Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell ; 160(1-2): 269-84, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25594183

RESUMO

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).


Assuntos
Osso e Ossos/citologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Intestino Delgado/citologia , Células-Tronco Mesenquimais/citologia , Animais , Cartilagem/metabolismo , Intestino Delgado/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
2.
Neurobiol Dis ; 134: 104696, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31783118

RESUMO

Cerebral dopamine neurotrophic factor (CDNF) is neuroprotective for nigrostriatal dopamine neurons and restores dopaminergic function in animal models of Parkinson's disease (PD). To understand the role of CDNF in mammals, we generated CDNF knockout mice (Cdnf-/-), which are viable, fertile, and have a normal life-span. Surprisingly, an age-dependent loss of enteric neurons occurs selectively in the submucosal but not in the myenteric plexus. This neuronal loss is a consequence not of increased apoptosis but of neurodegeneration and autophagy. Quantitatively, the neurodegeneration and autophagy found in the submucosal plexus in duodenum, ileum and colon of the Cdnf-/- mouse are much greater than in those of Cdnf+/+ mice. The selective vulnerability of submucosal neurons to the absence of CDNF is reminiscent of the tendency of pathological abnormalities to occur in the submucosal plexus in biopsies of patients with PD. In contrast, the number of substantia nigra dopamine neurons and dopamine and its metabolite concentrations in the striatum are unaltered in Cdnf-/- mice; however, there is an age-dependent deficit in the function of the dopamine system in Cdnf-/- male mice analyzed. This is observed as D-amphetamine-induced hyperactivity, aberrant dopamine transporter function, and as increased D-amphetamine-induced dopamine release demonstrating that dopaminergic axon terminal function in the striatum of the Cdnf-/- mouse brain is altered. The deficiencies of Cdnf-/- mice, therefore, are reminiscent of those seen in early stages of Parkinson's disease.


Assuntos
Encéfalo/patologia , Encéfalo/fisiologia , Dopamina/metabolismo , Sistema Nervoso Entérico/patologia , Sistema Nervoso Entérico/fisiopatologia , Fatores de Crescimento Neural/fisiologia , Neurônios/patologia , Neurônios/fisiologia , Animais , Apoptose , Autofagia , Feminino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural/genética
3.
Gastroenterology ; 153(4): 1068-1081.e7, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28711628

RESUMO

BACKGROUND & AIMS: When the glial fibrillary acidic protein (GFAP) promoter is used to express cellular toxins that eliminate glia in mice, intestinal epithelial permeability and proliferation increase; this led to the concept that glia are required for maintenance of the gastrointestinal epithelium. Many enteric glia, however, particularly in the mucosa, do not express GFAP. In contrast, virtually all enteric glia express proteolipid protein 1 (PLP1). We investigated whether elimination of PLP1-expressing cells compromises epithelial maintenance or gastrointestinal motility. METHODS: We generated mice that express tamoxifen-inducible Cre recombinase under control of the Plp1 promoter and carry the diptheria toxin subunit A (DTA) transgene in the Rosa26 locus (Plp1CreER;Rosa26DTA mice). In these mice, PLP1-expressing glia are selectively eliminated without affecting neighboring cells. We measured epithelial barrier function and gastrointestinal motility in these mice and littermate controls, and analyzed epithelial cell proliferation and ultrastructure from their intestinal tissues. To compare our findings with those from previous studies, we also eliminated glia with ganciclovir in GfapHSV-TK mice. RESULTS: Expression of DTA in PLP1-expressing cells selectively eliminated enteric glia from the small and large intestines, but caused no defects in epithelial proliferation, barrier integrity, or ultrastructure. In contrast, administration of ganciclovir to GfapHSV-TK mice eliminated fewer glia but caused considerable non-glial toxicity and epithelial cell death. Elimination of PLP1-expressing cells did not reduce survival of neurons in the intestine, but altered gastrointestinal motility in female, but not male, mice. CONCLUSIONS: Using the Plp1 promoter to selectively eliminate glia in mice, we found that enteric glia are not required for maintenance of the intestinal epithelium, but are required for regulation of intestinal motility in females. Previous observations supporting the concept that maintenance of the intestinal epithelium requires enteric glia can be attributed to non-glial toxicity in GfapHSV-TK mice and epithelial-cell expression of GFAP. Contrary to widespread notions, enteric glia are therefore not required for epithelial homeostasis. However, they regulate intestinal motility in a sex-dependent manner.


Assuntos
Sistema Nervoso Entérico/fisiologia , Motilidade Gastrointestinal , Mucosa Intestinal/fisiologia , Intestinos/inervação , Neuroglia/fisiologia , Animais , Proliferação de Células , Toxina Diftérica/genética , Toxina Diftérica/metabolismo , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/ultraestrutura , Feminino , Ganciclovir/toxicidade , Genótipo , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Homeostase , Integrases/genética , Integrases/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestrutura , Intestinos/efeitos dos fármacos , Intestinos/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Neuroglia/metabolismo , Neuroglia/ultraestrutura , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fenótipo , Regiões Promotoras Genéticas , RNA não Traduzido/genética , Fatores Sexuais , Fatores de Tempo
4.
J Neurosci ; 31(39): 13746-57, 2011 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-21957238

RESUMO

Trophic factor signaling is important for the migration, differentiation, and survival of enteric neurons during development. The mechanisms that regulate the maturation of enteric neurons in postnatal life, however, are poorly understood. Here, we show that transcriptional cofactor HIPK2 (homeodomain interacting protein kinase 2) is required for the maturation of enteric neurons and for regulating gliogenesis during postnatal development. Mice lacking HIPK2 display a spectrum of gastrointestinal (GI) phenotypes, including distention of colon and slowed GI transit time. Although loss of HIPK2 does not affect the enteric neurons in prenatal development, a progressive loss of enteric neurons occurs during postnatal life in Hipk2(-/-) mutant mice that preferentially affects the dopaminergic population of neurons in the caudal region of the intestine. The mechanism by which HIPK2 regulates postnatal enteric neuron development appears to involve the response of enteric neurons to bone morphogenetic proteins (BMPs). Specifically, compared to wild type mice, a larger proportion of enteric neurons in Hipk2(-/-) mutants have an abnormally high level of phosphorylated Smad1/5/8. Consistent with the ability of BMP signaling to promote gliogenesis, Hipk2(-/-) mutants show a significant increase in glia in the enteric nervous system. In addition, numbers of autophagosomes are increased in enteric neurons in Hipk2(-/-) mutants, and synaptic maturation is arrested. These results reveal a new role for HIPK2 as an important transcriptional cofactor that regulates the BMP signaling pathway in the maintenance of enteric neurons and glia, and further suggest that HIPK2 and its associated signaling mechanisms may be therapeutically altered to promote postnatal neuronal maturation.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Transporte/fisiologia , Dopamina/fisiologia , Sistema Nervoso Entérico/enzimologia , Neuroglia/fisiologia , Neurônios/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/fisiologia , Animais , Animais Recém-Nascidos , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/crescimento & desenvolvimento , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Neuroglia/enzimologia , Neurônios/citologia , Neurônios/enzimologia
5.
J Neurochem ; 107(6): 1709-21, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19014382

RESUMO

Acute exposure to amphetamines (AMPHs) collapses secretory vesicle pH gradients, which increases cytosolic catecholamine levels while decreasing the quantal size of catecholamine release during fusion events. AMPH and methamphetamine (METH), however, are retained in tissues over long durations. We used optical and electron microscopic probes to measure the effects of long-term METH exposure on secretory vesicle pH, and amperometry and intracellular patch electrochemistry to observe the effects on neurosecretion and cytosolic catecholamines in cultured rat chromaffin cells. In contrast to acute METH effects, exposure to the drug for 6-48 h at 10 microM and higher concentrations produced a concentration-dependent rebound hyperacidification of secretory vesicles. At 5-10 microM levels, prolonged METH increased the quantal size and reinstated exocytotic catecholamine release, although very high (> 100 microM) levels of the drug, while continuing to produce rebound hyperacidification, did not increase quantal size. Secretory vesicle rebound hyperacidification was temperature dependent with optimal response at approximately 37 degrees C, was not blocked by the transcription inhibitor, puromycin, and appears to be a general compensatory response to prolonged exposure with membranophilic weak bases, including AMPHs, methylphenidate, cocaine, and ammonia. Thus, under some conditions of prolonged exposure, AMPHs and other weak bases can enhance, rather than deplete, the vesicular release of catecholamines via a compensatory response resulting in vesicle acidification.


Assuntos
Catecolaminas/metabolismo , Células Cromafins/ultraestrutura , Inibidores da Captação de Dopamina/farmacologia , Metanfetamina/farmacologia , Vesículas Secretórias/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Cadaverina/análogos & derivados , Células Cultivadas , Citosol/efeitos dos fármacos , Citosol/metabolismo , Relação Dose-Resposta a Droga , Eletroquímica/métodos , Exocitose/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Microscopia Imunoeletrônica/métodos , Inibidores da Síntese de Proteínas/farmacologia , Puromicina/farmacologia , Ratos , Fatores de Tempo
6.
Cancer Cell ; 28(6): 800-814, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26585400

RESUMO

The regulation and stem cell origin of normal and neoplastic gastric glands are uncertain. Here, we show that Mist1 expression marks quiescent stem cells in the gastric corpus isthmus. Mist1(+) stem cells serve as a cell-of-origin for intestinal-type cancer with the combination of Kras and Apc mutation and for diffuse-type cancer with the loss of E-cadherin. Diffuse-type cancer development is dependent on inflammation mediated by Cxcl12(+) endothelial cells and Cxcr4(+) gastric innate lymphoid cells (ILCs). These cells form the perivascular gastric stem cell niche, and Wnt5a produced from ILCs activates RhoA to inhibit anoikis in the E-cadherin-depleted cells. Targeting Cxcr4, ILCs, or Wnt5a inhibits diffuse-type gastric carcinogenesis, providing targets within the neoplastic gastric stem cell niche.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Células-Tronco Neoplásicas/metabolismo , Nicho de Células-Tronco , Neoplasias Gástricas/metabolismo , Microambiente Tumoral , Animais , Anoikis , Antineoplásicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Transplante de Medula Óssea , Caderinas/metabolismo , Comunicação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Senescência Celular , Quimiocina CXCL12/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Humanos , Linfócitos/metabolismo , Linfócitos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Receptores CXCR4/metabolismo , Transdução de Sinais , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fatores de Tempo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
7.
J Clin Invest ; 124(3): 1283-95, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24487592

RESUMO

Doublecortin-like kinase 1 protein (DCLK1) is a gastrointestinal tuft cell marker that has been proposed to identify quiescent and tumor growth-sustaining stem cells. DCLK1⁺ tuft cells are increased in inflammation-induced carcinogenesis; however, the role of these cells within the gastrointestinal epithelium and their potential as cancer-initiating cells are poorly understood. Here, using a BAC-CreERT-dependent genetic lineage-tracing strategy, we determined that a subpopulation of DCLK1⁺ cells is extremely long lived and possesses rare stem cell abilities. Moreover, genetic ablation of Dclk1 revealed that DCLK1⁺ tuft cells contribute to recovery following intestinal and colonic injury. Surprisingly, conditional knockdown of the Wnt regulator APC in DCLK1⁺ cells was not sufficient to drive colonic carcinogenesis under normal conditions; however, dextran sodium sulfate-induced (DSS-induced) colitis promoted the development of poorly differentiated colonic adenocarcinoma in mice lacking APC in DCLK1⁺ cells. Importantly, colonic tumor formation occurred even when colitis onset was delayed for up to 3 months after induced APC loss in DCLK1⁺ cells. Thus, our data define an intestinal DCLK1⁺ tuft cell population that is long lived, quiescent, and important for intestinal homeostasis and regeneration. Long-lived DCLK1⁺ cells maintain quiescence even following oncogenic mutation, but are activated by tissue injury and can serve to initiate colon cancer.


Assuntos
Adenocarcinoma/patologia , Colo/patologia , Neoplasias do Colo/patologia , Células-Tronco Neoplásicas/fisiologia , Adenocarcinoma/metabolismo , Animais , Linhagem da Célula , Células Cultivadas , Colo/imunologia , Colo/inervação , Neoplasias do Colo/metabolismo , Toxina Diftérica/farmacologia , Quinases Semelhantes a Duplacortina , Homeostase , Mucosa Intestinal/imunologia , Mucosa Intestinal/inervação , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
8.
Neuron ; 74(2): 277-84, 2012 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-22542182

RESUMO

mTOR is a regulator of cell growth and survival, protein synthesis-dependent synaptic plasticity, and autophagic degradation of cellular components. When triggered by mTOR inactivation, macroautophagy degrades long-lived proteins and organelles via sequestration into autophagic vacuoles. mTOR further regulates synaptic plasticity, and neurodegeneration occurs when macroautophagy is deficient. It is nevertheless unknown whether macroautophagy modulates presynaptic function. We find that the mTOR inhibitor rapamycin induces formation of autophagic vacuoles in prejunctional dopaminergic axons with associated decreased axonal profile volumes, synaptic vesicle numbers, and evoked dopamine release. Evoked dopamine secretion was enhanced and recovery was accelerated in transgenic mice in which macroautophagy deficiency was restricted to dopaminergic neurons; rapamycin failed to decrease evoked dopamine release in the striatum of these mice. Macroautophagy that follows mTOR inhibition in presynaptic terminals, therefore, rapidly alters presynaptic structure and neurotransmission.


Assuntos
Autofagia/genética , Encéfalo/citologia , Regulação da Expressão Gênica/genética , Proteínas Associadas aos Microtúbulos/genética , Terminações Pré-Sinápticas/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralina/análogos & derivados , 8-Hidroxi-2-(di-n-propilamino)tetralina/farmacologia , Análise de Variância , Animais , Autofagia/efeitos dos fármacos , Proteína 7 Relacionada à Autofagia , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Eletroquímica , Regulação da Expressão Gênica/efeitos dos fármacos , Genótipo , Imunossupressores/farmacologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Proteínas Associadas aos Microtúbulos/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , RNA Mensageiro/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 289(6): G1148-63, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16037544

RESUMO

The aim of the current study was to identify enteric 5-HT(4) splice variants, locate enteric 5-HT(4) receptors, determine the relationship, if any, of the 5-HT(4) receptor to 5-HT(1P) activity, and to ascertain the function of 5-HT(4) receptors in enteric neurophysiology. 5-HT(4a), 5-HT(4b), 5-HT(4e), and 5-HT(4f) isoforms were found in mouse brain and gut. The ratio of 5-HT(4) expression to that of the neural marker, synaptophysin, was higher in gut than in brain but was similar in small and large intestines. Submucosal 5-HT(4) expression was higher than myenteric. Although transcripts encoding 5-HT(4a) and 5-HT(4b) isoforms were more abundant, those encoding 5-HT(4e) and 5-HT(4f) were myenteric plexus specific. In situ hybridization revealed the presence of transcripts encoding 5-HT(4) receptors in subsets of enteric neurons, interstitial cells of Cajal, and smooth muscle cells. IgY antibodies to mouse 5-HT(4) receptors were raised, affinity purified, and characterized. Nerve fibers in the circular muscle and the neuropil in ganglia of both plexuses were highly 5-HT(4) immunoreactive, although only a small subset of neurons contained 5-HT(4) immunoreactivity. No 5-HT(4)-immunoreactive nerves were detected in the mucosa. 5-HT and 5-HT(1P) agonists evoked a G protein-mediated long-lasting inward current that was neither mimicked by 5-HT(4) agonists nor blocked by 5-HT(4) antagonists. In contrast, the 5-HT(4) agonists renzapride and tegaserod increased the amplitudes of nicotinic evoked excitatory postsynaptic currents. Enteric neuronal 5-HT(4) receptors thus are presynaptic and probably exert their prokinetic effects by strengthening excitatory neurotransmission.


Assuntos
Sistema Nervoso Entérico/fisiologia , Isoformas de Proteínas/biossíntese , Receptores 5-HT4 de Serotonina/fisiologia , Processamento Alternativo , Animais , Sistema Nervoso Entérico/ultraestrutura , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Masculino , Camundongos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Receptores 5-HT4 de Serotonina/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinaptofisina/biossíntese
10.
J Physiol ; 542(Pt 2): 453-76, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12122145

RESUMO

Trapping of weak bases was utilized to evaluate stimulus-induced changes in the internal pH of the secretory vesicles of chromaffin cells and enteric neurons. The internal acidity of chromaffin vesicles was increased by the nicotinic agonist 1,1-dimethyl-4-phenyl-piperazinium iodide (DMPP; in vivo and in vitro) and by high K+ (in vitro); and in enteric nerve terminals by exposure to veratridine or a plasmalemmal [Ca2+]o receptor agonist (Gd3+). Stimulation-induced acidification of chromaffin vesicles was [Ca2+]o-dependent and blocked by agents that inhibit the vacuolar proton pump (vH+-ATPase) or flux through Cl- channels. Stimulation also increased the average volume of chromaffin vesicles and the proportion that displayed a clear halo around their dense cores (called active vesicles). Stimulation-induced increases in internal acidity and size were greatest in active vesicles. Stimulation of chromaffin cells in the presence of a plasma membrane marker revealed that membrane was internalized in endosomes but not in chromaffin vesicles. The stable expression of botulinum toxin E to prevent exocytosis did not affect the stimulation-induced acidification of the secretory vesicles of mouse neuroblastoma Neuro2A cells. Stimulation-induced acidification thus occurs independently of exocytosis. The quantal size of secreted catecholamines, measured by amperometry in cultured chromaffin cells, was found to be increased either by prior exposure to L-DOPA or stimulation by high K+, and decreased by inhibition of vH+-ATPase or flux through Cl- channels. These observations are consistent with the hypothesis that the content of releasable small molecules in secretory vesicles is increased when the driving force for their uptake is enhanced, either by increasing the transmembrane concentration or pH gradients.


Assuntos
Medula Suprarrenal/citologia , Células Cromafins/citologia , Concentração de Íons de Hidrogênio , Macrolídeos , Animais , Antibacterianos/farmacologia , Bovinos , Tamanho Celular/fisiologia , Células Cultivadas , Células Cromafins/ultraestrutura , Estimulação Elétrica , Eletroquímica/métodos , Homeostase/fisiologia , Imuno-Histoquímica , Masculino , Microscopia Imunoeletrônica , Organelas/efeitos dos fármacos , Organelas/fisiologia , Organelas/ultraestrutura , Potássio/farmacologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA