Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 42(2): e110553, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36504224

RESUMEN

Epithelial-mesenchymal transition (EMT) is pivotal in the initiation and development of cancer cell metastasis. We observed that the abundance of glycosphingolipids (GSLs), especially ganglioside subtypes, decreased significantly during TGF-ß-induced EMT in NMuMG mouse mammary epithelial cells and A549 human lung adenocarcinoma cells. Transcriptional profiling showed that TGF-ß/SMAD response genes and EMT signatures were strongly enriched in NMuMG cells, along with depletion of UDP-glucose ceramide glucosyltransferase (UGCG), the enzyme that catalyzes the initial step in GSL biosynthesis. Consistent with this finding, genetic or pharmacological inhibition of UGCG promoted TGF-ß signaling and TGF-ß-induced EMT. UGCG inhibition promoted A549 cell migration, extravasation in the zebrafish xenograft model, and metastasis in mice. Mechanistically, GSLs inhibited TGF-ß signaling by promoting lipid raft localization of the TGF-ß type I receptor (TßRI) and by increasing TßRI ubiquitination and degradation. Importantly, we identified ST3GAL5-synthesized a-series gangliosides as the main GSL subtype involved in inhibition of TGF-ß signaling and TGF-ß-induced EMT in A549 cells. Notably, ST3GAL5 is weakly expressed in lung cancer tissues compared to adjacent nonmalignant tissues, and its expression correlates with good prognosis.


Asunto(s)
Neoplasias Pulmonares , Factor de Crecimiento Transformador beta , Humanos , Animales , Ratones , Factor de Crecimiento Transformador beta/metabolismo , Gangliósidos , Transición Epitelial-Mesenquimal/genética , Pez Cebra/metabolismo , Neoplasias Pulmonares/metabolismo , Glicoesfingolípidos , Catálisis , Movimiento Celular , Línea Celular Tumoral
2.
FASEB J ; 29(7): 3027-39, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25852044

RESUMEN

Chronic low-grade inflammation associated with obesity contributes to insulin resistance and type 2 diabetes. Helminth parasites are the strongest natural inducers of type 2 immune responses, and short-lived infection with rodent nematodes was reported to improve glucose tolerance in obese mice. Here, we investigated the effects of chronic infection (12 weeks) with Schistosoma mansoni, a helminth that infects millions of humans worldwide, on whole-body metabolic homeostasis and white adipose tissue (WAT) immune cell composition in high-fat diet-induced obese C57BL/6 male mice. Our data indicate that chronic helminth infection reduced body weight gain (-62%), fat mass gain (-89%), and adipocyte size; lowered whole-body insulin resistance (-23%) and glucose intolerance (-16%); and improved peripheral glucose uptake (+25%) and WAT insulin sensitivity. Analysis of immune cell composition by flow cytometry and quantitative PCR (qPCR) revealed that S. mansoni promoted strong increases in WAT eosinophils and alternatively activated (M2) macrophages. Importantly, injections with S. mansoni-soluble egg antigens (SEA) recapitulated the beneficial effect of parasite infection on whole-body metabolic homeostasis and induced type 2 immune responses in WAT and liver. Taken together, we provide novel data suggesting that chronic helminth infection and helminth-derived molecules protect against metabolic disorders by promoting a T helper 2 (Th2) response, eosinophilia, and WAT M2 polarization.


Asunto(s)
Antígenos Helmínticos/administración & dosificación , Resistencia a la Insulina/inmunología , Obesidad/complicaciones , Obesidad/inmunología , Esquistosomiasis mansoni/complicaciones , Esquistosomiasis mansoni/inmunología , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/patología , Animales , Enfermedad Crónica , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina/fisiología , Hígado/inmunología , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/metabolismo , Células Th2/inmunología
3.
Diabetologia ; 58(1): 165-77, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25316433

RESUMEN

AIMS/HYPOTHESIS: South Asians have a higher risk of developing type 2 diabetes than Europeans. The underlying cause of this excess risk is still poorly understood but might be related to differences in the regulation of energy/nutrient-sensing pathways in metabolic tissues and subsequent changes in whole-body substrate metabolism. In this study, we investigated the whole-body and skeletal muscle metabolic adaptations to short-term energy restriction in South Asian and European volunteers. METHODS: Twenty-four middle-aged overweight South Asian and European men underwent a two-step hyperinsulinaemic-euglycaemic clamp, with skeletal muscle biopsies and indirect calorimetry before and after an 8 day diet very low in energy (very low calorie diet [VLCD]). Abdominal fat distribution and hepatic triacylglycerol content were assessed using MRI and MR spectroscopy. RESULTS: South Asian men had higher hepatic triacylglycerol content than European men, and exhibited elevated clamp insulin levels that probably reflect a lower insulin clearance rate. Despite higher insulin levels, endogenous glucose production rate was similar and glucose disposal rate (Rd) and nonoxidative glucose disposal rate (NOGD) were significantly lower in South Asian than European men, indicating impaired whole-body insulin sensitivity. Energy restriction decreased abdominal fat mass and hepatic triacylglycerol content in both groups. However, the shift induced by energy restriction from glucose towards lipid oxidation observed in European men was impaired in South Asian men, indicating whole-body metabolic inflexibility. Remarkably, although energy restriction improved hepatic insulin sensitivity in both groups, Rd improved only in South Asian men owing to higher NOGD. At the molecular level, an increase in insulin-induced activation of the skeletal muscle mTOR pathway was found in South Asian men, showing that skeletal muscle energy/nutrient-sensing pathways were differentially affected by energy restriction. CONCLUSIONS/INTERPRETATION: We conclude that South Asian men exhibit a different metabolic adaptation to short-term energy restriction than European men. TRIAL REGISTRATION: Dutch trial registry ( www.trialregister.nl ), trial number NTR 2473.


Asunto(s)
Adaptación Fisiológica/fisiología , Pueblo Asiatico , Restricción Calórica , Sobrepeso/dietoterapia , Sobrepeso/etnología , Sobrepeso/metabolismo , Población Blanca , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Asia/etnología , Restricción Calórica/etnología , Humanos , Metabolismo de los Lípidos , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Países Bajos , Transducción de Señal
4.
Am J Physiol Endocrinol Metab ; 307(10): E885-95, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25249505

RESUMEN

During fasting, rapid metabolic adaptations are required to maintain energy homeostasis. This occurs by a coordinated regulation of energy/nutrient-sensing pathways leading to transcriptional activation and repression of specific sets of genes. The aim of the study was to investigate how short-term fasting affects whole body energy homeostasis and skeletal muscle energy/nutrient-sensing pathways and transcriptome in humans. For this purpose, 12 young healthy men were studied during a 24-h fast. Whole body glucose/lipid oxidation rates were determined by indirect calorimetry, and blood and skeletal muscle biopsies were collected and analyzed at baseline and after 10 and 24 h of fasting. As expected, fasting induced a time-dependent decrease in plasma insulin and leptin levels, whereas levels of ketone bodies and free fatty acids increased. This was associated with a metabolic shift from glucose toward lipid oxidation. At the molecular level, activation of the protein kinase B (PKB/Akt) and mammalian target of rapamycin pathways was time-dependently reduced in skeletal muscle during fasting, whereas the AMP-activated protein kinase activity remained unaffected. Furthermore, we report some changes in the phosphorylation and/or content of forkhead protein 1, sirtuin 1, and class IIa histone deacetylase 4, suggesting that these pathways might be involved in the transcriptional adaptation to fasting. Finally, transcriptome profiling identified genes that were significantly regulated by fasting in skeletal muscle at both early and late time points. Collectively, our study provides a comprehensive map of the main energy/nutrient-sensing pathways and transcriptomic changes during short-term adaptation to fasting in human skeletal muscle.


Asunto(s)
Adaptación Fisiológica/fisiología , Metabolismo Energético/fisiología , Ayuno/metabolismo , Músculo Esquelético/metabolismo , Adaptación Fisiológica/genética , Calorimetría Indirecta , Metabolismo Energético/genética , Ácidos Grasos no Esterificados/sangre , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Homeostasis , Humanos , Insulina/sangre , Cuerpos Cetónicos/sangre , Leptina/sangre , Masculino , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Serina-Treonina Quinasas TOR/genética , Adulto Joven
5.
Lab Chip ; 24(3): 422-433, 2024 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-38087979

RESUMEN

Within the tumor microenvironment (TME), cancer cells use mechanotransduction pathways to convert biophysical forces to biochemical signals. However, the underlying mechanisms and functional significance of these pathways remain largely unclear. The upregulation of mechanosensitive pathways from biophysical forces such as interstitial flow (IF), leads to the activation of various cytokines, including transforming growth factor-ß (TGF-ß). TGF-ß promotes in part via a Smad-dependent signaling pathway the epithelial-mesenchymal transition (EMT) in cancer cells. The latter process is linked to increased cancer cell motility and invasion. Current research models have limited ability to investigate the combined effects of biophysical forces (such as IF) and cytokines (TGF-ß) in a 3D microenvironment. We used a 3D-matrix based microfluidic platform to demonstrate the potentiating effect of IF on exogenous TGF-ß induced upregulation of the Smad-signaling activity and the expression of mesenchymal marker vimentin in A549 lung cancer spheroids. To monitor this, we used stably integrated fluorescent based reporters into the A549 cancer cell genome. Our results demonstrate that IF enhances exogenous TGF-ß induced Smad-signaling activity in lung cancer spheroids embedded in a matrix microenvironment. In addition, we observed an increased cell motility for A549 spheroids when exposed to IF and TGF-ß. Our 3D-microfluidic model integrated with real-time imaging provides a powerful tool for investigating cancer cell signaling and motility associated with invasion characteristics in a physiologically relevant TME.


Asunto(s)
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/patología , Factor de Crecimiento Transformador beta/genética , Microfluídica , Mecanotransducción Celular , Línea Celular Tumoral , Transducción de Señal , Citocinas , Transición Epitelial-Mesenquimal , Movimiento Celular , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Microambiente Tumoral
6.
Am J Physiol Endocrinol Metab ; 304(9): E1012-21, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23512807

RESUMEN

Obesity in humans is often associated with metabolic inflexibility, but the underlying molecular mechanisms remain incompletely understood. The aim of the present study was to investigate how adaptation to prolonged fasting affects energy/nutrient-sensing pathways and metabolic gene expression in skeletal muscle from lean and obese individuals. Twelve lean and 14 nondiabetic obese subjects were fasted for 48 h. Whole body glucose/lipid oxidation rates were determined by indirect calorimetry, and blood and skeletal muscle biopsies were collected and analyzed. In response to fasting, body weight loss was similar in both groups, but the decrease in plasma insulin and leptin and the concomitant increase in growth hormone were significantly attenuated in obese subjects. The fasting-induced shift from glucose toward lipid oxidation was also severely blunted. At the molecular level, the expression of insulin receptor-ß (IRß) was lower in skeletal muscle from obese subjects at baseline, whereas the fasting-induced reductions in insulin signaling were similar in both groups. The protein expression of mitochondrial respiratory chain components, although not modified by fasting, was significantly reduced in obese subjects. Some minor differences in metabolic gene expression were observed at baseline and in response to fasting. Surprisingly, fasting reduced AMPK activity in lean but not in obese subjects, whereas the expression of AMPK subunits was not affected. We conclude that whole body metabolic inflexibility in response to prolonged fasting in obese humans is associated with lower skeletal muscle IRß and mitochondrial respiratory chain content as well as a blunted decline of AMPK activity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/fisiología , Transporte de Electrón/fisiología , Ayuno/metabolismo , Expresión Génica/fisiología , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Transducción de Señal/fisiología , Adulto , Antígenos CD/biosíntesis , Antígenos CD/genética , Western Blotting , Composición Corporal/fisiología , Peso Corporal/fisiología , Calorimetría Indirecta , Femenino , Glucosa/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Peroxidación de Lípido/fisiología , Masculino , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Insulina/biosíntesis , Receptor de Insulina/genética , Factores de Transcripción/metabolismo
7.
Immunol Cell Biol ; 91(7): 486-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23835553

RESUMEN

Recent reports have attributed an immunoregulatory role to the mammalian target of rapamycin (mTOR), a key serine/threonine protein kinase integrating input from growth factors and nutrients to promote cell growth and differentiation. In the present study, we investigated the role of the mTOR pathway in Th2 induction by human monocyte-derived dendritic cells (moDCs). Using a co-culture system of human lipopolysaccharide (LPS)-matured moDCs and allogeneic naive CD4(+) T cells, we show that inhibition of mTOR by the immunosuppressive drug rapamycin reduced moDC maturation and promoted Th2 skewing. Next, we investigated whether antigens from helminth parasites, the strongest natural inducers of Th2 responses, modulate moDCs via the mTOR pathway. In contrast to rapamycin, neither Schistosoma mansoni-soluble egg antigens (SEA) nor its major immunomodulatory component omega-1 affected the phosphorylation of S6 kinase (S6K) and 4E-binding protein 1 (4E-BP1), downstream targets of mTORC1. Finally, we found that the effects of rapamycin and SEA/omega-1 on Th2 skewing were additive, suggesting two distinct underlying molecular mechanisms. We conclude that conditioning human moDCs to skew immune responses towards Th2 can be achieved via an mTOR-dependent and -independent pathway triggered by rapamycin and helminth antigens, respectively.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Schistosoma mansoni/inmunología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos Helmínticos/inmunología , Proteínas de Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Proteínas del Huevo/inmunología , Proteínas del Helminto/inmunología , Humanos , Isoantígenos/inmunología , Lipopolisacáridos/inmunología , Fosfoproteínas/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/inmunología , Balance Th1 - Th2/efectos de los fármacos
8.
Signal Transduct Target Ther ; 7(1): 126, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35484112

RESUMEN

Ovo-like transcriptional repressor 1 (OVOL1) is a key mediator of epithelial lineage determination and mesenchymal-epithelial transition (MET). The cytokines transforming growth factor-ß (TGF-ß) and bone morphogenetic proteins (BMP) control the epithelial-mesenchymal plasticity (EMP) of cancer cells, but whether this occurs through interplay with OVOL1 is not known. Here, we show that OVOL1 is inversely correlated with the epithelial-mesenchymal transition (EMT) signature, and is an indicator of a favorable prognosis for breast cancer patients. OVOL1 suppresses EMT, migration, extravasation, and early metastatic events of breast cancer cells. Importantly, BMP strongly promotes the expression of OVOL1, which enhances BMP signaling in turn. This positive feedback loop is established through the inhibition of TGF-ß receptor signaling by OVOL1. Mechanistically, OVOL1 interacts with and prevents the ubiquitination and degradation of SMAD family member 7 (SMAD7), which is a negative regulator of TGF-ß type I receptor stability. Moreover, a small-molecule compound 6-formylindolo(3,2-b)carbazole (FICZ) was identified to activate OVOL1 expression and thereby antagonizing (at least in part) TGF-ß-mediated EMT and migration in breast cancer cells. Our results uncover a novel mechanism by which OVOL1 attenuates TGF-ß/SMAD signaling and maintains the epithelial identity of breast cancer cells.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Unión al ADN , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Factores de Transcripción , Factor de Crecimiento Transformador beta/genética
9.
J Vis Exp ; (168)2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33720132

RESUMEN

In response to specific external cues and the activation of certain transcription factors, endothelial cells can differentiate into a mesenchymal-like phenotype, a process that is termed endothelial to mesenchymal transition (EndMT). Emerging results have suggested that EndMT is causally linked to multiple human diseases, such as fibrosis and cancer. In addition, endothelial-derived mesenchymal cells may be applied in tissue regeneration procedures, as they can be further differentiated into various cell types (e.g., osteoblasts and chondrocytes). Thus, the selective manipulation of EndMT may have clinical potential. Like epithelial-mesenchymal transition (EMT), EndMT can be strongly induced by the secreted cytokine transforming growth factor-beta (TGF-ß), which stimulates the expression of so-called EndMT transcription factors (EndMT-TFs), including Snail and Slug. These EndMT-TFs then up- and downregulate the levels of mesenchymal and endothelial proteins, respectively. Here, we describe methods to investigate TGF-ß-induced EndMT in vitro, including a protocol to study the role of particular TFs in TGF-ß-induced EndMT. Using these techniques, we provide evidence that TGF-ß2 stimulates EndMT in murine pancreatic microvascular endothelial cells (MS-1 cells), and that the genetic depletion of Snail using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated gene editing, abrogates this phenomenon. This approach may serve as a model to interrogate potential modulators of endothelial biology, and can be used to perform genetic or pharmacological screens in order to identify novel regulators of EndMT, with potential application in human disease.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Células Endoteliales/metabolismo , Edición Génica , Mesodermo/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Técnica del Anticuerpo Fluorescente , Ratones , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/genética
10.
Front Cell Dev Biol ; 9: 616610, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33644053

RESUMEN

Endothelial-to-mesenchymal transition (EndMT) plays an important role in embryonic development and disease progression. Yet, how different members of the transforming growth factor-ß (TGF-ß) family regulate EndMT is not well understood. In the current study, we report that TGF-ß2, but not bone morphogenetic protein (BMP)9, triggers EndMT in murine endothelial MS-1 and 2H11 cells. TGF-ß2 strongly upregulates the transcription factor SNAIL, and the depletion of Snail is sufficient to abrogate TGF-ß2-triggered mesenchymal-like cell morphology acquisition and EndMT-related molecular changes. Although SLUG is not regulated by TGF-ß2, knocking out Slug also partly inhibits TGF-ß2-induced EndMT in 2H11 cells. Interestingly, in addition to SNAIL and SLUG, BMP9 stimulates inhibitor of DNA binding (ID) proteins. The suppression of Id1, Id2, or Id3 expression facilitated BMP9 in inducing EndMT and, in contrast, ectopic expression of ID1, ID2, or ID3 abrogated TGF-ß2-mediated EndMT. Altogether, our results show that SNAIL is critical and indispensable for TGF-ß2-mediated EndMT. Although SLUG is also involved in the EndMT process, it plays less of a crucial role in it. In contrast, ID proteins are essential for maintaining endothelial traits and repressing the function of SNAIL and SLUG during the EndMT process. These data suggest that the control over endothelial vs. mesenchymal cell states is determined, at least in part, by a balance between the expression of SNAIL/SLUG and ID proteins.

11.
J Vis Exp ; (164)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33191940

RESUMEN

Transforming growth factor-ß (TGF-ß) is a secreted multifunctional factor that plays a key role in intercellular communication. Perturbations of TGF-ß signaling can lead to breast cancer. TGF-ß elicits its effects on proliferation and differentiation via specific cell surface TGF-ß type I and type II receptors (i.e., TßRI and TßRII) that contain an intrinsic serine/threonine kinase domain. Upon TGF-ß-induced heteromeric complex formation, activated TßRI elicits intracellular signaling by phosphorylating SMAD2 and SMAD3. These activated SMADs form heteromeric complexes with SMAD4 to regulate specific target genes, including plasminogen activation inhibitor 1 (PAI-1, encoded by the SERPINE1 gene). The induction of epithelial-to-mesenchymal transition (EMT) allows epithelial cancer cells at the primary site or during colonization at distant sites to gain an invasive phenotype and drive tumor progression. TGF-ß acts as a potent inducer of breast cancer invasion by driving EMT. Here, we describe systematic methods to investigate TGF-ß signaling and EMT responses using premalignant human MCF10A-RAS (M2) cells and mouse NMuMG epithelial cells as examples. We describe methods to determine TGF-ß-induced SMAD2 phosphorylation by Western blotting, SMAD3/SMAD4-dependent transcriptional activity using luciferase reporter activity and SERPINE1 target gene expression by quantitative real-time-polymerase chain reaction (qRT-PCR). In addition, methods are described to examine TGF-ß-induced EMT by measuring changes in morphology, epithelial and mesenchymal marker expression, filamentous actin staining and immunofluorescence staining of E-cadherin. Two selective small molecule TGF-ß receptor kinase inhibitors, GW788388 and SB431542, were used to block TGF-ß-induced SMAD2 phosphorylation, target genes and changes in EMT marker expression. Moreover, we describe the transdifferentiation of mesenchymal breast Py2T murine epithelial tumor cells into adipocytes. Methods to examine TGF-ß-induced signaling and EMT in breast cancer may contribute to new therapeutic approaches for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Benzamidas/farmacología , Dioxoles/farmacología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Ratones , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Transcripción Genética/efectos de los fármacos
12.
Dis Model Mech ; 13(9)2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32820031

RESUMEN

Zebrafish models are well-established tools for investigating the underlying mechanisms of diseases. Here, we identified cercosporamide, a metabolite from the fungus Ascochyta aquiliqiae, as a potent bone morphogenetic protein receptor (BMPR) type I kinase inhibitor through a zebrafish embryo phenotypic screen. The developmental defects in zebrafish, including lack of the ventral fin, induced by cercosporamide were strikingly similar to the phenotypes caused by renowned small-molecule BMPR type I kinase inhibitors and inactivating mutations in zebrafish BMPRs. In mammalian cell-based assays, cercosporamide blocked BMP/SMAD-dependent transcriptional reporter activity and BMP-induced SMAD1/5-phosphorylation. Biochemical assays with a panel of purified recombinant kinases demonstrated that cercosporamide directly inhibited kinase activity of type I BMPRs [also called activin receptor-like kinases (ALKs)]. In mammalian cells, cercosporamide selectively inhibited constitutively active BMPR type I-induced SMAD1/5 phosphorylation. Importantly, cercosporamide rescued the developmental defects caused by constitutively active Alk2 in zebrafish embryos. We believe that cercosporamide could be the first of a new class of molecules with potential to be developed further for clinical use against diseases that are causally linked to overactivation of BMPR signaling, including fibrodysplasia ossificans progressiva and diffuse intrinsic pontine glioma.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Benzofuranos/farmacología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Pez Cebra/metabolismo , Animales , Benzofuranos/química , Benzofuranos/aislamiento & purificación , Bioensayo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/metabolismo , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
13.
Diabetes ; 55(12): 3221-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17130464

RESUMEN

Clinical insulin resistance is associated with decreased activation of phosphatidylinositol 3'-kinase (PI3K) and its downstream substrate protein kinase B (PKB)/Akt. However, its physiological protein substrates remain poorly characterized. In the present study, the effect of in vivo insulin action on phosphorylation of the PKB/Akt substrate 40 (PRAS40) was examined. In rat and mice, insulin stimulated PRAS40-Thr246 phosphorylation in skeletal and cardiac muscle, the liver, and adipose tissue in vivo. Physiological hyperinsulinemia increased PRAS40-Thr246 phosphorylation in human skeletal muscle biopsies. In cultured cell lines, insulin-mediated PRAS40 phosphorylation was prevented by the PI3K inhibitors wortmannin and LY294002. Immunohistochemical and immunofluorescence studies showed that phosphorylated PRAS40 is predominantly localized to the nucleus. Finally, in rats fed a high-fat diet (HFD), phosphorylation of PRAS40 was markedly reduced compared with low-fat diet-fed animals in all tissues examined. In conclusion, the current study identifies PRAS40 as a physiological target of in vivo insulin action. Phosphorylation of PRAS40 is increased by insulin in human, rat, and mouse insulin target tissues. In rats, this response is reduced under conditions of HFD-induced insulin resistance.


Asunto(s)
Proteínas en la Dieta , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Humanos , Insulina/fisiología , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar
14.
Mol Endocrinol ; 20(8): 1786-95, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16601071

RESUMEN

The stimulation of cells with physiological concentrations of insulin induces a variety of responses, e.g. an increase in glucose uptake, induction of glycogen and protein synthesis, and gene expression. One of the determinants regulating insulin-mediated gene expression may be activating transcription factor 2 (ATF2). Insulin activates ATF2 by phosphorylation of Thr69 and Thr71 via a two-step mechanism, in which ATF2-Thr71 phosphorylation precedes the induction of ATF2-Thr69+71 phosphorylation by several minutes. We previously found that in c-Jun N-terminal kinase (JNK)-/- fibroblasts, cooperation of the ERK1/2 and p38 pathways is required for two-step ATF2-Thr69+71 phosphorylation in response to growth factors. Because JNK is also capable of phosphorylating ATF2, we assessed the involvement of JNK, ERK1/2 and p38 in the insulin-induced two-step ATF2 phosphorylation in JNK-expressing A14 fibroblasts and 3T3L1-adipocytes. The induction of ATF2-Thr71 phosphorylation was sensitive to MAPK kinase (MEK) 1/2-inhibition with U0126, and this phosphorylation coincided with nuclear translocation of phosphorylated ERK1/2. Use of the JNK inhibitor SP600125 or expression of dominant-negative JNK-activator SAPK kinase (SEK1) prevented the induction of ATF2-Thr69+71, but not ATF2-Thr71 phosphorylation by insulin. ATF2-dependent transcription was also sensitive to SP-treatment. Abrogation of p38 activation with SB203580 or expression of dominant-negative MKK6 had no inhibitory effect on these events. In agreement with this, the onset of ATF2-Thr69+71 phosphorylation coincided with the nuclear translocation of phosphorylated JNK. Finally, in vitro kinase assays using nuclear extracts indicated that ERK1/2 preceded JNK translocation. We conclude that sequential activation and nuclear appearance of ERK1/2 and JNK, rather than p38, underlies the two-step insulin-induced ATF2 phosphorylation in JNK-expressing cells.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Insulina/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Fosforilación/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Células 3T3-L1 , Transporte Activo de Núcleo Celular , Animales , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Modelos Biológicos , Células 3T3 NIH , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Factores de Tiempo , Transfección
15.
Diabetes ; 66(12): 2942-2951, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28882901

RESUMEN

Obstructive sleep apnea syndrome is a highly prevalent disease resulting in transient respiratory arrest and chronic intermittent hypoxia (cIH). cIH is associated with insulin resistance and impaired metabolic homeostasis in rodents and humans, but the exact underlying mechanisms remain unclear. In the current study, we investigated the effects of 2 weeks of cIH (1-min cycle, fraction of inspired oxygen 21-5%, 8 h/day) on whole-body insulin sensitivity and glucose tolerance in lean mice. Although food intake and body weight were reduced compared with normoxia, cIH induced systemic insulin resistance in a hypoxia-inducible factor 1-independent manner and impaired insulin signaling in liver, white adipose tissue, and skeletal muscle. Unexpectedly, cIH improved whole-body glucose tolerance independently of changes in body weight and glucose-induced insulin response. This effect was associated with elevated phosphorylation of Thr172-AMPK and Ser237-TBC1 domain family member 1 (TBC1D1) in skeletal muscle, suggesting a tissue-specific AMPK-dependent increase in TBC1D1-driven glucose uptake. Remarkably, although food intake, body weight, and systemic insulin sensitivity were still affected, the improvement in glucose tolerance by cIH was abolished in muscle-specific AMPKα1α2-deficient mice. We conclude that cIH impairs insulin sensitivity while improving whole-body glucose tolerance by promoting specific activation of the skeletal muscle AMPK pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Hipoxia/metabolismo , Resistencia a la Insulina , Músculo Esquelético/enzimología , Animales , Enfermedad Crónica , Activación Enzimática , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
16.
Sci Rep ; 6: 24618, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27094951

RESUMEN

Obesity is associated with local tissue hypoxia and elevated hypoxia-inducible factor 1 alpha (HIF-1α) in metabolic tissues. Prolyl hydroxylases (PHDs) play an important role in regulating HIF-α isoform stability. In the present study, we investigated the consequence of whole-body PHD1 gene (Egln2) inactivation on metabolic homeostasis in mice. At baseline, PHD1-/- mice exhibited higher white adipose tissue (WAT) mass, despite lower body weight, and impaired insulin sensitivity and glucose tolerance when compared to age-matched wild-type (WT) mice. When fed a synthetic low-fat diet, PHD1-/- mice also exhibit a higher body weight gain and WAT mass along with glucose intolerance and systemic insulin resistance compared to WT mice. PHD1 deficiency led to increase in glycolytic gene expression, lipogenic proteins ACC and FAS, hepatic steatosis and liver-specific insulin resistance. Furthermore, gene markers of inflammation were also increased in the liver, but not in WAT or skeletal muscle, of PHD1-/- mice. As expected, high-fat diet (HFD) promoted obesity, hepatic steatosis, tissue-specific inflammation and systemic insulin resistance in WT mice but these diet-induced metabolic alterations were not exacerbated in PHD1-/- mice. In conclusion, PHD1 deficiency promotes hepatic steatosis and liver-specific insulin resistance but does not worsen the deleterious effects of HFD on metabolic homeostasis.


Asunto(s)
Hígado Graso/genética , Hígado Graso/metabolismo , Resistencia a la Insulina/genética , Procolágeno-Prolina Dioxigenasa/deficiencia , Adiposidad , Animales , Peso Corporal , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Dislipidemias/metabolismo , Metabolismo Energético , Hígado Graso/patología , Intolerancia a la Glucosa/metabolismo , Homeostasis , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Especificidad de Órganos/genética
17.
J Clin Endocrinol Metab ; 88(9): 4251-7, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12970295

RESUMEN

We analyzed the insulin receptor gene in four patients with leprechaunism and one with type A insulin resistance. We detected novel and previously reported mutations. The novel mutants were expressed in Chinese hamster ovary cells to evaluate the consequences for insulin receptor function. A type A insulin resistance patient from Morocco was homozygous for Arg252His mutation, similar to a previously described type A patient from Japan. A patient with leprechaunism was homozygous for the Ser323Leu mutation, previously identified in homozygous form in two patients with Rabson-Mendenhall syndrome. Phenotypic expression of this mutation is variable. A patient with leprechaunism is compound heterozygous for the previously described Arg1092Trp mutation and a nonsense mutation in codon 897. Another patient with leprechaunism was homozygous for a novel Asn431Asp mutation, which only partially reduces insulin proreceptor processing and activation of signaling cascades. The novel Leu93Gln mutation that fully disrupts proreceptor processing was found in one allele in a patient with leprechaunism. A nonsense mutation at codon 1122 was in the other allele. These results expand the number of pathogenic insulin receptor mutations and demonstrate the variability in their phenotypic expression. The biochemical analysis of mutant insulin receptors does not reliably predict whether the phenotype will be leprechaunism, the Rabson-Mendenhall syndrome, or type A insulin resistance. The previously reported correlation between fibroblast insulin binding and duration of patient survival was not observed.


Asunto(s)
Resistencia a la Insulina/genética , Mutación/fisiología , Receptor de Insulina/genética , Adolescente , Animales , Western Blotting , Células CHO , Células Cultivadas , Codón sin Sentido/genética , Codón sin Sentido/fisiología , Cricetinae , ADN/biosíntesis , ADN/genética , Femenino , Fibroblastos , Humanos , Hipoglucemiantes/farmacología , Lactante , Insulina/farmacología , Mutación/genética , Mutación Missense/genética , Mutación Missense/fisiología , Fenotipo , Fosfotirosina/metabolismo , Receptor de Insulina/biosíntesis , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
18.
Biochem Pharmacol ; 68(1): 105-12, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15183122

RESUMEN

Recently, it was shown that rottlerin inhibits insulin-stimulated glucose uptake and reduces intracellular adenosine triphosphate (ATP) levels in 3T3-L1 adipocytes, suggesting that these two events are causally linked. However, several other reports show that ATP-depletion induces glucose uptake in both muscle cells and adipocytes. In the present study, the mechanism of inhibition by rottlerin was studied in detail, in order to resolve this apparent discrepancy. It was found that rottlerin strongly reduces insulin-stimulated 2-deoxyglucose (2-DOG) uptake in 3T3-L1 adipocytes by a partial inhibition of the translocation of the insulin-responsive GLUT4 glucose transporter towards the plasma membrane (PM). Whereas the insulin-induced phosphatidyl-inositol-3' (PI-3') kinase signaling pathway is unaffected by rottlerin, Cbl tyrosine phosphorylation, which provides an essential, PI-3' kinase-independent signal towards GLUT4 translocation, is markedly attenuated. Furthermore, we also observed a direct inhibitory effect of rottlerin on insulin-induced glucose uptake in 3T3-L1 adipocytes. The direct inhibition of insulin-stimulated 2-DOG uptake by rottlerin displayed characteristics of uncompetitive inhibition: with the K(m(app)) of glucose uptake reduced from 1.6 to 0.9 mM and the V(max(app)) reduced from 5.2 to 1.0 nmol/minmg in the presence of rottlerin. In conclusion, rottlerin inhibits multiple steps involved in insulin-stimulated 2-DOG uptake in 3T3-L1 adipocytes. The observed reduction in GLUT4 translocation towards the PM and the uncompetitive inhibition of the glucose transport process provide alternative explanations for the inhibitory effects of rottlerin aside from the effects of rottlerin on intracellular levels of ATP.


Asunto(s)
Acetofenonas/farmacología , Adipocitos/efectos de los fármacos , Benzopiranos/farmacología , Glucosa/metabolismo , Insulina/farmacología , Proteínas Musculares , Transducción de Señal/efectos de los fármacos , Células 3T3 , Adipocitos/metabolismo , Animales , Transporte Biológico , Interacciones Farmacológicas , Transportador de Glucosa de Tipo 4 , Cinética , Ratones , Proteínas de Transporte de Monosacáridos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
19.
Diabetes ; 63(1): 248-58, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24357702

RESUMEN

South Asians (SAs) develop type 2 diabetes at a younger age and lower BMI compared with Caucasians (Cs). The underlying cause is still poorly understood but might result from an innate inability to adapt to the Westernized diet. This study aimed to compare the metabolic adaptation to a high-fat, high-calorie (HFHC) diet between both ethnicities. Twelve healthy, young lean male SAs and 12 matched Cs underwent a two-step hyperinsulinemic-euglycemic clamp with skeletal muscle biopsies and indirect calorimetry before and after a 5-day HFHC diet. Hepatic triglyceride content (HTG) and abdominal fat distribution were assessed using magnetic resonance imaging and spectroscopy. At baseline, SAs had higher insulin clamp levels than Cs, indicating reduced insulin clearance rate. Despite the higher insulin levels, endogenous glucose production was comparable between groups, suggesting lower hepatic insulin sensitivity in SAs. Furthermore, a 5-day HFHC diet decreased the insulin-stimulated (nonoxidative) glucose disposal rate only in SA. In skeletal muscle, no significant differences were found between groups in insulin/mammalian target of rapamycin signaling, metabolic gene expression, and mitochondrial respiratory chain content. Furthermore, no differences in (mobilization of) HTG and abdominal fat were detected. We conclude that HFHC feeding rapidly induces insulin resistance only in SAs. Thus, distinct adaptation to Western food may partly explain their propensity to develop type 2 diabetes.


Asunto(s)
Pueblo Asiatico , Dieta Alta en Grasa , Dieta/etnología , Resistencia a la Insulina/etnología , Metabolismo de los Lípidos/fisiología , Población Blanca , Adulto , Glucemia/metabolismo , Técnica de Clampeo de la Glucosa , Humanos , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Triglicéridos/metabolismo
20.
Diabetes ; 63(3): 880-91, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24270984

RESUMEN

Metformin is the first-line drug for the treatment of type 2 diabetes. Besides its well-characterized antihyperglycemic properties, metformin also lowers plasma VLDL triglyceride (TG). In this study, we investigated the underlying mechanisms in APOE*3-Leiden.CETP mice, a well-established model for human-like lipoprotein metabolism. We found that metformin markedly lowered plasma total cholesterol and TG levels, an effect mostly due to a decrease in VLDL-TG, whereas HDL was slightly increased. Strikingly, metformin did not affect hepatic VLDL-TG production, VLDL particle composition, and hepatic lipid composition but selectively enhanced clearance of glycerol tri[(3)H]oleate-labeled VLDL-like emulsion particles into brown adipose tissue (BAT). BAT mass and lipid droplet content were reduced in metformin-treated mice, pointing to increased BAT activation. In addition, both AMP-activated protein kinase α1 (AMPKα1) expression and activity and HSL and mitochondrial content were increased in BAT. Furthermore, therapeutic concentrations of metformin increased AMPK and HSL activities and promoted lipolysis in T37i differentiated brown adipocytes. Collectively, our results identify BAT as an important player in the TG-lowering effect of metformin by enhancing VLDL-TG uptake, intracellular TG lipolysis, and subsequent mitochondrial fatty acid oxidation. Targeting BAT might therefore be considered as a future therapeutic strategy for the treatment of dyslipidemia.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Hipoglucemiantes/farmacología , Lipoproteínas VLDL/metabolismo , Metformina/farmacología , Triglicéridos/sangre , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Células Cultivadas , Colesterol/sangre , Dislipidemias/tratamiento farmacológico , Femenino , Lipólisis/efectos de los fármacos , Lipoproteína Lipasa/fisiología , Ratones , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA