Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Am J Physiol Endocrinol Metab ; 327(3): E279-E289, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39017679

ABSTRACT

Fibroblast growth factor 19 (FGF19) is a hormone synthesized in enterocytes in response to bile acids. This review explores the pivotal role of FGF19 in metabolism, addressing the urgent global health concern of obesity and its associated pathologies, notably type 2 diabetes. The intriguing inverse correlation between FGF19 and body mass or visceral adiposity, as well as its rapid increase following bariatric surgery, emphasizes its potential as a therapeutic target. This article meticulously examines the impact of FGF19 on metabolism by gathering evidence primarily derived from studies conducted in animal models or cell lines, using both FGF19 treatment and genetic modifications. Overall, these studies demonstrate that FGF19 has antidiabetic and antiobesogenic effects. A thorough examination across metabolic tissues, including the liver, adipose tissue, skeletal muscle, and the central nervous system, is conducted, unraveling the intricate interplay of FGF19 across diverse organs. Moreover, we provide a comprehensive overview of clinical trials involving an FGF19 analog called aldafermin, emphasizing promising results in diseases such as nonalcoholic steatohepatitis and diabetes. Therefore, we aim to foster a deeper understanding of FGF19 role and encourage further exploration of its clinical applications, thereby advancing the field and offering innovative approaches to address the escalating global health challenge of obesity and related metabolic conditions.


Subject(s)
Diabetes Mellitus, Type 2 , Fibroblast Growth Factors , Obesity , Fibroblast Growth Factors/metabolism , Humans , Animals , Obesity/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Clinical Trials as Topic , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use
2.
Mol Cell Endocrinol ; 578: 112074, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37742789

ABSTRACT

Chloroquine diphosphate (CQ), a weak base used to inhibit autophagic flux and treat malaria and rheumatoid diseases, has been shown, through unknown mechanisms, to improve glucose and lipid homeostasis in patients and rodents. We investigate herein the molecular mechanisms underlying these CQ beneficial metabolic actions in diet-induced obese mice. For this, C57BL6/J mice fed with either a chow or a high-fat diet (HFD) and uncoupling protein 1 (UCP-1) KO and adipocyte Atg7-deficient mice fed with a HFD were treated or not with CQ (60 mg/kg of body weight/day) during 8 weeks and evaluated for body weight, adiposity, glucose homeostasis and brown and white adipose tissues (BAT and WAT) UCP-1 content. CQ reduced body weight gain and adipose tissue and liver masses in mice fed with a HFD, without altering food intake, oxygen consumption, respiratory exchange ratio, spontaneous motor activity and feces caloric content. CQ attenuated the insulin intolerance, hyperglycemia, hyperinsulinemia, hypertriglyceridemia and hypercholesterolemia induced by HFD intake, such effects that were associated with increases in serum and liver fibroblast growth factor 21 (FGF-21) and BAT and WAT UCP-1 content. Interestingly, CQ beneficial metabolic actions of reducing body weight and adiposity and improving glucose homeostasis were preserved in HFD-fed UCP-1 KO and adipocyte Atg7 deficient mice. CQ reduces body weight gain and adiposity and improves glucose homeostasis in diet-induced obese mice through mechanisms that might involve FGF-21, but not UCP1-mediated nonshivering thermogenesis or inhibition of adipocyte autophagy.

3.
Med Sci Educ ; 33(4): 913-924, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37546198

ABSTRACT

The mandatory isolation caused by COVID-19 required the adoption of emergency remote teaching, which caused difficulties for instructors, especially for those who use active learning that depends on student participation in class. This study aimed to investigate the ability of instructors to apply active learning effectively given the pandemic context. This was a cross-sectional observational study carried out in an undergraduate medical school. The sample was composed from one to three classes of 28 instructors that were observed synchronously. Each class was analyzed using a form created from an adaptation of the PORTAAL tool, aiming to evaluate quantitatively essential elements for active learning. We observed that the mean times devoted to activities and active participation of students were 54.8% and 33.1% of the total class time, respectively. Among the time spent in student interactions, the intra-group demanded the highest percentage of the class time. Additionally, 22.0% of the activities presented a high level in Bloom's taxonomy and there was a positive correlation between the percentage of activities at higher Bloom levels and the percentage of class time with student participation, intra-group or between-group interactions, supporting the use of higher-order cognitive skills in a collaborative and student-centered context. In conclusion, our findings indicate that some instructors were able to apply essential elements for an active and collaborative learning even during the emergency remote teaching.

4.
Hormones (Athens) ; 21(2): 195-208, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35247188

ABSTRACT

Obesity is a chronic condition of multifactorial etiology characterized by excessive body fat due to a calorie intake higher than energy expenditure. Given the intrinsic limitations of surgical interventions and the difficulties associated with lifestyle changes, pharmacological manipulation is currently one of the main therapies for metabolic diseases. Approaches aiming to promote energy expenditure through induction of thermogenesis have been explored and, in this context, brown adipose tissue (BAT) activation and browning have been shown to be promising strategies. Although such processes are physiologically stimulated by the sympathetic nervous system, not all situations that are known to increase adrenergic signaling promote a concomitant increase in BAT activation or browning in humans. Thus, a better understanding of factors involved in the thermogenesis attributed to these tissues is needed to enable the development of future therapies against obesity. Herein we carry out a critical review of original articles in humans under conditions previously known to trigger adrenergic responses-namely, cold, catecholamine-secreting tumor (pheochromocytoma and paraganglioma), burn injury, and adrenergic agonists-and discuss which of them are associated with increased BAT activation and browning. BAT is clearly stimulated in individuals exposed to cold or treated with high doses of the ß3-adrenergic agonist mirabegron, whereas browning is certainly induced in patients after burn injury or with pheochromocytoma, as well as in individuals treated with ß3-adrenergic agonist mirabegron for at least 10 weeks. Given the potential effect of increasing energy expenditure, adrenergic stimuli are promising strategies in the treatment of metabolic diseases.


Subject(s)
Adrenal Gland Neoplasms , Pheochromocytoma , Adipose Tissue, Brown/metabolism , Adrenal Gland Neoplasms/pathology , Adrenergic Agents/metabolism , Adrenergic Agonists/metabolism , Energy Metabolism , Humans , Nervous System/metabolism , Nervous System/pathology , Obesity/metabolism , Pheochromocytoma/pathology
5.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299197

ABSTRACT

In recent years, accumulating evidence has shown that the innate immune complement system is involved in several aspects of normal brain development and in neurodevelopmental disorders, including autism spectrum disorder (ASD). Although abnormal expression of complement components was observed in post-mortem brain samples from individuals with ASD, little is known about the expression patterns of complement molecules in distinct cell types in the developing autistic brain. In the present study, we characterized the mRNA and protein expression profiles of a wide range of complement system components, receptors and regulators in induced pluripotent stem cell (iPSC)-derived neural progenitor cells, neurons and astrocytes of individuals with ASD and neurotypical controls, which constitute in vitro cellular models that recapitulate certain features of both human brain development and ASD pathophysiology. We observed that all the analyzed cell lines constitutively express several key complement molecules. Interestingly, using different quantification strategies, we found that complement C4 mRNA and protein are expressed in significantly lower levels by astrocytes derived from ASD individuals compared to control astrocytes. As astrocytes participate in synapse elimination, and diminished C4 levels have been linked to defective synaptic pruning, our findings may contribute to an increased understanding of the atypically enhanced brain connectivity in ASD.


Subject(s)
Astrocytes/pathology , Autism Spectrum Disorder/pathology , Complement C4/metabolism , Induced Pluripotent Stem Cells/pathology , Neural Stem Cells/pathology , Neurons/pathology , Astrocytes/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Cells, Cultured , Complement C4/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism
6.
Article in English | MEDLINE | ID: mdl-34004356

ABSTRACT

The nutrient sensors peroxisome proliferator-activated receptor γ (PPARγ) and mechanistic target of rapamycin complex 1 (mTORC1) closely interact in the regulation of adipocyte lipid storage. The precise mechanisms underlying this interaction and whether this extends to other metabolic processes and the endocrine function of adipocytes are still unknown. We investigated herein the involvement of mTORC1 as a mediator of the actions of the PPARγ ligand rosiglitazone in subcutaneous inguinal white adipose tissue (iWAT) mass, endocrine function, lipidome, transcriptome and branched-chain amino acid (BCAA) metabolism. Mice bearing regulatory associated protein of mTOR (Raptor) deletion and therefore mTORC1 deficiency exclusively in adipocytes and littermate controls were fed a high-fat diet supplemented or not with the PPARγ agonist rosiglitazone (30 mg/kg/day) for 8 weeks and evaluated for iWAT mass, lipidome, transcriptome (Rnaseq), respiration and BCAA metabolism. Adipocyte mTORC1 deficiency not only impaired iWAT adiponectin transcription, synthesis and secretion, PEPCK mRNA levels, triacylglycerol synthesis and BCAA oxidation and mRNA levels of related proteins but also completely blocked the upregulation in these processes induced by pharmacological PPARγ activation with rosiglitazone. Mechanistically, adipocyte mTORC1 deficiency impairs PPARγ transcriptional activity by reducing PPARγ protein content, as well as by downregulating C/EBPα, a co-partner and facilitator of PPARγ. In conclusion, mTORC1 and PPARγ are essential partners involved in the regulation of subcutaneous adipose tissue adiponectin production and secretion and BCAA oxidative metabolism.


Subject(s)
Adiponectin/metabolism , Amino Acids, Branched-Chain/metabolism , Glycerol/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , PPAR gamma/metabolism , Subcutaneous Fat/metabolism , Up-Regulation , Animals , Mice , Oxidation-Reduction
7.
Front Neurosci ; 14: 23, 2020.
Article in English | MEDLINE | ID: mdl-32116493

ABSTRACT

Current evidence indicates that certain immune molecules such as components of the complement system are directly involved in neurobiological processes related to brain development, including neurogenesis, neuronal migration, synaptic remodeling, and response to prenatal or early postnatal brain insults. Consequently, complement system dysfunction has been increasingly implicated in disorders of neurodevelopmental origin, such as schizophrenia, autism spectrum disorder (ASD) and Rett syndrome. However, the mechanistic evidence for a causal relationship between impaired complement regulation and these disorders varies depending on the disease involved. Also, it is still unclear to what extent altered complement expression plays a role in these disorders through inflammation-independent or -dependent mechanisms. Furthermore, pathogenic mutations in specific complement components have been implicated in the etiology of 3MC syndrome, a rare autosomal recessive developmental disorder. The aims of this review are to discuss the current knowledge on the roles of the complement system in sculpting brain architecture and function during normal development as well as after specific inflammatory insults, such as maternal immune activation (MIA) during pregnancy, and to evaluate the existing evidence associating aberrant complement with developmental brain disorders.

8.
Mol Nutr Food Res ; 62(17): e1800283, 2018 09.
Article in English | MEDLINE | ID: mdl-30001482

ABSTRACT

SCOPE: To test whether myeloid cells Tsc1 deletion and therefore constitutive activation of the nutrient sensor mTORC1 protects from high-fat diet (HFD)-induced obesity, glucose intolerance, and adipose tissue inflammation. METHODS AND RESULTS: Mice with Tsc1 deletion in myeloid cells (MTsc1KO) and littermate controls (MTsc1WT) were fed with HFD for 8 weeks and evaluated for body weight, glucose homeostasis, and adipose tissue inflammation. MTsc1KO mice were protected from HFD-induced obesity and glucose intolerance. MTsc1KO, however, displayed, independently of the diet, abnormal behavior, episodes of intense movement, and muscle spasms followed by temporary paralysis. To investigate whether obesity protection was due to myeloid cells Tsc1 deletion, bone marrow was transplanted from MTsc1WT and MTsc1KO into irradiated C57BL6/J mice. Mice transplanted with MTsc1KO bone marrow displayed reduced body weight gain, adiposity, and inflammation, and enhanced energy expenditure, glucose tolerance and adipose tissue M2 macrophage content upon HFD feeding, in the absence of abnormal behavior. In vitro, Tsc1 deletion increased in a mTORC1-dependent manner macrophage polarization to M2 profile and mRNA levels of fatty acid binding protein 4 and PPARγ. CONCLUSION: Constitutive mTORC1 activation in myeloid cells protects mice from HFD-induced obesity, adipose tissue inflammation, and glucose intolerance by promoting macrophage polarization to M2 pro-resolution profile and increasing energy expenditure.


Subject(s)
Diet, High-Fat/adverse effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Myeloid Cells/metabolism , Obesity/etiology , Tuberous Sclerosis Complex 1 Protein/genetics , Adipose Tissue/pathology , Adipose Tissue/physiology , Animals , Cytokines/metabolism , Gene Expression Regulation , Macrophages/pathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Panniculitis/metabolism , Panniculitis/pathology , Tuberous Sclerosis Complex 1 Protein/metabolism , Weight Gain
9.
Hum Mutat ; 39(10): 1372-1383, 2018 10.
Article in English | MEDLINE | ID: mdl-29969175

ABSTRACT

The Reelin-DAB1 signaling pathway plays a crucial role in regulating neuronal migration and synapse function. Although many rare heterozygous variants in the Reelin gene (RELN) have been identified in patients with autism spectrum disorder (ASD), most variants are still of unknown clinical significance. Also, genetic data suggest that heterozygous variants in RELN alone appear to be insufficient to cause ASD. Here, we describe the identification and functional characterization of rare compound heterozygous missense variants in RELN in a patient with ASD in whom we have previously reported hyperfunctional mTORC1 signaling of yet unknown etiology. Using iPSC-derived neural progenitor cells (NPCs) from this patient, we provide experimental evidence that the identified variants are deleterious and lead to diminished Reelin secretion and impaired Reelin-DAB1 signal transduction. Also, our results suggest that mTORC1 pathway overactivation may function as a second hit event contributing to downregulation of the Reelin-DAB1 cascade in patient-derived NPCs, and that inhibition of mTORC1 by rapamycin attenuates Reelin-DAB1 signaling impairment. Taken together, our findings point to an abnormal interplay between Reelin-DAB1 and mTORC1 networks in nonsyndromic ASD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Genetic Variation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/chemistry , Alleles , Autism Spectrum Disorder/diagnosis , Biomarkers , Case-Control Studies , Cell Adhesion Molecules, Neuronal/chemistry , Child , Child, Preschool , Extracellular Matrix Proteins/chemistry , Female , Gene Expression , Heterozygote , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Models, Molecular , Nerve Tissue Proteins/chemistry , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Protein Conformation , Proto-Oncogene Proteins c-akt/metabolism , Reelin Protein , Serine Endopeptidases/chemistry , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism
10.
Einstein (Säo Paulo) ; 15(4): 507-511, Oct.-Dec. 2017. graf
Article in English | LILACS | ID: biblio-891425

ABSTRACT

ABSTRACT Obesity is characterized by an excessive increase in the adipose tissue mass, and is associated with higher incidence of several chronic metabolic diseases, such as type 2 diabetes. Therefore, its increasing prevalence is a public health concern, and it is important to better understand its etiology to develop new therapeutic strategies. Evidence accumulated over the years indicates that obesity is associated with a marked activation in adipose tissue of the mechanistic target of rapamycin complex 1 (mTORC1), a signaling pathway that controls lipid metabolism, and adipocyte formation and maintenance. Curiously, mTORC1 is also involved in the control of nonshivering thermogenesis and recruitment as well as browning of white adipose tissue. In this review, we explored mTORC1 functions in adipocytes and presented evidence, suggesting that mTORC1 may either increase or reduce adiposity, depending on the conditions and activation levels.


RESUMO A obesidade é caracterizada pelo aumento excessivo da massa de tecido adiposo, estando associada à maior incidência de diversas doenças metabólicas crônicas, como diabetes tipo 2. Sua crescente prevalência é uma questão de saúde pública, e faz-se importante compreender melhor sua etiologia, para desenvolver novas estratégias terapêuticas. As evidências acumuladas por muitos anos indicam que a obesidade está associada à significativa ativação no tecido adiposo do complexo 1 da proteína alvo mecanístico da rapamicina (mTORC1), uma via de sinalização que regula o metabolismo de lipídeos, bem como a formação e manutenção de adipócitos. Curiosamente, mTORC1 também está envolvido no controle da termogênese, independente do tremor muscular, e no recrutamento e browning de tecido adiposo branco. Nesta revisão, exploramos as diferentes funções do mTORC1 em adipócitos e apresentamos evidências que sugerem que o mTORC1 pode aumentar ou reduzir a adiposidade, dependendo das condições e de seu nível de ativação.


Subject(s)
Humans , Animals , Adiposity/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Obesity/metabolism , Adipose Tissue, Brown/metabolism , Adipocytes/metabolism , Thermogenesis/physiology , Diabetes Mellitus, Type 2/metabolism , Lipid Metabolism/physiology , Adipose Tissue, White/metabolism
11.
J Lipid Res ; 58(9): 1797-1807, 2017 09.
Article in English | MEDLINE | ID: mdl-28679588

ABSTRACT

Mechanistic target of rapamycin complex (mTORC)1 activity is increased in adipose tissue of obese insulin-resistant mice, but its role in the regulation of tissue inflammation is unknown. Herein, we investigated the effects of adipocyte mTORC1 deficiency on adipose tissue inflammation and glucose homeostasis. For this, mice with adipocyte raptor deletion and controls fed a chow or a high-fat diet were evaluated for body mass, adiposity, glucose homeostasis, and adipose tissue inflammation. Despite reducing adiposity, adipocyte mTORC1 deficiency promoted hepatic steatosis, insulin resistance, and adipose tissue inflammation (increased infiltration of macrophages, neutrophils, and B lymphocytes; crown-like structure density; TNF-α, interleukin (IL)-6, and monocyte chemoattractant protein 1 expression; IL-1ß protein content; lipid peroxidation; and de novo ceramide synthesis). The anti-oxidant, N-acetylcysteine, partially attenuated, whereas treatment with de novo ceramide synthesis inhibitor, myriocin, completely blocked adipose tissue inflammation and nucleotide oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3)-inflammasome activation, but not hepatic steatosis and insulin resistance induced by adipocyte raptor deletion. Rosiglitazone treatment, however, completely abrogated insulin resistance induced by adipocyte raptor deletion. In conclusion, adipocyte mTORC1 deficiency induces adipose tissue inflammation and NLRP3-inflammasome activation by promoting oxidative stress and de novo ceramide synthesis. Such adipose tissue inflammation, however, is not an underlying cause of the insulin resistance displayed by these mice.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/pathology , Ceramides/biosynthesis , Inflammasomes/metabolism , Mechanistic Target of Rapamycin Complex 1/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Adipocytes/drug effects , Adipocytes/pathology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Diet, High-Fat/adverse effects , Glucose/metabolism , Homeostasis/drug effects , Mechanistic Target of Rapamycin Complex 2/deficiency , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects
13.
Int J Mol Sci ; 18(3)2017 Mar 18.
Article in English | MEDLINE | ID: mdl-28335463

ABSTRACT

Whereas autism spectrum disorder (ASD) exhibits striking heterogeneity in genetics and clinical presentation, dysfunction of mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway has been identified as a molecular feature common to several well-characterized syndromes with high prevalence of ASD. Additionally, recent findings have also implicated mTORC1 signaling abnormalities in a subset of nonsyndromic ASD, suggesting that defective mTORC1 pathway may be a potential converging mechanism in ASD pathology across different etiologies. However, the mechanistic evidence for a causal link between aberrant mTORC1 pathway activity and ASD neurobehavioral features varies depending on the ASD form involved. In this review, we first discuss six monogenic ASD-related syndromes, including both classical and potentially novel mTORopathies, highlighting their contribution to our understanding of the neurobiological mechanisms underlying ASD, and then we discuss existing evidence suggesting that aberrant mTORC1 signaling may also play a role in nonsyndromic ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Multiprotein Complexes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , TOR Serine-Threonine Kinases/genetics
14.
Immunobiology ; 222(2): 261-271, 2017 02.
Article in English | MEDLINE | ID: mdl-27692982

ABSTRACT

Genetic- and diet-induced obesity and insulin resistance are associated with an increase in mechanistic target of rapamycin complex (mTORC) 1 activity in adipose tissue. We investigated herein the effects of pharmacological mTORC1 inhibition in the development of adipose tissue inflammation induced by high-fat diet (HFD) feeding, as well as in the polarization, metabolism and function of bone marrow-derived macrophages (BMDM). For this, C57BL/6J mice fed with a standard chow diet or a HFD (60% of calories from fat) and treated with either vehicle (0.1% Me2SO, 0.2% methylcellulose) or rapamycin (2mg/kg/ day, gavage) during 30days were evaluated for body weight, adiposity, glucose tolerance and adipose tissue inflammation. Although rapamycin did not affect the increase in body weight and adiposity, it exacerbated the glucose intolerance and adipose tissue inflammation induced by HFD feeding, as evidenced by the increased adipose tissue percentage of M1 macrophages, naive and activated cytotoxic T lymphocytes, and mRNA levels of proinflammatory molecules, such as TNF-α, IL-6 and MCP-1. In BMDM in vitro, pharmacological mTORC1 inhibition induced phosphorylation of NFκB p65 and spontaneous polarization of macrophages to a proinflammatory M1 profile, while it impaired M2 polarization induced by IL-4+IL-13, glycolysis and phagocytosis. Altogether, these findings indicate that mTORC1 activity is an important determinant of adipose tissue inflammatory profile and macrophage plasticity, metabolism and function.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Obesity/immunology , Obesity/metabolism , Panniculitis/immunology , Panniculitis/metabolism , Animals , Biomarkers , Cytokines/metabolism , Glucose/metabolism , Immunophenotyping , Inflammation Mediators/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Macrophages/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/pathology , Panniculitis/pathology , Phenotype , Sirolimus/pharmacology
15.
Einstein (Sao Paulo) ; 15(4): 507-511, 2017.
Article in English, Portuguese | MEDLINE | ID: mdl-29364369

ABSTRACT

Obesity is characterized by an excessive increase in the adipose tissue mass, and is associated with higher incidence of several chronic metabolic diseases, such as type 2 diabetes. Therefore, its increasing prevalence is a public health concern, and it is important to better understand its etiology to develop new therapeutic strategies. Evidence accumulated over the years indicates that obesity is associated with a marked activation in adipose tissue of the mechanistic target of rapamycin complex 1 (mTORC1), a signaling pathway that controls lipid metabolism, and adipocyte formation and maintenance. Curiously, mTORC1 is also involved in the control of nonshivering thermogenesis and recruitment as well as browning of white adipose tissue. In this review, we explored mTORC1 functions in adipocytes and presented evidence, suggesting that mTORC1 may either increase or reduce adiposity, depending on the conditions and activation levels.


Subject(s)
Adipocytes/metabolism , Adiposity/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Obesity/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Lipid Metabolism/physiology , Thermogenesis/physiology
16.
PLoS One ; 11(10): e0165115, 2016.
Article in English | MEDLINE | ID: mdl-27764229

ABSTRACT

INTRODUCTION: Impaired wound healing has been widely reported in diabetes. Linoleic acid (LA) accelerates the skin wound healing process in non-diabetic rats. However, LA has not been tested in diabetic animals. OBJECTIVES: We investigated whether oral administration of pure LA improves wound healing in streptozotocin-induced diabetic rats. METHODS: Dorsal wounds were induced in streptozotocin-induced type-1 diabetic rats treated or not with LA (0.22 g/kg b.w.) for 10 days. Wound closure was daily assessed for two weeks. Wound tissues were collected at specific time-points and used to measure fatty acid composition, and contents of cytokines, growth factors and eicosanoids. Histological and qPCR analyses were employed to examine the dynamics of cell migration during the healing process. RESULTS: LA reduced the wound area 14 days after wound induction. LA also increased the concentrations of cytokine-induced neutrophil chemotaxis (CINC-2αß), tumor necrosis factor-α (TNF-α) and leukotriene B4 (LTB4), and reduced the expression of macrophage chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1 (MIP-1). These results together with the histological analysis, which showed accumulation of leukocytes in the wound early in the healing process, indicate that LA brought forward the inflammatory phase and improved wound healing in diabetic rats. Angiogenesis was induced by LA through elevation in tissue content of key mediators of this process: vascular-endothelial growth factor (VEGF) and angiopoietin-2 (ANGPT-2). CONCLUSIONS: Oral administration of LA hastened wound closure in diabetic rats by improving the inflammatory phase and angiogenesis.


Subject(s)
Diabetes Mellitus, Experimental/complications , Linoleic Acid/administration & dosage , Neovascularization, Physiologic/drug effects , Wound Healing/drug effects , Administration, Oral , Angiopoietin-2/metabolism , Animals , Cell Movement/drug effects , Cytokines/metabolism , Gene Expression Regulation/drug effects , Linoleic Acid/pharmacology , Rats , Streptozocin , Vascular Endothelial Growth Factor A/metabolism
17.
Biochim Biophys Acta ; 1861(5): 430-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26923434

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1) loss of function reduces adiposity whereas partial mTORC1 inhibition enhances fat deposition. Herein we evaluated how constitutive mTORC1 activation in adipocytes modulates adiposity in vivo. Mice with constitutive mTORC1 activation in adipocytes induced by tuberous sclerosis complex (Tsc)1 deletion and littermate controls were evaluated for body mass, energy expenditure, glucose and fatty acid metabolism, mitochondrial function, mRNA and protein contents. Adipocyte-specific Tsc1 deletion reduced visceral, but not subcutaneous, fat mass, as well as adipocyte number and diameter, phenotypes that were associated with increased lipolysis, UCP-1 content (browning) and mRNA levels of pro-browning transcriptional factors C/EBPß and ERRα. Adipocyte Tsc1 deletion enhanced mitochondrial oxidative activity, fatty acid oxidation and the expression of PGC-1α and PPARα in both visceral and subcutaneous fat. In brown adipocytes, however, Tsc1 deletion did not affect UCP-1 content and basal respiration. Adipocyte Tsc1 deletion also reduced visceral adiposity and enhanced glucose tolerance, liver and muscle insulin signaling and adiponectin secretion in mice fed with purified low- or high-fat diet. In conclusion, adipocyte-specific Tsc1 deletion enhances mitochondrial activity, induces browning and reduces visceral adiposity in mice.


Subject(s)
Adipocytes, Brown/enzymology , Adipocytes, White/enzymology , Adipose Tissue, Brown/enzymology , Adiposity , Intra-Abdominal Fat/enzymology , Mitochondria/enzymology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adipocytes, Brown/ultrastructure , Adipocytes, White/ultrastructure , Adiponectin/deficiency , Adiponectin/genetics , Adipose Tissue, Brown/ultrastructure , Adiposity/genetics , Animals , Cell Respiration , Diet, Fat-Restricted , Diet, High-Fat , Energy Metabolism , Enzyme Activation , Gene Expression Regulation , Genotype , Glucose/metabolism , Insulin/metabolism , Intra-Abdominal Fat/ultrastructure , Lipolysis , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Oxidation-Reduction , Phenotype , Signal Transduction , Time Factors , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
18.
Mol Nutr Food Res ; 59(5): 957-67, 2015 May.
Article in English | MEDLINE | ID: mdl-25641959

ABSTRACT

SCOPE: We tested herein the hypothesis that peroxisome proliferator activated receptor γ (PPARγ) is a major mediator of omega-3 (n-3) protective actions against high-fat diet (HFD) induced obesity, glucose intolerance, and adipose tissue inflammation. METHODS AND RESULTS: C57BL6 wild-type and fat-1 transgenic (fat-1) mice were fed a low-fat diet (LFD) or HFD, treated or not with PPARγ antagonist, and evaluated for energy balance, adiposity, glucose tolerance, and adipose tissue inflammation. Fat-1 mice were protected from obesity, fasting hyperglycemia, glucose intolerance, and adipose tissue inflammation. PPARγ inhibition completely abolished fat-1 protection against HFD-induced glucose intolerance, but not obesity or adipose tissue inflammation. To investigate the role of myeloid cell as mediator of n-3 beneficial metabolic actions, mice with deletion (LyzM-PPARγ(KO)) or nondeletion (LyzM-PPARγ(WT)) of PPARγ in myeloid cells were fed either LFD or HFD (lard) or an HFD rich in n-3 (fish oil). Our findings indicate that myeloid cell associated PPARγ is not involved in the attenuation of HFD-induced glucose intolerance and adipose tissue inflammation induced by n-3. CONCLUSION: High endogenous n-3 fatty acid levels protect from HFD obesity, glucose intolerance, and adipose tissue inflammation. Among these, only protection against glucose intolerance is mediated by non-myeloid cell PPARγ.


Subject(s)
Adipose Tissue/pathology , Blood Glucose/analysis , Fatty Acids, Omega-3/administration & dosage , Obesity/prevention & control , PPAR gamma/physiology , Animals , Diet, High-Fat , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL
19.
Am J Physiol Endocrinol Metab ; 306(9): E1046-54, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24619883

ABSTRACT

mTOR inhibition with rapamycin induces a diabetes-like syndrome characterized by severe glucose intolerance, hyperinsulinemia, and hypertriglyceridemia, which is due to increased hepatic glucose production as well as reduced skeletal muscle glucose uptake and adipose tissue PPARγ activity. Herein, we tested the hypothesis that pharmacological PPARγ activation attenuates the diabetes-like syndrome associated with chronic mTOR inhibition. Rats treated with the mTOR inhibitor rapamycin (2 mg·kg(-1)·day(-1)) in combination or not with the PPARγ ligand rosiglitazone (15 mg·kg(-1)·day(-1)) for 15 days were evaluated for insulin secretion, glucose, insulin, and pyruvate tolerance, skeletal muscle and adipose tissue glucose uptake, and insulin signaling. Rosiglitazone corrected fasting hyperglycemia, attenuated the glucose and insulin intolerances, and abolished the increase in fasting plasma insulin and C-peptide levels induced by rapamycin. Surprisingly, rosiglitazone markedly increased the plasma insulin and C-peptide responses to refeeding in rapamycin-treated rats. Furthermore, rosiglitazone partially attenuated rapamycin-induced gluconeogenesis, as evidenced by the improved pyruvate tolerance and reduced mRNA levels of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase. Rosiglitazone also restored insulin's ability to stimulate glucose uptake and its incorporation into glycogen in skeletal muscle of rapamycin-treated rats, which was associated with normalization of Akt Ser(473) phosphorylation. However, the rapamycin-mediated impairments of adipose tissue glucose uptake and incorporation into triacylglycerol were unaffected by rosiglitazone. Our findings indicate that PPARγ activation ameliorates some of the disturbances in glucose homeostasis and insulin action associated with chronic rapamycin treatment by reducing gluconeogenesis and insulin secretion and restoring muscle insulin signaling and glucose uptake.


Subject(s)
Glucose Intolerance/prevention & control , PPAR gamma/agonists , Sirolimus/adverse effects , Thiazolidinediones/pharmacology , Animals , Cells, Cultured , Drug Antagonism , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Muscle, Skeletal/metabolism , PPAR gamma/metabolism , Rats , Rats, Sprague-Dawley , Rosiglitazone , TOR Serine-Threonine Kinases/antagonists & inhibitors
20.
Am J Physiol Regul Integr Comp Physiol ; 303(12): R1277-85, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23100029

ABSTRACT

Here, we investigated whether pharmacological PPARγ activation modulates key early events in brown adipose tissue (BAT) recruitment induced by acute cold exposure with the aim of unraveling the interrelationships between sympathetic and PPARγ signaling. Sprague-Dawley rats treated or not with the PPARγ ligand rosiglitazone (15 mg·kg(-1)·day(-1), 7 days) were kept at 23°C or exposed to cold (5°C) for 24 h and evaluated for BAT gene expression, sympathetic activity, thyroid status, and adrenergic signaling. Rosiglitazone did not affect the reduction in body weight gain and the increase in feed efficiency, Vo(2), and BAT sympathetic activity induced by 24-h cold exposure. Rosiglitazone strongly attenuated the increase in serum total and free T4 and T3 levels and BAT iodothyronine deiodinase type 2 (D2) and PGC-1α mRNA levels and potentiated the reduction in BAT thyroid hormone receptor (THR) ß mRNA levels induced by cold. Administration of T3 to rosiglitazone-treated rats exacerbated the cold-induced increase in energy expenditure but did not restore a proper activation of D2 and PGC-1α, nor further increased uncoupling protein 1 expression. Regarding adrenergic signaling, rosiglitazone did not affect the changes in BAT cAMP content and PKA activity induced by cold. Rosiglitazone alone or in combination with cold increased CREB binding to DNA, but it markedly reduced the expression of one of its major coactivators, CREB binding protein. In conclusion, pharmacological PPARγ activation impairs short-term cold elicitation of BAT adrenergic and thyroid signaling, which may result in abnormal tissue recruitment and thermogenic activity.


Subject(s)
Adipose Tissue, Brown/metabolism , Cold Temperature , Iodide Peroxidase/metabolism , PPAR gamma/metabolism , RNA-Binding Proteins/metabolism , Thyroid Gland/physiology , Transcription Factors/metabolism , Up-Regulation/physiology , Animals , Male , Models, Animal , PPAR gamma/agonists , PPAR gamma/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Sprague-Dawley , Rosiglitazone , Signal Transduction/drug effects , Signal Transduction/physiology , Thiazolidinediones/pharmacology , Thyroxine/blood , Time Factors , Triiodothyronine/blood , Iodothyronine Deiodinase Type II
SELECTION OF CITATIONS
SEARCH DETAIL