Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 585(7823): 102-106, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32848245

RESUMEN

Accumulating evidence indicates that gut microorganisms have a pathogenic role in autoimmune diseases, including in multiple sclerosis1. Studies of experimental autoimmune encephalomyelitis (an animal model of multiple sclerosis)2,3, as well as human studies4-6, have implicated gut microorganisms in the development or severity of multiple sclerosis. However, it remains unclear how gut microorganisms act on the inflammation of extra-intestinal tissues such as the spinal cord. Here we show that two distinct signals from gut microorganisms coordinately activate autoreactive T cells in the small intestine that respond specifically to myelin oligodendrocyte glycoprotein (MOG). After induction of experimental autoimmune encephalomyelitis in mice, MOG-specific CD4+ T cells are observed in the small intestine. Experiments using germ-free mice that were monocolonized with microorganisms from the small intestine demonstrated that a newly isolated strain in the family Erysipelotrichaceae acts similarly to an adjuvant to enhance the responses of T helper 17 cells. Shotgun sequencing of the contents of the small intestine revealed a strain of Lactobacillus reuteri that possesses peptides that potentially mimic MOG. Mice that were co-colonized with these two strains showed experimental autoimmune encephalomyelitis symptoms that were more severe than those of germ-free or monocolonized mice. These data suggest that the synergistic effects that result from the presence of these microorganisms should be considered in the pathogenicity of multiple sclerosis, and that further study of these microorganisms may lead to preventive strategies for this disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/microbiología , Microbioma Gastrointestinal/inmunología , Inflamación/patología , Médula Espinal/patología , Linfocitos T/inmunología , Linfocitos T/patología , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Vida Libre de Gérmenes , Inflamación/inmunología , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Intestino Delgado/patología , Limosilactobacillus reuteri/química , Limosilactobacillus reuteri/inmunología , Limosilactobacillus reuteri/patogenicidad , Masculino , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito/química , Glicoproteína Mielina-Oligodendrócito/inmunología , Médula Espinal/inmunología , Células Th17/inmunología , Células Th17/patología
2.
Nature ; 557(7707): 724-728, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29769726

RESUMEN

Microglia and astrocytes modulate inflammation and neurodegeneration in the central nervous system (CNS)1-3. Microglia modulate pro-inflammatory and neurotoxic activities in astrocytes, but the mechanisms involved are not completely understood4,5. Here we report that TGFα and VEGF-B produced by microglia regulate the pathogenic activities of astrocytes in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Microglia-derived TGFα acts via the ErbB1 receptor in astrocytes to limit their pathogenic activities and EAE development. Conversely, microglial VEGF-B triggers FLT-1 signalling in astrocytes and worsens EAE. VEGF-B and TGFα also participate in the microglial control of human astrocytes. Furthermore, expression of TGFα and VEGF-B in CD14+ cells correlates with the multiple sclerosis lesion stage. Finally, metabolites of dietary tryptophan produced by the commensal flora control microglial activation and TGFα and VEGF-B production, modulating the transcriptional program of astrocytes and CNS inflammation through a mechanism mediated by the aryl hydrocarbon receptor. In summary, we identified positive and negative regulators that mediate the microglial control of astrocytes. Moreover, these findings define a pathway through which microbial metabolites limit pathogenic activities of microglia and astrocytes, and suppress CNS inflammation. This pathway may guide new therapies for multiple sclerosis and other neurological disorders.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/microbiología , Microglía/metabolismo , Animales , Astrocitos/patología , Células Cultivadas , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/microbiología , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/prevención & control , Receptores ErbB/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Inflamación/prevención & control , Receptores de Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Receptores de Hidrocarburo de Aril/metabolismo , Simbiosis , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador alfa/metabolismo , Triptófano/deficiencia , Triptófano/metabolismo , Factor B de Crecimiento Endotelial Vascular/biosíntesis , Factor B de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Nature ; 551(7682): 585-589, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29143823

RESUMEN

A Western lifestyle with high salt consumption can lead to hypertension and cardiovascular disease. High salt may additionally drive autoimmunity by inducing T helper 17 (TH17) cells, which can also contribute to hypertension. Induction of TH17 cells depends on gut microbiota; however, the effect of salt on the gut microbiome is unknown. Here we show that high salt intake affects the gut microbiome in mice, particularly by depleting Lactobacillus murinus. Consequently, treatment of mice with L. murinus prevented salt-induced aggravation of actively induced experimental autoimmune encephalomyelitis and salt-sensitive hypertension by modulating TH17 cells. In line with these findings, a moderate high-salt challenge in a pilot study in humans reduced intestinal survival of Lactobacillus spp., increased TH17 cells and increased blood pressure. Our results connect high salt intake to the gut-immune axis and highlight the gut microbiome as a potential therapeutic target to counteract salt-sensitive conditions.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Lactobacillus/efectos de los fármacos , Lactobacillus/aislamiento & purificación , Cloruro de Sodio/farmacología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Animales , Autoinmunidad/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Heces/microbiología , Humanos , Hipertensión/inducido químicamente , Ácidos Indolacéticos/metabolismo , Indoles/metabolismo , Intestinos/citología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Intestinos/microbiología , Lactobacillus/inmunología , Activación de Linfocitos/efectos de los fármacos , Recuento de Linfocitos , Masculino , Ratones , Proyectos Piloto , Cloruro de Sodio/administración & dosificación , Simbiosis , Células Th17/citología , Triptófano/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(44): 27516-27527, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33077601

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. The etiology of MS is multifactorial, with disease risk determined by genetics and environmental factors. An emerging risk factor for immune-mediated diseases is an imbalance in the gut microbiome. However, the identity of gut microbes associated with disease risk, their mechanisms of action, and the interactions with host genetics remain obscure. To address these questions, we utilized the principal autoimmune model of MS, experimental autoimmune encephalomyelitis (EAE), together with a genetically diverse mouse model representing 29 unique host genotypes, interrogated by microbiome sequencing and targeted microbiome manipulation. We identified specific gut bacteria and their metabolic functions associated with EAE susceptibility, implicating short-chain fatty acid metabolism as a key element conserved across multiple host genotypes. In parallel, we used a reductionist approach focused on two of the most disparate phenotypes identified in our screen. Manipulation of the gut microbiome by transplantation and cohousing demonstrated that transfer of these microbiomes into genetically identical hosts was sufficient to modulate EAE susceptibility and systemic metabolite profiles. Parallel bioinformatic approaches identified Lactobacillus reuteri as a commensal species unexpectedly associated with exacerbation of EAE in a genetically susceptible host, which was functionally confirmed by bacterial isolation and commensal colonization studies. These results reveal complex interactions between host genetics and gut microbiota modulating susceptibility to CNS autoimmunity, providing insights into microbiome-directed strategies aimed at lowering the risk for autoimmune disease and underscoring the need to consider host genetics and baseline gut microbiome composition.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Microbioma Gastrointestinal/inmunología , Predisposición Genética a la Enfermedad , Interacciones Microbiota-Huesped/inmunología , Esclerosis Múltiple/genética , Animales , Autoinmunidad/genética , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Femenino , Variación Genética , Interacciones Microbiota-Huesped/genética , Humanos , Limosilactobacillus reuteri/inmunología , Masculino , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/microbiología
5.
Ann Neurol ; 89(6): 1195-1211, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33876477

RESUMEN

OBJECTIVE: This study was undertaken to investigate the gut microbiome in progressive multiple sclerosis (MS) and how it relates to clinical disease. METHODS: We sequenced the microbiota from healthy controls and relapsing-remitting MS (RRMS) and progressive MS patients and correlated the levels of bacteria with clinical features of disease, including Expanded Disability Status Scale (EDSS), quality of life, and brain magnetic resonance imaging lesions/atrophy. We colonized mice with MS-derived Akkermansia and induced experimental autoimmune encephalomyelitis (EAE). RESULTS: Microbiota ß-diversity differed between MS patients and controls but did not differ between RRMS and progressive MS or differ based on disease-modifying therapies. Disease status had the greatest effect on the microbiome ß-diversity, followed by body mass index, race, and sex. In both progressive MS and RRMS, we found increased Clostridium bolteae, Ruthenibacterium lactatiformans, and Akkermansia and decreased Blautia wexlerae, Dorea formicigenerans, and Erysipelotrichaceae CCMM. Unique to progressive MS, we found elevated Enterobacteriaceae and Clostridium g24 FCEY and decreased Blautia and Agathobaculum. Several Clostridium species were associated with higher EDSS and fatigue scores. Contrary to the view that elevated Akkermansia in MS has a detrimental role, we found that Akkermansia was linked to lower disability, suggesting a beneficial role. Consistent with this, we found that Akkermansia isolated from MS patients ameliorated EAE, which was linked to a reduction in RORγt+ and IL-17-producing γδ T cells. INTERPRETATION: Whereas some microbiota alterations are shared in relapsing and progressive MS, we identified unique bacteria associated with progressive MS and clinical measures of disease. Furthermore, elevated Akkermansia in MS may be a compensatory beneficial response in the MS microbiome. ANN NEUROL 2021;89:1195-1211.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Esclerosis Múltiple Crónica Progresiva/microbiología , Esclerosis Múltiple Crónica Progresiva/patología , Esclerosis Múltiple Recurrente-Remitente/microbiología , Esclerosis Múltiple Recurrente-Remitente/patología , Adulto , Akkermansia , Animales , Atrofia/patología , Encéfalo/patología , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Calidad de Vida
6.
Immunity ; 38(5): 1050-62, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23602766

RESUMEN

Cord factor, also called trehalose-6,6'-dimycolate (TDM), is a potent mycobacterial adjuvant. We herein report that the C-type lectin MCL (also called Clec4d) is a TDM receptor that is likely to arise from gene duplication of Mincle (also called Clec4e). Mincle is known to be an inducible receptor recognizing TDM, whereas MCL was constitutively expressed in myeloid cells. To examine the contribution of MCL in response to TDM adjuvant, we generated MCL-deficient mice. TDM promoted innate immune responses, such as granuloma formation, which was severely impaired in MCL-deficient mice. TDM-induced acquired immune responses, such as experimental autoimmune encephalomyelitis (EAE), was almost completely dependent on MCL, but not Mincle. Furthermore, by generating Clec4e(gfp) reporter mice, we found that MCL was also crucial for driving Mincle induction upon TDM stimulation. These results suggest that MCL is an FcRγ-coupled activating receptor that mediates the adjuvanticity of TDM.


Asunto(s)
Factores Cordón/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Lectinas Tipo C/inmunología , Proteínas de la Membrana/metabolismo , Receptores de IgG/inmunología , Adyuvantes Inmunológicos , Animales , Encefalomielitis Autoinmune Experimental/microbiología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Mycobacterium/inmunología , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología
7.
Eur J Immunol ; 50(12): 1863-1870, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33188704

RESUMEN

Recent literature indicates a potential importance of the gut microbiota for immune-mediated diseases. For instance, decreased diversity of commensals or an outgrowth of some bacterial strains, referred to as gut dysbiosis, was recently linked to hypertension, colitis, lupus, rheumatoid arthritis, and multiple sclerosis (MS). Studies in experimental autoimmune encephalomyelitis (EAE) as pivotal animal model of MS revealed a potential importance of microbial metabolites, including short-chain fatty acids or tryptophan metabolites. Both metabolites may influence the disease by modulation of the immune system, mainly by inducing Treg. These studies prompted researchers to investigate the contribution of the gut microbiota and microbial metabolites in the pathogenesis of MS. This review summarizes recent findings on the gut microbiota in MS patients and discusses the potential mechanisms how microbial metabolites may affect neuroinflammation. Many of these studies have been performed in the EAE model and were later reversely translated to humans. We also give a short summary on dietary high-salt effects on microbiota components and discuss the potential relevance of high-salt as a risk factor in MS.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inflamación/inmunología , Inflamación/microbiología , Microbiota/inmunología , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/microbiología
8.
Proc Natl Acad Sci U S A ; 114(40): 10713-10718, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28893978

RESUMEN

The gut microbiota regulates T cell functions throughout the body. We hypothesized that intestinal bacteria impact the pathogenesis of multiple sclerosis (MS), an autoimmune disorder of the CNS and thus analyzed the microbiomes of 71 MS patients not undergoing treatment and 71 healthy controls. Although no major shifts in microbial community structure were found, we identified specific bacterial taxa that were significantly associated with MS. Akkermansia muciniphila and Acinetobacter calcoaceticus, both increased in MS patients, induced proinflammatory responses in human peripheral blood mononuclear cells and in monocolonized mice. In contrast, Parabacteroides distasonis, which was reduced in MS patients, stimulated antiinflammatory IL-10-expressing human CD4+CD25+ T cells and IL-10+FoxP3+ Tregs in mice. Finally, microbiota transplants from MS patients into germ-free mice resulted in more severe symptoms of experimental autoimmune encephalomyelitis and reduced proportions of IL-10+ Tregs compared with mice "humanized" with microbiota from healthy controls. This study identifies specific human gut bacteria that regulate adaptive autoimmune responses, suggesting therapeutic targeting of the microbiota as a treatment for MS.


Asunto(s)
Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal , Leucocitos Mononucleares/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T/inmunología , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Leucocitos Mononucleares/microbiología , Leucocitos Mononucleares/patología , Masculino , Ratones , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/patología , Linfocitos T/microbiología , Linfocitos T/patología
9.
Proc Natl Acad Sci U S A ; 114(40): 10719-10724, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28893994

RESUMEN

There is emerging evidence that the commensal microbiota has a role in the pathogenesis of multiple sclerosis (MS), a putative autoimmune disease of the CNS. Here, we compared the gut microbial composition of 34 monozygotic twin pairs discordant for MS. While there were no major differences in the overall microbial profiles, we found a significant increase in some taxa such as Akkermansia in untreated MS twins. Furthermore, most notably, when transplanted to a transgenic mouse model of spontaneous brain autoimmunity, MS twin-derived microbiota induced a significantly higher incidence of autoimmunity than the healthy twin-derived microbiota. The microbial profiles of the colonized mice showed a high intraindividual and remarkable temporal stability with several differences, including Sutterella, an organism shown to induce a protective immunoregulatory profile in vitro. Immune cells from mouse recipients of MS-twin samples produced less IL-10 than immune cells from mice colonized with healthy-twin samples. IL-10 may have a regulatory role in spontaneous CNS autoimmunity, as neutralization of the cytokine in mice colonized with healthy-twin fecal samples increased disease incidence. These findings provide evidence that MS-derived microbiota contain factors that precipitate an MS-like autoimmune disease in a transgenic mouse model. They hence encourage the detailed search for protective and pathogenic microbial components in human MS.


Asunto(s)
Encéfalo/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Animales , Encéfalo/microbiología , Encéfalo/patología , Estudios de Cohortes , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Masculino , Metagenómica , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/patología , Adulto Joven
10.
Brain Behav Immun ; 82: 25-35, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31356922

RESUMEN

Currently, a combination of marijuana cannabinoids including delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) is used as a drug to treat muscle spasticity in patients with Multiple Sclerosis (MS). Because these cannabinoids can also suppress inflammation, it is unclear whether such patients benefit from suppression of neuroinflammation and if so, what is the mechanism through which cannabinoids act. In the currently study, we used a murine model of MS, experimental autoimmune encephalomyelitis (EAE), to study the role of gut microbiota in the attenuation of clinical signs of paralysis and inflammation caused by cannabinoids. THC + CBD treatment attenuated EAE and caused significant decrease in inflammatory cytokines such as IL-17 and IFN-γ while promoting the induction of anti-inflammatory cytokines such as IL-10 and TGF-ß. Use of 16S rRNA sequencing on bacterial DNA extracted from the gut revealed that EAE mice showed high abundance of mucin degrading bacterial species, such as Akkermansia muciniphila (A. muc), which was significantly reduced after THC + CBD treatment. Fecal Material Transfer (FMT) experiments confirmed that THC + CBD-mediated changes in the microbiome play a critical role in attenuating EAE. In silico computational metabolomics revealed that LPS biosynthesis, a key component in gram-negative bacteria such as A. muc, was found to be elevated in EAE mice which was confirmed by demonstrating higher levels of LPS in the brain, while treatment with THC + CBD reversed this trend. EAE mice treated with THC + CBD also had significantly higher levels of short chain fatty acids such as butyric, isovaleric, and valeric acids compared to naïve or disease controls. Collectively, our data suggest that cannabinoids may attenuate EAE and suppress neuroinflammation by preventing microbial dysbiosis seen during EAE and promoting healthy gut microbiota.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Cannabidiol/uso terapéutico , Cannabinoides/uso terapéutico , Cannabis/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Dronabinol/uso terapéutico , Disbiosis/complicaciones , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Microbioma Gastrointestinal/fisiología , Inflamación/complicaciones , Interferón gamma/inmunología , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple , ARN Ribosómico 16S/genética
11.
Acta Neurol Scand ; 139(3): 208-219, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30427062

RESUMEN

BACKGROUND: Multiple Sclerosis (MS) is a chronic immune-mediated neurological disease of the central nervous system with a complex and still not fully understood aetiology. In recent years, the gut microbiota and fermentative metabolites like short-chain fatty acids (SCFAs) have received increased attention in relation to the development and disease course of MS. This systematic review highlights and summarizes the existing literature within this field. METHODS: A systematic search in PubMed was conducted on 12 October 2017, to find published original studies on SCFAs and their impact on MS and the animal model of MS experimental autoimmune encephalomyelitis (EAE). Furthermore, all studies analysing the gut microbiota in MS patients were included. A total of 14 studies were eligible for this review. RESULTS: Short-chain fatty acids have been shown to ameliorate the disease course in EAE, but no studies specifically addressing the role of SCFAs in human MS patients were identified. However, some investigations have shown that the microbiota of MS patients is characterized by a reduction in SCFA-producing bacteria. CONCLUSIONS: Studies of EAE in mice suggest that SCFAs may play a role in the development and progression of EAE, but so far this has not been confirmed in humans. An aberrant gut microbiota in MS patients has been reported to be differentially abundant compared with healthy controls, although with little consistency in the bacterial taxa. Further investigations are required to elucidate the involvement of the gut microbiota and its metabolites, including potential beneficial effects of SCFAs, in the development and course of MS.


Asunto(s)
Ácidos Grasos Volátiles , Microbioma Gastrointestinal/fisiología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/microbiología , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Humanos , Ratones
12.
J Immunol ; 196(2): 703-14, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26673140

RESUMEN

Epidemiologic studies in humans have demonstrated that infection with helminth parasites is associated with a reduced risk of developing autoimmune diseases. Mechanistic studies in mice have linked the protective effect of helminths on autoimmunity to the suppressive activity of helminth-induced regulatory T cells (Tregs) or Th2 cells. In this study, we demonstrate that treatment of mice with Fasciola hepatica excretory-secretory products (FHES) attenuated the clinical signs of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Protection was associated with a significant reduction in the infiltration of pathogenic Th1 and Th17 cells into the brain. Although FHES enhanced anti-inflammatory cytokine and Th2 responses, protection against EAE was independent of IL-4, IL-10, and Tregs. However, administration of FHES induced production of the type 2 cytokines IL-33 and IL-5, which promoted accumulation of eosinophils. FHES-induced expansion of eosinophils and protection against EAE was lost in IL-33(-/-) mice and upon neutralization of IL-5. Furthermore, transfer of FHES-induced or IL-33-induced eosinophils conferred protection against EAE. In addition, treatment of mice with recombinant IL-33 attenuated autoimmunity, and this was dependent on IL-5. To our knowledge, this study is the first to report a role for helminth-induced IL-5 and IL-33 in protection against autoimmunity.


Asunto(s)
Antígenos Helmínticos/inmunología , Autoinmunidad/inmunología , Eosinofilia/inmunología , Interleucina-33/inmunología , Interleucina-5/inmunología , Células Th2/inmunología , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Citometría de Flujo , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
13.
Mediators Inflamm ; 2018: 8168717, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29805314

RESUMEN

The gut environment and gut microbiome dysbiosis have been demonstrated to significantly influence a range of disorders in humans, including obesity, diabetes, rheumatoid arthritis, and multiple sclerosis (MS). MS is an autoimmune disease affecting the central nervous system (CNS). The etiology of MS is not clear, and it should involve both genetic and extrinsic factors. The extrinsic factors responsible for predisposition to MS remain elusive. Recent studies on MS and its animal model, experimental autoimmune encephalomyelitis (EAE), have found that gastrointestinal microbiota may play an important role in the pathogenesis of MS/EAE. Thus, gut microbiome adjustment may be a future direction of treatment in MS. In this review, we discuss the characteristics of the gut microbiota, the connection between the brain and the gut, and the changes in gut microbiota in MS/EAE, and we explore the possibility of applying microbiota therapies in patients with MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Microbioma Gastrointestinal/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/microbiología , Animales , Humanos
14.
Nature ; 479(7374): 538-41, 2011 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22031325

RESUMEN

Active multiple sclerosis lesions show inflammatory changes suggestive of a combined attack by autoreactive T and B lymphocytes against brain white matter. These pathogenic immune cells derive from progenitors that are normal, innocuous components of the healthy immune repertoire but become autoaggressive upon pathological activation. The stimuli triggering this autoimmune conversion have been commonly attributed to environmental factors, in particular microbial infection. However, using the relapsing-remitting mouse model of spontaneously developing experimental autoimmune encephalomyelitis, here we show that the commensal gut flora-in the absence of pathogenic agents-is essential in triggering immune processes, leading to a relapsing-remitting autoimmune disease driven by myelin-specific CD4(+) T cells. We show further that recruitment and activation of autoantibody-producing B cells from the endogenous immune repertoire depends on availability of the target autoantigen, myelin oligodendrocyte glycoprotein (MOG), and commensal microbiota. Our observations identify a sequence of events triggering organ-specific autoimmune disease and these processes may offer novel therapeutic targets.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad/inmunología , Metagenoma/inmunología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Traslado Adoptivo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Encéfalo/inmunología , Encéfalo/patología , Movimiento Celular , Citocinas/inmunología , Dieta , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Vida Libre de Gérmenes/inmunología , Centro Germinal/citología , Centro Germinal/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Ratones , Esclerosis Múltiple Recurrente-Remitente/etiología , Proteínas de la Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Estómago/microbiología , Linfocitos T/inmunología
15.
Neurologia ; 32(3): 175-184, 2017 Apr.
Artículo en Inglés, Español | MEDLINE | ID: mdl-26383059

RESUMEN

INTRODUCTION: Multiple sclerosis (MS) is a demyelinating disease that affects young adults; in that age group, it represents the second leading cause of disability in our setting. Its precise aetiology has not been elucidated, but it is widely accepted to occur in genetically predisposed patients who are exposed to certain environmental factors. The discovery of the regulatory role played by intestinal microbiota in various autoimmune diseases has opened a new line of research in this field, which is discussed in this review. DEVELOPMENT: We reviewed published studies on the role of the microbiota in the development of both MS and its animal model, experimental autoimmune encephalomyelitis (EAE). In mice, it has been shown that intestinal microorganisms regulate the polarisation of T helper cells from Th1-Th17 up to Th2, the function of regulatory T cells, and the activity of B cells; they participate in the pathogenesis of EAE and contribute to its prevention and treatment. In contrast, evidence in humans is still scarce and mainly based on case-control studies that point to the presence of differences in certain bacterial communities. CONCLUSIONS: Multiple evidence points to the role of microbiota in EAE. Extrapolation of these results to MS is still in the early stages of research, and studies are needed to define which bacterial populations are associated with MS, the role they play in pathogenesis, and the therapeutic possibilities this knowledge offers us.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal/inmunología , Esclerosis Múltiple/microbiología , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/microbiología , Humanos , Ratones , Esclerosis Múltiple/inmunología
16.
Brain Behav Immun ; 50: 41-46, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26100487

RESUMEN

Infection with viral or bacterial pathogens has been linked with the development of multiple sclerosis (MS), while infection with helminth parasites has been associated protection against MS and other autoimmune diseases. Here we have used a murine model of MS, experimental autoimmune encephalomyelitis (EAE), to examine the effect of infection with the respiratory pathogen Bordetella pertussis infection on development of CNS inflammation. The data demonstrate that infection of mice with B. pertussis significantly attenuates the clinical course of EAE induced by active immunization or cell transfer. This was reflected in a significant reduction in VLA-4 and LFA-1 expression on T cells and infiltration of IL-17(+), IFN-γ(+) and IFN-γ(+)IL-17(+) CD4 T cells into the CNS. Infection with B. pertussis induced IL-10 production from dendritic cells in vitro and enhanced the frequency of IL-10-producing CD25(-)Foxp3(+/-) CD4(+) T cells in vivo. Furthermore, the suppressive effects of B. pertussis infection on EAE were lost in IL-10(-/-) mice. Our findings demonstrate that a bacterial infection of the respiratory tract can attenuate EAE by promoting production of the anti-inflammatory cytokine IL-10 that may suppress licensing of autoaggressive T cells in the lungs, thereby preventing their migration into the CNS.


Asunto(s)
Bordetella pertussis/patogenicidad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Interleucina-10/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Animales , Mediadores de Inflamación/metabolismo , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Células TH1/inmunología , Células Th17/inmunología , Células Th17/microbiología
17.
J Immunol ; 188(5): 2093-101, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22287719

RESUMEN

CFA is a strong adjuvant capable of stimulating cellular immune responses. Paradoxically, adjuvant immunotherapy by prior exposure to CFA or live mycobacteria suppresses the severity of experimental autoimmune encephalomyelitis (EAE) and spontaneous diabetes in rodents. In this study, we investigated immune responses during adjuvant immunotherapy of EAE. Induction of EAE in CFA-pretreated mice resulted in a rapid influx into the draining lymph nodes (dLNs) of large numbers of CD11b(+)Gr-1(+) myeloid cells, consisting of immature cells with ring-shaped nuclei, macrophages, and neutrophils. Concurrently, a population of mycobacteria-specific IFN-γ-producing T cells appeared in the dLNs. Immature myeloid cells in dLNs expressed the chemokines CXCL10 and CXCL16 in an IFN-γ-dependent manner. Subsequently, CD4(+) T cells coexpressing the cognate chemokine receptors CXCR3 and CXCR6 and myelin oligodendrocyte glycoprotein (MOG)-specific CD4(+) T cells accumulated within the chemokine-expressing dLNs, rather than within the CNS. Migration of CD4(+) T cells toward dLN cells was abolished by depleting the CD11b(+) cells and was also mediated by the CD11b(+) cells alone. In addition to altering the distribution of MOG-specific T cells, adjuvant treatment suppressed development of MOG-specific IL-17. Thus, adjuvant immunotherapy of EAE requires IFN-γ, which suppresses development of the Th17 response, and diverts autoreactive T cells away from the CNS toward immature myeloid cells expressing CXCL10 and CXCL16 in the lymph nodes.


Asunto(s)
Diferenciación Celular/inmunología , Quimiocina CXCL10/biosíntesis , Quimiocina CXCL6/biosíntesis , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Proteínas de la Mielina/inmunología , Células Mieloides/inmunología , Receptores CXCR3/biosíntesis , Receptores CXCR/biosíntesis , Subgrupos de Linfocitos T/inmunología , Animales , Inhibición de Migración Celular/inmunología , Quimiocina CXCL16 , Encefalomielitis Autoinmune Experimental/microbiología , Femenino , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/uso terapéutico , Interferón gamma/fisiología , Interferón gamma/uso terapéutico , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Glicoproteína Mielina-Oligodendrócito , Células Mieloides/microbiología , Células Mieloides/patología , Receptores CXCR6 , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
18.
Proc Natl Acad Sci U S A ; 108 Suppl 1: 4615-22, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20660719

RESUMEN

Although the effects of commensal bacteria on intestinal immune development seem to be profound, it remains speculative whether the gut microbiota influences extraintestinal biological functions. Multiple sclerosis (MS) is a devastating autoimmune disease leading to progressive deterioration of neurological function. Although the cause of MS is unknown, microorganisms seem to be important for the onset and/or progression of disease. However, it is unclear how microbial colonization, either symbiotic or infectious, affects autoimmunity. Herein, we investigate a role for the microbiota during the induction of experimental autoimmune encephalomyelitis (EAE), an animal model for MS. Mice maintained under germ-free conditions develop significantly attenuated EAE compared with conventionally colonized mice. Germ-free animals, induced for EAE, produce lower levels of the proinflammatory cytokines IFN-γ and IL-17A in both the intestine and spinal cord but display a reciprocal increase in CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Mechanistically, we show that gut dendritic cells from germ-free animals are reduced in the ability to stimulate proinflammatory T cell responses. Intestinal colonization with segmented filamentous bacteria (SFB) is known to promote IL-17 production in the gut; here, we show that SFBs also induced IL-17A-producing CD4(+) T cells (Th17) in the CNS. Remarkably, germ-free animals harboring SFBs alone developed EAE, showing that gut bacteria can affect neurologic inflammation. These findings reveal that the intestinal microbiota profoundly impacts the balance between pro- and antiinflammatory immune responses during EAE and suggest that modulation of gut bacteria may provide therapeutic targets for extraintestinal inflammatory diseases such as MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Intestinos/microbiología , Células Th17/inmunología , Animales , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Factores de Transcripción Forkhead/metabolismo , Vida Libre de Gérmenes , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Médula Espinal/metabolismo , Linfocitos T Reguladores/inmunología
19.
Microbiome ; 12(1): 114, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38915127

RESUMEN

BACKGROUND: Mediterranean diet rich in polyphenolic compounds holds great promise to prevent and alleviate multiple sclerosis (MS), a central nervous system autoimmune disease associated with gut microbiome dysbiosis. Health-promoting effects of natural polyphenols with low bioavailability could be attributed to gut microbiota reconstruction. However, its underlying mechanism of action remains elusive, resulting in rare therapies have proposed for polyphenol-targeted modulation of gut microbiota for the treatment of MS. RESULTS: We found that oral ellagic acid (EA), a natural polyphenol rich in the Mediterranean diet, effectively halted the progression of experimental autoimmune encephalomyelitis (EAE), the animal model of MS, via regulating a microbiota-metabolites-immunity axis. EA remodeled the gut microbiome composition and particularly increased the relative abundances of short-chain fatty acids -producing bacteria like Alloprevotella. Propionate (C3) was most significantly up-regulated by EA, and integrative modeling revealed a strong negative correlation between Alloprevotella or C3 and the pathological symptoms of EAE. Gut microbiota depletion negated the alleviating effects of EA on EAE, whereas oral administration of Alloprevotella rava mimicked the beneficial effects of EA on EAE. Moreover, EA directly promoted Alloprevotella rava (DSM 22548) growth and C3 production in vitro. The cell-free supernatants of Alloprevotella rava co-culture with EA suppressed Th17 differentiation by modulating acetylation in cell models. C3 can alleviate EAE development, and the mechanism may be through inhibiting HDAC activity and up-regulating acetylation thereby reducing inflammatory cytokines secreted by pathogenic Th17 cells. CONCLUSIONS: Our study identifies EA as a novel and potentially effective prebiotic for improving MS and other autoimmune diseases via the microbiota-metabolites-immunity axis. Video Abstract.


Asunto(s)
Ácido Elágico , Encefalomielitis Autoinmune Experimental , Microbioma Gastrointestinal , Esclerosis Múltiple , Propionatos , Ácido Elágico/farmacología , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/microbiología , Propionatos/metabolismo , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/microbiología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Femenino , Autoinmunidad/efectos de los fármacos , Disbiosis/microbiología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/inmunología , Humanos , Administración Oral
20.
J Autoimmun ; 40: 45-57, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22939403

RESUMEN

Heat shock proteins (Hsps) participate in the cellular response to stress and they are hiperexpressed in inflammatory conditions. They are also known to play a major role in immune modulation, controlling, for instance, autoimmune responses. In this study, we showed that oral administration of a recombinant Lactococcus lactis strain that produces and releases LPS-free Hsp65 prevented the development of experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice. This was confirmed by the reduced inflammatory cell infiltrate and absence of injury signs in the spinal cord. The effect was associated with reduced IL-17 and increased IL-10 production in mesenteric lymph node and spleen cell cultures. Hsp65-producing-L. lactis-fed mice had a remarkable increase in the number of natural and inducible CD4+Foxp3+ regulatory T (Treg) cells and CD4+LAP+ (Latency-associated peptide) Tregs - which express the membrane-bound TGF-ß - in spleen, inguinal and mesenteric lymph nodes as well as in spinal cord. Moreover, many Tregs co-expressed Foxp3 and LAP. In vivo depletion of LAP+ cells abrogated the effect of Hsp65-producing L. lactis in EAE prevention and worsened disease in medium-fed mice. Thus, Hsp65-L.lactis seems to boost this critical regulatory circuit involved in controlling EAE development in mice.


Asunto(s)
Proteínas Bacterianas/metabolismo , Chaperonina 60/metabolismo , Encefalomielitis Autoinmune Experimental , Lactococcus lactis/metabolismo , Mycobacterium leprae/genética , Linfocitos T Reguladores/metabolismo , Animales , Autoinmunidad , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Antígenos CD4/metabolismo , Chaperonina 60/biosíntesis , Chaperonina 60/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Factores de Transcripción Forkhead/metabolismo , Lactococcus lactis/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Médula Espinal/inmunología , Médula Espinal/metabolismo , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA