and then focus on whether and how these molecules might participate in the progression of neurodegenerative diseases.


Asunto(s)
Quimiocina CCL11/inmunología , Quimiocina CCL24/inmunología , Quimiocina CCL26/inmunología , Inmunidad Innata/inmunología , Enfermedades Neurodegenerativas/inmunología , Envejecimiento/inmunología , Animales , Quimiocina CCL11/sangre , Quimiocina CCL11/líquido cefalorraquídeo , Quimiocina CCL24/sangre , Quimiocina CCL24/líquido cefalorraquídeo , Quimiocina CCL26/sangre , Quimiocina CCL26/líquido cefalorraquídeo , Humanos , Enfermedades Neurodegenerativas/sangre , Enfermedades Neurodegenerativas/líquido cefalorraquídeo , Receptores CCR3/inmunología , Receptores CCR3/metabolismo
12.
J Neuroinflammation ; 15(1): 208, 2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012158

RESUMEN

BACKGROUND: Myelin oligodendrocyte glycoprotein (MOG)-reactive T-helper (Th)1 cells induce conventional experimental autoimmune encephalomyelitis (cEAE), characterized by ascending paralysis and monocyte-predominant spinal cord infiltrates, in C57BL/6 wildtype (WT) hosts. The same T cells induce an atypical form of EAE (aEAE), characterized by ataxia and neutrophil-predominant brainstem infiltrates, in syngeneic IFNγ receptor (IFNγR)-deficient hosts. Production of ELR+ CXC chemokines within the CNS is required for the development of aEAE, but not cEAE. The cellular source(s) and localization of ELR+ CXC chemokines in the CNS and the IFNγ-dependent pathways that regulate their production remain to be elucidated. METHODS: The spatial distribution of inflammatory lesions and CNS expression of the ELR+ CXC chemokines, CXCL1 and CXCL2, were determined via immunohistochemistry and/or in situ hybridization. Levels of CXCL1 and CXCL2, and their cognate receptor CXCR2, were measured in/on leukocyte subsets by flow cytometric and quantitative PCR (qPCR) analysis. Bone marrow neutrophils and macrophages were cultured with inflammatory stimuli in vitro prior to measurement of CXCL2 and CXCR2 by qPCR or flow cytometry. RESULTS: CNS-infiltrating neutrophils and monocytes, and resident microglia, are a prominent source of CXCL2 in the brainstem of IFNγRKO adoptive transfer recipients during aEAE. In WT transfer recipients, IFNγ directly suppresses CXCL2 transcription in microglia and myeloid cells, and CXCR2 transcription in CNS-infiltrating neutrophils. Consequently, infiltration of the brainstem parenchyma from the adjacent meninges is blocked during cEAE. CXCL2 directly stimulates its own expression in cultured neutrophils, which is enhanced by IL-1 and suppressed by IFNγ. CONCLUSIONS: We provide evidence for an IFNγ-regulated CXCR2/CXCL2 autocrine/paracrine feedback loop in innate immune cells that determines the location of CNS infiltrates during Th1-mediated EAE. When IFNγ signaling is impaired, myeloid cell production of CXCL2 increases, which promotes brainstem inflammation and results in clinical ataxia. IFNγ, produced within the CNS of WT recipients, suppresses myeloid cell CXCR2 and CXCL2 production, thereby skewing the location of neuroinflammatory infiltrates to the spinal cord and the clinical phenotype to an ascending paralysis. These data reveal a novel mechanism by which IFNγ and CXCL2 interact to direct regional recruitment of leukocytes in the CNS, resulting in distinct clinical presentations.


Asunto(s)
Encéfalo/metabolismo , Quimiocina CXCL2/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Interferón gamma/metabolismo , Transducción de Señal/fisiología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Proteína Ácida Fibrilar de la Glía/metabolismo , Interferón gamma/genética , Interferón gamma/farmacología , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/efectos de los fármacos , Monocitos/patología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Células Mieloides/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/patología , Tejido Parenquimatoso/patología , Fragmentos de Péptidos/toxicidad , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Mult Scler ; 24(1): 4-5, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29307302

RESUMEN

The Americas Committee for Treatment and Research in Multiple Sclerosis (ACTRIMS) Forum 2017 brought together a multidisciplinary team of basic and clinical researchers to discuss the impact of environmental, genetic, and epigenetic factors on multiple sclerosis (MS) susceptibility and clinical course. The series of articles in this special edition reflect the breadth and depth of the topics that were discussed.


Asunto(s)
Esclerosis Múltiple/etiología , Exposición a Riesgos Ambientales , Epigénesis Genética , Predisposición Genética a la Enfermedad , Humanos
14.
Proc Natl Acad Sci U S A ; 112(8): 2581-6, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25675510

RESUMEN

Innate immunity can facilitate nervous system regeneration, yet the underlying cellular and molecular mechanisms are not well understood. Here we show that intraocular injection of lipopolysaccharide (LPS), a bacterial cell wall component, or the fungal cell wall extract zymosan both lead to rapid and comparable intravitreal accumulation of blood-derived myeloid cells. However, when combined with retro-orbital optic nerve crush injury, lengthy growth of severed retinal ganglion cell (RGC) axons occurs only in zymosan-injected mice, and not in LPS-injected mice. In mice deficient for the pattern recognition receptor dectin-1 but not Toll-like receptor-2 (TLR2), zymosan-mediated RGC regeneration is greatly reduced. The combined loss of dectin-1 and TLR2 completely blocks the proregenerative effects of zymosan. In the retina, dectin-1 is expressed by microglia and dendritic cells, but not by RGCs. Dectin-1 is also present on blood-derived myeloid cells that accumulate in the vitreous. Intraocular injection of the dectin-1 ligand curdlan [a particulate form of ß(1, 3)-glucan] promotes optic nerve regeneration comparable to zymosan in WT mice, but not in dectin-1(-/-) mice. Particulate ß(1, 3)-glucan leads to increased Erk1/2 MAP-kinase signaling and cAMP response element-binding protein (CREB) activation in myeloid cells in vivo. Loss of the dectin-1 downstream effector caspase recruitment domain 9 (CARD9) blocks CREB activation and attenuates the axon-regenerative effects of ß(1, 3)-glucan. Studies with dectin-1(-/-)/WT reciprocal bone marrow chimeric mice revealed a requirement for dectin-1 in both retina-resident immune cells and bone marrow-derived cells for ß(1, 3)-glucan-elicited optic nerve regeneration. Collectively, these studies identify a molecular framework of how innate immunity enables repair of injured central nervous system neurons.


Asunto(s)
Axones/fisiología , Sistema Nervioso Central/patología , Inflamación/patología , Lectinas Tipo C/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , beta-Glucanos/efectos adversos , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Sistema Nervioso Central/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Fagocitosis/efectos de los fármacos , Tolerancia a Radiación/efectos de los fármacos , Retina/efectos de los fármacos , Retina/metabolismo , Receptor Toll-Like 2/metabolismo , Zimosan/farmacología
15.
J Immunol ; 195(6): 2552-9, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26238492

RESUMEN

Multiple sclerosis (MS) is believed to be initiated by myelin-reactive CD4(+) Th cells. IL-12-polarized Th1 cells, IL-23-polarized Th17 cells, and Th17 cells that acquire Th1 characteristics were each implicated in autoimmune pathogenesis. It is debated whether Th cells that can drive the development of demyelinating lesions are phenotypically diverse or arise from a single lineage. In the current study, we assessed the requirement of IL-12 or IL-23 stimulation, as well as Th plasticity, for the differentiation of T cells capable of inducing CNS axon damage. We found that stable murine Th1 and Th17 cells independently transfer experimental autoimmune encephalomyelitis (widely used as an animal model of MS) in the absence of IL-23 and IL-12, respectively. Plastic Th17 cells are particularly potent mediators of demyelination and axonopathy. In parallel studies, we identified MS patients who consistently mount either IFN-γ- or IL-17-skewed responses to myelin basic protein over the course of a year. Brain magnetic resonance imaging revealed that patients with mixed IFN-γ and IL-17 responses have relatively high T1 lesion burden, a measure of permanent axon damage. Our data challenge the dogma that IL-23 and Th17 plasticity are universally required for the development of experimental autoimmune encephalomyelitis. This study definitively demonstrates that autoimmune demyelinating disease can be driven by distinct Th-polarizing factors and effector subsets, underscoring the importance of a customized approach to the pharmaceutical management of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Células TH1/inmunología , Células Th17/inmunología , Traslado Adoptivo , Animales , Autoinmunidad/inmunología , Encéfalo/diagnóstico por imagen , Diferenciación Celular/inmunología , Enfermedades Desmielinizantes/inmunología , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-17/inmunología , Interleucina-23/inmunología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Básica de Mielina/inmunología , Nervio Óptico/inmunología , Nervio Óptico/patología , Radiografía , Células TH1/citología , Células TH1/trasplante , Células Th17/citología , Células Th17/trasplante
16.
Eur J Immunol ; 45(10): 2780-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26220255

RESUMEN

CD4(+) T-helper (Th) cells reactive against myelin antigens mediate the mouse model experimental autoimmune encephalomyelitis (EAE) and have been implicated in the pathogenesis of multiple sclerosis (MS). It is currently debated whether encephalitogenic Th cells are heterogeneous or arise from a single lineage. In the current study, we challenge the dogma that stimulation with the monokine IL-23 is universally required for the acquisition of pathogenic properties by myelin-reactive T cells. We show that IL-12-modulated Th1 cells readily produce IFN-γ and GM-CSF in the CNS of mice and induce a severe form of EAE via an IL-23-independent pathway. Th1-mediated EAE is characterized by monocyte-rich CNS infiltrates, elicits a strong proinflammatory cytokine response in the CNS, and is partially CCR2 dependent. Conversely, IL-23-modulated, stable Th17 cells induce EAE with a relatively mild course via an IL-12-independent pathway. These data provide definitive evidence that autoimmune disease can be driven by distinct CD4(+) T-helper-cell subsets and polarizing factors.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Interleucina-12/inmunología , Interleucina-23/inmunología , Esclerosis Múltiple/inmunología , Células TH1/inmunología , Animales , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-12/genética , Interleucina-23/genética , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Vaina de Mielina/genética , Vaina de Mielina/inmunología , Receptores CCR2/genética , Receptores CCR2/inmunología , Células TH1/patología , Células Th17/inmunología , Células Th17/patología
17.
J Neuroinflammation ; 13(1): 178, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27391474

RESUMEN

BACKGROUND: Neurodegeneration is believed to be the primary cause of permanent, long-term disability in patients with multiple sclerosis. The cause of neurodegeneration in multiple sclerosis appears to be multifactorial. One mechanism that has been implicated in the pathogenesis of neurodegeneration in multiple sclerosis is the targeting of neuronal and axonal antigens by autoantibodies. Multiple sclerosis patients develop antibodies to the RNA-binding protein, heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1), which is enriched in neurons. We hypothesized that anti-hnRNP A1 antibodies would contribute to neurodegeneration in an animal model of multiple sclerosis. METHODS: Following induction of experimental autoimmune encephalomyelitis (EAE) by direct immunization with myelin oligodendrocyte glycoprotein, mice were injected with anti-hnRNP A1 or control antibodies. Animals were examined clinically, and the central nervous system (CNS) tissues were tested for neurodegeneration with Fluoro-Jade C, a marker of degenerating neural elements. RESULTS: Injection of anti-hnRNP A1 antibodies in mice with EAE worsened clinical disease, altered the clinical disease phenotype, and caused neurodegeneration preferentially in the ventral spinocerebellar tract and deep white matter of the cerebellum in the CNS. Neurodegeneration in mice injected with hnRNP A1-M9 antibodies compared to control groups was consistent with "dying back" axonal degeneration. CONCLUSIONS: These data suggest that antibodies to the RNA-binding protein hnRNP A1 contribute to neurodegeneration in immune-mediated disease of the CNS.


Asunto(s)
Autoanticuerpos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/inmunología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Animales , Autoanticuerpos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/metabolismo
19.
Brain Behav Immun ; 54: 128-139, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26795429

RESUMEN

Elevated CXCL13 within the central nervous system (CNS) correlates with humoral responses in several neuroinflammatory diseases, yet its role is controversial. During coronavirus encephalomyelitis CXCL13 deficiency impaired CNS accumulation of memory B cells and antibody-secreting cells (ASC) but not naïve/early-activated B cells. However, despite diminished germinal center B cells and follicular helper T cells in draining lymph nodes, ASC in bone marrow and antiviral serum antibody were intact in the absence of CXCL13. The data demonstrate that CXCL13 is not essential in mounting effective peripheral humoral responses, but specifically promotes CNS accumulation of differentiated B cells.


Asunto(s)
Linfocitos B/inmunología , Sistema Nervioso Central/inmunología , Quimiocina CXCL13/inmunología , Infecciones por Coronavirus/inmunología , Encefalomielitis/inmunología , Animales , Linfocitos B/patología , Movimiento Celular/inmunología , Infecciones por Coronavirus/patología , Encefalomielitis/patología , Femenino , Cambio de Clase de Inmunoglobulina/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Colaboradores-Inductores/inmunología
20.
J Immunol ; 193(2): 564-70, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24928987

RESUMEN

The adoptive transfer of myelin-reactive T cells into wild-type hosts results in spinal cord inflammation and ascending paralysis, referred to as conventional experimental autoimmune encephalomyelitis (EAE), as opposed to brainstem inflammation and ataxia, which characterize disease in IFN-γRKO hosts (atypical EAE). In this article, we show that atypical EAE correlates with preferential upregulation of CXCL2 in the brainstem, and is driven by CXCR2-dependent recruitment of neutrophils. In contrast, conventional EAE is associated with upregulation of CCL2 in the spinal cord, and is driven by recruitment of monocytes via a partially CCR2-dependent pathway. This study illustrates how regional differences in chemokine expression within a target organ shape the spatial pattern and composition of autoimmune infiltrates, leading to disparate clinical outcomes.


Asunto(s)
Sistema Nervioso Central/inmunología , Quimiocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos de Diferenciación de Linfocitos T/metabolismo , Tronco Encefálico/inmunología , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Quimiocinas/biosíntesis , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Inmunofenotipificación , Inflamación/genética , Inflamación/metabolismo , Interleucina-12/inmunología , Interleucina-12/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Monocitos/inmunología , Monocitos/metabolismo , Glicoproteína Mielina-Oligodendrócito , Neutrófilos/inmunología , Neutrófilos/metabolismo , Fragmentos de Péptidos , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Interleucina-8B/inmunología , Receptores de Interleucina-8B/metabolismo , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA