Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Neurosci ; 40(5): 1162-1173, 2020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31889008

RESUMEN

Recovery after stroke is a multicellular process encompassing neurons, resident immune cells, and brain-invading cells. Stroke alters the gut microbiome, which in turn has considerable impact on stroke outcome. However, the mechanisms underlying gut-brain interaction and implications for long-term recovery are largely elusive. Here, we tested the hypothesis that short-chain fatty acids (SCFAs), key bioactive microbial metabolites, are the missing link along the gut-brain axis and might be able to modulate recovery after experimental stroke. SCFA supplementation in the drinking water of male mice significantly improved recovery of affected limb motor function. Using in vivo wide-field calcium imaging, we observed that SCFAs induced altered contralesional cortex connectivity. This was associated with SCFA-dependent changes in spine and synapse densities. RNA sequencing of the forebrain cortex indicated a potential involvement of microglial cells in contributing to the structural and functional remodeling. Further analyses confirmed a substantial impact of SCFAs on microglial activation, which depended on the recruitment of T cells to the infarcted brain. Our findings identified that microbiota-derived SCFAs modulate poststroke recovery via effects on systemic and brain resident immune cells.SIGNIFICANCE STATEMENT Previous studies have shown a bidirectional communication along the gut-brain axis after stroke. Stroke alters the gut microbiota composition, and in turn, microbiota dysbiosis has a substantial impact on stroke outcome by modulating the immune response. However, until now, the mediators derived from the gut microbiome affecting the gut-immune-brain axis and the molecular mechanisms involved in this process were unknown. Here, we demonstrate that short-chain fatty acids, fermentation products of the gut microbiome, are potent and proregenerative modulators of poststroke neuronal plasticity at various structural levels. We identified that this effect was mediated via circulating lymphocytes on microglial activation. These results identify short-chain fatty acids as a missing link along the gut-brain axis and as a potential therapeutic to improve recovery after stroke.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Ácidos Grasos Volátiles/administración & dosificación , Accidente Cerebrovascular/inmunología , Animales , Encéfalo/metabolismo , Femenino , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/metabolismo , Transcriptoma/efectos de los fármacos
2.
Gastroenterology ; 154(3): 624-636, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29031500

RESUMEN

BACKGROUND & AIMS: The enteric nervous system (ENS) regulates gastrointestinal function via different subtypes of neurons, organized into fine-tuned neural circuits. It is not clear how cell diversity is created within the embryonic ENS; information required for development of cell-based therapies and models of enteric neuropathies. We aimed to identify proteins that regulate ENS differentiation and network formation. METHODS: We generated and compared RNA expression profiles of the entire ENS, ENS progenitor cells, and non-ENS gut cells of mice, collected at embryonic days 11.5 and 15.5, when different subtypes of neurons are formed. Gastrointestinal tissues from R26ReYFP reporter mice crossed to Sox10-CreERT2 or Wnt1-Cre mice were dissected and the 6 populations of cells were isolated by flow cytometry. We used histochemistry to map differentially expressed proteins in mouse and human gut tissues at different stages of development, in different regions. We examined enteric neuronal diversity and gastric function in Wnt1-Cre x Sox6fl/fl mice, which do not express the Sox6 gene in the ENS. RESULTS: We identified 147 transcription and signaling factors that varied in spatial and temporal expression during development of the mouse ENS. Of the factors also analyzed in human ENS, most were conserved. We uncovered 16 signaling pathways (such as fibroblast growth factor and Eph/ephrin pathways). Transcription factors were grouped according to their specific expression in enteric progenitor cells (such as MEF2C), enteric neurons (such as SOX4), or neuron subpopulations (such as SATB1 and SOX6). Lack of SOX6 in the ENS reduced the numbers of gastric dopamine neurons and delayed gastric emptying. CONCLUSIONS: Using transcriptome and histochemical analyses of the developing mouse and human ENS, we mapped expression patterns of transcription and signaling factors. Further studies of these candidate determinants might elucidate the mechanisms by which enteric stem cells differentiate into neuronal subtypes and form distinct connectivity patterns during ENS development. We found expression of SOX6 to be required for development of gastric dopamine neurons.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Sistema Nervioso Entérico/metabolismo , Transducción de Señal , Estómago/inervación , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Comunicación Autocrina , Sistema Nervioso Entérico/embriología , Vaciamiento Gástrico , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genotipo , Edad Gestacional , Humanos , Ratones Noqueados , Comunicación Paracrina , Fenotipo , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Especificidad de la Especie , Factores de Transcripción/genética
3.
J Neurosci ; 36(28): 7428-40, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27413153

RESUMEN

UNLABELLED: Acute brain ischemia induces a local neuroinflammatory reaction and alters peripheral immune homeostasis at the same time. Recent evidence has suggested a key role of the gut microbiota in autoimmune diseases by modulating immune homeostasis. Therefore, we investigated the mechanistic link among acute brain ischemia, microbiota alterations, and the immune response after brain injury. Using two distinct models of acute middle cerebral artery occlusion, we show by next-generation sequencing that large stroke lesions cause gut microbiota dysbiosis, which in turn affects stroke outcome via immune-mediated mechanisms. Reduced species diversity and bacterial overgrowth of bacteroidetes were identified as hallmarks of poststroke dysbiosis, which was associated with intestinal barrier dysfunction and reduced intestinal motility as determined by in vivo intestinal bolus tracking. Recolonizing germ-free mice with dysbiotic poststroke microbiota exacerbates lesion volume and functional deficits after experimental stroke compared with the recolonization with a normal control microbiota. In addition, recolonization of mice with a dysbiotic microbiome induces a proinflammatory T-cell polarization in the intestinal immune compartment and in the ischemic brain. Using in vivo cell-tracking studies, we demonstrate the migration of intestinal lymphocytes to the ischemic brain. Therapeutic transplantation of fecal microbiota normalizes brain lesion-induced dysbiosis and improves stroke outcome. These results support a novel mechanism in which the gut microbiome is a target of stroke-induced systemic alterations and an effector with substantial impact on stroke outcome. SIGNIFICANCE STATEMENT: We have identified a bidirectional communication along the brain-gut microbiota-immune axis and show that the gut microbiota is a central regulator of immune homeostasis. Acute brain lesions induced dysbiosis of the microbiome and, in turn, changes in the gut microbiota affected neuroinflammatory and functional outcome after brain injury. The microbiota impact on immunity and stroke outcome was transmissible by microbiota transplantation. Our findings support an emerging concept in which the gut microbiota is a key regulator in priming the neuroinflammatory response to brain injury. These findings highlight the key role of microbiota as a potential therapeutic target to protect brain function after injury.


Asunto(s)
Disbiosis/etiología , Encefalitis/complicaciones , Encefalitis/etiología , Microbiota/fisiología , Accidente Cerebrovascular/complicaciones , Animales , Infarto Encefálico/etiología , Complejo CD3/metabolismo , Modelos Animales de Enfermedad , Disbiosis/inmunología , Disbiosis/microbiología , Heces/microbiología , Femenino , Enfermedades Gastrointestinales/etiología , Motilidad Gastrointestinal/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ileus/inmunología , Ileus/microbiología , Ileus/patología , Infarto de la Arteria Cerebral Media/complicaciones , Leucocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microbiota/inmunología , Accidente Cerebrovascular/etiología , Estructuras Linfoides Terciarias/patología
4.
J Neurosci ; 36(15): 4339-50, 2016 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-27076429

RESUMEN

The enteric nervous system (ENS) is organized into neural circuits within the gastrointestinal wall where it controls the peristaltic movements, secretion, and blood flow. Although proper gut function relies on the complex neuronal composition of the ENS, little is known about the transcriptional networks that regulate the diversification into different classes of enteric neurons and glia during development. Here we redefine the role of Ascl1 (Mash1), one of the few regulatory transcription factors described during ENS development. We show that enteric glia and all enteric neuronal subtypes appear to be derived from Ascl1-expressing progenitor cells. In the gut of Ascl1(-/-) mutant mice, neurogenesis is delayed and reduced, and posterior gliogenesis impaired. The ratio of neurons expressing Calbindin, TH, and VIP is selectively decreased while, for instance, 5-HT(+) neurons, which previously were believed to be Ascl1-dependent, are formed in normal numbers. Essentially the same differentiation defects are observed in Ascl1(KINgn2) transgenic mutants, where the proneural activity of Ngn2 replaces Ascl1, demonstrating that Ascl1 is required for the acquisition of specific enteric neuronal subtype features independent of its role in neurogenesis. In this study, we provide novel insights into the expression and function of Ascl1 in the differentiation process of specific neuronal subtypes during ENS development. SIGNIFICANCE STATEMENT: The molecular mechanisms underlying the generation of different neuronal subtypes during development of the enteric nervous system are poorly understood despite its pivotal function in gut motility and involvement in gastrointestinal pathology. This report identifies novel roles for the transcription factor Ascl1 in enteric gliogenesis and neurogenesis. Moreover, independent of its proneurogenic activity, Ascl1 is required for the normal expression of specific enteric neuronal subtype characteristics. Distinct enteric neuronal subtypes are formed in a temporally defined order, and we observe that the early-born 5-HT(+) neurons are generated in Ascl1(-/-) mutants, despite the delayed neurogenesis. Enteric nervous system progenitor cells may therefore possess strong intrinsic control over their specification at the initial waves of neurogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Sistema Nervioso Entérico/crecimiento & desarrollo , Neuronas/fisiología , Animales , Calbindinas/metabolismo , Diferenciación Celular/genética , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación/genética , Células-Madre Neurales/fisiología , Neurogénesis/genética , Neurogénesis/fisiología , Neuroglía/fisiología , Embarazo , Neuronas Serotoninérgicas/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
5.
Brain Behav Immun ; 66: 23-30, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28347867

RESUMEN

Experimental reproducibility between laboratories is a major translational obstacle worldwide, particularly in studies investigating immunomodulatory therapies in relation to brain disease. In recent years increasing attention has been drawn towards the gut microbiota as a key factor in immune cell polarization. Moreover, manipulation of the gut microbiota has been found effective in a diverse range of brain disorders. Within this study we aimed to test the impact of microbiota differences between mice from different sources on the post-stroke neuroinflammatory response. With this rationale, we have investigated the correlation between microbiota differences and the immune response in mice from three commercial breeders with the same genetic background (C57BL/6). While overall bacterial load was comparable, we detected substantial differences in species diversity and microbiota composition on lower taxonomic levels. Specifically, we investigated segmented filamentous bacteria (SFB)-which have been shown to promote T cell polarization-and found that they were absent in mice from one breeder but abundant in others. Our experiments revealed a breeder specific correlation between SFB presence and the ratio of Treg to Th17 cells. Moreover, recolonization of SFB-negative mice with SFB resulted in a T cell shift which mimicked the ratios found in SFB-positive mice. We then investigated the response to a known experimental immunotherapeutic approach, CD28 superagonist (CD28SA), which has been previously shown to expand the Treg population. CD28SA treatment had differing effects between mice from different breeders and was found to be ineffective at inducing Treg expansion in SFB-free mice. These changes directly corresponded to stroke outcome as mice lacking SFB had significantly larger brain infarcts. This study demonstrates the major impact of microbiota differences on T cell polarization in mice during ischemic stroke conditions, and following immunomodulatory therapies.


Asunto(s)
Microbioma Gastrointestinal , Accidente Cerebrovascular/inmunología , Linfocitos T/fisiología , Animales , Antígenos CD28/administración & dosificación , Antígenos CD28/inmunología , Encefalitis/inmunología , Encefalitis/metabolismo , Femenino , Inmunoterapia , Activación de Linfocitos , Masculino , Ratones Endogámicos C57BL , Reproducibilidad de los Resultados , Accidente Cerebrovascular/terapia
6.
iScience ; 24(10): 103095, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34622150

RESUMEN

The gut microbiome has been implicated as a key regulator of brain function in health and disease. But the impact of gut microbiota on functional brain connectivity is unknown. We used resting-state functional magnetic resonance imaging in germ-free and normally colonized mice under naive conditions and after ischemic stroke. We observed a strong, brain-wide increase of functional connectivity in germ-free animals. Graph theoretical analysis revealed significant higher values in germ-free animals, indicating a stronger and denser global network but with less structural organization. Breakdown of network function after stroke equally affected germ-free and colonized mice. Results from histological analyses showed changes in dendritic spine densities, as well as an immature microglial phenotype, indicating impaired microglia-neuron interaction in germ-free mice as potential cause of this phenomenon. These results demonstrate the substantial impact of bacterial colonization on brain-wide function and extend our so far mainly (sub) cellular understanding of the gut-brain axis.

7.
Elife ; 102021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33845942

RESUMEN

Previous studies have identified a crucial role of the gut microbiome in modifying Alzheimer's disease (AD) progression. However, the mechanisms of microbiome-brain interaction in AD were so far unknown. Here, we identify microbiota-derived short chain fatty acids (SCFA) as microbial metabolites which promote Aß deposition. Germ-free (GF) AD mice exhibit a substantially reduced Aß plaque load and markedly reduced SCFA plasma concentrations; conversely, SCFA supplementation to GF AD mice increased the Aß plaque load to levels of conventionally colonized (specific pathogen-free [SPF]) animals and SCFA supplementation to SPF mice even further exacerbated plaque load. This was accompanied by the pronounced alterations in microglial transcriptomic profile, including upregulation of ApoE. Despite increased microglial recruitment to Aß plaques upon SCFA supplementation, microglia contained less intracellular Aß. Taken together, our results demonstrate that microbiota-derived SCFA are critical mediators along the gut-brain axis which promote Aß deposition likely via modulation of the microglial phenotype.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal , Microglía/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Femenino , Masculino , Ratones , Organismos Libres de Patógenos Específicos
8.
J Cereb Blood Flow Metab ; 38(8): 1293-1298, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29846130

RESUMEN

Microbiome alterations have been shown to affect stroke outcome. However, to what extent the presence of a gut microbiome per se is affecting post-stroke neuroinflammation has not been tested. By comparing germfree mice with recolonized (Ex-GF) and conventional SPF mice, we were able to demonstrate that bacterial colonization reduces stroke volumes. Bacterial colonization increased cerebral expression of cytokines as well as microglia/macrophage cell counts in contrast to improved stroke outcome. Interestingly, the microbiome-mediated brain protection was absent in lymphocyte-deficient mice. These findings support the concept of lymphocyte-driven protective neuroinflammation after stroke under control of the microbiome.


Asunto(s)
Microbioma Gastrointestinal , Inflamación/inmunología , Neuroprotección , Accidente Cerebrovascular/inmunología , Linfocitos T/inmunología , Animales , Inmunidad , Inflamación/complicaciones , Inflamación/patología , Ratones , Factores Protectores , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/patología
9.
J Cereb Blood Flow Metab ; 37(8): 3015-3026, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27909266

RESUMEN

Despite the efficacy of neuroprotective approaches in animal models of stroke, their translation has so far failed from bench to bedside. One reason is presumed to be a low quality of preclinical study design, leading to bias and a low a priori power. In this study, we propose that the key read-out of experimental stroke studies, the volume of the ischemic damage as commonly measured by free-handed planimetry of TTC-stained brain sections, is subject to an unrecognized low inter-rater and test-retest reliability with strong implications for statistical power and bias. As an alternative approach, we suggest a simple, open-source, software-assisted method, taking advantage of automatic-thresholding techniques. The validity and the improvement of reliability by an automated method to tMCAO infarct volumetry are demonstrated. In addition, we show the probable consequences of increased reliability for precision, p-values, effect inflation, and power calculation, exemplified by a systematic analysis of experimental stroke studies published in the year 2015. Our study reveals an underappreciated quality problem in translational stroke research and suggests that software-assisted infarct volumetry might help to improve reproducibility and therefore the robustness of bench to bedside translation.


Asunto(s)
Infarto Encefálico/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Ataque Isquémico Transitorio/diagnóstico por imagen , Programas Informáticos , Animales , Encéfalo/irrigación sanguínea , Infarto Encefálico/etiología , Modelos Animales de Enfermedad , Ataque Isquémico Transitorio/complicaciones , Masculino , Ratones Endogámicos C57BL , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Investigación Biomédica Traslacional
10.
Nephrol News Issues ; 17(5): 73-6, 78, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12715628

RESUMEN

Though future clinical trials will help establish better-defined guidelines for L-carnitine as a therapeutic agent, clearly there are subsets of dialysis patients who can benefit from L-carnitine therapy, as evidenced by the case studies described in this article. In each of these cases, L-carnitine significantly improved the patient's quality of life by increasing muscle strength and energy levels, increasing hematocrit, minimizing intradialytic morbidities that interfere with dialysis prescription, or improving cardiac function. Given the limited side effects, a trial of L-carnitine with ongoing evaluation of beneficial effect is a reasonable therapeutic option for chronic dialysis patients with clinical conditions related to DCD.


Asunto(s)
Carnitina/deficiencia , Carnitina/uso terapéutico , Diálisis Renal/efectos adversos , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA