Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Gut Microbes ; 14(1): 2127446, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36179318

RESUMEN

The etiopathogenesis of multiple sclerosis (MS) is strongly affected by environmental factors such as diet and the gut microbiota. An isoflavone-rich (ISO) diet was previously shown to reduce the severity of MS in the animal model experimental autoimmune encephalomyelitis (EAE). Translation of this concept to clinical trial where dietary isoflavones may be recommended for MS patients will require preliminary evidence that providing the isoflavone-rich diet to people with MS (PwMS) who lack phytoestrogen-metabolizing bacteria has beneficial effects. We have previously shown that the gut microbiota of PwMS resembles the gut microbiota of mice raised under a phytoestrogen-free (phyto-free) diet in that it lacks phytoestrogen-metabolizing bacteria. To investigate the effects of phytoestrogens on the microbiota inflammatory response and EAE disease severity we switched the diet of mice raised under a phyto-free (PF) diet to an isoflavone-rich diet. Microbiota analysis showed that the change in diet from one that is ISO to one that is PF reduces beneficial bacteria such as Bifidobacterium species. In addition we observed functional differences in lipopolysaccharide (LPS) biosynthesis pathways. Moreover LPS extracted from feces of mice fed an ISO diet induced increased production of anti-inflammatory cytokines from bone marrow-derived macrophages relative to fecal-LPS isolated from mice fed a PF diet. Eventually mice whose diet was switched from a PF diet to an ISO diet trended toward reduced EAE severity and mortality. Overall we show that an isoflavone-rich diet specifically modulates LPS biosynthesis of the gut microbiota imparts an anti-inflammatory response and decreases disease severity.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Microbioma Gastrointestinal , Isoflavonas , Animales , Citocinas/metabolismo , Dieta , Encefalomielitis Autoinmune Experimental/microbiología , Inflamación , Isoflavonas/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Fitoestrógenos/farmacología
2.
J Neuroimmunol ; 360: 577700, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34482269

RESUMEN

Gut microbiota composition may affect the central nervous system (CNS) and immune function. Several studies have recently examined the possible link between gut microbiota composition and multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Most of these studies agree that patients with MS suffer from dysbiosis. Moreover, an altered proportion of certain phyla of bacteria was detected in the digestive tracts of these patients compared to healthy individuals. This review article gathers information from research papers that have examined the relationship between gut microbiota composition and MS and its possible mechanisms.


Asunto(s)
Eje Cerebro-Intestino , Disbiosis/complicaciones , Encefalomielitis Autoinmune Experimental/microbiología , Microbioma Gastrointestinal , Esclerosis Múltiple/microbiología , Animales , Eje Cerebro-Intestino/inmunología , Eje Cerebro-Intestino/fisiología , Modelos Animales de Enfermedad , Disbiosis/fisiopatología , Disbiosis/terapia , Encefalomielitis Autoinmune Experimental/fisiopatología , Trasplante de Microbiota Fecal , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Esclerosis Múltiple/etiología , Esclerosis Múltiple/fisiopatología , Esclerosis Múltiple/terapia , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/microbiología , Probióticos , Ratas , Vitamina D/uso terapéutico
3.
Front Immunol ; 12: 628629, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33796102

RESUMEN

Exercise therapy including endurance training and resistance training is a promising non-pharmacological therapy in patients with multiple sclerosis (MS). Recent studies have revealed that exercise exerts beneficial impacts on gut microbiota. However, the role of gut microbiota in the immune benefits of strength exercise (SE; one of resistance training) in central nervous system (CNS) autoimmunity is barely known. Here, we observed that 60-min SE ameliorated disease severity and neuropathology in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. SE increased the abundance and diversity of the gut microbiota, and decreased Firmicutes/Bacteroidetes ratio (F/B ratio) and intestinal mucosal permeability, and enrichment of several short-chain fatty acid (SCFA)-producing bacteria. Furthermore, SE reduced Th17 responses and increased Treg responses in the small intestine lymphoid tissues. Compared to the control group, microbiota-depleted mice receiving SE microbiome fecal transplants had lower disease severity and neuropathology scores. These results uncovered a protective role of SE in neuroimmunomodulation effects partly via changes to the gut microbiome.


Asunto(s)
Autoinmunidad , Bacterias/metabolismo , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Microbioma Gastrointestinal , Intestino Delgado/microbiología , Condicionamiento Físico Animal , Entrenamiento de Fuerza , Animales , Bacterias/inmunología , Disbiosis , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Intestino Delgado/inmunología , Ratones Endogámicos C57BL , Neuroinmunomodulación , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
4.
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
5.
Front Immunol ; 12: 623451, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679767

RESUMEN

Multiple sclerosis is a chronic autoimmune disease involving the central nervous system, and shows a high disability rate. Its pathogenesis is complicated, and there is no good treatment. In recent years, with in-depth studies on the regulation of gastrointestinal flora, the relationship between the mammalian immune system and the intestinal flora has been extensively explored. Changes in the composition and structure of the gastrointestinal flora can affect the characteristics and development of the host immune system and even induce a series of central nervous system inflammation events. The occurrence and development of multiple sclerosis are closely related to the continuous destruction of the intestinal barrier caused by intestinal dysbacteriosis. In this study, we analyzed Lactobacillus acidipiscis in a mouse model of experimental autoimmune encephalomyelitis (EAE). We found that the amount of L. acidipiscis in the intestinal tract was inversely proportional to the progress of EAE development. In addition, the number of CD4+ FOXP3+ regulatory T cells in the mesenteric lymph nodes of mice increased significantly after the mice were fed with L. acidipiscis, and the differentiation of CD4+ T cells to Th1 and Th17 cells was inhibited. However, the protective effect of L. acidipiscis was lost in γδ T cell-deficient mice and hence was concluded to depend on the presence of regulatory γδ T cells in the intestinal epithelium. Moreover, including L. acidipiscis enhanced the development of Vγ1+γδ T cells but suppressed that of Vγ4+γδ T cells. In summary, our results demonstrated the ability of L. acidipiscis to induce generation of regulatory γδ T cells that suppress the development of the encephalomyelitic Th1 and Th17 cells and the progress of EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Microbioma Gastrointestinal , Mucosa Intestinal/microbiología , Linfocitos Intraepiteliales/microbiología , Lactobacillus/inmunología , Probióticos , Animales , Diferenciación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Disbiosis , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/microbiología , Femenino , Genes Codificadores de la Cadena gamma de los Receptores de Linfocito T , Interacciones Huésped-Patógeno , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Lactobacillus/crecimiento & desarrollo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/microbiología , Células Th17/inmunología , Células Th17/metabolismo , Células Th17/microbiología
6.
Sci Immunol ; 6(56)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547052

RESUMEN

Interleukin-17A- (IL-17A) and IL-17F-producing CD4+ T helper cells (TH17 cells) are implicated in the development of chronic inflammatory diseases, such as multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). TH17 cells also orchestrate leukocyte invasion of the central nervous system (CNS) and subsequent tissue damage. However, the role of IL-17A and IL-17F as effector cytokines is still confused with the encephalitogenic function of the cells that produce these cytokines, namely, TH17 cells, fueling a long-standing debate in the neuroimmunology field. Here, we demonstrated that mice deficient for IL-17A/F lose their susceptibility to EAE, which correlated with an altered composition of their gut microbiota. However, loss of IL-17A/F in TH cells did not diminish their encephalitogenic capacity. Reconstitution of a wild-type-like intestinal microbiota or reintroduction of IL-17A specifically into the gut epithelium of IL-17A/F-deficient mice reestablished their susceptibility to EAE. Thus, our data demonstrated that IL-17A and IL-17F are not encephalitogenic mediators but rather modulators of intestinal homeostasis that indirectly alter CNS-directed autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal/inmunología , Interleucina-17/metabolismo , Esclerosis Múltiple/inmunología , Traslado Adoptivo , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Trasplante de Microbiota Fecal , Femenino , Humanos , Interleucina-17/genética , Masculino , Ratones , Ratones Noqueados , Esclerosis Múltiple/patología , Células Th17/inmunología , Células Th17/trasplante
7.
Gut Microbes ; 12(1): 1857515, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33382357

RESUMEN

Multiple sclerosis (MS) is a CNS autoimmune disease characterized by demyelination and inflammatory infiltration with a high disability rate. Increasing evidence has demonstrated the importance of gut microbiota as an environmental risk factor in MS and its animal model experimental autoimmune encephalomyelitis (EAE). Diet is the main determinant of gut microbiota composition and function, which greatly affects the shaping of microbial structure. Pomegranate peel, a waste product in the production of juice, is rich in health-promoting compounds. However, its individual constituents, immunoregulatory activities, and action associated with bacterial diversity in the gut microbiota are largely unknown. Here, the main nutrient ingredients of pomegranate peel extract (PPE) were identified as phenols, flavonoids, amino acids, carbohydrates, fatty acids, lipids, nucleotides, organic acids, alcohols, and vitamins via metabolomics evaluation. We found, for the first time, oral PPE (100 mg/kg/day) not only effectively relieves EAE, inhibits CNS inflammatory factor infiltration and myelin loss, but also reshapes gut microbiota. Furthermore, recipient EAE mice with fecal transplantation from the PPE-treated donor delayed the disease development significantly. 16S rRNA gene sequencing revealed the increased gut microbiota richness in PPE-treated group. Among them, Lactobacillaceae enriched significantly, while Alcaligenaceae and Acidaminococcacea decreased remarkably. In conclusion, our data demonstrated that gut microbiota mediated the beneficial effects of oral PPE on EAE, and provided new ideas for developing the prebiotic value of pomegranate peel for the treatment of autoimmune diseases.


Asunto(s)
Bacterias/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Esclerosis Múltiple/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Granada (Fruta)/química , Residuos/análisis , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/microbiología , Femenino , Frutas/química , Humanos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/microbiología , Extractos Vegetales/química
8.
Front Immunol ; 11: 578648, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33362764

RESUMEN

Gut microbiota has emerged as an important environmental factor in the pathobiology of multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system (CNS). Both genetic and environmental factors have been shown to play an important role in MS. Among genetic factors, the human leukocyte antigen (HLA) class II allele such as HLA-DR2, DR3, DR4, DQ6, and DQ8 show the association with the MS. We have previously used transgenic mice expressing MS susceptible HLA class II allele such as HLA-DR2, DR3, DQ6, and DQ8 to validate significance of HLA alleles in MS. Although environmental factors contribute to 2/3 of MS risk, less is known about them. Gut microbiota is emerging as an imporatnt environmental factor in MS pathogenesis. We and others have shown that MS patients have distinct gut microbiota compared to healthy control (HC) with a lower abundance of Prevotella. Additionally, the abundance of Prevotella increased in patients receiving disease-modifying therapies (DMTs) such as Copaxone and/or Interferon-beta (IFNß). We have previously identified a specific strain of Prevotella (Prevotella histicola), which can suppress experimental autoimmune encephalomyelitis (EAE) disease in HLA-DR3.DQ8 transgenic mice. Since Interferon-ß-1b [IFNß (Betaseron)] is a major DMTs used in MS patients, we hypothesized that treatment with the combination of P. histicola and IFNß would have an additive effect on the disease suppression. We observed that treatment with P. histicola suppressed disease as effectively as IFNß. Surprisingly, the combination of P. histicola and IFNß was not more effective than either treatment alone. P. histicola alone or in combination with IFNß increased the frequency and number of CD4+FoxP3+ regulatory T cells in the gut-associated lymphoid tissue (GALT). Treatment with P. histicola alone, IFNß alone, and in the combination decreased frequency of pro-inflammatory IFN-γ and IL17-producing CD4+ T cells in the CNS. Additionally, P. histicola alone or IFNß alone or the combination treatments decreased CNS pathology, characterized by reduced microglia and astrocytic activation. In conclusion, our study indicates that the human gut commensal P. histicola can suppress disease as effectively as commonly used MS drug IFNß and may provide an alternative treatment option for MS patients.


Asunto(s)
Antiinflamatorios/farmacología , Encefalomielitis Autoinmune Experimental/prevención & control , Microbioma Gastrointestinal , Interferón beta/farmacología , Intestinos/microbiología , Prevotella/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/metabolismo , Astrocitos/microbiología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/microbiología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/microbiología , Femenino , Factores de Transcripción Forkhead/metabolismo , Cadenas beta de HLA-DQ/genética , Cadenas HLA-DRB1/genética , Humanos , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Tejido Linfoide/microbiología , Masculino , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Microglía/microbiología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/microbiología
9.
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
10.
Benef Microbes ; 11(7): 669-684, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-33045841

RESUMEN

Multiple sclerosis (MS) is a neuro-inflammatory autoimmune disease of the central nervous system (CNS) that affects young adults. It is characterised by the development of demyelinating lesions and inflammation within the CNS. Although the causes of MS are still elusive, recent work using patient samples and experimental animal models has demonstrated a strong relationship between the gut microbiota and its contribution to CNS inflammation and MS. While there is no cure for MS, alteration of the gut microbiota composition through the use of probiotics is a very promising treatment. However, while most recent works have focused on the use of probiotics to modify pre-existing disease, little is known about its role in protecting from the establishment of MS. In this study, we determined whether colonisation with the probiotic bacterium Escherichia coli strain Nissle 1917 (EcN) could be used as a prophylactic strategy to prevent or alter the development of experimental autoimmune encephalomyelitis (EAE), a preclinical model of MS. We found that double gavage (two doses) of EcN before induction of EAE delayed disease onset and decreased disease severity. We also found that EcN-treated mice had decreased amounts of perivascular cuffing, CD4+ T cell infiltration into the CNS, together with significantly decreased absolute numbers of Th1 cells, and reduced activation of microglia. Although further studies are necessary to comprehend the exact protective mechanisms induced, our study supports a promising use of EcN as a probiotic for the prevention of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Escherichia coli/fisiología , Tracto Gastrointestinal/microbiología , Probióticos/administración & dosificación , Animales , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Recuento de Colonia Microbiana , Citocinas/sangre , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Escherichia coli/crecimiento & desarrollo , Heces/microbiología , Tracto Gastrointestinal/efectos de los fármacos , Inflamación , Ratones , Probióticos/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
11.
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
12.
Sci Rep ; 10(1): 15183, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938979

RESUMEN

The gut microbiome is known to be sensitive to changes in the immune system, especially during autoimmune diseases such as Multiple Sclerosis (MS). Our study examines the changes to the gut microbiome that occur during experimental autoimmune encephalomyelitis (EAE), an animal model for MS. We collected fecal samples at key stages of EAE progression and quantified microbial abundances with 16S V3-V4 amplicon sequencing. Our analysis of the data suggests that the abundance of commensal Lactobacillaceae decreases during EAE while other commensal populations belonging to the Clostridiaceae, Ruminococcaceae, and Peptostreptococcaceae families expand. Community analysis with microbial co-occurrence networks points to these three expanding taxa as potential mediators of gut microbiome dysbiosis. We also employed PICRUSt2 to impute MetaCyc Enzyme Consortium (EC) pathway abundances from the original microbial abundance data. From this analysis, we found that a number of imputed EC pathways responsible for the production of immunomodulatory compounds appear to be enriched in mice undergoing EAE. Our analysis and interpretation of results provides a detailed picture of the changes to the gut microbiome that are occurring throughout the course of EAE disease progression and helps to evaluate EAE as a viable model for gut dysbiosis in MS patients.


Asunto(s)
Clostridiaceae/fisiología , Disbiosis/microbiología , Encefalomielitis Autoinmune Experimental/microbiología , Heces/microbiología , Microbioma Gastrointestinal/genética , Lactobacillaceae/fisiología , Esclerosis Múltiple/microbiología , Peptostreptococcus/fisiología , ARN Ribosómico 16S/genética , Ruminococcus/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunomodulación , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
13.
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
14.
J Leukoc Biol ; 108(4): 1129-1137, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32779279

RESUMEN

Multiple sclerosis is a chronic and demyelinating disease of the central nervous system (CNS), most prevalent in women, and with an important social and economic cost worldwide. It is triggered by self-reacting lymphocytes that infiltrate the CNS and initiate neuroinflammation. Further, axonal loss and neuronal death takes place, leading to neurodegeneration and brain atrophy. The murine model for studying MS, experimental autoimmune encephalomyelitis (EAE), consists in immunizing mice with myelin-derived epitopes. APCs activate encephalitogenic T CD4 and CD8 lymphocytes that migrate mainly to the spinal cord resulting in neuroinflammation. Most of the knowledge on the pathophysiology and treatment of MS was obtained from EAE experiments, as Th17 cells, anti-alpha4 blocking Abs and the role of microbiota. Conversely, recent technology breakthroughs, such as CyTOF and single-cell RNA-seq, promise to revolutionize our understanding on the mechanisms involved both in MS and EAE. In fact, the importance of specific cellular populations and key molecules in MS/EAE is a constant matter of debate. It is well accepted that both Th1 and Th17 T CD4 lymphocytes play a relevant role in disease initiation after re-activation in situ. What is still under constant investigation, however, is the plasticity of the lymphocyte population, and the individual contribution of both resident and inflammatory cells for the progression or recovery of the disease. Thus, in this review, new findings obtained after single-cell analysis of blood and central nervous system infiltrating cells from MS/EAE and how they have contributed to a better knowledge on the cellular and molecular mechanisms of neuroinflammation are discussed.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Análisis de la Célula Individual , Células TH1/inmunología , Células Th17/inmunología , Animales , Linfocitos T CD8-positivos/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Humanos , Ratones , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Células TH1/patología , Células Th17/patología
15.
Life Sci ; 253: 117747, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32376270

RESUMEN

AIMS: Multiple sclerosis (MS) whose pathogenesis is still unclear is a chronic progressive disease in the central nervous system. Gut microbiota can directly or indirectly affect the immune system through the brain gut axis to engage in the occurrence and development of the disease. MATERIALS AND METHODS: C57BL/6 mice which were immunized by MOG35-55 to prepare experimental autoimmune encephalomyelitis (EAE) animal models were treated with rapamycin and MCC950 (CP-456773) in combination or separately. After sequencing the 16S rRNA V4 region of gut microbiota, the species, abundance and composition of gut microbiota were analyzed by Alpha diversity, Bata diversity and LEfSe analysis. The pathological changes and the expression of CD4 and CD8 of brain, large intestine and spleen were detected. KEY FINDINGS: The results showed that rapamycin and MCC950 could alleviate the progression of the disease by inducing autophagy and inhibiting the immune response. The Alpha diversity of EAE model group was no significant difference compering to control group while the number of OTUs was decreased. After the treatment by rapamycin and MCC950, the abundance and composition of gut microbiota was relatively recovered, which was close to that of normal mice. SIGNIFICANCE: Inhibiting immune cell-mediated inflammation and restoring the composition of gut microbiota may help to alleviate the clinical symptoms of multiple sclerosis. Furthermore, to research the regulatory effect between immune response and gut microbiota may be a new strategy for the prevention and treatment of multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Furanos/farmacología , Microbioma Gastrointestinal/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Sirolimus/farmacología , Sulfonamidas/farmacología , Animales , Encéfalo/inmunología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Femenino , Furanos/administración & dosificación , Compuestos Heterocíclicos de 4 o más Anillos , Indenos , Inflamación/inmunología , Inflamación/patología , Intestino Grueso/inmunología , Intestino Grueso/patología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/microbiología , ARN Ribosómico 16S , Sirolimus/administración & dosificación , Bazo/inmunología , Bazo/patología , Sulfonamidas/administración & dosificación , Sulfonas
16.
Nat Commun ; 11(1): 1913, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32312989

RESUMEN

The TAGAP gene locus has been linked to several infectious diseases or autoimmune diseases, including candidemia and multiple sclerosis. While previous studies have described a role of TAGAP in T cells, much less is known about its function in other cell types. Here we report that TAGAP is required for Dectin-induced anti-fungal signaling and proinflammatory cytokine production in myeloid cells. Following stimulation with Dectin ligands, TAGAP is phosphorylated by EPHB2 at tyrosine 310, which bridges proximal Dectin-induced EPHB2 activity to downstream CARD9-mediated signaling pathways. During Candida albicans infection, mice lacking TAGAP mount defective immune responses, impaired Th17 cell differentiation, and higher fungal burden. Similarly, in experimental autoimmune encephalomyelitis model of multiple sclerosis, TAGAP deficient mice develop significantly attenuated disease. In summary, we report that TAGAP plays an important role in linking Dectin-induced signaling to the promotion of effective T helper cell immune responses, during both anti-fungal host defense and autoimmunity.


Asunto(s)
Antifúngicos/inmunología , Candidiasis/inmunología , Diferenciación Celular , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/metabolismo , Receptor EphB2/metabolismo , Transducción de Señal/efectos de los fármacos , Células Th17/metabolismo , Animales , Antifúngicos/farmacología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/microbiología , Femenino , Proteínas Activadoras de GTPasa/genética , Humanos , Lectinas Tipo C/metabolismo , Masculino , Ratones Noqueados , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/inmunología , Fosforilación , Receptor EphB2/inmunología , Receptores Inmunológicos , Receptores de Reconocimiento de Patrones/metabolismo , Células Th17/inmunología
17.
Nat Commun ; 10(1): 4877, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31653831

RESUMEN

The interaction between the mammalian host and its resident gut microbiota is known to license adaptive immune responses. Nutritional constituents strongly influence composition and functional properties of the intestinal microbial communities. Here, we report that omission of a single essential amino acid - tryptophan - from the diet abrogates CNS autoimmunity in a mouse model of multiple sclerosis. Dietary tryptophan restriction results in impaired encephalitogenic T cell responses and is accompanied by a mild intestinal inflammatory response and a profound phenotypic shift of gut microbiota. Protective effects of dietary tryptophan restriction are abrogated in germ-free mice, but are independent of canonical host sensors of intracellular tryptophan metabolites. We conclude that dietary tryptophan restriction alters metabolic properties of gut microbiota, which in turn have an impact on encephalitogenic T cell responses. This link between gut microbiota, dietary tryptophan and adaptive immunity may help to develop therapeutic strategies for protection from autoimmune neuroinflammation.


Asunto(s)
Autoinmunidad/inmunología , Dieta , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal/inmunología , Linfocitos T/inmunología , Triptófano , Animales , Proteínas en la Dieta , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/microbiología , Microbioma Gastrointestinal/genética , Ratones , Esclerosis Múltiple , ARN Ribosómico 16S/genética
18.
Sci Rep ; 9(1): 12460, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31462662

RESUMEN

Diseases are often diagnosed once overt symptoms arise, ignoring the prior latent period when effective prevention may be possible. Experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis, exhibits such disease latency, but the molecular processes underlying this asymptomatic period remain poorly characterized. Gut microbes also influence EAE severity, yet their impact on the latent period remains unknown. Here, we show the latent period between immunization and EAE's overt symptom onset is characterized by distinct host responses as measured by stool proteomics. In particular, we found a transient increase in protease inhibitors that inversely correlated with disease severity. Vancomycin administration attenuated both EAE symptoms and protease inhibitor induction potentially by decreasing immune system reactivity, supporting a subset of the microbiota's role in modulating the host's latent period response. These results strengthen previous evidence of proteases and their inhibitors in EAE and highlight the utility stool-omics for revealing complex, dynamic biology.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Heces/microbiología , Microbioma Gastrointestinal , Regulación de la Expresión Génica , Esclerosis Múltiple , Inhibidores de Proteasas/metabolismo , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/patología , Proteómica , Vancomicina/farmacología
19.
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
20.
Neuropharmacology ; 157: 107685, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31247271

RESUMEN

Emerging evidence indicates that gut microbiota interacts with immune and nervous systems in the host and plays a critical role in the pathogenesis of multiple sclerosis (MS) and many psychiatric disorders such as depression and anxiety. The aim of this study was to explore the influence of gut bacterial depletion from early adolescence on adult immunological and neurobehavioral responses in mice with experimental-autoimmune-encephalomyelitis (EAE). We used an animal model of gut microbiota depletion induced by antibiotics from weaning to adulthood to assess clinical signs, cognitive function and depression-and anxiety-related symptoms in non-EAE and EAE-induced mice. We measured levels of interferon (IFN)-γ, interleukin (IL)-17A and IL-10 in serum, and BDNF, IL-1ß and tumor necrosis factor (TNF)-α) in the hippocampus. Antibiotic-treated mice displayed a significant delay in the onset of clinical symptoms of EAE. However, a higher severity of EAE was found between days 19-22 post-immunization in antibiotics-treated mice, while a reduction in the clinical signs of MS was observed at days 24-25 post-immunization. Antibiotic administration decreased IFN-γ and IL-17A levels and increased IL-10 in serum of EAE-induced mice. Antibiotic treatment significantly decreased hippocampal BDNF and enhanced learning and memory impairments in EAE-induced mice. However, no significant changes were found in non-EAE mice. Non-EAE and EAE mice treated with antibiotics exhibited increased anxiety-related behaviors, whereas depression-related symptoms and increased hippocampal TNF-α and IL-1ß were only observed in EAE-induced mice treated with antibiotics. This study supports the view that depletion of gut microbiota by antibiotics from weaning profoundly impacts adult immunological and neurobehavioral responses.


Asunto(s)
Ansiedad/microbiología , Depresión/microbiología , Encefalomielitis Autoinmune Experimental/microbiología , Microbioma Gastrointestinal/fisiología , Trastornos de la Memoria/microbiología , Esclerosis Múltiple/microbiología , Animales , Antibacterianos/farmacología , Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/complicaciones , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Hipocampo/metabolismo , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Trastornos de la Memoria/complicaciones , Ratones , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...