Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Gastroenterology ; 166(3): 437-449, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37995867

RESUMEN

BACKGROUND & AIMS: RET tyrosine kinase is necessary for enteric nervous system development. Loss-of-function RET mutations cause Hirschsprung disease (HSCR), in which infants are born with aganglionic bowel. Despite surgical correction, patients with HSCR often experience chronic defecatory dysfunction and enterocolitis, suggesting that RET is important after development. To test this hypothesis, we determined the location of postnatal RET and its significance in gastrointestinal (GI) motility. METHODS: RetCFP/+ mice and human transcriptional profiling data were studied to identify the enteric neuronal and epithelial cells that express RET. To determine whether RET regulates gut motility in vivo, genetic, and pharmacologic approaches were used to disrupt RET in all RET-expressing cells, a subset of enteric neurons, or intestinal epithelial cells. RESULTS: Distinct subsets of enteric neurons and enteroendocrine cells expressed RET in the adult intestine. RET disruption in the epithelium, rather than in enteric neurons, slowed GI motility selectively in male mice. RET kinase inhibition phenocopied this effect. Most RET+ epithelial cells were either enterochromaffin cells that release serotonin or L-cells that release peptide YY (PYY) and glucagon-like peptide 1 (GLP-1), both of which can alter motility. RET kinase inhibition exaggerated PYY and GLP-1 release in a nutrient-dependent manner without altering serotonin secretion in mice and human organoids. PYY receptor blockade rescued dysmotility in mice lacking epithelial RET. CONCLUSIONS: RET signaling normally limits nutrient-dependent peptide release from L-cells and this activity is necessary for normal intestinal motility in male mice. These effects could contribute to dysmotility in HSCR, which predominantly affects males, and uncovers a mechanism that could be targeted to treat post-prandial GI dysfunction.


Asunto(s)
Sistema Nervioso Entérico , Enfermedad de Hirschsprung , Lactante , Humanos , Masculino , Ratones , Animales , Péptido YY , Serotonina , Enfermedad de Hirschsprung/genética , Células Enteroendocrinas , Intestino Delgado , Péptido 1 Similar al Glucagón , Proteínas Proto-Oncogénicas c-ret/genética
2.
Gut ; 72(5): 870-881, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-35537812

RESUMEN

OBJECTIVE: Intestinal barrier loss is a Crohn's disease (CD) risk factor. This may be related to increased expression and enzymatic activation of myosin light chain kinase 1 (MLCK1), which increases intestinal paracellular permeability and correlates with CD severity. Moreover, preclinical studies have shown that MLCK1 recruitment to cell junctions is required for tumour necrosis factor (TNF)-induced barrier loss as well as experimental inflammatory bowel disease progression. We sought to define mechanisms of MLCK1 recruitment and to target this process pharmacologically. DESIGN: Protein interactions between FK506 binding protein 8 (FKBP8) and MLCK1 were assessed in vitro. Transgenic and knockout intestinal epithelial cell lines, human intestinal organoids, and mice were used as preclinical models. Discoveries were validated in biopsies from patients with CD and control subjects. RESULTS: MLCK1 interacted specifically with the tacrolimus-binding FKBP8 PPI domain. Knockout or dominant negative FKBP8 expression prevented TNF-induced MLCK1 recruitment and barrier loss in vitro. MLCK1-FKBP8 binding was blocked by tacrolimus, which reversed TNF-induced MLCK1-FKBP8 interactions, MLCK1 recruitment and barrier loss in vitro and in vivo. Biopsies of patient with CD demonstrated increased numbers of MLCK1-FKBP8 interactions at intercellular junctions relative to control subjects. CONCLUSION: Binding to FKBP8, which can be blocked by tacrolimus, is required for MLCK1 recruitment to intercellular junctions and downstream events leading to immune-mediated barrier loss. The observed increases in MLCK1 activity, MLCK1 localisation at cell junctions and perijunctional MLCK1-FKBP8 interactions in CD suggest that targeting this process may be therapeutic in human disease. These new insights into mechanisms of disease-associated barrier loss provide a critical foundation for therapeutic exploitation of FKBP8-MLCK1 interactions.


Asunto(s)
Enfermedad de Crohn , Animales , Humanos , Ratones , Células CACO-2 , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/metabolismo , Mucosa Intestinal/metabolismo , Ratones Noqueados , Quinasa de Cadena Ligera de Miosina/metabolismo , Tacrolimus/farmacología , Proteínas de Unión a Tacrolimus/metabolismo , Uniones Estrechas/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
3.
Development ; 140(19): 3939-49, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24046315

RESUMEN

Adipose tissue is formed at stereotypic times and locations in a diverse array of organisms. Once formed, the tissue is dynamic, responding to homeostatic and external cues and capable of a 15-fold expansion. The formation and maintenance of adipose tissue is essential to many biological processes and when perturbed leads to significant diseases. Despite this basic and clinical significance, understanding of the developmental biology of adipose tissue has languished. In this Review, we highlight recent efforts to unveil adipose developmental cues, adipose stem cell biology and the regulators of adipose tissue homeostasis and dynamism.


Asunto(s)
Tejido Adiposo/citología , Adipocitos/citología , Animales , Diferenciación Celular/fisiología , Humanos , Nicho de Células Madre/fisiología , Células Madre/citología
4.
Dev Cell ; 59(2): 244-261.e6, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38154460

RESUMEN

WNT morphogens trigger signaling pathways fundamental for embryogenesis, regeneration, and cancer. WNTs are modified with palmitoleate, which is critical for binding Frizzled (FZD) receptors and activating signaling. However, it is unknown how WNTs are released and spread from cells, given their strong lipid-dependent membrane attachment. We demonstrate that secreted FZD-related proteins and WNT inhibitory factor 1 are WNT carriers, potently releasing lipidated WNTs and forming active soluble complexes. WNT release occurs by direct handoff from the membrane protein WNTLESS to the carriers. In turn, carriers donate WNTs to glypicans and FZDs involved in WNT reception and to the NOTUM hydrolase, which antagonizes WNTs by lipid moiety removal. WNT transfer from carriers to FZDs is greatly facilitated by glypicans that serve as essential co-receptors in Wnt signaling. Thus, an extracellular network of carriers dynamically controls secretion, posttranslational regulation, and delivery of WNT morphogens, with important practical implications for regenerative medicine.


Asunto(s)
Glipicanos , Proteínas Wnt , Proteínas Wnt/metabolismo , Glipicanos/metabolismo , Vía de Señalización Wnt , Desarrollo Embrionario , Lípidos , Receptores Frizzled/química , Receptores Frizzled/metabolismo
5.
Cell Metab ; 6(3): 195-207, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17767906

RESUMEN

Adipose (Adp) is an evolutionarily conserved gene isolated from naturally occurring obese flies homozygous for an adp mutation. Here we show that the anti-obesity function of Adp (worm Y73E7A.9, fly adp, and murine Wdtc1) is conserved from worms to mammals. Further, Adp appears to inhibit fat formation in a dosage-sensitive manner. Adp heterozygous flies and Adp heterozygous mutant mice are obese and insulin resistant, as are mice that express a dominant negative form of Adp in fat cells. Conversely, fat-restricted Adp transgenic mice are lean and display improved metabolic profiles. A transient transgenic increase in Adp activity in adult fly fat tissues reduces fat accumulation, indicating therapeutic potential. ADP may elicit these anti-adipogenic functions by regulating chromatin dynamics and gene transcription, as it binds both histones and HDAC3 and inhibits PPARgamma activity. Thus Adp appears to be involved in an ancient pathway that regulates fat accumulation.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Obesidad/genética , Proteínas/genética , Proteínas/metabolismo , Tejido Adiposo/anatomía & histología , Tejido Adiposo/fisiología , Animales , Caenorhabditis elegans/anatomía & histología , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Drosophila/antagonistas & inhibidores , Femenino , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/prevención & control , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Obesidad/prevención & control , Proteínas/antagonistas & inhibidores
6.
Nat Commun ; 13(1): 261, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017529

RESUMEN

Enteroendocrine (EE) cells are the most abundant hormone-producing cells in humans and are critical regulators of energy homeostasis and gastrointestinal function. Challenges in converting human intestinal stem cells (ISCs) into functional EE cells, ex vivo, have limited progress in elucidating their role in disease pathogenesis and in harnessing their therapeutic potential. To address this, we employed small molecule targeting of the endocannabinoid receptor signaling pathway, JNK, and FOXO1, known to mediate endodermal development and/or hormone production, together with directed differentiation of human ISCs from the duodenum and rectum. We observed marked induction of EE cell differentiation and gut-derived expression and secretion of SST, 5HT, GIP, CCK, GLP-1 and PYY upon treatment with various combinations of three small molecules: rimonabant, SP600125 and AS1842856. Robust differentiation strategies capable of driving human EE cell differentiation is a critical step towards understanding these essential cells and the development of cell-based therapeutics.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Antracenos/farmacología , Cromogranina A/metabolismo , Endocannabinoides/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Péptido YY/metabolismo , Quinolonas/farmacología , Rimonabant/farmacología , Transducción de Señal , Somatostatina/metabolismo , Factores de Transcripción/metabolismo
7.
J Clin Endocrinol Metab ; 107(4): e1610-e1619, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-34791361

RESUMEN

CONTEXT: Pseudohypoparathyroidism type Ib (PHP1B) is characterized by hypocalcemia and hyperphosphatemia due to parathyroid hormone resistance in the proximal renal tubules. Maternal pathogenic STX16/GNAS variants leading to maternal epigenetic GNAS changes impair expression of the stimulatory G protein alpha-subunit (Gsα) thereby causing autosomal dominant PHP1B. In contrast, genetic defects responsible for sporadic PHP1B (sporPHP1B) remain mostly unknown. OBJECTIVE: Determine whether PHP1B encountered after in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) causes GNAS remethylation defects similar to those in sporPHP1B. DESIGN: Retrospective analysis. RESULTS: Nine among 36 sporPHP1B patients investigated since 2000, all with loss of methylation (LOM) at the 3 maternal GNAS differentially methylated regions (DMRs) and gain of methylation at the paternal NESP DMR, had been conceived through IVF or ICSI. Besides abnormal GNAS methylation, IVF/ICSI PHP1B cases revealed no additional imprinting defects. Three of these PHP1B patients have dizygotic twins, and 4 have IVF/ICSI-conceived siblings, all with normal GNAS methylation; 2 unaffected younger siblings were conceived naturally. CONCLUSION: Sporadic and IVF/ICSI-conceived PHP1B patients revealed indistinguishable epigenetic changes at all 4 GNAS DMRs, thus suggesting a similar underlying disease mechanism. Given that remethylation at the 3 maternal DMRs occurs during oogenesis, male factors are unlikely to cause LOM postfertilization. Instead, at least some of the sporPHP1B variants could be caused by a defect or defects in an oocyte-expressed gene that is required for fertility and for re-establishing maternal GNAS methylation imprints. It remains uncertain, however, whether the lack of GNAS remethylation alone and the resulting reduction in Gsα expression is sufficient to impair oocyte maturation.


Asunto(s)
Cromograninas , Seudohipoparatiroidismo , Cromograninas/genética , Metilación de ADN , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Masculino , Oogénesis , Seudohipoparatiroidismo/genética , Estudios Retrospectivos , Seudohipoparatiroidismo
8.
Nat Commun ; 13(1): 6786, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36351897

RESUMEN

Toxin B (TcdB) is a major exotoxin responsible for diseases associated with Clostridioides difficile infection. Its sequence variations among clinical isolates may contribute to the difficulty in developing effective therapeutics. Here, we investigate receptor-binding specificity of major TcdB subtypes (TcdB1 to TcdB12). We find that representative members of subtypes 2, 4, 7, 10, 11, and 12 do not recognize the established host receptor, frizzled proteins (FZDs). Using a genome-wide CRISPR-Cas9-mediated screen, we identify tissue factor pathway inhibitor (TFPI) as a host receptor for TcdB4. TFPI is recognized by a region in TcdB4 that is homologous to the FZD-binding site in TcdB1. Analysis of 206 TcdB variant sequences reveals a set of six residues within this receptor-binding site that defines a TFPI binding-associated haplotype (designated B4/B7) that is present in all TcdB4 members, a subset of TcdB7, and one member of TcdB2. Intragenic micro-recombination (IR) events have occurred around this receptor-binding region in TcdB7 and TcdB2 members, resulting in either TFPI- or FZD-binding capabilities. Introduction of B4/B7-haplotype residues into TcdB1 enables dual recognition of TFPI and FZDs. Finally, TcdB10 also recognizes TFPI, although it does not belong to the B4/B7 haplotype, and shows species selectivity: it recognizes TFPI of chicken and to a lesser degree mouse, but not human, dog, or cattle versions. These findings identify TFPI as a TcdB receptor and reveal IR-driven changes on receptor-specificity among TcdB variants.


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Animales , Bovinos , Perros , Ratones , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/química , Clostridioides difficile/genética , Recombinación Genética , Humanos
9.
BMJ Case Rep ; 12(3)2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30898952

RESUMEN

11-year old twin boy found to have idiopathic precocious puberty after routine well-child examination revealed discordant pubertal growth between the two brothers.


Asunto(s)
Pubertad Precoz/diagnóstico , Gemelos Dicigóticos , Acné Vulgar , Niño , Gráficos de Crecimiento , Cabello , Humanos , Masculino , Examen Físico
10.
Brain Res ; 1140: 96-104, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16359645

RESUMEN

Nitric oxide (NO) is a diffusible messenger molecule produced primarily by neuronal nitric oxide synthase (nNOS) in the central nervous system. Both nNOS expression and NO production are regulated by calcium ions. Leaner and tottering mice carry a mutation in the pore forming subunit (alpha1A) of P/Q-type voltage-gated calcium ion channels, which decreases calcium ion current through the affected channels and disrupts calcium homeostasis. We have previously shown that nNOS expression is altered in adult leaner and tottering cerebella. In addition, leaner and tottering mice have been shown to have abnormal cerebellar granule cell-Purkinje cell synapses and leaner cerebellar granule cells undergo abnormal apoptosis during early postnatal development. Since NO production has been linked to several developmental roles including neuronal cell death, synaptogenesis and neuronal cell survival, our objective was to evaluate the expression of nNOS in developing leaner and tottering cerebella. Our results show that nNOS is differentially expressed in leaner and tottering cerebella compared to wild type cerebella and compared to each other. In whole cerebella, Western blotting revealed a significant increase in nNOS expression at postnatal day 12 in tottering but not leaner or wild type cerebella. At the cellular level the NADPH-diaphorase marker for nNOS revealed a significant increase in nNOS expression in basket cell interneurons in both mutant mice. nNOS expression in granule cells in the internal granule cell layer in tottering mice was increased at P12, while granule cells of leaner mice exhibited decreased nNOS expression at P20. The changes in nNOS expression at P12 did not correlate with a change in overall NO production, but rather maintained wild type NO concentrations. These findings suggest that changes in nNOS expression in the leaner and tottering cerebella are compensatory in nature with NO most likely functioning as a calcium-regulated neuroprotective/neurotrophic factor in postnatal cerebellar development.


Asunto(s)
Canales de Calcio Tipo P/genética , Canales de Calcio Tipo Q/genética , Cerebelo/enzimología , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones Mutantes Neurológicos/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Cerebelo/citología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Deshidrogenasa/metabolismo , Neuronas/enzimología , Óxido Nítrico Sintasa de Tipo I/genética , Fenotiazinas
11.
Am Psychol ; 62(2): 131-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17324038

RESUMEN

In 1974, a story was published about clandestine research done by John B. Watson that was judged to be so reprehensible that it was offered as the real reason he was fired from his faculty position at Johns Hopkins University in 1920, at perhaps the peak of his academic career. Watson's dismissal from Johns Hopkins may have been the most important event in his career, and it almost certainly altered the history of American psychology. Thus, this story has great significance. The claims of the story, however, have never been validated or invalidated. This article examines the evidence for and against the existence of such research and discusses Watson's academic dismissal in light of that evidence.


Asunto(s)
Investigación Conductal/historia , Psicología/historia , Conducta Sexual/historia , Baltimore , Investigación Conductal/métodos , Behaviorismo/historia , Historia del Siglo XX , Humanos , Conducta Sexual/fisiología , Universidades/historia
12.
Alcohol ; 39(2): 65-72, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17134658

RESUMEN

Previous studies have reported that alcohol exposure reduces the number of neuronal-like pheochromocytoma (PC12) cells in culture. In this study, the interactive effect of coexposure of alcohol and nicotine on PC12 cell numbers was examined in comparison with the effect derived from alcohol or nicotine exposure individually. Moreover, the role of apoptosis in mediating changes in PC12 cell numbers was also investigated. It was hypothesized that alcohol would result in cell loss, and the presence of nicotine would attenuate the damaging effects of alcohol. PC12 cells were exposed to alcohol (100 mM), nicotine (10 microM), or both alcohol and nicotine for 24, 48, 72, or 96 h. Caspase-3 activity and DNA fragmentation, markers for apoptotic cell death, were measured to determine the role of apoptosis in mediating decreases in PC12 cell numbers. The findings indicated that both alcohol and nicotine exposure significantly decreased PC12 cell numbers when compared with the control treatment. Furthermore, the coexposure of these two drugs caused a significantly greater decrease in cell numbers when compared with cells exposed to either alcohol or nicotine alone. This additive effect was related to the duration of exposure with a marked reduction in cell numbers following 96 h of coexposure to alcohol and nicotine. Neither alcohol nor nicotine exposure appeared to alter caspase-3 activity or DNA fragmentation levels, suggesting that the reduction in PC12 cell numbers following alcohol and/or nicotine exposure may possibly be due to factors other than apoptosis, such as interference with proliferation rates.


Asunto(s)
Etanol/farmacología , Factor de Crecimiento Nervioso/farmacología , Nicotina/farmacología , Células PC12/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Interacciones Farmacológicas , Ratas
13.
PLoS One ; 11(3): e0152129, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27015423

RESUMEN

Exercise decreases adiposity and improves metabolic health; however, the physiological and molecular underpinnings of these phenomena remain unknown. Here, we investigate the effect of endurance training on adipose progenitor lineage commitment. Using mice with genetically labeled adipose progenitors, we show that these cells react to exercise by decreasing their proliferation and differentiation potential. Analyses of mouse models that mimic the skeletal muscle adaptation to exercise indicate that muscle, in a non-autonomous manner, regulates adipose progenitor homeostasis, highlighting a role for muscle-derived secreted factors. These findings support a humoral link between skeletal muscle and adipose progenitors and indicate that manipulation of adipose stem cell function may help address obesity and diabetes.


Asunto(s)
Tejido Adiposo/citología , Músculo Esquelético/fisiología , Condicionamiento Físico Animal , Células Madre/citología , Células 3T3-L1 , Adaptación Fisiológica , Adipocitos/citología , Tejido Adiposo/metabolismo , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Proliferación Celular , Medios de Cultivo Condicionados , Diabetes Mellitus/metabolismo , Prueba de Tolerancia a la Glucosa , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis , Masculino , Ratones , Ratones Transgénicos , Obesidad/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Resistencia Física/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Trombospondinas/metabolismo
14.
Brain Res Dev Brain Res ; 160(2): 231-8, 2005 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-16256207

RESUMEN

It has been suggested that developmental alcohol-induced brain damage is mediated through increases in oxidative stress. In this study, the concentrations of malondialdehyde (MDA) and reduced glutathione (GSH) were measured to indicate alcohol-mediated oxidative stress. In addition, the ability of two known antioxidants, melatonin (MEL) and lazaroid U-83836E (U), to attenuate alcohol-induced oxidative stress was investigated. Sprague-Dawley rat pups were randomly assigned to six artificially-reared groups, ALC (alcohol), MEL, MEL/ALC, U, U/ALC, and GC (gastrostomy control), and one normal suckle control (to control for artificial-rearing effects on the dependent variables). The daily dosages for ALC, MEL, and U were 6 g/kg, 20 mg/kg, and 20 mg/kg, respectively. Alcohol was administered in 2 consecutive feedings, and antioxidant (MEL or U) was administered for a total of 4 consecutive feedings (2 feedings prior to and 2 feedings concurrently with alcohol). The animals received treatment from postnatal days (PD) 4 through 9. Cerebellar, hippocampal, and cortical samples were collected on PD 9 and analyzed for MDA and GSH content. The results indicated that MDA concentrations in the cerebellum were significantly elevated in animals receiving alcohol; however, MDA levels in the hippocampus and cortex were not affected by alcohol treatment. Additionally, GSH levels in the cerebellum were significantly elevated in groups receiving alcohol, regardless of antioxidant treatment. Neither antioxidant was able to protect against alcohol-induced alterations of MDA or GSH. These findings suggest that alcohol might increase GSH levels indirectly as a compensatory mechanism designed to protect the brain from oxidative-stress-mediated insult.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Cerebelo , Etanol/farmacología , Glutatión/metabolismo , Malondialdehído/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Química Encefálica/efectos de los fármacos , Depresores del Sistema Nervioso Central/sangre , Cerebelo/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Cromanos/administración & dosificación , Interacciones Farmacológicas , Etanol/sangre , Melatonina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Piperazinas/administración & dosificación , Ratas , Ratas Sprague-Dawley
15.
Nat Commun ; 5: 5196, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25330806

RESUMEN

Oestrogen, often via oestrogen receptor alpha (ERα) signalling, regulates metabolic physiology, highlighted by post-menopausal temperature dysregulation (hot flashes), glucose intolerance, increased appetite and reduced metabolic rate. Here we show that ERα signalling has a role in adipose lineage specification in mice. ERα regulates adipose progenitor identity and potency, promoting white adipogenic lineage commitment. White adipose progenitors lacking ERα reprogramme and enter into smooth muscle and brown adipogenic fates. Mechanistic studies highlight a TGFß programme involved in progenitor reprogramming downstream of ERα signalling. The observed reprogramming has profound metabolic outcomes; both female and male adipose-lineage ERα-mutant mice are lean, have improved glucose sensitivity and are resistant to weight gain on a high-fat diet. Further, they are hypermetabolic, hyperphagic and hyperthermic, all consistent with a brown phenotype. Together, these findings indicate that ERα cell autonomously regulates adipose lineage commitment, brown fat and smooth muscle cell formation, and systemic metabolism, in a manner relevant to prevalent metabolic diseases.


Asunto(s)
Tejido Adiposo Pardo/citología , Diferenciación Celular , Estrógenos/metabolismo , Miocitos del Músculo Liso/citología , Transducción de Señal , Células Madre/metabolismo , Tejido Adiposo/citología , Tejido Adiposo Blanco/citología , Animales , Linaje de la Célula , Proliferación Celular , Separación Celular , Receptor alfa de Estrógeno/metabolismo , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Mutantes , Mutación , Neovascularización Fisiológica , Fenotipo , Distribución Aleatoria , Células Madre/citología , Factor de Crecimiento Transformador beta/metabolismo
16.
Mol Metab ; 2(3): 227-42, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24049737

RESUMEN

Our data demonstrate that estrogens, estrogen receptor-α (ERα), and estrogen receptor-ß (ERß) regulate adipose tissue distribution, inflammation, fibrosis, and glucose homeostasis, by determining that αERKO mice have increased adipose tissue inflammation and fibrosis prior to obesity onset. Selective deletion of adipose tissue ERα in adult mice using a novel viral vector technology recapitulated the findings in the total body ERα null mice. Generation of a novel mouse model, lacking ERα specifically from adipocytes (AdipoERα), demonstrated increased markers of fibrosis and inflammation, especially in the males. Additionally, we found that the beneficial effects of estrogens on adipose tissue require adipocyte ERα. Lastly, we determined the role of ERß in regulating inflammation and fibrosis, by breeding the AdipoERα into the ßERKO background and found that in the absence of adipocyte ERα, ERß has a protective role. These data suggest that adipose tissue and adipocyte ERα protects against adiposity, inflammation, and fibrosis in both males and females.

17.
Cell Metab ; 15(4): 492-504, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22482731

RESUMEN

Adipose tissues provide circulating nutrients and hormones. We present in vivo mouse studies highlighting roles for Wnt signals in both aspects of metabolism. ß-catenin activation in PPARγ-expressing fat progenitors (PBCA) decreased fat mass and induced fibrotic replacement of subcutaneous fat specifically. In spite of lipodystrophy, PBCA mice did not develop the expected diabetes and hepatosteatosis, but rather exhibited improved glucose metabolism and normal insulin sensitivity. Glucose uptake was increased in muscle independently of insulin, associated with cell-surface translocation of glucose transporters and AMPK activation. Ex vivo assays showed these effects were likely secondary to blood-borne signals since PBCA sera or conditioned media from PBCA fat progenitors enhanced glucose uptake and activated AMPK in muscle cultures. Thus, adipose progenitor Wnt activation dissociates lipodystrophy from dysfunctional metabolism and highlights a fat-muscle endocrine axis, which may represent a potential therapy to lower blood glucose and improve metabolism.


Asunto(s)
Adipocitos/metabolismo , Adipocitos/patología , Glucosa/metabolismo , Músculos/metabolismo , Células Madre/metabolismo , Células Madre/patología , Vía de Señalización Wnt , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/efectos de los fármacos , Adiposidad/efectos de los fármacos , Animales , Transporte Biológico/efectos de los fármacos , Compartimento Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Insulina/metabolismo , Lipodistrofia/metabolismo , Lipodistrofia/patología , Ratones , Ratones Mutantes , Músculos/efectos de los fármacos , Mutación/genética , PPAR gamma/metabolismo , Células Madre/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
19.
Cell Metab ; 14(1): 116-22, 2011 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-21723509

RESUMEN

White adipose tissue regulates metabolism; the importance of this control is highlighted by the ongoing pandemic of obesity and associated complications such as diabetes, atherosclerosis, and cancer. White adipose tissue maintenance is a dynamic process, yet very little is known about how pharmacologic stimuli affect such plasticity. Combining in vivo lineage marking and BrdU labeling strategies, we found that rosiglitazone, a member of the thiazolidinedione class of glucose-lowering medicines, markedly increases the evolution of adipose progenitors into adipocytes. Notably, chronic rosiglitazone administration disrupts the adipogenic and self-renewal capacities of the stem cell compartment and alters its molecular characteristics. These data unravel unknown aspects of adipose dynamics and provide a basis to manipulate the adipose lineage for therapeutic ends.


Asunto(s)
Tejido Adiposo Blanco/citología , Tiazolidinedionas/farmacología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Ratones , Células Madre/citología , Células Madre/metabolismo
20.
Mol Cell Biol ; 31(23): 4692-705, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21947280

RESUMEN

Osteoclasts are bone-resorbing cells essential for skeletal development, homeostasis, and regeneration. They derive from hematopoietic progenitors in the monocyte/macrophage lineage and differentiate in response to RANKL. However, the precise nature of osteoclast progenitors is a longstanding and important question. Using inducible peroxisome proliferator-activated receptor γ (PPARγ)-tTA TRE-GFP (green fluorescent protein) reporter mice, we show that osteoclast progenitors reside specifically in the PPARγ-expressing hematopoietic bone marrow population and identify the quiescent PPARγ(+) cells as osteoclast progenitors. Importantly, two PPARγ-tTA TRE-Cre-controlled genetic models provide compelling functional evidence. First, Notch activation in PPARγ(+) cells causes high bone mass due to impaired osteoclast precursor proliferation. Second, selective ablation of PPARγ(+) cells by diphtheria toxin also causes high bone mass due to decreased osteoclast numbers. Furthermore, PPARγ(+) cells respond to both pathological and pharmacological resorption-enhancing stimuli. Mechanistically, PPARγ promotes osteoclast progenitors by activating GATA2 transcription. These findings not only identify the long-sought-after osteoclast progenitors but also establish unprecedented tools for their visualization, isolation, characterization, and genetic manipulation.


Asunto(s)
Células de la Médula Ósea/citología , Factor de Transcripción GATA2/metabolismo , Osteoclastos/citología , PPAR gamma/metabolismo , Células Madre/citología , Fosfatasa Ácida/genética , Fosfatasa Ácida/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Colágeno Tipo I/orina , Femenino , Factor de Transcripción GATA2/genética , Expresión Génica , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Tamaño de los Órganos , Osteocalcina/sangre , Osteoclastos/metabolismo , PPAR gamma/genética , Fragmentos de Péptidos/orina , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Radiografía , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Madre/metabolismo , Fosfatasa Ácida Tartratorresistente , Tibia/anatomía & histología , Tibia/citología , Tibia/diagnóstico por imagen , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA