Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Cell Stress Chaperones ; 29(1): 175-200, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38331164

ABSTRACT

The heat shock response (HSR) is an ancient and evolutionarily conserved mechanism designed to restore cellular homeostasis following proteotoxic challenges. However, it has become increasingly evident that disruptions in energy metabolism also trigger the HSR. This interplay between proteostasis and energy regulation is rooted in the fundamental need for ATP to fuel protein synthesis and repair, making the HSR an essential component of cellular energy management. Recent findings suggest that the origins of proteostasis-defending systems can be traced back over 3.6 billion years, aligning with the emergence of sugar kinases that optimized glycolysis around 3.594 billion years ago. This evolutionary connection is underscored by the spatial similarities between the nucleotide-binding domain of HSP70, the key player in protein chaperone machinery, and hexokinases. The HSR serves as a hub that integrates energy metabolism and resolution of inflammation, further highlighting its role in maintaining cellular homeostasis. Notably, 5'-adenosine monophosphate-activated protein kinase emerges as a central regulator, promoting the HSR during predominantly proteotoxic stress while suppressing it in response to predominantly metabolic stress. The complex relationship between 5'-adenosine monophosphate-activated protein kinase and the HSR is finely tuned, with paradoxical effects observed under different stress conditions. This delicate equilibrium, known as caloristasis, ensures that cellular homeostasis is maintained despite shifting environmental and intracellular conditions. Understanding the caloristatic controlling switch at the heart of this interplay is crucial. It offers insights into a wide range of conditions, including glycemic control, obesity, type 2 diabetes, cardiovascular and neurodegenerative diseases, reproductive abnormalities, and the optimization of exercise routines. These findings highlight the profound interconnectedness of proteostasis and energy metabolism in cellular function and adaptation.


Subject(s)
Diabetes Mellitus, Type 2 , Proteostasis , Humans , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response , Adenosine Monophosphate/metabolism , Protein Kinases/metabolism
2.
Gac. méd. boliv ; 46(1)2023.
Article in Spanish | LILACS-Express | LILACS | ID: biblio-1448292

ABSTRACT

La isoforma AMPKß2 (proteína quinasa activada por AMP) favorece la homeostasis glucémica a través de un mecanismo independiente de insulina. Muchos "importagogos" de glucosa como SC4 actúan como activadores de AMPK, pero su consumo prolongado se asocia a efectos indeseables. Objetivo: en este trabajo se utilizó el acoplamiento molecular para analizar la posible interacción entre sapogeninas y AMPK. Métodos: se ha procedido a la preparación de los blancos proteicos, preparación de los ligandos y el acoplamiento molecular. Resultados: los resultados mostraron que ocho sapogeninas presentes en Chenopodium quinoa interactúan en el mismo sitio de unión que SC4 correspondiente al sitio ADaM de AMPK. Estas interacciones puntuaron valores de ΔG que oscilan entre -6,2 y -7,7 kcal/mol, siendo el ácido serjánico la sapogenina con el ΔG más bajo. La adición de grupos hidrofílicos como -OH y -COOH en la estructura química del ácido serjánico mejoró su afinidad de unión a la isoforma AMPKß2 abriendo la posibilidad de generar fármacos semi-sintéticos a partir de compuestos naturales con mayor actividad biológica y mejor especificidad.


The AMPKP2 (AMP-activated protein kinase) isoform promotes glycemic homeostasis through an insulin-independent mechanism. Many glucose "importers" such as SC4 act as AMPK activators, but their prolonged consumption is associated with undesirable effects. Objetive: in this work, molecular docking was used to analyze the possible interaction between sapogenins and AMPK. Methods: the preparation of the protein blanks, preparation of the ligands and molecular coupling have been carried out. Results: the results showed that eight sapogenins present in Chenopodium quinoa interact at the same binding site as SC4 corresponding to the ADaM site of AMPK. These interactions scored ΔG values ranging from -6.2 to -7.7 kcal/mol, with Serjanic acid being the sapogenin with the lowest ΔG. The addition of hydrophilic groups such as -OH and -COOH in the chemical structure of Serjanic acid improved its binding affinity to the AMPKß2 isoform opening the possibility of generating semi-synthetic drugs from natural compounds with higher biological activity and better specificity.

3.
J Mol Endocrinol ; 66(3): 207-221, 2021 03.
Article in English | MEDLINE | ID: mdl-33640872

ABSTRACT

Recent studies have provided evidence that triiodothyronine (T3) might play an effective role in the recovery of ischemic myocardium, through the preservation of mitochondrial function and the improvement of energy substrate metabolism. To this respect, it has been suggested that T3 could activate AMP-activated protein kinase (AMPK), the cellular 'fuel-gauge' enzyme, although its role has yet to be elucidated. The aim of the present study was to investigate the effects produced by acute treatment with T3 (60 nM) and the pharmacological inhibition of AMPK by compound C on isolated rat left atria subjected to 75 min simulated ischemia-75 min reperfusion. Results showed that T3 increased AMPK activation during simulated ischemia-reperfusion, while compound C prevented it. At the end of simulated reperfusion, acute T3 treatment increased contractile function recovery and cellular viability conservation. Mitochondrial ultrastructure was better preserved in the presence of T3 as well as mitochondrial ATP production rate and tissue ATP content. Calcium retention capacity, a parameter widely used as an indicator of the resistance of mitochondrial permeability transition pore (MPTP) to opening, and GSK-3ß phosphorylation, a master switch enzyme that limits MPTP opening, were increased by T3 administration. All these beneficial effects exerted by T3 acute treatment were prevented when compound C was co-administrated. The present study provided original evidence that T3 enhances intrinsic activation of AMPK during myocardial ischemia-reperfusion, being this enzyme involved, at least in part, in the protective effects exerted by T3, contributing to mitochondrial structure and function preservation, post-ischemic contractile recovery and conservation of cellular viability.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Myocardium/pathology , Triiodothyronine/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cell Survival/drug effects , Diastole/drug effects , Female , Glycogen Synthase Kinase 3 beta/metabolism , Heart Atria/ultrastructure , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/physiopathology , Phosphorylation/drug effects , Rats, Sprague-Dawley , Systole/drug effects , Triiodothyronine/pharmacology
4.
Biol Res ; 54(1): 3, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33546773

ABSTRACT

BACKGROUND: Testosterone regulates nutrient and energy balance to maintain protein synthesis and metabolism in cardiomyocytes, but supraphysiological concentrations induce cardiac hypertrophy. Previously, we determined that testosterone increased glucose uptake-via AMP-activated protein kinase (AMPK)-after acute treatment in cardiomyocytes. However, whether elevated glucose uptake is involved in long-term changes of glucose metabolism or is required during cardiomyocyte growth remained unknown. In this study, we hypothesized that glucose uptake and glycolysis increase in testosterone-treated cardiomyocytes through AMPK and androgen receptor (AR). METHODS: Cultured cardiomyocytes were stimulated with 100 nM testosterone for 24 h, and hypertrophy was verified by increased cell size and mRNA levels of ß-myosin heavy chain (ß-mhc). Glucose uptake was assessed by 2-NBDG. Glycolysis and glycolytic capacity were determined by measuring extracellular acidification rate (ECAR). RESULTS: Testosterone induced cardiomyocyte hypertrophy that was accompanied by increased glucose uptake, glycolysis enhancement and upregulated mRNA expression of hexokinase 2. In addition, testosterone increased AMPK phosphorylation (Thr172), while inhibition of both AMPK and AR blocked glycolysis and cardiomyocyte hypertrophy induced by testosterone. Moreover, testosterone supplementation in adult male rats by 5 weeks induced cardiac hypertrophy and upregulated ß-mhc, Hk2 and Pfk2 mRNA levels. CONCLUSION: These results indicate that testosterone stimulates glucose metabolism by activation of AMPK and AR signaling which are critical to induce cardiomyocyte hypertrophy.


Subject(s)
AMP-Activated Protein Kinases , Glucose/metabolism , Myocytes, Cardiac , Receptors, Androgen/metabolism , Testosterone/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Hypertrophy , Male , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Signal Transduction
5.
Biol. Res ; 54: 3-3, 2021. graf, ilus
Article in English | LILACS | ID: biblio-1505792

ABSTRACT

BACKGROUND: Testosterone regulates nutrient and energy balance to maintain protein synthesis and metabolism in cardiomyocytes, but supraphysiological concentrations induce cardiac hypertrophy. Previously, we determined that testosterone increased glucose uptake­via AMP-activated protein kinase (AMPK)­after acute treatment in cardiomyocytes. However, whether elevated glucose uptake is involved in long-term changes of glucose metabolism or is required during cardiomyocyte growth remained unknown. In this study, we hypothesized that glucose uptake and glycolysis increase in testosterone-treated cardiomyocytes through AMPK and androgen receptor (AR). METHODS: Cultured cardiomyocytes were stimulated with 100 nM testosterone for 24 h, and hypertrophy was verified by increased cell size and mRNA levels of ß-myosin heavy chain (ß-mhc). Glucose uptake was assessed by 2-NBDG. Glycolysis and glycolytic capacity were determined by measuring extracellular acidification rate (ECAR). RESULTS: Testosterone induced cardiomyocyte hypertrophy that was accompanied by increased glucose uptake, glycolysis enhancement and upregulated mRNA expression of hexokinase 2. In addition, testosterone increased AMPK phosphorylation (Thr172), while inhibition of both AMPK and AR blocked glycolysis and cardiomyocyte hypertrophy induced by testosterone. Moreover, testosterone supplementation in adult male rats by 5 weeks induced cardiac hypertrophy and upregulated ß-mhc, Hk2 and Pfk2 mRNA levels. CONCLUSION: These results indicate that testosterone stimulates glucose metabolism by activation of AMPK and AR signaling which are critical to induce cardiomyocyte hypertrophy.


Subject(s)
Animals , Male , Rats , Testosterone/pharmacology , Receptors, Androgen/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Signal Transduction , Cells, Cultured , Hypertrophy , Myocardium/pathology
6.
Am J Physiol Renal Physiol ; 318(5): F1229-F1236, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32249610

ABSTRACT

Metformin, an AMP-activated protein kinase (AMPK) activator, has been shown in previous studies to reduce kidney fibrosis in different models of experimental chronic kidney disease (CKD). However, in all of these studies, the administration of metformin was initiated before the establishment of renal disease, which is a condition that does not typically occur in clinical settings. The aim of the present study was to investigate whether the administration of metformin could arrest the progression of established renal disease in a well-recognized model of CKD, the subtotal kidney nephrectomy (Nx) model. Adult male Munich-Wistar rats underwent either Nx or sham operations. After the surgery (30 days), Nx rats that had systolic blood pressures of >170 mmHg and albuminuria levels of >40 mg/24 h were randomized to a no-treatment condition or to a treatment condition with metformin (300 mg·kg-1·day-1) for a period of either 60 or 120 days. After 60 days of treatment, we did not observe any differences in kidney disease parameters between Nx metformin-treated and untreated rats. However, after 120 days, Nx rats that had been treated with metformin displayed significant reductions in albuminuria levels and in markers of renal fibrosis. These effects were independent of any other effects on blood pressure or glycemia. In addition, treatment with metformin was also able to activate kidney AMPK and therefore improve mitochondrial biogenesis. It was concluded that metformin can arrest the progression of established kidney disease in the Nx model, likely via the activation of AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Enzyme Activators/pharmacology , Kidney/drug effects , Metformin/pharmacology , Nephrectomy , Renal Insufficiency, Chronic/prevention & control , Albuminuria/etiology , Albuminuria/metabolism , Albuminuria/prevention & control , Animals , Disease Models, Animal , Disease Progression , Enzyme Activation , Fibrosis , Hypertension/etiology , Hypertension/metabolism , Hypertension/prevention & control , Kidney/enzymology , Kidney/pathology , Kidney/surgery , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Organelle Biogenesis , Rats, Wistar , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Time Factors
7.
Article in English | MEDLINE | ID: mdl-32152146

ABSTRACT

OBJECTIVE: Obesity is associated with metabolic abnormalities, including insulin resistance and dyslipidemias. Previous studies demonstrated that genistein intake modifies the gut microbiota in mice by selectively increasing Akkermansia muciniphila, leading to reduction of metabolic endotoxemia and insulin sensitivity. However, it is not known whether the consumption of genistein in humans with obesity could modify the gut microbiota reducing the metabolic endotoxemia and insulin sensitivity. RESEARCH DESIGN AND METHODS: 45 participants with a Homeostatic Model Assessment (HOMA) index greater than 2.5 and body mass indices of ≥30 and≤40 kg/m2 were studied. Patients were randomly distributed to consume (1) placebo treatment or (2) genistein capsules (50 mg/day) for 2 months. Blood samples were taken to evaluate glucose concentration, lipid profile and serum insulin. Insulin resistance was determined by means of the HOMA for insulin resistance (HOMA-IR) index and by an oral glucose tolerance test. After 2 months, the same variables were assessed including a serum metabolomic analysis, gut microbiota, and a skeletal muscle biopsy was obtained to study the gene expression of fatty acid oxidation. RESULTS: In the present study, we show that the consumption of genistein for 2 months reduced insulin resistance in subjects with obesity, accompanied by a modification of the gut microbiota taxonomy, particularly by an increase in the Verrucomicrobia phylum. In addition, subjects showed a reduction in metabolic endotoxemia and an increase in 5'-adenosine monophosphate-activated protein kinase phosphorylation and expression of genes involved in fatty acid oxidation in skeletal muscle. As a result, there was an increase in circulating metabolites of ß-oxidation and ω-oxidation, acyl-carnitines and ketone bodies. CONCLUSIONS: Change in the gut microbiota was accompanied by an improvement in insulin resistance and an increase in skeletal muscle fatty acid oxidation. Therefore, genistein could be used as a part of dietary strategies to control the abnormalities associated with obesity, particularly insulin resistance; however, long-term studies are needed.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anti-Obesity Agents/administration & dosage , Gastrointestinal Microbiome/drug effects , Genistein/administration & dosage , Insulin Resistance , Muscle, Skeletal/drug effects , Obesity/metabolism , Obesity/microbiology , Double-Blind Method , Fatty Acids/metabolism , Humans , Muscle, Skeletal/metabolism
8.
IUBMB Life ; 71(9): 1211-1220, 2019 09.
Article in English | MEDLINE | ID: mdl-31091354

ABSTRACT

Liver preconditioning (PC) refers to the development of an enhanced tolerance to injuring stimuli. For example, the protection from ischemia-reperfusion (IR) in the liver that is obtained by previous maneuvers triggering beneficial molecular and functional changes. Recently, we have assessed the PC effects of thyroid hormone (T3; single dose of 0.1 mg/kg) and n-3 long-chain polyunsaturated fatty acids (n-3 LCPUFAs; daily doses of 450 mg/kg for 7 days) that abrogate IR injury to the liver. This feature is also achieved by a combined T3 and the n-3 LCPUFA docosahexaenoic acid (DHA) using a reduced period of supplementation of the FA (daily doses of 300 mg/kg for 3 days) and half of the T3 dosage (0.05 mg/kg). T3 -dependent protective mechanisms include (i) the reactive oxygen species (ROS)-dependent activation of transcription factors nuclear factor-κB (NF-κB), AP-1, signal transducer and activator of transcription 3, and nuclear factor erythroid-2-related factor 2 (Nrf2) upregulating the expression of protective proteins. (ii) ROS-induced endoplasmic reticulum stress affording proper protein folding. (iii) The autophagy response to produce FAs for oxidation and ATP supply and amino acids for protein synthesis. (iv) Downregulation of inflammasome nucleotide-bonding oligomerization domain leucine-rich repeat containing family pyrin containing 3 and interleukin-1ß expression to prevent inflammation. N-3 LCPUFAs induce antioxidant responses due to Nrf2 upregulation, with inflammation resolution being related to production of oxidation products and NF-κB downregulation. Energy supply to achieve liver PC is met by the combined DHA plus T3 protocol through upregulation of AMPK coupled to peroxisome proliferator-activated receptor-γ coactivator 1α signaling. In conclusion, DHA plus T3 coadministration favors hepatic bioenergetics and lipid homeostasis that is of crucial importance in acute and clinical conditions such as IR, which may be extended to long-term or chronic situations including steatosis in obesity and diabetes. © 2019 IUBMB Life, 71(9):1211-1220, 2019.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Reperfusion Injury/diet therapy , Stress, Physiological/drug effects , Thyroid Hormones/therapeutic use , Dietary Supplements , Energy Metabolism/drug effects , Fatty Liver/diet therapy , Fatty Liver/pathology , Fatty Liver/prevention & control , Humans , Inflammasomes/drug effects , Inflammasomes/genetics , Ischemic Preconditioning , Liver/drug effects , Liver/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control
9.
Am J Physiol Cell Physiol ; 314(6): C702-C711, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29443552

ABSTRACT

The renin-angiotensin system (RAS) plays a pivotal role in the pathogenesis of cardiovascular diseases. New members of this system have been characterized and shown to have biologically relevant actions. Alamandine and its receptor MrgD are recently identified components of RAS. In the cardiovascular system, alamandine actions included vasodilation, antihypertensive, and antifibrosis effects. Currently, the actions of alamandine on cardiomyocytes are unknown. Here our goal was twofold: 1) to unravel the signaling molecules activated by the alamandine/MrgD axis in cardiomyocytes; and 2) to evaluate the ability of this axis to prevent angiotensin II (ANG II)-induced hypertrophy. In cardiomyocytes from C57BL/6 mice, alamandine treatment induced an increase in nitric oxide (NO) production, which was blocked by d-Pro7-ANG-(1-7), a MrgD antagonist. This NO rise correlated with increased phosphorylation of AMPK. Alamandine-induced NO production was preserved in Mas-/- myocytes and lost in MrgD-/- cells. Binding of fluorescent-labeled alamandine was observed in wild-type cells, but it was dramatically reduced in MrgD-/- myocytes. We also assessed the consequences of prolonged alamandine exposure to cultured neonatal rat cardiomyocytes (NRCMs) treated with ANG II. Treatment of NRCMs with alamandine prevented ANG II-induced hypertrophy. Moreover, the antihypertrophic actions of alamandine were mediated via MrgD and NO, since they could be prevented by d-Pro7-ANG-(1-7) or inhibitors of NO synthase or AMPK. ß-Alanine, a MrgD agonist, recapitulated alamandine's cardioprotective effects in cardiomyocytes. Our data show that alamandine via MrgD induces AMPK/NO signaling to counterregulate ANG II-induced hypertrophy. These findings highlight the therapeutic potential of the alamandine/MrgD axis in the heart.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Angiotensin II/toxicity , Cardiomegaly/prevention & control , Myocytes, Cardiac/drug effects , Nerve Tissue Proteins/agonists , Nitric Oxide/metabolism , Oligopeptides/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cells, Cultured , Enzyme Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/metabolism , Oligopeptides/metabolism , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
10.
Pflugers Arch ; 470(2): 367-383, 2018 02.
Article in English | MEDLINE | ID: mdl-29032506

ABSTRACT

AMP-activated protein kinase (AMPK) is a serine-threonine kinase that functions primarily as a metabolic sensor to coordinate anabolic and catabolic processes in the cell, via phosphorylation of multiple proteins involved in metabolic pathways, aimed to re-establish energy homeostasis at a cell-autonomous level. Myocardial ischemia and reperfusion represents a metabolic stress situation for myocytes. Whether AMPK plays a critical role in the metabolic and functional responses involved in these conditions remains uncertain. In this study, in order to gain a deeper insight into the role of endogenous AMPK activation during myocardial ischemia and reperfusion, we explored the effects of the pharmacological inhibition of AMPK on contractile function rat, contractile reserve, tissue lactate production, tissue ATP content, and cellular viability. For this aim, isolated atria subjected to simulated 75 min ischemia-75 min reperfusion (Is-Rs) in the presence or absence of the pharmacological inhibitor of AMPK (compound C) were used. Since in most clinical situations of ischemia-reperfusion the heart is exposed to high levels of fatty acids, the influence of palmitate present in the incubation medium was also investigated. The present results suggest that AMPK activity significantly increases during Is, remaining activated during Rs. The results support that intrinsic activation of AMPK has functional protective effects in the reperfused atria when glucose is the only available energetic substrate whereas it is deleterious when palmitate is also available. Cellular viability was not affected by either of these conditions.


Subject(s)
Energy Metabolism , Heart Atria/metabolism , Myocardial Reperfusion Injury/metabolism , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Adenosine Triphosphate/metabolism , Animals , Atrial Function , Fatty Acids/metabolism , Female , Glucose/metabolism , Lactic Acid/metabolism , Myocardial Contraction , Rats , Rats, Sprague-Dawley
11.
Pharmacol Res ; 132: 168-175, 2018 06.
Article in English | MEDLINE | ID: mdl-29253525

ABSTRACT

Normal liver function includes a number of metabolic processes, secretion of cellular mediators and its role in immunobiology; these require a high energy supply, which is further enhanced under adverse conditions triggering hepatic disorders or injury due to the operation of counteracting mechanisms. Alterations in oxygen availability, such as ischemia-reperfusion (IR) leading to liver inflammation and high-fat diet (HFD)-induced hepatic steatosis, are noxious responses encountered in hepatic surgery and obesity, respectively. Several strategies have been developed to attenuate or prevent these disorders, including thyroid hormone (T3), docosahexaenoic acid (DHA) and extra virgin olive oil (EVOO). These hormetic agents that exert beneficial effects in the low dose range were shown to abrogate IR-induced liver injury effectively in the case of T3, DHA, or their combined administration, whereas DHA plus EVOO attenuate HFD-induced hepatic steatosis, although they can induce adverse effects in other experimental settings. The use of combined hepatoprotective protocols (DHA + T3 or DHA + EVOO) using low doses or reduced supplementation periods is characterized by the stimulation of different types of molecular defensive mechanisms and similar signaling processes that exhibit synergism, thus constituting suitable experimental liver pharmacological preconditioning strategies with possible future clinical applications.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Olive Oil/therapeutic use , Reperfusion Injury/drug therapy , Triiodothyronine/therapeutic use , Animals , Diet, High-Fat , Drug Therapy, Combination , Humans , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Reperfusion Injury/metabolism
12.
Br J Nutr ; 119(2): 131-142, 2018 01.
Article in English | MEDLINE | ID: mdl-29268800

ABSTRACT

This study investigates the effects of replacing dietary casein by soya protein on the underlying mechanisms involved in the impaired metabolic fate of glucose and lipid metabolisms in the heart of dyslipidaemic rats chronically fed (8 months) a sucrose-rich (62·5 %) diet (SRD). To test this hypothesis, Wistar rats were fed an SRD for 4 months. From months 4 to 8, half the animals continued with the SRD and the other half were fed an SRD in which casein was substituted by soya. The control group received a diet with maize starch as the carbohydrate source. Compared with the SRD-fed group, the following results were obtained. First, soya protein significantly (P<0·001) reduced the plasma NEFA levels and normalised dyslipidaemia and glucose homoeostasis, improving insulin resistance. The protein levels of fatty acid translocase at basal state and under insulin stimulation and the protein levels and activity of muscle-type carnitine palmitoyltransferase 1 were normalised. Second, a significant (P<0·001) reduction of TAG, long-chain acyl CoA and diacylglycerol levels was observed in the heart muscle. Third, soya protein significantly increased (P<0·01) GLUT4 protein level under insulin stimulation and normalised glucose phosphorylation and oxidation. A reduction of phosphorylated AMP protein kinase protein level was recorded without changes in uncoupling protein 2 and PPARα. Fourth, hydroxyproline concentration decreased in the left ventricle and hypertension was normalised. The new information provided shows the beneficial effects of soya protein upon the altered pathways of glucose and lipid metabolism in the heart muscle of this rat model.


Subject(s)
Dyslipidemias/metabolism , Glucose/metabolism , Hypertension/metabolism , Lipid Metabolism/drug effects , Myocardium/metabolism , Soybean Proteins/administration & dosage , Animals , Carnitine O-Palmitoyltransferase/analysis , Dietary Proteins/administration & dosage , Dietary Sucrose/administration & dosage , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Glucose/administration & dosage , Hydroxyproline/analysis , Insulin/blood , Insulin Resistance/physiology , Male , Myocardium/enzymology , PPAR alpha/analysis , Rats , Rats, Wistar
13.
Immunol Lett ; 184: 92-97, 2017 04.
Article in English | MEDLINE | ID: mdl-28109981

ABSTRACT

Thyroid hormone (T3) induces liver preconditioning (PC) against ischemia-reperfusion (IR), a response energetically supported by AMP-activated protein kinase (AMPK) upregulation. The aim of this work is to evaluate the influence of T3 on IR-induced liver NLRP3 inflammasome activation and the relevance of AMPK activity on liver injury by the use of the AMPK inhibitor compound C (CC). Male Sprague-Dawley rats were given 0.1mgT3/kg (time zero) and 10mg CC/kg (time zero and 24h) or the respective vehicles, and subjected to 1h ischemia-20h reperfusion 48h after hormone treatment. Measurements included parameters of liver injury, hepatic levels of mRNAs (qPCR) and proteins (Western Blot or ELISA). IR induced substantial distortion of liver architecture, hepatocyte necrosis, and neutrophil infiltration with increased serum aspartate aminotransferase (AST) levels. T3 suppressed IR liver injury and AST enhancement, effects that were reverted by CC. Concomitantly, IR-induced liver mRNA and protein expression of NLRP3 and interleukin-1ß (IL-1ß) were restrained by T3, whereas CC eliminated T3-dependent PC. In conclusion, in vivo T3 administration triggers liver PC against IR injury by suppressing the inflammatory response associated with hepatic NLRP3 and IL-1ß upregulation, with AMPK playing a causal role regulating energy dynamics to upkeep PC.


Subject(s)
Inflammasomes/metabolism , Liver Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/metabolism , Thyroid Hormones/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Gene Expression , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Liver Diseases/genetics , Liver Diseases/pathology , Male , Rats , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction/drug effects , Thyroid Hormones/pharmacology , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
14.
Braz. j. pharm. sci ; 52(4): 591-594, Oct.-Dec. 2016.
Article in English | LILACS | ID: biblio-951874

ABSTRACT

ABSTRACT Various studies have linked metformin, a universally antidiabetic drug, with semen quality; however, such a direct link has not been established. This review systematically addresses and summarizes the effect of metformin on semen quality, particularly sperm function. We searched the MEDLINE electronic database for English articles and abstracts containing the key words 'metformin' and 'sperm', and relevant articles were reviewed. In summary, metformin appears to have improved and provided positive impact on sperm quality. This effect may be due to the ability of metformin to reduce oxidative stress and lipid peroxidation, enhance 5'-AMP activated protein kinase activity, and restore the normal levels of pituitary-gonadal hormones. However, further clinical research is still necessary to confirm such effect.


Subject(s)
Semen/metabolism , Metformin/analysis , Metformin/adverse effects , Testosterone/pharmacology , Oxidative Stress
15.
Int Immunopharmacol ; 38: 298-305, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27344038

ABSTRACT

Obesity and insulin resistance have been associated with deterioration in asthma outcomes. High oxidative stress and deficient activation of AMP-activated protein kinase (AMPK) have emerged as important regulators linking insulin resistance and inflammation. This study aimed to evaluate the effects of resveratrol on obesity-associated allergic pulmonary inflammation. Male C57/Bl6 mice fed with high-fat diet to induce obesity (obese group) or standard-chow diet (lean group) were treated or not with resveratrol (100mg/kg/day, two weeks). Mice were sensitized and challenged with ovalbumin (OVA). At 48h thereafter, bronchoalveolar lavage fluid was performed, and lungs collected for morphological studies and Western blot analysis. Treatment of obese mice with resveratrol significantly reduced hyperglycemia and insulin resistance, as well as the body measures (body mass, fat mass, % fat, and body area). OVA-challenge promoted a higher increase in pulmonary eosinophil infiltration in obese compared with lean mice, which was nearly abrogated by resveratrol treatment. Resveratrol markedly increased the phosphorylated AMPK expression in lung tissues of obese compared with lean mice. Resveratrol reduced the p47phox expression and reactive-oxygen species (ROS) production, and elevated the superoxide dismutase (SOD) levels in lung tissues of obese mice. The increased pulmonary levels of TNF-α and inducible nitric oxide synthase (iNOS) in obese mice were also normalized after resveratrol treatment. In lean mice, resveratrol failed to affect the levels of fasting glucose, p47phox, ROS levels, TNF-α, iNOS and phosphorylated AMPK. Resveratrol exhibits protective effects in obesity-associated lung inflammation that is accompanied by local AMPK activation and antioxidant property.


Subject(s)
Antioxidants/therapeutic use , Asthma/drug therapy , Eosinophils/physiology , Lung/drug effects , Obesity/drug therapy , Pneumonia/drug therapy , Stilbenes/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Disease Progression , Lung/pathology , Mice , Mice, Inbred Strains , Resveratrol
16.
J Renin Angiotensin Aldosterone Syst ; 16(3): 481-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-23950549

ABSTRACT

INTRODUCTION: adenosine monophosphate-activated protein kinase (AMPK) plays a prominent role as a metabolic stress sensor, and it has recently been suggested that the renin-angiotensin system, in addition to its role in stress regulation, may play a significant role in regulating the AMPK system. This study aimed to evaluate the effects of candesartan, an angiotensin II receptor blocker, on cardiac and hepatic AMPK activity basally as well as after surgical stress under general anesthesia. MATERIALS AND METHODS: Male Wistar rats were treated with 5 mg/kg/day candesartan in their drinking water for two weeks. Levels of cardiac and hepatic AMPK activity were determined, using a kinase activity assay, basally and after surgical stress under general anesthesia. RESULTS: Chronic administration of candesartan increased hepatic AMPK activity approximately 4 times (p<0.05) while no significant change was demonstrated in cardiac AMPK. Cardiac and hepatic AMPK activities were not significantly increased by surgical stress alone performed under anesthesia. However, chronic treatment with candesartan decreased AMPK activity in both liver and heart after surgical stress under anesthesia (p<0.01 for both comparisons). CONCLUSIONS: While chronic candesartan treatment may stimulate AMPK activity in certain organs such as the liver, when combined with surgical stress under anesthesia it inhibits pathways regulating AMPK activity.


Subject(s)
Adenylate Kinase/metabolism , Benzimidazoles/pharmacology , Laparotomy , Liver/enzymology , Myocardium/enzymology , Stress, Physiological , Tetrazoles/pharmacology , Animals , Benzimidazoles/administration & dosage , Biphenyl Compounds , Liver/drug effects , Male , Rats, Wistar , Stress, Physiological/drug effects , Tetrazoles/administration & dosage
17.
World J Gastroenterol ; 20(46): 17416-25, 2014 Dec 14.
Article in English | MEDLINE | ID: mdl-25516653

ABSTRACT

AIM: To investigate the redox dependency and promotion of downstream targets in thyroid hormone (T3)-induced AMP-activated protein kinase (AMPK) signaling as cellular energy sensor to limit metabolic stresses in the liver. METHODS: Fed male Sprague-Dawley rats were given a single ip dose of 0.1 mg T3/kg or T3 vehicle (NaOH 0.1 N; controls) and studied at 8 or 24 h after treatment. Separate groups of animals received 500 mg N-acetylcysteine (NAC)/kg or saline ip 30 min prior T3. Measurements included plasma and liver 8-isoprostane and serum ß-hydroxybutyrate levels (ELISA), hepatic levels of mRNAs (qPCR), proteins (Western blot), and phosphorylated AMPK (ELISA). RESULTS: T3 upregulates AMPK signaling, including the upstream kinases Ca(2+)-calmodulin-dependent protein kinase kinase-ß and transforming growth factor-ß-activated kinase-1, with T3-induced reactive oxygen species having a causal role due to its suppression by pretreatment with the antioxidant NAC. Accordingly, AMPK targets acetyl-CoA carboxylase and cyclic AMP response element binding protein are phosphorylated, with the concomitant carnitine palmitoyltransferase-1α (CPT-1α) activation and higher expression of peroxisome proliferator-activated receptor-γ co-activator-1α and that of the fatty acid oxidation (FAO)-related enzymes CPT-1α, acyl-CoA oxidase 1, and acyl-CoA thioesterase 2. Under these conditions, T3 induced a significant increase in the serum levels of ß-hydroxybutyrate, a surrogate marker for hepatic FAO. CONCLUSION: T3 administration activates liver AMPK signaling in a redox-dependent manner, leading to FAO enhancement as evidenced by the consequent ketogenic response, which may constitute a key molecular mechanism regulating energy dynamics to support T3 preconditioning against ischemia-reperfusion injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fatty Acids/metabolism , Liver/drug effects , Triiodothyronine/pharmacology , 3-Hydroxybutyric Acid/blood , AMP-Activated Protein Kinases/genetics , Animals , Antioxidants/pharmacology , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Enzyme Activation , Gene Expression Regulation, Enzymologic , Injections, Intraperitoneal , Liver/enzymology , Male , Oxidation-Reduction , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Time Factors , Triiodothyronine/administration & dosage
18.
Biochem Biophys Res Commun ; 441(4): 923-8, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24216105

ABSTRACT

Mammalian target of rapamycin (mTOR) complex is a key regulator of autophagy, cell growth and proliferation. Here, we studied the effects of arginine vasopressin (AVP) on mTOR activation in vascular smooth muscle cells cultured in high glucose concentration. AVP induced the mTOR phosphorylation in A-10 cells grown in high glucose, in contrast to cells cultured in normal glucose; wherein, only basal phosphorylation was observed. The AVP-induced mTOR phosphorylation was inhibited by a PI3K inhibitor. Moreover, the AVP-induced mTOR activation inhibited autophagy and increased thymidine incorporation in cells grown in high glucose. This increase was abolished by rapamycin which inhibits the mTORC1 complex formation. Our results suggest that AVP stimulates mTOR phosphorylation by activating the PI3K/Akt signaling pathway and, subsequently, inhibits autophagy and raises cell proliferation in A-10 cells maintained in high glucose concentration.


Subject(s)
Arginine Vasopressin/pharmacology , Glucose/metabolism , Hyperglycemia/enzymology , Multiprotein Complexes/metabolism , Myocytes, Smooth Muscle/drug effects , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Chromones/pharmacology , Glucose/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Morpholines/pharmacology , Myocytes, Smooth Muscle/enzymology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats
SELECTION OF CITATIONS
SEARCH DETAIL