Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cell ; 176(5): 1098-1112.e18, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30794774

RESUMEN

Increased levels of intestinal bile acids (BAs) are a risk factor for colorectal cancer (CRC). Here, we show that the convergence of dietary factors (high-fat diet) and dysregulated WNT signaling (APC mutation) alters BA profiles to drive malignant transformations in Lgr5-expressing (Lgr5+) cancer stem cells and promote an adenoma-to-adenocarcinoma progression. Mechanistically, we show that BAs that antagonize intestinal farnesoid X receptor (FXR) function, including tauro-ß-muricholic acid (T-ßMCA) and deoxycholic acid (DCA), induce proliferation and DNA damage in Lgr5+ cells. Conversely, selective activation of intestinal FXR can restrict abnormal Lgr5+ cell growth and curtail CRC progression. This unexpected role for FXR in coordinating intestinal self-renewal with BA levels implicates FXR as a potential therapeutic target for CRC.


Asunto(s)
Neoplasias Intestinales/metabolismo , Células Madre Neoplásicas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Línea Celular , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Ácido Desoxicólico/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Intestinales/genética , Intestinos , Hígado , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/fisiología , Organoides/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Factores de Riesgo , Transducción de Señal , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/metabolismo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología
2.
Nature ; 586(7830): 606-611, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32814902

RESUMEN

Islets derived from stem cells hold promise as a therapy for insulin-dependent diabetes, but there remain challenges towards achieving this goal1-6. Here we generate human islet-like organoids (HILOs) from induced pluripotent stem cells and show that non-canonical WNT4 signalling drives the metabolic maturation necessary for robust ex vivo glucose-stimulated insulin secretion. These functionally mature HILOs contain endocrine-like cell types that, upon transplantation, rapidly re-establish glucose homeostasis in diabetic NOD/SCID mice. Overexpression of the immune checkpoint protein programmed death-ligand 1 (PD-L1) protected HILO xenografts such that they were able to restore glucose homeostasis in immune-competent diabetic mice for 50 days. Furthermore, ex vivo stimulation with interferon-γ induced endogenous PD-L1 expression and restricted T cell activation and graft rejection. The generation of glucose-responsive islet-like organoids that are able to avoid immune detection provides a promising alternative to cadaveric and device-dependent therapies in the treatment of diabetes.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Evasión Inmune , Islotes Pancreáticos/citología , Islotes Pancreáticos/inmunología , Organoides/citología , Organoides/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Línea Celular , Epigénesis Genética , Femenino , Glucosa/metabolismo , Rechazo de Injerto , Xenoinjertos , Homeostasis , Humanos , Tolerancia Inmunológica , Secreción de Insulina , Trasplante de Islotes Pancreáticos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Organoides/trasplante , Linfocitos T/citología , Linfocitos T/inmunología , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt4/metabolismo , Proteína Wnt4/farmacología
4.
Cell Metab ; 34(1): 171-183.e6, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986332

RESUMEN

Inexorable increases in insulin resistance, lipolysis, and hepatic glucose production (HGP) are hallmarks of type 2 diabetes. Previously, we showed that peripheral delivery of exogenous fibroblast growth factor 1 (FGF1) has robust anti-diabetic effects mediated by the adipose FGF receptor (FGFR) 1. However, its mechanism of action is not known. Here, we report that FGF1 acutely lowers HGP by suppressing adipose lipolysis. On a molecular level, FGF1 inhibits the cAMP-protein kinase A axis by activating phosphodiesterase 4D (PDE4D), which separates it mechanistically from the inhibitory actions of insulin via PDE3B. We identify Ser44 as an FGF1-induced regulatory phosphorylation site in PDE4D that is modulated by the feed-fast cycle. These findings establish the FGF1/PDE4 pathway as an alternate regulator of the adipose-HGP axis and identify FGF1 as an unrecognized regulator of fatty acid homeostasis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Humanos , Insulina/metabolismo , Lipólisis/fisiología
6.
Nat Rev Endocrinol ; 13(10): 599-609, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28664920

RESUMEN

A hypercaloric diet combined with a sedentary lifestyle is a major risk factor for the development of insulin resistance, type 2 diabetes mellitus (T2DM) and associated comorbidities. Standard treatment for T2DM begins with lifestyle modification, and includes oral medications and insulin therapy to compensate for progressive ß-cell failure. However, current pharmaceutical options for T2DM are limited in that they do not maintain stable, durable glucose control without the need for treatment intensification. Furthermore, each medication is associated with adverse effects, which range from hypoglycaemia to weight gain or bone loss. Unexpectedly, fibroblast growth factor 1 (FGF1) and its low mitogenic variants have emerged as potentially safe candidates for restoring euglycaemia, without causing overt adverse effects. In particular, a single peripheral injection of FGF1 can lower glucose to normal levels within hours, without the risk of hypoglycaemia. Similarly, a single intracerebroventricular injection of FGF1 can induce long-lasting remission of the diabetic phenotype. This Review discusses potential mechanisms by which centrally administered FGF1 improves central glucose-sensing and peripheral glucose uptake in a sustained manner. Specifically, we explore the potential crosstalk between FGF1 and glucose-sensing neuronal circuits, hypothalamic neural stem cells and synaptic plasticity. Finally, we highlight therapeutic considerations of FGF1 and compare its metabolic actions with FGF15 (rodents), FGF19 (humans) and FGF21.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Factor 1 de Crecimiento de Fibroblastos/administración & dosificación , Resistencia a la Insulina , Insulina/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/diagnóstico , Modelos Animales de Enfermedad , Femenino , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Estudios de Seguimiento , Humanos , Masculino , Ratones Endogámicos C57BL , Distribución Aleatoria , Rol , Resultado del Tratamiento
7.
J Clin Invest ; 127(3): 1061-1074, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28218624

RESUMEN

MicroRNAs (miRNAs) are negative modulators of gene expression that fine-tune numerous biological processes. miRNA loss-of-function rarely results in highly penetrant phenotypes, but rather, influences cellular responses to physiologic and pathophysiologic stresses. Here, we have reported that a single member of the evolutionarily conserved miR-7 family, miR-7a2, is essential for normal pituitary development and hypothalamic-pituitary-gonadal (HPG) function in adulthood. Genetic deletion of mir-7a2 causes infertility, with low levels of gonadotropic and sex steroid hormones, small testes or ovaries, impaired spermatogenesis, and lack of ovulation in male and female mice, respectively. We found that miR-7a2 is highly expressed in the pituitary, where it suppresses golgi glycoprotein 1 (GLG1) expression and downstream bone morphogenetic protein 4 (BMP4) signaling and also reduces expression of the prostaglandin F2a receptor negative regulator (PTGFRN), an inhibitor of prostaglandin signaling and follicle-stimulating hormone (FSH) and luteinizing hormone (LH) secretion. Our results reveal that miR-7a2 critically regulates sexual maturation and reproductive function by interconnecting miR-7 genomic circuits that regulate FSH and LH synthesis and secretion through their effects on pituitary prostaglandin and BMP4 signaling.


Asunto(s)
Hipogonadismo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Infertilidad/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Femenino , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/metabolismo , Hormonas Esteroides Gonadales/genética , Hormonas Esteroides Gonadales/metabolismo , Hipogonadismo/genética , Infertilidad/genética , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Ovario/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Testículo/metabolismo
8.
Sci Rep ; 6: 38611, 2016 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-27934886

RESUMEN

Inactivation of transcription factor Foxa1 in mice results in neonatal mortality of unknown cause. Here, we report that ablation of Foxa1 causes impaired development and loss of the subthalamic nucleus (STN). Functional deficits in the STN have been implicated in the etiology of Huntington's and Parkinson's disease. We show that neuronal ablation by Synapsin1-Cre-mediated Foxa1 deletion is sufficient to induce hyperlocomotion in mice. Transcriptome profiling of STN neurons in conditional Foxa1 knockout mice revealed changes in gene expression reminiscent of those in neurodegenerative diseases. We identified Ppargc1a, a transcriptional co-activator that is implicated in neurodegeneration, as a Foxa1 target. These findings were substantiated by the observation of Foxa1-dependent demise of STN neurons in conditional models of Foxa1 mutant mice. Finally, we show that the spontaneous firing activity of Foxa1-deficient STN neurons is profoundly impaired. Our data reveal so far elusive roles of Foxa1 in the development and maintenance of STN function.


Asunto(s)
Factor Nuclear 3-alfa del Hepatocito/genética , Núcleo Subtalámico/fisiología , Animales , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Genes Esenciales , Genes Reporteros , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Hipercinesia/genética , Masculino , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Especificidad de Órganos/genética , Organogénesis/genética , Núcleo Subtalámico/embriología , Transcriptoma
9.
Cell Metab ; 17(3): 436-47, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23416070

RESUMEN

Circulating levels of insulin and glucagon reflect the nutritional state of animals and elicit regulatory responses in the liver that maintain glucose and lipid homeostasis. The transcription factor Foxa2 activates lipid metabolism and ketogenesis during fasting and is inhibited via insulin-PI3K-Akt signaling-mediated phosphorylation at Thr156 and nuclear exclusion. Here we show that, in addition, Foxa2 is acetylated at the conserved residue Lys259 following inhibition of histone deacetylases (HDACs) class I-III and the cofactors p300 and SirT1 are involved in Foxa2 acetylation and deacetylation, respectively. Physiologically, fasting states and glucagon stimulation are sufficient to induce Foxa2 acetylation. Introduction of the acetylation-mimicking (K259Q) or -deficient (K259R) mutations promotes or inhibits Foxa2 activity, respectively, and adenoviral expression of Foxa2-K259Q augments expression of genes involved in fatty acid oxidation and ketogenesis. Our study reveals a molecular mechanism by which glucagon signaling activates a fasting response through acetylation of Foxa2.


Asunto(s)
Ayuno/fisiología , Glucagón/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Metabolismo de los Lípidos/fisiología , Hígado/fisiología , Acetilación , Adenoviridae , Animales , Compuestos Azo , Línea Celular , Inmunoprecipitación de Cromatina , Ayuno/metabolismo , Vectores Genéticos , Células Hep G2 , Factor Nuclear 3-beta del Hepatocito/genética , Histona Desacetilasas/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Hígado/metabolismo , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Mutagénesis/genética , Mutagénesis Sitio-Dirigida , Interferencia de ARN , Sirtuina 1/metabolismo , Factores de Transcripción p300-CBP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA