Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
3.
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
4.
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
5.
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.

6.
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
7.
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
8.
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
9.
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
10.
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
11.
Sci Rep ; 10(1): 19374, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33168920

RESUMO

Small-molecule inhibitors of non-canonical IκB kinases TANK-binding kinase 1 (TBK1) and IκB kinase ε (IKKε) have shown to stimulate ß-cell regeneration in multiple species. Here we demonstrate that TBK1 is predominantly expressed in ß-cells in mammalian islets. Proteomic and transcriptome analyses revealed that genetic silencing of TBK1 increased expression of proteins and genes essential for cell proliferation in INS-1 832/13 rat ß-cells. Conversely, TBK1 overexpression decreased sensitivity of ß-cells to the elevation of cyclic AMP (cAMP) levels and reduced proliferation of ß-cells in a manner dependent on the activity of cAMP-hydrolyzing phosphodiesterase 3 (PDE3). While the mitogenic effect of (E)3-(3-phenylbenzo[c]isoxazol-5-yl)acrylic acid (PIAA) is derived from inhibition of TBK1, PIAA augmented glucose-stimulated insulin secretion (GSIS) and expression of ß-cell differentiation and proliferation markers in human embryonic stem cell (hESC)-derived ß-cells and human islets. TBK1 expression was increased in ß-cells upon diabetogenic insults, including in human type 2 diabetic islets. PIAA enhanced expression of cell cycle control molecules and ß-cell differentiation markers upon diabetogenic challenges, and accelerated restoration of functional ß-cells in streptozotocin (STZ)-induced diabetic mice. Altogether, these data suggest the critical function of TBK1 as a ß-cell autonomous replication barrier and present PIAA as a valid therapeutic strategy augmenting functional ß-cells.


Assuntos
Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Células Secretoras de Insulina/enzimologia , Proteínas Serina-Treonina Quinases/biossíntese , Regeneração , Animais , Linhagem Celular Tumoral , Inativação Gênica , Células-Tronco Embrionárias Humanas/enzimologia , Humanos , Insulina/genética , Insulina/metabolismo , Secreção de Insulina , Proteínas Serina-Treonina Quinases/genética , Ratos
12.
Nat Commun ; 11(1): 2241, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32382023

RESUMO

The generation of pancreatic cell types from renewable cell sources holds promise for cell replacement therapies for diabetes. Although most effort has focused on generating pancreatic beta cells, considerable evidence indicates that glucagon secreting alpha cells are critically involved in disease progression and proper glucose control. Here we report on the generation of stem cell-derived human pancreatic alpha (SC-alpha) cells from pluripotent stem cells via a transient pre-alpha cell intermediate. These pre-alpha cells exhibit a transcriptional profile similar to mature alpha cells and although they produce proinsulin protein, they do not secrete significant amounts of processed insulin. Compound screening identified a protein kinase c activator that promotes maturation of pre-alpha cells into SC-alpha cells. The resulting SC-alpha cells do not express insulin, share an ultrastructure similar to cadaveric alpha cells, express and secrete glucagon in response to glucose and some glucagon secretagogues, and elevate blood glucose upon transplantation in mice.


Assuntos
Técnicas de Cultura de Células/métodos , Células Secretoras de Glucagon/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Western Blotting , Diferenciação Celular/fisiologia , Linhagem Celular , Eletrofisiologia , Imunofluorescência , Humanos , Pâncreas/citologia
13.
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
14.
JCI Insight ; 5(9)2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32267250

RESUMO

The worldwide prevalence of type 2 diabetes (T2D) is increasing. Despite normal to higher bone density, patients with T2D paradoxically have elevated fracture risk resulting, in part, from poor bone quality. Advanced glycation endproducts (AGEs) and inflammation as a consequence of enhanced receptor for AGE (RAGE) signaling are hypothesized culprits, although the exact mechanisms underlying skeletal dysfunction in T2D are unclear. Lack of inducible models that permit environmental (in obesity) and temporal (after skeletal maturity) control of T2D onset has hampered progress. Here, we show in C57BL/6 mice that a onetime pharmacological intervention (streptozotocin, STZ) initiated in adulthood combined with high-fat diet-induced (HFD-induced) obesity caused hallmark features of human adult-onset T2D, including prolonged hyperglycemia, insulin resistance, and pancreatic ß cell dysfunction, but not complete destruction. In addition, HFD/STZ (i.e., T2D) resulted in several changes in bone quality that closely mirror those observed in humans, including compromised bone microarchitecture, reduced biomechanical strength, impaired bone material properties, altered bone turnover, and elevated levels of the AGE CML in bone and blood. Furthermore, T2D led to the premature accumulation of senescent osteocytes with a unique proinflammatory signature. These findings highlight the RAGE pathway and senescent cells as potential targets to treat diabetic skeletal fragility.


Assuntos
Osso e Ossos , Diabetes Mellitus Tipo 2/metabolismo , Osteócitos , Animais , Densidade Óssea , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Senescência Celular , Modelos Animais de Doenças , Produtos Finais de Glicação Avançada/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteócitos/metabolismo , Osteócitos/patologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo
15.
Sci Rep ; 9(1): 18570, 2019 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-31796771

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

16.
Stem Cell Reports ; 13(2): 307-321, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31378674

RESUMO

Generation of functional ß cells from pluripotent sources would accelerate diagnostic and therapeutic applications for diabetes research and therapy. However, it has been challenging to generate competent ß cells with dynamic insulin-secretory capacity to glucose and incretin stimulations. We introduced transcription factors, critical for ß-cell development and function, in differentiating human induced pluripotent stem cells (PSCs) and assessed the impact on the functionality of derived ß-cell (psBC) progeny. A perifusion system revealed stepwise transduction of the PDX1, NEUROG3, and MAFA triad (PNM) enabled in vitro generation of psBCs with glucose and GLP-1 responsiveness within 3 weeks. PNM transduction upregulated genes associated with glucose sensing, insulin secretion, and ß-cell maturation. In recipient diabetic mice, PNM-transduced psBCs showed glucose-responsive insulin secretion as early as 1 week post transplantation. Thus, enhanced pre-emptive ß-cell specification of PSCs by PNM drives generation of glucose- and incretin-responsive psBCs in vitro, offering a competent tissue-primed biotherapy.


Assuntos
Diabetes Mellitus Experimental/terapia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Glucose/farmacologia , Células-Tronco Pluripotentes Induzidas/transplante , Secreção de Insulina/efeitos dos fármacos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Peptídeo C/metabolismo , Diferenciação Celular , Diabetes Mellitus Experimental/induzido quimicamente , Regulação da Expressão Gênica , Teste de Tolerância a Glucose , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Camundongos , Camundongos SCID , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Transativadores/genética , Transativadores/metabolismo , Transdução Genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
17.
Sci Rep ; 8(1): 15587, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30349097

RESUMO

ß-cell proliferation induction is a promising therapeutic strategy to restore ß-cell mass. By screening small molecules in a transgenic zebrafish model of type 1 diabetes, we identified inhibitors of non-canonical IκB kinases (IKKs), TANK-binding kinase 1 (TBK1) and IκB kinase ε (IKKε), as enhancers of ß-cell regeneration. The most potent ß-cell regeneration enhancer was a cinnamic acid derivative (E)-3-(3-phenylbenzo[c]isoxazol-5-yl)acrylic acid (PIAA), which, acting through the cAMP-dependent protein kinase A (PKA), stimulated ß-cell-specific proliferation by increasing cyclic AMP (cAMP) levels and mechanistic target of rapamycin (mTOR) activity. A combination of PIAA and cilostamide, an inhibitor of ß-cell-enriched cAMP hydrolyzing enzyme phosphodiesterase (PDE) 3, enhanced ß-cell proliferation, whereas overexpression of PDE3 blunted the mitogenic effect of PIAA in zebrafish. PIAA augmented proliferation of INS-1ß-cells and ß-cells in mammalian islets including human islets with elevation in cAMP levels and insulin secretion. PIAA improved glycemic control in streptozotocin (STZ)-induced diabetic mice with increases in ß-cell proliferation, ß-cell area, and insulin content in the pancreas. Collectively, these data reveal an evolutionarily conserved and critical role of TBK1/IKKε suppression in expanding functional ß-cell mass.


Assuntos
Proliferação de Células/efeitos dos fármacos , Quinase I-kappa B/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Regeneração/efeitos dos fármacos , Animais , Cinamatos/metabolismo , Humanos , Quinolonas/metabolismo , Ratos Endogâmicos Lew , Peixe-Zebra
20.
Dis Model Mech ; 11(6)2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29915142

RESUMO

High-fat diet (HFD)-fed mouse models have been widely used to study early type 2 diabetes. Decreased ß-cell glucokinase (GCK) expression has been observed in HFD-induced diabetes. However, owing to its crucial roles in glucose metabolism in the liver and in islet ß-cells, the contribution of decreased GCK expression to the development of HFD-induced diabetes is unclear. Here, we employed a ß-cell-targeted gene transfer vector and determined the impact of ß-cell-specific increase in GCK expression on ß-cell function and glucose handling in vitro and in vivo Overexpression of GCK enhanced glycolytic flux, ATP-sensitive potassium channel activation and membrane depolarization, and increased proliferation in Min6 cells. ß-cell-targeted GCK transduction did not change glucose handling in chow-fed C57BL/6 mice. Although adult mice fed a HFD showed reduced islet GCK expression, impaired glucose tolerance and decreased glucose-stimulated insulin secretion (GSIS), ß-cell-targeted GCK transduction improved glucose tolerance and restored GSIS. Islet perifusion experiments verified restored GSIS in isolated HFD islets by GCK transduction. Thus, our data identify impaired ß-cell GCK expression as an underlying mechanism for dysregulated ß-cell function and glycemic control in HFD-induced diabetes. Our data also imply an etiological role of GCK in diet-induced diabetes.This article has an associated First Person interview with the first author of the paper.


Assuntos
Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/patologia , Glucoquinase/metabolismo , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Animais , Cálcio/metabolismo , Proliferação de Células , Dependovirus/metabolismo , Diabetes Mellitus Experimental/genética , Dieta Hiperlipídica , Glucose/metabolismo , Teste de Tolerância a Glucose , Glicólise , Insulina/metabolismo , Espaço Intracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais , Transdução Genética , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...