Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Metab ; 33(6): 1155-1170.e10, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33951475

RESUMO

Pathologies of the micro- and macrovascular systems are a hallmark of the metabolic syndrome, which can lead to chronically elevated blood pressure. However, the underlying pathomechanisms involved still need to be clarified. Here, we report that an obesity-associated increase in serum leptin triggers the select expansion of the micro-angioarchitecture in pre-autonomic brain centers that regulate hemodynamic homeostasis. By using a series of cell- and region-specific loss- and gain-of-function models, we show that this pathophysiological process depends on hypothalamic astroglial hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling downstream of leptin signaling. Importantly, several distinct models of HIF1α-VEGF pathway disruption in astrocytes are protected not only from obesity-induced hypothalamic angiopathy but also from sympathetic hyperactivity or arterial hypertension. These results suggest that hyperleptinemia promotes obesity-induced hypertension via a HIF1α-VEGF signaling cascade in hypothalamic astrocytes while establishing a novel mechanistic link that connects hypothalamic micro-angioarchitecture with control over systemic blood pressure.


Assuntos
Astrócitos/metabolismo , Hipertensão/metabolismo , Hipotálamo/metabolismo , Leptina/fisiologia , Obesidade/metabolismo , Animais , Astrócitos/patologia , Feminino , Hipotálamo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
J Clin Invest ; 131(8)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33630762

RESUMO

Bardet-Biedl syndrome (BBS) is a rare autosomal recessive disorder caused by mutations in genes encoding components of the primary cilium and is characterized by hyperphagic obesity. To investigate the molecular basis of obesity in human BBS, we developed a cellular model of BBS using induced pluripotent stem cell-derived (iPSC-derived) hypothalamic arcuate-like neurons. BBS mutations BBS1M390R and BBS10C91fsX95 did not affect neuronal differentiation efficiency but caused morphological defects, including impaired neurite outgrowth and longer primary cilia. Single-cell RNA sequencing of BBS1M390R hypothalamic neurons identified several downregulated pathways, including insulin and cAMP signaling and axon guidance. Additional studies demonstrated that BBS1M390R and BBS10C91fsX95 mutations impaired insulin signaling in both human fibroblasts and iPSC-derived neurons. Overexpression of intact BBS10 fully restored insulin signaling by restoring insulin receptor tyrosine phosphorylation in BBS10C91fsX95 neurons. Moreover, mutations in BBS1 and BBS10 impaired leptin-mediated p-STAT3 activation in iPSC-derived hypothalamic neurons. Correction of the BBS mutation by CRISPR rescued leptin signaling. POMC expression and neuropeptide production were decreased in BBS1M390R and BBS10C91fsX95 iPSC-derived hypothalamic neurons. In the aggregate, these data provide insights into the anatomic and functional mechanisms by which components of the BBSome in CNS primary cilia mediate effects on energy homeostasis.


Assuntos
Síndrome de Bardet-Biedl/metabolismo , Chaperoninas/metabolismo , Hipotálamo/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação de Sentido Incorreto , Neurônios/metabolismo , Sistemas do Segundo Mensageiro , Substituição de Aminoácidos , Animais , Síndrome de Bardet-Biedl/genética , Chaperoninas/genética , AMP Cíclico/genética , AMP Cíclico/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética
3.
J Clin Invest ; 126(5): 1897-910, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27064284

RESUMO

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.


Assuntos
Alelos , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Regulação da Expressão Gênica , Íntrons , Obesidade , Polimorfismo de Nucleotídeo Único , Proteínas , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Proteínas do Citoesqueleto , Metabolismo Energético/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Camundongos , Camundongos Transgênicos , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Proteínas/genética , Proteínas/metabolismo , Proteína p130 Retinoblastoma-Like/biossíntese , Proteína p130 Retinoblastoma-Like/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
4.
J Clin Invest ; 125(2): 796-808, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25555215

RESUMO

The hypothalamus is the central regulator of systemic energy homeostasis, and its dysfunction can result in extreme body weight alterations. Insights into the complex cellular physiology of this region are critical to the understanding of obesity pathogenesis; however, human hypothalamic cells are largely inaccessible for direct study. Here, we developed a protocol for efficient generation of hypothalamic neurons from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) obtained from patients with monogenetic forms of obesity. Combined early activation of sonic hedgehog signaling followed by timed NOTCH inhibition in human ESCs/iPSCs resulted in efficient conversion into hypothalamic NKX2.1+ precursors. Application of a NOTCH inhibitor and brain-derived neurotrophic factor (BDNF) further directed the cells into arcuate nucleus hypothalamic-like neurons that express hypothalamic neuron markers proopiomelanocortin (POMC), neuropeptide Y (NPY), agouti-related peptide (AGRP), somatostatin, and dopamine. These hypothalamic-like neurons accounted for over 90% of differentiated cells and exhibited transcriptional profiles defined by a hypothalamic-specific gene expression signature that lacked pituitary markers. Importantly, these cells displayed hypothalamic neuron characteristics, including production and secretion of neuropeptides and increased p-AKT and p-STAT3 in response to insulin and leptin. Our results suggest that these hypothalamic-like neurons have potential for further investigation of the neurophysiology of body weight regulation and evaluation of therapeutic targets for obesity.


Assuntos
Diferenciação Celular , Hipotálamo/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios , Obesidade/metabolismo , Antígenos de Diferenciação/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Proteínas Hedgehog/metabolismo , Humanos , Hipotálamo/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Proteínas Nucleares/metabolismo , Obesidade/patologia , Pró-Opiomelanocortina/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/metabolismo
5.
Cell Metab ; 19(5): 767-79, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24807221

RESUMO

Common polymorphisms in the first intron of FTO are associated with increased body weight in adults. Previous studies have suggested that a CUX1-regulatory element within the implicated FTO region controls expression of FTO and the nearby ciliary gene, RPGRIP1L. Given the role of ciliary genes in energy homeostasis, we hypothesized that mice hypomorphic for Rpgrip1l would display increased adiposity. We find that Rpgrip1l⁺/⁻ mice are hyperphagic and fatter, and display diminished suppression of food intake in response to leptin administration. In the hypothalamus of Rpgrip1l⁺/⁻ mice, and in human fibroblasts with hypomorphic mutations in RPGRIP1L, the number of AcIII-positive cilia is diminished, accompanied by impaired convening of the leptin receptor to the vicinity of the cilium, and diminished pStat3 in response to leptin. These findings suggest that RPGRIP1L may be partly or exclusively responsible for the obesity susceptibility signal at the FTO locus.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Adiposidade/genética , Cílios/genética , Polimorfismo Genético/genética , Proteínas/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Células Cultivadas , Ingestão de Alimentos/genética , Feminino , Fibroblastos/metabolismo , Humanos , Hipotálamo , Íntrons , Leptina/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores para Leptina/genética , Fator de Transcrição STAT3/genética , Aumento de Peso/genética
6.
J Biol Chem ; 286(3): 2155-70, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21037323

RESUMO

The first intron of FTO contains common single nucleotide polymorphisms associated with body weight and adiposity in humans. In an effort to identify the molecular basis for this association, we discovered that FTO and RPGRIP1L (a ciliary gene located in close proximity to the transcriptional start site of FTO) are regulated by isoforms P200 and P110 of the transcription factor, CUX1. This regulation occurs via a single AATAAATA regulatory site (conserved in the mouse) within the FTO intronic region associated with adiposity in humans. Single nucleotide polymorphism rs8050136 (located in this regulatory site) affects binding affinities of P200 and P110. Promoter-probe analysis revealed that binding of P200 to this site represses FTO, whereas binding of P110 increases transcriptional activity from the FTO as well as RPGRIP1L minimal promoters. Reduced expression of Fto or Rpgrip1l affects leptin receptor isoform b trafficking and leptin signaling in N41 mouse hypothalamic or N2a neuroblastoma cells in vitro. Leptin receptor clusters in the vicinity of the cilium of arcuate hypothalamic neurons in C57BL/6J mice treated with leptin, but not in fasted mice, suggesting a potentially important role of the cilium in leptin signaling that is, in part, regulated by FTO and RPGRIP1L. Decreased Fto/Rpgrip1l expression in the arcuate hypothalamus coincides with decreased nuclear enzymatic activity of a protease (cathepsin L) that has been shown to cleave full-length CUX1 (P200) to P110. P200 disrupts (whereas P110 promotes) leptin receptor isoform b clustering in the vicinity of the cilium in vitro. Clustering of the receptor coincides with increased leptin signaling as reflected in protein levels of phosphorylated Stat3 (p-Stat3). Association of the FTO locus with adiposity in humans may reflect functional consequences of A/C alleles at rs8050136. The obesity-risk (A) allele shows reduced affinity for the FTO and RPGRIP1L transcriptional activator P110, leading to the following: 1) decreased FTO and RPGRIP1L mRNA levels; 2) reduced LEPR trafficking to the cilium; and, as a consequence, 3) a diminished cellular response to leptin.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Hipotálamo/metabolismo , Proteínas Nucleares/metabolismo , Receptores para Leptina/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Tecido Adiposo/metabolismo , Adiposidade/genética , Alelos , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Catepsina L/genética , Catepsina L/metabolismo , Linhagem Celular Tumoral , Proteínas de Homeodomínio/genética , Humanos , Íntrons/genética , Camundongos , Camundongos Mutantes , Oxigenases de Função Mista , Proteínas Nucleares/genética , Obesidade/genética , Obesidade/metabolismo , Oxo-Ácido-Liases/genética , Oxo-Ácido-Liases/metabolismo , Fosforilação/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores para Leptina/genética , Proteínas Repressoras/genética , Elementos de Resposta , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Fator de Transcrição STAT3/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 294(4): R1185-96, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18256137

RESUMO

Two recent, large whole-genome association studies (GWAS) in European populations have associated a approximately 47-kb region that contains part of the FTO gene with high body mass index (BMI). The functions of FTO and adjacent FTM in human biology are not clear. We examined expression of these genes in organs of mice segregating for monogenic obesity mutations, exposed to underfeeding/overfeeding, and to 4 degrees C. Fto/Ftm expression was reduced in mesenteric adipose tissue of mice segregating for the Ay, Lep ob, Lepr db, Cpe fat, or tub mutations, and there was a similar trend in other tissues. These effects were not due to adiposity per se. Hypothalamic Fto and Ftm expression were decreased by fasting in lean and obese animals and by cold exposure in lean mice. The fact that responses of Fto and Ftm expression to these manipulations were almost indistinguishable suggested that the genes might be coregulated. The putative overlapping regulatory region contains at least two canonical CUTL1 binding sites. One of these nominal CUTL1 sites includes rs8050136, a SNP associated with high body mass. The A allele of rs8050136 preferentially bound CUTL1[corrected] in human fibroblast DNA. 70% knockdown of CUTL1 expression in human fibroblasts decreased FTO and FTM expression by 90 and 65%, respectively. Animals and humans with various genetic interruptions of FTO or FTM have phenotypes reminiscent of aspects of the Bardet-Biedl obesity syndrome, a confirmed "ciliopathy." FTM has recently been shown to be a ciliary basal body protein.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Adiposidade/genética , Regulação da Expressão Gênica , Obesidade/genética , Oxo-Ácido-Liases/genética , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Síndrome de Bardet-Biedl/genética , Síndrome de Bardet-Biedl/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Ingestão de Alimentos , Embrião de Mamíferos/metabolismo , Metabolismo Energético/genética , Jejum/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotermia Induzida , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Oxigenases de Função Mista , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Obesidade/metabolismo , Oxo-Ácido-Liases/metabolismo , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Células Estromais/metabolismo , Fatores de Transcrição , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA