Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 56(8): 1712-1726, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37557080

RESUMEN

The enteric nervous system is largely autonomous, and the central nervous system is compartmentalized behind the blood-brain barrier. Yet the intestinal microbiota shapes gut function, local and systemic immune responses, and central nervous system functions including cognition and mood. In this review, we address how the gut microbiota can profoundly influence neural and immune networks. Although many of the interactions between these three systems originate in the intestinal mucosa, intestinal function and immunity are modulated by neural pathways that connect the gut and brain. Furthermore, a subset of microbe-derived penetrant molecules enters the brain and regulates central nervous system function. Understanding how these seemingly isolated entities communicate has the potential to open up new avenues for therapies and interventions.


Asunto(s)
Sistema Nervioso Entérico , Microbioma Gastrointestinal , Microbiota , Sistema Nervioso Central , Encéfalo
2.
Nat Immunol ; 18(2): 116-122, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28092371

RESUMEN

Interactions between the nervous system and immune system are required for organ function and homeostasis. Evidence suggests that enteric neurons and intestinal immune cells share common regulatory mechanisms and can coordinate their responses to developmental challenges and environmental aggressions. These discoveries shed light on the physiology of system interactions and open novel perspectives for therapy designs that target underappreciated neurological-immunological commonalities. Here we highlight findings that address the importance of neuroimmune cell units (NICUs) in intestinal development, homeostasis and disease.


Asunto(s)
Sistema Inmunológico , Enfermedades Intestinales/inmunología , Mucosa Intestinal/inmunología , Intestinos/inmunología , Sistema Nervioso , Neurogénesis , Neuroinmunomodulación , Animales , Homeostasis , Humanos , Enfermedades Intestinales/terapia , Intestinos/embriología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Microbiota , Cresta Neural , Neuroglía/fisiología
3.
Nature ; 599(7883): 125-130, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34671159

RESUMEN

Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.


Asunto(s)
Homeostasis , Intestinos/inmunología , Intestinos/fisiología , Neuroglía/inmunología , Neuroglía/fisiología , Regeneración , Adventicia/inmunología , Adventicia/parasitología , Animales , Quimiocina CXCL10/inmunología , Duodeno/inmunología , Duodeno/parasitología , Duodeno/patología , Duodeno/fisiología , Femenino , Gliosis , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Interferón gamma/inmunología , Intestinos/parasitología , Intestinos/patología , Masculino , Ratones , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología
4.
Nature ; 578(7794): 284-289, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32025031

RESUMEN

Neural control of the function of visceral organs is essential for homeostasis and health. Intestinal peristalsis is critical for digestive physiology and host defence, and is often dysregulated in gastrointestinal disorders1. Luminal factors, such as diet and microbiota, regulate neurogenic programs of gut motility2-5, but the underlying molecular mechanisms remain unclear. Here we show that the transcription factor aryl hydrocarbon receptor (AHR) functions as a biosensor in intestinal neural circuits, linking their functional output to the microbial environment of the gut lumen. Using nuclear RNA sequencing of mouse enteric neurons that represent distinct intestinal segments and microbiota states, we demonstrate that the intrinsic neural networks of the colon exhibit unique transcriptional profiles that are controlled by the combined effects of host genetic programs and microbial colonization. Microbiota-induced expression of AHR in neurons of the distal gastrointestinal tract enables these neurons to respond to the luminal environment and to induce expression of neuron-specific effector mechanisms. Neuron-specific deletion of Ahr, or constitutive overexpression of its negative feedback regulator CYP1A1, results in reduced peristaltic activity of the colon, similar to that observed in microbiota-depleted mice. Finally, expression of Ahr in the enteric neurons of mice treated with antibiotics partially restores intestinal motility. Together, our experiments identify AHR signalling in enteric neurons as a regulatory node that integrates the luminal environment with the physiological output of intestinal neural circuits to maintain gut homeostasis and health.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Intestinos/fisiología , Neuronas/fisiología , Peristaltismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Femenino , Vida Libre de Gérmenes , Intestinos/inervación , Ligandos , Masculino , Ratones , Vías Nerviosas , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Transcriptoma/genética
5.
Nature ; 548(7669): 582-587, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28847002

RESUMEN

Multiple populations of wake-promoting neurons have been characterized in mammals, but few sleep-promoting neurons have been identified. Wake-promoting cell types include hypocretin and GABA (γ-aminobutyric-acid)-releasing neurons of the lateral hypothalamus, which promote the transition to wakefulness from non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. Here we show that a subset of GABAergic neurons in the mouse ventral zona incerta, which express the LIM homeodomain factor Lhx6 and are activated by sleep pressure, both directly inhibit wake-active hypocretin and GABAergic cells in the lateral hypothalamus and receive inputs from multiple sleep-wake-regulating neurons. Conditional deletion of Lhx6 from the developing diencephalon leads to decreases in both NREM and REM sleep. Furthermore, selective activation and inhibition of Lhx6-positive neurons in the ventral zona incerta bidirectionally regulate sleep time in adult mice, in part through hypocretin-dependent mechanisms. These studies identify a GABAergic subpopulation of neurons in the ventral zona incerta that promote sleep.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sueño/fisiología , Factores de Transcripción/metabolismo , Zona Incerta/citología , Ácido gamma-Aminobutírico/metabolismo , Animales , Linaje de la Célula , Neuronas GABAérgicas/efectos de los fármacos , Eliminación de Gen , Hipocampo/citología , Hipocampo/fisiología , Proteínas con Homeodominio LIM/deficiencia , Proteínas con Homeodominio LIM/efectos de los fármacos , Proteínas con Homeodominio LIM/genética , Masculino , Ratones , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Orexinas/metabolismo , Terminales Presinápticos/metabolismo , Sueño/efectos de los fármacos , Sueño/genética , Sueño REM/efectos de los fármacos , Sueño REM/genética , Sueño REM/fisiología , Factores de Tiempo , Factores de Transcripción/deficiencia , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Vigilia/efectos de los fármacos , Vigilia/genética , Vigilia/fisiología , Zona Incerta/efectos de los fármacos , Zona Incerta/fisiología
7.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25079316

RESUMEN

Mesenchymal stem cells occupy niches in stromal tissues where they provide sources of cells for specialized mesenchymal derivatives during growth and repair. The origins of mesenchymal stem cells have been the subject of considerable discussion, and current consensus holds that perivascular cells form mesenchymal stem cells in most tissues. The continuously growing mouse incisor tooth offers an excellent model to address the origin of mesenchymal stem cells. These stem cells dwell in a niche at the tooth apex where they produce a variety of differentiated derivatives. Cells constituting the tooth are mostly derived from two embryonic sources: neural crest ectomesenchyme and ectodermal epithelium. It has been thought for decades that the dental mesenchymal stem cells giving rise to pulp cells and odontoblasts derive from neural crest cells after their migration in the early head and formation of ectomesenchymal tissue. Here we show that a significant population of mesenchymal stem cells during development, self-renewal and repair of a tooth are derived from peripheral nerve-associated glia. Glial cells generate multipotent mesenchymal stem cells that produce pulp cells and odontoblasts. By combining a clonal colour-coding technique with tracing of peripheral glia, we provide new insights into the dynamics of tooth organogenesis and growth.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Incisivo/citología , Células Madre Mesenquimatosas/citología , Neuroglía/citología , Animales , Rastreo Celular , Células Clonales/citología , Pulpa Dental/citología , Femenino , Incisivo/embriología , Masculino , Ratones , Modelos Biológicos , Cresta Neural/citología , Odontoblastos/citología , Regeneración , Células de Schwann/citología
8.
Glia ; 67(6): 1167-1178, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30730592

RESUMEN

Coordination of gastrointestinal function relies on joint efforts of enteric neurons and glia, whose crosstalk is vital for the integration of their activity. To investigate the signaling mechanisms and to delineate the spatial aspects of enteric neuron-to-glia communication within enteric ganglia we developed a method to stimulate single enteric neurons while monitoring the activity of neighboring enteric glial cells. We combined cytosolic calcium uncaging of individual enteric neurons with calcium imaging of enteric glial cells expressing a genetically encoded calcium indicator and demonstrate that enteric neurons signal to enteric glial cells through pannexins using paracrine purinergic pathways. Sparse labeling of enteric neurons and high-resolution analysis of the structural relation between neuronal cell bodies, varicose release sites and enteric glia uncovered that this form of neuron-to-glia communication is contained between the cell body of an enteric neuron and its surrounding enteric glial cells. Our results reveal the spatial and functional foundation of neuro-glia units as an operational cellular assembly in the enteric nervous system.


Asunto(s)
Comunicación Celular/fisiología , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Animales , Células Cultivadas , Sistema Nervioso Entérico/química , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroglía/química , Neuronas/química
9.
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
10.
PLoS Genet ; 12(11): e1006439, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27902697

RESUMEN

Hirschsprung disease (HSCR) is characterized by absence of enteric neurons from the distal colon and severe intestinal dysmotility. To understand the pathophysiology and genetics of HSCR we developed a unique zebrafish model that allows combined genetic, developmental and in vivo physiological studies. We show that ret mutant zebrafish exhibit cellular, physiological and genetic features of HSCR, including absence of intestinal neurons, reduced peristalsis, and varying phenotype expressivity in the heterozygous state. We perform live imaging experiments using a UAS-GAL4 binary genetic system to drive fluorescent protein expression in ENS progenitors. We demonstrate that ENS progenitors migrate at reduced speed in ret heterozygous embryos, without changes in proliferation or survival, establishing this as a principal pathogenic mechanism for distal aganglionosis. We show, using live imaging of actual intestinal movements, that intestinal motility is severely compromised in ret mutants, and partially impaired in ret heterozygous larvae, and establish a clear correlation between neuron position and organised intestinal motility. We exploited the partially penetrant ret heterozygous phenotype as a sensitised background to test the influence of a candidate modifier gene. We generated mapk10 loss-of-function mutants, which show reduced numbers of enteric neurons. Significantly, we show that introduction of mapk10 mutations into ret heterozygotes enhanced the ENS deficit, supporting MAPK10 as a HSCR susceptibility locus. Our studies demonstrate that ret heterozygous zebrafish is a sensitized model, with many significant advantages over existing murine models, to explore the pathophysiology and complex genetics of HSCR.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Enfermedad de Hirschsprung/genética , Proteína Quinasa 10 Activada por Mitógenos/genética , Proteínas Proto-Oncogénicas c-ret/genética , Pez Cebra/genética , Animales , Colon/inervación , Colon/patología , Modelos Animales de Enfermedad , Sistema Nervioso Entérico/patología , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/patología , Humanos , Mutación , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Proteínas Proto-Oncogénicas c-ret/metabolismo
11.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25839327

RESUMEN

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Asunto(s)
Epistasis Genética/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética , Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Semaforinas/genética , Animales , Secuencia de Bases , Estudio de Asociación del Genoma Completo , Ratones , Datos de Secuencia Molecular , Semaforinas/deficiencia , Semaforinas/metabolismo , Análisis de Secuencia de ADN
12.
Immunity ; 31(5): 705-10, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19836266

RESUMEN

Strong evidence is emerging that the nervous and immune systems share mechanisms of gene regulation, signaling, cell communication, and supracellular organization. This brings to the fore many questions, not least of which is the developmental and evolutionary origin of the commonalities between the two systems. By providing answers to these questions, immunologists and neurobiologists increasingly expose the mechanistic and conceptual affinities of their respective fields and facilitate the understanding of fundamental principles that govern the organization of complex cellular systems. The current essay and reviews in Immunity and Neuron attempt to communicate to the wider scientific community a series of examples relating to commonalities between the immune and nervous system and enhance the dialog and exchange of ideas between the two fields.


Asunto(s)
Evolución Molecular , Sistema Inmunológico , Sistema Nervioso , Animales , Humanos
13.
Proc Natl Acad Sci U S A ; 117(33): 19624-19626, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32759220
14.
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
15.
Dev Biol ; 417(2): 158-67, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27112528

RESUMEN

The gastrointestinal (GI) tract is innervated by intrinsic enteric neurons and by extrinsic efferent and afferent nerves. The enteric (intrinsic) nervous system (ENS) in most regions of the gut consists of two main ganglionated layers; myenteric and submucosal ganglia, containing numerous types of enteric neurons and glial cells. Axons arising from the ENS and from extrinsic neurons innervate most layers of the gut wall and regulate many gut functions. The majority of ENS cells are derived from vagal neural crest cells (NCCs), which proliferate, colonize the entire gut, and first populate the myenteric region. After gut colonization by vagal NCCs, the extrinsic nerve fibers reach the GI tract, and Schwann cell precursors (SCPs) enter the gut along the extrinsic nerves. Furthermore, a subpopulation of cells in myenteric ganglia undergoes a radial (inward) migration to form the submucosal plexus, and the intrinsic and extrinsic innervation to the mucosal region develops. Here, we focus on recent progress in understanding the developmental processes that occur after the gut is colonized by vagal ENS precursors, and provide an up-to-date overview of molecular mechanisms regulating the development of the intrinsic and extrinsic innervation of the GI tract.


Asunto(s)
Sistema Nervioso Entérico , Tracto Gastrointestinal/inervación , Neurogénesis/fisiología , Neuronas Aferentes/citología , Neuronas Eferentes/citología , Animales , Movimiento Celular , Sistema Nervioso Entérico/anatomía & histología , Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/crecimiento & desarrollo , Tracto Gastrointestinal/embriología , Humanos , Ratones , Cresta Neural/embriología , Transducción de Señal
16.
Dev Biol ; 409(2): 392-405, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26658318

RESUMEN

Neural crest cells comprise a multipotent, migratory cell population that generates a diverse array of cell and tissue types, during vertebrate development. Enteric Nervous System controls the function of the gastrointestinal tract and is mainly derived from the vagal and sacral neural crest cells. Deregulation on self-renewal and differentiation of the enteric neural crest cells is evident in enteric nervous system disorders, such as Hirschsprung disease, characterized by the absence of ganglia in a variable length of the distal bowel. Here we show that Geminin is essential for Enteric Nervous System generation as mice that lacked Geminin expression specifically in neural crest cells revealed decreased generation of vagal neural crest cells, and enteric neural crest cells (ENCCs). Geminin-deficient ENCCs showed increased apoptosis and decreased cell proliferation during the early stages of gut colonization. Furthermore, decreased number of committed ENCCs in vivo and the decreased self-renewal capacity of enteric progenitor cells in vitro, resulted in almost total aganglionosis resembling a severe case of Hirschsprung disease. Our results suggest that Geminin is an important regulator of self-renewal and survival of enteric nervous system progenitor cells.


Asunto(s)
Sistema Nervioso Entérico/patología , Geminina/metabolismo , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/patología , Cresta Neural/metabolismo , Células Madre/metabolismo , Animales , Recuento de Células , Muerte Celular , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células , Geminina/deficiencia , Eliminación de Gen , Intestinos/patología , Ratones , Cresta Neural/citología , Neuroglía/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo
17.
Dev Biol ; 417(2): 229-51, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27059883

RESUMEN

Over the last 20 years, there has been increasing focus on the development of novel stem cell based therapies for the treatment of disorders and diseases affecting the enteric nervous system (ENS) of the gastrointestinal tract (so-called enteric neuropathies). Here, the idea is that ENS progenitor/stem cells could be transplanted into the gut wall to replace the damaged or absent neurons and glia of the ENS. This White Paper sets out experts' views on the commonly used methods and approaches to identify, isolate, purify, expand and optimize ENS stem cells, transplant them into the bowel, and assess transplant success, including restoration of gut function. We also highlight obstacles that must be overcome in order to progress from successful preclinical studies in animal models to ENS stem cell therapies in the clinic.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Sistema Nervioso Entérico/patología , Tracto Gastrointestinal/patología , Enfermedad de Hirschsprung/terapia , Seudoobstrucción Intestinal/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Animales , Modelos Animales de Enfermedad , Tracto Gastrointestinal/inervación , Guías como Asunto , Enfermedad de Hirschsprung/patología , Humanos , Seudoobstrucción Intestinal/patología
18.
Gastroenterology ; 151(5): 836-844, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27521479

RESUMEN

The gastrointestinal (GI) tract is essential for the absorption of nutrients, induction of mucosal and systemic immune responses, and maintenance of a healthy gut microbiota. Key aspects of gastrointestinal physiology are controlled by the enteric nervous system (ENS), which is composed of neurons and glial cells. The ENS is exposed to and interacts with the outer (microbiota, metabolites, and nutrients) and inner (immune cells and stromal cells) microenvironment of the gut. Although the cellular blueprint of the ENS is mostly in place by birth, the functional maturation of intestinal neural networks is completed within the microenvironment of the postnatal gut, under the influence of gut microbiota and the mucosal immune system. Recent studies have shown the importance of molecular interactions among microbiota, enteric neurons, and immune cells for GI homeostasis. In addition to its role in GI physiology, the ENS has been associated with the pathogenesis of neurodegenerative disorders, such as Parkinson's disease, raising the possibility that microbiota-ENS interactions could offer a viable strategy for influencing the course of brain diseases. Here, we discuss recent advances on the role of microbiota and the immune system on the development and homeostasis of the ENS, a key relay station along the gut-brain axis.


Asunto(s)
Sistema Nervioso Entérico/inmunología , Sistema Nervioso Entérico/microbiología , Microbioma Gastrointestinal , Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/fisiología , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Homeostasis/fisiología , Humanos , Mucosa Intestinal/embriología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiología , Enfermedad de Parkinson/etiología
19.
BMC Biol ; 14(1): 94, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27776507

RESUMEN

BACKGROUND: In vertebrate organisms, the neural crest (NC) gives rise to multipotential and highly migratory progenitors which are distributed throughout the embryo and generate, among other structures, the peripheral nervous system, including the intrinsic neuroglial networks of the gut, i.e. the enteric nervous system (ENS). The majority of enteric neurons and glia originate from vagal NC-derived progenitors which invade the foregut mesenchyme and migrate rostro-caudally to colonise the entire length of the gut. Although the migratory behaviour of NC cells has been studied extensively, it remains unclear how their properties and response to microenvironment change as they navigate through complex cellular terrains to reach their target embryonic sites. RESULTS: Using conditional gene inactivation in mice we demonstrate here that the cell cycle-dependent protein Geminin (Gem) is critical for the survival of ENS progenitors in a stage-dependent manner. Gem deletion in early ENS progenitors (prior to foregut invasion) resulted in cell-autonomous activation of DNA damage response and p53-dependent apoptosis, leading to severe intestinal aganglionosis. In contrast, ablation of Gem shortly after ENS progenitors had invaded the embryonic gut did not result in discernible survival or migratory deficits. In contrast to other developmental systems, we obtained no evidence for a role of Gem in commitment or differentiation of ENS lineages. The stage-dependent resistance of ENS progenitors to mutation-induced genotoxic stress was further supported by the enhanced survival of post gut invasion ENS lineages to γ-irradiation relative to their predecessors. CONCLUSIONS: Our experiments demonstrate that, in mammals, NC-derived ENS lineages are sensitive to genotoxic stress in a stage-specific manner. Following gut invasion, ENS progenitors are distinctly resistant to Gem ablation and irradiation in comparison to their pre-enteric counterparts. These studies suggest that the microenvironment of the embryonic gut protects ENS progenitors and their progeny from genotoxic stress.


Asunto(s)
Daño del ADN/efectos de los fármacos , Sistema Nervioso Entérico/citología , Geminina/farmacología , Cresta Neural/citología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Sistema Nervioso Entérico/efectos de los fármacos , Femenino , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Neurogénesis/efectos de los fármacos , Embarazo
20.
Cereb Cortex ; 25(9): 2370-82, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24626607

RESUMEN

Cortical interneurons are characterized by extraordinary functional and morphological diversity. Although tremendous progress has been made in uncovering molecular and cellular mechanisms implicated in interneuron generation and function, several questions still remain open. Rho-GTPases have been implicated as intracellular mediators of numerous developmental processes such as cytoskeleton organization, vesicle trafficking, transcription, cell cycle progression, and apoptosis. Specifically in cortical interneurons, we have recently shown a cell-autonomous and stage-specific requirement for Rac1 activity within proliferating interneuron precursors. Conditional ablation of Rac1 in the medial ganglionic eminence leads to a 50% reduction of GABAergic interneurons in the postnatal cortex. Here we examine the additional role of Rac3 by analyzing Rac1/Rac3 double-mutant mice. We show that in the absence of both Rac proteins, the embryonic migration of medial ganglionic eminence-derived interneurons is further impaired. Postnatally, double-mutant mice display a dramatic loss of cortical interneurons. In addition, Rac1/Rac3-deficient interneurons show gross cytoskeletal defects in vitro, with the length of their leading processes significantly reduced and a clear multipolar morphology. We propose that in the absence of Rac1/Rac3, cortical interneurons fail to migrate tangentially towards the pallium due to defects in actin and microtubule cytoskeletal dynamics.


Asunto(s)
Axones/fisiología , Corteza Cerebral/citología , Interneuronas/citología , Microtúbulos/fisiología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Animales Recién Nacidos , Axones/ultraestructura , Ciclo Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Interneuronas/metabolismo , Interneuronas/ultraestructura , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Eminencia Media/citología , Ratones , Ratones Transgénicos , Microtúbulos/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Paclitaxel/farmacología , Embarazo , Factor Nuclear Tiroideo 1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Moduladores de Tubulina/farmacología , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA