Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 325(5): E438-E447, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37702737

RESUMEN

Excess glucocorticoid (GC) signaling in adipose tissue is a key driver of insulin resistance and hepatic steatosis, but underlying mechanisms have not been fully elucidated. Signal transducer and activator of transcription 5 (STAT5) signaling in adipocytes has also been implicated in the progression of similar metabolic disturbances. Although STAT5 has been shown to interact with the glucocorticoid receptor (GR) in many cell types including adipocytes, the relevance of the STAT5/GR complex has not been investigated in adipocytes. Adult male and female adipocyte-specific STAT5 knockout (STAT5AKO) and floxed mice were given corticosterone (CORT) or vehicle in their drinking water for 1 wk and examined for differences in their metabolic responses to GC excess. CORT-induced lipolysis, insulin resistance, and changes in body composition were comparable between genotypes and in both sexes. Adipocyte STAT5 is not necessary for GC-mediated progression of metabolic disease.NEW & NOTEWORTHY Both STAT5 and glucocorticoid receptor contribute to metabolic processes and type 2 diabetes, in large part, due to their functions in adipocytes. These two transcription factors can form a complex and function together. Our novel studies determined the role of adipocyte STAT5 in glucocorticoid-induced diabetes. We observed that STAT5 in adipocytes is not needed for glucocorticoid-induced diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Enfermedades Metabólicas , Factor de Transcripción STAT5 , Animales , Femenino , Masculino , Ratones , Adipocitos/metabolismo , Corticosterona/farmacología , Corticosterona/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucocorticoides/efectos adversos , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Resistencia a la Insulina/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Enfermedades Metabólicas/inducido químicamente , Enfermedades Metabólicas/genética
2.
Physiol Genomics ; 54(8): 319-324, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35816649

RESUMEN

Preeclampsia (PE), a pregnancy-specific disorder, is characterized by maternal hypertension and proteinuria or another accompanying sign/symptom of multiorgan dysfunction. Maternal symptoms resolve with delivery of the baby and, importantly, the placenta. Therefore, the placenta plays a causal role in PE. However, the precise cause of abnormal placental development and/or function is unknown. Women with obesity have an increased risk of developing PE that is potentially related to the increased inflammation that accompanies increased maternal adiposity. Furthermore, inflammatory adipokines, i.e., leptin, have been linked to the development of systemic inflammation, hypertension, and other adverse outcomes associated with PE. Rodent models that recapitulate key pathophysiological features of the maternal and fetal syndrome have been used translationally to study PE. This review covers inflammatory adipokines, immune cells, and impaired placental development associated with PE in women and in rodent models of PE that use functional genomics to test causation.


Asunto(s)
Hipertensión , Preeclampsia , Adipoquinas , Tejido Adiposo , Femenino , Humanos , Hipertensión/complicaciones , Inflamación/complicaciones , Obesidad , Placenta , Placentación , Preeclampsia/etiología , Embarazo
3.
Int J Mol Sci ; 23(7)2022 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-35409014

RESUMEN

Herbal remedies are increasing in popularity as treatments for metabolic conditions such as obesity and Type 2 Diabetes. One potential therapeutic option is fenugreek seeds (Trigonella foenum-graecum), which have been used for treating high cholesterol and Type 2 diabetes. A proposed mechanism for these benefits is through alterations in the microbiome, which impact mammalian host metabolic function. This study used untargeted metabolomics to investigate the fenugreek-induced alterations in the intestinal, liver, and serum profiles of mice fed either a 60% high-fat or low-fat control diet each with or without fenugreek supplementation (2% w/w) for 14 weeks. Metagenomic analyses of intestinal contents found significant alterations in the relative composition of the gut microbiome resulting from fenugreek supplementation. Specifically, Verrucomicrobia, a phylum containing beneficial bacteria which are correlated with health benefits, increased in relative abundance with fenugreek. Metabolomics partial least squares discriminant analysis revealed substantial fenugreek-induced changes in the large intestines. However, it was observed that while the magnitude of changes was less, significant modifications were present in the liver tissues resulting from fenugreek supplementation. Further analyses revealed metabolic processes affected by fenugreek and showed broad ranging impacts in multiple pathways, including carnitine biosynthesis, cholesterol and bile acid metabolism, and arginine biosynthesis. These pathways may play important roles in the beneficial effects of fenugreek.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Trigonella , Animales , Colesterol , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Suplementos Dietéticos , Mamíferos , Ratones , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico
4.
FASEB J ; 34(1): 41-65, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914647

RESUMEN

While great interest in health effects of natural product (NP) including dietary supplements and foods persists, promising preclinical NP research is not consistently translating into actionable clinical trial (CT) outcomes. Generally considered the gold standard for assessing safety and efficacy, CTs, especially phase III CTs, are costly and require rigorous planning to optimize the value of the information obtained. More effective bridging from NP research to CT was the goal of a September, 2018 transdisciplinary workshop. Participants emphasized that replicability and likelihood of successful translation depend on rigor in experimental design, interpretation, and reporting across the continuum of NP research. Discussions spanned good practices for NP characterization and quality control; use and interpretation of models (computational through in vivo) with strong clinical predictive validity; controls for experimental artefacts, especially for in vitro interrogation of bioactivity and mechanisms of action; rigorous assessment and interpretation of prior research; transparency in all reporting; and prioritization of research questions. Natural product clinical trials prioritized based on rigorous, convergent supporting data and current public health needs are most likely to be informative and ultimately affect public health. Thoughtful, coordinated implementation of these practices should enhance the knowledge gained from future NP research.


Asunto(s)
Productos Biológicos/farmacología , Investigación Biomédica Traslacional/normas , Animales , Evaluación Preclínica de Medicamentos , Etnobotánica , Humanos
5.
J Nat Prod ; 84(4): 1078-1086, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33830759

RESUMEN

Two new diprenylated coumaric acid isomers (1a and 1b) and two known congeners, capillartemisin A (2) and B (3), were isolated from Artemisia scoparia as bioactive markers using bioactivity-guided HPLC fractionation. Their structures were determined by spectroscopic means, including 1D and 2D NMR methods and LC-MS, with their purity assessed by 1D 1H pure shift qNMR spectroscopic analysis. The bioactivity of compounds was evaluated by enhanced accumulation of lipids, as measured using Oil Red O staining, and by increased expression of several adipocyte marker genes, including adiponectin in 3T3-L1 adipocytes relative to untreated negative controls. Compared to the plant's 80% EtOH extract, these purified compounds showed significant but still weaker inhibition of TNFα-induced lipolysis in 3T3-L1 adipocytes. This suggests that additional bioactive substances are responsible for the multiple metabolically favorable effects on adipocytes observed with Artemisia scoparia extract.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Artemisia/química , Ácidos Cumáricos/farmacología , Células 3T3-L1 , Adiponectina/metabolismo , Animales , Ácidos Cumáricos/aislamiento & purificación , Lipólisis/efectos de los fármacos , Ratones , Fitoquímicos/aislamiento & purificación , Fitoquímicos/farmacología , Prenilación , Factor de Necrosis Tumoral alfa/metabolismo
6.
Clin Sci (Lond) ; 134(6): 629-639, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32219346

RESUMEN

Adipocytes and adipose tissue are not inert and make substantial contributions to systemic metabolism by influencing energy homeostasis, insulin sensitivity, and lipid storage. In addition to well-studied hormones such as insulin, there are numerous hormones, cytokines, and growth factors that modulate adipose tissue function. Many endocrine mediators utilize the JAK-STAT pathway to mediate dozens of biological processes, including inflammation and immune responses. JAKs and STATs can modulate both adipocyte development and mature adipocyte function. Of the seven STAT family members, four STATs are expressed in adipocytes and regulated during adipogenesis (STATs 1, 3, 5A, and 5B). These STATs have been shown to play influential roles in adipose tissue development and function. STAT6, in contrast, is highly expressed in both preadipocytes and mature adipocytes, but is not considered to play a major role in regulating adipose tissue function. This review will summarize the latest research that pertains to the functions of STATs in adipocytes and adipose tissue.


Asunto(s)
Adipocitos/metabolismo , Factores de Transcripción STAT/metabolismo , Adipocitos/citología , Adipogénesis , Animales , Humanos , Factores de Transcripción STAT/genética , Transducción de Señal
7.
J Biol Chem ; 292(48): 19733-19742, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982698

RESUMEN

STAT5 proteins play a role in adipocyte development and function, but their specific functions are largely unknown. To this end, we used an unbiased MS-based approach to identify novel STAT5-interacting proteins. We observed that STAT5A bound the E1ß and E2 subunits of the pyruvate dehydrogenase complex (PDC). Whereas STAT5A typically localizes to the cytosol or nucleus, PDC normally resides within the mitochondrial matrix where it converts pyruvate to acetyl-CoA. We employed affinity purification and immunoblotting to validate the interaction between STAT5A and PDC subunits in murine and human cultured adipocytes, as well as in adipose tissue. We found that multiple PDC subunits interact with hormone-activated STAT5A in a dose- and time-dependent manner that coincides with tyrosine phosphorylation of STAT5. Using subcellular fractionation and immunofluorescence microscopy, we observed that PDC-E2 is present within the adipocyte nucleus where it associates with STAT5A. Because STAT5A is a transcription factor, we used chromatin immunoprecipitation (ChIP) to assess PDC's ability to interact with STAT5 DNA-binding sites. These analyses revealed that PDC-E2 is bound to a STAT5-binding site in the promoter of the STAT5 target gene cytokine-inducible SH2-containing protein (cish). We have demonstrated a compelling interaction between STAT5A and PDC subunits in adipocytes under physiological conditions. There is previous evidence that PDC localizes to cancer cell nuclei where it plays a role in histone acetylation. On the basis of our ChIP data and these previous findings, we hypothesize that PDC may modulate STAT5's ability to regulate gene expression by controlling histone or STAT5 acetylation.


Asunto(s)
Tejido Adiposo/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Factor de Transcripción STAT5/metabolismo , Células 3T3-L1 , Tejido Adiposo/citología , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica
8.
Am J Physiol Endocrinol Metab ; 315(5): E1053-E1061, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30153067

RESUMEN

An ethanolic extract of Artemisia scoparia (SCO) has metabolically favorable effects on adipocyte development and function in vitro and in vivo. In diet-induced obese mice, SCO supplementation significantly reduced fasting glucose and insulin levels. Given the importance of adipocyte lipolysis in metabolic health, we hypothesized that SCO modulates lipolysis in vitro and in vivo. Free fatty acids and glycerol were measured in the sera of mice fed a high-fat diet with or without SCO supplementation. In cultured 3T3-L1 adipocytes, the effects of SCO on lipolysis were assessed by measuring glycerol and free fatty acid release. Microarray analysis, qPCR, and immunoblotting were used to assess gene expression and protein abundance. We found that SCO supplementation of a high-fat diet in mice substantially reduces circulating glycerol and free fatty acid levels, and we observed a cell-autonomous effect of SCO to significantly attenuate tumor necrosis factor-α (TNFα)-induced lipolysis in cultured adipocytes. Although several prolipolytic and antilipolytic genes were identified by microarray analysis of subcutaneous and visceral adipose tissue from SCO-fed mice, regulation of these genes did not consistently correlate with SCO's ability to reduce lipolytic metabolites in sera or cell culture media. However, in the presence of TNFα in cultured adipocytes, SCO induced antilipolytic changes in phosphorylation of hormone-sensitive lipase and perilipin. Together, these data suggest that the antilipolytic effects of SCO on adipose tissue play a role in the ability of this botanical extract to improve whole body metabolic parameters and support its use as a dietary supplement to promote metabolic resiliency.


Asunto(s)
Adipocitos/efectos de los fármacos , Artemisia , Lipólisis/efectos de los fármacos , Extractos Vegetales/farmacología , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Células Cultivadas , Ácidos Grasos no Esterificados/sangre , Glicerol/sangre , Ratones , Perilipina-1/metabolismo , Fosforilación/efectos de los fármacos , Esterol Esterasa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
9.
Proc Natl Acad Sci U S A ; 112(25): E3300-9, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-26056297

RESUMEN

The correlations between intramyocellular lipid (IMCL), decreased fatty acid oxidation (FAO), and insulin resistance have led to the hypothesis that impaired FAO causes accumulation of lipotoxic intermediates that inhibit muscle insulin signaling. Using a skeletal muscle-specific carnitine palmitoyltransferase-1 KO model, we show that prolonged and severe mitochondrial FAO inhibition results in increased carbohydrate utilization, along with reduced physical activity; increased circulating nonesterified fatty acids; and increased IMCLs, diacylglycerols, and ceramides. Perhaps more importantly, inhibition of mitochondrial FAO also initiates a local, adaptive response in muscle that invokes mitochondrial biogenesis, compensatory peroxisomal fat oxidation, and amino acid catabolism. Loss of its major fuel source (lipid) induces an energy deprivation response in muscle coordinated by signaling through AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) to maintain energy supply for locomotion and survival. At the whole-body level, these adaptations result in resistance to obesity.


Asunto(s)
Adaptación Fisiológica , Ácidos Grasos no Esterificados/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/fisiología , Oxidación-Reducción
10.
Endocr Res ; 43(4): 275-283, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29768065

RESUMEN

AIM OF THE STUDY: The regulation and actions of fibroblast growth factor 21 (FGF21) are responsive to energy status and macronutrient balance, and investigations of FGF21 in normal pregnancy, which could be informative for FGF21 biology, are seldom. The goal of our study was to examine FGF21 levels in a contemporary healthy, pregnant population. METHODS: We phenotyped 43 women with overweight and obesity during pregnancy for weight, body composition, and fasting blood. Serum FGF21 was measured during the first and third trimesters. Placentas were collected at delivery. RESULTS: Maternal FGF21 concentrations were positively correlated with body mass index and adiposity, but not lean mass or glucose homeostasis. FGF21 concentrations significantly increased from the first to third trimester of pregnancy (0.105 vs. 0.256 ng/mL, p < 0.0001). Changes in FGF21 concentrations across pregnancy were not associated with changes in body weight or composition but inversely with the change in fasting glucose. FGF21 mRNA levels in placenta were very low and do not likely contribute to FGF21 in the maternal circulation. CONCLUSIONS: FGF21 increases throughout pregnancy in our healthy cohort with overweight and obesity, independent of the placenta, and does not appear to be sensing the changes in energy balance (reflected in the change in maternal energy stores), but changes in macronutrient status. Thus, we propose FGF21 may be a potential signal of maternal nutrient status in pregnancy.


Asunto(s)
Adiposidad/fisiología , Metabolismo Energético/fisiología , Factores de Crecimiento de Fibroblastos/sangre , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Obesidad/sangre , Sobrepeso/sangre , Adulto , Índice de Masa Corporal , Femenino , Humanos , Estado Nutricional , Embarazo , Adulto Joven
11.
J Biol Chem ; 291(33): 17066-76, 2016 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-27325693

RESUMEN

Oncostatin M (OSM) is a multifunctional gp130 cytokine. Although OSM is produced in adipose tissue, it is not produced by adipocytes. OSM expression is significantly induced in adipose tissue from obese mice and humans. The OSM-specific receptor, OSM receptor ß (OSMR), is expressed in adipocytes, but its function remains largely unknown. To better understand the effects of OSM in adipose tissue, we knocked down Osmr expression in adipocytes in vitro using siRNA. In vivo, we generated a mouse line lacking Osmr in adiponectin-expressing cells (OSMR(FKO) mice). The effects of OSM on gene expression were also assessed in vitro and in vivo OSM exerts proinflammatory effects on cultured adipocytes that are partially rescued by Osmr knockdown. Osm expression is significantly increased in adipose tissue T cells of high fat-fed mice. In addition, adipocyte Osmr expression is increased following high fat feeding. OSMR(FKO) mice exhibit increased insulin resistance and adipose tissue inflammation and have increased lean mass, femoral length, and bone volume. Also, OSMR(FKO) mice exhibit increased expression of Osm, the T cell markers Cd4 and Cd8, and the macrophage markers F4/80 and Cd11c Interestingly, the same proinflammatory genes induced by OSM in adipocytes are induced in the adipose tissue of the OSMR(FKO) mouse, suggesting that increased expression of proinflammatory genes in adipose tissue arises both from adipocytes and other cell types. These findings suggest that adipocyte OSMR signaling is involved in the regulation of adipose tissue homeostasis and that, in obesity, OSMR ablation may exacerbate insulin resistance by promoting adipose tissue inflammation.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Oncostatina M/metabolismo , Paniculitis/metabolismo , Transducción de Señal , Células 3T3-L1 , Adipocitos/patología , Tejido Adiposo/patología , Animales , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Antígenos CD4/genética , Antígenos CD4/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Ratones , Ratones Mutantes , Obesidad/patología , Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Paniculitis/genética , Paniculitis/patología
12.
J Biol Chem ; 290(21): 13401-16, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25851902

RESUMEN

Glucocorticoids signal through the glucocorticoid receptor (GR) and are administered clinically for a variety of situations, including inflammatory disorders, specific cancers, rheumatoid arthritis, and organ/tissue transplantation. However, glucocorticoid therapy is also associated with additional complications, including steroid-induced diabetes. We hypothesized that modification of the steroid backbone is one strategy to enhance the therapeutic potential of GR activation. Toward this goal, two commercially unavailable, thiobenzothiazole-containing derivatives of hydrocortisone (termed MS4 and MS6) were examined using 832/13 rat insulinoma cells as well as rodent and human islets. We found that MS4 had transrepression properties but lacked transactivation ability, whereas MS6 retained both transactivation and transrepression activities. In addition, MS4 and MS6 both displayed anti-inflammatory activity. Furthermore, MS4 displayed reduced impact on islet ß-cell function in both rodent and human islets. Similar to dexamethasone, MS6 promoted adipocyte development in vitro, whereas MS4 did not. Moreover, neither MS4 nor MS6 activated the Pck1 (Pepck) gene in primary rat hepatocytes. We conclude that modification of the functional groups attached to the D-ring of the hydrocortisone steroid molecule produces compounds with altered structure-function GR agonist activity with decreased impact on insulin secretion and reduced adipogenic potential but with preservation of anti-inflammatory activity.


Asunto(s)
Antiinflamatorios/farmacología , Bencimidazoles/farmacología , Benzotiazoles/farmacología , Hidrocortisona/análogos & derivados , Hidrocortisona/farmacología , Inflamación/tratamiento farmacológico , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inmunología , Tiazoles/química , Células 3T3-L1 , Animales , Antiinflamatorios/síntesis química , Apoptosis/efectos de los fármacos , Bencimidazoles/síntesis química , Benzotiazoles/síntesis química , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dexametasona/farmacología , Perfilación de la Expresión Génica , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/metabolismo , Humanos , Hidrocortisona/síntesis química , Técnicas para Inmunoenzimas , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Metabolómica , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno/efectos de los fármacos , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Biochim Biophys Acta ; 1849(6): 637-52, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25882704

RESUMEN

Enhanced leukocytic infiltration into pancreatic islets contributes to inflammation-based diminutions in functional ß-cell mass. Insulitis (aka islet inflammation), which can be present in both T1DM and T2DM, is one factor influencing pancreatic ß-cell death and dysfunction. IL-1ß, an inflammatory mediator in both T1DM and T2DM, acutely (within 1h) induced expression of the CCL20 gene in rat and human islets and clonal ß-cell lines. Transcriptional induction of CCL20 required the p65 subunit of NF-κB to replace the p50 subunit at two functional κB sites within the CCL20 proximal gene promoter. The NF-κB p50 subunit prevents CCL20 gene expression during unstimulated conditions and overexpression of p50 reduces CCL20, but enhances cyclooxygenase-2 (COX-2), transcript accumulation after exposure to IL-1ß. We also identified differential recruitment of specific co-activator molecules to the CCL20 gene promoter, when compared with the CCL2 and COX2 genes, revealing distinct transcriptional requirements for individual NF-κB responsive genes. Moreover, IL-1ß, TNF-α and IFN-γ individually increased the expression of CCR6, the receptor for CCL20, on the surface of human neutrophils. We further found that the chemokine CCL20 is elevated in serum from both genetically obese db/db mice and in C57BL6/J mice fed a high-fat diet. Taken together, these results are consistent with a possible activation of the CCL20-CCR6 axis in diseases with inflammatory components. Thus, interfering with this signaling pathway, either at the level of NF-κB-mediated chemokine production, or downstream receptor activation, could be a potential therapeutic target to offset inflammation-associated tissue dysfunction in obesity and diabetes.


Asunto(s)
Quimiocina CCL20/genética , Diabetes Mellitus/genética , Inflamación/genética , Obesidad/genética , Factor de Transcripción ReIA/genética , Animales , Quimiocina CCL20/biosíntesis , Quimiocina CCL20/metabolismo , Diabetes Mellitus/patología , Humanos , Inmunidad Innata/genética , Inflamación/patología , Resistencia a la Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Obesos , FN-kappa B/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Ratas , Receptores CCR6/genética , Transducción de Señal/genética , Factor de Transcripción ReIA/biosíntesis , Factor de Transcripción ReIA/metabolismo
14.
J Cell Physiol ; 231(7): 1562-74, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26566083

RESUMEN

Knowledge concerning mechanisms that control proliferation and differentiation of preadipocytes is essential to our understanding of adipocyte hyperplasia and the development of obesity. Evidence has shown that temporal regulation of mitogen-activated protein kinase (MAPK) phosphorylation and dephosphorylation is critical for coupling extracellular stimuli to cellular growth and differentiation. Using differentiating 3T3-L1 preadipocytes as a model of adipocyte hyperplasia, we examined a role for dual-specificity phosphatase 1 (DUSP1) on the timely modulation of MAPK signaling during states of growth arrest, proliferation, and differentiation. Using real-time reverse transcription PCR (qRT-PCR), we report that DUSP1 is induced during early preadipocyte proliferation concomitant with ERK and p38 dephosphorylation. As deactivation of ERK and p38 is essential for the progression of adipocyte differentiation, we further showed that de novo mRNA synthesis was required for ERK and p38 dephosphorylation, suggesting a role for "inducible" phosphatases in regulating MAPK signaling. Pharmacological and genetic inhibition of DUSP1 markedly increased ERK and p38 phosphorylation during early adipocyte differentiation. Based on these findings, we postulated that loss of DUSP1 would block adipocyte hyperplasia. However, genetic loss of DUSP1 was not sufficient to prevent preadipocyte proliferation or differentiation, suggesting a role for other phosphatases in the regulation of adipogenesis. In support of this, qRT-PCR identified several MAPK-specific DUSPs induced during early (DUSP2, -4, -5, & -6), mid (DUSP4 & -16) and late (DUSP9) stages of adipocyte differentiation. Collectively, these data suggest an important role for DUSPs in regulating MAPK dephosphorylation, with an emphasis on DUSP1, during early adipogenesis.


Asunto(s)
Diferenciación Celular/genética , Fosfatasa 1 de Especificidad Dual/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/genética , Obesidad/genética , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proliferación Celular/genética , Fosfatasa 1 de Especificidad Dual/genética , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Obesidad/patología , Fosforilación , ARN Mensajero/biosíntesis
15.
Am J Physiol Endocrinol Metab ; 310(2): E129-36, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26601851

RESUMEN

Adiponectin is a hormone secreted from adipocytes that plays an important role in insulin sensitivity and protects against metabolic syndrome. Growth hormone (GH) and prolactin (PRL) are potent STAT5 activators that regulate the expression of several genes in adipocytes. Studies have shown that the secretion of adiponectin from adipose tissue is decreased by treatment with PRL and GH. In this study, we demonstrate that 3T3-L1 adipocytes treated with GH or PRL exhibit a reduction in adiponectin protein levels. Furthermore, we identified three putative STAT5 binding sites in the murine adiponectin promoter and show that only one of these, located at -3,809, binds nuclear protein in a GH- or PRL-dependent manner. Mutation of the STAT5 binding site reduced PRL-dependent protein binding, and supershift analysis revealed that STAT5A and -5B, but not STAT1 and -3, bind to this site in response to PRL. Chromatin immunoprecipitation (IP) analysis demonstrated that only STAT5A, and not STAT1 and -3, bind to the murine adiponectin promoter in a GH-dependent manner in vivo. Adiponectin promoter/reporter constructs were responsive to GH, and chromatin IP analysis reveals that STAT5 binds the adiponectin promoter in vivo following GH stimulation. Overall, these data strongly suggest that STAT5 activators regulate adiponectin transcription through the binding of STAT5 to the -3,809 site that leads to decreased adiponectin expression and secretion. These mechanistic observations are highly consistent with studies in mice and humans that have high GH or PRL levels that are accompanied by lower circulating levels of adiponectin.


Asunto(s)
Adipocitos/efectos de los fármacos , Adiponectina/metabolismo , Hormona del Crecimiento/farmacología , Prolactina/farmacología , Factor de Transcripción STAT5/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Ratones , Regiones Promotoras Genéticas
16.
J Biol Chem ; 289(9): 5960-9, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24391115

RESUMEN

Lipocalin-2 (LCN2) is secreted from adipocytes, and its expression is up-regulated in obese and diabetic mice and humans. LCN2 expression and secretion have been shown to be induced by two proinflammatory cytokines, IFNγ and TNFα, in cultured murine and human adipocytes. In these studies, we demonstrated that IFNγ and TNFα induced LCN2 expression and secretion in vivo. Although we observed a strong induction of LCN2 expression and secretion from white adipose tissue (WAT) depots, the induction of LCN2 varied among different insulin-sensitive tissues. Knockdown experiments also demonstrated that STAT1 is required for IFNγ-induced lipocalin-2 expression in murine adipocytes. Similarly, knockdown of p65 in adipocytes demonstrated the necessity of the NF-κB signaling pathway for TNFα-mediated effects on LCN2. Activation of ERKs by IFNγ and TNFα also affected STAT1 and NF-κB signaling through modulation of serine phosphorylation. ERK activation-induced serine phosphorylation of both STAT1 and p65 mediated the additive effects of IFNγ and TNFα on LCN2 expression. Our results suggest that these same mechanisms occur in humans as we observed STAT1 and NF-κB binding to the human LCN2 promoter in chromatin immunoprecipitation assays performed in human fat cells. These studies substantially increase our knowledge regarding the requirements and mechanisms used by proinflammatory cytokines to induce LCN2 expression.


Asunto(s)
Proteínas de Fase Aguda/biosíntesis , Adipocitos/metabolismo , Tejido Adiposo Blanco/metabolismo , Regulación de la Expresión Génica/fisiología , Lipocalinas/biosíntesis , Proteínas Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Elementos de Respuesta/fisiología , Células 3T3-L1 , Proteínas de Fase Aguda/genética , Adipocitos/citología , Tejido Adiposo Blanco/citología , Animales , Técnicas de Silenciamiento del Gen , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Lipocalina 2 , Lipocalinas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Proteínas Oncogénicas/genética , Proteínas Proto-Oncogénicas/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Biochim Biophys Acta ; 1842(3): 431-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23735217

RESUMEN

Adipocytes play important roles in lipid storage, energy homeostasis and whole body insulin sensitivity. The JAK-STAT (Janus Kinase-Signal Transducer and Activator of Transcription) pathway mediates a variety of physiological processes including development, hematopoiesis, and inflammation. Although the JAK-STAT signaling pathway occurs in all cells, this pathway can mediate cell specific responses. Studies in the last two decades have identified hormones and cytokines that activate the JAK-STAT signaling pathway. These cytokines and hormones have profound effects on adipocytes. The content of this review will introduce the types of adipocytes and immune cells that make up adipose tissue, the impact of obesity on adipose cellular composition and function, and the general constituents of the JAK-STAT pathway and how its activators regulate adipose tissue development and physiology. A summary of the identification of STAT target genes in adipocytes reveals how these transcription factors impact various areas of adipocyte metabolism including insulin action, modulation of lipid stores, and glucose homeostasis. Lastly, we will evaluate exciting new data linking the JAK-STAT pathway and brown adipose tissue and consider the future outlook in this area of investigation. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Insulina/metabolismo , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Adipocitos/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Humanos , Insulina/fisiología , Metabolismo de los Lípidos , Transducción de Señal
18.
Am J Physiol Endocrinol Metab ; 309(3): E205-13, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26058863

RESUMEN

Adipose tissue has the largest capacity to store energy in the body and provides energy through the release of free fatty acids during times of energy need. Different types of immune cells are recruited to adipose tissue under various physiological conditions, indicating that these cells contribute to the regulation of adipose tissue. One major pathway influenced by a number of immune cells is the release of free fatty acids through lipolysis during both physiological (e.g., cold stress) and pathophysiological processes (e.g., obesity, type 2 diabetes). Adipose tissue expansion during obesity leads to immune cell infiltration and adipose tissue remodeling, a homeostatic process that promotes inflammation in adipose tissue. The release of proinflammatory cytokines stimulates lipolysis and causes insulin resistance, leading to adipose tissue dysfunction and systemic disruptions of metabolism. This review focuses on the interactions of cytokines and other inflammatory molecules that regulate adipose tissue lipolysis during physiological and pathophysiological states.


Asunto(s)
Adipocitos/metabolismo , Citocinas/metabolismo , Lipólisis , Modelos Biológicos , Paniculitis/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Adipocitos/inmunología , Animales , Humanos , Resistencia a la Insulina , Activación de Macrófagos , Paniculitis/inmunología , Receptores de Reconocimiento de Patrones/agonistas
19.
PLoS Biol ; 10(11): e1001436, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209380

RESUMEN

Obesity is a condition characterized by excess adipose tissue that results from positive energy balance and is the most common metabolic disorder in the industrialized world. The obesity epidemic shows no sign of slowing, and it is increasingly a global problem. Serious clinical problems associated with obesity include an increased risk for type 2 diabetes, atherosclerosis, and cancer. Hence, understanding the origin and development of adipocytes and adipose tissue will be critical to the analysis and treatment of metabolic diseases. Historically, albeit incorrectly, adipocytes were thought to be inert cells whose singular function was lipid storage. It is now known that adipocytes have other critical functions; the most important include sensitivity to insulin and the ability to produce and secrete adipocyte-specific endocrine hormones that regulate energy homeostasis in other tissues. Today, adipocytes are recognized as critical regulators of whole-body metabolism and known to be involved in the pathogenesis of a variety of metabolic diseases. All cells come from other cells and many cells arise from precursor cells. Adipocytes are not created from other adipocytes, but they arise from precursor cells. In the last two decades, scientists have discovered the function of many proteins that influence the ability of precursor cells to become adipocytes. If the expansion of the adipose tissue is the problem, it seems logical that adipocyte development inhibitors could be a viable anti-obesity therapeutic. However, factors that block adipocyte development and limit adipocyte expansion also impair metabolic health. This notion may be counterintuitive, but several lines of evidence support the idea that blocking adipocyte development is unhealthy. For this reason it is clear that we need a better understanding of adipocyte development.


Asunto(s)
Adipocitos/citología , Adipogénesis , Tejido Adiposo/metabolismo , Metabolismo de los Lípidos , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Fármacos Antiobesidad/metabolismo , Linaje de la Célula , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Metabolismo Energético , Humanos , Obesidad/metabolismo , Obesidad/patología , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
J Strength Cond Res ; 29(11): 3097-104, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26274372

RESUMEN

The effects of resistance training on adiponectin have thus far yielded equivocal results. However, the effect of periodized resistance training on plasma adiponectin in offspring of type 2 diabetic and nondiabetic (ND) parents (first-degree family history) has yet to be determined. Untrained healthy young men with and without a first-degree family history of type 2 diabetes were assigned to an exercise intervention group (E) or an active control group (C). The E group performed a 12-week periodized resistance training program, whereas the C group did not participate in any structured exercise program. Fasting plasma samples were obtained pre- and posttraining. Total plasma adiponectin changed significantly in the E group (33.7 ± 14.7%, p = 0.025) but not in the C group (10.8 ± 19.2%). Offspring of type 2 diabetic parents subjects had significantly greater improvements in plasma adiponectin (69.3 ± 34.8%) compared with ND subjects (-3.2 ± 29.9%, p = 0.046). Regular aerobic exercise was significantly correlated with average plasma adiponectin (r = 0.32), whereas first-degree family history of type 2 diabetes (r = -0.58) and decreases in body fat percentage (r = -0.77) were inversely correlated with average plasma adiponectin. Periodized high-intensity resistance training seems to increase plasma adiponectin, particularly in individuals with a first-degree family history of type 2 diabetes or those who experience a significant loss of fat mass. The direct correlation between regular aerobic exercise and adiponectin further suggests that a combination of aerobic and resistance training yields greater improvements in plasma adiponectin compared with resistance training alone.


Asunto(s)
Adiponectina/sangre , Entrenamiento de Fuerza/métodos , Adolescente , Adulto , Distribución de la Grasa Corporal , Diabetes Mellitus Tipo 2 , Humanos , Masculino , Distribución Aleatoria , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA