Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38733989

RESUMEN

The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.


Asunto(s)
Cerebelo , Neuronas , Retina , Animales , Femenino , Masculino , Ratones , Cerebelo/metabolismo , Cerebelo/irrigación sanguínea , Cerebelo/citología , Canales Iónicos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Vasos Retinianos/metabolismo
2.
Wiley Interdiscip Rev Dev Biol ; 10(1): e379, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32267095

RESUMEN

The central nervous system (CNS) is composed of precisely assembled circuits which support a variety of physiological functions and behaviors. These circuits include multiple subtypes of neurons with unique morphologies, electrical properties, and molecular identities. How these component parts are precisely wired-up has been a topic of great interest to the field of developmental neurobiology and has implications for our understanding of the etiology of many neurological disorders and mental illnesses. To date, many molecules involved in synaptic choice and specificity have been identified, including members of several families of cell-adhesion molecules (CAMs), which are cell-surface molecules that mediate cell-cell contacts and subsequent intracellular signaling. One favored hypothesis is that unique expression patterns of CAMs define specific neuronal subtype populations and determine compatible pre- and postsynaptic neuronal partners based on the expression of these unique CAMs. The mouse retina has served as a beautiful model for investigations into mammalian CAM interactions due to its well-defined neuronal subtypes and distinct circuits. Moreover, the retina is readily amenable to visualization of circuit organization and electrophysiological measurement of circuit function. The advent of recent genetic, genomic, and imaging technologies has opened the field up to large-scale, unbiased approaches for identification of new molecular determinants of synaptic specificity. Thus, building on the foundation of work reviewed here, we can expect rapid expansion of the field, harnessing the mouse retina as a model to understand the molecular basis for synaptic specificity and functional circuit assembly. This article is categorized under: Nervous System Development > Vertebrates: General Principles Nervous System Development > Vertebrates: Regional Development.


Asunto(s)
Retina/fisiología , Sinapsis/fisiología , Animales , Humanos
3.
G3 (Bethesda) ; 8(8): 2825-2832, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-29950427

RESUMEN

The role of Notch signaling in cell-fate decisions has been studied extensively; however, this pathway is also active in adult tissues, including the nervous system. Notch signaling modulates a wide range of behaviors and processes of the nervous system in the nematode Caenorhabditis elegans, but there is no evidence for Notch signaling directly altering synaptic strength. Here, we demonstrate Notch-mediated regulation of synaptic activity at the C. elegans neuromuscular junction (NMJ). For this, we used aldicarb, an inhibitor of the enzyme acetylcholinesterase, and assessed paralysis rates of animals with altered Notch signaling. Notch receptors LIN-12 and GLP-1 are required for normal NMJ function; they regulate NMJ activity in an opposing fashion. Complete loss of LIN-12 skews the excitation/inhibition balance at the NMJ toward increased activity, whereas partial loss of GLP-1 has the opposite effect. Specific Notch ligands and co-ligands are also required for proper NMJ function. The role of LIN-12 is independent of cell-fate decisions; manipulation of LIN-12 signaling through RNAi knockdown or overexpression of the co-ligand OSM-11 after development alters NMJ activity. We demonstrate that LIN-12 modulates GABA signaling in this paradigm, as loss of GABA signaling suppresses LIN-12 gain-of-function defects. Further analysis, in vivo and in silico, suggests that LIN-12 may modulate transcription of the GABAB receptor GBB-2 Our findings confirm a non-developmental role for the LIN-12/Notch receptor in regulating synaptic signaling and identify the GABAB receptor GBB-2 as a potential Notch transcriptional target in the C. elegans nervous system.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Unión Neuromuscular/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo , Aldicarb/farmacología , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Inhibidores de la Colinesterasa/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mutación , Unión Neuromuscular/efectos de los fármacos , Receptores Notch/genética , Transducción de Señal/efectos de los fármacos
4.
Sci Rep ; 7(1): 2525, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28566702

RESUMEN

Mechanisms mediating adult enteric neurogenesis are largely unknown. Using inflammation-associated neurogenesis models and a transgenic approach, we aimed to understand the cell-source for new neurons in infectious and inflammatory colitis. Dextran sodium sulfate (DSS) and Citrobacter rodentium colitis (CC) was induced in adult mice and colonic neurons were quantified. Sox2GFP and PLP1GFP mice confirmed the cell-type specificity of these markers. Sox2CreER:YFP and PLP1creER:tdT mice were used to determine the fate of these cells after colitis. Sox2 expression was investigated in colonic neurons of human patients with Clostridium difficile or ulcerative colitis. Both DSS and CC led to increased colonic neurons. Following colitis in adult Sox2CreER:YFP mice, YFP initially expressed predominantly by glia becomes expressed by neurons following colitis, without observable DNA replication. Similarly in PLP1CreER:tdT mice, PLP1 cells that co-express S100b but not RET also give rise to neurons following colitis. In human colitis, Sox2-expressing neurons increase from 1-2% to an average 14% in colitis. The new neurons predominantly express calretinin, thus appear to be excitatory. These results suggest that colitis promotes rapid enteric neurogenesis in adult mice and humans through differentiation of Sox2- and PLP1-expressing cells, which represent enteric glia and/or neural progenitors. Further defining neurogenesis will improve understanding and treatment of injury-associated intestinal motility/sensory disorders.


Asunto(s)
Colitis/genética , Inflamación/genética , Proteína Proteolipídica de la Mielina/genética , Factores de Transcripción SOXB1/genética , Animales , Citrobacter rodentium/patogenicidad , Clostridioides difficile/patogenicidad , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Colon/inervación , Colon/metabolismo , Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inflamación/inducido químicamente , Inflamación/microbiología , Inflamación/patología , Ratones , Ratones Transgénicos , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/metabolismo , Neuronas/patología , Subunidad beta de la Proteína de Unión al Calcio S100/genética
5.
J Anat ; 230(4): 567-574, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28116763

RESUMEN

The development of the enteric nervous system (ENS) and intestinal smooth muscle occurs in a spatially and temporally correlated manner, but how they influence each other is unknown. In the developing mid-gut of the chick embryo, we find that α-smooth muscle actin expression, indicating early muscle differentiation, occurs after the arrival of migrating enteric neural crest-derived cells (ENCCs). In contrast, hindgut smooth muscle develops prior to ENCC arrival. Smooth muscle development is normal in experimentally aganglionic hindguts, suggesting that proper development and patterning of the muscle layers does not rely on the ENS. However, inhibiting early smooth muscle development severely disrupts ENS patterning without affecting ENCC proliferation or apoptosis. Our results demonstrate that early intestinal smooth muscle differentiation is required for patterning the developing ENS.


Asunto(s)
Sistema Nervioso Entérico/embriología , Intestinos/embriología , Músculo Liso/embriología , Animales , Embrión de Pollo , Pollos , Sistema Nervioso Entérico/anatomía & histología , Intestinos/anatomía & histología , Músculo Liso/anatomía & histología , Técnicas de Cultivo de Órganos
6.
Pediatr Res ; 81(5): 838-846, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28060794

RESUMEN

BACKGROUND: Enteric neural stem/progenitor cells (ENSCs) offer an innovative approach to treating Hirschsprung disease (HSCR) and other enteric neuropathies. However, postnatal-derived human ENSCs have not been thoroughly characterized and their behavior in the embryonic and postnatal intestinal environment is unknown. METHODS: ENSCs were isolated from the intestines of 25 patients undergoing bowel resection, including 7 children with HSCR. Neuronal differentiation and proliferation of ENSCs from submucosal and myenteric plexuses from patients with and without HSCR were characterized. ENSC migration and differentiation were studied following transplantation into embryonic chick neural crest, embryonic chick hindgut, and postnatal mouse aganglionic colon. RESULTS: The proliferative and neurogenic potential of ENSCs from HSCR intestine is equivalent to that of non-HSCR controls. Similarly, no difference was observed between myenteric- and submucosal-derived ENSCs. Postnatal ENSCs transplanted to embryonic neural crest pathways and to aneural hindgut migrate normally and differentiate into appropriate neural crest-derived cell types. ENSCs in postnatal mouse aganglionic colon differentiate into neurons and glia both ex vivo and in vivo. CONCLUSIONS: ENSCs isolated from the postnatal intestine of patients with and without HSCR can behave like embryonic neural crest-derived cells. These results support the feasibility of cell-based therapy for future treatment of neurointestinal disease.


Asunto(s)
Movimiento Celular , Proliferación Celular , Enfermedad de Hirschsprung/patología , Intestino Grueso/inervación , Plexo Mientérico/patología , Células-Madre Neurales/patología , Neurogénesis , Nicho de Células Madre , Plexo Submucoso/patología , Adolescente , Animales , Células Cultivadas , Embrión de Pollo , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Enfermedad de Hirschsprung/cirugía , Humanos , Lactante , Recién Nacido , Masculino , Ratones Endogámicos C57BL , Células-Madre Neurales/trasplante , Esferoides Celulares , Trasplante de Células Madre , Adulto Joven
7.
J Surg Res ; 206(2): 451-459, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27884342

RESUMEN

BACKGROUND: Enteric neurospheres derived from postnatal intestine represent a promising avenue for cell replacement therapy to treat Hirschsprung disease and other neurointestinal diseases. We describe a simple method to improve the neuronal yield of spontaneously formed gut-derived neurospheres. MATERIALS AND METHODS: Enteric neurospheres were formed from the small and large intestines of mouse and human subjects. Neurosphere size, neural crest cell content, cell migration, neuronal differentiation, and neuronal proliferation in culture were analyzed. The effect of supplemental neurotrophic factors, including glial cell line-derived neurotrophic factor (GDNF) and endothelin-3, was also assessed. RESULTS: Mouse small intestine-derived neurospheres contained significantly more P75-expressing neural crest-derived cells (49.9 ± 15.3% versus 21.6 ± 11.9%, P < 0.05) and gave rise to significantly more Tuj1-expressing neurons than colon-derived neurospheres (69.9 ± 8.6% versus 46.2 ± 15.6%, P < 0.05). A similar pattern was seen in neurospheres isolated from human small and large intestine (32.6 ± 17.5% versus 10.2 ± 8.2% neural crest cells, P < 0.05; 29.7 ± 16.4% versus 16.0 ± 13.5% enteric neurons, P < 0.05). The addition of GDNF to the culture media further improved the neurogenic potential of small intestinal neurospheres (75.9 ± 4.0% versus 67.8 ± 5.8%, P < 0.05) whereas endothelin-3 had no effect. CONCLUSIONS: Enteric neurospheres formed from small intestine and supplemented with GDNF yield an enriched population of neural crest-derived progenitor cells and give rise to a high density of enteric neurons.


Asunto(s)
Sistema Nervioso Entérico/citología , Células-Madre Neurales/trasplante , Neurogénesis/fisiología , Adolescente , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Niño , Sistema Nervioso Entérico/fisiología , Femenino , Enfermedades Gastrointestinales/terapia , Enfermedad de Hirschsprung/terapia , Humanos , Lactante , Intestino Grueso/citología , Intestino Grueso/fisiología , Intestino Delgado/citología , Intestino Delgado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/fisiología , Adulto Joven
8.
J Pediatr Surg ; 51(11): 1834-1838, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27570241

RESUMEN

PURPOSE: Recent evidence suggests that patients with Hirschsprung disease (HD) have abnormal neurotransmitter expression in the ganglionated proximal colon. These alterations may cause persistent bowel dysfunction even after pullthrough surgery. We sought to quantify the proportion of nitrergic neurons in the ganglionic colon of HD patients and relate these findings to functional outcome. METHODS: The proximal resection margin from 17 patients with colonic HD who underwent a pullthrough procedure and colorectal tissue from 4 age-matched controls were immunohistochemically examined to quantify the proportion of nitrergic neurons. The incidence of constipation, incontinence, and enterocolitis in HD patients was assessed retrospectively and correlated with the proportion of nitric oxide synthase (NOS) expressing neurons. Neuronal subtypes in the ganglionic colon of the Edrnb-/- mouse model of HD were also studied. RESULTS: Mice with HD had a significantly higher proportion of NOS+ neurons in ganglionic colon than normal littermates (32.0±5.6% vs. 19.8±1.2%, p<0.01). Patients with HD also had significantly more NOS+ neurons than controls (18.4±4.6% vs. 13.1±1.9%, p<0.01). Patients who experienced constipation or enterocolitis postoperatively tended toward a higher proportion of NOS+ neurons (21.4±3.9% vs. 17.1±4.1%, p=0.06). Furthermore, patients with a proportion of NOS+ neurons above the median of all HD patients (18.3%) were significantly more likely to have constipation than those below the median (75% vs. 14%, p<0.05). CONCLUSION: An overabundance of nitrergic neurons in the proximal resection margin is associated with HD and may predict bowel dysfunction following pullthrough surgery.


Asunto(s)
Procedimientos Quirúrgicos del Sistema Digestivo/métodos , Enfermedad de Hirschsprung/cirugía , Plexo Mientérico/metabolismo , Neuronas Nitrérgicas/patología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Femenino , Enfermedad de Hirschsprung/diagnóstico , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Masculino , Ratones , Ratones Endogámicos C57BL , Plexo Mientérico/patología , Neuronas Nitrérgicas/metabolismo
9.
Biomaterials ; 88: 1-11, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26922325

RESUMEN

Cell therapy offers an innovative approach for treating enteric neuropathies. Postnatal gut-derived enteric neural stem/progenitor cells (ENSCs) represent a potential autologous source, but have a limited capacity for proliferation and neuronal differentiation. Since serotonin (5-HT) promotes enteric neuronal growth during embryonic development, we hypothesized that serotonin receptor agonism would augment growth of neurons from transplanted ENSCs. Postnatal ENSCs were isolated from 2 to 4 week-old mouse colon and cultured with 5-HT4 receptor agonist (RS67506)-loaded liposomal nanoparticles. ENSCs were co-cultured with mouse colon explants in the presence of RS67506-loaded (n = 3) or empty nanoparticles (n = 3). ENSCs were also transplanted into mouse rectum in vivo with RS67506-loaded (n = 8) or blank nanoparticles (n = 4) confined in a thermosensitive hydrogel, Pluronic F-127. Neuronal density and proliferation were analyzed immunohistochemically. Cultured ENSCs gave rise to significantly more neurons in the presence of RS67506-loaded nanoparticles. Similarly, colon explants had significantly increased neuronal density when RS67506-loaded nanoparticles were present. Finally, following in vivo cell delivery, co-transplantation of ENSCs with 5-HT4 receptor agonist-loaded nanoparticles led to significantly increased neuronal density and proliferation. We conclude that optimization of postnatal ENSCs can support their use in cell-based therapies for neurointestinal diseases.


Asunto(s)
Preparaciones de Acción Retardada/química , Sistema Nervioso Entérico/citología , Células-Madre Neurales/trasplante , Neurogénesis/efectos de los fármacos , Poloxámero/química , Agonistas del Receptor de Serotonina 5-HT4/administración & dosificación , Agonistas del Receptor de Serotonina 5-HT4/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Sistema Nervioso Entérico/efectos de los fármacos , Sistema Nervioso Entérico/fisiología , Tracto Gastrointestinal/inervación , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Liposomas/química , Ratones Endogámicos C57BL , Nanopartículas/química , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/terapia , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Temperatura
10.
Development ; 143(2): 264-75, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26674309

RESUMEN

The enteric nervous system (ENS) develops from neural crest cells that migrate along the intestine, differentiate into neurons and glia, and pattern into two plexuses within the gut wall. Inductive interactions between epithelium and mesenchyme regulate gut development, but the influence of these interactions on ENS development is unknown. Epithelial-mesenchymal recombinations were constructed using avian hindgut mesenchyme and non-intestinal epithelium from the bursa of Fabricius. These recombinations led to abnormally large and ectopically positioned ganglia. We hypothesized that sonic hedgehog (Shh), a secreted intestinal epithelial protein not expressed in the bursa, mediates this effect. Inhibition of Shh signaling, by addition of cyclopamine or a function-blocking antibody, resulted in large, ectopic ganglia adjacent to the epithelium. Shh overexpression, achieved in ovo using Shh-encoding retrovirus and in organ culture using recombinant protein, led to intestinal aganglionosis. Shh strongly induced the expression of versican and collagen type IX, whereas cyclopamine reduced expression of these chondroitin sulfate proteoglycans that are known to be inhibitory to neural crest cell migration. Shh also inhibited enteric neural crest-derived cell (ENCC) proliferation, promoted neuronal differentiation, and reduced expression of Gdnf, a key regulator of ENS formation. Ptc1 and Ptc2 were not expressed by ENCCs, and migration of isolated ENCCs was not inhibited by Shh protein. These results suggest that epithelial-derived Shh acts indirectly on the developing ENS by regulating the composition of the intestinal microenvironment.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Matriz Extracelular/metabolismo , Proteínas Hedgehog/metabolismo , Animales , Apoptosis/fisiología , Movimiento Celular , Pollos , Proteínas Hedgehog/genética , Inmunohistoquímica , Hibridación in Situ , Ratones , Receptores Patched , Receptor Patched-1 , Codorniz , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...