Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 116
1.
Am J Physiol Endocrinol Metab ; 326(2): E134-E147, 2024 02 01.
Article En | MEDLINE | ID: mdl-38117265

Vertical sleeve gastrectomy (VSG) restores glucose homeostasis in obese mice and humans. In addition, the increased fibroblast growth factor (FGF)15/19 circulating level postsurgery has been implicated in this effect. However, the impact of FGF15/19 on pancreatic islets remains unclear. Using a diet-induced obese mice model, we demonstrate that VSG attenuates insulin hypersecretion in isolated pancreatic islets, likely due to morphological alterations in the endocrine pancreas such as reduction in islet, ß-cell, and α-cell mass. In addition, VSG relieves gene expression of endoplasmic reticulum (ER) stress and inflammation markers in islets from obese mice. Incubation of INS-1E ß-cells with serum from obese mice induced dysfunction and cell death, whereas these conditions were not induced with serum from obese mice submitted to VSG, implicating the involvement of a humoral factor. Indeed, VSG increased FGF15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E cells treated with the serum from these mice. Moreover, exposing INS-1E cells to an FGFR inhibitor abolished the effects of VSG serum on insulin secretion and cell death. Also, recombinant FGF19 prevents INS-1E cells from dysfunction and death induced by serum from obese mice. These findings indicate that the amelioration of glucose-insulin homeostasis promoted by VSG is mediated, at least in part, by FGF15/19. Therefore, approaches promoting FGF15/19 release or action may restore pancreatic islet function in obesity.NEW & NOTEWORTHY Vertical sleeve gastrectomy (VSG) decreases insulin secretion, endoplasmic reticulum (ER) stress, and inflammation in pancreatic islets from obese mice. In addition, VSG increased fibroblast growth factor (FGF)15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E ß-cells treated with the serum from these mice. Serum from operated mice protects INS-1E cells from dysfunction and apoptosis, which was mediated by FGF15/19.


Insulin-Secreting Cells , Insulin , Mice , Humans , Animals , Insulin/metabolism , Mice, Obese , Insulin-Secreting Cells/metabolism , Glucose/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Gastrectomy , Inflammation/metabolism , Homeostasis
2.
Ann Anat ; 241: 151891, 2022 Apr.
Article En | MEDLINE | ID: mdl-35114378

Cell-to-cell interactions mediated by intercellular junctions (IJs) are crucial for beta-cell functioning and proper insulin secretion, however, their role in type-2 diabetes is still unclear. This work aimed to evaluate the cellular distribution and expression of proteins associated with adherens (AJs) and gap junctions (GJs) in pancreatic islets of C57BL6 mice fed a high-fat (HF) diet. The administration of HF diet for 30 days induced an increase in body weight, post-prandial glycemia, insulinemia, glucose intolerance, and moderate insulin resistance associated with mild perturbations in insulin secretion. The intercellular content of the AJ-associated proteins (namely, E-, N-cadherins, and α-, ß-catenins) was significantly higher in islet cells of HF-fed mice. Inversely, the gap junctional content of Cx36 was significantly decreased, as revealed by immunofluorescence, which was paralleled by a reduction in the frequency of calcium oscillations in islets of prediabetic mice. In conclusion, the endocrine pancreas displays significant changes in the content of several junctional proteins at the cell-cell contact region following short-term HF diet administration, indicating that IJs may be involved in the adaptive response of beta cells seen during this state.


Insulin-Secreting Cells , Islets of Langerhans , Animals , Cell Adhesion Molecules/metabolism , Diet, High-Fat/adverse effects , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL
3.
Life Sci ; 291: 120239, 2022 Feb 15.
Article En | MEDLINE | ID: mdl-34942163

Aim Investigate whether inheritance of improved skeletal muscle mitochondrial function and its association with glycemic control are multigenerational benefits of exercise. MAIN METHODS: Male Swiss mice were subjected to 8 weeks of endurance training and mated with untrained females. KEY FINDINGS: Trained fathers displayed typical endurance training-induced adaptations. Remarkably, offspring from trained fathers also exhibited higher endurance performance, mitochondrial oxygen consumption, glucose tolerance and insulin sensitivity. However, PGC-1α expression was not increased in the offspring. In the offspring, the expression of the co-repressor NCoR1 was reduced, increasing activation of PGC-1α target genes. These effects correlated with higher DNA methylation at the NCoR1 promoter in both, the sperm of trained fathers and in the skeletal muscle of their offspring. SIGNIFICANCE: Higher skeletal muscle mitochondrial function is inherited by epigenetic de-activation of a key PGC-1α co-repressor.


Mitochondria/metabolism , Physical Conditioning, Animal/physiology , Physical Exertion/physiology , Animals , DNA Methylation , Epigenesis, Genetic/genetics , Female , Male , Mice , Mitochondria/physiology , Muscle, Skeletal/physiology , Nuclear Receptor Co-Repressor 1/metabolism , Oxygen Consumption/physiology , Paternal Inheritance/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/physiology , Physical Conditioning, Animal/methods , RNA, Messenger/genetics
4.
Life Sci ; 285: 120020, 2021 Nov 15.
Article En | MEDLINE | ID: mdl-34624320

AIMS: The bile acid (BA), tauroursodeoxycholic acid (TUDCA) regulates glucose homeostasis; however, it is not clear whether its effects on insulin signaling are due to its direct interaction with the insulin receptor (IR) or through activation of the G-coupled BA receptor, TGR5. We, herein, investigated whether the actions of TUDCA on glucose homeostasis occur via IR or TGR5 activation. MAIN METHODS: Glucose homeostasis was evaluated in high-fat diet (HFD)-obese or control (CTL) mice, after 30 days or one intraperitoneal (ip) injection of 300 mg/kg TUDCA, respectively. Molecular docking was performed to investigate the potential binding of TUDCA on the IR and TGR5. KEY FINDINGS: After 30 days of TUDCA treatment, HFD mice exhibited improvements in glucose tolerance and insulin sensitivity, which were abolished when these rodents received the IR antagonist, S961. Molecular docking experiments showed that TUDCA demonstrates high binding affinity for TGR5 and IR and strongly interacts with the insulin binding sites 1 and 2 of the IR. Consistent with this potential agonist activity of TUDCA on IR, CTL mice displayed increased hepatic phosphorylation of AKT after an ip injection of TUDCA. This effect was not associated with altered glycemia in CTL mice and was dependent on IR activation, as S961 prevented hepatic AKT activation by TUDCA. Furthermore, TUDCA activated the hepatic protein kinase A (PKA) and cAMP response element-binding protein (CREB) pathway in CTL mice, even after the administration of S961. SIGNIFICANCE: We provide novel evidence that TUDCA may be an agonist of the IR, in turn activating AKT and contributing, at least in part, to its beneficial effects upon glucose homeostasis.


Glucose/metabolism , Receptor, Insulin/agonists , Taurochenodeoxycholic Acid/pharmacology , Animals , Binding Sites , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose Tolerance Test , Homeostasis/drug effects , Male , Mice , Molecular Docking Simulation , Obesity/metabolism , Protein Binding , Receptor, Insulin/chemistry , Receptors, G-Protein-Coupled/metabolism , Taurochenodeoxycholic Acid/administration & dosage
5.
Sci Rep ; 11(1): 18114, 2021 09 13.
Article En | MEDLINE | ID: mdl-34518585

Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia. While cognitive deficits remain the major manifestation of AD, metabolic and non-cognitive abnormalities, such as alterations in food intake, body weight and energy balance are also present, both in AD patients and animal models. In this sense, the tauroursodeoxycholic acid (TUDCA) has shown beneficial effects both in reducing the central and cognitive markers of AD, as well as in attenuating the metabolic disorders associated with it. We previously demonstrated that TUDCA improves glucose homeostasis and decreases the main AD neuromarkers in the streptozotocin-induced AD mouse model (Stz). Besides that, TUDCA-treated Stz mice showed lower body weight and adiposity. Here, we investigated the actions of TUDCA involved in the regulation of body weight and adiposity in Stz mice, since the effects of TUDCA in hypothalamic appetite control and energy homeostasis have not yet been explored in an AD mice model. The TUDCA-treated mice (Stz + TUDCA) displayed lower food intake, higher energy expenditure (EE) and respiratory quotient. In addition, we observed in the hypothalamus of the Stz + TUDCA mice reduced fluorescence and gene expression of inflammatory markers, as well as normalization of the orexigenic neuropeptides AgRP and NPY expression. Moreover, leptin-induced p-JAK2 and p-STAT3 signaling in the hypothalamus of Stz + TUDCA mice was improved, accompanied by reduced acute food intake after leptin stimulation. Taken together, we demonstrate that TUDCA treatment restores energy metabolism in Stz mice, a phenomenon that is associated with reduced food intake, increased EE and improved hypothalamic leptin signaling. These findings suggest treatment with TUDCA as a promising therapeutic intervention for the control of energy homeostasis in AD individuals.


Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Energy Metabolism/drug effects , Homeostasis , Streptozocin/adverse effects , Taurochenodeoxycholic Acid/pharmacology , Adiposity , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Biomarkers , Body Weight , Disease Management , Disease Models, Animal , Gene Expression , Immunohistochemistry , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Leptin/metabolism , Male , Mice , Organ Specificity , Signal Transduction , Thermogenesis
6.
Front Endocrinol (Lausanne) ; 12: 679492, 2021.
Article En | MEDLINE | ID: mdl-34054736

Hyperinsulinemia is frequently associated with aging and may cause insulin resistance in elderly. Since insulin secretion and clearance decline with age, hyperinsulinemia seems to be maintained, primarily, due to a decrease in the insulin clearance. To investigate these aging effects, 3- and 18-month-old male C57BL/6 mice were subjected to intraperitoneal glucose and insulin tolerance tests (ipGTT and ipITT) and, during the ipGTT, plasma c-peptide and insulin were measure to evaluate in vivo insulin clearance. Glucose-stimulated insulin secretion in isolated pancreatic islets was also assessed, and liver samples were collected for molecular analyses (western blot). Although insulin sensitivity was not altered in the old mice, glucose tolerance, paradoxically, seems to be increased, accompanied by higher plasma insulin, during ipGTT. While insulin secretion did not increase, insulin clearance was reduced in the old mice, as suggested by the lower c-peptide:insulin ratio, observed during ipGTT. Carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1) and insulin-degrading enzyme (IDE), as well as the activity of this enzyme, were reduced in the liver of old mice, justifying the decreased insulin clearance observed in these mice. Therefore, loss of hepatic CEACAM1 and IDE function may be directly related to the decline in insulin clearance during aging.


Aging/metabolism , Glucose/pharmacology , Insulin Secretion/drug effects , Insulin/metabolism , Islets of Langerhans/drug effects , Animals , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Glucose Tolerance Test , Insulin/blood , Insulin Resistance/physiology , Insulin Secretion/physiology , Insulysin/metabolism , Islets of Langerhans/metabolism , Liver/metabolism , Male , Mice
7.
Life Sci ; 272: 119252, 2021 May 01.
Article En | MEDLINE | ID: mdl-33636170

Bear bile has been used in Traditional Chinese Medicine for thousands of years due to its therapeutic potential and clinical applications. The tauroursodeoxycholic acid (TUDCA), one of the acids found in bear bile, is a hydrophilic bile acid and naturally produced in the liver by conjugation of taurine to ursodeoxycholic acid (UDCA). Several studies have shown that TUDCA has neuroprotective action in several models of neurodegenerative disorders (ND), including Alzheimer's disease, Parkinson's disease, and Huntington's disease, based on its potent ability to inhibit apoptosis, attenuate oxidative stress, and reduce endoplasmic reticulum stress in different experimental models of these illnesses. Our research extends the knowledge of the bile acid TUDCA actions in ND and the mechanisms and pathways involved in its cytoprotective effects on the brain, providing a novel perspective and opportunities for treatment of these diseases.


Neurodegenerative Diseases/drug therapy , Taurochenodeoxycholic Acid/pharmacology , Alzheimer Disease/drug therapy , Animals , Apoptosis/drug effects , Bile/metabolism , Bile Acids and Salts/metabolism , Endoplasmic Reticulum Stress/drug effects , Humans , Medicine, Chinese Traditional/methods , Taurochenodeoxycholic Acid/metabolism , Ursodeoxycholic Acid/metabolism , Ursodeoxycholic Acid/pharmacology
8.
Metabolism ; 116: 154701, 2021 03.
Article En | MEDLINE | ID: mdl-33417894

BACKGROUND: Protein malnutrition in childhood predisposes individuals to vascular and pancreatic endocrine dysfunction, thus increasing the risk of diabetes and hypertension. Because taurine may reduce cardiometabolic risk, we hypothesized that taurine treatment has a beneficial effect on the pancreatic vasculature during protein restriction. METHODS AND RESULTS: Weaned mice were fed a normal or a low-protein diet and were treated with or without taurine for 3 months. The lieno-pancreatic artery (LPA) from low-protein diet-treated mice exhibited impaired endothelium-dependent relaxation to acetylcholine that was associated with decreased endothelium-derived hyperpolarization (EDH), hydrogen sulfide (H2S) production, and H2S-synthesizing CBS expression and impaired vasorelaxation to an H2S-donor, NaHS. These changes were prevented by taurine treatment. We compared the effects of taurine with the effects of the direct vasodilator hydralazine and found that both normalized blood pressure and the endothelial vasodilator function of the LPA in the mice fed a protein-restricted diet. However, only taurine restored the CBS expression in the LPA and insulin secretion in response to high glucose. The LPA supplies the pancreas and shares morphometry with the mesenteric resistance artery (MRA). However, in the MRA, low-protein diet-induced endothelial dysfunction is driven by impaired NOS-derived NO with no changes in H2S signaling. CONCLUSIONS: The results suggest that taurine protects against protein malnutrition-induced endothelial dysfunction in the LPA by upregulating the CBS-H2S pathway. Considering the importance of the pancreatic vasculature for endocrine islet activity, taurine may be a potential therapy for the vascular and metabolic dysfunction associated with malnutrition and comorbidities.


Endothelium, Vascular/drug effects , Hydrogen Sulfide/pharmacology , Malnutrition/complications , Pancreas/drug effects , Protein Deficiency/complications , Taurine/therapeutic use , Animals , Blood Pressure/drug effects , Dietary Proteins/administration & dosage , Endothelium, Vascular/physiopathology , Malnutrition/drug therapy , Malnutrition/physiopathology , Mice , Mice, Inbred C57BL , Pancreas/blood supply , Pancreas/physiopathology , Protein Deficiency/drug therapy , Protein Deficiency/physiopathology , Vasodilation/drug effects
9.
Mol Cell Endocrinol ; 521: 111116, 2021 02 05.
Article En | MEDLINE | ID: mdl-33321116

Alzheimer's disease (AD) is a neurodegenerative disorder and the major cause of dementia. According to predictions of the World Health Organization, more than 150 million people worldwide will suffer from dementia by 2050. An increasing number of studies have associated AD with type 2 diabetes mellitus (T2DM), since most of the features found in T2DM are also observed in AD, such as insulin resistance and glucose intolerance. In this sense, some bile acids have emerged as new therapeutic targets to treat AD and metabolic disorders. The taurine conjugated bile acid, tauroursodeoxycholic (TUDCA), reduces amyloid oligomer accumulation and improves cognition in APP/PS1 mice model of AD, and also improves glucose-insulin homeostasis in obese and type 2 diabetic mice. Herein, we investigated the effect of TUDCA upon glucose metabolism in streptozotocin-induced AD mice model (Stz). The Stz mice that received 300 mg/kg TUDCA during 10 days (Stz + TUDCA), showed improvement in glucose tolerance and insulin sensitivity, reduced fasted and fed glycemia, increased islet mass and ß-cell area, as well as increased glucose-stimulated insulin secretion, compared with Stz mice that received only PBS. Stz + TUDCA mice also displayed lower neuroinflammation, reduced protein content of amyloid oligomer in the hippocampus, improved memory test and increased protein content of insulin receptor ß-subunit in the hippocampus. In conclusion, TUDCA treatment enhanced glucose homeostasis in the streptozotocin-induced Alzheimer's disease mice model, pointing this bile acid as a good strategy to counteract glucose homeostasis disturbance in AD pathology.


Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Bile Acids and Salts/metabolism , Blood Glucose/drug effects , Hippocampus/drug effects , Insulin-Secreting Cells/drug effects , Taurochenodeoxycholic Acid/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Glucose/metabolism , Glucose/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/drug therapy , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Memory and Learning Tests , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Streptozocin/toxicity , Taurochenodeoxycholic Acid/administration & dosage
10.
Int J Mol Sci ; 23(1)2021 Dec 21.
Article En | MEDLINE | ID: mdl-35008427

BACKGROUND/AIMS: Epigenetic regulation is considered the main molecular mechanism underlying the developmental origin of health and disease's (DOHAD) hypothesis. Previous studies that have investigated the role of paternal exercise on the metabolic health of the offspring did not control for the amount and intensity of the training or possible effects of adaptation to exercise and produced conflicting results regarding the benefits of parental exercise to the next generation. We employed a precisely regulated exercise regimen to study the transgenerational inheritance of improved metabolic health. METHODS: We subjected male mice to a well-controlled exercise -training program to investigate the effects of paternal exercise on glucose tolerance and insulin sensitivity in their adult progeny. To investigate the molecular mechanisms of epigenetic inheritance, we determined chromatin markers in the skeletal muscle of the offspring and the paternal sperm. RESULTS: Offspring of trained male mice exhibited improved glucose homeostasis and insulin sensitivity. Paternal exercise modulated the DNA methylation profile of PI3Kca and the imprinted H19/Igf2 locus at specific differentially methylated regions (DMRs) in the skeletal muscle of the offspring, which affected their gene expression. Remarkably, a similar DNA methylation profile at the PI3Kca, H19, and Igf2 genes was present in the progenitor sperm indicating that exercise-induced epigenetic changes that occurred during germ cell development contributed to transgenerational transmission. CONCLUSION: Paternal exercise might be considered as a strategy that could promote metabolic health in the offspring as the benefits can be inherited transgenerationally.


Class I Phosphatidylinositol 3-Kinases/genetics , DNA Methylation , Insulin Resistance/genetics , Insulin-Like Growth Factor II/genetics , Physical Conditioning, Animal/methods , RNA, Long Noncoding/genetics , Spermatozoa/chemistry , Animals , Epigenesis, Genetic , Female , Glucose Tolerance Test , High-Throughput Nucleotide Sequencing , Male , Mice , Models, Animal , Oxygen Consumption , Paternal Inheritance , Sequence Analysis, DNA , Spermatozoa/metabolism
11.
An Acad Bras Cienc ; 92(4): e20201382, 2020.
Article En | MEDLINE | ID: mdl-33237150

D-pinitol is one of the major inositol found in plants and studies suggest its potential hypoglycemic and hypolipidemic actions in diabetic rodents. Here, we investigated the actions of D-pinitol on adiposity, and in lipid and glycemic homeostasis in monosodium glutamate (MSG)-obese mice. Swiss mice received daily subcutaneous injections of MSG [(4g/kg of body weight (BW)] or saline [1.25g/kg BW; control (CTL)] during their first five days of life. From 90-120 day-old, half of the MSG and CTL groups received 50 mg D-pinitol/kg BW/day (MPIN and CPIN groups) or vehicle (saline; MSG and CTL groups) by gavage. MSG mice displayed higher abdominal adiposity and hepatic triglycerides (TG) deposition, and increased hepatic expression of lipogenic genes (SREBP-1c, ACC-1 and FASN), but downregulation in AMPKα mRNA. MSG mice also exhibited hyperinsulinemia, islet hypersecretion and hypertrophy, glucose intolerance and insulin resistance. D-pinitol did not change adiposity, glucose intolerance, insulin resistance, but increased hepatic triglycerides (TG) content in MPIN mice, which was associated with increases in gene expressions of SREBP-1c and FASN, but reduction in AMPKα. Furthermore, D-pinitol enhanced insulin secretion in MPIN and CPIN groups. Therefore, D-pinitol enhanced glucose-induced insulin secretion, which may account to enhances hepatic lipogenesis and TG deposition in MPIN mice.


Lipid Metabolism , Sodium Glutamate , Animals , Blood Glucose , Inositol/analogs & derivatives , Insulin Secretion , Lipids , Mice , Mice, Obese
12.
Life Sci ; 256: 118012, 2020 Sep 01.
Article En | MEDLINE | ID: mdl-32593710

AIMS: Bisphenol (BP)-A exposure can impair glucose and lipid metabolism. However, it is unclear whether this endocrine disruptor (ED) modulates these processes in postmenopause, a period with organic changes that increase the risk for metabolic diseases. Herein, we evaluated the effects of BPA exposure on adiposity, glucose homeostasis and hepatic steatosis in ovariectomized (OVX) mice fed on a high-fat diet (HFD). MAIN METHODS: Adult Swiss female mice were OVX and submitted to a normolipidic diet or HFD and drinking water without [control (OVX CTL) and OVX HFD groups, respectively] or with 1 µg/mL BPA (OVX CBPA and OVX HBPA groups, respectively), for 3 months. KEY FINDINGS: OVX HFD females displayed increased adiposity, glucose intolerance, insulin resistance and moderate hepatic steatosis. This effect was associated with a high hepatic expression of genes involved in lipogenesis (Srebf1 and Scd1), ß-oxidation (Cpt1a) and endoplasmic reticulum (ER) stress (Hspa5 and Hyou1). BPA did not alter adiposity or glucose homeostasis disruptions induced by HFD. However, this ED triggered severe steatosis, exacerbating hepatic fat and collagen depositions in OVX HBPA, in association with a reduction in Mttp mRNA, and up-regulation of genes involved in ß-oxidation (Acox1 and Acadvl), mitochondrial uncoupling (Ucp2), ER stress (Hyou1 and Atf6) and chronic liver injury (Tgfb1and Casp8). Furthermore, BPA caused mild steatosis in OVX CBPA females, increasing the hepatic total lipids and mRNAs for Srebf1, Scd1, Hspa5, Hyou1 and Atf6. SIGNIFICANCE: BPA aggravated hepatic steatosis in OVX mice. Especially when combined with a HFD, BPA caused NAFLD progression, which was partly mediated by chronic ER stress and the TGF-ß1 pathway.


Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Endoplasmic Reticulum Stress/drug effects , Non-alcoholic Fatty Liver Disease/chemically induced , Phenols/toxicity , Adiposity/drug effects , Animals , Diet, High-Fat , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum Chaperone BiP , Female , Glucose/metabolism , Insulin Resistance , Lipogenesis/drug effects , Mice , Non-alcoholic Fatty Liver Disease/pathology , Ovariectomy
13.
Nitric Oxide ; 94: 48-53, 2020 01 01.
Article En | MEDLINE | ID: mdl-31669041

Taurine is a semiessential amino acid found at high concentrations in mammalian plasma and cells, where it regulates cellular functions such as ion flux, controls cell volume and serves as a substrate for conjugated bile acids (BAs). Exogenous administration of both taurine and taurine-conjugated BAs have also been implicated in the modulation of cardiovascular functions. This brief review summarizes the role of taurine and taurine-conjugated BAs in vascular relaxation through the modulation of endothelium-derived nitric oxide (NO). The effects of taurine on vascular health are controversial. However, in the presence of cardiometabolic risk factors, it has been proposed that taurine can increase vascular NO levels by increasing eNOS expression, eNOS phosphorylation on Ser1177, NO bioavailability, the level of antioxidative defense, and the l-arginine/NOS inhibitor asymmetric dimethylarginine (ADMA) ratio. The taurine-conjugated BA-mediated activation of Farnesoid X receptor (FXR), G protein-coupled BA receptor (TGR5) and/or muscarinic 3 receptor (M3) was also reported to increase vascular NO production. FXR activation increases eNOS expression and may reduce ADMA formation, while TGR5 increases mobilization of Ca2+ and phosphorylation of eNOS and Akt in endothelial cells. Furthermore, taurine and taurine-conjugated BAs might regulate NO synthesis and activity by enhancing H2S generation. Several studies have demonstrated the beneficial effects of both taurine and taurine-conjugated BAs in reversing the endothelial dysfunction associated with diabetes, atherosclerosis, hypertension, obesity, malnutrition, and smoking. In addition, taurine-conjugated BAs have emerged as a potential treatment for portal hypertension. Despite these favorable findings, there is a need to further explore the mechanisms and signaling pathways underlying the endothelial effects of taurine and taurine-conjugated BAs. Here, we summarize the main findings regarding the effects of taurine and taurine-conjugated BAs on the endothelial dysfunction associated with altered NO metabolism in cardiovascular diseases.


Bile Acids and Salts/metabolism , Endothelial Cells/metabolism , Nitric Oxide/biosynthesis , Taurine/metabolism , Animals , Humans
14.
Mol Metab ; 29: 124-135, 2019 11.
Article En | MEDLINE | ID: mdl-31668384

OBJECTIVE: Dietary restriction (DR) improves health and prolongs lifespan in part by upregulating type III endoribonuclease DICER in adipose tissue. In this study, we aimed to specifically test which missing dietary component was responsible for DICER upregulation. METHODS: We performed a nutrient screen in mouse preadipocytes and validated the results in vivo using different kinds of dietary interventions in wild type or genetically modified mice and worms, also testing the requirement of DICER on the effects of the diets. RESULTS: We found that sulfur amino acid restriction (i.e., methionine or cysteine) is sufficient to increase Dicer mRNA expression in preadipocytes. Consistently, while DR increases DICER expression in adipose tissue of mice, this effect is blunted by supplementation of the diet with methionine, cysteine, or casein, but not with a lipid or carbohydrate source. Accordingly, dietary methionine or protein restriction mirrors the effects of DR. These changes are associated with alterations in serum adiponectin. We also found that DICER controls and is controlled by adiponectin. In mice, DICER plays a role in methionine restriction-induced upregulation of Ucp1 in adipose tissue. In C. elegans, DR and a model of methionine restriction also promote DICER expression in the intestine (an analog of the adipose tissue) and prolong lifespan in a DICER-dependent manner. CONCLUSIONS: We propose an evolutionary conserved mechanism in which dietary sulfur amino acid restriction upregulates DICER levels in adipose tissue leading to beneficial health effects.


Cysteine/deficiency , DEAD-box RNA Helicases/metabolism , Methionine/deficiency , Adipocytes/cytology , Adipocytes/metabolism , Adiponectin/blood , Adiponectin/metabolism , Adipose Tissue, Beige/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Diet/methods , Diet/veterinary , Intestinal Mucosa/metabolism , Longevity , Male , Mice, Inbred C57BL , Mice, Knockout , Ribonuclease III/genetics , Ribonuclease III/metabolism , Uncoupling Protein 1/metabolism , Up-Regulation
15.
Adv Physiol Educ ; 43(3): 266-269, 2019 Sep 01.
Article En | MEDLINE | ID: mdl-31166122

Obesity and diabetes are two of the biggest public health problems in the modern world. One possible way to combat the rising prevalence of these diseases is through the spread of awareness about its consequences and how to prevent them. Therefore, educational interventions focused on teaching the physiological basis of these conditions might be valuable tools. However, most scholar curriculums lack high-quality material devoted to this topic. Thus we developed an educational booklet, composed of playful elements, targeted toward high school students and destined for application in classrooms. The efficacy of the developed material was validated through a pretest-posttest design, in which the students had to answer a 10-question test. After booklet completion, students had better outcomes, with an increase in the percentage of correct answers in 7 out of 10 questions contained in the test (P < 0.05). Thus we developed an effective material for usage in the high school classroom to spread the awareness of the risks of metabolic diseases and how to prevent them.


Diabetes Mellitus/physiopathology , Health Education/methods , Obesity/physiopathology , Pamphlets , Play and Playthings/psychology , Students/psychology , Adolescent , Brazil/epidemiology , Diabetes Mellitus/epidemiology , Female , Humans , Male , Obesity/epidemiology , Schools
16.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1126-1137, 2019 06 01.
Article En | MEDLINE | ID: mdl-30738810

In experimental obesity, the hypothalamus is affected by an inflammatory response activated by dietary saturated fats. This inflammation is triggered as early as one day after exposure to a high-fat diet, and during its progression, there is recruitment of inflammatory cells from the systemic circulation. The objective of the present study was identifying chemokines potentially involved in the development of hypothalamic diet-induced inflammation. In order to identify chemokines potentially involved in this process, we performed a real-time PCR array that determined Ackr2 as one of the transcripts undergoing differential regulation in obese-prone as compared to obese-resistant mice fed a high-fat diet for three days. ACKR2 is a decoy receptor that acts as an inhibitor of the signals generated by several CC inflammatory chemokines. Our results show that Ackr2 expression is rapidly induced after exposure to dietary fats both in obese-prone and obese-resistant mice. In immunofluorescence studies, ACKR2 was detected in hypothalamic neurons expressing POMC and NPY and also in microglia and astrocytes. The lentiviral overexpression of ACKR2 in the hypothalamus reduced diet-induced hypothalamic inflammation; however, there was no change in spontaneous caloric intake and body mass. Nevertheless, the overexpression of ACKR2 resulted in improvement of glucose tolerance, which was accompanied by reduced insulin secretion and increased whole body insulin sensitivity. Thus, ACKR2 is a decoy chemokine receptor expressed in most hypothalamic cells that is modulated by dietary intervention and acts to reduce diet-induced inflammation, leading to improved glucose tolerance due to improved insulin action.


Gene Expression Profiling , Glucose/metabolism , Hypothalamus/metabolism , Inflammation/genetics , Obesity/genetics , Receptors, Chemokine/genetics , Animals , Astrocytes/metabolism , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat/adverse effects , Glucose Tolerance Test , Hypothalamus/cytology , Inflammation/etiology , Inflammation/metabolism , Insulin Resistance/genetics , Male , Mice , Neurons/metabolism , Obesity/etiology , Obesity/metabolism , Receptors, Chemokine/metabolism
17.
Eur J Nutr ; 58(2): 551-563, 2019 Mar.
Article En | MEDLINE | ID: mdl-29374793

PURPOSE: Obesity predisposes to cardiovascular and metabolic diseases. The amino acid, L-taurine (Tau), regulates glucose and lipid homeostasis and vascular function. Here we investigated whether Tau supplementation prevents endothelial dysfunction in the thoracic aortas of monosodium glutamate-induced obese (MSG) rats. METHODS: Male rats received subcutaneous injections of MSG (4 mg/kg body weight/day) or saline (control group, CTL) during the first five days of life. From 21 to 150 days of age, the rats were distributed into the groups: CTL, MSG, and CTL and MSG supplemented with 2.5% Tau in their drinking water (CTAU and MTAU). RESULTS: At 150-days old, MSG rats presented massive abdominal fat deposition, hypertriglyceridemia, hyperinsulinemia, glucose intolerance and high plasma levels of malondialdehyde (MDA), a lipid peroxidation marker. Tau supplementation attenuated fat accumulation in perigonadal adipose tissue and prevented the increase in triglycerides and MDA plasma levels. Aortic rings of MSG rats presented reduced vasodilation in response to acetylcholine (ACh). No modifications in insulin-induced vasodilatation, or Akt and eNOS phosphorylation, were observed in MSG aortas; thoracic aortas from MSG rats presented reduced tunica media thickness, with a lower aortic wall thickness/lumen diameter ratio and decreased total collagen content. Tau supplementation restored ACh-induced vasodilation and collagen content. CONCLUSIONS: Our study presents the first evidence that Tau prevents disruptions in vascular reactivity and in extracellular matrix composition in thoracic aortas of MSG-obese rats. The vascular protective actions of Tau may be linked to reduced lipid peroxidation and a reduction in cardiovascular risk factors, such as abdominal fat and hypertriglyceridemia.


Aorta, Thoracic/drug effects , Dietary Supplements , Endothelium, Vascular/drug effects , Hypothalamus/metabolism , Obesity/physiopathology , Taurine/pharmacology , Animals , Aorta, Thoracic/metabolism , Disease Models, Animal , Male , Rats , Rats, Wistar , Taurine/administration & dosage
18.
J Cell Physiol ; 234(5): 7019-7031, 2019 05.
Article En | MEDLINE | ID: mdl-30317580

Obesity predisposes to glucose intolerance and type 2 diabetes (T2D). This disease is often characterized by insulin resistance, changes in insulin clearance, and ß-cell dysfunction. However, studies indicate that, for T2D development, disruptions in glucagon physiology also occur. Herein, we investigated the involvement of glucagon in impaired glycemia control in monosodium glutamate (MSG)-obese mice. Male Swiss mice were subcutaneously injected daily, during the first 5 days after birth, with MSG (4 mg/g body weight [BW]) or saline (1.25 mg/g BW). At 90 days of age, MSG-obese mice were hyperglycemic, hyperinsulinemic, and hyperglucagonemic and had lost the capacity to increase their insulin/glucagon ratio when transitioning from the fasting to fed state, exacerbating hepatic glucose output. Furthermore, hepatic protein expressions of phosphorylated (p)-protein kinase A (PKA) and cAMP response element-binding protein (pCREB), and of phosphoenolpyruvate carboxykinase (PEPCK) enzyme were higher in fed MSG, before and after glucagon stimulation. Increased pPKA and phosphorylated hormone-sensitive lipase content were also observed in white fat of MSG. MSG islets hypersecreted glucagon in response to 11.1 and 0.5 mmol/L glucose, a phenomenon that persisted in the presence of insulin. Additionally, MSG α cells were hypertrophic displaying increased α-cell mass and immunoreactivity to phosphorylated mammalian target of rapamycin (pmTOR) protein. Therefore, severe glucose intolerance in MSG-obese mice was associated with increased hepatic glucose output, in association with hyperglucagonemia, caused by the refractory actions of glucose and insulin in α cells and via an effect that may be due to enhanced mTOR activation.


Blood Glucose/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/blood , Glucose Intolerance/blood , Insulin Resistance , Insulin/blood , Obesity/blood , Sodium Glutamate , Adipose Tissue, White/metabolism , Animals , Biomarkers/blood , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Glucose Intolerance/chemically induced , Glucose Intolerance/physiopathology , Liver/metabolism , Male , Mice , Obesity/chemically induced , Obesity/physiopathology , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Phosphorylation , TOR Serine-Threonine Kinases/metabolism
19.
J Cell Physiol ; 234(6): 9802-9809, 2019 06.
Article En | MEDLINE | ID: mdl-30370604

Human life expectancy is increasing faster lately and, consequently, the number of patients with age-related diseases such as type 2 diabetes (T2D) is rising every year. Cases of hyperinsulinemia have been extensively reported in elderly subjects and this alteration in blood insulin concentration is postulated to be a cause of insulin resistance, which in some cases triggers T2D onset. Thus, it is important to know the underlying mechanisms of age-dependent hyperinsulinemia to find new strategies to prevent T2D in elderly subjects. Two processes control blood insulin concentration: Insulin secretion by the endocrine portion of the pancreas and insulin clearance, which occurs mainly in the liver by the action of the insulin-degrading enzyme (IDE). Here, we demonstrated that 10-month-old mice (old) display increased body and fat pad weight, compared with 3-month-old mice (control), and these alterations were accompanied by glucose and insulin intolerance. We also confirm hyperinsulinemia in the old mice, which was related to increased insulin secretion but not to reduced insulin clearance. Although no changes in insulin clearance were observed, IDE activity was lower in the liver of old compared with the control mice. However, this decreased IDE activity was compensated by increased expression of IDE protein in the liver, thus explaining the similar insulin clearance observed in both groups. In conclusion, at the beginning of aging, 10-month-old mice do not display any alterations in insulin clearance. Therefore, hyperinsulinemia is initiated primarily due to a higher insulin secretion in the age-related metabolic dysfunction in mice.


Aging , Glucose/metabolism , Hyperinsulinism/etiology , Insulin/metabolism , Animals , Area Under Curve , Blood Glucose , Body Weight , Glucose/pharmacology , Homeostasis , Hyperinsulinism/metabolism , Insulin/blood , Insulysin , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL
20.
Amino Acids ; 50(11): 1511-1524, 2018 Nov.
Article En | MEDLINE | ID: mdl-30206707

The beneficial actions of L-taurine (Tau) against glucose intolerance, obesity, type 2 diabetes (T2D), and non-alcoholic fat liver disease (NAFLD) have been linked to its antioxidant and anti-inflammatory effects, which ameliorate tissue insulin sensitivity. Importantly, there are several lines of evidence that indicate a direct action of Tau on the endocrine pancreas to regulate the secretion and paracrine actions of insulin, glucagon, and somatostatin. Furthermore, Tau can also ameliorate glucose metabolism through the enhancement of insulin signaling. However, some of the benefits of Tau upon intermediary metabolism may manifest via considerable antagonism of the action of insulin. Therefore, this review discusses the mechanisms of action by which Tau may regulate endocrine pancreatic morphofunction, and glucose and lipid homeostasis.


Glucose/metabolism , Lipid Metabolism/physiology , Pancreas/metabolism , Taurine/metabolism , Animals , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Signal Transduction/physiology
...