Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Am J Physiol Cell Physiol ; 320(5): C822-C841, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33439778

RESUMEN

Adipocytes are specialized cells with pleiotropic roles in physiology and pathology. Several types of fat cells with distinct metabolic properties coexist in various anatomically defined fat depots in mammals. White, beige, and brown adipocytes differ in their handling of lipids and thermogenic capacity, promoting differences in size and morphology. Moreover, adipocytes release lipids and proteins with paracrine and endocrine functions. The intrinsic properties of adipocytes pose specific challenges in culture. Mature adipocytes float in suspension culture due to high triacylglycerol content and are fragile. Moreover, a fully differentiated state, notably acquirement of the unilocular lipid droplet of white adipocyte, has so far not been reached in two-dimensional culture. Cultures of mouse and human-differentiated preadipocyte cell lines and primary cells have been established to mimic white, beige, and brown adipocytes. Here, we survey various models of differentiated preadipocyte cells and primary mature adipocyte survival describing main characteristics, culture conditions, advantages, and limitations. An important development is the advent of three-dimensional culture, notably of adipose spheroids that recapitulate in vivo adipocyte function and morphology in fat depots. Challenges for the future include isolation and culture of adipose-derived stem cells from different anatomic location in animal models and humans differing in sex, age, fat mass, and pathophysiological conditions. Further understanding of fat cell physiology and dysfunction will be achieved through genetic manipulation, notably CRISPR-mediated gene editing. Capturing adipocyte heterogeneity at the single-cell level within a single fat depot will be key to understanding diversities in cardiometabolic parameters among lean and obese individuals.


Asunto(s)
Adipocitos/fisiología , Tejido Adiposo/fisiología , Adipogénesis , Tejido Adiposo/citología , Animales , Comunicación Celular , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular , Humanos , Fenotipo , Especificidad de la Especie , Esferoides Celulares , Técnicas de Cultivo de Tejidos
2.
FASEB J ; 33(5): 6269-6280, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30768368

RESUMEN

Paternal preconceptional high-fat diet (HFD) alters whole-body glucose and energy homeostasis over several generations, which may be mediated by altered transcriptomic profiles of metabolic organs. We investigated the effect of paternal HFD on the hepatic transcriptomic and metabolic signatures of female grand-offspring. Paternal HFD strongly impacted the liver transcriptome of the second-generation offspring. Gene set enrichment analysis (GSEA) revealed grandpaternal-HFD altered the TNF-α signaling via NFκB pathway, independent of the grand-offspring's diet. Reduction in the hepatic cytokine levels, including the TNF-α, as well as NFκB content and activity, suggest that the basal inflammatory response in F2 rats is disturbed. GSEA also show altered expression of various genes annotated to the fatty acid metabolism. Grandpaternal-HFD reduced G0/G1 switch gene 2 (G0S2) expression, concomitant with reduced hepatic triglyceride content in F2 rats. In conclusion, the hepatic transcriptome is altered in grand-offspring from HFD-fed grandfathers. Altered TNF-α/NFκB signaling and levels of inflammatory cytokines indicate grandpaternal HFD impacts hepatic immunometabolism. Overall, our findings indicate that paternal exposure to environmental factors transgenerationally reprograms metabolism in a tissue-specific manner, affecting the development and health of future generations.-De Castro Barbosa, T., Alm, P. S., Krook, A., Barrès, R., Zierath, J. R. Paternal high-fat diet transgenerationally impacts hepatic immunometabolism.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hígado/metabolismo , Herencia Paterna , Transcriptoma , Animales , Epigénesis Genética , Femenino , Masculino , FN-kappa B/genética , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
FASEB J ; 32(8): 4158-4171, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29565738

RESUMEN

Maternal polycystic ovary syndrome (PCOS), a condition associated with hyperandrogenism, is suggested to increase anxiety-like behavior in the offspring. Because PCOS is closely linked to obesity, we investigated the impact of an adverse hormonal or metabolic maternal environment and offspring obesity on anxiety in the offspring. The obese PCOS phenotype was induced by chronic high-fat-high-sucrose (HFHS) consumption together with prenatal dihydrotestosterone exposure in mouse dams. Anxiety-like behavior was assessed in adult offspring with the elevated-plus maze and open-field tests. The influence of maternal androgens and maternal and offspring diet on genes implicated in anxiety were analyzed in the amygdala and hypothalamus with real-time PCR ( n = 47). Independent of diet, female offspring exposed to maternal androgens were more anxious and displayed up-regulation of adrenoceptor α 1B in the amygdala and up-regulation of hypothalamic corticotropin-releasing hormone ( Crh). By contrast, male offspring exposed to a HFHS maternal diet had increased anxiety-like behavior and showed up-regulation of epigenetic markers in the amygdala and up-regulation of hypothalamic Crh. Overall, there were substantial sex differences in gene expression in the brain. These findings provide novel insight into how maternal androgens and obesity exert sex-specific effects on behavior and gene expression in the offspring of a PCOS mouse model.-Manti, M., Fornes, R., Qi, X., Folmerz, E., Lindén Hirschberg, A., de Castro Barbosa, T., Maliqueo, M., Benrick, A., Stener-Victorin, E. Maternal androgen excess and obesity induce sexually dimorphic anxiety-like behavior in the offspring.


Asunto(s)
Andrógenos/metabolismo , Ansiedad/etiología , Ansiedad/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Animales , Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Expresión Génica/fisiología , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Relaciones Madre-Hijo , Síndrome del Ovario Poliquístico/metabolismo , Caracteres Sexuales , Regulación hacia Arriba/fisiología
4.
Am J Physiol Endocrinol Metab ; 310(1): E51-60, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26530149

RESUMEN

Decrease of AMPK-related signal transduction and insufficient lipid oxidation contributes to the pathogenesis of obesity and type 2 diabetes. Previously, we identified that diacylglycerol kinase-δ (DGKδ), an enzyme involved in triglyceride biosynthesis, is reduced in skeletal muscle from type 2 diabetic patients. Here, we tested the hypothesis that DGKδ plays a role in maintaining appropriate AMPK action in skeletal muscle and energetic aspects of contraction. Voluntary running activity was reduced in DGKδ(+/-) mice, but glycogen content and mitochondrial markers were unaltered, suggesting that DGKδ deficiency affects skeletal muscle energetics but not mitochondrial protein abundance. We next determined the role of DGKδ in AMPK-related signal transduction and lipid metabolism in isolated skeletal muscle. AMPK activation and signaling were reduced in DGKδ(+/-) mice, concomitant with impaired lipid oxidation and elevated incorporation of free fatty acids into triglycerides. Strikingly, DGKδ deficiency impaired work performance, as evident by altered force production and relaxation dynamics in response to repeated contractions. In conclusion, DGKδ deficiency impairs AMPK signaling and lipid metabolism, thereby highlighting the deleterious role of excessive lipid metabolites in the development of peripheral insulin resistance and type 2 diabetes pathogenesis. DGKδ deficiency also influences skeletal muscle energetics, which may lead to low physical activity levels in type 2 diabetes.


Asunto(s)
Adenilato Quinasa/metabolismo , Diacilglicerol Quinasa/fisiología , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Músculo Esquelético/metabolismo , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diacilglicerol Quinasa/genética , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/genética , Condicionamiento Físico Animal/fisiología , Transducción de Señal/genética
5.
Am J Physiol Cell Physiol ; 307(9): C774-87, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25143351

RESUMEN

Two-dimensional difference gel electrophoresis (2-D DIGE)-based proteome analysis has revealed intrinsic insulin resistance in myotubes derived from type 2 diabetic patients. Using 2-D DIGE-based proteome analysis, we identified a subset of insulin-resistant proteins involved in protein turnover in skeletal muscle of type 2 diabetic patients, suggesting aberrant regulation of the protein homeostasis maintenance system underlying metabolic disease. We then validated the role of the ubiquitin-proteasome system (UPS) in myotubes to investigate whether impaired proteasome function may lead to metabolic arrest or insulin resistance. Myotubes derived from muscle biopsies obtained from people with normal glucose tolerance (NGT) or type 2 diabetes were exposed to the proteasome inhibitor bortezomib (BZ; Velcade) without or with insulin. BZ exposure increased protein carbonylation and lactate production yet impaired protein synthesis and UPS function in myotubes from type 2 diabetic patients, marking the existence of an insulin-resistant signature that was retained in cultured myotubes. In conclusion, BZ treatment further exacerbates insulin resistance and unmasks intrinsic features of metabolic disease in myotubes derived from type 2 diabetic patients. Our results highlight the existence of a confounding inherent abnormality in cellular protein dynamics in metabolic disease, which is uncovered through concurrent inhibition of the proteasome system.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Músculo Esquelético/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Ácidos Borónicos/farmacología , Bortezomib , Células Cultivadas , Diabetes Mellitus Tipo 2/enzimología , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Glucógeno/biosíntesis , Humanos , Insulina/farmacología , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Estrés Oxidativo/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Carbonilación Proteica/efectos de los fármacos , Proteoma/metabolismo , Pirazinas/farmacología , Interferencia de ARN , Transducción de Señal
6.
Metabolism ; 158: 155939, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38843995

RESUMEN

BACKGROUND AND AIM: Diacylglycerol kinase (DGK) isoforms catalyze an enzymatic reaction that removes diacylglycerol (DAG) and thereby terminates protein kinase C signaling by converting DAG to phosphatidic acid. DGKδ (type II isozyme) downregulation causes insulin resistance, metabolic inflexibility, and obesity. Here we determined whether DGKδ overexpression prevents these metabolic impairments. METHODS: We generated a transgenic mouse model overexpressing human DGKδ2 under the myosin light chain promoter (DGKδ TG). We performed deep metabolic phenotyping of DGKδ TG mice and wild-type littermates fed chow or high-fat diet (HFD). Mice were also provided free access to running wheels to examine the effects of DGKδ overexpression on exercise-induced metabolic outcomes. RESULTS: DGKδ TG mice were leaner than wild-type littermates, with improved glucose tolerance and increased skeletal muscle glycogen content. DGKδ TG mice were protected against HFD-induced glucose intolerance and obesity. DGKδ TG mice had reduced epididymal fat and enhanced lipolysis. Strikingly, DGKδ overexpression recapitulated the beneficial effects of exercise on metabolic outcomes. DGKδ overexpression and exercise had a synergistic effect on body weight reduction. Microarray analysis of skeletal muscle revealed common gene ontology signatures of exercise and DGKδ overexpression that were related to lipid storage, extracellular matrix, and glycerophospholipids biosynthesis pathways. CONCLUSION: Overexpression of DGKδ induces adaptive changes in both skeletal muscle and adipose tissue, resulting in protection against HFD-induced obesity. DGKδ overexpression recapitulates exercise-induced adaptations on energy homeostasis and skeletal muscle gene expression profiles.

7.
Cell Rep Med ; 5(1): 101348, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-38151020

RESUMEN

The discovery of exercise-regulated circulatory factors has fueled interest in organ crosstalk, especially between skeletal muscle and adipose tissue, and the role in mediating beneficial effects of exercise. We studied the adipose tissue transcriptome in men and women with normal glucose tolerance or type 2 diabetes following an acute exercise bout, revealing substantial exercise- and time-dependent changes, with sustained increase in inflammatory genes in type 2 diabetes. We identify oncostatin-M as one of the most upregulated adipose-tissue-secreted factors post-exercise. In cultured human adipocytes, oncostatin-M enhances MAPK signaling and regulates lipolysis. Oncostatin-M expression arises predominantly from adipose tissue immune cell fractions, while the corresponding receptors are expressed in adipocytes. Oncostatin-M expression increases in cultured human Thp1 macrophages following exercise-like stimuli. Our results suggest that immune cells, via secreted factors such as oncostatin-M, mediate a crosstalk between skeletal muscle and adipose tissue during exercise to regulate adipocyte metabolism and adaptation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Femenino , Humanos , Masculino , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Lipólisis
8.
Adipocyte ; : 2283213, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37982546

RESUMEN

BACKGROUND: Mature adipocytes are notoriously difficult to study ex vivo and alternative cell culture systems have therefore been developed. One of the most common models are human adipose progenitor cells (hAPCs). Unfortunately, these display replicative senescence after prolonged culture conditions, which limits their use in mechanistic studies. METHODS: Herein, we knocked in human telomerase reverse transcriptase (TERT) into the AAVS1 locus of CD55+ hAPCs derived from abdominal subcutaneous adipose tissue and characterized the cells before and after differentiation into adipocytes. RESULTS: Immortalized TERT-hAPCs retained proliferative and adipogenic capacities comparable to those of early-passage wild type hAPCs for > 80 passages. In line with this, our integrative transcriptomic and proteomic analyses revealed that TERT-hAPCs displayed robust adipocyte expression profiles in comparison to wild type hAPCs. This was confirmed by functional analyses of lipid turnover where TERT-hAPCs exhibited pronounced responses to insulin and pro-lipolytic stimuli such as isoprenaline, dibutyrul cAMP and tumour necrosis factor alpha. In addition, TERT-hAPCs could be readily cultured in both standard 2D and 3D-cultures and proteomic analyses revealed that the spheroid culture conditions improved adipogenesis. CONCLUSION: Through descriptive and functional studies, we demonstrate that immortalization of human CD55+ hAPCs is feasible and results in cells with stable proliferative and adipogenic capacities over multiple passages. As these cells are cryopreservable, they provide the additional advantage over primary cells of allowing repeated studies in both 2D and 3D model systems with the same genetic background. (234/250).

9.
Diabetologia ; 59(1): 5-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26489734
10.
Adv Sci (Weinh) ; 8(16): e2100106, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34165908

RESUMEN

Obesity and type 2 diabetes are strongly associated with adipose tissue dysfunction and impaired adipogenesis. Understanding the molecular underpinnings that control adipogenesis is thus of fundamental importance for the development of novel therapeutics against metabolic disorders. However, translational approaches are hampered as current models do not accurately recapitulate adipogenesis. Here, a scaffold-free versatile 3D adipocyte culture platform with chemically defined conditions is presented in which primary human preadipocytes accurately recapitulate adipogenesis. Following differentiation, multi-omics profiling and functional tests demonstrate that 3D adipocyte cultures feature mature molecular and cellular phenotypes similar to freshly isolated mature adipocytes. Spheroids exhibit physiologically relevant gene expression signatures with 4704 differentially expressed genes compared to conventional 2D cultures (false discovery rate < 0.05), including the concerted expression of factors shaping the adipogenic niche. Furthermore, lipid profiles of >1000 lipid species closely resemble patterns of the corresponding isogenic mature adipocytes in vivo (R2 = 0.97). Integration of multi-omics signatures with analyses of the activity profiles of 503 transcription factors using global promoter motif inference reveals a complex signaling network, involving YAP, Hedgehog, and TGFß signaling, that links the organotypic microenvironment in 3D culture to the activation and reinforcement of PPARγ and CEBP activity resulting in improved adipogenesis.


Asunto(s)
Adipogénesis/fisiología , Tejido Adiposo/patología , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Humanos , Transducción de Señal/fisiología
11.
J Cachexia Sarcopenia Muscle ; 12(5): 1232-1248, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34342159

RESUMEN

BACKGROUND: Type 2 diabetes and obesity are often seen concurrently with skeletal muscle wasting, leading to further derangements in function and metabolism. Muscle wasting remains an unmet need for metabolic disease, and new approaches are warranted. The neuropeptide urocortin 2 (UCN2) and its receptor corticotropin releasing factor receptor 2 (CRHR2) are highly expressed in skeletal muscle and play a role in regulating energy balance, glucose metabolism, and muscle mass. The aim of this study was to investigate the effects of modified UCN2 peptides as a pharmaceutical therapy to counteract the loss of skeletal muscle mass associated with obesity and casting immobilization. METHODS: High-fat-fed mice (C57Bl/6J; 26 weeks old) and ob/ob mice (11 weeks old) were injected daily with a PEGylated (Compound A) and non-PEGylated (Compound B) modified human UCN2 at 0.3 mg/kg subcutaneously for 14 days. A separate group of chow-fed C57Bl/6J mice (12 weeks old) was subjected to hindlimb cast immobilization and, after 1 week, received daily injections with Compound A. In vivo functional tests were performed to measure protein synthesis rates and skeletal muscle function. Ex vivo functional and molecular tests were performed to measure contractile force and signal transduction of catabolic and anabolic pathways in skeletal muscle. RESULTS: Skeletal muscles (extensor digitorum longus, soleus, and tibialis anterior) from high-fat-fed mice treated with Compound A were ~14% heavier than muscles from vehicle-treated mice. Chronic treatment with modified UCN2 peptides altered the expression of structural genes and transcription factors in skeletal muscle in high-fat diet-induced obesity including down-regulation of Trim63 and up-regulation of Nr4a2 and Igf1 (P < 0.05 vs. vehicle). Signal transduction via both catabolic and anabolic pathways was increased in tibialis anterior muscle, with increased phosphorylation of ribosomal protein S6 at Ser235/236 , FOXO1 at Ser256 , and ULK1 at Ser317 , suggesting that UCN2 treatment modulates protein synthesis and degradation pathways (P < 0.05 vs. vehicle). Acutely, a single injection of Compound A in drug-naïve mice had no effect on the rate of protein synthesis in skeletal muscle, as measured via the surface sensing of translation method, while the expression of Nr4a3 and Ppargc1a4 was increased (P < 0.05 vs. vehicle). Compound A treatment prevented the loss of force production from disuse due to casting. Compound B treatment increased time to fatigue during ex vivo contractions of fast-twitch extensor digitorum longus muscle. Compound A and B treatment increased lean mass and rates of skeletal muscle protein synthesis in ob/ob mice. CONCLUSIONS: Modified human UCN2 is a pharmacological candidate for the prevention of the loss of skeletal muscle mass associated with obesity and immobilization.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético , Obesidad/tratamiento farmacológico , Obesidad/etiología , Péptidos , Urocortinas
12.
Am J Physiol Endocrinol Metab ; 298(1): E8-E16, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19861587

RESUMEN

Expression of an activated form of calcineurin in skeletal muscle selectively up-regulates slow-fiber-specific gene expression. Here, we tested the hypothesis that expression of activated calcineurin in skeletal muscle influences body composition, energy homeostasis, and exercise performance. Using transgenic mice expressing activated calcineurin (CnA*) in skeletal muscle (MCK-CnA* transgenic mice), we determined whether skeletal muscle reprogramming by calcineurin activation affects exercise performance and skeletal muscle mitochondrial function. Body weight and extensor digitorum longus (EDL) skeletal muscle weight were reduced 10% in MCK-CnA* mice compared with wild-type littermates. Basal oxygen consumption, food intake, and voluntary exercise behavior were unchanged between MCK-CnA* and wild-type mice. However, when total energy expenditure was normalized by fat-free mass, energy expenditure was increased in MCK-CnA* mice. An endurance performance treadmill running test revealed MCK-CnA* mice are fatigue resistant and run 50% farther before exhaustion. After a standardized exercise bout, glycogen and triglyceride content in EDL muscle was higher in MCK-CnA* vs. wild-type mice. Mitochondrial respiratory capacity was increased 35% in EDL muscle from resting MCK-CnA* mice. In conclusion, our results provide evidence to support the hypothesis that calcineurin activation in skeletal muscle increases mitochondrial oxidative function and energy substrate storage, which contributes to enhanced endurance exercise performance. These adaptive changes occur as a consequence of a lifelong expression of a constitutively active calcineurin and mimic the response to chronic endurance training.


Asunto(s)
Calcineurina/genética , Calcineurina/metabolismo , Mitocondrias/fisiología , Músculo Esquelético/fisiología , Resistencia Física/fisiología , Aciltransferasas/metabolismo , Animales , Peso Corporal/fisiología , Diacilglicerol O-Acetiltransferasa/metabolismo , Transporte de Electrón/fisiología , Metabolismo Energético/fisiología , Proteínas de Transporte de Ácidos Grasos/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucógeno/metabolismo , Ratones , Ratones Transgénicos , Contracción Muscular/fisiología , Músculo Esquelético/anatomía & histología , Tamaño de los Órganos/fisiología , Consumo de Oxígeno/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal/fisiología , Transactivadores/metabolismo , Factores de Transcripción , Triglicéridos/metabolismo
13.
Diabetes ; 68(7): 1403-1414, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31010957

RESUMEN

The neuropeptide urocortin 2 (UCN2) and its receptor corticotropin-releasing hormone receptor 2 (CRHR2) are highly expressed in skeletal muscle and play a role in regulating energy balance and glucose metabolism. We investigated a modified UCN2 peptide as a potential therapeutic agent for the treatment of obesity and insulin resistance, with a specific focus on skeletal muscle. High-fat-fed mice (C57BL/6J) were injected daily with a PEGylated UCN2 peptide (compound A) at 0.3 mg/kg subcutaneously for 14 days. Compound A reduced body weight, food intake, whole-body fat mass, and intramuscular triglycerides compared with vehicle-treated controls. Furthermore, whole-body glucose tolerance was improved by compound A treatment, with increased insulin-stimulated Akt phosphorylation at Ser473 and Thr308 in skeletal muscle, concomitant with increased glucose transport into extensor digitorum longus and gastrocnemius muscle. Mechanistically, this is linked to a direct effect on skeletal muscle because ex vivo exposure of soleus muscle from chow-fed lean mice to compound A increased glucose transport and insulin signaling. Moreover, exposure of GLUT4-Myc-labeled L6 myoblasts to compound A increased GLUT4 trafficking. Our results demonstrate that modified UCN2 peptides may be efficacious in the treatment of type 2 diabetes by acting as an insulin sensitizer in skeletal muscle.


Asunto(s)
Glucosa/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Urocortinas/farmacología , Animales , Western Blotting , Composición Corporal/efectos de los fármacos , Electroporación , Células HEK293 , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Urocortinas/química
14.
Rev Inst Med Trop Sao Paulo ; 49(6): 371-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18157404

RESUMEN

BACKGROUND: The pathogenesis of chronic hepatitis C is still a matter of debate. CD4+ and CD8+ T lymphocytes (TL) are typically observed within the portal and periportal spaces of affected livers, but their functional role in hepatitis C progression has not been fully elucidated. METHODS: CD4+ and CD8+ TL were quantified by immunohistochemistry in portal and periportal spaces of 39 liver biopsies from patients with chronic hepatitis C. They were associated to demographic data, histological parameters, laboratory findings of patients and hepatitis C genotypes. RESULTS: There was high numbers of CD4+ and CD8+ TL from which the density of CD4+ T was higher than CD8+ TL in portal and periportal spaces. CD4+ and CD8+ TL were directly correlated to intensity of interface hepatitis. CD8+ TL correlated to serum enzyme levels. CONCLUSION: The high numbers of CD4+ and CD8+ TL in portal and periportal spaces and their correlation to interface hepatitis suggest that hepatitis C evolution depends on the action of intrahepatic T lymphocytes, lending support to the notion of an immune-mediated mechanism in the pathogenesis of chronic hepatitis C.


Asunto(s)
Relación CD4-CD8 , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Hígado/virología , Adolescente , Adulto , Progresión de la Enfermedad , Femenino , Genotipo , Hepacivirus/genética , Hepatitis C Crónica/patología , Humanos , Inmunohistoquímica , Hígado/irrigación sanguínea , Hígado/patología , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
15.
Life Sci ; 191: 17-23, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-28993146

RESUMEN

L-Arginine has emerged as an important supplement for athletes and non-athletes in order to improve performance. Arginine has been extensively used as substrate for nitric oxide synthesis, leading to increased vasodilatation and hormonal secretion. However, the chronic consumption of arginine has been shown to impair insulin sensitivity. In the present study, we aimed to evaluate whether chronic arginine supplementation associated with exercise training would have a beneficial impact on insulin sensitivity. We, therefore, treated Wistar rats for 4weeks with arginine, associated or not with exercise training (treadmill). We assessed the somatotropic activation, by evaluating growth hormone (GH) gene expression and protein content in the pituitary, as well is GH concentration in the serum. Additionally, we evaluate whole-body insulin sensitivity, by performing an insulin tolerance test. Skeletal muscle morpho-physiological parameters were also assessed. Insulin sensitivity was impaired in the arginine-treated rats. However, exercise training reversed the negative effects of arginine. Arginine and exercise training increased somatotropic axis function, muscle mass and body weight gain. The combination arginine and exercise training further decreased total fat mass. Our results confirm that chronic arginine supplementation leads to insulin resistance, which can be reversed in the association with exercise training. We provide further evidence that exercise training is an important tool to improve whole-body metabolism.


Asunto(s)
Arginina/efectos adversos , Suplementos Dietéticos/efectos adversos , Resistencia a la Insulina , Músculo Esquelético/fisiología , Animales , Regulación de la Expresión Génica , Hormona del Crecimiento/análisis , Hormona del Crecimiento/sangre , Hormona del Crecimiento/genética , Insulina/metabolismo , Masculino , Condicionamiento Físico Animal , Esfuerzo Físico , Ratas Wistar
16.
Mol Metab ; 6(7): 621-630, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28702319

RESUMEN

OBJECTIVE: Parental nutrition and lifestyle impact the metabolic phenotype of the offspring. We have reported that grandpaternal chronic high-fat diet (HFD) transgenerationally impairs glucose metabolism in subsequent generations. Here we determined whether grandpaternal diet transgenerationally impacts the transcriptome and lipidome in skeletal muscle. Our aim was to identify tissue-specific pathways involved in transgenerational inheritance of environmental-induced phenotypes. METHODS: F0 male Sprague-Dawley rats were fed a HFD or chow for 12 weeks before breeding with chow-fed females to generate the F1 generation. F2 offspring were generated by mating F1 males fed a chow diet with an independent line of chow-fed females. F1 and F2 offspring were fed chow or HFD for 12 weeks. Transcriptomic and LC-MS lipidomic analyses were performed in extensor digitorum longus muscle from F2-females rats. Gene set enrichment analysis (GSEA) was performed to determine pathways reprogrammed by grandpaternal diet. RESULTS: GSEA revealed an enrichment of the unfolded protein response pathway in skeletal muscle of grand-offspring from HFD-fed grandfathers compared to grand-offspring of chow-fed males. Activation of the stress sensor (ATF6α), may be a pivotal point whereby this pathway is activated. Interestingly, skeletal muscle from F1-offspring was not affected in a similar manner. No major changes were observed in the skeletal muscle lipidome profile due to grandpaternal diet. CONCLUSIONS: Grandpaternal HFD-induced obesity transgenerationally affected the skeletal muscle transcriptome. This finding further highlights the impact of parental exposure to environmental factors on offspring's development and health.


Asunto(s)
Dieta Alta en Grasa , Impresión Genómica , Músculo Esquelético/metabolismo , Obesidad/genética , Respuesta de Proteína Desplegada/genética , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Animales , Femenino , Glucosa/metabolismo , Metabolismo de los Lípidos , Masculino , Ratas , Ratas Sprague-Dawley , Transcriptoma
17.
Life Sci ; 79(15): 1444-9, 2006 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-16723138

RESUMEN

Arginine (Arg) presents a potent growth hormone (GH) releasing activity. In vivo and in vitro studies carried out in our laboratory have demonstrated that acute treatment with Arg also increases GH gene expression. Taking into account the recognizable diabetogenic role of GH and that Arg increases insulin release, this study aimed at evaluating the effects of oral chronic administration of Arg on GH gene expression, by Northern blotting analysis, and on the insulin sensitivity, by means of the Insulin Tolerance Test (ITT), blood glucose decay rate (kitt) and insulin plasma concentration. We demonstrated that rats that consumed Arg ( approximately 35 mg/day) in drinking water, during 4 weeks, presented an increase in GH mRNA content (p < 0.01), a decreased peripheral response to insulin, as shown by the reduced blood glucose decay rate (p < 0.05), and a higher insulin plasma concentration (p < 0.01) than control group. Arg treatment did not modify the animals' food and water intake, while it decreased the heart rate and the arterial blood pressure compared to control group (p < 0.05). According to the results presented herein we conclude that chronic oral administration of arginine increases GH gene expression and induces insulin resistance. The arterial blood pressure decrease has already been pointed out in the literature, and seems to occur in response to the dilating effect of nitric oxide generated from Arg, as well as from NO generation in central and peripheral neuronal populations that express NOS and are involved in the autonomic regulation of the cardiac function.


Asunto(s)
Arginina/administración & dosificación , Hormona del Crecimiento/genética , Resistencia a la Insulina/genética , Administración Oral , Animales , Arginina/farmacología , Glucemia/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Insulina/sangre , Masculino , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
18.
Mol Metab ; 5(3): 184-197, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26977389

RESUMEN

OBJECTIVES: Chronic and high consumption of fat constitutes an environmental stress that leads to metabolic diseases. We hypothesized that high-fat diet (HFD) transgenerationally remodels the epigenome of spermatozoa and metabolism of the offspring. METHODS: F0-male rats fed either HFD or chow diet for 12 weeks were mated with chow-fed dams to generate F1 and F2 offspring. Motile spermatozoa were isolated from F0 and F1 breeders to determine DNA methylation and small non-coding RNA (sncRNA) expression pattern by deep sequencing. RESULTS: Newborn offspring of HFD-fed fathers had reduced body weight and pancreatic beta-cell mass. Adult female, but not male, offspring of HFD-fed fathers were glucose intolerant and resistant to HFD-induced weight gain. This phenotype was perpetuated in the F2 progeny, indicating transgenerational epigenetic inheritance. The epigenome of spermatozoa from HFD-fed F0 and their F1 male offspring showed common DNA methylation and small non-coding RNA expression signatures. Altered expression of sperm miRNA let-7c was passed down to metabolic tissues of the offspring, inducing a transcriptomic shift of the let-7c predicted targets. CONCLUSION: Our results provide insight into mechanisms by which HFD transgenerationally reprograms the epigenome of sperm cells, thereby affecting metabolic tissues of offspring throughout two generations.

19.
Metabolism ; 62(1): 79-89, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22889511

RESUMEN

OBJECTIVE: The amino acid Arginine (Arg) is the main biological precursor of nitric oxide (NO) and has been described to improve insulin sensitivity in diabetes and obesity. We investigated the molecular mechanisms involved in the long-term effects of Arg on glucose and lipid metabolism. MATERIALS AND METHODS: L6 myotubes were treated with Arg (7 mmol/L) for 6 days. D-Mannitol (7 mmol/L) was used as control; spermine NONOate (10 µmol/L) and L-NAME (100 µmol/L) were used to evaluate the NO/c-GMP pathway role. Basal and insulin-induced (120 nmol/L) glycogen synthesis, glucose uptake and lipid oxidation, c-GMP and nitrite levels, and the intracellular signaling pathways were evaluated. RESULTS: Arg-treatment increased: 1) basal and insulin-stimulated glycogen synthesis; 2) glucose uptake; 3) palmitate oxidation; 4) p-Akt (Ser(473)), total and plasma membrane GLUT4 content, total and p-AMPK-α and p-ACC (Ser(79)), p-GSK-3α/ß (Ser(21/9)) and 5) nitrite and c-GMP levels. L-NAME treatment suppressed Arg effects on: 1) nitrite and c-GMP content; 2) glycogen synthesis and glucose uptake; 3) basal and insulin-stimulated p-Akt (Ser(473)), total and p-AMPK-α and ACC, and nNOS expression. CONCLUSION: We provide evidence that Arg improves glucose and lipid metabolism in skeletal muscle, in parallel with increased phosphorylation of Akt and AMPK-α. These effects were mediated by the NO/c-GMP pathway. Thus, arginine treatment enhances signal transduction and has a beneficial effect of metabolism in skeletal muscle through direct activation of Akt and AMPK pathways.


Asunto(s)
Glucosa/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Espermina/análogos & derivados , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Arginina/antagonistas & inhibidores , Arginina/farmacología , Western Blotting , Línea Celular , Transportador de Glucosa de Tipo 4/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Espermina/farmacología
20.
Endocrinology ; 150(5): 2080-6, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19106217

RESUMEN

We have shown that rats chronically treated with Arginine (Arg), although normoglycemic, exhibit hyperinsulinemia and decreased blood glucose disappearance rate after an insulin challenge. Attempting to investigate the processes underlying these alterations, male Wistar rats were treated with Arg (35 mg/d), in drinking water, for 4 wk. Rats were then acutely stimulated with insulin, and the soleus and extensorum digitalis longus muscles, white adipose tissue (WAT), and liver were excised for total and/or phosphorylated insulin receptor (IR), IR substrate 1/2, Akt, Janus kinase 2, signal transducer and activator of transcription (STAT) 1/3/5, and p85alpha/55alpha determination. Muscles and WAT were also used for plasma membrane (PM) and microsome evaluation of glucose transporter (GLUT) 4 content. Pituitary GH mRNA, GH, and liver IGF-I mRNA expression were estimated. It was shown that Arg treatment: 1) did not affect phosphotyrosine-IR, whereas it decreased phosphotyrosine-IR substrate 1/2 and phosphoserine-Akt content in all tissues studied, indicating that insulin signaling is impaired at post-receptor level; 2) decreased PM GLUT4 content in both muscles and WAT; 3) increased the pituitary GH mRNA, GH, and liver IGF-I mRNA expression, the levels of phosphotyrosine-STAT5 in both muscles, phosphotyrosine-Janus kinase 2 in extensorum digitalis longus, phosphotyrosine-STAT3 in liver, and WAT as well as total p85alpha in soleus, indicating that GH signaling is enhanced in these tissues; and 4) increased p55alpha total content in muscles, WAT, and liver. The present findings provide the molecular mechanisms by which insulin resistance and, by extension, reduced GLUT4 content in PM of muscles and WAT take place after chronic administration of Arg, and further suggest a putative role for GH in its genesis, considering its diabetogenic effect.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Arginina/farmacología , Hormona del Crecimiento/fisiología , Insulina/metabolismo , Hígado/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Regulación hacia Abajo/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA