Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
FASEB J ; 29(2): 671-83, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25389133

RESUMEN

Retinoic acid signaling is required for maintaining a range of cellular processes, including cell differentiation, proliferation, and apoptosis. We investigated the actions of all-trans-retinoic acid (atRA) signaling in pancreatic ß-cells of adult mice. atRA signaling was ablated in ß-cells by overexpressing a dominant-negative retinoic acid receptor (RAR)-α mutant (RARdn) using an inducible Cre-Lox system under the control of the pancreas duodenal homeobox gene promoter. Our studies establish that hypomorphism for RAR in ß-cells leads to an age-dependent decrease in plasma insulin in the fed state and in response to a glucose challenge. Glucose-stimulated insulin secretion was also impaired in islets isolated from mice expressing RARdn. Among genes that are atRA responsive, Glut2 and Gck mRNA levels were decreased in isolated islets from RARdn-expressing mice. Histologic analyses of RARdn-expressing pancreata revealed a decrease in ß-cell mass and insulin per ß-cell 1 mo after induction of the RARdn. Our results indicate that atRA signaling mediated by RARs is required in the adult pancreas for maintaining both ß-cell function and mass, and provide insights into molecular mechanisms underlying these actions.


Asunto(s)
Glucemia/metabolismo , Células Secretoras de Insulina/citología , Insulina/metabolismo , Receptores de Ácido Retinoico/metabolismo , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Genotipo , Homeostasis , Insulina/sangre , Secreción de Insulina , Integrasas/metabolismo , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/metabolismo , Receptor alfa de Ácido Retinoico , Transducción de Señal , Tretinoina/metabolismo
2.
JCI Insight ; 4(3)2019 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-30728336

RESUMEN

Intronic polymorphisms in the α-ketoglutarate-dependent dioxygenase gene (FTO) that are highly associated with increased body weight have been implicated in the transcriptional control of a nearby ciliary gene, retinitis pigmentosa GTPase regulator-interacting protein-1 like (RPGRIP1L). Previous studies have shown that congenital Rpgrip1l hypomorphism in murine proopiomelanocortin (Pomc) neurons causes obesity by increasing food intake. Here, we show by congenital and adult-onset Rpgrip1l deletion in Pomc-expressing neurons that the hyperphagia and obesity are likely due to neurodevelopmental effects that are characterized by a reduction in the Pomc/Neuropeptide Y (Npy) neuronal number ratio and marked increases in arcuate hypothalamic-paraventricular hypothalamic (ARH-PVH) axonal projections. Biallelic RPGRIP1L mutations result in fewer cilia-positive human induced pluripotent stem cell-derived (iPSC-derived) neurons and blunted responses to Sonic Hedgehog (SHH). Isogenic human ARH-like embryonic stem cell-derived (ESc-derived) neurons homozygous for the obesity-risk alleles at rs8050136 or rs1421085 have decreased RPGRIP1L expression and have lower numbers of POMC neurons. RPGRIP1L overexpression increases POMC cell number. These findings suggest that apparently functional intronic polymorphisms affect hypothalamic RPGRIP1L expression and impact development of POMC neurons and their derivatives, leading to hyperphagia and increased adiposity.

3.
PLoS One ; 13(5): e0197548, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29847571

RESUMEN

We have previously reported that Ildr2 knockdown via adenovirally-delivered shRNA causes hepatic steatosis in mice. In the present study we investigated hepatic biochemical and anatomic phenotypes of Cre-mediated Ildr2 knock-out mice. Liver-specific Ildr2 knock-out mice were generated in C57BL/6J mice segregating for a floxed (exon 1) allele of Ildr2, using congenital and acute (10-13-week-old male mice) Cre expression. In addition, Ildr2 shRNA was administered to Ildr2 knock-out mice to test the effects of Ildr2 shRNA, per se, in the absence of Ildr2 expression. RNA sequencing was performed on livers of these knockdown and knockout mice. Congenital and acute liver-specific and hepatocyte-specific knockout mice did not develop hepatic steatosis. However, administration of Ildr2 shRNA to Ildr2 knock-out mice did cause hepatic steatosis, indicating that the Ildr2 shRNA had apparent "off-target" effects on gene(s) other than Ildr2. RNA sequencing and BLAST sequence alignment revealed Dgka as a candidate gene mediating these "off-target" effects. Ildr2 shRNA is 63% homologous to the Dgka gene, and Dgka expression decreased only in mice displaying hepatic steatosis. Dgka encodes diacylglycerol kinase (DGK) alpha, one of a family of DGKs which convert diacylglycerides to phosphatidic acid for second messenger signaling. Dgka knockdown mice would be expected to accumulate diacylglyceride, contributing to the observed hepatic steatosis. We conclude that ILDR2 plays a negligible role in hepatic steatosis. Rather, hepatic steatosis observed previously in Ildr2 knockdown mice was likely due to shRNA targeting of Dgka and/or other "off-target" genes. We propose that the gene candidates identified in this follow-up study may lead to identification of novel regulators of hepatic lipid metabolism.


Asunto(s)
Proteínas de la Membrana/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Diacilglicerol Quinasa/genética , Diacilglicerol Quinasa/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Hepatocitos/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/patología , ARN Interferente Pequeño/genética , Análisis de Secuencia de ARN , Triglicéridos/metabolismo
4.
J Clin Invest ; 127(1): 293-305, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27941249

RESUMEN

Prader-Willi syndrome (PWS) is caused by a loss of paternally expressed genes in an imprinted region of chromosome 15q. Among the canonical PWS phenotypes are hyperphagic obesity, central hypogonadism, and low growth hormone (GH). Rare microdeletions in PWS patients define a 91-kb minimum critical deletion region encompassing 3 genes, including the noncoding RNA gene SNORD116. Here, we found that protein and transcript levels of nescient helix loop helix 2 (NHLH2) and the prohormone convertase PC1 (encoded by PCSK1) were reduced in PWS patient induced pluripotent stem cell-derived (iPSC-derived) neurons. Moreover, Nhlh2 and Pcsk1 expression were reduced in hypothalami of fasted Snord116 paternal knockout (Snord116p-/m+) mice. Hypothalamic Agrp and Npy remained elevated following refeeding in association with relative hyperphagia in Snord116p-/m+ mice. Nhlh2-deficient mice display growth deficiencies as adolescents and hypogonadism, hyperphagia, and obesity as adults. Nhlh2 has also been shown to promote Pcsk1 expression. Humans and mice deficient in PC1 display hyperphagic obesity, hypogonadism, decreased GH, and hypoinsulinemic diabetes due to impaired prohormone processing. Here, we found that Snord116p-/m+ mice displayed in vivo functional defects in prohormone processing of proinsulin, pro-GH-releasing hormone, and proghrelin in association with reductions in islet, hypothalamic, and stomach PC1 content. Our findings suggest that the major neuroendocrine features of PWS are due to PC1 deficiency.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento/metabolismo , Neuronas/metabolismo , Síndrome de Prader-Willi/metabolismo , Proinsulina/metabolismo , Proproteína Convertasa 1/deficiencia , Precursores de Proteínas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Femenino , Hormona Liberadora de Hormona del Crecimiento/genética , Humanos , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patología , Hipogonadismo/genética , Hipogonadismo/metabolismo , Hipogonadismo/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones Noqueados , Neuronas/patología , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/patología , Proinsulina/genética , Precursores de Proteínas/genética , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo
5.
J Clin Invest ; 126(5): 1897-910, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-27064284

RESUMEN

Noncoding polymorphisms in the fat mass and obesity-associated (FTO) gene represent common alleles that are strongly associated with effects on food intake and adiposity in humans. Previous studies have suggested that the obesity-risk allele rs8050136 in the first intron of FTO alters a regulatory element recognized by the transcription factor CUX1, thereby leading to decreased expression of FTO and retinitis pigmentosa GTPase regulator-interacting protein-1 like (RPGRIP1L). Here, we evaluated the effects of rs8050136 and another potential CUX1 element in rs1421085 on expression of nearby genes in human induced pluripotent stem cell-derived (iPSC-derived) neurons. There were allele-dosage effects on FTO, RPGRIP1L, and AKT-interacting protein (AKTIP) expression, but expression of other vicinal genes, including IRX3, IRX5, and RBL2, which have been implicated in mediating functional effects, was not altered. In vivo manipulation of CUX1, Fto, and/or Rpgrip1l expression in mice affected adiposity in a manner that was consistent with CUX1 influence on adiposity via remote effects on Fto and Rpgrip1l expression. In support of a mechanism, mice hypomorphic for Rpgrip1l exhibited hyperphagic obesity, as the result of diminished leptin sensitivity in Leprb-expressing neurons. Together, the results of this study indicate that the effects of FTO-associated SNPs on energy homeostasis are due in part to the effects of these genetic variations on hypothalamic FTO, RPGRIP1L, and possibly other genes.


Asunto(s)
Alelos , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Regulación de la Expresión Génica , Intrones , Obesidad , Polimorfismo de Nucleótido Simple , Proteínas , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Proteínas del Citoesqueleto , Metabolismo Energético/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Ratones , Ratones Transgénicos , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Proteínas/genética , Proteínas/metabolismo , Proteína p130 Similar a la del Retinoblastoma/biosíntesis , Proteína p130 Similar a la del Retinoblastoma/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
6.
Obesity (Silver Spring) ; 17(1): 126-35, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18997673

RESUMEN

Perturbations in the functional integrity of the leptin axis are obvious candidates for mediation of altered adiposity. In a large number of genetic association studies in humans, the nonconservative LEPR Q223R allele has been inconsistently associated with adiposity. Subtle, long-term effects of such genetic variants can be obscured by effects of the environment and other confounders that render definitive inferences difficult to reach. We directly assessed the biological effects of this variant in 129P3/J mice segregating for the humanized Lepr allele at codon 223. No effects of this allele were detected on body weight, composition, or energy expenditure in animals fed diets of varying fat content over periods as long as 235 days. In vitro, Q223R did not affect leptin signaling as reflected by activation of STAT3. We conclude that Q223R is unlikely to play a significant role in regulation of human adiposity. This approach to vetting of human allelic variation might be more widely used.


Asunto(s)
Tejido Adiposo/fisiología , Composición Corporal/genética , Obesidad/genética , Receptores de Leptina/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Cartilla de ADN , Dieta con Restricción de Grasas , Células Madre Embrionarias/fisiología , Exones , Femenino , Regulación de la Expresión Génica , Variación Genética , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda