Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
2.
Cell ; 142(2): 309-19, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20655471

RESUMEN

Global energy balance in mammals is controlled by the actions of circulating hormones that coordinate fuel production and utilization in metabolically active tissues. Bone-derived osteocalcin, in its undercarboxylated, hormonal form, regulates fat deposition and is a potent insulin secretagogue. Here, we show that insulin receptor (IR) signaling in osteoblasts controls osteoblast development and osteocalcin expression by suppressing the Runx2 inhibitor Twist2. Mice lacking IR in osteoblasts have low circulating undercarboxylated osteocalcin and reduced bone acquisition due to decreased bone formation and deficient numbers of osteoblasts. With age, these mice develop marked peripheral adiposity and hyperglycemia accompanied by severe glucose intolerance and insulin resistance. The metabolic abnormalities in these mice are improved by infusion of undercarboxylated osteocalcin. These results indicate the existence of a bone-pancreas endocrine loop through which insulin signaling in the osteoblast ensures osteoblast differentiation and stimulates osteocalcin production, which in turn regulates insulin sensitivity and pancreatic insulin secretion.


Asunto(s)
Osteoblastos/metabolismo , Osteogénesis , Receptor de Insulina/metabolismo , Adiposidad , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Resistencia a la Insulina , Masculino , Ratones , Osteoblastos/citología , Osteocalcina/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Proteína 1 Relacionada con Twist/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 324(6): L836-L848, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37070742

RESUMEN

Right ventricular (RV) adaptation is the principal determinant of outcomes in pulmonary arterial hypertension (PAH), however, RV function is challenging to assess. RV responses to hemodynamic stressors are particularly difficult to interrogate without invasive testing. This study sought to identify metabolomic markers of in vivo right ventricular function and exercise performance in PAH. Consecutive subjects with PAH (n = 23) underwent rest and exercise right heart catheterization with multibeat pressure volume loop analysis. Pulmonary arterial blood was collected at rest and during exercise. Mass spectrometry-based targeted metabolomics were performed, and metabolic associations with hemodynamics and comprehensive measures of RV function were determined using sparse partial least squares regression. Metabolite profiles were compared with N-terminal prohormone of B-type natriuretic peptide (NT-proBNP) measurements for accuracy in modeling ventriculo-arterial parameters. Thirteen metabolites changed in abundance with exercise, including metabolites reflecting increased arginine bioavailability, precursors of catecholamine and nucleotide synthesis, and branched-chain amino acids. Higher resting arginine bioavailability predicted more favorable exercise hemodynamics and pressure-flow relationships. Subjects with more severe PAH augmented arginine bioavailability with exercise to a greater extent than subjects with less severe PAH. We identified relationships between kynurenine pathway metabolism and impaired ventriculo-arterial coupling, worse RV diastolic function, lower RV contractility, diminished RV contractility with exercise, and RV dilation with exercise. Metabolite profiles outperformed NT-proBNP in modeling RV contractility, diastolic function, and exercise performance. Specific metabolite profiles correspond to RV functional measurements only obtainable via invasive pressure-volume loop analysis and predict RV responses to exercise. Metabolic profiling may inform discovery of RV functional biomarkers.NEW & NOTEWORTHY In this cohort of patients with pulmonary arterial hypertension (PAH), we investigate metabolomic associations with comprehensive right ventricular (RV) functional measurements derived from multibeat RV pressure-volume loop analysis. Our results show that tryptophan metabolism, particularly the kynurenine pathway, is linked to intrinsic RV function and PAH pathobiology. Findings also highlight the importance of arginine bioavailability in the cardiopulmonary system's response to exercise stress. Metabolite profiles selected via unbiased analysis outperformed N-terminal prohormone of B-type natriuretic peptide (NT-proBNP) in predicting load-independent measures of RV function at rest and cardiopulmonary system performance under stress. Overall, this work suggests the potential for select metabolites to function as disease-specific biomarkers, offers insights into PAH pathobiology, and informs discovery of potentially targetable RV-centric pathways.


Asunto(s)
Hipertensión Arterial Pulmonar , Disfunción Ventricular Derecha , Humanos , Péptido Natriurético Encefálico , Función Ventricular Derecha/fisiología , Quinurenina , Hipertensión Pulmonar Primaria Familiar , Biomarcadores , Arginina
4.
Am J Physiol Lung Cell Mol Physiol ; 325(5): L617-L627, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37786941

RESUMEN

Understanding metabolic evolution underlying pulmonary arterial hypertension (PAH) development may clarify pathobiology and reveal disease-specific biomarkers. Patients with systemic sclerosis (SSc) are regularly surveilled for PAH, presenting an opportunity to examine metabolic change as disease develops in an at-risk cohort. We performed mass spectrometry-based metabolomics on longitudinal serum samples collected before and near SSc-PAH diagnosis, compared with time-matched SSc subjects without PAH, in a SSc surveillance cohort. We validated metabolic differences in a second cohort and determined metabolite-phenotype relationships. In parallel, we performed serial metabolomic and hemodynamic assessments as the disease developed in a preclinical model. For differentially expressed metabolites, we investigated corresponding gene expression in human and rodent PAH lungs. Kynurenine and its ratio to tryptophan (kyn/trp) increased over the surveillance period in patients with SSc who developed PAH. Higher kyn/trp measured two years before diagnostic right heart catheterization increased the odds of SSc-PAH diagnosis (OR 1.57, 95% CI 1.05-2.36, P = 0.028). The slope of kyn/trp rise during SSc surveillance predicted PAH development and mortality. In both clinical and experimental PAH, higher kynurenine pathway metabolites correlated with adverse pulmonary vascular and RV measurements. In human and rodent PAH lungs, expression of TDO2, which encodes tryptophan 2,3 dioxygenase (TDO), a protein that catalyzes tryptophan conversion to kynurenine, was significantly upregulated and tightly correlated with pulmonary hypertensive features. Upregulated kynurenine pathway metabolism occurs early in PAH, localizes to the lung, and may be modulated by TDO2. Kynurenine pathway metabolites may be candidate PAH biomarkers and TDO warrants exploration as a potential novel therapeutic target.NEW & NOTEWORTHY Our study shows an early increase in kynurenine pathway metabolism in at-risk subjects with systemic sclerosis who develop pulmonary arterial hypertension (PAH). We show that kynurenine pathway upregulation precedes clinical diagnosis and that this metabolic shift is associated with increased disease severity and shorter survival times. We also show that gene expression of TDO2, an enzyme that generates kynurenine from tryptophan, rises with PAH development.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Esclerodermia Sistémica , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/complicaciones , Quinurenina , Triptófano , Esclerodermia Sistémica/complicaciones , Hipertensión Pulmonar Primaria Familiar , Biomarcadores
5.
FASEB J ; 36(6): e22347, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35579659

RESUMEN

C1q/TNF-related proteins (CTRP1-15) constitute a conserved group of secreted proteins of the C1q family with diverse functions. In vitro studies have shown that CTRP11/C1QL4 can inhibit adipogenesis, antagonize myoblast fusion, and promote testosterone synthesis and secretion. Whether CTRP11 is required for these processes in vivo remains unknown. Here, we show that knockout (KO) mice lacking CTRP11 have normal skeletal muscle mass and function, and testosterone level, suggesting that CTRP11 is dispensable for skeletal muscle development and testosterone production. We focused our analysis on whether this nutrient-responsive secreted protein plays a role in controlling sugar and fat metabolism. At baseline when mice are fed a standard chow, CTRP11 deficiency affects metabolic parameters in a sexually dimorphic manner. Only Ctrp11-KO female mice have significantly higher fasting serum ketones and reduced physical activity. In the refeeding phase following food withdrawal, Ctrp11-KO female mice have reduced food intake and increased metabolic rate and energy expenditure, highlighting CTRP11's role in fasting-refeeding response. When challenged with a high-fat diet to induce obesity and metabolic dysfunction, CTRP11 deficiency modestly exacerbates obesity-induced glucose intolerance, with more pronounced effects seen in Ctrp11-KO male mice. Switching to a low-fat diet after obesity induction results in greater fat loss in wild type relative to KO male mice, suggesting impaired response to obesity reversal and reduced metabolic flexibility in the absence of CTRP11. Collectively, our data provide genetic evidence for novel sex-dependent metabolic regulation by CTRP11, but note the overall modest contribution of CTRP11 to systemic energy homeostasis.


Asunto(s)
Complemento C1/metabolismo , Complemento C1q , Dieta Alta en Grasa , Animales , Complemento C1q/metabolismo , Metabolismo Energético/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Testosterona
6.
Am J Physiol Endocrinol Metab ; 322(6): E480-E493, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35403439

RESUMEN

Secreted proteins of the C1q/TNF-related protein (CTRP) family play diverse functions in different organ systems. In the brain, CTRP14/C1QL1 is required for the proper establishment and maintenance of synapses between climbing fibers and cerebellar Purkinje cells. Beyond the central nervous system, the function of CTRP14 is largely unknown. A recent genome-wide association study has implicated CTRP14/C1QL1 as a candidate gene associated with total body fat mass. Here, we explored the potential metabolic roles of CTRP14. We show that Ctrp14 expression in peripheral tissues is dynamically regulated by fasting-refeeding and high-fat feeding. In the chow-fed basal state, Ctrp14 deletion modestly reduces glucose tolerance in knockout (KO) male mice and affects physical activity in a sex- and nutritional state-dependent manner. In the ad libitum fed state, Ctrp14 KO male mice have lower physical activity. In contrast, female KO mice have increased physical activity in the fasted and refed states. In response to an obesogenic diet, CTRP14-deficient mice of either sex gained similar weight and are indistinguishable from wild-type littermates in body composition, lipid profiles, and insulin sensitivity. Ambulatory activity, however, is reduced in Ctrp14 KO male mice. Food intake is also reduced in Ctrp14 KO male mice in the refed period following food deprivation. Meal pattern analyses indicate that decreased caloric intake from fasting to refeeding is due, in part, to smaller meal size. We conclude that CTRP14 is largely dispensable for metabolic homeostasis, but highlight context-dependent and sexually dimorphic metabolic responses of Ctrp14 deletion affecting physical activity and ingestive behaviors.NEW & NOTEWORTHY CTRP14 is a secreted protein whose function in the peripheral tissues is largely unknown. We show that the expression of Ctrp14 in peripheral tissues is regulated by metabolic and nutritional state. We generated mice lacking CTRP14 and show that CTRP14 deficiency alters physical activity and food intake in response to fasting and refeeding. Our data has provided new and valuable information on the physiological function of CTRP14.


Asunto(s)
Ayuno , Resistencia a la Insulina , Animales , Complemento C1q/genética , Dieta Alta en Grasa , Ingestión de Alimentos/genética , Femenino , Estudio de Asociación del Genoma Completo , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Noqueados
7.
Neurobiol Dis ; 171: 105808, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35779777

RESUMEN

Wallerian degeneration (WD) is a conserved axonal self-destruction program implicated in several neurological diseases. WD is driven by the degradation of the NAD+ synthesizing enzyme NMNAT2, the buildup of its substrate NMN, and the activation of the NAD+ degrading SARM1, eventually leading to axonal fragmentation. The regulation and amenability of these events to therapeutic interventions remain unclear. Here we explored pharmacological strategies that modulate NMN and NAD+ metabolism, namely the inhibition of the NMN-synthesizing enzyme NAMPT, activation of the nicotinic acid riboside (NaR) salvage pathway and inhibition of the NMNAT2-degrading DLK MAPK pathway in an axotomy model in vitro. Results show that NAMPT and DLK inhibition cause a significant but time-dependent delay of WD. These time-dependent effects are related to NMNAT2 degradation and changes in NMN and NAD+ levels. Supplementation of NAMPT inhibition with NaR has an enhanced effect that does not depend on timing of intervention and leads to robust protection up to 4 days. Additional DLK inhibition extends this even further to 6 days. Metabolite analyses reveal complex effects indicating that NAMPT and MAPK inhibition act by reducing NMN levels, ameliorating NAD+ loss and suppressing SARM1 activity. Finally, the axonal NAD+/NMN ratio is highly predictive of cADPR levels, extending previous cell-free evidence on the allosteric regulation of SARM1. Our findings establish a window of axon protection extending several hours following injury. Moreover, we show prolonged protection by mixed treatments combining MAPK and NAMPT inhibition that proceed via complex effects on NAD+ metabolism and inhibition of SARM1.


Asunto(s)
Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida-Nucleótido Adenililtransferasa , Degeneración Walleriana , Animales , Proteínas del Dominio Armadillo/metabolismo , Axones/patología , Proteínas del Citoesqueleto/metabolismo , Humanos , Mamíferos/metabolismo , NAD/metabolismo , Degeneración Nerviosa/patología , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Inhibidores de Proteínas Quinasas , Degeneración Walleriana/metabolismo
8.
Glia ; 68(10): 1987-2000, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32173924

RESUMEN

Tanycytes are radial glial cells located in the mediobasal hypothalamus. Recent studies have proposed that tanycytes play an important role in hypothalamic control of energy homeostasis, although this has not been directly tested. Here, we report the phenotype of mice in which tanycytes of the arcuate nucleus and median eminence were conditionally ablated in adult mice. Although the cerebrospinal fluid-hypothalamic barrier was rendered more permeable following tanycyte ablation, neither the blood-hypothalamic barrier nor leptin-induced pSTAT3 activation in hypothalamic parenchyma were affected. We observed a significant increase in visceral fat distribution accompanying insulin insensitivity in male mice, without significant effect on either body weight or food intake. A high-fat diet tended to accelerate overall body weight gain in tanycyte-ablated mice, but the development of visceral adiposity and insulin insensitivity was comparable to wildtype. Thermoneutral housing exacerbated fat accumulation and produced a shift away from fat oxidation in tanycyte-ablated mice. These results clarify the extent to which tanycytes regulate energy balance, and demonstrate a role for tanycytes in regulating fat metabolism.


Asunto(s)
Tejido Adiposo/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Células Ependimogliales/metabolismo , Eliminación de Gen , Eminencia Media/metabolismo , Obesidad/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/química , Metabolismo Energético/fisiología , Células Ependimogliales/química , Masculino , Eminencia Media/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética
9.
Am J Physiol Endocrinol Metab ; 319(6): E1084-E1100, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33017221

RESUMEN

Central and peripheral mechanisms are both required for proper control of energy homeostasis. Among circulating plasma proteins, C1q/TNF-related proteins (CTRPs) have recently emerged as important regulators of sugar and fat metabolism. CTRP4, expressed in brain and adipose tissue, is unique among the family members in having two tandem globular C1q domains. We previously showed that central administration of recombinant CTRP4 suppresses food intake, suggesting a central nervous system role in regulating ingestive physiology. Whether this effect is pharmacological or physiological remains unclear. We used a loss-of-function knockout (KO) mouse model to clarify the physiological role of CTRP4. Under basal conditions, CTRP4 deficiency increased serum cholesterol levels and impaired glucose tolerance in male but not female mice fed a control low-fat diet. When challenged with a high-fat diet, male and female KO mice responded differently to weight gain and had different food intake patterns. On an obesogenic diet, male KO mice had similar weight gain as wild-type littermates. When fed ad libitum, KO male mice had greater meal number, shorter intermeal interval, and reduced satiety ratio. Female KO mice, in contrast, had lower body weight and adiposity. In the refeeding period following food deprivation, female KO mice had significantly higher food intake due to longer meal duration and reduced satiety ratio. Collectively, our data provide genetic evidence for a sex-dependent physiological role of CTRP4 in modulating food intake patterns and systemic energy metabolism.


Asunto(s)
Adipoquinas/genética , Adipoquinas/fisiología , Adiposidad/genética , Ingestión de Alimentos/genética , Adipoquinas/farmacología , Animales , Recuento de Células Sanguíneas , Colesterol/sangre , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Femenino , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , Respuesta de Saciedad , Caracteres Sexuales , Aumento de Peso/genética
10.
FASEB J ; 33(12): 14734-14747, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31689372

RESUMEN

Cytokines and chemokines play diverse roles in different organ systems. Family with sequence similarity 19, member A1-5 (FAM19A1-A5; also known as TAFA1-5) is a group of conserved chemokine-like proteins enriched in the CNS of mice and humans. Their functions are only beginning to emerge. Here, we show that the expression of Fam19a1-a5 in different mouse brain regions are induced or suppressed by unfed and refed states. The striking nutritional regulation of Fam19a family members in the brain suggests a potential central role in regulating metabolism. Using a knockout (KO) mouse model, we show that loss of FAM19A1 results in sexually dimorphic phenotypes. In male mice, FAM19A1 deficiency alters food intake patterns during the light and dark cycle. Fam19a1 KO mice are hyperactive, and locomotor hyperactivity is more pronounced in female KO mice. Behavior tests indicate that Fam19a1 KO female mice have reduced anxiety and sensitivity to pain. Spatial learning and exploration, however, is preserved in Fam19a1 KO mice. Altered behaviors are associated with elevated norepinephrine and dopamine turnover in the striatum. Our results establish an in vivo function of FAM19A1 and highlight central roles for this family of neurokines in modulating animal physiology and behavior.-Lei, X., Liu, L., Terrillion, C. E., Karuppagounder, S. S., Cisternas, P., Lay, M., Martinelli, D. C., Aja, S., Dong, X., Pletnikov, M. V., Wong, G. W. FAM19A1, a brain-enriched and metabolically responsive neurokine, regulates food intake patterns and mouse behaviors.


Asunto(s)
Quimiocinas/fisiología , Cuerpo Estriado/metabolismo , Ingestión de Alimentos , Locomoción , Aprendizaje Espacial , Animales , Células Cultivadas , Quimiocinas/genética , Dopamina/metabolismo , Conducta Exploratoria , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Ratas , Factores Sexuales
11.
Proc Natl Acad Sci U S A ; 114(52): E11238-E11247, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29229807

RESUMEN

Sclerostin has traditionally been thought of as a local inhibitor of bone acquisition that antagonizes the profound osteoanabolic capacity of activated Wnt/ß-catenin signaling, but serum sclerostin levels in humans exhibit a correlation with impairments in several metabolic parameters. These data, together with the increased production of sclerostin in mouse models of type 2 diabetes, suggest an endocrine function. To determine whether sclerostin contributes to the coordination of whole-body metabolism, we examined body composition, glucose homeostasis, and fatty acid metabolism in Sost-/- mice as well as mice that overproduce sclerostin as a result of adeno-associated virus expression from the liver. Here, we show that in addition to dramatic increases in bone volume, Sost-/- mice exhibit a reduction in adipose tissue accumulation in association with increased insulin sensitivity. Sclerostin overproduction results in the opposite metabolic phenotype due to adipocyte hypertrophy. Additionally, Sost-/- mice and those administered a sclerostin-neutralizing antibody are resistant to obesogenic diet-induced disturbances in metabolism. This effect appears to be the result of sclerostin's effects on Wnt signaling and metabolism in white adipose tissue. Since adipocytes do not produce sclerostin, these findings suggest an unexplored endocrine function for sclerostin that facilitates communication between the skeleton and adipose tissue.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Composición Corporal , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glicoproteínas/metabolismo , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/patología , Tejido Adiposo/patología , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Noqueados
12.
Glycobiology ; 27(2): 129-139, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27683310

RESUMEN

Sialyltransferases are a family of 20 gene products in mice and humans that transfer sialic acid from its activated precursor, CMP-sialic acid, to the terminus of glycoprotein and glycolipid acceptors. ST3Gal-II (coded by the St3gal2 gene) transfers sialic acid preferentially to the three positions of galactose on the Galß1-3GalNAc terminus of gangliosides GM1 and GD1b to synthesize GD1a and GT1b, respectively. Mice with a targeted disruption of St3gal2 unexpectedly displayed late-onset obesity and insulin resistance. At 3 months of age, St3gal2-null mice were the same weight as their wild type (WT) counterparts, but by 13 months on standard chow they were visibly obese, 22% heavier and with 37% greater fat/lean ratio than WT mice. St3gal2-null mice became hyperglycemic and displayed impaired glucose tolerance by 9 months of age. They had sharply reduced insulin responsiveness despite equivalent pancreatic islet morphology. Analyses of insulin receptor (IR) tyrosine kinase substrate IRS-1 and downstream target Akt revealed decreased insulin-induced phosphorylation in adipose tissue but not liver or skeletal muscle of St3gal2-null mice. Thin-layer chromatography and mass spectrometry revealed altered ganglioside profiles in the adipose tissue of St3gal2-null mice compared to WT littermates. Metabolically, St3gal2-null mice display a reduced respiratory exchange ratio compared to WT mice, indicating a preference for lipid oxidation as an energy source. Despite their altered metabolism, St3gal2-null mice were hyperactive. We conclude that altered ganglioside expression in adipose tissue results in diminished IR sensitivity and late-onset obesity.


Asunto(s)
Resistencia a la Insulina/genética , Ácido N-Acetilneuramínico/metabolismo , Obesidad/genética , Sialiltransferasas/genética , Tejido Adiposo/metabolismo , Animales , Modelos Animales de Enfermedad , Galactosa/metabolismo , Gangliósidos/biosíntesis , Gangliósidos/genética , Prueba de Tolerancia a la Glucosa , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Metabolismo de los Lípidos/genética , Ratones , Ratones Noqueados , Ácido N-Acetilneuramínico/genética , Obesidad/patología , beta-Galactosida alfa-2,3-Sialiltransferasa
13.
Biochem Biophys Res Commun ; 470(4): 818-23, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26802467

RESUMEN

The regulation of food intake is important for body energy homeostasis. Hypothalamic insulin signaling decreases food intake by upregulating the expression of anorexigenic neuropeptides and downregulating the expression of orexigenic neuropeptides. INS-2, a Mn(2+) chelate of 4-O-(2-amino-2-deoxy-ß-D-galactopyranosyl)-3-O-methyl-D-chiro-inositol, acts as an insulin mimetic and sensitizer. We found that intracerebroventricular injection of INS-2 decreased body weight and food intake in mice. In hypothalamic neuronal cell lines, INS-2 downregulated the expression of neuropeptide Y (NPY), an orexigenic neuropeptide, but upregulated the expression of proopiomelanocortin (POMC), an anorexigenic neuropeptide, via modulation of the AKT-forkhead box-containing protein-O1 (FoxO1) pathway. Pretreatment of these cells with INS-2 enhanced the action of insulin on downstream signaling, leading to a further decrease in NPY expression and increase in POMC expression. These data indicate that INS-2 reduces food intake by regulating the expression of the hypothalamic neuropeptide genes through the AKT-FoxO1 pathway downstream of insulin.


Asunto(s)
Disacáridos/administración & dosificación , Ingestión de Alimentos/fisiología , Factores de Transcripción Forkhead/metabolismo , Hipotálamo/fisiología , Insulina/metabolismo , Neuropéptidos/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Línea Celular , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Proteína Forkhead Box O1 , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hipotálamo/efectos de los fármacos , Fosfatos de Inositol/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Polisacáridos/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
14.
Arch Biochem Biophys ; 591: 57-65, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26701603

RESUMEN

The Keap1/Nrf2 pathway, known to regulate the expression of a series of cytoprotective and antioxidant genes, has been studied in the context of obesity and type 2 diabetes; diseases that are characterized by chronic oxidative stress. There is increasing evidence, however, that the transcription factor Nrf2 can crosstalk with pathways not directly related to cytoprotection. Our present work focuses on the effect of Nrf2 on hepatic gluconeogenesis and lipogenesis, two metabolic processes which are dysregulated in the obese/diabetic state. To this end, a genetic mouse model of Nrf2 pathway activation was used (Keap1-hypo; both Keap1 alleles are hypomorphic) and was exposed to a 3-month high-fat diet along with the relevant control wild-type mice. The Keap1-hypo mice were partially protected from obesity, had lower fasting glucose and insulin levels and developed less liver steatosis compared to the wild-type. Key gluconeogenic and lipogenic enzymes were repressed in the Keap1-hypo livers with concomitant activated Ampk signaling. Primary Keap1-hypo hepatocyte cultures also show increased Ampk signaling and repressed glucose production. In conclusion, increased Keap1/Nrf2 signaling in the liver is accompanied by repressed gluconeogenesis and lipogenesis that can, at least partially, explain the ameliorated diabetic phenotype in the Keap1-hypo mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Grasas de la Dieta/metabolismo , Gluconeogénesis/fisiología , Lipogénesis/fisiología , Hígado/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas del Citoesqueleto/genética , Dieta Alta en Grasa/métodos , Proteína 1 Asociada A ECH Tipo Kelch , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/fisiología
15.
Am J Physiol Endocrinol Metab ; 306(7): E779-90, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24473438

RESUMEN

Transgenic overexpression of CTRP9, a secreted hormone downregulated in obesity, confers striking protection against diet-induced obesity and type 2 diabetes. However, the physiological relevance of this adiponectin-related plasma protein remains undefined. Here, we used gene targeting to establish the metabolic function of CTRP9 in a physiological context. Mice lacking CTRP9 were obese and gained significantly more body weight when fed standard laboratory chow. Increased food intake, due in part to upregulated expression of hypothalamic orexigenic neuropeptides, contributed to greater adiposity in CTRP9 knockout mice. Although the frequency of food intake remained unchanged, CTRP9 knockout mice increased caloric intake by increasing meal size and decreasing satiety ratios. The absence of CTRP9 also resulted in peripheral tissue insulin resistance, leading to increased fasting insulin levels, impaired hepatic insulin signaling, and reduced insulin tolerance. Increased expression of lipogenic genes, combined with enhanced caloric intake, contributed to hepatic steatosis in CTRP9 knockout mice. Loss of CTRP9 also resulted in reduced skeletal muscle AMPK activation and mitochondrial content. Together, these results provide the genetic evidence for a physiological role of CTRP9 in controlling energy balance via central and peripheral mechanisms.


Asunto(s)
Adiponectina/fisiología , Ingestión de Alimentos/genética , Hígado Graso/genética , Glicoproteínas/fisiología , Resistencia a la Insulina/genética , Adiponectina/genética , Adiposidad/genética , Animales , Peso Corporal/genética , Metabolismo Energético/genética , Femenino , Eliminación de Gen , Marcación de Gen , Glicoproteínas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
J Biol Chem ; 287(2): 1576-87, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22086915

RESUMEN

We previously described the adipokine CTRP1, which has up-regulated expression following exposure to the anti-diabetic drug rosiglitazone and increased circulating levels in adiponectin-null mice (Wong, G. W., Krawczyk, S. A., Kitidis-Mitrokostas, C., Revett, T., Gimeno, R., and Lodish, H. F. (2008) Biochem. J. 416, 161-177). Although recombinant CTRP1 lowers blood glucose in mice, its physiological function, mechanisms of action, and roles in metabolic stress remain unknown. Here, we show that circulating levels of CTRP1 are strikingly reduced in diet-induced obese mice. Overexpressing CTRP1 in transgenic mice improved insulin sensitivity and decreased high-fat diet-induced weight gain. Reduced adiposity resulted from enhanced fatty acid oxidation and energy expenditure, effects mediated by AMP-activated protein kinase (AMPK). In skeletal muscle of transgenic mice, AMPKα and its downstream target, acetyl-CoA carboxylase (ACC), were hyperphosphorylated, indicative of AMPK activation and ACC inhibition. Inactivation of ACC promotes mitochondrial fat oxidation. Consistent with the direct effect of CTRP1 on AMPK signaling, recombinant CTRP1 administration acutely stimulated muscle AMPKα and ACC phosphorylation in vivo. In isolated soleus muscle, recombinant CTRP1 activated AMPK signaling to increase fatty acid oxidation ex vivo, an effect abrogated by an AMPK inhibitor. These results provide the first in vivo evidence that CTRP1 is a novel regulator of fatty acid metabolism.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Adipoquinas/metabolismo , Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Acetil-CoA Carboxilasa/genética , Adipoquinas/genética , Adiposidad/genética , Animales , Grasas de la Dieta , Ácidos Grasos/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Oxidación-Reducción , Transducción de Señal/fisiología
17.
Eur J Neurosci ; 37(7): 1033-47, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23360481

RESUMEN

Estrogen-related receptors (ERRs) α, ß and γ are orphan nuclear hormone receptors with no known ligands. Little is known concerning the role of ERRß in energy homeostasis, as complete ERRß-null mice die mid-gestation. We generated two viable conditional ERRß-null mouse models to address its metabolic function. Whole-body deletion of ERRß in Sox2-Cre:ERRß(lox/lox) mice resulted in major alterations in body composition, metabolic rate, meal patterns and voluntary physical activity levels. Nestin-Cre:ERRß(lox/lox) mice exhibited decreased expression of ERRß in hindbrain neurons, the predominant site of expression, decreased neuropeptide Y (NPY) gene expression in the hindbrain, increased lean body mass, insulin sensitivity, increased energy expenditure, decreased satiety and decreased time between meals. In the absence of ERRß, increased ERRγ signaling decreased satiety and the duration of time between meals, similar to meal patterns observed for both the Sox2-Cre:ERRß(lox/lox) and Nestin-Cre:ERRß(lox/lox) strains of mice. Central and/or peripheral ERRγ signaling may modulate these phenotypes by decreasing NPY gene expression. Overall, the relative expression ratio between ERRß and ERRγ may be important in modulating ingestive behavior, specifically satiety, gene expression, as well as whole-body energy balance.


Asunto(s)
Metabolismo Energético , Neuropéptido Y/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Animales , Peso Corporal , Eliminación de Gen , Expresión Génica , Resistencia a la Insulina , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuropéptido Y/genética , Fenotipo , Rombencéfalo/metabolismo , Saciedad , Transducción de Señal
18.
Am J Physiol Gastrointest Liver Physiol ; 305(3): G214-24, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23744740

RESUMEN

CTRP3 is a secreted plasma protein of the C1q family that helps regulate hepatic gluconeogenesis and is downregulated in a diet-induced obese state. However, the role of CTRP3 in regulating lipid metabolism has not been established. Here, we used a transgenic mouse model to address the potential function of CTRP3 in ameliorating high-fat diet-induced metabolic stress. Both transgenic and wild-type mice fed a high-fat diet showed similar body weight gain, food intake, and energy expenditure. Despite similar adiposity to wild-type mice upon diet-induced obesity (DIO), CTRP3 transgenic mice were strikingly resistant to the development of hepatic steatosis, had reduced serum TNF-α levels, and demonstrated a modest improvement in systemic insulin sensitivity. Additionally, reduced hepatic triglyceride levels were due to decreased expression of enzymes (GPAT, AGPAT, and DGAT) involved in triglyceride synthesis. Importantly, short-term daily administration of recombinant CTRP3 to DIO mice for 5 days was sufficient to improve the fatty liver phenotype, evident as reduced hepatic triglyceride content and expression of triglyceride synthesis genes. Consistent with a direct effect on liver cells, recombinant CTRP3 treatment reduced fatty acid synthesis and neutral lipid accumulation in cultured rat H4IIE hepatocytes. Together, these results establish a novel role for CTRP3 hormone in regulating hepatic lipid metabolism and highlight its protective function and therapeutic potential in attenuating hepatic steatosis.


Asunto(s)
Adipoquinas/fisiología , Dieta Alta en Grasa , Hígado Graso/etiología , Hígado Graso/prevención & control , Triglicéridos/metabolismo , Adipoquinas/genética , Adipoquinas/farmacología , Animales , Ácidos Grasos/biosíntesis , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/química , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Proteínas Recombinantes/farmacología , Triglicéridos/análisis , Triglicéridos/genética , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/sangre
19.
Am J Physiol Regul Integr Comp Physiol ; 305(5): R522-33, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23842676

RESUMEN

CTRP9 is a secreted multimeric protein of the C1q family and the closest paralog of the insulin-sensitizing adipokine, adiponectin. The metabolic function of this adipose tissue-derived plasma protein remains largely unknown. Here, we show that the circulating levels of CTRP9 are downregulated in diet-induced obese mice and upregulated upon refeeding. Overexpressing CTRP9 resulted in lean mice that dramatically resisted weight gain induced by a high-fat diet, largely through decreased food intake and increased basal metabolism. Enhanced fat oxidation in CTRP9 transgenic mice resulted from increases in skeletal muscle mitochondrial content, expression of enzymes involved in fatty acid oxidation (LCAD and MCAD), and chronic AMPK activation. Hepatic and skeletal muscle triglyceride levels were substantially decreased in transgenic mice. Consequently, CTRP9 transgenic mice had a greatly improved metabolic profile with markedly reduced fasting insulin and glucose levels. The high-fat diet-induced obesity, insulin resistance, and hepatic steatosis observed in wild-type mice were prevented in transgenic mice. Consistent with the in vivo data, recombinant protein significantly enhanced fat oxidation in L6 myotubes via AMPK activation and reduced lipid accumulation in H4IIE hepatocytes. Collectively, these data establish CTRP9 as a novel metabolic regulator and a new component of the metabolic network that links adipose tissue to lipid metabolism in skeletal muscle and liver.


Asunto(s)
Adiponectina/metabolismo , Tejido Adiposo/fisiopatología , Grasas de la Dieta/efectos adversos , Glicoproteínas/metabolismo , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/fisiopatología , Obesidad/etiología , Obesidad/fisiopatología , Adiponectina/genética , Animales , Glicoproteínas/genética , Masculino , Enfermedades Metabólicas/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/prevención & control
20.
Am J Physiol Regul Integr Comp Physiol ; 304(12): R1085-95, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23576617

RESUMEN

Disruption of finely coordinated neuropeptide signals in the hypothalamus can result in altered food intake and body weight. We identified neuron-derived neurotrophic factor (NENF) as a novel secreted protein through a large-scale screen aimed at identifying novel secreted hypothalamic proteins that regulate food intake. We observed robust Nenf expression in hypothalamic nuclei known to regulate food intake, and its expression was altered under the diet-induced obese (DIO) condition relative to the fed state. Hypothalamic Nenf mRNA was regulated by brain-derived neurotrophic factor (BDNF) signaling, itself an important regulator of appetite. Delivery of purified recombinant BDNF into the lateral cerebral ventricle decreased hypothalamic Nenf expression, while pharmacological inhibition of trkB signaling increased Nenf mRNA expression. Furthermore, recombinant NENF administered via an intracerebroventricular cannula decreased food intake and body weight and increased hypothalamic Pomc and Mc4r mRNA expression. Importantly, the appetite-suppressing effect of NENF was abrogated in obese mice fed a high-fat diet, demonstrating a diet-dependent modulation of NENF function. We propose the existence of a regulatory circuit involving BDNF, NENF, and melanocortin signaling. Our study validates the power of using an integrated experimental and bioinformatic approach to identify novel CNS-derived proteins with appetite-modulating function and reveals NENF as an important central modulator of food intake.


Asunto(s)
Apetito/fisiología , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Obesidad/fisiopatología , Transducción de Señal/fisiología , Animales , Apetito/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/farmacología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/farmacología , Obesidad/inducido químicamente , Obesidad/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA