Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 315(5): E833-E847, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29944388

RESUMEN

ß-Klotho (encoded by Klb) is an obligate coreceptor, mediating both fibroblast growth factor (FGF)15 and FGF21 signaling. Klb-/- mice are refractory to metabolic FGF15 and FGF21 action and exhibit derepressed (increased) bile acid (BA) synthesis. Here, we deeply phenotyped male Klb-/- mice on a pure C57BL/6J genetic background, fed a chow diet focusing on metabolic aspects. This aims to better understand the physiological consequences of concomitant FGF15 and FGF21 signaling deficiency, in particular on the gut-liver axis. Klb-/- mice present permanent growth restriction independent of adiposity and energy balance. Klb-/- mice also exhibit few changes in carbohydrate metabolism, combining normal gluco-tolerance, insulin sensitivity, and fasting response with increased gluconeogenic capacity and decreased glycogen mobilization. Livers of Klb-/- mice reveal pathologic features, including a proinflammatory status and initiation of fibrosis. These defects are associated to a massive shift in BA composition in the enterohepatic system and blood circulation featured by a large excess of microbiota-derived deoxycholic acid, classically known for its genotoxicity in the gastrointestinal tract. In conclusion, ß-Klotho is a gatekeeper of hepatic integrity through direct action (mediating FGF21 anti-inflammatory signaling) and indirect mechanisms (mediating FGF15 signaling that maintains BA level and composition).


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Peso Corporal/fisiología , Tracto Gastrointestinal/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Adiposidad/genética , Animales , Metabolismo Energético/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Gluconeogénesis/fisiología , Cuerpos Cetónicos/sangre , Proteínas Klotho , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Transducción de Señal/fisiología
2.
Mol Genet Metab ; 112(1): 64-72, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24685552

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) are pentameric ligand-gated cation channels well characterized in neuronal signal transmission. Moreover, recent studies have revealed nAChR expression in nonneuronal cell types throughout the body, including tissues involved in metabolism. In the present study, we screen gene expression of nAChR subunits in pancreatic islets and adipose tissues. Mice pancreatic islets present predominant expression of α7 and ß2 nAChR subunits but at a lower level than in central structures. Characterization of glucose and energy homeostasis in α7ß2nAChR(-/-) mice revealed no major defect in insulin secretion and sensitivity but decreased glycemia apparently unrelated to gluconeogenesis or glycogenolysis. α7ß2nAChR(-/-) mice presented an increase in lean and bone body mass and a decrease in fat storage with normal body weight. These observations were associated with elevated spontaneous physical activity in α7ß2nAChR(-/-) mice, mainly due to elevation in fine vertical (rearing) activity while their horizontal (ambulatory) activity remained unchanged. In contrast to α7nAChR(-/-) mice presenting glucose intolerance and insulin resistance associated to excessive inflammation of adipose tissue, the present metabolic phenotyping of α7ß2nAChR(-/-) mice revealed a metabolic improvement possibly linked to the increase in spontaneous physical activity related to central ß2nAChR deficiency.


Asunto(s)
Tejido Adiposo/metabolismo , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Índice Glucémico , Humanos , Insulina/metabolismo , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Receptores Nicotínicos/genética
3.
Pediatr Res ; 75(3): 415-23, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24346113

RESUMEN

BACKGROUND: Lipopolysaccharide (LPS) injection in the corpus callosum (CC) of rat pups results in diffuse white matter injury similar to the main neuropathology of preterm infants. The aim of this study was to characterize the structural and metabolic markers of acute inflammatory injury by high-field magnetic resonance imaging (MRI) magnetic resonance spectroscopy (MRS) in vivo. METHODS: Twenty-four hours after a 1-mg/kg injection of LPS in postnatal day 3 rat pups, diffusion tensor imaging and proton nuclear magnetic spectroscopy ((1)H NMR) were analyzed in conjunction to determine markers of cell death and inflammation using immunohistochemistry and gene expression. RESULTS: MRI and MRS in the CC revealed an increase in lactate and free lipids and a decrease of the apparent diffusion coefficient. Detailed evaluation of the CC showed a marked apoptotic response assessed by fractin expression. Interestingly, the degree of reduction in the apparent diffusion coefficient correlated strongly with the natural logarithm of fractin expression, in the same region of interest. LPS injection further resulted in increased activated microglia clustered in the cingulum, widespread astrogliosis, and increased expression of genes for interleukin (IL)-1, IL-6, and tumor necrosis factor. CONCLUSION: This model was able to reproduce the typical MRI hallmarks of acute diffuse white matter injury seen in preterm infants and allowed the evaluation of in vivo biomarkers of acute neuropathology after inflammatory challenge.


Asunto(s)
Biomarcadores/metabolismo , Encefalitis/diagnóstico , Leucoencefalopatías/diagnóstico , Animales , Imagen de Difusión Tensora , Humanos , Inmunohistoquímica , Recien Nacido Prematuro , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Ácido Láctico/metabolismo , Lipopolisacáridos , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
4.
Pediatr Res ; 75(1-1): 51-61, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24213624

RESUMEN

BACKGROUND: Intrauterine growth restriction (IUGR) is a major risk factor for both perinatal and long-term morbidity. Bovine lactoferrin (bLf) is a major milk glycoprotein considered as a pleiotropic functional nutrient. The impact of maternal supplementation with bLf on IUGR-induced sequelae, including inadequate growth and altered cerebral development, remains unknown. METHODS: IUGR was induced through maternal dexamethasone infusion (100 µg/kg during last gestational week) in rats. Maternal supplementation with bLf (0.85% in food pellet) was provided during both gestation and lactation. Pup growth was monitored, and Pup brain metabolism and gene expression were studied using in vivo (1)H NMR spectroscopy, quantitative PCR, and microarray in the hippocampus at postnatal day (PND)7. RESULTS: Maternal bLf supplementation did not change gestational weight but increased the birth body weight of control pups (4%) with no effect on the IUGR pups. Maternal bLf supplementation allowed IUGR pups to recover a normalized weight at PND21 (weaning) improving catch-up growth. Significantly altered levels of brain metabolites (γ-aminobutyric acid, glutamate, N-acetylaspartate, and N-acetylaspartylglutamate) and transcripts (brain-derived neurotrophic factor (BDNF), divalent metal transporter 1 (DMT-1), and glutamate receptors) in IUGR pups were normalized with maternal bLf supplementation. CONCLUSION: Our data suggest that maternal bLf supplementation is a beneficial nutritional intervention able to revert some of the IUGR-induced sequelae, including brain hippocampal changes.


Asunto(s)
Encéfalo/efectos de los fármacos , Suplementos Dietéticos , Crecimiento/efectos de los fármacos , Lactoferrina/administración & dosificación , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Dexametasona/administración & dosificación , Femenino , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/prevención & control , Expresión Génica/efectos de los fármacos , Lactancia , Lactoferrina/farmacología , Reacción en Cadena de la Polimerasa , Embarazo , Ratas , Aumento de Peso/efectos de los fármacos
5.
Sci Adv ; 10(1): eadj4686, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38170783

RESUMEN

Type 1 diabetes mellitus (T1DM) is characterized by insulin deficiency leading to hyperglycemia and several metabolic defects. Insulin therapy remains the cornerstone of T1DM management, yet it increases the risk of life-threatening hypoglycemia and the development of major comorbidities. Here, we report an insulin signaling-independent pathway able to improve glycemic control in T1DM rodents. Co-treatment with recombinant S100 calcium-binding protein A9 (S100A9) enabled increased adherence to glycemic targets with half as much insulin and without causing hypoglycemia. Mechanistically, we demonstrate that the hyperglycemia-suppressing action of S100A9 is due to a Toll-like receptor 4-dependent increase in glucose uptake in specific skeletal muscles (i.e., soleus and diaphragm). In addition, we found that T1DM mice have abnormal systemic inflammation, which is resolved by S100A9 therapy alone (or in combination with low insulin), hence uncovering a potent anti-inflammatory action of S100A9 in T1DM. In summary, our findings reveal the S100A9-TLR4 skeletal muscle axis as a promising therapeutic target for improving T1DM treatment.


Asunto(s)
Diabetes Mellitus Tipo 1 , Hiperglucemia , Hipoglucemia , Animales , Ratones , Insulina/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Hipoglucemia/complicaciones , Hipoglucemia/tratamiento farmacológico , Hiperglucemia/tratamiento farmacológico , Calgranulina B
6.
Front Endocrinol (Lausanne) ; 13: 971745, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36313762

RESUMEN

Interleukin-18 (IL-18) is a classical member of the IL-1 superfamily of cytokines. As IL-1ß, IL-18 precursor is processed by inflammasome/caspase-1 into a mature and biologically active form. IL-18 binds to its specific receptor composed of two chains (IL-18Rα and IL-18Rß) to trigger a similar intracellular signaling pathway as IL-1, ultimately leading to activation of NF-κB and inflammatory processes. Independently of this IL-1-like signaling, IL-18 also specifically induces IFN-γ production, driving the Th1 immune response. In circulation, IL-18 binds to the IL-18 binding protein (IL-18BP) with high affinity, letting only a small fraction of free IL-18 able to trigger receptor-mediated signaling. In contrast to other IL-1 family members, IL-18 is produced constitutively by different cell types, suggesting implications in normal physiology. If the roles of IL-18 in inflammatory processes and infectious diseases are well described, recent experimental studies in mice have highlighted the action of IL-18 signaling in the control of energy homeostasis, pancreatic islet immunity and liver integrity during nutritional stress. At the same time, clinical observations implicate IL-18 in various metabolic diseases including obesity, type 1 and 2 diabetes and nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH). In the present review, we summarize and discuss both the physiological actions of IL-18 in metabolism and its potential roles in pathophysiological mechanisms leading to the most common human metabolic disorders, such as obesity, diabetes and NAFLD/NASH.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Enfermedad del Hígado Graso no Alcohólico , Humanos , Ratones , Animales , Interleucina-18 , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad
7.
Mol Cell Endocrinol ; 541: 111503, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-34763008

RESUMEN

Pancreatic beta cell dysfunction is a hallmark of type 2 diabetes. Growth differentiation factor 15 (GDF15), which is an energy homeostasis regulator, has been shown to improve several metabolic parameters in the context of diabetes. However, its effects on pancreatic beta-cell remain to be identified. We, therefore, performed experiments using cell models and histological sectioning of wild-type and knock-out GDF15 mice to determine the effect of GDF15 on insulin secretion and cell viability. A bioinformatics analysis was performed to identify GDF15-correlated genes. GDF15 prevents glucotoxicity-mediated altered glucose-stimulated insulin secretion (GSIS) and connexin-36 downregulation. Inhibition of endogenous GDF15 reduced GSIS in cultured mouse beta-cells under standard conditions while it had no impact on GSIS in cells exposed to glucolipotoxicity, which is a diabetogenic condition. Furthermore, this inhibition exacerbated glucolipotoxicity-reduced cell survival. This suggests that endogenous GDF15 in beta-cell is required for cell survival but not GSIS in the context of glucolipotoxicity.


Asunto(s)
Conexinas/genética , Glucosa/efectos adversos , Factor 15 de Diferenciación de Crecimiento/fisiología , Células Secretoras de Insulina/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Conexinas/metabolismo , Citoprotección/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Glucosa/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Secreción de Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína delta-6 de Union Comunicante
8.
Transl Res ; 227: 75-88, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32711187

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the most common hepatic disorder related to type 2 diabetes (T2D). The disease can evolve toward nonalcoholic steatohepatitis (NASH), a state of hepatic inflammation and fibrosis. There is presently no drug that effectively improves and/or prevents NAFLD/NASH/fibrosis. GLP-1 receptor agonists (GLP-1Ra) are effective in treating T2D. As with the endogenous gut incretins, GLP-1Ra potentiate glucose-induced insulin secretion. In addition, GLP-1Ra limit food intake and weight gain, additional beneficial properties in the context of obesity/insulin-resistance. Nevertheless, these pleiotropic effects of GLP-1Ra complicate the elucidation of their direct action on the liver. In the present study, we used the classical methionine-choline deficient (MCD) dietary model to investigate the potential direct hepatic actions of the GLP-1Ra liraglutide. A 4-week infusion of liraglutide (570 µg/kg/day) did not impact body weight, fat accretion or glycemic control in MCD-diet fed mice, confirming the suitability of this model for avoiding confounding factors. Liraglutide treatment did not prevent lipid deposition in the liver of MCD-fed mice but limited the accumulation of C16 and C24-ceramide/sphingomyelin species. In addition, liraglutide treatment alleviated hepatic inflammation (in particular accumulation of M1 pro-inflammatory macrophages) and initiation of fibrosis. Liraglutide also influenced the composition of gut microbiota induced by the MCD-diet. This included recovery of a normal Bacteroides proportion and, among the Erysipelotrichaceae family, a shift between Allobaculum and Turicibacter genera. In conclusion, liraglutide prevents accumulation of C16 and C24-ceramides/sphingomyelins species, inflammation and initiation of fibrosis in MCD-diet-fed mice liver, suggesting beneficial hepatic actions independent of weight loss and global hepatic steatosis.


Asunto(s)
Colina/administración & dosificación , Dieta , Receptor del Péptido 1 Similar al Glucagón/agonistas , Inflamación/prevención & control , Liraglutida/farmacología , Hígado/efectos de los fármacos , Metionina/administración & dosificación , Animales , Liraglutida/uso terapéutico , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico
9.
Sci Rep ; 9(1): 16810, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31728041

RESUMEN

Lipotoxicity is a key player in the pathogenesis of nonalcoholic steatohepatitis (NASH), a progressive subtype of nonalcoholic fatty liver disease (NAFLD). In the present study, we combine histological, transcriptional and lipidomic approaches to dissociate common and specific alterations induced by two classical dietary NASH models (atherogenic (ATH) and methionine/choline deficient (MCD) diet) in C57BL/6J male mice. Despite a similar degree of steatosis, MCD-fed mice showed more pronounced liver damage and a worsened pro-inflammatory and pro-fibrogenic environment than ATH-fed mice. Regarding lipid metabolism, the ATH diet triggered hepatic counter regulatory mechanisms, while the MCD diet worsened liver lipid accumulation by a concomitant increase in lipid import and reduction in lipid export. Liver lipidomics revealed sphingolipid enrichment in both NASH models that was accompanied by an upregulation of the ceramide biosynthesis pathway and a significant rise in dihydroceramide levels. In contrast, the phospholipid composition was not substantially altered by the ATH diet, whereas the livers of MCD-fed mice presented a reduced phosphatidylcholine to phosphatidylethanolamine (PC/PE) ratio and a strong depletion in phospholipids containing the sum of 34-36 carbons in their fatty acid chains. Therefore, the assessment of liver damage at the histological and transcriptional level combined with a lipidomic analysis reveals sphingolipids as shared mediators in liver lipotoxicity and pathogenesis of NASH.


Asunto(s)
Aterosclerosis/metabolismo , Dieta/efectos adversos , Redes Reguladoras de Genes/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Esfingolípidos/metabolismo , Animales , Aterosclerosis/inducido químicamente , Aterosclerosis/genética , Colina/química , Dieta Aterogénica/efectos adversos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Lipidómica , Masculino , Metionina/deficiencia , Ratones , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/genética , Fosfatidilcolinas/metabolismo
10.
Endocrinology ; 149(12): 6289-99, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18687784

RESUMEN

Despite medical advice, 20-30% of female smokers continue to smoke during pregnancy. Epidemiological studies have associated maternal smoking with increased risk of obesity and type-2 diabetes in the offspring. In the present study, we investigated the impact of prenatal nicotine exposure (3 mg/kg in Sprague Dawley rats via osmotic Alzet minipumps) on the early endocrine pancreas and adipose tissue development in rat pups before weaning. Body weight, fat deposition, food intake and food efficiency, cold tolerance, spontaneous physical activity, glucose utilization, and insulin sensitivity were also examined at adulthood. Prenatal nicotine exposure led to a decrease in endocrine pancreatic islet size and number at 7 d of life (postnatal d 7), which corroborates with a decrease in gene expression of specific transcription factors such as pancreatic and duodenal homeobox 1, Pax-6, Nkx6.1, and of hormones such as insulin and glucagon. The prenatal nicotine exposure also led to an increase in epididymal white adipose tissue weight at weaning (postnatal d 21), and marked hypertrophy of adipocytes, with increased gene expression of proadipogenic transcription factors such as CAAT-enhancer-binding protein-alpha, peroxisome proliferator activated receptor-gamma, and sterol regulatory element binding protein-1C. These early tissue alterations led to significant metabolic consequences, as shown by increased body weight and fat deposition, increased food efficiency on high-fat diet, cold intolerance, reduced physical activity, and glucose intolerance combined with insulin resistance observed at adulthood. These results prove a direct association between fetal nicotine exposure and offspring metabolic syndrome with early signs of dysregulations of adipose tissue and pancreatic development.


Asunto(s)
Peso Corporal/efectos de los fármacos , Glucosa/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Nicotina/toxicidad , Efectos Tardíos de la Exposición Prenatal/metabolismo , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/metabolismo , Animales , Calorimetría , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Femenino , Estimulantes Ganglionares/administración & dosificación , Estimulantes Ganglionares/toxicidad , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Islotes Pancreáticos/crecimiento & desarrollo , Masculino , Nicotina/administración & dosificación , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley
11.
Endocr Rev ; 39(6): 960-989, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30124818

RESUMEN

Discovered 20 years ago, fibroblast growth factor (FGF)19, and its mouse ortholog FGF15, were the first members of a new subfamily of FGFs able to act as hormones. During fetal life, FGF15/19 is involved in organogenesis, affecting the development of the ear, eye, heart, and brain. At adulthood, FGF15/19 is mainly produced by the ileum, acting on the liver to repress hepatic bile acid synthesis and promote postprandial nutrient partitioning. In rodents, pharmacologic doses of FGF19 induce the same antiobesity and antidiabetic actions as FGF21, with these metabolic effects being partly mediated by the brain. However, activation of hepatocyte proliferation by FGF19 has long been a challenge to its therapeutic use. Recently, genetic reengineering of the molecule has resolved this issue. Despite a global overlap in expression pattern and function, murine FGF15 and human FGF19 exhibit several differences in terms of regulation, molecular structure, signaling, and biological properties. As most of the knowledge originates from the use of FGF19 in murine models, differences between mice and humans in the biology of FGF15/19 have to be considered for a successful translation from bench to bedside. This review summarizes the basic knowledge concerning FGF15/19 in mice and humans, with a special focus on regulation of production, morphogenic properties, hepatocyte growth, bile acid homeostasis, as well as actions on glucose, lipid, and energy homeostasis. Moreover, implications and therapeutic perspectives concerning FGF19 in human diseases (including obesity, type 2 diabetes, hepatic steatosis, biliary disorders, and cancer) are also discussed.


Asunto(s)
Enfermedades de las Vías Biliares/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hígado Graso/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/uso terapéutico , Neoplasias/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Animales , Humanos
12.
Thyroid ; 28(6): 780-798, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29742982

RESUMEN

BACKGROUND: The thyroid gland has a special relationship with oxidative stress. While generation of oxidative substances is part of normal iodide metabolism during thyroid hormone synthesis, the gland must also defend itself against excessive oxidation in order to maintain normal function. Antioxidant and detoxification enzymes aid thyroid cells to maintain homeostasis by ameliorating oxidative insults, including during exposure to excess iodide, but the factors that coordinate their expression with the cellular redox status are not known. The antioxidant response system comprising the ubiquitously expressed NFE2-related transcription factor 2 (Nrf2) and its redox-sensitive cytoplasmic inhibitor Kelch-like ECH-associated protein 1 (Keap1) defends tissues against oxidative stress, thereby protecting against pathologies that relate to DNA, protein, and/or lipid oxidative damage. Thus, it was hypothesized that Nrf2 should also have important roles in maintaining thyroid homeostasis. METHODS: Ubiquitous and thyroid-specific male C57BL6J Nrf2 knockout (Nrf2-KO) mice were studied. Plasma and thyroids were harvested for evaluation of thyroid function tests by radioimmunoassays and of gene and protein expression by real-time polymerase chain reaction and immunoblotting, respectively. Nrf2-KO and Keap1-KO clones of the PCCL3 rat thyroid follicular cell line were generated using CRISPR/Cas9 technology and were used for gene and protein expression studies. Software-predicted Nrf2 binding sites on the thyroglobulin enhancer were validated by site-directed in vitro mutagenesis and chromatin immunoprecipitation. RESULTS: The study shows that Nrf2 mediates antioxidant transcriptional responses in thyroid cells and protects the thyroid from oxidation induced by iodide overload. Surprisingly, it was also found that Nrf2 has a dramatic impact on both the basal abundance and the thyrotropin-inducible intrathyroidal abundance of thyroglobulin (Tg), the precursor protein of thyroid hormones. This effect is mediated by cell-autonomous regulation of Tg gene expression by Nrf2 via its direct binding to two evolutionarily conserved antioxidant response elements in an upstream enhancer. Yet, despite upregulating Tg levels, Nrf2 limits Tg iodination both under basal conditions and in response to excess iodide. CONCLUSIONS: Nrf2 exerts pleiotropic roles in the thyroid gland to couple cell stress defense mechanisms to iodide metabolism and the thyroid hormone synthesis machinery, both under basal conditions and in response to excess iodide.


Asunto(s)
Antioxidantes/metabolismo , Yodo/sangre , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Tiroglobulina/sangre , Glándula Tiroides/metabolismo , Animales , Línea Celular , Citoplasma/metabolismo , Homeostasis , Humanos , Yoduros/química , Proteína 1 Asociada A ECH Tipo Kelch/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo , Oxígeno/química , Regiones Promotoras Genéticas , Ratas , Especies Reactivas de Oxígeno/metabolismo , Tiroglobulina/genética , Hormonas Tiroideas/metabolismo
13.
JCI Insight ; 2(8)2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28422755

RESUMEN

ß-Klotho (encoded by Klb) is the obligate coreceptor mediating FGF21 and FGF15/19 signaling. Klb-/- mice are refractory to beneficial action of pharmacological FGF21 treatment including stimulation of glucose utilization and thermogenesis. Here, we investigated the energy homeostasis in Klb-/- mice on high-fat diet in order to better understand the consequences of abrogating both endogenous FGF15/19 and FGF21 signaling during caloric overload. Surprisingly, Klb-/- mice are resistant to diet-induced obesity (DIO) owing to enhanced energy expenditure and BAT activity. Klb-/- mice exhibited not only an increase but also a shift in bile acid (BA) composition featured by activation of the classical (neutral) BA synthesis pathway at the expense of the alternative (acidic) pathway. High hepatic production of cholic acid (CA) results in a large excess of microbiota-derived deoxycholic acid (DCA). DCA is specifically responsible for activating the TGR5 receptor that stimulates BAT thermogenic activity. In fact, combined gene deletion of Klb and Tgr5 or antibiotic treatment abrogating bacterial conversion of CA into DCA both abolish DIO resistance in Klb-/- mice. These results suggested that DIO resistance in Klb-/- mice is caused by high levels of DCA, signaling through the TGR5 receptor. These data also demonstrated that gut microbiota can regulate host thermogenesis via conversion of primary into secondary BA. Pharmacologic or nutritional approaches to selectively modulate BA composition may be a promising target for treating metabolic disorders.

14.
EMBO Mol Med ; 9(10): 1379-1397, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28754744

RESUMEN

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic form of isolated gonadotropin-releasing hormone (GnRH) deficiency caused by mutations in > 30 genes. Fibroblast growth factor receptor 1 (FGFR1) is the most frequently mutated gene in CHH and is implicated in GnRH neuron development and maintenance. We note that a CHH FGFR1 mutation (p.L342S) decreases signaling of the metabolic regulator FGF21 by impairing the association of FGFR1 with ß-Klotho (KLB), the obligate co-receptor for FGF21. We thus hypothesized that the metabolic FGF21/KLB/FGFR1 pathway is involved in CHH Genetic screening of 334 CHH patients identified seven heterozygous loss-of-function KLB mutations in 13 patients (4%). Most patients with KLB mutations (9/13) exhibited metabolic defects. In mice, lack of Klb led to delayed puberty, altered estrous cyclicity, and subfertility due to a hypothalamic defect associated with inability of GnRH neurons to release GnRH in response to FGF21. Peripheral FGF21 administration could indeed reach GnRH neurons through circumventricular organs in the hypothalamus. We conclude that FGF21/KLB/FGFR1 signaling plays an essential role in GnRH biology, potentially linking metabolism with reproduction.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Síndrome de Kallmann/genética , Proteínas de la Membrana/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Células COS , Caenorhabditis elegans/genética , Chlorocebus aethiops , Estudios de Cohortes , Femenino , Factores de Crecimiento de Fibroblastos/genética , Hormona Liberadora de Gonadotropina/genética , Células HEK293 , Humanos , Hipotálamo/metabolismo , Proteínas Klotho , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Neuronas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
15.
Diabetes ; 54(12): 3503-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16306368

RESUMEN

Interleukin (IL)-1 is a regulator of inflammation but is also implicated in the control of energy homeostasis. Because the soluble IL-1 receptor antagonist (IL-1Ra) is markedly increased in the serum of obese patients and is overexpressed in white adipose tissue in obesity, we studied the metabolic consequences of genetic IL-1Ra ablation in mice. We have shown that IL-1Ra-/- mice have a lean phenotype due to decreased fat mass, related to a defect in adipogenesis and increased energy expenditure. The adipocytes were smaller in these animals, and the expression of genes involved in adipogenesis was reduced. Energy expenditure as measured by indirect calorimetry was elevated, and weight loss in response to a 24-h fast was increased in IL-1Ra-/- animals compared with wild-type mice. Lipid oxidation of IL-1Ra-/- mice was higher during the light period, reflecting their reduction in diurnal food intake. Interestingly, IL-1Ra-/- and IL-1Ra+/- mice presented an attenuation in high-fat diet-induced caloric hyperphagia, indicating a better adaptation to hypercaloric alimentation, which is in line with the role of IL-1Ra as a mediator of leptin resistance. Taken together, we show that IL-1Ra is an important regulator of adipogenesis, food intake, and energy expenditure.


Asunto(s)
Tejido Adiposo/anatomía & histología , Ingestión de Energía , Metabolismo Energético , Sialoglicoproteínas/deficiencia , Sialoglicoproteínas/metabolismo , Pérdida de Peso/fisiología , Tejido Adiposo/fisiología , Animales , Composición Corporal , Amplificación de Genes , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1/sangre , Interleucina-6/sangre , Intrones , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Sialoglicoproteínas/genética , Aumento de Peso
16.
Endocrinology ; 157(10): 3800-3808, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27471776

RESUMEN

Type 1 diabetes (T1DM) results from destruction of most insulin-secreting pancreatic ß-cells. The persistence of ß-cells decades after the onset of the disease indicates that the resistance of individual cells to the autoimmune insult is heterogeneous and might depend on the metabolic status of a cell at a given moment. The aim of this study is to investigate whether activation of nicotinic acetylcholine receptors (nACh-Rs) could increase ß-cell resistance against the adverse environment prevailing at the onset of T1DM. Here, we show that nACh-R activation by nicotine and choline, 2 agonists of the receptor, decreases murine and human ß-cell apoptosis induced by proinflammatory cytokines known to be present in the islet environment at the onset of T1DM. The protective mechanism activated by nicotine and choline involves attenuation of mitochondrial outer membrane permeabilization via modulation of endoplasmic reticulum stress, of the activity of B-cell lymphoma 2 family proteins and cytoplasmic calcium levels. Local inflammation and endoplasmic reticulum stress being key determinants of ß-cell death in T1DM, we conclude that pharmacological activation of nACh-R could represent a valuable therapeutic option in the modulation of ß-cell death in T1DM.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Islotes Pancreáticos/efectos de los fármacos , Agonistas Nicotínicos/uso terapéutico , Animales , Calcio/metabolismo , Caspasa 3/metabolismo , Colina , Técnicas de Cultivo , Evaluación Preclínica de Medicamentos , Estrés del Retículo Endoplásmico , Femenino , Regulación de la Expresión Génica , Genes bcl-2 , Humanos , Ratones , Ratones Endogámicos C57BL , Membranas Mitocondriales/metabolismo , Nicotina , Agonistas Nicotínicos/farmacología , Óxido Nítrico/metabolismo
17.
Diabetes ; 52(5): 1104-10, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12716739

RESUMEN

The secreted form of the interleukin-1 receptor antagonist (IL-1Ra) is an acute-phase protein intervening in the counterregulation of inflammatory processes. We previously showed that this cytokine antagonist is upregulated in the serum of obese patients, correlating with BMI and insulin resistance. In this study, we examined the expression pattern of IL-1Ra and showed that it is highly expressed not only in liver and spleen, but also in white adipose tissue (WAT), where it is upregulated in obesity. In WAT of obese humans, IL-1Ra was also markedly increased. Moreover, human WAT explants secreted IL-1Ra into the medium, a process that could be stimulated fivefold by interferon-beta. Finally, lipopolysaccharide administration induced a long-lasting expression of IL-1Ra in mouse WAT, suggesting that adipose tissue is an important source of IL-1Ra in both obesity and inflammation. In summary, we demonstrated that WAT is one of the most important sources of IL-1Ra quantitatively, suggesting that this tissue could represent a novel target for anti-inflammatory treatment. Moreover, it can be speculated that IL-1Ra, whose production is markedly increased in WAT in obese individuals, contributes further to weight gain because of its endocrine and paracrine effects on the hypothalamus and adipocytes, respectively.


Asunto(s)
Tejido Adiposo/fisiopatología , Regulación de la Expresión Génica , Obesidad/inmunología , Sialoglicoproteínas/genética , Tejido Adiposo/efectos de los fármacos , Animales , Humanos , Inflamación/genética , Inflamación/inmunología , Interferón beta/farmacología , Proteína Antagonista del Receptor de Interleucina 1 , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/genética , Técnicas de Cultivo de Órganos , ARN Mensajero/genética , Valores de Referencia , Sialoglicoproteínas/efectos de los fármacos , Sialoglicoproteínas/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/fisiología
18.
PLoS One ; 10(7): e0132136, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26244509

RESUMEN

Di-(2-ethylhexyl)phtalate (DEHP) is a plasticizer with endocrine disrupting properties found ubiquitously in the environment and altering reproduction in rodents. Here we investigated the impact of prenatal exposure to DEHP on spermatogenesis and DNA sperm methylation in two distinct, selected, and sequenced mice strains. FVB/N and C57BL/6J mice were orally exposed to 300 mg/kg/day of DEHP from gestation day 9 to 19. Prenatal DEHP exposure significantly decreased spermatogenesis in C57BL/6J (fold-change = 0.6, p-value = 8.7*10-4), but not in FVB/N (fold-change = 1, p-value = 0.9). The number of differentially methylated regions (DMRs) by DEHP-exposure across the entire genome showed increased hyper- and decreased hypo-methylation in C57BL/6J compared to FVB/N. At the promoter level, three important subsets of genes were massively affected. Promoters of vomeronasal and olfactory receptors coding genes globally followed the same trend, more pronounced in the C57BL/6J strain, of being hyper-methylated in DEHP related conditions. In contrast, a large set of micro-RNAs were hypo-methylated, with a trend more pronounced in the FVB/N strain. We additionally analyze both the presence of functional genetic variations within genes that were associated with the detected DMRs and that could be involved in spermatogenesis, and DMRs related with the DEHP exposure that affected both strains in an opposite manner. The major finding in this study indicates that prenatal exposure to DEHP can decrease spermatogenesis in a strain-dependent manner and affects sperm DNA methylation in promoters of large sets of genes putatively involved in both sperm chemotaxis and post-transcriptional regulatory mechanisms.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Dietilhexil Ftalato/efectos adversos , Disruptores Endocrinos/efectos adversos , Plastificantes/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Espermatogénesis/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Embarazo , Especificidad de la Especie
19.
J Clin Endocrinol Metab ; 89(6): 2652-8, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15181037

RESUMEN

Adipose tissue is the source of production and site of action of several pro- and antiinflammatory cytokines. We have recently shown that white adipose tissue (WAT) is a major producer of the antiinflammatory IL-1 receptor antagonist (IL-1Ra). Because IL-1Ra serum levels are elevated 7-fold in human obesity and an excess of this protein has been implicated in the acquired resistance to leptin and insulin, we investigated the regulation of IL-1Ra in human WAT. We demonstrate that IL-1Ra is mainly produced by adipocytes, rather than the stromal fraction of WAT, and that IL-1alpha and beta, as well as interferon-beta (IFN-beta), strongly up-regulate the expression and secretion of IL-1Ra in WAT. Moreover, human WAT expresses the receptors and proteins known to be required for the action of IL-1 (IL-1 receptor type I, IL-1 receptor accessory protein) and IFN-beta (IFN-alpha/beta receptor subunits 1 and 2). Finally, human WAT actively secretes these regulatory cytokines, suggesting that they up-regulate IL-1Ra through a local autocrine/paracrine action, which is hypothesized to play a regulatory role in adipogenesis and metabolism.


Asunto(s)
Tejido Adiposo/metabolismo , Factores Inmunológicos/farmacología , Interferón beta/farmacología , Interleucina-1/farmacología , Interleucina-4/farmacología , Sialoglicoproteínas/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Expresión Génica/fisiología , Humanos , Factores Inmunológicos/genética , Interferón beta/genética , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1/genética , Interleucina-4/genética , Sialoglicoproteínas/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
20.
J Clin Endocrinol Metab ; 88(12): 5921-6, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14671191

RESUMEN

Riboflavin-responsive, multiple acylcoenzyme A dehydrogenase deficiency (RR-MAD), a lipid storage myopathy, is characterized by, among others, a decrease in fatty acid (FA) beta-oxidation capacity. Muscle uncoupling protein 3 (UCP3) is up-regulated under conditions that either increase the levels of circulating free FA and/or decrease FA beta-oxidation. Using a relatively large cohort of seven RR-MAD patients, we aimed to better characterize the metabolic disturbances of this disease and to explore the possibility that it might increase UCP3 expression. A battery of biochemical and molecular tests were performed, which demonstrated decreases in FA beta-oxidation and in the activities of respiratory chain complexes I and II. These metabolic alterations were associated with increases of 3.1- and 1.7-fold in UCP3 mRNA and protein expression, respectively. All parameters were restored to control values after riboflavin treatment. We postulate that the up-regulation of UCP3 in RR-MAD is due to the accumulation of muscle FA/acylCoA. RR-MAD is an optimal model to support the hypothesis that UCP3 is involved in the outward translocation of an excess of FA from the mitochondria and to show that, in humans, the effects of FA on UCP3 expression are direct and independent of fatty acid beta-oxidation.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Proteínas Portadoras/metabolismo , Ácidos Grasos/metabolismo , Riboflavina/uso terapéutico , Adolescente , Adulto , Proteínas Portadoras/genética , Estudios de Cohortes , Complejo I de Transporte de Electrón/deficiencia , Complejo II de Transporte de Electrones/deficiencia , Femenino , Humanos , Canales Iónicos , Metabolismo de los Lípidos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Oxidación-Reducción , ARN Mensajero/metabolismo , Proteína Desacopladora 3
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA