Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Am J Physiol Endocrinol Metab ; 318(6): E856-E865, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32315211

RESUMO

Obesity is associated with several chronic comorbidities, one of which is type 2 diabetes mellitus (T2DM). The pathogenesis of obesity and T2DM is influenced by alterations in diet macronutrient composition, which regulate energy expenditure, metabolic function, glucose homeostasis, and pancreatic islet cell biology. Recent studies suggest that increased intake of dietary carbohydrates plays a previously underappreciated role in the promotion of obesity and consequent metabolic dysfunction. Thus, in this study, we utilized mouse models to test the hypothesis that dietary carbohydrates modulate energetic, metabolic, and islet adaptions to high-fat diets. To address this, we exposed C57BL/6J mice to 12 wk of 3 eucaloric high-fat diets (>60% calories from fat) with varying total carbohydrate (1-20%) and sucrose (0-20%) content. Our results show that severe restriction of dietary carbohydrates characteristic of ketogenic diets reduces body fat accumulation, enhances energy expenditure, and reduces prevailing glycemia and insulin resistance compared with carbohydrate-rich, high-fat diets. Moreover, severe restriction of dietary carbohydrates also results in functional, morphological, and molecular changes in pancreatic islets highlighted by restricted capacity for ß-cell mass expansion and alterations in insulin secretory response. These studies support the hypothesis that low-carbohydrate/high-fat diets provide antiobesogenic benefits and suggest further evaluation of the effects of these diets on ß-cell biology in humans.


Assuntos
Dieta Hiperlipídica , Dieta Cetogênica , Carboidratos da Dieta , Metabolismo Energético , Hiperglicemia/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Obesidade/metabolismo , Adaptação Fisiológica , Tecido Adiposo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Dieta com Restrição de Gorduras , Sacarose Alimentar , Teste de Tolerância a Glucose , Secreção de Insulina , Camundongos
2.
Physiology (Bethesda) ; 33(2): 138-150, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29412061

RESUMO

The epidemic of Type 2 diabetes mellitus necessitates development of novel therapeutic and preventative strategies to attenuate expansion of this debilitating disease. Evidence links the circadian system to various aspects of diabetes pathophysiology and treatment. The aim of this review will be to outline the rationale for therapeutic targeting of the circadian system in the treatment and prevention of Type 2 diabetes mellitus and consequent metabolic comorbidities.


Assuntos
Ritmo Circadiano , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Animais , Relógios Circadianos , Diabetes Mellitus Tipo 2/prevenção & controle , Homeostase , Humanos
3.
Am J Physiol Endocrinol Metab ; 313(2): E213-E221, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28465284

RESUMO

Metabolic state and circadian clock function exhibit a complex bidirectional relationship. Circadian disruption increases propensity for metabolic dysfunction, whereas common metabolic disorders such as obesity and type 2 diabetes (T2DM) are associated with impaired circadian rhythms. Specifically, alterations in glucose availability and glucose metabolism have been shown to modulate clock gene expression and function in vitro; however, to date, it is unknown whether development of diabetes imparts deleterious effects on the suprachiasmatic nucleus (SCN) circadian clock and SCN-driven outputs in vivo. To address this question, we undertook studies in aged diabetic rats transgenic for human islet amyloid polypeptide, an established nonobese model of T2DM (HIP rat), which develops metabolic defects closely recapitulating those present in patients with T2DM. HIP rats were also cross-bred with a clock gene reporter rat model (Per1:luciferase transgenic rat) to permit assessment of the SCN and the peripheral molecular clock function ex vivo. Utilizing these animal models, we examined effects of diabetes on 1) behavioral circadian rhythms, 2) photic entrainment of circadian activity, 3) SCN and peripheral tissue molecular clock function, and 4) melatonin secretion. We report that circadian activity, light-induced entrainment, molecular clockwork, as well as melatonin secretion are preserved in the HIP rat model of T2DM. These results suggest that despite the well-characterized ability of glucose to modulate circadian clock gene expression acutely in vitro, SCN clock function and key behavioral and physiological outputs appear to be preserved under chronic diabetic conditions characteristic of nonobese T2DM.


Assuntos
Comportamento Animal/fisiologia , Ritmo Circadiano/genética , Diabetes Mellitus Tipo 2 , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Luz , Masculino , Proteínas Circadianas Period/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia
4.
Hum Mol Genet ; 24(10): 3004, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25753258

RESUMO

In this article, Figure 2F was incorrect. The correct panel is shown below. The authors sincerely apologise for this error.

5.
Diabetologia ; 59(4): 734-43, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26762333

RESUMO

AIMS/HYPOTHESIS: Obesity and consequent insulin resistance are known risk factors for type 2 diabetes. A compensatory increase in beta cell function and mass in response to insulin resistance permits maintenance of normal glucose homeostasis, whereas failure to do so results in beta cell failure and type 2 diabetes. Recent evidence suggests that the circadian system is essential for proper metabolic control and regulation of beta cell function. We set out to address the hypothesis that the beta cell circadian clock is essential for the appropriate functional and morphological beta cell response to insulin resistance. METHODS: We employed conditional deletion of the Bmal1 (also known as Arntl) gene (encoding a key circadian clock transcription factor) in beta cells using the tamoxifen-inducible CreER(T) recombination system. Upon adulthood, Bmal1 deletion in beta cells was achieved and mice were exposed to either chow or high fat diet (HFD). Changes in diurnal glycaemia, glucose tolerance and insulin secretion were longitudinally monitored in vivo and islet morphology and turnover assessed by immunofluorescence. Isolated islet experiments in vitro were performed to delineate changes in beta cell function and transcriptional regulation of cell proliferation. RESULTS: Adult Bmal1 deletion in beta cells resulted in failed metabolic adaptation to HFD characterised by fasting and diurnal hyperglycaemia, glucose intolerance and loss of glucose-stimulated insulin secretion. Importantly, HFD-induced beta cell expansion was absent following beta cell Bmal1 deletion indicating impaired beta cell proliferative and regenerative potential, which was confirmed by assessment of transcriptional profiles in isolated islets. CONCLUSION/INTERPRETATION: Results of the study suggest that the beta cell circadian clock is a novel regulator of compensatory beta cell expansion and function in response to increased insulin demand associated with diet-induced obesity.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Obesidade/metabolismo , Fatores de Transcrição ARNTL/genética , Animais , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Sobrevivência Celular/fisiologia , Ritmo Circadiano/fisiologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Hiperglicemia/genética , Hiperglicemia/metabolismo , Masculino , Camundongos , Obesidade/etiologia
6.
Physiol Genomics ; 48(9): 677-87, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27495157

RESUMO

Pancreatic islet failure is a characteristic feature of impaired glucose control in diabetes mellitus. Circadian control of islet function is essential for maintaining proper glucose homeostasis. Circadian variations in transcriptional pathways have been described in diverse cell types and shown to be critical for optimization of cellular function in vivo. In the current study, we utilized Short Time Series Expression Miner (STEM) analysis to identify diurnally expressed transcripts and biological pathways from mouse islets isolated at 4 h intervals throughout the 24 h light-dark cycle. STEM analysis identified 19 distinct chronological model profiles, and genes belonging to each profile were subsequently annotated to significantly enriched Kyoto Encyclopedia of Genes and Genomes biological pathways. Several transcriptional pathways essential for proper islet function (e.g., insulin secretion, oxidative phosphorylation), cell survival (e.g., insulin signaling, apoptosis) and cell proliferation (DNA replication, homologous recombination) demonstrated significant time-dependent variations. Notably, KEGG pathway analysis revealed "protein processing in endoplasmic reticulum - mmu04141" as one of the most enriched time-dependent pathways in islets. This study provides unique data set on time-dependent diurnal profiles of islet gene expression and biological pathways, and suggests that diurnal variation of the islet transcriptome is an important feature of islet homeostasis, which should be taken into consideration for optimal experimental design and interpretation of future islet studies.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Ilhotas Pancreáticas/fisiologia , Transcriptoma/fisiologia , Animais , Glicemia/metabolismo , Proliferação de Células/fisiologia , Expressão Gênica/fisiologia , Homeostase/fisiologia , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia
7.
J Histotechnol ; 39(2): 59-65, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27231405

RESUMO

Laser capture microdissection (LCM) is a powerful method to isolate specific populations of cells for subsequent analysis such as gene expression profiling, for example, microarrays or ribonucleic (RNA)-Seq. This technique has been applied to frozen as well as formalin-fixed, paraffin-embedded (FFPE) specimens with variable outcomes regarding quality and quantity of extracted RNA. The goal of the study was to develop the methods to isolate high-quality RNA from islets of Langerhans and pancreatic duct glands (PDG) isolated by LCM. We report an optimized protocol for frozen sections to minimize RNA degradation and maximize recovery of expected transcripts from the samples using quantitative real-time polymerase chain reaction (RT-PCR) by adding RNase inhibitors at multiple steps during the experiment. This technique reproducibly delivered intact RNA (RIN values 6-7). Using quantitative RT-PCR, the expected profiles of insulin, glucagon, mucin6 (Muc6), and cytokeratin-19 (CK-19) mRNA in PDGs and pancreatic islets were detected. The described experimental protocol for frozen pancreas tissue might also be useful for other tissues with moderate to high levels of intrinsic ribonuclease (RNase) activity.

9.
Curr Diab Rep ; 14(4): 474, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24532160

RESUMO

Type 2 diabetes mellitus (T2DM) is a complex metabolic disease characterized by the loss of beta-cell secretory function and mass. The pathophysiology of beta-cell failure in T2DM involves a complex interaction between genetic susceptibilities and environmental risk factors. One environmental condition that is gaining greater appreciation as a risk factor for T2DM is the disruption of circadian rhythms (eg, shift-work and sleep loss). In recent years, circadian disruption has become increasingly prevalent in modern societies and consistently shown to augment T2DM susceptibility (partly mediated through its effects on pancreatic beta-cells). Since beta-cell failure is essential for development of T2DM, we will review current work from epidemiologic, clinical, and animal studies designed to gain insights into the molecular and physiological mechanisms underlying the predisposition to beta-cell failure associated with circadian disruption. Elucidating the role of circadian clocks in regulating beta-cell health will add to our understanding of T2DM pathophysiology and may contribute to the development of novel therapeutic and preventative approaches.


Assuntos
Glicemia/metabolismo , Ritmo Circadiano , Diabetes Mellitus Tipo 2/fisiopatologia , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Animais , Relógios Circadianos , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Emprego , Estresse do Retículo Endoplasmático , Exposição Ambiental , Feminino , Predisposição Genética para Doença , Humanos , Inflamação/metabolismo , Masculino , Estresse Oxidativo
10.
Aging (Albany NY) ; 15(19): 9984-10009, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37819791

RESUMO

Cellular senescence and circadian dysregulation are biological hallmarks of aging. Whether they are coordinately regulated has not been thoroughly studied. We hypothesize that BMAL1, a pioneer transcription factor and master regulator of the molecular circadian clock, plays a role in the senescence program. Here, we demonstrate BMAL1 is significantly upregulated in senescent cells and has altered rhythmicity compared to non-senescent cells. Through BMAL1-ChIP-seq, we show that BMAL1 is uniquely localized to genomic motifs associated with AP-1 in senescent cells. Integration of BMAL1-ChIP-seq data with RNA-seq data revealed that BMAL1 presence at AP-1 motifs is associated with active transcription. Finally, we showed that BMAL1 contributes to AP-1 transcriptional control of key features of the senescence program, including altered regulation of cell survival pathways, and confers resistance to drug-induced apoptosis. Overall, these results highlight a previously unappreciated role of the core circadian clock component BMAL1 on the molecular phenotype of senescent cells.


Assuntos
Fatores de Transcrição ARNTL , Relógios Circadianos , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Fator de Transcrição AP-1/genética , Regulação da Expressão Gênica , Relógios Circadianos/genética , Senescência Celular/genética , Ritmo Circadiano
11.
Gastroenterology ; 141(1): 150-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21334333

RESUMO

BACKGROUND & AIMS: Glucagon-like peptide-1-based therapy is gaining widespread use for type 2 diabetes, although there are concerns about risks for pancreatitis and pancreatic and thyroid cancers. There are also concerns that dipeptidyl peptidase-4 inhibitors could cause cancer, given their effects on immune function. METHODS: We examined the US Food and Drug Administration's database of reported adverse events for those associated with the dipeptidyl peptidase-4 inhibitor sitagliptin and the glucagon-like peptide-1 mimetic exenatide, from 2004-2009; data on adverse events associated with 4 other medications were compared as controls. The primary outcomes measures were rates of reported pancreatitis, pancreatic and thyroid cancer, and all cancers associated with sitagliptin or exenatide, compared with other therapies. RESULTS: Use of sitagliptin or exenatide increased the odds ratio for reported pancreatitis 6-fold as compared with other therapies (P<2×10(-16)). Pancreatic cancer was more commonly reported among patients who took sitagliptin or exenatide as compared with other therapies (P<.008, P<9×10(-5)). All other cancers occurred similarly among patients who took sitagliptin compared with other therapies (P=.20). CONCLUSIONS: These data are consistent with case reports and animal studies indicating an increased risk for pancreatitis with glucagon-like peptide-1-based therapy. The findings also raise caution about the potential long-term actions of these drugs to promote pancreatic cancer.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Hipoglicemiantes/efeitos adversos , Neoplasias Pancreáticas/induzido quimicamente , Pancreatite/induzido quimicamente , Peptídeos/efeitos adversos , Pirazinas/efeitos adversos , Receptores de Glucagon/agonistas , Neoplasias da Glândula Tireoide/induzido quimicamente , Triazóis/efeitos adversos , Peçonhas/efeitos adversos , Sistemas de Notificação de Reações Adversas a Medicamentos , Diabetes Mellitus Tipo 2/epidemiologia , Exenatida , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Razão de Chances , Neoplasias Pancreáticas/epidemiologia , Pancreatite/epidemiologia , Medição de Risco , Fatores de Risco , Fosfato de Sitagliptina , Neoplasias da Glândula Tireoide/epidemiologia , Estados Unidos/epidemiologia , United States Food and Drug Administration
12.
Front Endocrinol (Lausanne) ; 13: 842603, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35355560

RESUMO

Our ever-changing modern environment is a significant contributor to the increased prevalence of many chronic diseases, and particularly, type 2 diabetes mellitus (T2DM). Although the modern era has ushered in numerous changes to our daily living conditions, changes in "what" and "when" we eat appear to disproportionately fuel the rise of T2DM. The pancreatic islet is a key biological controller of an organism's glucose homeostasis and thus plays an outsized role to coordinate the response to environmental factors to preserve euglycemia through a delicate balance of endocrine outputs. Both successful and failed adaptation to dynamic environmental stimuli has been postulated to occur due to changes in the transcriptional and epigenetic regulation of pathways associated with islet secretory function and survival. Therefore, in this review we examined and evaluated the current evidence elucidating the key epigenetic mechanisms and transcriptional programs underlying the islet's coordinated response to the interaction between the timing and the composition of dietary nutrients common to modern lifestyles. With the explosion of next generation sequencing, along with the development of novel informatic and -omic approaches, future work will continue to unravel the environmental-epigenetic relationship in islet biology with the goal of identifying transcriptional and epigenetic targets associated with islet perturbations in T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Ilhotas Pancreáticas , Diabetes Mellitus Tipo 2/complicações , Dieta , Epigênese Genética , Homeostase , Humanos , Ilhotas Pancreáticas/metabolismo
13.
Cell Death Dis ; 13(4): 353, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35428762

RESUMO

Pancreatic ß-cell failure in type 2 diabetes mellitus (T2DM) is associated with impaired regulation of autophagy which controls ß-cell development, function, and survival through clearance of misfolded proteins and damaged organelles. However, the mechanisms responsible for defective autophagy in T2DM ß-cells remain unknown. Since recent studies identified circadian clock transcriptional repressor REV-ERBα as a novel regulator of autophagy in cancer, in this study we set out to test whether REV-ERBα-mediated inhibition of autophagy contributes to the ß-cell failure in T2DM. Our study provides evidence that common diabetogenic stressors (e.g., glucotoxicity and cytokine-mediated inflammation) augment ß-cell REV-ERBα expression and impair ß-cell autophagy and survival. Notably, pharmacological activation of REV-ERBα was shown to phenocopy effects of diabetogenic stressors on the ß-cell through inhibition of autophagic flux, survival, and insulin secretion. In contrast, negative modulation of REV-ERBα was shown to provide partial protection from inflammation and glucotoxicity-induced ß-cell failure. Finally, using bioinformatic approaches, we provide further supporting evidence for augmented REV-ERBα activity in T2DM human islets associated with impaired transcriptional regulation of autophagy and protein degradation pathways. In conclusion, our study reveals a previously unexplored causative relationship between REV-ERBα expression, inhibition of autophagy, and ß-cell failure in T2DM.


Assuntos
Relógios Circadianos , Diabetes Mellitus Tipo 2 , Autofagia/genética , Ritmo Circadiano/fisiologia , Diabetes Mellitus Tipo 2/genética , Humanos , Inflamação , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo
14.
Hum Mol Genet ; 18(13): 2388-99, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19386626

RESUMO

Recent human genetics studies have revealed that common variants of the TCF7L2 (T-cell factor 7-like 2, formerly known as TCF4) gene are strongly associated with type 2 diabetes mellitus (T2DM). We have shown that TCF7L2 expression in the beta-cells is correlated with function and survival of the insulin-producing pancreatic beta-cell. In order to understand how variations in TCF7L2 influence diabetes progression, we investigated its mechanism of action in the beta-cell. We show robust differences in TCF7L2 expression between healthy controls and models of T2DM. While mRNA levels were approximately 2-fold increased in isolated islets from the diabetic db/db mouse, the Vancouver Diabetic Fatty (VDF) Zucker rat and the high fat/high sucrose diet-treated mouse compared with the non-diabetic controls, protein levels were decreased. A similar decrease was observed in pancreatic sections from patients with T2DM. In parallel, expression of the receptors for glucagon-like peptide 1 (GLP-1R) and glucose-dependent insulinotropic polypeptide (GIP-R) was decreased in islets from humans with T2DM as well as in isolated human islets treated with siRNA to TCF7L2 (siTCF7L2). Also, insulin secretion stimulated by glucose, GLP-1 and GIP, but not KCl or cyclic adenosine monophosphate (cAMP) was impaired in siTCF7L2-treated isolated human islets. Loss of TCF7L2 resulted in decreased GLP-1 and GIP-stimulated AKT phosphorylation, and AKT-mediated Foxo-1 phosphorylation and nuclear exclusion. Our findings suggest that beta-cell function and survival are regulated through an interplay between TCF7L2 and GLP-1R/GIP-R expression and signaling in T2DM.


Assuntos
Regulação para Baixo , Células Secretoras de Insulina/fisiologia , Receptores dos Hormônios Gastrointestinais/genética , Receptores de Glucagon/genética , Fatores de Transcrição TCF/metabolismo , Idoso , Animais , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Ratos , Ratos Zucker , Receptores dos Hormônios Gastrointestinais/metabolismo , Receptores de Glucagon/metabolismo , Transdução de Sinais , Fatores de Transcrição TCF/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição
15.
Diabetes ; 70(1): 143-154, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33087455

RESUMO

Type 2 diabetes mellitus (T2DM) is characterized by ß-cell dysfunction as a result of impaired glucose-stimulated insulin secretion (GSIS). Studies show that ß-cell circadian clocks are important regulators of GSIS and glucose homeostasis. These observations raise the question about whether enhancement of the circadian clock in ß-cells will confer protection against ß-cell dysfunction under diabetogenic conditions. To test this, we used an approach by first generating mice with ß-cell-specific inducible overexpression of Bmal1 (core circadian transcription factor; ß-Bmal1 OV ). We subsequently examined the effects of ß-Bmal1 OV on the circadian clock, GSIS, islet transcriptome, and glucose metabolism in the context of diet-induced obesity. We also tested the effects of circadian clock-enhancing small-molecule nobiletin on GSIS in mouse and human control and T2DM islets. We report that ß-Bmal1 OV mice display enhanced islet circadian clock amplitude and augmented in vivo and in vitro GSIS and are protected against obesity-induced glucose intolerance. These effects were associated with increased expression of purported BMAL1-target genes mediating insulin secretion, processing, and lipid metabolism. Furthermore, exposure of isolated islets to nobiletin enhanced ß-cell secretory function in a Bmal1-dependent manner. This work suggests therapeutic targeting of the circadian system as a potential strategy to counteract ß-cell failure under diabetogenic conditions.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Intolerância à Glucose/metabolismo , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Obesidade/metabolismo , Fatores de Transcrição ARNTL/genética , Animais , Glicemia/metabolismo , Relógios Circadianos/genética , Ritmo Circadiano/genética , Flavonas/farmacologia , Glucose/farmacologia , Intolerância à Glucose/etiologia , Intolerância à Glucose/genética , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Obesidade/complicações , Obesidade/genética
16.
Endocrinology ; 162(1)2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32455427

RESUMO

Intrinsic ß-cell circadian clocks are important regulators of insulin secretion and overall glucose homeostasis. Whether the circadian clock in ß-cells is perturbed following exposure to prodiabetogenic stressors such as proinflammatory cytokines, and whether these perturbations are featured during the development of diabetes, remains unknown. To address this, we examined the effects of cytokine-mediated inflammation common to the pathophysiology of diabetes, on the physiological and molecular regulation of the ß-cell circadian clock. Specifically, we provide evidence that the key diabetogenic cytokine IL-1ß disrupts functionality of the ß-cell circadian clock and impairs circadian regulation of glucose-stimulated insulin secretion. The deleterious effects of IL-1ß on the circadian clock were attributed to impaired expression of key circadian transcription factor Bmal1, and its regulator, the NAD-dependent deacetylase, Sirtuin 1 (SIRT1). Moreover, we also identified that Type 2 diabetes in humans is associated with reduced immunoreactivity of ß-cell BMAL1 and SIRT1, suggestive of a potential causative link between islet inflammation, circadian clock disruption, and ß-cell failure. These data suggest that the circadian clock in ß-cells is perturbed following exposure to proinflammatory stressors and highlights the potential for therapeutic targeting of the circadian system for treatment for ß-cell failure in diabetes.


Assuntos
Relógios Circadianos/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Interleucina-1beta/metabolismo , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Idoso , Animais , Relógios Circadianos/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Células Secretoras de Insulina/metabolismo , Insulinoma , Interleucina-1beta/efeitos adversos , Interleucina-1beta/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Ratos , Sirtuínas/genética , Sirtuínas/metabolismo
17.
Cell Rep ; 36(8): 109613, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34433033

RESUMO

Coordinated communication among pancreatic islet cells is necessary for maintenance of glucose homeostasis. In diabetes, chronic exposure to pro-inflammatory cytokines has been shown to perturb ß cell communication and function. Compelling evidence has implicated extracellular vesicles (EVs) in modulating physiological and pathological responses to ß cell stress. We report that pro-inflammatory ß cell small EVs (cytokine-exposed EVs [cytoEVs]) induce ß cell dysfunction, promote a pro-inflammatory islet transcriptome, and enhance recruitment of CD8+ T cells and macrophages. Proteomic analysis of cytoEVs shows enrichment of the chemokine CXCL10, with surface topological analysis depicting CXCL10 as membrane bound on cytoEVs to facilitate direct binding to CXCR3 receptors on the surface of ß cells. CXCR3 receptor inhibition reduced CXCL10-cytoEV binding and attenuated ß cell dysfunction, inflammatory gene expression, and leukocyte recruitment to islets. This work implies a significant role of pro-inflammatory ß cell-derived small EVs in modulating ß cell function, global gene expression, and antigen presentation through activation of the CXCL10/CXCR3 axis.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Quimiocina CXCL10/metabolismo , Vesículas Extracelulares/metabolismo , Receptores CXCR3/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus/patologia , Células Secretoras de Insulina/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL
18.
Commun Biol ; 4(1): 594, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-34012065

RESUMO

Type 2 diabetes is characterized by ß and α cell dysfunction. We used phasor-FLIM (Fluorescence Lifetime Imaging Microscopy) to monitor oxidative phosphorylation and glycolysis in living islet cells before and after glucose stimulation. In healthy cells, glucose enhanced oxidative phosphorylation in ß cells and suppressed oxidative phosphorylation in α cells. In Type 2 diabetes, glucose increased glycolysis in ß cells, and only partially suppressed oxidative phosphorylation in α cells. FLIM uncovers key perturbations in glucose induced metabolism in living islet cells and provides a sensitive tool for drug discovery in diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Imagem Molecular/métodos , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/patologia , Células Secretoras de Glucagon/efeitos dos fármacos , Glicólise , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Microscopia de Fluorescência , Fosforilação Oxidativa , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos
19.
J Clin Invest ; 131(17)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34623331

RESUMO

Pancreatic ß cell failure in type 2 diabetes mellitus (T2DM) is attributed to perturbations of the ß cell's transcriptional landscape resulting in impaired glucose-stimulated insulin secretion. Recent studies identified SLC4A4 (a gene encoding an electrogenic Na+-coupled HCO3- cotransporter and intracellular pH regulator, NBCe1) as one of the misexpressed genes in ß cells of patients with T2DM. Thus, in the current study, we set out to test the hypothesis that misexpression of SLC4A4/NBCe1 in T2DM ß cells contributes to ß cell dysfunction and impaired glucose homeostasis. To address this hypothesis, we first confirmed induction of SLC4A4/NBCe1 expression in ß cells of patients with T2DM and demonstrated that its expression was associated with loss of ß cell transcriptional identity, intracellular alkalinization, and ß cell dysfunction. In addition, we generated a ß cell-selective Slc4a4/NBCe1-KO mouse model and found that these mice were protected from diet-induced metabolic stress and ß cell dysfunction. Importantly, improved glucose tolerance and enhanced ß cell function in Slc4a4/NBCe1-deficient mice were due to augmented mitochondrial function and increased expression of genes regulating ß cell identity and function. These results suggest that increased ß cell expression of SLC4A4/NBCe1 in T2DM plays a contributory role in promotion of ß cell failure and should be considered as a potential therapeutic target.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Expressão Gênica , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Intolerância à Glucose/prevenção & controle , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Obesidade/genética , Obesidade/metabolismo , Simportadores de Sódio-Bicarbonato/deficiência , Simportadores de Sódio-Bicarbonato/genética , Estresse Fisiológico
20.
Sci Adv ; 7(51): eabg6856, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34910509

RESUMO

Circadian rhythm disruption (CD) is associated with impaired glucose homeostasis and type 2 diabetes mellitus (T2DM). While the link between CD and T2DM remains unclear, there is accumulating evidence that disruption of fasting/feeding cycles mediates metabolic dysfunction. Here, we used an approach encompassing analysis of behavioral, physiological, transcriptomic, and epigenomic effects of CD and consequences of restoring fasting/feeding cycles through time-restricted feeding (tRF) in mice. Results show that CD perturbs glucose homeostasis through disruption of pancreatic ß cell function and loss of circadian transcriptional and epigenetic identity. In contrast, restoration of fasting/feeding cycle prevented CD-mediated dysfunction by reestablishing circadian regulation of glucose tolerance, ß cell function, transcriptional profile, and reestablishment of proline and acidic amino acid­rich basic leucine zipper (PAR bZIP) transcription factor DBP expression/activity. This study provides mechanistic insights into circadian regulation of ß cell function and corresponding beneficial effects of tRF in prevention of T2DM.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa