Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731952

RESUMEN

Porphyromonas gingivalis (Pg), a Gram-negative oral pathogen, promotes and accelerates periodontitis-associated gut disorders. Intestinal epithelial barrier dysfunction is crucial in the pathogenesis of intestinal and systemic diseases. In this study, we sought to elucidate the protective role of cinnamaldehyde (CNM, an activator of Nrf2) against P. gingivalis (W83) and Pg-derived lipopolysaccharide (Pg-LPS) induced intestinal epithelial barrier dysfunction via antioxidative mechanisms in IEC-6 cells. IEC-6 (ATCC, CRL-1592) cells were pretreated with or without CNM (100 µM), in the presence or absence of P. gingivalis (strain W83, 109 MOI) or Pg-LPS (1, 10, and 100 µg/mL), respectively, between 0-72 h time points by adopting a co-culture method. Intestinal barrier function, cytokine secretion, and intestinal oxidative stress protein markers were analyzed. P. gingivalis or Pg-LPS significantly (p < 0.05) increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels expressing oxidative stress damage. Pg-LPS, as well as Pg alone, induces inflammatory cytokines via TLR-4 signaling. Furthermore, infection reduced Nrf2 and NAD(P)H quinone dehydrogenase 1 (NQO1). Interestingly, inducible nitric oxide synthase (iNOS) protein expression significantly (p < 0.05) increased with Pg-LPS or Pg infection, with elevated levels of nitric oxide (NO). CNM treatment suppressed both Pg- and Pg-LPS-induced intestinal oxidative stress damage by reducing ROS, MDA, and NO production. Furthermore, CNM treatment significantly upregulated the expression of tight junction proteins via increasing the phosphorylation levels of PI3K/Akt/Nrf2 suppressing inflammatory cytokines. CNM protected against Pg infection-induced intestinal epithelial barrier dysfunction by activating the PI3K/Akt-mediated Nrf2 signaling pathway in IEC-6 cells.


Asunto(s)
Acroleína , Mucosa Intestinal , Factor 2 Relacionado con NF-E2 , Óxido Nítrico , Fosfatidilinositol 3-Quinasas , Porphyromonas gingivalis , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Factor 2 Relacionado con NF-E2/metabolismo , Acroleína/análogos & derivados , Acroleína/farmacología , Animales , Transducción de Señal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Porphyromonas gingivalis/patogenicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Óxido Nítrico/metabolismo , Línea Celular , Lipopolisacáridos , Estrés Oxidativo/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Citocinas/metabolismo
2.
Sci Adv ; 8(39): eabo3991, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36170368

RESUMEN

Recent genome-wide association studies corroborate classical research on developmental programming indicating that obesity is primarily a neurodevelopmental disease strongly influenced by nutrition during critical ontogenic windows. Epigenetic mechanisms regulate neurodevelopment; however, little is known about their role in establishing and maintaining the brain's energy balance circuitry. We generated neuron and glia methylomes and transcriptomes from male and female mouse hypothalamic arcuate nucleus, a key site for energy balance regulation, at time points spanning the closure of an established critical window for developmental programming of obesity risk. We find that postnatal epigenetic maturation is markedly cell type and sex specific and occurs in genomic regions enriched for heritability of body mass index in humans. Our results offer a potential explanation for both the limited ontogenic windows for and sex differences in sensitivity to developmental programming of obesity and provide a rich resource for epigenetic analyses of developmental programming of energy balance.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Hipotálamo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Índice de Masa Corporal , Epigénesis Genética , Epigenómica , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Obesidad/genética , Obesidad/metabolismo
3.
Nat Commun ; 12(1): 3525, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112797

RESUMEN

Contrasting to the established role of the hypothalamic agouti-related protein (AgRP) neurons in feeding regulation, the neural circuit and signaling mechanisms by which they control energy expenditure remains unclear. Here, we report that energy expenditure is regulated by a subgroup of AgRP neurons that send non-collateral projections to neurons within the dorsal lateral part of dorsal raphe nucleus (dlDRN) expressing the melanocortin 4 receptor (MC4R), which in turn innervate nearby serotonergic (5-HT) neurons. Genetic manipulations reveal a bi-directional control of energy expenditure by this circuit without affecting food intake. Fiber photometry and electrophysiological results indicate that the thermo-sensing MC4RdlDRN neurons integrate pre-synaptic AgRP signaling, thereby modulating the post-synaptic serotonergic pathway. Specifically, the MC4RdlDRN signaling elicits profound, bi-directional, regulation of body weight mainly through sympathetic outflow that reprograms mitochondrial bioenergetics within brown and beige fat while feeding remains intact. Together, we suggest that this AgRP neural circuit plays a unique role in persistent control of energy expenditure and body weight, hinting next-generation therapeutic approaches for obesity and metabolic disorders.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Conducción Nerviosa/fisiología , Neuronas Serotoninérgicas/fisiología , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Peso Corporal , Cromatografía Liquida , Ingestión de Alimentos/fisiología , Metabolismo Energético/genética , Masculino , Ratones , Conducción Nerviosa/efectos de los fármacos , Conducción Nerviosa/efectos de la radiación , Obesidad/metabolismo , Optogenética , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/efectos de la radiación , Serotonina/metabolismo , Serotonina/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Espectrometría de Masas en Tándem , Temperatura
4.
Mol Psychiatry ; 26(7): 2837-2853, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33767348

RESUMEN

The high comorbidity between obesity and mental disorders, such as depression and anxiety, often exacerbates metabolic and neurological symptoms significantly. However, neural mechanisms that underlie reciprocal control of feeding and mental states are largely elusive. Here we report that melanocortin 4 receptor (MC4R) neurons located in the dorsal bed nucleus of the stria terminus (dBNST) engage in the regulation of mentally associated weight gain by receiving GABAergic projections from hypothalamic AgRP neurons onto α5-containing GABAA receptors and serotonergic afferents onto 5-HT3 receptors. Chronic treatment with a high-fat diet (HFD) significantly blunts the hyperexcitability of AgRP neurons in response to not only hunger but also anxiety and depression-like stimuli. Such HFD-mediated desensitization reduces GABAergic outputs from AgRP neurons to downstream MC4RdBNST neurons, resulting in severe mental dysregulation. Genetic enhancement of the GABAAR-α5 or suppression of the 5-HT3R within the MC4RdBNST neurons not only abolishes HFD-induced anxiety and depression but also robustly reduces body weight by suppression of food intake. To gain further translational insights, we revealed that combined treatment of zonisamide (enhancing the GABAAR-α5 signaling) and granisetron (a selective 5-HT3R antagonist) alleviates mental dysfunction and yields a robust reversal of diet-induced obesity by reducing total calorie intake and altering food preference towards a healthy low-fat diet. Our results unveil a neural mechanism for reciprocal control of appetite and mental states, which culminates in a novel zonisamide-granisetron cocktail therapy for potential tackling the psychosis-obesity comorbidity.


Asunto(s)
Trastorno Depresivo , Serotonina , Proteína Relacionada con Agouti , Ansiedad , Depresión , Humanos , Obesidad , Ácido gamma-Aminobutírico
5.
Nat Commun ; 8(1): 713, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28959025

RESUMEN

Ghrelin is the only known circulating orexigenic hormone. It is primarily secreted by the stomach and acts at its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), in the hypothalamus to signal hunger and promote food intake. The melanocortin receptor accessory protein 2 (MRAP2) was previously shown to regulate energy homeostasis through the modulation of the activity of the melanocortin-4 receptor and prokineticin receptors. In this study we identify MRAP2 as a partner of ghrelin-GHSR1a signaling. We show that MRAP2 interacts with GHSR1a and potentiates ghrelin-stimulated signaling both in vitro and in vivo. We demonstrate that in the absence of MRAP2, fasting fails to activate agouti-related protein neurons. In addition, we show that the orexigenic effect of ghrelin is lost in mice lacking MRAP2. Our results suggest that MRAP2 is an important modulator of the energy homeostasis machinery that operates through the regulation of multiple GPCRs throughout the hypothalamus.Melanocortin receptor accessory protein 2 (MRAP2) is an adaptor protein that contributes to melanocortin-4 receptor and prokineticin receptor 1 signalling. Here the authors show that MRAP2 also regulates ghrelin receptor signalling in the hypothalamus and starvation sensing in mice.


Asunto(s)
Hambre/fisiología , Proteínas Modificadoras de la Actividad de Receptores/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Ingestión de Alimentos , Metabolismo Energético , Ghrelina/metabolismo , Homeostasis , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Modificadoras de la Actividad de Receptores/genética , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Ghrelina/genética
6.
Proc Natl Acad Sci U S A ; 113(13): 3645-50, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26976589

RESUMEN

Currently available inducible Cre/loxP systems, despite their considerable utility in gene manipulation, have pitfalls in certain scenarios, such as unsatisfactory recombination rates and deleterious effects on physiology and behavior. To overcome these limitations, we designed a new, inducible gene-targeting system by introducing an in-frame nonsense mutation into the coding sequence of Cre recombinase (nsCre). Mutant mRNAs transcribed from nsCre transgene can be efficiently translated into full-length, functional Cre recombinase in the presence of nonsense suppressors such as aminoglycosides. In a proof-of-concept model, GABA signaling from hypothalamic neurons expressing agouti-related peptide (AgRP) was genetically inactivated within 4 d after treatment with a synthetic aminoglycoside. Disruption of GABA synthesis in AgRP neurons in young adult mice led to a dramatic loss of body weight due to reduced food intake and elevated energy expenditure; they also manifested glucose intolerance. In contrast, older mice with genetic inactivation of GABA signaling by AgRP neurons had only transient reduction of feeding and body weight; their energy expenditure and glucose tolerance were unaffected. These results indicate that GABAergic signaling from AgRP neurons plays a key role in the control of feeding and metabolism through an age-dependent mechanism. This new genetic technique will augment current tools used to elucidate mechanisms underlying many physiological and neurological processes.


Asunto(s)
Metabolismo Energético/genética , Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Ácido gamma-Aminobutírico/fisiología , Proteína Relacionada con Agouti/deficiencia , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/fisiología , Animales , Ingeniería Genética , Glutamato Descarboxilasa/deficiencia , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/fisiología , Hipotálamo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal
7.
Elife ; 52016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26829592

RESUMEN

The Melanocortin Receptor Accessory Protein 2 (MRAP2) is an important regulator of energy homeostasis and its loss causes severe obesity in rodents. MRAP2 mediates its action in part through the potentiation of the MC4R, however, it is clear that MRAP2 is expressed in tissues that do not express MC4R, and that the deletion of MRAP2 does not recapitulate the phenotype of Mc4r KO mice. Consequently, we hypothesized that other GPCRs involved in the control of energy homeostasis are likely to be regulated by MRAP2. In this study we identified PKR1 as the first non-melanocortin GPCR to be regulated by MRAP2. We show that MRAP2 significantly and specifically inhibits PKR1 signaling. We also demonstrate that PKR1 and MRAP2 co-localize in neurons and that Mrap2 KO mice are hypersensitive to PKR1 stimulation. This study not only identifies new partners of MRAP2 but also a new pathway through which MRAP2 regulates energy homeostasis.


Asunto(s)
Ingestión de Alimentos , Proteínas Modificadoras de la Actividad de Receptores/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Ratones , Ratones Noqueados , Neuronas/química
8.
Proc Natl Acad Sci U S A ; 110(36): 14765-70, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23964123

RESUMEN

Diphtheria toxin-mediated, acute ablation of hypothalamic neurons expressing agouti-related protein (AgRP) in adult mice leads to anorexia and starvation within 7 d that is caused by hyperactivity of neurons within the parabrachial nucleus (PBN). Because NMDA glutamate receptors are involved in various synaptic plasticity-based behavioral modifications, we hypothesized that modulation of the NR2A and NR2B subunits of the NMDA receptor in PBN neurons could contribute to the anorexia phenotype. We observed by Western blot analyses that ablation of AgRP neurons results in enhanced expression of NR2B along with a modest suppression of NR2A. Interestingly, systemic administration of LiCl in a critical time window before AgRP neuron ablation abolished the anorectic response. LiCl treatment suppressed NR2B levels in the PBN and ameliorated the local Fos induction that is associated with anorexia. This protective role of LiCl on feeding was blunted in vagotomized mice. Chronic infusion of RO25-6981, a selective NR2B inhibitor, into the PBN recapitulated the role of LiCl in maintaining feeding after AgRP neuron ablation. We suggest that the accumulation of NR2B subunits in the PBN contributes to aphagia in response to AgRP neuron ablation and may be involved in other forms of anorexia.


Asunto(s)
Apetito/fisiología , Neuronas/fisiología , Puente/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Adyuvantes Inmunológicos/farmacología , Proteína Relacionada con Agouti/deficiencia , Proteína Relacionada con Agouti/genética , Animales , Anorexia/genética , Anorexia/fisiopatología , Anorexia/prevención & control , Apetito/efectos de los fármacos , Western Blotting , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Trastornos de Deglución/genética , Trastornos de Deglución/fisiopatología , Trastornos de Deglución/prevención & control , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Cloruro de Litio/farmacología , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenoles , Piperidinas/farmacología , Puente/citología , Puente/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Rombencéfalo/citología , Rombencéfalo/metabolismo , Rombencéfalo/fisiología , Factores de Tiempo , Vagotomía
9.
Endocrinology ; 152(3): 890-902, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21239438

RESUMEN

Defective melanocortin signaling causes hyperphagic obesity in humans and the melanocortin-4 receptor knockout mouse (MC4R(-/-)). The human disease most commonly presents, however, as haploinsufficiency of the MC4R. This study validates the MC4R(+/-) mouse as a model of the human disease in that, like the MC4R(-/-), the MC4R(+/-) mouse also exhibits a sustained hyperphagic response to dietary fat. Furthermore, both saturated and monounsaturated fats elicit this response. N-acylphosphatidylethanolamine (NAPE) is a signaling lipid induced after several hours of high-fat feeding, that, if dysregulated, might explain the feeding behavior in melanocortin obesity syndrome. Remarkably, however, MC4R(-/-) mice produce elevated levels of NAPE and are fully responsive to the anorexigenic activity of NAPE and oleoylethanolamide. Interestingly, additional differences in N-acylethanolamine (NAE) biochemistry were seen in MC4R(-/-) animals, including reduced plasma NAE levels and elevated hypothalamic levels of fatty acid amide hydrolase expression. Thus, while reduced expression of NAPE or NAE does not explain the high-fat hyperphagia in the melanocortin obesity syndrome, alterations in this family of signaling lipids are evident. Analysis of the microstructure of feeding behavior in response to dietary fat in the MC4R(-/-) and MC4R(+/-) mice indicates that the high-fat hyperphagia involves defective satiation and an increased rate of food intake, suggesting defective satiety signaling and enhanced reward value of dietary fat.


Asunto(s)
Grasas de la Dieta/metabolismo , Hiperfagia/genética , Hiperfagia/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Femenino , Regulación de la Expresión Génica/fisiología , Heterocigoto , Masculino , Ratones , Ratones Noqueados , Mutación , Receptor de Melanocortina Tipo 1/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(1): 355-60, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21169216

RESUMEN

Melanocortin-4 receptor (MC4R) is critical for energy homeostasis, and the paraventricular nucleus of the hypothalamus (PVN) is a key site of MC4R action. Most studies suggest that leptin regulates PVN neurons indirectly, by binding to receptors in the arcuate nucleus or ventromedial hypothalamus and regulating release of products like α-melanocyte-stimulating hormone (α-MSH), neuropeptide Y (NPY), glutamate, and GABA from first-order neurons onto the MC4R PVN cells. Here, we investigate mechanisms underlying regulation of activity of these neurons under various metabolic states by using hypothalamic slices from a transgenic MC4R-GFP mouse to record directly from MC4R neurons. First, we show that in vivo leptin levels regulate the tonic firing rate of second-order MC4R PVN neurons, with fasting increasing firing frequency in a leptin-dependent manner. We also show that, although leptin inhibits these neurons directly at the postsynaptic membrane, α-MSH and NPY potently stimulate and inhibit the cells, respectively. Thus, in contrast with the conventional model of leptin action, the primary control of MC4R PVN neurons is unlikely to be mediated by leptin action on arcuate NPY/agouti-related protein and proopiomelanocortin neurons. We also show that the activity of MC4R PVN neurons is controlled by the constitutive activity of the MC4R and that expression of the receptor mRNA and α-MSH sensitivity are both stimulated by leptin. Thus, leptin acts multinodally on arcuate nucleus/PVN circuits to regulate energy homeostasis, with prominent mechanisms involving direct control of both membrane conductances and gene expression in the MC4R PVN neuron.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Metabolismo Energético/fisiología , Homeostasis/fisiología , Leptina/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal/fisiología , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Electrofisiología , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Melanocortinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Neurológicos , Neuronas/metabolismo , Neuronas/fisiología , Neuropéptido Y/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Reacción en Cadena de la Polimerasa , alfa-MSH/metabolismo
11.
Mol Endocrinol ; 24(12): 2366-81, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20943814

RESUMEN

Fasting-induced suppression of thyroid hormone levels is an adaptive response to reduce energy expenditure in both humans and mice. This suppression is mediated by the hypothalamic-pituitary-thyroid axis through a reduction in TRH levels expressed in neurons of the paraventricular nucleus of the hypothalamus (PVN). TRH gene expression is positively regulated by leptin. Whereas decreased leptin levels during fasting lead to a reduction in TRH gene expression, the mechanisms underlying this process are still unclear. Indeed, evidence exists that TRH neurons in the PVN are targeted by leptin indirectly via the arcuate nucleus, whereas correlative evidence for a direct action exists as well. Here we provide both in vivo and in vitro evidence that the activity of hypothalamic-pituitary-thyroid axis is regulated by both direct and indirect leptin regulation. We show that both leptin and α-MSH induce significant neuronal activity mediated through a postsynaptic mechanism in TRH-expressing neurons of PVN. Furthermore, we provide in vivo evidence indicating the contribution of each pathway in maintaining serum levels of thyroid hormone.


Asunto(s)
Adiposidad/fisiología , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Hormona Liberadora de Tirotropina/biosíntesis , Adiposidad/efectos de los fármacos , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Ayuno/sangre , Ayuno/metabolismo , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Leptina/metabolismo , Leptina/farmacología , Melanocortinas/agonistas , Melanocortinas/farmacología , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Neuropéptido Y/farmacología , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Ratas , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Hormonas Tiroideas/sangre , Hormonas Tiroideas/metabolismo , Hormona Liberadora de Tirotropina/antagonistas & inhibidores , Hormona Liberadora de Tirotropina/genética , alfa-MSH/metabolismo , alfa-MSH/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...