Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(19): e2215590120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126693

RESUMEN

Chronic stress induces depression- and anxiety-related behaviors, which are common mental disorders accompanied not only by dysfunction of the brain but also of the intestine. Activating transcription factor 4 (ATF4) is a stress-induced gene, and we previously show that it is important for gut functions; however, the contribution of the intestinal ATF4 to stress-related behaviors is not known. Here, we show that chronic stress inhibits the expression of ATF4 in gut epithelial cells. ATF4 overexpression in the colon relieves stress-related behavioral alterations in male mice, as measured by open-field test, elevated plus-maze test, and tail suspension test, whereas intestine-specific ATF4 knockout induces stress-related behavioral alterations in male mice. Furthermore, glutamatergic neurons are inhibited in the paraventricular thalamus (PVT) of two strains of intestinal ATF4-deficient mice, and selective activation of these neurons alleviates stress-related behavioral alterations in intestinal ATF4-deficient mice. The highly expressed gut-secreted peptide trefoil factor 3 (TFF3) is chosen from RNA-Seq data from ATF4 deletion mice and demonstrated decreased in gut epithelial cells, which is directly regulated by ATF4. Injection of TFF3 reverses stress-related behaviors in ATF4 knockout mice, and the beneficial effects of TFF3 are blocked by inhibiting PVT glutamatergic neurons using DREADDs. In summary, this study demonstrates the function of ATF4 in the gut-brain regulation of stress-related behavioral alterations, via TFF3 modulating PVT neural activity. This research provides evidence of gut signals regulating stress-related behavioral alterations and identifies possible drug targets for the treatment of stress-related behavioral disorders.


Asunto(s)
Factor de Transcripción Activador 4 , Tálamo , Masculino , Animales , Ratones , Factor de Transcripción Activador 4/metabolismo , Tálamo/metabolismo , Neuronas/metabolismo , Ratones Noqueados , Colon/metabolismo
2.
Commun Biol ; 6(1): 50, 2023 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-36641530

RESUMEN

Psychiatric disorders, such as anxiety, are associated with inflammatory bowel disease (IBD), however, the neural mechanisms regulating this comorbidity are unknown. Here, we show that hypothalamic agouti-related protein (AgRP) neuronal activity is suppressed under chronic restraint stress (CRS), a condition known to increase anxiety and colitis susceptibility. Consistently, chemogenic activation or inhibition of AgRP neurons reverses or mimics CRS-induced increase of anxiety-like behaviors and colitis susceptibility, respectively. Furthermore, CRS inhibits AgRP neuronal activity by suppressing the expression of c-Jun. Moreover, overexpression of c-Jun in these neurons protects against the CRS-induced effects, and knockdown of c-Jun in AgRP neurons (c-Jun∆AgRP) promotes anxiety and colitis susceptibility. Finally, the levels of secreted protein thrombospondin 1 (THBS1) are negatively associated with increased anxiety and colitis, and supplementing recombinant THBS1 rescues colitis susceptibility in c-Jun∆AgRP mice. Taken together, these results reveal critical roles of hypothalamic AgRP neuron-derived c-Jun in orchestrating stress-induced anxiety and colitis susceptibility.


Asunto(s)
Colitis , Hipotálamo , Ratones , Animales , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Ansiedad/etiología , Neuronas/fisiología , Colitis/genética , Colitis/metabolismo
3.
Mol Metab ; 42: 101084, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32971298

RESUMEN

OBJECTIVE: Although the hypothalamus is crucial for peripheral metabolism control, the signals in specific neurons involved remain poorly understood. The aim of our current study was to explore the role of the hypothalamic gene mothers against decapentaplegic homolog 7 (Smad7) in peripheral glucose disorders. METHODS: We studied glucose metabolism in high-fat diet (HFD)-fed mice and middle-aged mice with Cre-mediated recombination causing 1) overexpression of Smad7 in hypothalamic proopiomelanocortin (POMC) neurons, 2) deletion of Smad7 in POMC neurons, and 3) overexpression of protein kinase B (AKT) in arcuate nucleus (ARC) in Smad7 overexpressed mice. Intracerebroventricular (ICV) cannulation of insulin was used to test the hypothalamic insulin sensitivity in the mice. Hypothalamic primary neurons were used to investigate the mechanism of Smad7 regulating hypothalamic insulin signaling. RESULTS: We found that Smad7 expression was increased in POMC neurons in the hypothalamic ARC of HFD-fed or middle-aged mice. Furthermore, overexpression of Smad7 in POMC neurons disrupted the glucose balance, and deletion of Smad7 in POMC neurons prevented diet- or age-induced glucose disorders, which was likely to be independent of changes in body weight or food intake. Moreover, the effect of Smad7 was reversed by overexpression of AKT in the ARC. Finally, Smad7 decreased AKT phosphorylation by activating protein phosphatase 1c in hypothalamic primary neurons. CONCLUSIONS: Our results demonstrated that an excess of central Smad7 in POMC neurons disrupts glucose balance by attenuating hypothalamic insulin signaling. In addition, we found that this regulation was mediated by the activity of protein phosphatase 1c.


Asunto(s)
Glucosa/metabolismo , Proopiomelanocortina/metabolismo , Proteína smad7/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Dieta Alta en Grasa , Metabolismo Energético , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Hipotálamo/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Obesidad/metabolismo , Proopiomelanocortina/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteína smad7/genética
4.
Gastroenterology ; 156(4): 1098-1111, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30452920

RESUMEN

BACKGROUND & AIMS: Activating transcription factor 4 (ATF4) regulates genes involved in the inflammatory response, amino acid metabolism, autophagy, and endoplasmic reticulum stress. We investigated whether its activity is altered in patients with inflammatory bowel diseases (IBDs) and mice with enterocolitis. METHODS: We obtained biopsy samples during endoscopy from inflamed and/or uninflamed regions of the colon from 21 patients with active Crohn's disease (CD), 22 patients with active ulcerative colitis (UC), and 38 control individuals without IBD and of the ileum from 19 patients with active CD and 8 individuals without IBD in China. Mice with disruption of Atf4 specifically in intestinal epithelial cells (Atf4ΔIEC mice) and Atf4-floxed mice (controls) were given dextran sodium sulfate (DSS) to induce colitis. Some mice were given injections of recombinant defensin α1 (DEFA1) and supplementation of l-alanyl-glutamine or glutamine in drinking water. Human and mouse ileal and colon tissues were analyzed by quantitative real-time polymerase chain reaction, immunoblots, and immunohistochemistry. Serum and intestinal epithelial cell (IEC) amino acids were measured by high-performance liquid chromatography-tandem mass spectrometry. Levels of ATF4 were knocked down in IEC-18 cells with small interfering RNAs. Microbiomes were analyzed in ileal feces from mice by using 16S ribosomal DNA sequencing. RESULTS: Levels of ATF4 were significantly decreased in inflamed intestinal mucosa from patients with active CD or active UC compared with those from uninflamed regions or intestinal mucosa from control individuals. ATF4 was also decreased in colonic epithelia from mice with colitis vs mice without colitis. Atf4ΔIEC mice developed spontaneous enterocolitis and colitis of greater severity than control mice after administration of DSS. Atf4ΔIEC mice had decreased serum levels of glutamine and reduced levels of antimicrobial peptides, such as Defa1, Defa4, Defa5, Camp, and Lyz1, in ileal Paneth cells. Atf4ΔIEC mice had alterations in ileal microbiomes compared with control mice; these changes were reversed by administration of glutamine. Injections of DEFA1 reduced the severity of spontaneous enteritis and DSS-induced colitis in Atf4ΔIEC mice. We found that expression of solute carrier family 1 member 5 (SLC1A5), a glutamine transporter, was directly regulated by ATF4 in cell lines. Overexpression of SLC1A5 in IEC-18 or primary IEC cells increased glutamine uptake and expression of antimicrobial peptides. Knockdown of ATF4 in IEC-18 cells increased expression of inflammatory cytokines, whereas overexpression of SLC1A5 in the knockdown cells reduced cytokine expression. Levels of SLC1A5 were decreased in inflamed intestinal mucosa of patients with CD and UC and correlated with levels of ATF4. CONCLUSIONS: Levels of ATF4 are decreased in inflamed intestinal mucosa from patients with active CD or UC. In mice, ATF4 deficiency reduces glutamine uptake by intestinal epithelial cells and expression of antimicrobial peptides by decreasing transcription of Slc1a5. ATF4 might therefore be a target for the treatment of IBD.


Asunto(s)
Factor de Transcripción Activador 4/deficiencia , Péptidos Catiónicos Antimicrobianos/metabolismo , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Glutamina/metabolismo , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Adolescente , Adulto , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Animales , Estudios de Casos y Controles , Línea Celular , Colitis/inducido químicamente , Colitis/metabolismo , Colitis Ulcerosa/sangre , Colitis Ulcerosa/patología , Colon/citología , Colon/metabolismo , Enfermedad de Crohn/sangre , Enfermedad de Crohn/patología , Células Epiteliales , Femenino , Técnicas de Silenciamiento del Gen , Glutamina/sangre , Glutamina/farmacología , Humanos , Íleon/citología , Íleon/metabolismo , Íleon/microbiología , Mucosa Intestinal/metabolismo , Masculino , Ratones , Microbiota/efectos de los fármacos , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Células de Paneth/metabolismo , Adulto Joven
5.
PLoS Biol ; 16(5): e2004225, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29750781

RESUMEN

p38 has long been known as a central mediator of protein kinase A (PKA) signaling in brown adipocytes, which positively regulate the transcription of uncoupling protein 1 (UCP-1). However, the physiological role of p38 in adipose tissues, especially the white adipose tissue (WAT), is largely unknown. Here, we show that mice lacking p38α in adipose tissues display a lean phenotype, improved metabolism, and resistance to diet-induced obesity. Surprisingly, ablation of p38α causes minimal effects on brown adipose tissue (BAT) in adult mice, as evident from undetectable changes in UCP-1 expression, mitochondrial function, body temperature (BT), and energy expenditure. In contrast, genetic ablation of p38α in adipose tissues not only markedly facilitates the browning in WAT upon cold stress but also prevents diet-induced obesity. Consistently, pharmaceutical inhibition of p38α remarkably enhances the browning of WAT and has metabolic benefits. Furthermore, our data suggest that p38α deficiency promotes white-to-beige adipocyte reprogramming in a cell-autonomous manner. Mechanistically, inhibition of p38α stimulates the UCP-1 transcription through PKA and its downstream cAMP-response element binding protein (CREB), which form a positive feedback loop that functions to reinforce the white-to-beige phenotypic switch during cold exposure. Together, our study reveals that inhibition of p38α is able to promote WAT browning and confer metabolic benefits. Our study also indicates that p38α in WAT represents an exciting pharmacological target to combat obesity and metabolic diseases.


Asunto(s)
Tejido Adiposo/metabolismo , Imidazoles/uso terapéutico , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Obesidad/metabolismo , Piridinas/uso terapéutico , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Reprogramación Celular , Frío , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dieta Alta en Grasa , Evaluación Preclínica de Medicamentos , Imidazoles/farmacología , Ratones , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/genética , Obesidad/prevención & control , Fenotipo , Piridinas/farmacología , Termogénesis
6.
Diabetes ; 67(4): 569-580, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29321171

RESUMEN

Although the central nervous system has been implicated in glucocorticoid-induced gain of fat mass, the underlying mechanisms are poorly understood. The aim of this study was to investigate the possible involvement of hypothalamic serum- and glucocorticoid-regulated kinase 1 (SGK1) in glucocorticoid-increased adiposity. It is well known that SGK1 expression is induced by acute glucocorticoid treatment, but it is interesting that we found its expression to be decreased in the arcuate nucleus of the hypothalamus, including proopiomelanocortin (POMC) neurons, following chronic dexamethasone (Dex) treatment. To study the role of SGK1 in POMC neurons, we produced mice that developed or experienced adult-onset SGK1 deletion in POMC neurons (PSKO). As observed in Dex-treated mice, PSKO mice exhibited increased adiposity and decreased energy expenditure. Mice overexpressing constitutively active SGK1 in POMC neurons consistently had the opposite phenotype and did not experience Dex-increased adiposity. Finally, Dex decreased hypothalamic α-melanocyte-stimulating hormone (α-MSH) content and its precursor Pomc expression via SGK1/FOXO3 signaling, and intracerebroventricular injection of α-MSH or adenovirus-mediated FOXO3 knockdown in the arcuate nucleus largely reversed the metabolic alterations in PSKO mice. These results demonstrate that POMC SGK1/FOXO3 signaling mediates glucocorticoid-increased adiposity, providing new insights into the mechanistic link between glucocorticoids and fat accumulation and important hints for possible treatment targets for obesity.


Asunto(s)
Adiposidad/efectos de los fármacos , Dexametasona/farmacología , Proteína Forkhead Box O3/genética , Glucocorticoides/farmacología , Proteínas Inmediatas-Precoces/genética , Neuronas/efectos de los fármacos , Proopiomelanocortina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Adiposidad/genética , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/efectos de los fármacos , Proteína Forkhead Box O3/metabolismo , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , alfa-MSH/efectos de los fármacos , alfa-MSH/metabolismo
7.
Nutrients ; 9(8)2017 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-28933767

RESUMEN

Hemin is a breakdown product of hemoglobin. It has been reported that the injection of hemin improves lipid metabolism and insulin sensitivity in various genetic models. However, the effect of hemin supplementation in food on lipid metabolism and insulin sensitivity is still unclear, and whether hemin directly affects cellular insulin sensitivity is yet to be elucidated. Here we show that hemin enhances insulin-induced phosphorylation of insulin receptors, Akt, Gsk3ß, FoxO1 and cytoplasmic translocation of FoxO1 in cultured primary hepatocytes under insulin-resistant conditions. Furthermore, hemin diminishes the accumulation of triglyceride and increases in free fatty acid content in primary hepatocytes induced by palmitate. Oral administration of hemin decreases body weight, energy intake, blood glucose and triglyceride levels, and improves insulin and glucose tolerance as well as hepatic insulin signaling and hepatic steatosis in male mice fed a high-fat diet. In addition, hemin treatment decreases the mRNA and protein levels of some hepatic genes involved in lipogenic regulation, fatty acid synthesis and storage, and increases the mRNA level and enzyme activity of CPT1 involved in fatty acid oxidation. These data demonstrate that hemin can improve lipid metabolism and insulin sensitivity in both cultured hepatocytes and mice fed a high-fat diet, and show the potential beneficial effects of hemin from food on lipid and glucose metabolism.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/prevención & control , Hemina/farmacología , Hepatocitos/efectos de los fármacos , Resistencia a la Insulina , Insulina/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Animales , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Metabolismo Energético/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos/genética , Masculino , Ratones Endogámicos C57BL , Ácido Palmítico/farmacología , Fosforilación , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/agonistas , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Triglicéridos/sangre , Pérdida de Peso/efectos de los fármacos
8.
Autophagy ; 13(6): 1088-1089, 2017 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-28350524

RESUMEN

ATF4 (activating transcription factor 4) is an important transcription factor that has many biological functions, while its role in hypothalamic POMC (pro-opiomelanocortin-α) neurons in the regulation of energy homeostasis has not been explored. We recently discovered that mice with an Atf4 deletion specific to POMC neurons (PAKO mice) are lean and have higher energy expenditure. Furthermore, these mice are resistant to high-fat diet (HFD)-induced obesity and obesity-related metabolic disorders. Mechanistically, we found the expression of ATG5 (autophagy-related 5) is upregulated in POMC neurons of PAKO mice, and ATF4 regulates ATG5 expression by binding directly to its promoter. Mice with Atf4 and Atg5 double knockout in POMC neurons have reduced energy expenditure and gain more fat mass compared with PAKO mice under a HFD. Finally, the effect of Atf4 knockout in POMC neurons is possibly mediated by enhanced ATG5-dependent macroautophagy/autophagy and α-melanocyte-stimulating hormone (α-MSH) production in the hypothalamus. Together, this work not only identifies a beneficial role for ATF4 in hypothalamic POMC neurons in the regulation of obesity, but also provides a new potential therapeutic target for obesity and obesity-related metabolic diseases.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Proteína 5 Relacionada con la Autofagia/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Proopiomelanocortina/metabolismo , Transducción de Señal , Animales , Autofagia , Ratones , Modelos Biológicos
9.
Diabetes ; 66(5): 1146-1158, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28213613

RESUMEN

Although many biological functions of activating transcription factor 4 (ATF4) have been identified, a role of hypothalamic ATF4 in the regulation of energy homeostasis is poorly understood. In this study, we showed that hypothalamic proopiomelanocortin (POMC) neuron-specific ATF4 knockout (PAKO) mice are lean and have higher energy expenditure. Furthermore, PAKO mice were resistant to high-fat diet-induced obesity, glucose intolerance, and leptin resistance. Moreover, the expression of autophagy protein 5 (ATG5) was increased or decreased by ATF4 knockdown or overexpression, respectively, and ATF4 inhibited the transcription of ATG5 by binding to the basic zipper-containing protein sites on its promoter. Importantly, mice with double knockout of ATF4 and ATG5 in POMC neurons gained more fat mass and reduced energy expenditure compared with PAKO mice under a high-fat diet. Finally, the effect of ATF4 deletion in POMC neurons was possibly mediated via enhanced ATG5-dependent autophagy and α-melanocyte-stimulating hormone production in the hypothalamus. Taken together, these results identify the beneficial role of hypothalamic ATF4/ATG5 axis in the regulation of energy expenditure, obesity, and obesity-related metabolic disorders, which suggests that ATF4/ATG5 axis in the hypothalamus may be a new potential therapeutic target for treating obesity and obesity-related metabolic diseases.


Asunto(s)
Factor de Transcripción Activador 4/genética , Tejido Adiposo/metabolismo , Proteína 5 Relacionada con la Autofagia/genética , Metabolismo Energético/genética , Hipotálamo/metabolismo , Neuronas/metabolismo , Factor de Transcripción Activador 4/metabolismo , Tejido Adiposo/patología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Proteína 5 Relacionada con la Autofagia/metabolismo , Glucemia/metabolismo , Western Blotting , Corticosterona/metabolismo , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hormona del Crecimiento/metabolismo , Hipotálamo/citología , Insulina/metabolismo , Resistencia a la Insulina , Leptina/metabolismo , Ratones , Ratones Noqueados , Norepinefrina/metabolismo , Tamaño de los Órganos , Proopiomelanocortina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
10.
Diabetes ; 66(3): 640-650, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27993927

RESUMEN

Although many functions of activating transcription factor 4 (ATF4) are identified, a role of ATF4 in the hypothalamus in regulating energy homeostasis is unknown. Here, we generated adult-onset agouti-related peptide neuron-specific ATF4 knockout (AgRP-ATF4 KO) mice and found that these mice were lean, with improved insulin and leptin sensitivity and decreased hepatic lipid accumulation. Furthermore, AgRP-ATF4 KO mice showed reduced food intake and increased energy expenditure, mainly because of enhanced thermogenesis in brown adipose tissue. Moreover, AgRP-ATF4 KO mice were resistant to high-fat diet-induced obesity, insulin resistance, and liver steatosis and maintained at a higher body temperature under cold stress. Interestingly, the expression of FOXO1 was directly regulated by ATF4 via binding to the cAMP-responsive element site on its promoter in hypothalamic GT1-7 cells. Finally, Foxo1 expression was reduced in the arcuate nucleus (ARC) of the hypothalamus of AgRP-ATF4 KO mice, and adenovirus-mediated overexpression of FOXO1 in ARC increased the fat mass in AgRP-ATF4 KO mice. Collectively, our data demonstrate a novel function of ATF4 in AgRP neurons of the hypothalamus in energy balance and lipid metabolism and suggest hypothalamic ATF4 as a potential drug target for treating obesity and its related metabolic disorders.


Asunto(s)
Factor de Transcripción Activador 4/genética , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/genética , Resistencia a la Insulina/genética , Hígado/metabolismo , Neuronas/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Dieta Alta en Grasa , Ingestión de Alimentos/genética , Proteína Forkhead Box O1/metabolismo , Homeostasis , Hipotálamo/citología , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo
11.
Oncotarget ; 7(39): 63679-63689, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27579768

RESUMEN

Substantial studies on fatty acid synthase (FASN) have focused on its role in regulating lipid metabolism and researchers have a great interest in treating cancer with dietary manipulation of amino acids. In the current study, we found that leucine deprivation caused the FASN-dependent anticancer effect. Here we showed that leucine deprivation inhibited cell proliferation and induced apoptosis of MDA-MB-231 and MCF-7 breast cancer cells. In an in vivo tumor xenograft model, the leucine-free diet suppressed the growth of human breast cancer tumors and triggered widespread apoptosis of the cancer cells. Further study indicated that leucine deprivation decreased expression of lipogenic gene FASN in vitro and in vivo. Over-expression of FASN or supplementation of palmitic acid (the product of FASN action) blocked the effects of leucine deprivation on cell proliferation and apoptosis in vitro and in vivo. Moreover, leucine deprivation suppressed the FASN expression via regulating general control non-derepressible (GCN)2 and sterol regulatory element-binding protein 1C (SREBP1C). Taken together, our study represents proof of principle that anticancer effects can be obtained with strategies to deprive tumors of leucine via suppressing FASN expression, which provides important insights in prevention of breast cancer via metabolic intervention.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Acido Graso Sintasa Tipo I/metabolismo , Leucina/metabolismo , Animales , Neoplasias de la Mama/genética , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Ácidos Grasos/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
12.
Open Biol ; 6(8)2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27558934

RESUMEN

Although numerous functions of inositol-requiring enzyme 1α (IRE1α) have been identified, a role of IRE1α in pro-opiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus is largely unknown. Here, we showed that mice lacking IRE1α specifically in POMC neurons (PIKO) are lean and resistant to high-fat diet-induced obesity and obesity-related insulin resistance, liver steatosis and leptin resistance. Furthermore, PIKO mice had higher energy expenditure, probably due to increased thermogenesis in brown adipose tissue. Additionally, α-melanocyte-stimulating hormone production was increased in the hypothalamus of PIKO mice. These results demonstrate that IRE1α in POMC neurons plays a critical role in the regulation of obesity and obesity-related metabolic disorders. Our results also suggest that IRE1α is not only an endoplasmic reticulum stress sensor, but also a new potential therapeutic target for obesity and obesity-related metabolic diseases.


Asunto(s)
Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Neuronas/metabolismo , Obesidad/prevención & control , Proopiomelanocortina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Técnicas de Inactivación de Genes , Hipotálamo/metabolismo , Ratones , Obesidad/inducido químicamente , Termogénesis , Delgadez/genética , alfa-MSH/metabolismo
13.
Diabetologia ; 57(10): 2136-44, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25064125

RESUMEN

AIMS/HYPOTHESIS: Recent studies have revealed the crucial role of the central nervous system (CNS), especially the hypothalamus, in the regulation of insulin sensitivity in peripheral tissues. The aim of our current study was to investigate the possible involvement of hypothalamic prolactin receptors (PRLRs) in the regulation of hepatic insulin sensitivity. METHODS: We employed overexpression of PRLRs in mouse hypothalamus via intracerebroventricular injection of adenovirus expressing PRLR and inhibition of PRLRs via adenovirus expressing short-hairpin RNA (shRNA) specific for PRLRs in vivo. Selective hepatic vagotomy was employed to verify the important role of the vagus nerve in mediating signals from the brain to peripheral organs. In addition, a genetic insulin-resistant animal model, the db/db mouse, was used in our study to investigate the role of hypothalamic PRLRs in regulating whole-body insulin sensitivity. RESULTS: Overexpression of PRLRs in the hypothalamus improved hepatic insulin sensitivity in mice and inhibition of hypothalamic PRLRs had the opposite effect. In addition, we demonstrated that hypothalamic PRLR-improved insulin sensitivity was significantly attenuated by inhibiting the activity of signal transducer and activator of transcription 5 (STAT5) in the CNS and by selective hepatic vagotomy. Finally, overexpression of PRLRs significantly ameliorated insulin resistance in db/db mice. CONCLUSIONS/INTERPRETATION: Our study identifies a novel central pathway involved in the regulation of hepatic insulin sensitivity, mediated by hypothalamic PRLR/STAT5 signalling and the vagus nerve, thus demonstrating an important role for hypothalamic PRLRs under conditions of insulin resistance.


Asunto(s)
Hígado/metabolismo , Receptores de Prolactina/metabolismo , Factor de Transcripción STAT5/metabolismo , Nervio Vago/metabolismo , Animales , Células Cultivadas , Hipotálamo/metabolismo , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Prolactina/genética , Factor de Transcripción STAT5/genética
14.
J Biol Chem ; 289(3): 1779-87, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24302741

RESUMEN

Leptin signaling in the hypothalamus is crucial in energy homeostasis. We have previously shown that dietary deprivation of the essential amino acid leucine in mice stimulates fat loss by increasing energy expenditure. The involvement of leptin signaling in this regulation, however, has not been reported. Here, we show that leucine deprivation promotes leptin signaling in mice maintained on an otherwise normal diet and restores leptin responses in mice maintained on a high fat diet, a regimen known to induce leptin resistance. In addition, we found that leucine deprivation stimulated energy expenditure, and fat loss was largely blocked in db/db mice homozygous for a mutation in leptin receptor and a knock-in mouse line Y3F with abrogation of leptin receptor Tyr(1138)-mediated signal transducer and activator transcript 3 signaling. Overall, our studies describe a novel link between hypothalamic leptin signaling and stimulation of energy expenditure under leucine deprivation.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Leptina/metabolismo , Leucina/deficiencia , Transducción de Señal , Animales , Grasas de la Dieta/farmacología , Leptina/genética , Ratones , Ratones Mutantes , Mutación , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
15.
Diabetes ; 62(7): 2230-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23454693

RESUMEN

Recent studies have revealed that the central nervous system, particularly the hypothalamus, is critical for regulating insulin sensitivity in peripheral tissues. The aim of our current study is to investigate the possible involvement of hypothalamic activating transcription factor 4 (ATF4) in the regulation of insulin sensitivity in the liver. Here, we show that overexpression of ATF4 in the hypothalamus resulting from intracerebroventricular injection of adenovirus expressing ATF4 induces hepatic insulin resistance in mice and that inhibition of hypothalamic ATF4 by intracerebroventricular adenovirus expressing a dominant-negative ATF4 variant has the opposite effect. We also show that hypothalamic ATF4-induced insulin resistance is significantly blocked by selective hepatic vagotomy or by inhibiting activity of the mammalian target of rapamycin (mTOR) downstream target S6K1. Finally, we show that inhibition of hypothalamic ATF4 reverses hepatic insulin resistance induced by acute brain endoplasmic reticulum (ER) stress. Taken together, our study describes a novel central pathway regulating hepatic insulin sensitivity that is mediated by hypothalamic ATF4/mTOR/S6K1 signaling and the vagus nerve and demonstrates an important role for hypothalamic ATF4 in brain ER stress-induced hepatic insulin resistance. These results may lead to the identification of novel therapeutic targets for treating insulin resistance and associated metabolic diseases.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Hipotálamo/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Nervio Vago/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Estrés del Retículo Endoplásmico/fisiología , Masculino , Ratones , Neuronas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Vagotomía
16.
Diabetes ; 61(10): 2461-71, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22787141

RESUMEN

It is well established that the central nervous system (CNS), especially the hypothalamus, plays an important role in regulating energy homeostasis and lipid metabolism. We have previously shown that hypothalamic corticotropin-releasing hormone (CRH) is critical for stimulating fat loss in response to dietary leucine deprivation. The molecular mechanisms underlying the CNS regulation of leucine deprivation-stimulated fat loss are, however, still largely unknown. Here, we used intracerebroventricular injection of adenoviral vectors to identify a novel role for hypothalamic p70 S6 kinase 1 (S6K1), a major downstream effector of the kinase mammalian target of rapamycin, in leucine deprivation stimulation of energy expenditure. Furthermore, we show that the effect of hypothalamic S6K1 is mediated by modulation of Crh expression in a melanocortin-4 receptor-dependent manner. Taken together, our studies provide a new perspective for understanding the regulation of energy expenditure by the CNS and the importance of cross-talk between nutritional control and regulation of endocrine signals.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leucina/deficiencia , Receptor de Melanocortina Tipo 4/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Animales , Hígado , Fosforilación , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Transducción de Señal/fisiología
17.
Mol Endocrinol ; 25(9): 1624-35, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21719534

RESUMEN

We previously showed that leucine deprivation decreases abdominal fat mass largely by increasing energy expenditure, as demonstrated by increased lipolysis in white adipose tissue (WAT) and uncoupling protein 1 (UCP1) expression in brown adipose tissue (BAT). The goal of the present study was to investigate the possible involvement of central nervous system (CNS) in this regulation and elucidate underlying molecular mechanisms. For this purpose, levels of genes and proteins related to lipolysis in WAT and UCP1 expression in BAT were analyzed in wild-type mice after intracerebroventricular administration of leucine or corticotrophin-releasing hormone antibodies, or in mice deleted for three ß-adrenergic receptors, after being maintained on a leucine-deficient diet for 7 d. Here, we show that intracerebroventricular administration of leucine significantly attenuates abdominal fat loss and blocks activation of hormone sensitive lipase in WAT and induction of UCP1 in BAT in leucine-deprived mice. Furthermore, we provide evidence that leucine deprivation stimulates fat loss by increasing expression of corticotrophin-releasing hormone in the hypothalamus via activation of stimulatory G protein/cAMP/protein kinase A/cAMP response element-binding protein pathway. Finally, we show that the effect of leucine deprivation on fat loss is mediated by activation of the sympathetic nervous system. These results suggest that CNS plays an important role in regulating fat loss under leucine deprivation and thereby provide novel and important insights concerning the importance of CNS leucine in the regulation of energy homeostasis.


Asunto(s)
Adiposidad , Hormona Liberadora de Corticotropina/metabolismo , Hipotálamo/metabolismo , Leucina/deficiencia , Sistema Nervioso Simpático/metabolismo , Grasa Abdominal/efectos de los fármacos , Grasa Abdominal/metabolismo , Tejido Adiposo Pardo/metabolismo , Adiposidad/efectos de los fármacos , Animales , Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Hormona Liberadora de Corticotropina/inmunología , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Metabolismo Energético/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/enzimología , Inyecciones Intraventriculares , Leucina/administración & dosificación , Leucina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/deficiencia , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos
18.
Biochem J ; 438(2): 283-9, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21644928

RESUMEN

Chronic feeding of HCD (high-carbohydrate diet) is one of the major contributors to the prevailing of metabolic diseases. ATF4 (activating transcription factor 4) has been shown to play an important role in the regulation of glucose metabolism and obesity development; however, it is unclear how ATF4(-/-) mice respond to HCD. In the present study, we show that 8 weeks of HCD results in significant higher accumulation of TAGs (triacylglycerols) in livers and impairment in glucose tolerance in ATF4(+/+) mice, but not in ATF4(-/-) mice, compared with those on a normal diet. Meanwhile, energy expenditure is further enhanced by HCD in ATF4(-/-) mice. Moreover, we show that ATF4 deficiency suppresses HCD-induced SCD1 (stearoyl-CoA desaturase 1) expression, furthermore, oral supplementation of the main product of SCD1 oleate (18:1) increases TAG accumulation in livers of ATF4(-/-) mice. Taken together, these results suggest that ATF4 deficiency is protective for HCD-induced hepatic steatosis and impairment of glucose tolerance and insulin sensitivity. Furthermore, the resistance to hepatic steatosis is at least in part due to suppression of SCD1 expression under HCD.


Asunto(s)
Factor de Transcripción Activador 4/deficiencia , Dieta , Hígado Graso/prevención & control , Factor de Transcripción Activador 4/metabolismo , Tejido Adiposo/efectos de los fármacos , Administración Oral , Animales , Carbohidratos de la Dieta , Suplementos Dietéticos , Metabolismo Energético/efectos de los fármacos , Hígado Graso/inducido químicamente , Prueba de Tolerancia a la Glucosa , Insulina/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones , Ácido Oléico/administración & dosificación , Ácido Oléico/farmacología , Estearoil-CoA Desaturasa/metabolismo , Triglicéridos/metabolismo
19.
Adv Nutr ; 2(5): 428-39, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22332084

RESUMEN

Nutrition-related health issues have emerged as a major threat to public health since the rebirth of the economy in China starting in the 1980s. To meet this challenge, the Chinese Academy of Sciences established the Institute for Nutritional Sciences (INS) at Shanghai, China ≈ 8 y ago. The mission of the INS is to apply modern technologies and concepts in nutritional research to understand the molecular mechanism and provide means of intervention in the combat against nutrition-related diseases, including type 2 diabetes, metabolic syndrome, obesity, cardiovascular diseases, and many types of cancers. Through diligent and orchestrated efforts by INS scientists, graduate students, and research staff in the past few years, the INS has become the leading institution in China in the areas of basic nutritional research and metabolic regulation. Scientists at the INS have made important progress in many areas, including the characterization of genetic and nutritional properties of the Chinese population, metabolic control associated with nutrient sensing, molecular mechanisms underlying glucose and lipid metabolism, regulation of metabolism by adipokines and inflammatory pathways, disease intervention using functional foods or extracts of Chinese herbs, and many biological studies related to carcinogenesis. The INS will continue its efforts in understanding the optimal nutritional needs for Chinese people and the molecular causes associated with metabolic diseases, thus paving the way for effective and individualized intervention in the future. This review highlights the major research endeavors undertaken by INS scientists in recent years.


Asunto(s)
Academias e Institutos/organización & administración , Estado Nutricional , Salud Pública/métodos , Adipoquinas/genética , Adipoquinas/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , China , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/prevención & control , Medicamentos Herbarios Chinos/uso terapéutico , Regulación de la Expresión Génica , Humanos , Síndrome Metabólico/etiología , Síndrome Metabólico/prevención & control , Neoplasias/etiología , Neoplasias/prevención & control , Trastornos Nutricionales/complicaciones , Trastornos Nutricionales/prevención & control , Obesidad/etiología , Obesidad/prevención & control
20.
Endocrinology ; 145(5): 2221-7, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14764630

RESUMEN

The regulation of TRH gene expression in the paraventricular nucleus of the hypothalamus (PVH) by leptin is critical for normal function of the thyroid axis in rodents and humans. The TRH neuron in the PVH expresses both leptin and melanocortin-4 receptors, suggesting that both signaling systems may regulate TRH gene expression in vivo. Indeed, the TRH promoter responds to both of these signaling pathways in cell culture through identified cis-acting elements, which include signal transducer and activator of transcription (STAT) 3 and cAMP-response element binding protein binding sites that mediate leptin and melanocortin responses, respectively. To determine whether leptin signaling can directly target the TRH promoter in vivo, we developed a chromatin immunoprecipitation assay to use on leptin-treated animals. After a single injection of leptin in fasting animals, we could detect a significant increase in TRH gene expression in the PVH that correlated well with the induction of phosphorylated-STAT3 in the hypothalamus. Furthermore, using a STAT3 antibody, we could immunoprecipitate the STAT-binding site containing regions of both the TRH promoter and the promoter of the suppressor of cytokine signaling-3 gene, another well-defined target of leptin action. In contrast, upstream regions of these promoters that lack STAT sites were not precipitated. Taken together these experiments demonstrate that STAT3 mediates transcriptional effects of leptin in vivo and that the TRH promoter is a likely direct site of leptin action. In addition, these experiments demonstrate that chromatin immunoprecipitation can be used to characterize leptin-signaling in vivo.


Asunto(s)
Leptina/farmacología , Regiones Promotoras Genéticas/genética , Hormona Liberadora de Tirotropina/genética , Animales , Cromatina , ADN/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/química , Técnicas de Inmunoadsorción , Ratones , Ratones Obesos , Obesidad/genética , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/química , Fosforilación , Proteínas Represoras/genética , Factor de Transcripción STAT3 , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/análisis , Transactivadores/metabolismo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA