Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(48): 30335-30343, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33199621

RESUMO

Implementation of regular physical activity helps in the maintenance of a healthy metabolic profile both in humans and mice through molecular mechanisms not yet completely defined. Here, we show that high-intensity interval training (HIIT) modifies the microRNA (miRNA) profile of circulating exosomes in mice, including significant increases in miR-133a and miR-133b Importantly, treatment of sedentary mice with exosomes isolated from the plasma of trained mice improves glucose tolerance, insulin sensitivity, and decreases plasma levels of triglycerides. Moreover, exosomes isolated from the muscle of trained mice display similar changes in miRNA content, and their administration to sedentary mice reproduces the improvement of glucose tolerance. Exosomal miRNAs up-regulated by HIIT target insulin-regulated transcription factor forkhead box O1 (FoxO1) and, accordingly, expression of FoxO1 is decreased in the liver of trained and exosome-treated mice. Treatment with exosomes transfected with a miR-133b mimic or with a specific siRNA targeting FoxO1 recapitulates the metabolic effects observed in trained mice. Overall, our data suggest that circulating exosomes released by the muscle carry a specific miRNA signature that is modified by exercise and induce expression changes in the liver that impact whole-body metabolic profile.


Assuntos
Regulação para Baixo/genética , Exossomos/metabolismo , Proteína Forkhead Box O1/genética , Treinamento Intervalado de Alta Intensidade , Resistência à Insulina , Fígado/metabolismo , MicroRNAs/metabolismo , Músculos/metabolismo , Animais , Exossomos/ultraestrutura , Proteína Forkhead Box O1/metabolismo , Gluconeogênese , Glucose/metabolismo , Metabolismo dos Lipídeos , Masculino , Metabolômica , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Condicionamento Físico Animal
2.
Int J Mol Sci ; 24(3)2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36768391

RESUMO

Obesity and type 2 diabetes are associated with defects of insulin action in different tissues or alterations in ß-cell secretory capacity that may be triggered by environmental challenges, inadequate lifestyle choices, or an underlying genetic predisposition. In addition, recent data shows that obesity may also be caused by perturbations of the gut microbiota, which then affect metabolic function and energy homeostasis in the host. Maintenance of metabolic homeostasis in complex organisms such as mammals requires organismal-level communication, including between the different organs and the gut microbiota. Extracellular vesicles (EVs) have been identified in all domains of life and have emerged as crucial players in inter-organ and inter-kingdom crosstalk. Interestingly, EVs found in edible vegetables or in milk have been shown to influence gut microbiota or tissue function in mammals. Moreover, there is a multidirectional crosstalk mediated by EVs derived from gut microbiota and body organs that has implications for host health. Untangling this complex signaling network may help implement novel therapies for the treatment of metabolic disease.


Assuntos
Diabetes Mellitus Tipo 2 , Vesículas Extracelulares , Animais , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Obesidade/metabolismo , Vesículas Extracelulares/metabolismo , Mamíferos , Comunicação
3.
Int J Mol Sci ; 23(23)2022 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-36499248

RESUMO

Most cells release extracellular vesicles (EVs) that can be detected circulating in blood. We and others have shown that the microRNA contents of these vesicles induce transcriptomic changes in acceptor cells, contributing to the adjustment of metabolic homeostasis in response to environmental demands. Here, we explore the potential for modulating obesity- and exercise-derived EV-microRNAs to treat the metabolic dysfunction associated with obesity in mice. Treatment with EV-miRNAs alleviated glucose intolerance and insulin resistance in obese mice to an extent similar to that of high-intensity interval training, although only exercise improved cardiorespiratory fitness and decreased body weight. Mechanistically, EV-miRNAs decreased fatty acid and cholesterol biosynthesis pathways in the liver, reducing hepatic steatosis and increasing insulin sensitivity, resulting in decreased glycemia and triglyceridemia. Our data suggest that manipulation of EV-miRNAs may be a viable strategy to alleviate metabolic dysfunction in obese and diabetic patients who are unable to exercise, although actual physical activity is needed to improve cardiorespiratory fitness.


Assuntos
Vesículas Extracelulares , Intolerância à Glucose , Resistência à Insulina , MicroRNAs , Camundongos , Animais , MicroRNAs/metabolismo , Obesidade/complicações , Obesidade/terapia , Vesículas Extracelulares/metabolismo , Intolerância à Glucose/metabolismo
4.
Proc Natl Acad Sci U S A ; 115(48): 12158-12163, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30429322

RESUMO

Obesity is frequently associated with metabolic disease. Here, we show that obesity changes the miRNA profile of plasma exosomes in mice, including increases in miR-122, miR-192, miR-27a-3p, and miR-27b-3p Importantly, treatment of lean mice with exosomes isolated from obese mice induces glucose intolerance and insulin resistance. Moreover, administration of control exosomes transfected with obesity-associated miRNA mimics strongly induces glucose intolerance in lean mice and results in central obesity and hepatic steatosis. Expression of the candidate target gene Ppara is decreased in white adipose tissue but not in the liver of mimic-treated (MIMIC) mice, and this is accompanied by increased circulating free fatty acids and hypertriglyceridemia. Treatment with a specific siRNA targeting Ppara transfected into exosomes recapitulates the phenotype induced by obesity-associated miRNAs. Importantly, simultaneously reducing free fatty acid plasma levels in MIMIC mice with either the lipolysis inhibitor acipimox or the PPARα agonist fenofibrate partially protects against these metabolic alterations. Overall, our data highlight the central role of obesity-associated exosomal miRNAs in the etiopathogeny of glucose intolerance and dyslipidemia.


Assuntos
Exossomos/genética , Glucose/metabolismo , Metabolismo dos Lipídeos , MicroRNAs/genética , Obesidade/genética , Obesidade/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Exossomos/metabolismo , Ácidos Graxos não Esterificados/sangue , Intolerância à Glucose , Humanos , Insulina/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , MicroRNAs/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo
5.
Diabetes Metab Res Rev ; 35(3): e3107, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30513130

RESUMO

Diabetes is a group of metabolic diseases characterized by elevated blood glucose levels that drive the development of life-threatening complications. Diabetes results from a situation of insufficient insulin action, either by deficient production of the hormone by the pancreas, or by the development of insulin resistance in peripheral tissues such as liver, muscle, or the adipose depots. Communication between organs is thus central to the maintenance of glucose homoeostasis. Recently, several studies are evidencing that small vesicles called exosomes released by, amongst other, the adipose tissue can regulate gene expression in other tissues, hence modulating interorgan crosstalk. Therefore, exosomes participate in the development of diabetes and its associated complications. Their study holds the potential of providing us with novel biomarkers for the early diagnosis and stratification of patients at risk of developing diabetes, hence allowing the timely implementation of more personalized therapies. On the other hand, the molecular dissection of the pathways initiated by exosomes under situations of metabolic stress could help to gain a deeper knowledge of the pathophysiology of diabetes and its associated metabolic diseases.


Assuntos
Biomarcadores/metabolismo , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/terapia , Exossomos/metabolismo , Animais , Diabetes Mellitus/metabolismo , Humanos
6.
FASEB J ; 31(12): 5296-5306, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28821639

RESUMO

Human islet amyloid polypeptide (hIAPP) aggregation is associated with ß-cell dysfunction and death in type 2 diabetes (T2D). we aimed to determine whether in vivo treatment with chemical chaperone 4-phenylbutyrate (PBA) ameliorates hIAPP-induced ß-cell dysfunction and islet amyloid formation. Oral administration of PBA in hIAPP transgenic (hIAPP Tg) mice expressing hIAPP in pancreatic ß cells counteracted impaired glucose homeostasis and restored glucose-stimulated insulin secretion. Moreover, PBA treatment almost completely prevented the transcriptomic alterations observed in hIAPP Tg islets, including the induction of genes related to inflammation. PBA also increased ß-cell viability and improved insulin secretion in hIAPP Tg islets cultured under glucolipotoxic conditions. Strikingly, PBA not only prevented but even reversed islet amyloid deposition, pointing to a direct effect of PBA on hIAPP. This was supported by in silico calculations uncovering potential binding sites of PBA to monomeric, dimeric, and pentameric fibrillar structures, and by in vitro assays showing inhibition of hIAPP fibril formation by PBA. Collectively, these results uncover a novel beneficial effect of PBA on glucose homeostasis by restoring ß-cell function and preventing amyloid formation in mice expressing hIAPP in ß cells, highlighting the therapeutic potential of PBA for the treatment of T2D.-Montane, J., de Pablo, S., Castaño, C., Rodríguez-Comas, J., Cadavez, L., Obach, M., Visa, M., Alcarraz-Vizán, G., Sanchez-Martinez, M., Nonell-Canals, A., Parrizas, M., Servitja, J.-M., Novials, A. Amyloid-induced ß-cell dysfunction and islet inflammation are ameliorated by 4-phenylbutyrate (PBA) treatment.


Assuntos
Amiloide/toxicidade , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Fenilbutiratos/farmacologia , Animais , Teste de Tolerância a Glucose , Humanos , Imuno-Histoquímica , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/ultraestrutura , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Ilhotas Pancreáticas/citologia , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase em Tempo Real
7.
J Biol Chem ; 288(39): 28230-42, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-23943621

RESUMO

Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-activated nuclear receptor and a master regulator of adipogenesis. Microsomal prostaglandin E (PGE) synthase-1 (mPGES-1) is an inducible enzyme that couples with cyclooxygenase-2 for the biosynthesis of PGE2. In this study we demonstrate the existence of a coordinate functional interaction between PPARγ and mPGES-1 in controlling the process of pre-adipocyte differentiation in white adipose tissue (WAT). Adipocyte-specific PPARγ knock-out mice carrying an aP2 promoter-driven Cre recombinase transgene showed a blunted response to the adipogenic effects of a high fat diet. Pre-adipocytes from these knock-out mice showed loss of PPARγ and were resistant to rosiglitazone-induced WAT differentiation. In parallel, WAT from these mice showed increased expression of uncoupling protein 1, a mitochondrial enzyme that dissipates chemical energy as heat. Adipose tissue from mice lacking PPARγ also showed mPGES-1 up-regulation and increased PGE2 levels. In turn, PGE2 suppressed PPARγ expression and blocked rosiglitazone-induced pre-adipocyte differentiation toward white adipocytes while directly elevating uncoupling protein 1 expression and pre-adipocyte differentiation into mature beige/brite adipocytes. Consistently, pharmacological mPGES-1 inhibition directed pre-adipocyte differentiation toward white adipocytes while suppressing differentiation into beige/brite adipocytes. This browning effect was reproduced in knockdown experiments using a siRNA directed against mPGES-1. The effects of PGE2 on pre-adipocyte differentiation were not seen in mice lacking PPARγ in adipose tissue and were not mirrored by other eicosanoids (i.e. leukotriene B4). Taken together, these findings identify PGE2 as a key regulator of white-to-brown adipogenesis and suggest the existence of a coordinate regulation of adipogenesis between PPARγ and mPGES-1.


Assuntos
Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Regulação Enzimológica da Expressão Gênica , Oxirredutases Intramoleculares/metabolismo , Microssomos/enzimologia , PPAR gama/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipogenia , Tecido Adiposo/enzimologia , Animais , Diferenciação Celular , Eicosanoides/metabolismo , Feminino , Homeostase , Inflamação/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Prostaglandina-E Sintases , Prostaglandinas/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo
8.
J Neuroinflammation ; 11: 126, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25051986

RESUMO

BACKGROUND: Aging is characterized by a low-grade systemic inflammation that contributes to the pathogenesis of neurodegenerative disorders such as Alzheimer's disease (AD). However, little knowledge is currently available on the molecular processes leading to chronic neuroinflammation. In this context, recent studies have described the role of chromatin regulators in inflammation and longevity including the REST corepressor (Rcor)-2 factor, which seems to be involved in an inflammatory suppressive program. METHODS: To assess the impact of Rcor2 in age-related inflammation, gene expression levels were quantified in different tissues and ages of the spontaneous senescence-accelerated P8 mouse (P8) using the SAMR1 mouse (R1) as a control. Specific siRNA transfection in P8 and R1 astrocyte cultures was used to determine Rcor2 involvement in the modulation of neuroinflammation. The effect of lipopolysaccharide (LPS) treatment on Rcor2 levels and neuroinflammation was analyzed both in vivo and in vitro. RESULTS: P8 mice presented a dramatic decrease in Rcor2 gene expression compared with R1 controls in splenocytes, an alteration also observed in the brain cortex, hippocampus and primary astrocytes of these mice. Rcor2 reduction in astrocytes was accompanied by an increased basal expression of the interleukin (Il)-6 gene. Strikingly, intraperitoneal LPS injection in R1 mice downregulated Rcor2 in the hippocampus, with a concomitant upregulation of tumor necrosis factor (Tnf-α), Il1-ß and Il6 genes. A negative correlation between Rcor2 and Il6 gene expression was also verified in LPS-treated C6 glioma cells. Knock down of Rcor2 by siRNA transfection (siRcor2) in R1 astrocytes upregulated Il6 gene expression while siRcor2 further increased Il6 expression in P8 astrocytes. Moreover, LPS activation provoked a further downregulation of Rcor2 and an amplified induction of Il6 in siRcor2-tranfected astrocytes. CONCLUSIONS: Data presented here show interplay between Rcor2 downregulation and increased inflammation and suggest that Rcor2 may be a key regulator of inflammaging.


Assuntos
Envelhecimento/genética , Regulação da Expressão Gênica/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Repressoras/metabolismo , Análise de Variância , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/anatomia & histologia , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Proteínas Correpressoras , Citocinas/sangue , Citocinas/genética , Citocinas/metabolismo , Encefalite/induzido quimicamente , Encefalite/patologia , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Interleucina-6/sangue , Lipopolissacarídeos/farmacologia , Masculino , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Proteínas do Tecido Nervoso/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas Repressoras/genética
9.
Curr Opin Clin Nutr Metab Care ; 15(4): 342-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22617562

RESUMO

PURPOSE OF REVIEW: Epigenetic regulation plays an essential role in cell differentiation, by allowing the establishment and maintenance of the gene-expression pattern of the mature cell type. Because of its importance in chronic diseases, adipogenesis is one of the best-studied differentiation processes. The hormonal and transcriptional cascades governing the differentiation of the adipocytes are well known, but the role of epigenetic mechanisms is only starting to emerge. In this review, we intend to summarize the recently described epigenetic events that participate in adipogenesis and their connections with the main factors that constitute the classical transcriptional cascade. RECENT FINDINGS: The advent of high-throughput technologies has made possible the exhaustive analysis of the epigenetic phenomenons taking place during adipogenesis. The cooperative recruitment of CCAAT/enhancer-binding protein (C/EBPß) and other early proadipogenic transcription factors to transcription factor hotspots shortly after induction of adipogenesis is required to establish a transient epigenomic state that then informs the recruitment of the later adipogenic transcription factors peroxisome proliferator-activated receptor (PPARγ) and C/EBPα to their target genes. SUMMARY: Epigenetic marks and chromatin-modifying proteins contribute to adipogenesis and, through regulation of the phenotypic maintenance of the mature adipocytes, to the control of metabolism.


Assuntos
Adipócitos/citologia , Adipogenia/genética , Epigênese Genética , Obesidade/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Adipócitos/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Cromatina/metabolismo , Humanos , PPAR gama/metabolismo , Fenótipo
10.
Muscle Nerve ; 45(1): 128-30, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22190319

RESUMO

The protein kinase product of the gene mutated in myotonic dystrophy 1 (DMPK) is reported to play a role in cardiac pathophysiology. To gain insight into the molecular mechanisms modulated by DMPK, we characterize the impact of DMPK ablation in the context of cardiac ß-adrenergic function. Our data demonstrate that DMPK knockout mice present altered ß-agonist-induced responses and suggest that this is due, at least in part, to a reduced density of ß(1)-adrenergic receptors in cardiac plasma membranes.


Assuntos
Proteínas Serina-Treonina Quinases/deficiência , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ecocardiografia , Isoproterenol/farmacologia , Camundongos , Camundongos Knockout , Miocárdio/citologia , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Distrofia Miotônica/fisiopatologia , Miotonina Proteína Quinase , Fosforilação/efeitos dos fármacos , Receptores Adrenérgicos beta/sangue , Serina/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteínas de Transporte Vesicular/metabolismo
11.
Cells ; 12(1)2022 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-36611962

RESUMO

Obesity and other closely associated diseases, such as metabolic-associated fatty liver disease (MAFLD) and type 2 diabetes, give rise to a common biometric and metabolic phenotype resulting from a different etiopathogenesis. To characterize the first stages of metabolic dysfunction induced by either obesity or hepatic steatosis, we compared two animal models of short-term feeding with either high-fat (HFD) or high-sucrose (SAC) diets. Using transcriptomic, metabolic, and calorimetric analyses, we determined that a short-term HFD leads to obesity and then hepatic steatosis through lipid storage, whereas SAC increases gluconeogenesis and de novo lipogenesis, resulting in hepatic steatosis followed later by obesity. Plasma exosomal miRNA profiles differed between HFD and SAC mice, and the injection of exosomes from HFD or SAC mice reproduced some transcriptomic and metabolic features of the donor mice. Finally, we exploited our data to identify circulating miR-22-3p as a candidate biomarker for MAFLD patient stratification. In conclusion, dietary challenges affecting adipose or hepatic metabolism regulate the abundance of exosomal miRNAs in plasma, which in turn modulate gene expression, helping the organism to adapt.


Assuntos
Diabetes Mellitus Tipo 2 , Exossomos , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Diabetes Mellitus Tipo 2/metabolismo , Exossomos/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/metabolismo , Ração Animal
12.
J Biol Chem ; 285(39): 30034-41, 2010 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-20656681

RESUMO

Epigenetic mechanisms, in particular the enzymatic modification of histones, are a crucial element of cell differentiation, a regulated process that allows a precursor cell basically to turn into a different cell type while maintaining the same genetic equipment. We have previously described that the promoters of adipogenic genes display significant levels of dimethylation at the Lys(4) of histone H3 (H3K4) in preadipocytes, where these genes are still silenced, thus maintaining the chromatin of the precursor cell in a receptive state. Here, we show that the expression of several histone demethylases and methyltransferases increases during adipogenesis, suggesting an important role for these proteins in this process. Knockdown of the H3K4/K9 demethylase LSD1 results in markedly decreased differentiation of 3T3-L1 preadipocytes. This outcome is associated with decreased H3K4 dimethylation and increased H3K9 dimethylation at the promoter of transcription factor cebpa, whose expression must be induced >200-fold upon stimulation of differentiation. Thus, our data suggest that LSD1 acts to maintain a permissive state of the chromatin in this promoter by opposing the action of a H3K9 methyltransferase. Knockdown of H3K9 methyltransferase SETDB1 produced the opposite results, by decreasing H3K9 dimethylation and increasing H3K4 dimethylation levels at the cebpa promoter and favoring differentiation. These findings indicate that the histone methylation status of adipogenic genes as well as the expression and function of the proteins involved in its maintenance play a crucial role in adipogenesis.


Assuntos
Adipócitos/metabolismo , Adipogenia/fisiologia , Diferenciação Celular/fisiologia , Epigênese Genética/fisiologia , Oxirredutases N-Desmetilantes/metabolismo , Células-Tronco/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Cromatina/genética , Cromatina/metabolismo , Técnicas de Silenciamento de Genes , Histona Desmetilases , Histona-Lisina N-Metiltransferase , Histonas/genética , Histonas/metabolismo , Camundongos , Oxirredutases N-Desmetilantes/genética , Regiões Promotoras Genéticas/fisiologia , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Células-Tronco/citologia
13.
Sci Rep ; 11(1): 11878, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-34088954

RESUMO

Amyloid deposits in pancreatic islets, mainly formed by human islet amyloid polypeptide (hIAPP) aggregation, have been associated with loss of ß-cell mass and function, and are a pathological hallmark of type 2 diabetes (T2D). Treatment with chaperones has been associated with a decrease in endoplasmic reticulum stress leading to improved glucose metabolism. The aim of this work was to investigate whether the chemical chaperone 4-phenylbutyrate (PBA) prevents glucose metabolism abnormalities and amyloid deposition in obese agouti viable yellow (Avy) mice that overexpress hIAPP in ß cells (Avy hIAPP mice), which exhibit overt diabetes. Oral PBA treatment started at 8 weeks of age, when Avy hIAPP mice already presented fasting hyperglycemia, glucose intolerance, and impaired insulin secretion. PBA treatment strongly reduced the severe hyperglycemia observed in obese Avy hIAPP mice in fasting and fed conditions throughout the study. This effect was paralleled by a decrease in hyperinsulinemia. Importantly, PBA treatment reduced the prevalence and the severity of islet amyloid deposition in Avy hIAPP mice. Collectively, these results show that PBA treatment elicits a marked reduction of hyperglycemia and reduces amyloid deposits in obese and diabetic mice, highlighting the potential of chaperones for T2D treatment.


Assuntos
Hiperglicemia/tratamento farmacológico , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/metabolismo , Obesidade/tratamento farmacológico , Fenilbutiratos/farmacologia , Amiloide/metabolismo , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Feminino , Intolerância à Glucose/metabolismo , Teste de Tolerância a Glucose , Homeostase , Hiperglicemia/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Obesidade/metabolismo
14.
Mol Metab ; 53: 101251, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34015524

RESUMO

OBJECTIVE: Pancreatic ß-cell dysfunction is a central feature in the pathogenesis of type 2 diabetes (T2D). Accumulating evidence indicates that ß-site APP-cleaving enzyme 2 (BACE2) inhibition exerts a beneficial effect on ß-cells in different models of T2D. Thus, targeting BACE2 may represent a potential therapeutic strategy for the treatment of this disease. Here, we aimed to investigate the effects of BACE2 suppression on glucose homeostasis in a model of diet-induced obesity. METHODS: BACE2 knock-out (BKO) and wild-type (WT) mice were fed with a high-fat diet (HFD) for 2 or 16 weeks. Body weight, food intake, respiratory exchange ratio, locomotor activity, and energy expenditure were determined. Glucose homeostasis was evaluated by glucose and insulin tolerance tests. ß-cell proliferation was assessed by Ki67-positive nuclei, and ß-cell function was determined by measuring glucose-stimulated insulin secretion. Leptin sensitivity was evaluated by quantifying food intake and body weight after an intraperitoneal leptin injection. Neuropeptide gene expression and insulin signaling in the mediobasal hypothalamus were determined by qPCR and Akt phosphorylation, respectively. RESULTS: After 16 weeks of HFD feeding, BKO mice exhibited an exacerbated body weight gain and hyperphagia, in comparison to WT littermates. Glucose tolerance was similar in both groups, whereas HFD-induced hyperinsulinemia, insulin resistance, and ß-cell expansion were more pronounced in BKO mice. In turn, leptin-induced food intake inhibition and hypothalamic insulin signaling were impaired in BKO mice, regardless of the diet, in accordance with deregulation of the expression of hypothalamic neuropeptide genes. Importantly, BKO mice already showed increased ß-cell proliferation and glucose-stimulated insulin secretion with respect to WT littermates after two weeks of HFD feeding, before the onset of obesity. CONCLUSIONS: Collectively, these results reveal that BACE2 suppression in an obesogenic setting leads to exacerbated body weight gain, hyperinsulinemia, and insulin resistance. Thus, we conclude that inhibition of BACE2 may aggravate the adverse metabolic effects associated with obesity.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Obesidade/metabolismo , Animais , Dieta/efeitos adversos , Masculino , Camundongos , Camundongos Transgênicos
15.
Diabetes ; 70(1): 240-254, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33097483

RESUMO

Innovative biomarkers are needed to improve the management of patients with type 2 diabetes mellitus (T2DM). Blood circulating miRNAs have been proposed as a potential tool to detect T2DM complications, but the lack of tissue specificity, among other reasons, has hampered their translation to clinical settings. Extracellular vesicle (EV)-shuttled miRNAs have been proposed as an alternative approach. Here, we adapted an immunomagnetic bead-based method to isolate plasma CD31+ EVs to harvest vesicles deriving from tissues relevant for T2DM complications. Surface marker characterization showed that CD31+ EVs were also positive for a range of markers typical of both platelets and activated endothelial cells. After characterization, we quantified 11 candidate miRNAs associated with vascular performance and shuttled by CD31+ EVs in a large (n = 218) cross-sectional cohort of patients categorized as having T2DM without complications, having T2DM with complications, and control subjects. We found that 10 of the tested miRNAs are affected by T2DM, while the signature composed by miR-146a, -320a, -422a, and -451a efficiently identified T2DM patients with complications. Furthermore, another CD31+ EV-shuttled miRNA signature, i.e., miR-155, -320a, -342-3p, -376, and -422a, detected T2DM patients with a previous major adverse cardiovascular event. Many of these miRNAs significantly correlate with clinical variables held to play a key role in the development of complications. In addition, we show that CD31+ EVs from patients with T2DM are able to promote the expression of selected inflammatory mRNAs, i.e., CCL2, IL-1α, and TNFα, when administered to endothelial cells in vitro. Overall, these data suggest that the miRNA cargo of plasma CD31+ EVs is largely affected by T2DM and related complications, encouraging further research to explore the diagnostic potential and the functional role of these alterations.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Vesículas Extracelulares/metabolismo , MicroRNAs/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Estudos Transversais , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
16.
Methods Mol Biol ; 456: 231-47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18516565

RESUMO

Chromatin, long thought to be no more than a scaffold supporting DNA compaction inside the cell nucleus, has emerged in the last few years as a major regulatory element involved in the control of gene expression both acutely during interphase and programmatically throughout complex processes of development and differentiation. Adipogenesis is the result of an intertwined network of transcription factors and coregulators with chromatin-modifying activities and offers an excellent model for the study of transcriptional regulation. In this regard, electrophoretic mobility shift assay and immunoprecipitation of chromatin are complementary methods that can be used to study the binding of nuclear proteins to DNA and to characterize how these proteins interact with and modify chromatin to regulate gene expression and, more globally, cell differentiation. This chapter provides some strategies to perform these two assays using 3T3-L1 cells and rodent primary preadipocytes and adipocytes.


Assuntos
Adipócitos/fisiologia , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Transcrição Gênica , Células 3T3-L1 , Animais , Imunoprecipitação da Cromatina/instrumentação , Imunoprecipitação da Cromatina/métodos , Ensaio de Desvio de Mobilidade Eletroforética/instrumentação , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Camundongos
17.
Diabetes ; 55(6): 1869-73, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731855

RESUMO

Recently, it has been shown that mutations in the P2 promoter of the hepatocyte nuclear factor (HNF)-4 alpha gene (HNF4A) cause maturity-onset diabetes of the young (MODY), while single nucleotide polymorphisms in this locus are associated with type 2 diabetes. In this study, we examined 1,189 bp of the P2 promoter and the associated exon 1D of HNF4A for variations associated with diabetes in 114 patients with type 2 diabetes, 72 MODYX probands, and 85 women with previous gestational diabetes mellitus. A -192c/g mutation was found in five patients. We screened 1,587 diabetic subjects and 4,812 glucose-tolerant subjects for the -192c/g mutation and identified 5 diabetic and 1 glucose-tolerant mutation carriers (P=0.004). Examination of the families showed that carriers of the -192c/g mutation had a significantly impaired glucose-stimulated insulin release and lower levels of serum total cholesterol compared with matched control subjects. Furthermore, the mutation disrupted the binding of an unidentified sequence-specific DNA binding complex present in human islet extracts. Also, two novel linked polymorphisms in the P2 promoter at positions -1107g/t and -858c/t were identified. These variants were not significantly associated with type 2 diabetes or any pre-diabetic traits. In conclusion, a rare, novel mutation that disrupts a protein binding site in the pancreatic HNF4A promoter associates with late-onset diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Fator 4 Nuclear de Hepatócito/genética , Mutação , Regiões Promotoras Genéticas/genética , Adulto , Fatores Etários , Idoso , Sítios de Ligação , Glicemia/análise , Índice de Massa Corporal , Diabetes Mellitus Tipo 2/sangue , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Genótipo , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Fatores Sexuais
18.
J Clin Invest ; 110(6): 827-33, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12235114

RESUMO

Mutations in the genes encoding hepatocyte nuclear factor 4alpha (HNF-4alpha) and HNF-1alpha impair insulin secretion and cause maturity onset diabetes of the young (MODY). HNF-4alpha is known to be an essential positive regulator of HNF-1alpha. More recent data demonstrates that HNF-4alpha expression is dependent on HNF-1alpha in mouse pancreatic islets and exocrine cells. This effect is mediated by binding of HNF-1alpha to a tissue-specific promoter (P2) located 45.6 kb upstream from the previously characterized Hnf4alpha promoter (P1). Here we report that the expression of HNF-4alpha in human islets and exocrine cells is primarily mediated by the P2 promoter. Furthermore, we describe a G --> A mutation in a conserved nucleotide position of the HNF-1alpha binding site of the P2 promoter, which cosegregates with MODY. The mutation results in decreased affinity for HNF-1alpha, and consequently in reduced HNF-1alpha-dependent activation. These findings provide genetic evidence that HNF-1alpha serves as an upstream regulator of HNF-4alpha and interacts directly with the P2 promoter in human pancreatic cells. Furthermore, they indicate that this regulation is essential to maintain normal pancreatic function.


Assuntos
Ilhotas Pancreáticas/fisiologia , Fosfoproteínas/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Adolescente , Adulto , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Criança , Pré-Escolar , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Genes Reporter , Fator 1 Nuclear de Hepatócito , Fator 1-alfa Nuclear de Hepatócito , Fator 1-beta Nuclear de Hepatócito , Fator 4 Nuclear de Hepatócito , Humanos , Lactente , Masculino , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Linhagem , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
19.
Best Pract Res Clin Endocrinol Metab ; 30(5): 591-601, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27923453

RESUMO

Incidence of diabetes and other metabolic disorders is increasing worldwide, with almost half the cases remaining undiagnosed. This is cause for concern as poor management of glucose or lipid levels causes tissue damage that may result in micro- or macrovascular complications. Current methods of diagnosing metabolic disorders do not provide any clues on disease aetiology or their posterior evolution and incidence of complications, which are the main cause of disease-associated morbidity. Circulating microRNAs found in blood change with the physiological condition of the organism and may help to: (1) identify people at risk of developing metabolic disease, (2) diagnose diabetes or other metabolic disorders on the basis of their aetiology, (3) predict the development of complications, and (4) monitor response to treatment. Results published to date show promise in this direction but technical issues must still be honed in order to warrant their application in the clinical practice.


Assuntos
Doenças Metabólicas/sangue , MicroRNAs/sangue , Biomarcadores/sangue , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo
20.
Mol Cell Endocrinol ; 420: 57-65, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26607804

RESUMO

Human islet amyloid polypeptide (hIAPP) is the major component of amyloid deposits in islets of type 2 diabetic patients. hIAPP misfolding and aggregation is one of the factors that may lead to ß-cell dysfunction and death. Endogenous chaperones are described to be important for the folding and functioning of proteins. Here, we examine the effect of the endoplasmic reticulum chaperone protein disulfide isomerase (PDI) on ß-cell dysfunction. Among other chaperones, PDI was found to interact with hIAPP in human islet lysates. Furthermore, intrinsically recovered PDI levels were able to restore the effect of high glucose- and palmitate-induced ß-cell dysfunction by increasing 3.9-fold the glucose-stimulated insulin secretion levels and restoring insulin content up to basal control values. Additionally, PDI transduction decreased induced apoptosis by glucolipotoxic conditions. This approach could reveal a new therapeutic target and aid in the development of strategies to improve ß-cell dysfunction in type 2 diabetic patients.


Assuntos
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Amiloide/metabolismo , Animais , Apoptose/efeitos dos fármacos , Glucose/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos Transgênicos , Chaperonas Moleculares/metabolismo , Ácido Palmítico/farmacologia , Ligação Proteica/efeitos dos fármacos , Extratos de Tecidos/metabolismo , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA