Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Neuroinflammation ; 16(1): 221, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31727092

RESUMEN

BACKGROUND: Obese mice on a high-fat diet (HFD) display signs of inflammation in the hypothalamic arcuate nucleus (ARC), a critical area for controlling systemic energy metabolism. This has been suggested as a key mechanism of obesity-associated hypothalamic dysfunction. We reported earlier that bone marrow-derived macrophages accumulate in the ARC to sustain hypothalamic inflammation upon chronic exposure to an HFD. However, the mechanism underlying hypothalamic macrophage accumulation has remained unclear. METHODS: We investigated whether circulating monocytes or myeloid precursors contribute to hypothalamic macrophage expansion during chronic HFD feeding. To trace circulating myeloid cells, we generated mice that express green fluorescent protein (GFP) in their lysozyme M-expressing myeloid cells (LysMGFP mice). We conducted parabiosis and bone marrow transplantation experiments using these animals. Mice received an HFD for 12 or 30 weeks and were then sacrificed to analyze LysMGFP cells in the hypothalamus. Hypothalamic vascular permeability in the HFD-fed obese mice was also tested by examining the extravascular leakage of Evans blue and fluorescence-labeled albumin. The timing of LysMGFP cell entry to the hypothalamus during development was also evaluated. RESULTS: Our parabiosis and bone marrow transplantation experiments revealed a significant infiltration of circulating LysMGFP cells into the liver, skeletal muscle, choroid plexus, and leptomeninges but not in the hypothalamic ARC during chronic HFD feeding, despite increased hypothalamic vascular permeability. These results suggested that the recruitment of circulating monocytes is not a major mechanism for maintaining and expanding the hypothalamic macrophage population in diet-induced obesity. We demonstrated instead that LysMGFP cells infiltrate the hypothalamus during its development. LysMGFP cells appeared in the hypothalamic area from the late embryonic period. This cellular pool suddenly increased immediately after birth, peaked at the postnatal second week, and adopted an adult pattern of distribution after weaning. CONCLUSIONS: Bone marrow-derived macrophages mostly populate the hypothalamus in early postnatal life and may maintain their pool without significant recruitment of circulating monocytes throughout life, even under conditions of chronic HFD feeding.


Asunto(s)
Hipotálamo/metabolismo , Macrófagos/metabolismo , Obesidad/metabolismo , Animales , Trasplante de Médula Ósea , Permeabilidad Capilar , Dieta Alta en Grasa , Metabolismo Energético , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Masculino , Ratones , Parabiosis
2.
J Biol Chem ; 290(29): 18146-18155, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26041775

RESUMEN

Terminally differentiated neurons have a single, primary cilium. The primary cilia of hypothalamic neurons play a critical role in sensing metabolic signals. We recently showed that mice with leptin deficiency or resistance have shorter cilia in the hypothalamic neurons, and leptin treatment elongates cilia in hypothalamic neurons. Here, we investigated the molecular mechanisms by which leptin controls ciliary length in hypothalamic neurons. In N1 hypothalamic neuronal cells, leptin treatment increased the expression of intraflagellar transport proteins. These effects occurred via phosphatase and tensin homolog/glycogen synthase kinase-3ß-mediated inhibition of the transcriptional factor RFX1. Actin filament dynamics were also involved in leptin-promoted ciliary elongation. Both leptin and cytochalasin-D treatment induced F-actin disruption and cilium elongation in hypothalamic neurons that was completely abrogated by co-treatment with the F-actin polymerizer phalloidin. Our findings suggest that leptin elongates hypothalamic neuronal cilia by stimulating the production of intraflagellar transport proteins and destabilizing actin filaments.


Asunto(s)
Actinas/metabolismo , Cilios/metabolismo , Hipotálamo/citología , Leptina/metabolismo , Neuronas/citología , Actinas/ultraestructura , Animales , Línea Celular , Línea Celular Tumoral , Cilios/ultraestructura , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Fosfohidrolasa PTEN/metabolismo
3.
EMBO Rep ; 15(7): 801-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24825475

RESUMEN

Hypothalamic leptin signaling plays a central role in maintaining body weight homeostasis. Here, we show that clusterin/ApoJ, recently identified as an anorexigenic neuropeptide, is an important regulator in the hypothalamic leptin signaling pathway. Coadministration of clusterin potentiates the anorexigenic effect of leptin and boosts leptin-induced hypothalamic Stat3 activation. In cultured neurons, clusterin enhances receptor binding and subsequent endocytosis of leptin. These effects are mainly mediated through the LDL receptor-related protein-2 (Lrp2). Notably, inhibition of hypothalamic clusterin, Lrp2 or endocytosis abrogates anorexia and hypothalamic Stat3 activation caused by leptin. These findings propose a novel regulatory mechanism in central leptin signaling pathways.


Asunto(s)
Clusterina/metabolismo , Endocitosis/fisiología , Leptina/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Transducción de Señal , Animales , Clusterina/deficiencia , Clusterina/genética , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Unión Proteica , Receptores de Leptina/metabolismo
4.
Exp Physiol ; 99(9): 1104-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24972836

RESUMEN

Insulin resistance is a hallmark feature of type 2 diabetes and obesity. In addition to the classical view that insulin resistance in the liver, muscle and fat disrupts glucose homeostasis, studies in the past decade have illustrated that insulin resistance in the hypothalamus dysregulates hepatic glucose production and food intake, leading to type 2 diabetes and obesity. This invited review argues that in addition to the hypothalamus, insulin signalling in the dorsal vagal complex regulates hepatic glucose production and food intake. A thorough understanding of the physiological and pathophysiological mechanisms of insulin action in the hypothalamus and dorsal vagal complex is necessary in order to identify therapeutic targets for obesity and type 2 diabetes.


Asunto(s)
Hipotálamo/metabolismo , Insulina/metabolismo , Transducción de Señal , Núcleo Solitario/metabolismo , Nervio Vago/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Ingestión de Alimentos , Metabolismo Energético , Gluconeogénesis , Humanos , Hipotálamo/fisiopatología , Resistencia a la Insulina , Hígado/metabolismo , Obesidad/metabolismo , Obesidad/fisiopatología , Núcleo Solitario/fisiopatología , Nervio Vago/fisiopatología
5.
Yonsei Med J ; 65(2): 55-69, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38288646

RESUMEN

Mitochondria function as platforms for bioenergetics, nutrient metabolism, intracellular signaling, innate immunity regulators, and modulators of stem cell activity. Thus, the decline in mitochondrial functions causes or correlates with diabetes mellitus and many aging-related diseases. Upon stress or damage, the mitochondria elicit a series of adaptive responses to overcome stress and restore their structural integrity and functional homeostasis. These adaptive responses to low-level or transient mitochondrial stress promote health and resilience to upcoming stress. Beneficial effects of low-grade mitochondrial stress, termed mitohormesis, have been observed in various organisms, including mammals. Accumulated evidence indicates that treatments boosting mitohormesis have therapeutic potential in various human diseases accompanied by mitochondrial stress. Here, we review multiple cellular signaling pathways and interorgan communication mechanisms through which mitochondrial stress leads to advantageous outcomes. We also discuss the relevance of mitohormesis in obesity, diabetes, metabolic liver disease, aging, and exercise.


Asunto(s)
Promoción de la Salud , Enfermedades Metabólicas , Animales , Humanos , Mitocondrias/metabolismo , Envejecimiento , Ejercicio Físico/fisiología , Estrés Oxidativo , Mamíferos
6.
BMB Rep ; 57(3): 149-154, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37817436

RESUMEN

The stomach has emerged as a crucial endocrine organ in the regulation of feeding since the discovery of ghrelin. Gut-derived hormones, such as ghrelin and cholecystokinin, can act through the vagus nerve. We previously reported the satiety effect of hypothalamic clusterin, but the impact of peripheral clusterin remains unknown. In this study, we administered clusterin intraperitoneally to mice and observed its ability to suppress fasting-driven food intake. Interestingly, we found its synergism with cholecystokinin and antagonism with ghrelin. These effects were accompanied by increased c-fos immunoreactivity in nucleus tractus solitarius, area postrema, and hypothalamic paraventricular nucleus. Notably, truncal vagotomy abolished this response. The stomach expressed clusterin at high levels among the organs, and gastric clusterin was detected in specific enteroendocrine cells and the submucosal plexus. Gastric clusterin expression decreased after fasting but recovered after 2 hours of refeeding. Furthermore, we confirmed that stomachspecific overexpression of clusterin reduced food intake after overnight fasting. These results suggest that gastric clusterin may function as a gut-derived peptide involved in the regulation of feeding through the gut-brain axis. [BMB Reports 2024; 57(3): 149-154].


Asunto(s)
Ingestión de Alimentos , Ghrelina , Ratones , Animales , Ghrelina/farmacología , Ingestión de Alimentos/fisiología , Clusterina/farmacología , Colecistoquinina/farmacología , Estómago , Conducta Alimentaria
7.
BMB Rep ; 57(7): 342, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39079709

RESUMEN

[Erratum to: BMB Reports 2024; 57(3): 149-154, PMID: 37817436, PMCID: PMC10979347] The BMB Reports would like to correct in BMB Rep. 57(3):149-154, titled "Stomach clusterin as a gut-derived feeding regulator". This research was supported by the Creative-Pioneering Researchers Program through Seoul National University. Since grant name and number are incorrect, this information has now been corrected as follows: This work was supported by the National Research Foundation of Korea funded by the Korean government (2020R1A2C3004843, 2022M3E5E8017213 to M-S.K., 2020R1C1C10 08033 to O.K.) and by Creative-Pioneering Researchers Program through Seoul National University (to O.K.). The authors apologize for any inconvenience or confusion that may be caused by this error. The ACKNOWLEDGEMENTS of Original PDF version have been corrected.

8.
Nat Commun ; 15(1): 2102, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453901

RESUMEN

Nicotinamide adenine dinucleotide (NAD)+ serves as a crucial coenzyme in numerous essential biological reactions, and its cellular availability relies on the activity of the nicotinamide phosphoribosyltransferase (NAMPT)-catalyzed salvage pathway. Here we show that treatment with saturated fatty acids activates the NAD+ salvage pathway in hypothalamic astrocytes. Furthermore, inhibition of this pathway mitigates hypothalamic inflammation and attenuates the development of obesity in male mice fed a high-fat diet (HFD). Mechanistically, CD38 functions downstream of the NAD+ salvage pathway in hypothalamic astrocytes burdened with excess fat. The activation of the astrocytic NAMPT-NAD+-CD38 axis in response to fat overload induces proinflammatory responses in the hypothalamus. It also leads to aberrantly activated basal Ca2+ signals and compromised Ca2+ responses to metabolic hormones such as insulin, leptin, and glucagon-like peptide 1, ultimately resulting in dysfunctional hypothalamic astrocytes. Our findings highlight the significant contribution of the hypothalamic astrocytic NAD+ salvage pathway, along with its downstream CD38, to HFD-induced obesity.


Asunto(s)
Grasas de la Dieta , NAD , Masculino , Ratones , Animales , NAD/metabolismo , Grasas de la Dieta/metabolismo , Astrocitos/metabolismo , Obesidad/metabolismo , Hipotálamo/metabolismo , Citocinas/metabolismo
9.
Neuroendocrinology ; 98(3): 233-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080898

RESUMEN

Clusterin is a sulfated glycoprotein abundantly expressed in the pituitary gland and hypothalamus of mammals. However, its physiological role in neuroendocrine function is largely unknown. In the present study, we investigated the effects of intracerebroventricular (ICV) administration of clusterin on plasma pituitary hormone levels in normal rats. Single ICV injection of clusterin provoked neurohormonal changes seen under acute stress condition: increased plasma adrenocorticotropic hormone (ACTH), corticosterone, GH and prolactin levels and decreased LH and FSH levels. Consistently, hypothalamic and pituitary clusterin expression levels were upregulated following a restraint stress, suggesting an involvement of endogenous clusterin in stress-induced neurohormonal changes. In the pituitary intermediate lobe, clusterin was coexpressed with proopiomelanocortin (POMC), a precursor of ACTH. Treatment of clusterin in POMC expressing AtT-20 pituitary cells increased basal and corticotropin-releasing hormone (CRH)-stimulated POMC promoter activities and intracellular cAMP levels. Furthermore, clusterin treatment triggered ACTH secretion from AtT-20 cells in a CRH-dependent manner, indicating that increased clusterin under stressful conditions may augment CRH-stimulated ACTH production and release. In summary, hypothalamic and pituitary clusterin may function as a modulator of neurohormonal responses under stressful conditions.


Asunto(s)
Clusterina/fisiología , Hipotálamo/metabolismo , Neurotransmisores/biosíntesis , Hipófisis/metabolismo , Hormona Adrenocorticotrópica/antagonistas & inhibidores , Hormona Adrenocorticotrópica/biosíntesis , Hormona Adrenocorticotrópica/metabolismo , Animales , Clusterina/administración & dosificación , Clusterina/sangre , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Neurotransmisores/antagonistas & inhibidores , Neurotransmisores/metabolismo , Hipófisis/efectos de los fármacos , Proopiomelanocortina/antagonistas & inhibidores , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/sangre , Estrés Psicológico/prevención & control , Estrés Psicológico/psicología , Regulación hacia Arriba/fisiología
10.
Nat Commun ; 14(1): 1994, 2023 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-37031230

RESUMEN

Nicotinamide adenine dinucleotide (NAD+) is an essential cofactor of critical enzymes including protein deacetylase sirtuins/SIRTs and its levels in mammalian cells rely on the nicotinamide phosphoribosyltransferase (NAMPT)-mediated salvage pathway. Intracellular NAMPT (iNAMPT) is secreted and found in the blood as extracellular NAMPT (eNAMPT). In the liver, the iNAMPT-NAD+ axis oscillates in a circadian manner and regulates the cellular clockwork. Here we show that the hypothalamic NAD+ levels show a distinct circadian fluctuation with a nocturnal rise in lean mice. This rhythm is in phase with that of plasma eNAMPT levels but not with that of hypothalamic iNAMPT levels. Chemical and genetic blockade of eNAMPT profoundly inhibit the nighttime elevations in hypothalamic NAD+ levels as well as those in locomotor activity (LMA) and energy expenditure (EE). Conversely, elevation of plasma eNAMPT by NAMPT administration increases hypothalamic NAD+ levels and stimulates LMA and EE via the hypothalamic NAD+-SIRT-FOXO1-melanocortin pathway. Notably, obese animals display a markedly blunted circadian oscillation in blood eNAMPT-hypothalamic NAD+-FOXO1 axis as well as LMA and EE. Our findings indicate that the eNAMPT regulation of hypothalamic NAD+ biosynthesis underlies circadian physiology and that this system can be significantly disrupted by obesity.


Asunto(s)
Citocinas , NAD , Ratones , Animales , NAD/metabolismo , Citocinas/metabolismo , Hígado/metabolismo , Metabolismo Energético , Ritmo Circadiano , Locomoción , Mamíferos/metabolismo
11.
Mol Cells ; 45(4): 169-176, 2022 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-35387896

RESUMEN

A primary cilium, a hair-like protrusion of the plasma membrane, is a pivotal organelle for sensing external environmental signals and transducing intracellular signaling. An interesting linkage between cilia and obesity has been revealed by studies of the human genetic ciliopathies Bardet-Biedl syndrome and Alström syndrome, in which obesity is a principal manifestation. Mouse models of cell type-specific cilia dysgenesis have subsequently demonstrated that ciliary defects restricted to specific hypothalamic neurons are sufficient to induce obesity and hyperphagia. A potential mechanism underlying hypothalamic neuron cilia-related obesity is impaired ciliary localization of G protein-coupled receptors involved in the regulation of appetite and energy metabolism. A well-studied example of this is melanocortin 4 receptor (MC4R), mutations in which are the most common cause of human monogenic obesity. In the paraventricular hypothalamus neurons, a blockade of ciliary trafficking of MC4R as well as its downstream ciliary signaling leads to hyperphagia and weight gain. Another potential mechanism is reduced leptin signaling in hypothalamic neurons with defective cilia. Leptin receptors traffic to the periciliary area upon leptin stimulation. Moreover, defects in cilia formation hamper leptin signaling and actions in both developing and differentiated hypothalamic neurons. The list of obesity-linked ciliary proteins is expending and this supports a tight association between cilia and obesity. This article provides a brief review on the mechanism of how ciliary defects in hypothalamic neurons facilitate obesity.


Asunto(s)
Cilios , Leptina , Animales , Cilios/metabolismo , Humanos , Hiperfagia/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones , Obesidad/genética , Obesidad/metabolismo
12.
J Biol Chem ; 285(13): 9706-9715, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20097762

RESUMEN

Anorexia and weight loss are prevalent in infectious diseases. To investigate the molecular mechanisms underlying these phenomena, we established animal models of infection-associated anorexia by administrating bacterial and viral products, lipopolysaccharide (LPS) and human immunodeficiency virus-1 transactivator protein (Tat). In these models, we found that the nuclear factor-kappaB (NF-kappaB), a pivotal transcription factor for inflammation-related proteins, was activated in the hypothalamus. In parallel, administration of LPS and Tat increased hypothalamic pro-inflammatory cytokine production, which was abrogated by inhibition of hypothalamic NF-kappaB. In vitro, NF-kappaB activation directly stimulated the transcriptional activity of pro-opiomelanocortin (POMC), a precursor of anorexigenic melanocortin, and mediated the stimulatory effects of LPS, Tat, and pro-inflammatory cytokines on POMC transcription, implying the involvement of NF-kappaB in controlling feeding behavior. Consistently, hypothalamic injection of LPS and Tat caused a significant reduction in food intake and body weight, which was prevented by blockade of NF-kappaB and melanocortin. Furthermore, disruption of I kappaB kinase-beta, an upstream kinase of NF-kappaB, in POMC neurons attenuated LPS- and Tat-induced anorexia. These findings suggest that infection-associated anorexia and weight loss are mediated via NF-kappaB activation in hypothalamic POMC neurons. In addition, hypothalamic NF-kappaB was activated by leptin, an important anorexigenic hormone, and mediates leptin-stimulated POMC transcription, indicating that hypothalamic NF-kappaB also serves as a downstream signaling pathway of leptin.


Asunto(s)
Anorexia/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Animales , Línea Celular Tumoral , Humanos , Lipopolisacáridos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos , Modelos Biológicos
13.
Cell Metab ; 33(2): 334-349.e6, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33535098

RESUMEN

Low-grade mitochondrial stress can promote health and longevity, a phenomenon termed mitohormesis. Here, we demonstrate the opposing metabolic effects of low-level and high-level mitochondrial ribosomal (mitoribosomal) stress in hypothalamic proopiomelanocortin (POMC) neurons. POMC neuron-specific severe mitoribosomal stress due to Crif1 homodeficiency causes obesity in mice. By contrast, mild mitoribosomal stress caused by Crif1 heterodeficiency in POMC neurons leads to high-turnover metabolism and resistance to obesity. These metabolic benefits are mediated by enhanced thermogenesis and mitochondrial unfolded protein responses (UPRmt) in distal adipose tissues. In POMC neurons, partial Crif1 deficiency increases the expression of ß-endorphin (ß-END) and mitochondrial DNA-encoded peptide MOTS-c. Central administration of MOTS-c or ß-END recapitulates the adipose phenotype of Crif1 heterodeficient mice, suggesting these factors as potential mediators. Consistently, regular running exercise at moderate intensity stimulates hypothalamic MOTS-c/ß-END expression and induces adipose tissue UPRmt and thermogenesis. Our findings indicate that POMC neuronal mitohormesis may underlie exercise-induced high-turnover metabolism.


Asunto(s)
Hipotálamo/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Condicionamiento Físico Animal , Proopiomelanocortina/metabolismo , Animales , Línea Celular Tumoral , Metabolismo Energético , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
14.
Nat Commun ; 11(1): 5772, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33188191

RESUMEN

Hypothalamic neurons including proopiomelanocortin (POMC)-producing neurons regulate body weights. The non-motile primary cilium is a critical sensory organelle on the cell surface. An association between ciliary defects and obesity has been suggested, but the underlying mechanisms are not fully understood. Here we show that inhibition of ciliogenesis in POMC-expressing developing hypothalamic neurons, by depleting ciliogenic genes IFT88 and KIF3A, leads to adulthood obesity in mice. In contrast, adult-onset ciliary dysgenesis in POMC neurons causes no significant change in adiposity. In developing POMC neurons, abnormal cilia formation disrupts axonal projections through impaired lysosomal protein degradation. Notably, maternal nutrition and postnatal leptin surge have a profound impact on ciliogenesis in the hypothalamus of neonatal mice; through these effects they critically modulate the organization of hypothalamic feeding circuits. Our findings reveal a mechanism of early life programming of adult adiposity, which is mediated by primary cilia in developing hypothalamic neurons.


Asunto(s)
Adiposidad , Cilios/metabolismo , Hipotálamo/embriología , Hipotálamo/metabolismo , Lisosomas/metabolismo , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/metabolismo , Axones/metabolismo , Metabolismo Energético , Femenino , Glucosa/metabolismo , Leptina/metabolismo , Desnutrición/patología , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Neurogénesis , Obesidad/metabolismo , Obesidad/patología , Organogénesis , Proopiomelanocortina/metabolismo , Proteolisis
15.
Neurosci Lett ; 681: 105-109, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-29857041

RESUMEN

Leptin plays a pivotal role in the central control of energy balance through leptin receptors expressed on specific hypothalamic nuclei. Leptin suppresses food intake and body weight and ameliorates hyperglycemia by acting on the AgRP and POMC neurons of the arcuate nucleus. Leptin action on POMC neurons are essential for control of body weight and blood glucose levels and are known to be mediated by JAK-STAT3 and PI3K signalling pathway thus increase POMC mRNA and intrinsic excitability. The effects of leptin on AgRP neurons are not as clear although it has been reported to hyperpolarize AgRP neurons through change of K+ conductance. Using cell-attached patch and whole cell patch configuration, we directly assessed neuronal response to leptin in GFP labelled AgRP or POMC neurons in mice after 18 h of food deprivation. We found leptin has a direct effect on POMC neuron through increased intrinsic excitability and decreased inhibitory synaptic inputs. However, leptin does not have any effect on intrinsic excitability of AgRP neurons in fasted condition although food deprivation induced increase of firing frequency of AgRP neurons. In conclusion, leptin probably has a direct and acute effect on POMC neurons but not on AgRP neurons to control their excitability within feeding-regulatory neuronal circuitry.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Privación de Alimentos/fisiología , Hipotálamo/metabolismo , Leptina/farmacología , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Femenino , Hipotálamo/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos
16.
J Appl Physiol (1985) ; 125(3): 715-722, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29952246

RESUMEN

Angiopoietin-like protein 4 (Angptl4)/fasting-induced adipose factor (Fiaf) expression levels are increased by exercise in skeletal muscle. We have previously shown that Angptl4 regulates food intake and energy expenditure via modulation of hypothalamic AMP-activated protein kinase (AMPK) activity. AMPK is an important signaling molecule that integrates skeletal muscle metabolism during exercise. Therefore, we investigated the involvement of Angptl4 in exercise-induced AMPK activation in skeletal muscle. Angptl4 protein and mRNA expression levels were significantly increased in the gastrocnemius and soleus muscles of mice following a 50-min running bout. Treatment of C2C12 myotubes with Angptl4 increased phosphorylation of AMPK and acetyl-CoA carboxylase (ACC), which were markers of AMPK activation, and the mitochondrial maximum respiratory capacity. Treadmill exercise increased AMPK and ACC phosphorylation in the gastrocnemius of normal mice; this phosphorylation increase was attenuated in mice lacking Angptl4. Endurance to swimming and hanging was also reduced in Angptl4 knockout mice. Taken together, our current data demonstrate that exercise-induced upregulation of skeletal muscle Angptl4 is critical for AMPK activation and exercise tolerance. These findings unveil a new role for skeletal muscle Angptl4 in exercise physiology. NEW & NOTEWORTHY 1) Angiopoietin-like protein 4 (Angptl4) treatment activates AMP-activated protein kinase (AMPK) signaling in skeletal muscle cells. 2) Angptl4 increases the maximum mitochondrial oxidative capacity through AMPK activation in skeletal muscle cells. 3) Lack of Angptl4 mitigates exercise-induced skeletal muscle AMPK activation. 4) Angptl4-deficient mice show a lower endurance to exercise.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Proteína 4 Similar a la Angiopoyetina/genética , Músculo Esquelético/enzimología , Músculo Esquelético/fisiología , Esfuerzo Físico/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Proteína 4 Similar a la Angiopoyetina/metabolismo , Animales , Activación Enzimática , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Musculares/enzimología , Mitocondrias Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Resistencia Física/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Natación/fisiología
17.
Metabolism ; 88: 51-60, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30179604

RESUMEN

BACKGROUND: Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase SIRT1 is an important regulator of hypothalamic neuronal function. Thus, an adequate hypothalamic NAD content is critical for maintaining normal energy homeostasis. METHODS: We investigated whether NAD supplementation increases hypothalamic NAD levels and affects energy metabolism in mice. Furthermore, we investigated the mechanisms underlying the effects of exogenous NAD on central metabolism upon entering the hypothalamus. RESULTS: Central and peripheral NAD administration suppressed fasting-induced hyperphagia and weight gain in mice. Extracellular NAD was imported into N1 hypothalamic neuronal cells in a connexin 43-dependent and CD73-independent manner. Consistent with the in vitro data, inhibition of hypothalamic connexin 43 blocked hypothalamic NAD uptake and NAD-induced anorexia. Exogenous NAD suppressed NPY and AgRP transcriptional activity, which was mediated by SIRT1 and FOXO1. CONCLUSIONS: Exogenous NAD is effectively transported to the hypothalamus via a connexin 43-dependent mechanism and increases hypothalamic NAD content. Therefore, NAD supplementation is a potential therapeutic method for metabolic disorders characterized by hypothalamic NAD depletion.


Asunto(s)
Conexina 43/metabolismo , Metabolismo Energético/efectos de los fármacos , Hipotálamo/efectos de los fármacos , NAD/farmacología , Proteína Relacionada con Agouti/genética , Animales , Transporte Biológico , Hiperfagia/prevención & control , Hipotálamo/citología , Hipotálamo/metabolismo , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Masculino , Ratones Endogámicos C57BL , NAD/administración & dosificación , Neuronas/metabolismo , Neuropéptido Y/genética , Sirtuina 1/metabolismo , Transcripción Genética/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
18.
Cell Rep ; 25(4): 934-946.e5, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30355499

RESUMEN

Obesity-associated metabolic alterations are closely linked to low-grade inflammation in peripheral organs, in which macrophages play a central role. Using genetic labeling of myeloid lineage cells, we show that hypothalamic macrophages normally reside in the perivascular area and circumventricular organ median eminence. Chronic consumption of a high-fat diet (HFD) induces expansion of the monocyte-derived macrophage pool in the hypothalamic arcuate nucleus (ARC), which is significantly attributed to enhanced proliferation of macrophages. Notably, inducible nitric oxide synthase (iNOS) is robustly activated in ARC macrophages of HFD-fed obese mice. Hypothalamic macrophage iNOS inhibition completely abrogates macrophage accumulation and activation, proinflammatory cytokine overproduction, reactive astrogliosis, blood-brain-barrier permeability, and lipid accumulation in the ARC of obese mice. Moreover, central iNOS inhibition improves obesity-induced alterations in systemic glucose metabolism without affecting adiposity. Our findings suggest a critical role for hypothalamic macrophage-expressed iNOS in hypothalamic inflammation and abnormal glucose metabolism in cases of overnutrition-induced obesity.


Asunto(s)
Hipotálamo/patología , Inflamación/enzimología , Macrófagos/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Obesidad/enzimología , Animales , Núcleo Arqueado del Hipotálamo/patología , Barrera Hematoencefálica/patología , Proliferación Celular , Dieta Alta en Grasa , Glucosa/metabolismo , Inflamación/patología , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Obesidad/patología , Células RAW 264.7
19.
Diabetes ; 64(4): 1142-53, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25338813

RESUMEN

Hypothalamic lipid sensing is important for the maintenance of energy balance. Angiopoietin-like protein 3 (Angptl3) critically regulates the clearance of circulating lipids by inhibiting lipoprotein lipase (LPL). The current study demonstrated that Angptl3 is highly expressed in the neurons of the mediobasal hypothalamus, an important area in brain lipid sensing. Suppression of hypothalamic Angptl3 increased food intake but reduced energy expenditure and fat oxidation, thereby promoting weight gain. Consistently, intracerebroventricular (ICV) administration of Angptl3 caused the opposite metabolic changes, supporting an important role for hypothalamic Angptl3 in the control of energy balance. Notably, ICV Angptl3 significantly stimulated hypothalamic LPL activity. Moreover, coadministration of the LPL inhibitor apolipoprotein C3 antagonized the effects of Angptl3 on energy metabolism, indicating that LPL activation is critical for the central metabolic actions of Angptl3. Increased LPL activity is expected to promote lipid uptake by hypothalamic neurons, leading to enhanced brain lipid sensing. Indeed, ICV injection of Angptl3 increased long-chain fatty acid (LCFA) and LCFA-CoA levels in the hypothalamus. Furthermore, inhibitors of hypothalamic lipid-sensing pathways prevented Angptl3-induced anorexia and weight loss. These findings identify Angptl3 as a novel regulator of the hypothalamic lipid-sensing pathway.


Asunto(s)
Angiopoyetinas/metabolismo , Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Hipotálamo/metabolismo , Lipoproteína Lipasa/metabolismo , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/genética , Angiopoyetinas/farmacología , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Masculino , Ratones , Interferencia de ARN , Ratas , Ratas Sprague-Dawley
20.
J Clin Invest ; 124(5): 2193-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24667636

RESUMEN

The majority of mammalian cells have nonmotile primary cilia on their surface that act as antenna-like sensory organelles. Genetic defects that result in ciliary dysfunction are associated with obesity in humans and rodents, which suggests that functional cilia are important for controlling energy balance. Here we demonstrated that neuronal cilia lengths were selectively reduced in hypothalami of obese mice with leptin deficiency and leptin resistance. Treatment of N1 hypothalamic neuron cells with leptin stimulated cilia assembly via inhibition of the tumor suppressors PTEN and glycogen synthase kinase 3ß (GSK3ß). Induction of short cilia in the hypothalamus of adult mice increased food intake and decreased energy expenditure, leading to a positive energy balance. Moreover, mice with short hypothalamic cilia exhibited attenuated anorectic responses to leptin, insulin, and glucose, which indicates that leptin-induced cilia assembly is essential for sensing these satiety signals by hypothalamic neurons. These data suggest that leptin governs the sensitivity of hypothalamic neurons to metabolic signals by controlling the length of the cell's antenna.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Neuronas/metabolismo , Animales , Anorexia/genética , Anorexia/metabolismo , Línea Celular , Cilios/genética , Cilios/metabolismo , Glucosa/genética , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipotálamo/citología , Insulina/genética , Insulina/metabolismo , Leptina , Ratones , Ratones Noqueados , Neuronas/citología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA