Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell Metab ; 35(3): 438-455.e7, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36889283

RESUMEN

Until menopause, women have a lower propensity to develop metabolic diseases than men, suggestive of a protective role for sex hormones. Although a functional synergy between central actions of estrogens and leptin has been demonstrated to protect against metabolic disturbances, the underlying cellular and molecular mechanisms mediating this crosstalk have remained elusive. By using a series of embryonic, adult-onset, and tissue/cell-specific loss-of-function mouse models, we document an unprecedented role of hypothalamic Cbp/P300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 1 (Cited1) in mediating estradiol (E2)-dependent leptin actions that control feeding specifically in pro-opiomelanocortin (Pomc) neurons. We reveal that within arcuate Pomc neurons, Cited1 drives leptin's anorectic effects by acting as a co-factor converging E2 and leptin signaling via direct Cited1-ERα-Stat3 interactions. Together, these results provide new insights on how melanocortin neurons integrate endocrine inputs from gonadal and adipose axes via Cited1, thereby contributing to the sexual dimorphism in diet-induced obesity.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Leptina , Ratones , Animales , Femenino , Leptina/metabolismo , Estradiol/farmacología , Proopiomelanocortina/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo
2.
J Clin Invest ; 130(11): 6093-6108, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32780722

RESUMEN

Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 2/enzimología , Fosfatasas de Especificidad Dual/metabolismo , Hipotálamo/enzimología , Resistencia a la Insulina , MAP Quinasa Quinasa 4/metabolismo , Transducción de Señal , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Fosfatasas de Especificidad Dual/genética , MAP Quinasa Quinasa 4/genética , Ratones , Ratones Noqueados
3.
J Med Chem ; 61(24): 11144-11157, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30525586

RESUMEN

Celastrol is a natural pentacyclic triterpene used in traditional Chinese medicine with significant weight-lowering effects. Celastrol-administered mice at 100 µg/kg decrease food consumption and body weight via a leptin-dependent mechanism, yet its molecular targets in this pathway remain elusive. Here, we demonstrate in vivo that celastrol-induced weight loss is largely mediated by the inhibition of leptin negative regulators protein tyrosine phosphatase (PTP) 1B (PTP1B) and T-cell PTP (TCPTP) in the arcuate nucleus (ARC) of the hypothalamus. We show in vitro that celastrol binds reversibly and inhibits noncompetitively PTP1B and TCPTP. NMR data map the binding site to an allosteric site in the catalytic domain that is in proximity of the active site. By using a panel of PTPs implicated in hypothalamic leptin signaling, we show that celastrol additionally inhibited PTEN and SHP2 but had no activity toward other phosphatases of the PTP family. These results suggest that PTP1B and TCPTP in the ARC are essential for celastrol's weight lowering effects in adult obese mice.


Asunto(s)
Fármacos Antiobesidad/farmacología , Obesidad/tratamiento farmacológico , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Triterpenos/farmacología , Sitio Alostérico , Animales , Fármacos Antiobesidad/metabolismo , Dominio Catalítico , Dieta Alta en Grasa/efectos adversos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratones Transgénicos , Obesidad/etiología , Triterpenos Pentacíclicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/química , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Relación Estructura-Actividad , Triterpenos/química , Triterpenos/metabolismo , Pérdida de Peso/efectos de los fármacos
4.
Diabetes ; 67(11): 2456-2465, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30158241

RESUMEN

Celastrol, a plant-derived constituent of traditional Chinese medicine, has been proposed to offer significant potential as an antiobesity drug. However, the molecular mechanism for this activity is unknown. We show that the weight-lowering effects of celastrol are driven by decreased food consumption. Although young Lep ob mice respond with a decrease in food intake and body weight, adult Lep db and Lep ob mice are unresponsive to celastrol, suggesting that functional leptin signaling in adult mice is required to elicit celastrol's catabolic actions. Protein tyrosine phosphatase 1 (PTP1B), a leptin negative-feedback regulator, has been previously reported to be one of celastrol's targets. However, we found that global PTP1B knockout (KO) and wild-type (WT) mice have comparable weight loss and hypophagia when treated with celastrol. Increased levels of uncoupling protein 1 (UCP1) in subcutaneous white and brown adipose tissue suggest celastrol-induced thermogenesis as a further mechanism. However, diet-induced obese UCP1 WT and KO mice have comparable weight loss upon celastrol treatment, and celastrol treatment has no effect on energy expenditure under ambient housing or thermoneutral conditions. Overall, our results suggest that celastrol-induced weight loss is hypophagia driven and age-dependently mediated by functional leptin signaling. Our data encourage reconsideration of therapeutic antiobesity strategies built on leptin sensitization.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Obesidad/metabolismo , Extractos Vegetales/farmacología , Triterpenos/farmacología , Proteína Desacopladora 1/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Ratones Noqueados , Obesidad/genética , Triterpenos Pentacíclicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Desacopladora 1/genética
5.
J Neuroinflammation ; 15(1): 35, 2018 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-29422055

RESUMEN

ᅟ: Astrocytosis is a reactive process involving cellular, molecular, and functional changes to facilitate neuronal survival, myelin preservation, blood brain barrier function and protective glial scar formation upon brain insult. The overall pro- or anti-inflammatory impact of reactive astrocytes appears to be driven in a context- and disease-driven manner by modulation of astrocytic Ca2+ homeostasis and activation of Ca2+/calmodulin-activated serine/threonine phosphatase calcineurin. Here, we aimed to assess whether calcineurin is dispensable for astrocytosis in the hypothalamus driven by prolonged high fat diet (HFD) feeding. Global deletion of calcineurin A beta (gene name: Ppp3cb) led to a decrease of glial fibrillary acidic protein (GFAP)-positive cells in the ventromedial hypothalamus (VMH), dorsomedial hypothalamus (DMH), and arcuate nucleus (ARC) of mice exposed chronically to HFD. The concomitant decrease in Iba1-positive microglia in the VMH further suggests a modest impact of Ppp3cb deletion on microgliosis. Pharmacological inhibition of calcineurin activity by Fk506 had no impact on IBA1-positive microglia in hypothalami of mice acutely exposed to HFD for 1 week. However, Fk506-treated mice displayed a decrease in GFAP levels in the ARC. In vivo effects could not be replicated in cell culture, where calcineurin inhibition by Fk506 had no effect on astrocytic morphology, astrocytic cell death, GFAP, and vimentin protein levels or microglia numbers in primary hypothalamic astrocytes and microglia co-cultures. Further, adenoviral overexpression of calcineurin subunit Ppp3r1 in primary glia culture did not lead to an increase in GFAP fluorescence intensity. Overall, our results point to a prominent role of calcineurin in mediating hypothalamic astrocytosis as response to acute and chronic HFD exposure. Moreover, discrepant findings in vivo and in cell culture indicate the necessity of studying astrocytes in their "natural" environment, i.e., preserving an intact hypothalamic microenvironment with neurons and non-neuronal cells in close proximity.


Asunto(s)
Calcineurina/deficiencia , Dieta Alta en Grasa/efectos adversos , Gliosis/metabolismo , Gliosis/prevención & control , Hipotálamo/metabolismo , Animales , Astrocitos/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Gliosis/patología , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Biomed Chromatogr ; 32(4)2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29166705

RESUMEN

Monoamines, acting as hormones and neurotransmitters, play a critical role in multiple physiological processes ranging from cognitive function and mood to sympathetic nervous system activity, fight-or-flight response and glucose homeostasis. In addition to brain and blood, monoamines are abundant in several tissues, and dysfunction in their synthesis or signaling is associated with various pathological conditions. It was our goal to develop a method to detect these compounds in peripheral murine tissues. In this study, we employed a high-performance liquid chromatography method using electrochemical detection that allows not only detection of catecholamines but also a detailed analysis of nine monoamines and metabolites in murine tissues. Simple tissue extraction procedures were optimized for muscle (gastrocnemius, extensor digitorum longus and soleus), liver, pancreas and white adipose tissue in the range of weight 10-200 mg. The system allowed a limit of detection between 0.625 and 2.5 pg µL-1 for monoamine analytes and their metabolites, including dopamine, 3,4-dihydroxyphenylacetic acid, 3-methoxytyramine, homovanillic acid, norepinephrine, epinephrine, 3-methoxy-4-hydroxyphenylglycol, serotonin and 5-hydroxyindoleacetic acid. Typical concentrations for different monoamines and their metabolization products in these tissues are presented for C57Bl/6 J mice fed a high-fat diet.


Asunto(s)
Monoaminas Biogénicas/análisis , Monoaminas Biogénicas/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Técnicas Electroquímicas/métodos , Tejido Adiposo Blanco/química , Animales , Monoaminas Biogénicas/química , Dieta Alta en Grasa , Sistema Digestivo/química , Hipotálamo/química , Límite de Detección , Modelos Lineales , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/química , Especificidad de Órganos , Reproducibilidad de los Resultados
7.
Neuroscience ; 357: 241-254, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28627418

RESUMEN

Elevated levels of oxidative stress and neuronal inflammation in the hypothalamus or ventral midbrain, respectively, represent common denominators for obesity and Parkinson's Disease (PD). However, little is known about defense mechanisms that protect neurons in these regions from oxidative damage. Here, we aimed to assess whether murine Gpx4, a crucial antioxidant enzyme that protects neurons from membrane damage and ferroptosis, is critical for the protection from neuronal inflammation in two distinct pathophysiologic diseases, namely metabolic dysfunction in diet-induced obesity or PD. Gpx4 was deleted from either AgRP or POMC neurons in the hypothalamus, essential for metabolic homeostasis, or from dopaminergic neurons in the ventral midbrain, governing behaviors such as anxiety or voluntary movement. To induce a pro-inflammatory environment, AgRP and POMC neuron-specific Gpx4 knockout mice were subjected to high-fat high-sucrose (HFHS) diet. To exacerbate oxidative stress in dopaminergic neurons of the ventral midbrain, we systemically co-deleted the PD-related gene DJ-1. Gpx4 was dispensable for the maintenance of cellular health and function of POMC neurons, even in mice exposed to obesogenic conditions. In contrast, HFHS-fed mice with Gpx4 deletion from AgRP neurons displayed increased body adiposity. Gpx4 expression and activity were diminished in the hypothalamus of HFHS-fed mice compared to standard diet-fed controls. Gpx4 deletion from dopaminergic neurons induced anxiety behavior, and diminished spontaneous locomotor activity when DJ-1 was co-deleted. Overall, these data suggest a physiological role for Gpx4 in balancing metabolic control signals and inflammation in AgRP but not POMC neurons. Moreover, Gpx4 appears to constitute an important rheostat against neuronal dysfunction and PD-like symptoms in dopaminergic circuitry within the ventral midbrain.


Asunto(s)
Ansiedad/enzimología , Peso Corporal/fisiología , Glutatión Peroxidasa/deficiencia , Actividad Motora/fisiología , Obesidad/enzimología , Trastornos Parkinsonianos/enzimología , Adiposidad/fisiología , Animales , Ansiedad/inmunología , Ansiedad/patología , Conducta Animal/fisiología , Dieta Alta en Grasa , Sacarosa en la Dieta , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/patología , Femenino , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Hipotálamo/enzimología , Hipotálamo/inmunología , Hipotálamo/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/patología , Estrés Oxidativo/fisiología , Trastornos Parkinsonianos/inmunología , Trastornos Parkinsonianos/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo , Caracteres Sexuales , Glutatión Peroxidasa GPX1
8.
Nat Commun ; 7: 10782, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26923837

RESUMEN

Hypothalamic leptin signalling has a key role in food intake and energy-balance control and is often impaired in obese individuals. Here we identify histone deacetylase 5 (HDAC5) as a regulator of leptin signalling and organismal energy balance. Global HDAC5 KO mice have increased food intake and greater diet-induced obesity when fed high-fat diet. Pharmacological and genetic inhibition of HDAC5 activity in the mediobasal hypothalamus increases food intake and modulates pathways implicated in leptin signalling. We show HDAC5 directly regulates STAT3 localization and transcriptional activity via reciprocal STAT3 deacetylation at Lys685 and phosphorylation at Tyr705. In vivo, leptin sensitivity is substantially impaired in HDAC5 loss-of-function mice. Hypothalamic HDAC5 overexpression improves leptin action and partially protects against HFD-induced leptin resistance and obesity. Overall, our data suggest that hypothalamic HDAC5 activity is a regulator of leptin signalling that adapts food intake and body weight to our dietary environment.


Asunto(s)
Hipotálamo/metabolismo , Leptina/metabolismo , Animales , Glucemia , Línea Celular , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Prueba de Tolerancia a la Glucosa , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Infusiones Intraventriculares , Resistencia a la Insulina , Captura por Microdisección con Láser , Leptina/genética , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Endogámicos , Ratones Noqueados , Neuronas/fisiología , Ratas , Ratas Wistar
9.
Diabetes ; 63(4): 1422-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24379349

RESUMEN

We recently reported restoration of leptin responsiveness in diet-induced obese (DIO) mice using a pharmacologically optimized, polyethylene-glycolated (PEG)-leptin analog in combination with exendin-4 or FGF21. However, the return of leptin action required discontinuation of high-fat diet (HFD) exposure. Here we assess whether a single peptide possessing balanced coagonism at the glucagon-like peptide 1 (GLP-1) and glucagon receptors can restore leptin responsiveness in DIO mice maintained on a HFD. DIO mice were treated with PEG-GLP-1/glucagon (30 nmol/kg every fourth day) to induce an ∼15% body weight loss, upon which they were randomized to continue PEG-GLP-1/glucagon therapy or reassigned to receive supplemental daily PEG-leptin (185 nmol/kg/day). The addition of PEG-leptin to PEG-GLP-1/glucagon resulted in an ∼18% greater weight loss as compared with PEG-GLP-1/glucagon alone and was accompanied by further decreases in food intake and improved glucose and lipid metabolism. The beneficial effect of PEG-leptin supplementation occurred after an initial body weight loss similar to what we previously reported following reduced dietary fat along with PEG-leptin and exendin-4 or FGF21 cotreatment. In summary, we report that GLP-1/glucagon coagonism restores leptin responsiveness in mice maintained on a HFD, thus emphasizing the translational value of this polypharmacotherapy for the treatment of obesity and diabetes.


Asunto(s)
Péptido 1 Similar al Glucagón/agonistas , Péptido 1 Similar al Glucagón/uso terapéutico , Leptina/agonistas , Obesidad/tratamiento farmacológico , Receptores de Glucagón/agonistas , Animales , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Glucagón/agonistas , Glucagón/uso terapéutico , Leptina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Polietilenglicoles/uso terapéutico , Pérdida de Peso
10.
Exp Biol Med (Maywood) ; 234(5): 513-21, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19234057

RESUMEN

Selenium (Se) plays a critical role in testis, sperm, and reproduction, and testis Se levels are remarkably maintained in Se deficiency. In most other tissues, Se levels decrease dramatically as do levels of most selenoproteins and levels of a subset of Se-regulated selenoprotein mRNAs. Because of the recent identification of key molecules in the targeted trafficking of Se to the testis, we examined the hierarchy of Se regulation in testis by determining the dietary Se regulation of the full testis selenoproteome in rats fed graded levels of Se (0 to 0.8 microg Se/g) as Na2SeO3 for 28 d. Se status did not significantly affect testis weight or glutathione peroxidase 4 (Gpx4) activity (P>0.05). qRT-PCR analysis of selenoprotein mRNA expression revealed that 21 of the 24 selenoprotein mRNAs and ApoER2 mRNA (the selenoprotein P [Sepp1] receptor) were also not regulated significantly by dietary Se status. In contrast, Gpx1 activity decreased to 28% of Se-adequate levels, and mRNA levels for Gpx1, Sepp1, and Sepw1 (selenoprotein W) decreased significantly in Se-deficient rats to 45, 46, and 55%, respectively, of Se-adequate plateau levels. Overlap of hyperbolic Gpx4 activity and Sepw1 mRNA response curves with testis Se concentration, all with minimum dietary Se requirements<0.016 microg Se/g, showed the priority for synthesis of Gpx4. Higher minimum dietary Se requirements of 0.04 microg Se/g for Gpx1 activity and Sepp1 mRNA, and the even higher minimum dietary Se requirement of 0.08 microg Se/g for Gpx1 mRNA, suggest that the hierarchy of these biomarkers reflects distinct, lower priority pools, cell types, and roles for Se within the testis.


Asunto(s)
Suplementos Dietéticos , Regulación Enzimológica de la Expresión Génica , Glutatión Peroxidasa/biosíntesis , ARN Mensajero/biosíntesis , Selenio/metabolismo , Testículo/enzimología , Animales , Transporte Biológico/efectos de los fármacos , Proteínas Relacionadas con Receptor de LDL , Masculino , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Ratas , Receptores de Lipoproteína/biosíntesis , Reproducción/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenio/deficiencia , Selenio/farmacología , Selenoproteína W/biosíntesis , Espermatozoides/enzimología , Glutatión Peroxidasa GPX1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA