Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cell Biochem Funct ; 40(8): 903-913, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36169111

RESUMEN

Semaglutide (GLP-1 agonist) was approved for treating obesity. Although the effects on weight loss and metabolism are known, the responses of adipocytes to semaglutide are yet limited. C57BL/6 male mice (n = 20/group) were fed a control diet (C) or a high-fat (HF) diet for 16 weeks and then separated into four groups (n = 10/group) for an additional four weeks: C, C diet and semaglutide, HF, and HF diet and semaglutide. Epididymal white adipose tissue (eWAT) and subcutaneous white adipose tissue (sWAT) fat pads were studied with biochemistry, immunohistochemistry/fluorescence, stereology, and reverse transcription-quantitative polymerase chain reaction. In obese mice, semaglutide reduced the fat pad masses (eWAT, -55%; sWAT, -40%), plasmatic cytokines, and proinflammatory gene expressions: tumor necrosis factor-alpha (-60%); interleukin (IL)-6 (-55%); IL-1 beta (-40%); monocyte chemoattractant protein-1 (-90%); and leptin (-80%). Semaglutide also lessened endoplasmic reticulum (ER) stress genes of activating transcription factor-4 (-85%), CCAAT enhancer-binding protein homologous protein (-55%), and growth arrest and DNA damage-inducible gene 45 (-45%). The obese mice's adipocyte hypertrophy and macrophage infiltration were equally reduced by semaglutide. Semaglutide enhanced multiloculation and uncoupled protein 1 (UCP1) labeling in obese mice: peroxisome proliferator-activated receptor-alpha (+560%) and gamma (+150%), fibronectin type III domain-containing protein 5 (+215%), peroxisome proliferator-activated receptor-alpha coactivator (+110%), nuclear respiratory factor 1 (+260%), and mitochondrial transcription factor A (+120%). Semaglutide also increased thermogenetic gene expressions for the browning phenotype maintenance: beta-3 adrenergic receptor (+520%), PR domain containing 16 (+90%), and Ucp1 (+110%). In conclusion, semaglutide showed significant beneficial effects beyond weight loss, directly on fat pads and adipocytes of obese mice, remarkably anti-inflammatory, and reduced adipocyte size and ER stress. Besides, semaglutide activated adipocyte browning, improving UCP1, mitochondrial biogenesis, and thermogenic marker expressions help weight loss.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón , Grasa Intraabdominal , Animales , Masculino , Ratones , Dieta Alta en Grasa , Estrés del Retículo Endoplásmico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Inflamación/tratamiento farmacológico , Grasa Intraabdominal/metabolismo , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Receptores Activados del Proliferador del Peroxisoma , Grasa Subcutánea , Pérdida de Peso , Tejido Adiposo Pardo
2.
Hepatol Res ; 47(9): 928-940, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27653239

RESUMEN

AIM: The aim of this study was to investigate the effects of rosuvastatin in a model of diet-induced obesity and non-alcoholic fatty liver disease, with attention to the activation of hepatic stellate cells (HSCs). METHOD: Male C57BL/6 mice received a control diet (C; 10% energy as lipids) or a high-fat diet (HF; 50% energy as lipids) for 12 weeks, followed by 7 weeks of treatment. Group CR received control diet + rosuvastatin; group HFR received high-fat diet + rosuvastatin. RESULTS: The HF group showed higher insulin, total cholesterol, triacylglycerol, and leptin levels than the C group, all of which were significantly diminished by rosuvastatin in the HFR group. The HF group had greater steatosis and activated HSCs than the C group, whereas rosuvastatin diminished the steatosis (less 21%, P < 0.001) and significantly inhibited the activation of the HSCs in the HFR group compared to the HF group. The sterol regulatory element-binding protein-1 and the peroxisome proliferator-activated receptor (PPAR)-γ protein expressions were increased in HF animals and reduced after treatment in the HFR group. By contrast, low PPAR-α and carnitine palmitoyltransferase-1 expressions were found in the HF group, and were restored by rosuvastatin treatment in the HFR group. CONCLUSION: Rosuvastatin mitigated hepatic steatosis by modulating PPAR balance, favoring PPAR-α over PPAR-γ downstream effects. The effects were accompanied by a diminishing of insulin resistance, the anti-inflammatory adipokine profile, and HSC activation, avoiding non-alcoholic fatty liver disease progression and non-alcoholic steatohepatitis onset in this model.

3.
Metab Brain Dis ; 32(5): 1735-1745, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28681199

RESUMEN

The brain is very sensitive to metabolic dysfunctions induced by diets high in saturated fatty acids, leading to neuroinflammation. The liraglutide has been found to have neuroprotective effects. However, its neuroprotective action in a model of palmitate-induced neuroinflammation had not yet been evaluated. Mice were intracerebroventricular (ICV) infused with palmitate and received subcutaneous liraglutide. The hippocampal dentate gyrus and CA1 regions were analyzed (morphology and inflammation-related proteins in microglia and astrocyte by confocal microscopy). Also, a real-time PCR was performed to measure the levels of tumor necrosis factor (TNF) alpha and interleukin (IL) 6. Palmitate ICV infusion resulted in pronounced inflammation response in the hippocampus, reactive microgliosis, and astrogliosis, with hypertrophied IBA1 immunoreactive microglia, increased microglial density with ameboid shape, decreased in the number of branches and junctions and increased the major histocompatibility complex (MHC) II expression. Also, we observed in the hippocampus of ICV palmitate infused mice an elevation in the pro-inflammatory cytokine levels TNFalpha and IL6. Liraglutide induced the neuroprotective microglial phenotype, characterized by an increased microglia complexity (enlarged Feret's diameter), an improved number of both cell junctions and processes, and lower circularity, accompanied by a significant reduction in TNFalpha and IL6 expressions. The study provides evidence that liraglutide may be a suitable treatment against the palmitate-induced neuroinflammation, which it is characterized by the reactive microgliosis and astrogliosis, as well as increased pro-inflammatory cytokines, which has been described as one of the primary causes of several pathologies of the central nervous system.


Asunto(s)
Hipocampo/patología , Inflamación/tratamiento farmacológico , Liraglutida/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Región CA1 Hipocampal/patología , Proteínas de Unión al Calcio/metabolismo , Giro Dentado/patología , Genes MHC Clase II/genética , Gliosis/patología , Inflamación/prevención & control , Inyecciones Intraventriculares , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/ultraestructura , Palmitatos , Reacción en Cadena de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo
4.
Hepatol Res ; 44(4): 450-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23607320

RESUMEN

AIM: Weight cycling (WC) is frequent in obesity treatment. We evaluated the degree of regression of the liver damage in WC. METHODS: C57BL/6 male mice received standard chow (SC, 10% energy from lipids) or high-fat diet (HF, 60% energy from lipids) for 6 months (SC6 or HF6) or a diet that alternated every 2 months (SC2/HF2/SC2 or HF2/SC2/HF2). RESULTS: The body mass gain followed the HF intake and induced WC in the animals. The liver alanine aminotransferase, triglyceride and cholesterol levels were higher in the groups receiving the HF diet for any period. The plasma insulin and glucose levels were the highest in the HF6 and HF2/SC2/HF2 groups. Any HF intake increased the liver mass. All the groups had some degree of liver steatosis, with the SC6 group exhibiting the lowest level (∼23% compared with 50-70%). The activated hepatic stellate cells were sparse throughout the liver sections from the HF6 and HF2/SC2/HF2 groups. The lowest sterol regulatory element-binding protein-1c (SREBP-1c) level was detected in the SC6 group. The peroxisome proliferator-activated receptor (PPAR)-α expression was higher in the SC6 and SC2/HF2/SC2 groups than in the HF6 and HF2/SC2/HF2 groups that showed reduced expression. CONCLUSION: WC induced by diet leads to adverse response in the liver, including biochemical and molecular alterations that are not reversed during the lean period of the WC, which must be maintained for a long period to allow the liver to recover from the damage associated with obesity.

5.
Brain Res ; 1846: 149225, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39243951

RESUMEN

We studied the effect of semaglutide (glucagon-like peptide type 1 agonist) on hypothalamic pro-inflammatory genes in diet-induced obese mice. Male C57BL/6J mice were fed a control (C) or high-fat (HF) diet for 16 weeks, then divided into six groups and maintained for an additional four-week study: C, C+semaglutide (CS), C pair-feeding (CP), HF, HF+semaglutide (HFS), and HF pair-feeding (HFP).Weight gain (WG), food efficiency (FE), and plasmatic biochemistry were determined. The hypothalamus was removed and prepared for molecular analysis. Semaglutide reduced WG and FE in the HF group. High cytokines levels (tumor necrosis factor alpha, TNF alpha, monocyte chemoattractant protein 1, MCP1, and Resistin) in HF mice were reduced in HFS mice. High pro-inflammatory gene expressions were seen in HF (toll-like receptor 4, Tlr4; Mcp1; interleukin 6, Il6; Tnfa), inflammasome complex (Pirina domain-containing receptor 3, Nlrp3; Caspase 1, Il1b, Il18), and microglial activation (ionized calcium-binding adapter molecule 1, Iba1; cluster differentiation 68, Cd68; argirase 1, Arg1) but mitigated in HFS. The principal components analysis (PCA) based on these markers in a PC1 x PC2 scatterplot put HF and HFP together but far away from a cluster formed by C and HFS, indicating little significance of weight loss (HFP) but decisive action of semaglutide (HFS) in the results. In conclusion, semaglutide benefits hypothalamic pro-inflammatory genes, inflammasome complex, and microglial activation independent of the weight loss effect. Since GLP-1 receptor agonists such as semaglutide are already indicated to treat obesity and diabetes, the potential translational effects on neuroinflammation should be considered.

6.
Eur J Nutr ; 52(8): 1891-900, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23291721

RESUMEN

PURPOSE: This study aimed to evaluate the role of caffeine chronic administration during gestation of C57BL/6 mice on cardiac remodeling and the expression of components of the renin-angiotensin system (RAS) in male offspring as adults. METHODS: Pregnant C57BL/6 female mice were divided into two groups (n = 10): Control group (C), dams were injected with the vehicle only (saline 0.9% NaCl); Caffeine group (CF), dams received daily a subcutaneous injection of 20 mg/kg of caffeine/day (1 mg/mL saline). Pups had free access to standard chow since weaning to 3 months of age, when they were killed. RESULTS: CF group showed increased energy expenditure (+7%) with consequent reduction in body mass (BM) gain (-18%), increased blood pressure (+48%), and higher heart rate (+10%) than C group. The ratio between LV mass/BM was greater (+10%), with bigger cardiomyocytes (+40%), and reduced vascularization (-25%) in CF group than in C group. In the LV, the expression of angiotensin-converting enzyme (+30%), Angiotensin II (AngII) (+60%), AngII receptor (ATR)-1 (+77%) were higher, and the expression of ATR-2 was lower (-46%; P < 0.05) in CF group than in C group. In the kidney, the expressions of renin (+128%) and ATR-1 (+88%) were higher in CF group than in C group. CONCLUSIONS: Chronic administration of caffeine to pregnant dams led to persistent activation of local RAS in the kidney and heart of the offspring, which, in turn, leads to high BP and adverse cardiac remodeling. These findings highlight the urge to encourage pregnant women to avoid food or medicines containing caffeine.


Asunto(s)
Cafeína/administración & dosificación , Cafeína/efectos adversos , Corazón/efectos de los fármacos , Corazón/embriología , Exposición Materna/efectos adversos , Angiotensina II/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Hipertensión/etiología , Hipertensión/fisiopatología , Riñón/efectos de los fármacos , Riñón/embriología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Renina/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos
7.
Int J Mol Sci ; 14(11): 21873-86, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24196354

RESUMEN

The chronic intake of fructose has been linked to insulin resistance, obesity, dyslipidemia and nonalcoholic fatty liver disease (NAFLD), which in turn, may progress to nonalcoholic steatohepatitis (NASH). We aimed to evaluate the magnitude of the effects of the chronic consumption of high-fructose (HFr) and high fat (HF) alone or combined. Four groups of male mice were fed different diets for 16 weeks: standard chow (9% fat: SC), HF diet (42% fat), HFr diet (34% fructose) and HF/HFr diet (42% fat, 34% fructose). The food intake was not different among the groups, and the body mass was not greater in the HFr group than in the SC group. The homeostasis model assessment for insulin resistance (HOMA-IR), as well as plasmatic total cholesterol and triglycerides were greater in the groups HF, HFr, and HF/HFr group than in the SC group. We observed in the groups HF, HFr and HF/HFr, compared to the group SC, nonalcoholic fatty liver disease (NAFLD) with a predominance of lipogenesis mediated by SREBP-1c and PPAR-γ, and a reduction of the oxidation mediated by PPAR-α. We also observed an increase in gluconeogenesis mediated by the GLUT-2 and the PEPCK. Importantly, we identified areas of necroinflammation indicating a transition from NAFLD to nonalcoholic steatohepatitis in the HFr and HF/HFr groups. This study is relevant in demonstrating that fructose consumption, even in the absence of obesity, causes serious and deleterious changes in the liver with the presence of the dyslipidemia, insulin resistance (IR), and NAFLD with areas of necroinflammation. These conditions are associated with a poor prognosis.


Asunto(s)
Hígado Graso/metabolismo , Fructosa/metabolismo , Resistencia a la Insulina , Hígado/efectos de los fármacos , Animales , Dieta Alta en Grasa , Hígado Graso/patología , Fructosa/administración & dosificación , Humanos , Hígado/metabolismo , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico , Obesidad/metabolismo , Obesidad/patología , Sobrepeso/metabolismo , Sobrepeso/patología
8.
Biochem Pharmacol ; 217: 115852, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37832793

RESUMEN

We studied the effect of cotadutide, a dual agonist glucagon-like peptide 1 (GLP1)/Glucagon, on interscapular brown adipose tissue (iBAT) remodeling and thermogenesis of obese mice. Twelve-week-old male C57BL/6 mice were fed a control diet (C group, n = 20) or a high-fat diet (HF group, n = 20) for ten weeks. Then, animals were redivided, adding cotadutide treatment: C, CC, HF, and HFC for four additional weeks. The multilocular brown adipocyte structure showed fat conversion (whitening), hypertrophy, and structural disarray in the HF group, which was reverted in cotadutide-treated animals. Cotadutide enhances the body temperature, thermogenesis, and sympathetic innervation (peroxisome proliferator-activated receptor-α, ß3 adrenergic receptor, interleukin 6, and uncoupled protein 1), reduces pro-inflammatory markers (disintegrin and metallopeptidase domain, morphogenetic protein 8a, and neuregulin 4), and improves angiogenesis (vascular endothelial growth factor A, and perlecan). In addition, cotadutide enhances lipolysis (perilipin and cell death-inducing DNA fragmentation factor α), mitochondrial biogenesis (nuclear respiratory factor 1, transcription factor A mitochondrial, mitochondrial dynamin-like GTPase, and peroxisome proliferator-activated receptor gamma coactivator 1α), and mitochondrial fusion/fission (dynamin-related protein 1, mitochondrial fission protein 1, and parkin RBR E3 ubiquitin protein ligase). Cotadutide reduces endoplasmic reticulum stress (activating transcription factor 4, C/EBP homologous protein, and growth arrest and DNA-damage inducible), and extracellular matrix markers (lysyl oxidase, collagen type I α1, collagen type VI α3, matrix metallopeptidases 2 and 9, and hyaluronan synthases 1 and 2). In conclusion, the experimental evidence is compelling in demonstrating cotadutide's thermogenic effect on obese mice's iBAT, contributing to unraveling its action mechanisms and the possible translational benefits.


Asunto(s)
Tejido Adiposo Pardo , Factor A de Crecimiento Endotelial Vascular , Ratones , Animales , Masculino , Tejido Adiposo Pardo/metabolismo , Ratones Obesos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Adipocitos Marrones , Dieta Alta en Grasa/efectos adversos , Termogénesis , Dinaminas/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo
9.
Clin Sci (Lond) ; 119(6): 239-50, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20415664

RESUMEN

The aim of the present study was to evaluate the effects of monotherapies and combinations of drugs on insulin sensitivity, adipose tissue morphology, and pancreatic and hepatic remodelling in C57BL/6 mice fed on a very HF (high-fat) diet. Male C57BL/6 mice were fed on an HF (60% lipids) diet or SC (standard chow; 10% lipids) diet for 10 weeks, after which time the following drug treatments began: HF-T (HF diet treated with telmisartan; 5.2 mg x kg-1 of body weight x day-1), HF-S (HF diet treated with sitagliptin; 1.08 g x kg-1 of body weight.day-1), HF-M (HF diet treated with metformin; 310.0 mg x kg-1 of body weight x day-1), HF-TM (HF diet treated with telmisartan+metformin), HF-TS (HF diet treated with telmisartan+sitagliptin) and HF-SM (HF diet treated with sitagliptin+metformin). Treated groups also had free access to the HF diet, and treatments lasted for 6 weeks. Morphometry, stereological tools, immunostaining, ELISA, Western blot analysis and electron microscopy were used. The HF diet yielded an overweight phenotype, an increase in oral glucose intolerance, hyperinsulinaemia, hypertrophied islets and adipocytes, stage 2 steatosis (>33%), and reduced liver PPAR-alpha (peroxisome-proliferator-activated receptor-alpha) and GLUT-2 (glucose transporter-2) levels, concomitant with enhanced SREBP-1 (sterol-regulatory-element-binding protein-1) expression (P<0.0001). Conversely, all drug treatments resulted in significant weight loss, a reversal of insulin resistance, islet and adipocyte hypertrophy, and alleviated hepatic steatosis. Only the HF-T and HF-TS groups had body weights similar to the SC group at the end of the experiment, and the latter treatment reversed hepatic steatosis. Increased PPAR-alpha immunostaining in parallel with higher GLUT-2 and reduced SREBP-1 expression may explain the favourable hepatic outcomes. Restoration of adipocyte size was consistent with higher adiponectin levels and lower TNF-alpha (tumour necrosis factor-alpha) levels (P<0.0001) in the drug-treated groups. In conclusion, all of the drug treatments were effective in controlling the metabolic syndrome. The best results were achieved using telmisartan and sitagliptin as monotherapies or as a dual treatment, combining partial PPAR-gamma agonism and PPAR-alpha activation in the liver with extended incretin action.


Asunto(s)
Bencimidazoles/uso terapéutico , Benzoatos/uso terapéutico , Grasas de la Dieta/administración & dosificación , Síndrome Metabólico/tratamiento farmacológico , Metformina/uso terapéutico , Obesidad/tratamiento farmacológico , Pirazinas/uso terapéutico , Triazoles/uso terapéutico , Adipocitos/patología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Ingestión de Energía/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina , Hígado/fisiopatología , Hígado/ultraestructura , Masculino , Síndrome Metabólico/patología , Síndrome Metabólico/fisiopatología , Ratones , Ratones Endogámicos C57BL , Obesidad/patología , Obesidad/fisiopatología , Páncreas/fisiopatología , Fosfato de Sitagliptina , Telmisartán
10.
Am J Obstet Gynecol ; 203(5): 495.e1-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20822767

RESUMEN

OBJECTIVE: This work aimed to verify the hypothesis that maternal intake of high-fat diet in critical periods of pregnancy and/or suckling period predisposes nonalcoholic fatty liver disease in adult C57BL/6 mice offspring. STUDY DESIGN: Male pups were divided into 5 groups: (1) SC, from standard chow-fed dams; (2) G, from high-fat chow (HF)-fed dams during the gestation (G) period; (3) L, from HF-fed dams during the lactation (L) period; (4) GL, from HF-fed dams during the gestation and lactation (GL) periods; and (5) GL/HF, from HF-fed dams during GL, maintaining an HF diet from postweaning to adulthood. We analyzed body mass, plasma blood, and liver structure. RESULTS: The G offspring showed insulin resistance and lower glucose transporter-2 expression. Hepatic steatosis was present in the G, L, GL, and mainly in GL/HF offspring. Sterol regulatory element-binding protein-1c expression was higher in G, GL, and GL/HF offspring. CONCLUSION: Programming by HF chow predisposes hepatic adverse remodeling in the liver of adult offspring.


Asunto(s)
Grasas de la Dieta/metabolismo , Hígado Graso/etiología , Hígado/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Alanina Transaminasa/metabolismo , Fosfatasa Alcalina/metabolismo , Análisis de Varianza , Animales , Glucemia , Western Blotting , Peso Corporal , Susceptibilidad a Enfermedades/metabolismo , Ensayo de Inmunoadsorción Enzimática , Hígado Graso/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Resistencia a la Insulina , Masculino , Ratones , Embarazo , Radioinmunoensayo , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Nutr Biochem ; 70: 164-173, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31207355

RESUMEN

Fructose may induce an endocrine dysfunction in adipose tissue in rodents. Browning is identified by deposits of beige adipocytes in subcutaneous white adipose tissue (sWAT). We study the effects of the high-intensity interval training (HIIT) on the formation of beige adipocytes in the sWAT of mice fed a high-fructose diet. Sixty male mice (3 months old; C57BL/6) were fed two diets for 18 weeks (n=30 each): control diet (C) or high-fructose diet (F). At the 10th week, for an additional 8-week period, the groups were (n=15 each) nontrained (NT) or trained (HIIT): C-NT, C-HIIT, F-NT and F-HIIT. We evaluated body mass, energy expenditure and molecular analyses for browning and thermogenic markers in sWAT. The HIIT groups showed significantly lower body mass and increased energy expenditure. The consumption of fructose was linked with an increased sWAT mass. However, HIIT caused a reduction of sWAT mass compared to the NT groups. Energy intake was parallel in the groups, regardless of the diet type and HIIT. Fructose was related to higher glucose and insulin levels and hypertrophied sWAT adipocytes, but HIIT decreased both glucose and insulin levels and led to the appearance of brown fat-like adipocytes dispersed in sWAT with higher expression of browning markers. Also, fructose reduced the sWAT markers of mitochondrial biogenesis and beta-oxidation, which were enhanced by HIIT. In conclusion, HIIT might stimulate the sWAT browning in mice fed a high-fructose diet associated with beneficial changes in mitochondrial biogenesis and beta-oxidation markers, contributing to a whole-body metabolic improvement.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Dieta , Condicionamiento Físico Animal , Grasa Subcutánea/metabolismo , Adipocitos/citología , Animales , Glucemia/metabolismo , Metabolismo de los Hidratos de Carbono , Metabolismo Energético , Fructosa/administración & dosificación , Insulina/metabolismo , Resistencia a la Insulina , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Oxígeno/metabolismo , Termogénesis
12.
J Steroid Biochem Mol Biol ; 192: 105399, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31175967

RESUMEN

The fructose added to soft drinks and processed food, as well as frequent detection of vitamin D deficiency in the body, are two insults increasingly considered to cause lesions in target organs. We studied the liver after a chronic high-fructose diet deficient and supplemented with vitamin D. Sixty C57BL/6 mature male mice were allocated into six groups (n = 10) for ten weeks: control (C), control deficient in vitamin D (CDD), control supplemented with vitamin D (CDS), fructose (F), fructose deficient in vitamin D (FDD), and fructose supplemented with vitamin D (FDS). The gene expressions of vitamin D receptor and CYP27B1 and 25 hydroxyvitamin D plasma level ensured that the diets caused vitamin D deficiency or supplementation. Body mass did not change, but blood pressure (BP) increased in CDD, F, and FDD, whereas BP was controlled in FDS. Insulinemia, insulin tolerance and resistance were seen in both vitamin D deficiency and fructose groups but improved with vitamin D supplementation. The steatosis and fibrosis were observed in the CDD, F and FDD groups. Also, F and FDD showed activation of stellate cells (HSC). Lipogenesis and inflammation gene expressions were enhanced in the CDD, F and FDD groups, but diminished with vitamin D supplementation. In conclusion, we demonstrated the adverse effects of vitamin D deficiency on metabolism, liver steatosis and, combined with fructose intake, liver interstitial fibrosis with hepatic stellate cell activation, and alteration of the lipogenesis, beta-oxidation, and liver inflammation. All these data improved when vitamin D was supplemented in the animals.


Asunto(s)
Suplementos Dietéticos , Hígado Graso/prevención & control , Fructosa/farmacología , Resistencia a la Insulina , Lipogénesis , Deficiencia de Vitamina D/complicaciones , Vitamina D/administración & dosificación , Animales , Hígado Graso/etiología , Hígado Graso/patología , Fructosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Vitaminas/administración & dosificación
13.
Clin Sci (Lond) ; 114(5): 381-92, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17927565

RESUMEN

Substantial evidence suggests that poor intrauterine milieu elicited by maternal nutritional disturbance may programme susceptibility in the fetus to later development of chronic diseases, such as obesity, hypertension, cardiovascular disease and diabetes. One of the most interesting features of fetal programming is the evidence from several studies that the consequences may not be limited to the first-generation offspring and that it can be passed transgenerationally. In the present study, female rats (F0) were fed either a normal-protein diet [control diet (C); 19 g of protein/100 g of diet] or a low-protein diet [restricted diet (R); 5 g of protein/100 g of diet]. The offspring were termed according to the period and the types of diet the dams were fed, i.e. CC, RC, CR and RR (first letter indicates the diet during gestation and the second the diet during lactation). At 3 months of age, F1 females were bred to proven males, outside the experiment, to produce F2 offspring. At weaning, F2 offspring were divided by gender. RC1 offspring (with the number indicating the filial generation) were born with low birthweight, but afterwards they had catch-up growth, reaching the weight of the CC1 offspring. The increased glycaemia in RC1 offspring was associated with insulin resistance. CR1 and RR1 offspring had impaired growth with no changes in glucose metabolism. RC2 offspring had high BM (body mass) at birth, which was sustained over the whole experiment in male offspring. The F2 generation had more alteration in glucose metabolism than the F1 generation. CR2 and RC2 offspring had hyperglycaemia accompanied by hyperinsulinaemia and insulin resistance in both genders. CR2 offspring had an increase in body adiposity with hyperleptinaemia. In conclusion, low protein during gestation improves BM, fat mass and growth rate in F1 rats, but has adverse effects on glucose and leptin metabolism, resulting in insulin resistance in adult F1 and F2 offspring. Low protein during lactation has adverse effects on glucose, insulin and leptin metabolism, resulting in insulin resistance in adult F2 offspring. These findings suggest that low protein during gestation and/or lactation can be passed transgenerationally to the second generation.


Asunto(s)
Glucemia/metabolismo , Dieta con Restricción de Proteínas , Trastornos Nutricionales en el Feto/fisiopatología , Lactancia/fisiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Biometría , Femenino , Desarrollo Fetal/fisiología , Crecimiento/fisiología , Hiperglucemia/embriología , Hiperglucemia/genética , Hiperinsulinismo/embriología , Hiperinsulinismo/genética , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Leptina/sangre , Masculino , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal/fisiología , Ratas
14.
Biomed Pharmacother ; 107: 878-888, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30257400

RESUMEN

AIMS: A prevalence of vitamin D deficiency has been reported in association with the postmenopause. Thus, we aimed to experimentally study the effect of the vitamin D deficiency and ovariectomy, alone or combined, in the liver damage. MAIN METHODS: Three-months-old female mice C57BL/6 with bilateral ovariectomy (Ovx group, n = 30) or a sham procedure (n = 30) were separated feeding control diet (C, n = 15) or a diet restricted in vitamin D (D-, n = 15) during additional 12 weeks. KEY FINDINGS: Body mass (BM) and blood pressure (BP) were higher in Ovx than in C animals, but highest in Ovx (D-) that also showed glucose intolerance/ insulin resistance. Plasmatic lipids, alanine aspartase transferase, and hepatic steatosis were increased because of the combination of Ovx and D-. However, D- had little implication in the changes of the BM and BP, but affected hepatic steatosis. Gene and protein expressions demonstrated an impaired glucose uptake in the liver because of Ovx and D-, and an increase in lipogenesis and decrease in beta-oxidation in the liver associated more to the Ovx, but also evident in D-. Also, interleukin 6 and tumor necrosis factor alpha showed an enhancement due to dietary restriction of vitamin D. SIGNIFICANCE: The findings demonstrated that ovariectomy and dietary restriction of vitamin D are inducers of harmful effects on the liver of mice, enhancing lipogenesis and inflammation and compromising beta-oxidation. The treatment of vitamin D deficiency is simple and not costly and can reduce the impact of menopause on metabolism and especially the liver.


Asunto(s)
Hígado Graso/etiología , Inflamación/etiología , Hígado/patología , Deficiencia de Vitamina D/complicaciones , Animales , Hígado Graso/patología , Femenino , Inflamación/patología , Resistencia a la Insulina/fisiología , Lípidos/sangre , Lipogénesis/fisiología , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Oxidación-Reducción
15.
J Appl Physiol (1985) ; 123(4): 699-709, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28495843

RESUMEN

Fructose-rich caloric sweeteners induce adverse changes in the metabolism of humans. The study evaluated the effects of high-intensity interval training (HIIT) on a fructose feeding model, focusing on the liver, white adipose tissue (WAT), skeletal muscle, and their interplay. Male C57BL/6 mice were fed for 18 wk one of the following diets: control (C; 5% of total energy from fructose) or fructose (F; 55% of total energy from fructose). In the 10th week, for an additional 8-wk period, the groups were divided into nontrained (NT) or HIIT groups, totaling four groups: C-NT, C-HIIT, F-NT, and F-HIIT. At the end of the experiment, fructose consumption in the F-NT group led to a high systolic blood pressure, high plasma triglycerides, insulin resistance with glucose intolerance, and lower insulin sensitivity. We also observed liver steatosis, adipocyte hypertrophy, and diminished gene expressions of peroxisome proliferator-activated receptor-γ coactivator 1-α and fibronectin type III domain containing 5 (FNDC5; irisin) in this F-NT group. These results were accompanied by decreased gene expressions of nuclear respiratory factor 1 and mitochondrial transcription factor A (markers of mitochondrial biogenesis), and peroxisome proliferator-activated receptor-α and carnitine palmitoyltransferase 1 (markers of ß-oxidation). HIIT improved all of these data in the C-HIIT and F-HIIT groups. In conclusion, in mice fed a fructose diet, HIIT improved body mass, blood pressure, glucose metabolism, and plasma triglycerides. Liver, WAT, and skeletal muscle were positively modulated by HIIT, indicating HIIT as a coadjutant treatment for diseases affecting these tissues.NEW & NOTEWORTHY We investigated the effects of high-intensity interval training (HIIT) in mice fed a fructose-rich diet and the resulting severe negative effect on the liver, white adipose tissue (WAT), and skeletal muscle, which reduced the expression of fibronectin type III domain containing 5 (FNDC5, irisin) and PGC1α and, consequently, affected markers of mitochondrial biogenesis and ß-oxidation. Because HIIT may block these adverse effects in all of these three tissues, it might be suggested that it functions as a coadjutant treatment in combatting the alterations caused by high-fructose intake.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Fructosa/administración & dosificación , Entrenamiento de Intervalos de Alta Intensidad , Hígado/metabolismo , Condicionamiento Físico Animal , Animales , Presión Sanguínea , Dieta , Fibronectinas/metabolismo , Intolerancia a la Glucosa , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Triglicéridos/sangre
16.
J Pediatr (Rio J) ; 93(6): 551-559, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28822233

RESUMEN

OBJECTIVE: To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children. SOURCES: A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication. SUMMARY OF FINDINGS: Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA. CONCLUSIONS: Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.


Asunto(s)
Epigénesis Genética/genética , Padre , Obesidad/genética , Obesidad/metabolismo , Animales , Femenino , Humanos , Masculino , Modelos Animales
17.
Menopause ; 13(1): 87-95, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16607103

RESUMEN

OBJECTIVE: To study the combined effects of ovariectomy and regular exercise training on hypertension and on cardiac and aortic remodeling in spontaneously hypertensive rats (SHR). DESIGN: Three-month-old female spontaneously hypertensive rats (SHR) were ovariectomized (ovx) or were left intact (int) and divided in four groups (n = 7): sedentary (sed-ovx), exercise-trained (ex-ovx), sedentary intact (sed-int), and exercise-trained intact (ex-int). The exercise protocol was performed on a motor treadmill for 13 weeks. Blood pressure (BP), left ventricular myocardium and aortic wall were studied by light microscopy and stereology. RESULTS: Exercise-trained SHR showed a BP reduction of more than 15% compared with the matched sedentary SHR (sed-int: 210 +/- 5 mm Hg, sed-ovx: 225 +/- 4 mm Hg, ex-int: 178 +/- 2 mm Hg, ex-ovx: 180 +/- 3 mm Hg, P < 0.001). Ovariectomy caused adverse cardiac and aortic wall remodeling, including cardiomyocyte hypertrophy, myocardial interstitial reparative fibrosis and vascularization impairment with loss of cardiomyocytes, and aortic tunica media hypertrophy. Exercise training showed beneficial effects, mainly reduced BP, decreased cardiac hypertrophy due to hypertension, and increased myocardial vascularization. Ovariectomy accelerated cardiomyocyte loss in SHR while exercise training offset this process. Exercise training was the main factor influencing the improvement of intramyocardial arteries length density and significantly reduced the aortic wall thickness and increased the density of smooth muscle cell nuclei per tunica media unit area. CONCLUSIONS: In ovariectomized SHR, exercise training exerts beneficial effects diminishing adverse cardiac and aortic wall remodeling, mainly by reducing interstitial myocardial fibrosis, improving myocardial vascularization, and sustaining the number of cardiomyocytes.


Asunto(s)
Cardiomegalia/prevención & control , Hipertensión/prevención & control , Ovariectomía , Condicionamiento Físico Animal , Remodelación Ventricular/fisiología , Animales , Aorta/patología , Biometría , Vasos Coronarios , Femenino , Hipertrofia , Miocardio/patología , Ratas , Ratas Endogámicas SHR , Túnica Media/patología
18.
Artículo en Inglés | MEDLINE | ID: mdl-16919431

RESUMEN

We studied the effects of edible oils intake on the renal cortical structure of streptozotocin-induced diabetic (Db) and non-diabetic spontaneously hypertensive rats (SHR). Male SHR divided into 5 groups were studied during 6 weeks: one non-diabetic SHR group and four diabetic SHR groups (three groups received by gavage olive, palm or fish oil). Kidneys were analyzed by light microscopy and stereology. Oils intake did not change the plasma glucose levels. The blood pressure (BP) was lower in SHR-Db than in SHR, but SHR-Db-fish oil showed the lowest BP. Creatinine clearance was different between diabetic SHR and non-diabetic SHR, but not between treated SHR-Db and untreated SHR-Db. The renal cortex showed scars surrounding obsolete glomeruli with inflammatory infiltrate mainly in untreated SHR-Db. The olive oil, palm oil and mainly fish oil intake retard the usual loss of glomeruli and attenuate the renal cortex adverse remodeling of Db and non-Db SHR.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Aceites de Pescado/administración & dosificación , Hipertensión/fisiopatología , Corteza Renal/efectos de los fármacos , Aceites de Plantas/administración & dosificación , Regeneración/efectos de los fármacos , Animales , Glucemia/análisis , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Creatinina/orina , Diabetes Mellitus Experimental/complicaciones , Hipertensión/complicaciones , Corteza Renal/fisiología , Corteza Renal/ultraestructura , Masculino , Aceite de Oliva , Aceite de Palma , Ratas , Ratas Endogámicas SHR , Estreptozocina
19.
J Sports Med Phys Fitness ; 56(5): 655-63, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27285355

RESUMEN

BACKGROUND: Controlling obesity and other comorbidities in the population is a challenge in modern society. High-intensity interval training (HIIT) combines short periods of high-intensity exercise with long recovery periods or a low-intensity exercise. The aim was to assess the impact of HIIT in the context of diet-induced obesity in the animal model. METHODS: C57BL/6 mice were fed one of the two diets: standard chow (lean group [LE]) or a high-fat diet (obese group [OB]). After twelve weeks, the animals were divided into non-trained groups (LE-NT and OB-NT) and trained groups (LE-T and OB-T), and began an exercise protocol. For biochemical analysis of inflammatory and lipid profile, we used a colorimetric enzymatic method and an automatic spectrophotometer. One-way ANOVA was used for statistical analysis of the experimental groups with Holm-Sidak post-hoc Test. Two-way ANOVA analyzed the interactions between diet and HIIT protocol. RESULTS: HIIT leads to significant reductions in body mass, blood glucose, glucose tolerance and hepatic lipid profile in T-groups compared to NT-groups. HIIT was able to reduce plasma levels of inflammatory cytokines. Additionally, HIIT improves the insulin immunodensity in the islets, reduces the adiposity and the hepatic steatosis in the T-groups. HIIT improves beta-oxidation and peroxisome proliferator-activated receptor (PPAR)-alpha and reduces lipogenesis and PPAR-gamma levels in the liver. In skeletal muscle, HIIT improves PPAR-alpha and glucose transporter-4 and reduces PPAR-gamma levels. CONCLUSIONS: HIIT leads to attenuate the adverse effects caused by a chronic ingestion of a high-fat diet.


Asunto(s)
Comorbilidad , Entrenamiento de Intervalos de Alta Intensidad , Ratones Obesos/metabolismo , Obesidad/etiología , Hipernutrición , Natación , Adiposidad , Animales , Glucemia/metabolismo , Insulina/sangre , Resistencia a la Insulina , Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo
20.
Obesity (Silver Spring) ; 24(3): 626-33, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26916241

RESUMEN

OBJECTIVE: The neuroprotective effects of liraglutide (200 µg/kg, twice daily, subcutaneous administration) in the hypothalamic arcuate nucleus (ARC) of diet-induced obese mice were investigated. METHODS: C57BL/6 mice were separated into groups: standard chow treated with vehicle or liraglutide and the respective liraglutide pair-fed group; high-fat diet treated with vehicle or liraglutide and the respective pair-fed group. Body mass (BM) evolution, carbohydrate metabolism, leptin resistance, proteins involved in energetic balance, apoptosis, and microglia in the ARC were studied. RESULTS: Obese animals showed glucose intolerance, resistance to insulin and to anorexigenic effect of leptin, and microgliosis accompanied by elevated Bax/Bcl2 ratio in the ARC. Liraglutide improved the carbohydrate metabolism, BM loss, and the activation of pro-opiomelanocortin (POMC) and cocaine and amphetamine-regulated transcript (CART) in the ARC. The liraglutide enhanced leptin sensitivity and diminished the microgliosis with decrease in Bax/Bcl2 ratio. CONCLUSIONS: Liraglutide activates central anorexigenic pathways, thereby diminishing the energy intake of obese mice and improving the metabolic parameters related to obesity. Liraglutide is a relevant neuroprotective agent, which can decrease the microgliosis and stimulate the anti-apoptotic pathway, a significant effect in the treatment of obesity and its comorbidities. Some benefits of liraglutide are independent of the BM loss, which usually accompanies the drug administration.


Asunto(s)
Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Hipoglucemiantes/farmacología , Leptina/metabolismo , Liraglutida/farmacología , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ingestión de Energía , Hipotálamo/metabolismo , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Proopiomelanocortina/metabolismo , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda