Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
eNeuro ; 8(5)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34535504

RESUMEN

The hedgehog signaling pathway is best known for its role in developmental patterning of the neural tube and limb bud. More recently, hedgehog signaling has been recognized for its roles in growth of adult tissues and maintenance of progenitor cell niches. However, the role of hedgehog signaling in fully differentiated cells like neurons in the adult brain is less clear. In mammals, coordination of hedgehog pathway activity relies on primary cilia and patients with ciliopathies such as Bardet-Biedl and Alström syndrome exhibit clinical features clearly attributable to errant hedgehog such as polydactyly. However, these ciliopathies also present with features not clearly associated with hedgehog signaling such as hyperphagia-associated obesity. How hedgehog signaling may contribute to feeding behavior is complex and unclear, but cilia are critical for proper energy homeostasis. Here, we provide a detailed analysis of the expression of core components of the hedgehog signaling pathway in the adult mouse hypothalamus with an emphasis on feeding centers. We show that hedgehog pathway genes continue to be expressed in differentiated neurons important for the regulation of feeding behavior. Furthermore, we demonstrate for the first time that pathway activity is regulated at the transcriptional level by fasting. These data suggest that hedgehog signaling is involved in the proper functioning of brain regions that regulate feeding behavior and that hedgehog pathway dysfunction may play a role in the obesity observed in certain ciliopathies.


Asunto(s)
Ayuno , Proteínas Hedgehog , Animales , Cilios/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Hipotálamo/metabolismo , Ratones , Transducción de Señal
2.
Semin Cell Dev Biol ; 110: 43-50, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32466971

RESUMEN

An emerging number of rare genetic disorders termed ciliopathies are associated with pediatric obesity. It is becoming clear that the mechanisms associated with cilia dysfunction and obesity in these syndromes are complex. In addition to ciliopathic syndromic forms of obesity, several cilia-associated signaling gene mutations also lead to morbid obesity. While cilia have critical and diverse functions in energy homeostasis including their roles in centrally mediated food intake as well as in peripheral tissues, many questions remain. Here, we briefly discuss the syndromic ciliopathies and monoallelic cilia signaling gene mutations associated with obesity. We also describe potential ways cilia may be involved in common obesity. We discuss how neuronal cilia impact food intake potentially through leptin signaling and changes in ciliary G protein-coupled receptor (GPCR) signaling. We highlight several recent studies that have implicated the potential for cilia in peripheral tissues such as adipose and the pancreas to contribute to metabolic dysfunction. Then we discuss the potential for cilia to impact energy homeostasis through their roles in both development and adult tissue homeostasis. The studies discussed in this review highlight how a comprehensive understanding of the requirement of cilia for the regulation of diverse biological functions will contribute to our understanding of common forms of obesity.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Cilios/metabolismo , Ciliopatías/genética , Leptina/genética , Obesidad Mórbida/genética , Obesidad Infantil/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Adulto , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Niño , Cilios/patología , Ciliopatías/metabolismo , Ciliopatías/patología , Ingestión de Alimentos/genética , Regulación de la Expresión Génica , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Leptina/metabolismo , Neuronas/metabolismo , Neuronas/patología , Obesidad Mórbida/metabolismo , Obesidad Mórbida/patología , Páncreas/metabolismo , Páncreas/patología , Obesidad Infantil/metabolismo , Obesidad Infantil/patología , Transducción de Señal
3.
Mol Metab ; 30: 131-139, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31767164

RESUMEN

OBJECTIVE: Fibroblast growth factor 19 (FGF19) is a postprandial hormone which plays diverse roles in the regulation of bile acid, glucose, and lipid metabolism. Administration of FGF19 to obese/diabetic mice lowers body weight, improves insulin sensitivity, and enhances glycemic control. The primary target organ of FGF19 is the liver, where it regulates bile acid homeostasis in response to nutrient absorption. In contrast, the broader pharmacologic actions of FGF19 are proposed to be driven, in part, by the recruitment of the thermogenic protein uncoupling protein 1 (UCP1) in white and brown adipose tissue. However, the precise contribution of UCP1-dependent thermogenesis to the therapeutic actions of FGF19 has not been critically evaluated. METHODS: Using WT and germline UCP1 knockout mice, the primary objective of the current investigation was to determine the in vivo pharmacology of FGF19, focusing on its thermogenic and anti-obesity activity. RESULTS: We report that FGF19 induced mRNA expression of UCP1 in adipose tissue and show that this effect is required for FGF19 to increase caloric expenditure. However, we demonstrate that neither UCP1 induction nor an elevation in caloric expenditure are necessary for FGF19 to induce weight loss in obese mice. In contrast, the anti-obesity action of FGF19 appeared to be associated with its known physiological role. In mice treated with FGF19, there was a significant reduction in the mRNA expression of genes associated with hepatic bile acid synthesis enzymes, lowered levels of hepatic bile acid species, and a significant increase in fecal energy content, all indicative of reduced lipid absorption in animals treated with FGF19. CONCLUSION: Taken together, we report that the anti-obesity effect of FGF19 occurs in the absence of UCP1. Our data suggest that the primary way in which exogenous FGF19 lowers body weight in mice may be through the inhibition of bile acid synthesis and subsequently a reduction of dietary lipid absorption.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Proteína Desacopladora 1/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Peso Corporal , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa , Metabolismo Energético , Factores de Crecimiento de Fibroblastos/genética , Resistencia a la Insulina , Metabolismo de los Lípidos , Lipogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Termogénesis , Proteína Desacopladora 1/genética
4.
Front Cell Neurosci ; 13: 266, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31249512

RESUMEN

Primary cilia dysfunction has been associated with hyperphagia and obesity in both ciliopathy patients and mouse models of cilia perturbation. Neurons throughout the brain possess these solitary cellular appendages, including in the feeding centers of the hypothalamus. Several cell biology questions associated with primary neuronal cilia signaling are challenging to address in vivo. Here we utilize primary hypothalamic neuronal cultures to study ciliary signaling in relevant cell types. Importantly, these cultures contain neuronal populations critical for appetite and satiety such as pro-opiomelanocortin (POMC) and agouti related peptide (AgRP) expressing neurons and are thus useful for studying signaling involved in feeding behavior. Correspondingly, these cultured neurons also display electrophysiological activity and respond to both local and peripheral signals that act on the hypothalamus to influence feeding behaviors, such as leptin and melanin concentrating hormone (MCH). Interestingly, we found that cilia mediated hedgehog signaling, generally associated with developmental processes, can influence ciliary GPCR signaling (Mchr1) in terminally differentiated neurons. Specifically, pharmacological activation of the hedgehog-signaling pathway using the smoothened agonist, SAG, attenuated the ability of neurons to respond to ligands (MCH) of ciliary GPCRs. Understanding how the hedgehog pathway influences cilia GPCR signaling in terminally differentiated neurons could reveal the molecular mechanisms associated with clinical features of ciliopathies, such as hyperphagia-associated obesity.

5.
FASEB J ; 33(1): 1440-1455, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30133325

RESUMEN

The transition zone (TZ) is a domain at the base of the cilium that is involved in maintaining ciliary compartment-specific sensory and signaling activity by regulating cilia protein composition. Mutations in TZ proteins result in cilia dysfunction, often causing pleiotropic effects observed in a group of human diseases classified as ciliopathies. The purpose of this study is to describe the importance of the TZ component Meckel-Grüber syndrome 6 ( Mks6) in several organ systems and tissues regarding ciliogenesis and cilia maintenance using congenital and conditional mutant mouse models. Similar to MKS, congenital loss of Mks6 is embryonic lethal, displaying cilia loss and altered cytoskeletal microtubule modifications but only in specific cell types. Conditional Mks6 mutants have a variable cystic kidney phenotype along with severe retinal degeneration with mislocalization of phototransduction cascade proteins. However, other phenotypes, such as anosmia and obesity, which are typically associated with cilia and TZ dysfunction, were not evident. These data indicate that despite Mks6 being a core TZ component, it has tissue- or cell type-specific functions important for cilia formation and cilia sensory and signaling activities. Lewis, W. R., Bales, K. L., Revell, D. Z., Croyle, M. J., Engle, S. E., Song, C. J., Malarkey, E. B., Uytingco, C. R., Shan, D., Antonellis, P. J., Nagy, T. R., Kesterson, R. A., Mrug, M. M., Martens, J. R., Berbari, N. F., Gross, A. K., Yoder, B. K. Mks6 mutations reveal tissue- and cell type-specific roles for the cilia transition zone.


Asunto(s)
Cilios/metabolismo , Proteínas del Citoesqueleto/genética , Mutación , Acetilación , Animales , Trastornos de la Motilidad Ciliar/genética , Citoplasma/metabolismo , Encefalocele/genética , Femenino , Genes Letales , Enfermedades Renales Quísticas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Trastornos del Olfato/genética , Fenotipo , Enfermedades Renales Poliquísticas/genética , Degeneración Retiniana/genética , Retinitis Pigmentosa/genética , Tubulina (Proteína)/metabolismo , Aumento de Peso/genética
6.
Genesis ; 56(8): e23217, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29806135

RESUMEN

The neuropeptide, melanin concentrating hormone (MCH), and its G protein-coupled receptor, melanin concentrating hormone receptor 1 (Mchr1), are expressed centrally in adult rodents. MCH signaling has been implicated in diverse behaviors such as feeding, sleep, anxiety, as well as addiction and reward. While a model utilizing the Mchr1 promoter to drive constitutive expression of Cre recombinase (Mchr1-Cre) exists, there is a need for an inducible Mchr1-Cre to determine the roles for this signaling pathway in neural development and adult neuronal function. Here, we generated a BAC transgenic mouse where the Mchr1 promotor drives expression of tamoxifen inducible CreER recombinase. Many aspects of the Mchr1-Cre expression pattern are recapitulated by the Mchr1-CreER model, though there are also notable differences. Most strikingly, compared to the constitutive model, the new Mchr1-CreER model shows strong expression in adult animals in hypothalamic brain regions involved in feeding behavior but diminished expression in regions involved in reward, such as the nucleus accumbens. The inducible Mchr1-CreER allele will help reveal the potential for Mchr1 signaling to impact neural development and subsequent behavioral phenotypes, as well as contribute to the understanding of the MCH signaling pathway in terminally differentiated adult neurons and the diverse behaviors that it influences.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Receptores de Somatostatina/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Integrasas , Melaninas/metabolismo , Ratones , Ratones Transgénicos , Modelos Animales , Neuronas/metabolismo , Neuropéptidos/metabolismo , Hormonas Hipofisarias/metabolismo , Receptores de Somatostatina/metabolismo , Transducción de Señal , Tamoxifeno
7.
J Neurosci ; 34(1): 305-12, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24381291

RESUMEN

The precise morphology of the mechanosensitive hair bundle requires seamless integration of actin and microtubule networks. Here, we identify Acf7a (actin crosslinking family protein 7a) as a protein positioned to bridge these distinct cytoskeletal networks in hair cells. By imaging Acf7a-Citrine fusion protein in zebrafish and immunolabeling of vestibular and cochlear mouse hair cells, we show that Acf7a and ACF7 circumscribe, underlie, and are interwoven into the cuticular plate (CP), and they also encircle the basal body of the kinocilium. In cochlear hair cells, ACF7 localization is graded, with the highest concentration near each fonticulus--an area free of F-actin in the region of the CP that contains the basal body. During hair-cell development and regeneration, Acf7a precedes formation of the hair bundle and CP. Finally, electron tomography demonstrates that the ends of microtubules insert into the CP and are decorated with filamentous linkers connecting microtubules to the CP. These observations are consistent with ACF7 being a linker protein, which may shape the cytoskeleton of the hair cell early during hair-bundle genesis.


Asunto(s)
Actinas/análisis , Células Ciliadas Auditivas/química , Proteínas de Microfilamentos/análisis , Tubulina (Proteína)/análisis , Máculas Acústicas , Actinas/ultraestructura , Animales , Animales Modificados Genéticamente , Pollos , Citoesqueleto/química , Citoesqueleto/ultraestructura , Femenino , Células Ciliadas Auditivas/ultraestructura , Masculino , Ratones , Proteínas de Microfilamentos/ultraestructura , Especificidad de la Especie , Tubulina (Proteína)/ultraestructura , Pez Cebra
8.
Am J Hum Genet ; 93(1): 132-40, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23768514

RESUMEN

Previously, DFNB89, a locus associated with autosomal-recessive nonsyndromic hearing impairment (ARNSHI), was mapped to chromosomal region 16q21-q23.2 in three unrelated, consanguineous Pakistani families. Through whole-exome sequencing of a hearing-impaired individual from each family, missense mutations were identified at highly conserved residues of lysyl-tRNA synthetase (KARS): the c.1129G>A (p.Asp377Asn) variant was found in one family, and the c.517T>C (p.Tyr173His) variant was found in the other two families. Both variants were predicted to be damaging by multiple bioinformatics tools. The two variants both segregated with the nonsyndromic-hearing-impairment phenotype within the three families, and neither mutation was identified in ethnically matched controls or within variant databases. Individuals homozygous for KARS mutations had symmetric, severe hearing impairment across all frequencies but did not show evidence of auditory or limb neuropathy. It has been demonstrated that KARS is expressed in hair cells of zebrafish, chickens, and mice. Moreover, KARS has strong localization to the spiral ligament region of the cochlea, as well as to Deiters' cells, the sulcus epithelium, the basilar membrane, and the surface of the spiral limbus. It is hypothesized that KARS variants affect aminoacylation in inner-ear cells by interfering with binding activity to tRNA or p38 and with tetramer formation. The identification of rare KARS variants in ARNSHI-affected families defines a gene that is associated with ARNSHI.


Asunto(s)
Pérdida Auditiva/enzimología , Pérdida Auditiva/genética , Lisina-ARNt Ligasa/genética , Mutación Missense , Animales , Estudios de Casos y Controles , Pollos , Cóclea/metabolismo , Cóclea/patología , Biología Computacional/métodos , Consanguinidad , Femenino , Ligamiento Genético , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Haplotipos , Homocigoto , Humanos , Lisina-ARNt Ligasa/metabolismo , Masculino , Ratones , Linaje , Aminoacilación de ARN de Transferencia , Pez Cebra/embriología , Pez Cebra/metabolismo
9.
Endocrinology ; 152(8): 2996-3004, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21712364

RESUMEN

Fibroblast growth factor (FGF21) plays an important role in regulating hepatic oxidation of fatty acids and gluconeogenesis in response to fasting and during consumption of a ketogenic diet. However, the metabolic pathways through which FGF21 regulates hepatic function are not well defined. To identify the effects of FGF21 on the liver in vivo, we administered FGF21 to mice and analyzed acute effects on signaling and gene expression. We found that FGF21 acts directly on the liver to stimulate phosphorylation of fibroblast growth factor receptor substrate 2 and ERK1/2. Acute FGF21 treatment induced hepatic expression of key regulators of gluconeogenesis, lipid metabolism, and ketogenesis including glucose-6-phosphatase, phosphoenol pyruvate carboxykinase, 3-hydroxybutyrate dehydrogenase type 1, and carnitine palmitoyltransferase 1α. In addition, injection of FGF21 was associated with decreased circulating insulin and free fatty acid levels. FGF21 treatment induced mRNA and protein expression of peroxisome proliferator-activated receptor-γ coactivator (PGC-1α), suggesting that PGC-1α may play a role in regulating FGF21 action. However, studies using mice with liver-specific ablation of PGC-1α revealed the same regulation of gluconeogenic gene expression by FGF21 as seen in wild-type mice, indicating that PGC-1α is not necessary for the effect of FGF21 on glucose metabolism. These data demonstrate that FGF21 acts directly on the liver to modulate hepatic metabolism. The direct effects we examined are not dependent on PGC-1α. In addition, FGF21 treatment is associated with decreased serum insulin levels that my affect hepatic function.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Hígado/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/fisiología , Gluconeogénesis , Humanos , Proteínas Klotho , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , PPAR alfa/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transactivadores/genética , Transactivadores/fisiología , Factores de Transcripción
10.
Diabetes ; 59(11): 2781-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20682689

RESUMEN

OBJECTIVE: Fibroblast growth factor 21 (FGF21) is a key mediator of fatty acid oxidation and lipid metabolism. Pharmacological doses of FGF21 improve glucose tolerance, lower serum free fatty acids, and lead to weight loss in obese mice. Surprisingly, however, FGF21 levels are elevated in obese ob/ob and db/db mice and correlate positively with BMI in humans. However, the expected beneficial effects of endogenous FGF21 to increase glucose tolerance and reduce circulating triglycerides are absent in obesity. RESEARCH DESIGN AND METHODS: To test the hypothesis that obesity is a state of FGF21 resistance, we evaluated the response of obese mice to exogenous FGF21 administration. In doing this, we assessed the impact of diet-induced obesity on FGF21 signaling and resultant transcriptional events in the liver and white adipose tissue. We also analyzed the physiologic impact of FGF21 resistance by assessing serum parameters that are acutely regulated by FGF21. RESULTS: When obese mice are treated with FGF21, they display both a significantly attenuated signaling response as assessed by extracellular mitogen-activated protein kinase 1 and 2 (ERK1/2) phosphorylation as well as an impaired induction of FGF21 target genes, including cFos and EGR1. These effects were seen in both liver and fat. Similarly, changes in serum parameters such as the decline in glucose and free fatty acids are attenuated in FGF21-treated DIO mice. CONCLUSIONS: These data demonstrate that DIO mice have increased endogenous levels of FGF21 and respond poorly to exogenous FGF21. We therefore propose that obesity is an FGF21-resistant state.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Obesidad/sangre , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/fisiología , Animales , Resistencia a Medicamentos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Inyecciones Intravenosas , Hígado/efectos de los fármacos , Hígado/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , ARN Mensajero/sangre , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...