Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Diabetes ; 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38905124

RESUMO

Exocrine-to-endocrine crosstalk in the pancreas is crucial to maintain beta cell function. However, the molecular mechanisms underlying this crosstalk are largely undefined. Trefoil factor 2 (Tff2) is a secreted factor known to promote the proliferation of beta cells in vitro, but its physiological role in vivo in the pancreas is unknown. Also, it remains unclear which pancreatic cell type expresses Tff2 protein. We therefore created a mouse model with a conditional knockout of Tff2 in the murine pancreas. We find that the Tff2 protein is preferentially expressed in acinar but not ductal or endocrine cells. Tff2 deficiency in the pancreas reduces beta cell mass on embryonic day 16.5. However, homozygous mutant mice are born without a reduction of beta cells and with acinar Tff3 compensation by day 7. When mice are aged to 1 year, both male and female homozygous and male heterozygous mutants develop impaired glucose tolerance without affected insulin sensitivity. Perifusion analysis reveals that the second phase of glucose-stimulated insulin secretion from islets is reduced in aged homozygous mutant compared to controls. Collectively, these results demonstrate a previously unknown role of Tff2 as an exocrine acinar cell-derived protein required for maintaining functional endocrine beta cells in mice.

2.
Int J Mol Sci ; 25(3)2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38338783

RESUMO

Type 2 diabetes (T2D) is a heterogenous disease, and conventionally, peripheral insulin resistance (IR) was thought to precede islet ß-cell dysfunction, promoting progression from prediabetes to T2D. New evidence suggests that T2D-lean individuals experience early ß-cell dysfunction without significant IR. Regardless of the primary event (i.e., IR vs. ß-cell dysfunction) that contributes to dysglycemia, significant early-onset oxidative damage and mitochondrial dysfunction in multiple metabolic tissues may be a driver of T2D onset and progression. Oxidative stress, defined as the generation of reactive oxygen species (ROS), is mediated by hyperglycemia alone or in combination with lipids. Physiological oxidative stress promotes inter-tissue communication, while pathological oxidative stress promotes inter-tissue mis-communication, and new evidence suggests that this is mediated via extracellular vesicles (EVs), including mitochondria containing EVs. Under metabolic-related stress conditions, EV-mediated cross-talk between ß-cells and skeletal muscle likely trigger mitochondrial anomalies leading to prediabetes and T2D. This article reviews the underlying molecular mechanisms in ROS-related pathogenesis of prediabetes, including mitophagy and mitochondrial dynamics due to oxidative stress. Further, this review will describe the potential of various therapeutic avenues for attenuating oxidative damage, reversing prediabetes and preventing progression to T2D.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Doenças Mitocondriais , Estado Pré-Diabético , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estresse Oxidativo/fisiologia , Comunicação
3.
J Korean Acad Nurs ; 53(1): 69-86, 2023 Feb.
Artigo em Coreano | MEDLINE | ID: mdl-36898686

RESUMO

PURPOSE: This study aimed to evaluate the effects of digital health interventions on the psychotic symptoms among people with severe mental illness in the community. METHODS: A systematic review and meta-analysis were conducted in accordance with the Cochrane Intervention Research Systematic Review Manual and PRISMA. A literature search was conducted of published randomized controlled trials (RCTs) for digital health interventions from January 2022 to April 2022. RevMan software 5.3 was used for quality assessment and meta-analysis. RESULTS: A total 14 studies out of 9,864 studies were included in the review, and 13 were included in meta-analysis. The overall effect size of digital health interventions on psychotic symptoms was -0.21 (95% CI = -0.32 to -0.10). Sub-analysis showed that the reduction of the psychotic symptoms was effective in the schizophrenia spectrum group (SMD = -.0.22; 95% CI = -.0.36 to -0.09), web (SMD = -0.41; 95% CI = -0.82 to 0.01), virtual reality (SMD = -0.33; 95% CI = -0.56 to -0.10), mobile (SMD = -0.15; 95% CI = -0.28 to -0.03), intervention period of less than 3 months (SMD = -0.23; 95% CI = -0.35 to -0.11), and non-treatment group (SMD = -0.23; 95% CI = -0.36 to -0.11). CONCLUSION: These findings suggest that digital health interventions alleviate psychotic symptoms in patients with severe mental illnesses. However, well-designed digital health studies should be conducted in the future.


Assuntos
Transtornos Mentais , Humanos , Depressão
4.
Int J Mol Sci ; 25(1)2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38203312

RESUMO

Diet-related lipotoxic stress is a significant driver of skeletal muscle insulin resistance (IR) and type 2 diabetes (T2D) onset. ß2-adrenergic receptor (ß-AR) agonism promotes insulin sensitivity in vivo under lipotoxic stress conditions. Here, we established an in vitro paradigm of lipotoxic stress using palmitate (Palm) in rat skeletal muscle cells to determine if ß-AR agonism could cooperate with double C-2-like domain beta (DOC2B) enrichment to promote skeletal muscle insulin sensitivity under Palm-stress conditions. Previously, human T2D skeletal muscles were shown to be deficient for DOC2B, and DOC2B enrichment resisted IR in vivo. Our Palm-stress paradigm induced IR and ß-AR resistance, reduced DOC2B protein levels, triggered cytoskeletal cofilin phosphorylation, and reduced GLUT4 translocation to the plasma membrane (PM). By enhancing DOC2B levels in rat skeletal muscle, we showed that the deleterious effects of palmitate exposure upon cofilin, insulin, and ß-AR-stimulated GLUT4 trafficking to the PM and glucose uptake were preventable. In conclusion, we revealed a useful in vitro paradigm of Palm-induced stress to test for factors that can prevent/reverse skeletal muscle dysfunctions related to obesity/pre-T2D. Discerning strategies to enrich DOC2B and promote ß-AR agonism can resist skeletal muscle IR and halt progression to T2D.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Humanos , Animais , Ratos , Músculo Esquelético , Fatores de Despolimerização de Actina , Palmitatos/farmacologia , Glucose , Proteínas de Ligação ao Cálcio , Proteínas do Tecido Nervoso
5.
Diabetes ; 71(6): 1246-1260, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35377441

RESUMO

Double C2 domain Β (DOC2b) protein is required for glucose-stimulated insulin secretion (GSIS) in ß-cells, the underlying mechanism of which remains unresolved. Our biochemical analysis using primary human islets and human and rodent clonal ß-cells revealed that DOC2b is tyrosine phosphorylated within 2 min of glucose stimulation, and Src family kinase member YES is required for this process. Biochemical and functional analysis using DOC2bY301 mutants revealed the requirement of Y301 phosphorylation for the interaction of DOC2b with YES kinase and increased content of VAMP2, a protein on insulin secretory granules, at the plasma membrane (PM), concomitant with DOC2b-mediated enhancement of GSIS in ß-cells. Coimmunoprecipitation studies demonstrated an increased association of DOC2b with ERM family proteins in ß-cells following glucose stimulation or pervanadate treatment. Y301 phosphorylation-competent DOC2b was required to increase ERM protein activation, and ERM protein knockdown impaired DOC2b-mediated boosting of GSIS, suggesting that tyrosine-phosphorylated DOC2b regulates GSIS via ERM-mediated granule localization to the PM. Taken together, these results demonstrate the glucose-induced posttranslational modification of DOC2b in ß-cells, pinpointing the kinase, site of action, and downstream signaling events and revealing a regulatory role of YES kinase at various steps in GSIS. This work will enhance the development of novel therapeutic strategies to restore glucose homeostasis in diabetes.


Assuntos
Proteínas de Ligação ao Cálcio , Células Secretoras de Insulina , Proteínas de Ligação ao Cálcio/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Tirosina/metabolismo
6.
Indoor Air ; 32(3): e13013, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35347791

RESUMO

The association between particulate matter and children's increased blood pressure is inconsistent, and few studies have evaluated indoor exposure, accounting for time-activity. The present study aimed to examine the association between personal short-term exposure to PM2.5 and blood pressure in children. We conducted a panel study with up to three physical examinations during different seasons of 2018 (spring, summer, and fall) among 52 children. The indoor PM2.5 concentration was continuously measured at home and classroom of each child using indoor air quality monitors. The outdoor PM2.5 concentration was measured from the nearest monitoring station. We constructed a mixed effect model to analyze the association of short-term indoor and outdoor PM2.5 exposure accounting for time-activity of each participant with blood pressure. The average PM2.5 concentration was 34.3 ± 9.2 µg/m3 and it was highest in the spring. The concentration measured at homes was generally higher than that measured at outdoor monitoring station. A 10-µg/m3 increment of the up to previous 3-day mean (lag0-3) PM2.5 concentration was associated with 2.7 mmHg (95%CI = 0.8, 4.0) and 2.1 mmHg (95%CI = 0.3, 4.0) increases in systolic and diastolic blood pressure, respectively. In a panel study comprehensively evaluating both indoor and outdoor exposures, which enabled more accurate exposure assessment, we observed a statistically significant association between blood pressure and PM2.5 exposure in children.


Assuntos
Poluentes Atmosféricos , Poluição do Ar em Ambientes Fechados , Poluentes Atmosféricos/análise , Poluição do Ar em Ambientes Fechados/efeitos adversos , Poluição do Ar em Ambientes Fechados/análise , Pressão Sanguínea , Criança , Exposição Ambiental/análise , Monitoramento Ambiental , Humanos , Material Particulado/análise
7.
Front Endocrinol (Lausanne) ; 13: 821849, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35222279

RESUMO

Skeletal muscle accounts for ~80% of insulin-stimulated glucose uptake. The Group I p21-activated kinase 1 (PAK1) is required for the non-canonical insulin-stimulated GLUT4 vesicle translocation in skeletal muscle cells. We found that the abundances of PAK1 protein and its downstream effector in muscle, ARPC1B, are significantly reduced in the skeletal muscle of humans with type 2 diabetes, compared to the non-diabetic controls, making skeletal muscle PAK1 a candidate regulator of glucose homeostasis. Although whole-body PAK1 knockout mice exhibit glucose intolerance and are insulin resistant, the contribution of skeletal muscle PAK1 in particular was unknown. As such, we developed inducible skeletal muscle-specific PAK1 knockout (skmPAK1-iKO) and overexpression (skmPAK1-iOE) mouse models to evaluate the role of PAK1 in skeletal muscle insulin sensitivity and glucose homeostasis. Using intraperitoneal glucose tolerance and insulin tolerance testing, we found that skeletal muscle PAK1 is required for maintaining whole body glucose homeostasis. Moreover, PAK1 enrichment in GLUT4-myc-L6 myoblasts preserves normal insulin-stimulated GLUT4 translocation under insulin resistance conditions. Unexpectedly, skmPAK1-iKO also showed aberrant plasma insulin levels following a glucose challenge. By applying conditioned media from PAK1-enriched myotubes or myoblasts to ß-cells in culture, we established that a muscle-derived circulating factor(s) could enhance ß-cell function. Taken together, these data suggest that PAK1 levels in the skeletal muscle can regulate not only skeletal muscle insulin sensitivity, but can also engage in tissue crosstalk with pancreatic ß-cells, unveiling a new molecular mechanism by which PAK1 regulates whole-body glucose homeostasis.


Assuntos
Diabetes Mellitus Tipo 2 , Quinases Ativadas por p21 , Animais , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Homeostase , Camundongos , Músculo Esquelético/metabolismo , Transdução de Sinais , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
8.
Nat Commun ; 13(1): 424, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35058456

RESUMO

Mitochondrial dysfunction is implicated in skeletal muscle insulin resistance. Syntaxin 4 (STX4) levels are reduced in human diabetic skeletal muscle, and global transgenic enrichment of STX4 expression improves insulin sensitivity in mice. Here, we show that transgenic skeletal muscle-specific STX4 enrichment (skmSTX4tg) in mice reverses established insulin resistance and improves mitochondrial function in the context of diabetogenic stress. Specifically, skmSTX4tg reversed insulin resistance caused by high-fat diet (HFD) without altering body weight or food consumption. Electron microscopy of wild-type mouse muscle revealed STX4 localisation at or proximal to the mitochondrial membrane. STX4 enrichment prevented HFD-induced mitochondrial fragmentation and dysfunction through a mechanism involving STX4-Drp1 interaction and elevated AMPK-mediated phosphorylation at Drp1 S637, which favors fusion. Our findings challenge the dogma that STX4 acts solely at the plasma membrane, revealing that STX4 localises at/proximal to and regulates the function of mitochondria in muscle. These results establish skeletal muscle STX4 enrichment as a candidate therapeutic strategy to reverse peripheral insulin resistance.


Assuntos
Dinaminas/metabolismo , Exocitose , Resistência à Insulina , Dinâmica Mitocondrial , Músculo Esquelético/metabolismo , Proteínas Qa-SNARE/metabolismo , Adenilato Quinase/metabolismo , Animais , Respiração Celular , Ciclo do Ácido Cítrico , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dieta Hiperlipídica , Doxiciclina/farmacologia , Feminino , Glucose/metabolismo , Homeostase , Masculino , Metaboloma , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Músculo Esquelético/ultraestrutura , Especificidade de Órgãos , Fosforilação , Fosfosserina/metabolismo , Condicionamento Físico Animal
9.
Restor Dent Endod ; 46(4): e54, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34909418

RESUMO

OBJECTIVES: The purpose of this study was to quantify phase transformation after hydrofluoric acid (HF) etching at various concentrations on the surface of yttria-stabilized tetragonal zirconia polycrystal (Y-TZP), and to evaluate changes in bonding strength before and after thermal cycling. MATERIALS AND METHODS: A group whose Y-TZP surface was treated with tribochemical silica abrasion (TS) was used as the control. Y-TZP specimens from each experimental group were etched with 5%, 10%, 20%, and 40% HF solutions at room temperature for 10 minutes. First, to quantify the phase transformation, Y-TZP specimens (n = 5) treated with TS, 5%, 10%, 20% and 40% HF solutions were subjected to X-ray diffraction. Second, to evaluate the change in bond strength before and after thermal cycling, zirconia primer and MDP-containing resin cement were sequentially applied to the Y-TZP specimen. After 5,000 thermal cycles for half of the Y-TZP specimens, shear bond strength was measured for all experimental groups (n = 10). RESULTS: The monoclinic phase content in the 40% HF-treated group was higher than that of the 5%, 10%, and 20% HF-treated groups, but lower than that of TS-treated group (p < 0.05). The 40% HF-treated group showed significantly higher bonding strength than the TS, 5%, and 10% HF-treated groups, even after thermal cycling (p < 0.05). CONCLUSIONS: Through this experiment, the group treated with SiO2 containing air-borne abrasion on the Y-TZP surface showed higher phase transformation and higher reduction in bonding strength after thermal cycling compared to the group treated with high concentration HF.

10.
Diabetes ; 70(12): 2837-2849, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34556496

RESUMO

Syntaxin 4 (STX4), a plasma membrane-localized SNARE protein, regulates human islet ß-cell insulin secretion and preservation of ß-cell mass. We found that human type 1 diabetes (T1D) and NOD mouse islets show reduced ß-cell STX4 expression, consistent with decreased STX4 expression, as a potential driver of T1D phenotypes. To test this hypothesis, we generated inducible ß-cell-specific STX4-expressing NOD mice (NOD-ißSTX4). Of NOD-ißSTX4 mice, 73% had sustained normoglycemia vs. <20% of control NOD (NOD-Ctrl) mice by 25 weeks of age. At 12 weeks of age, before diabetes conversion, NOD-ißSTX4 mice demonstrated superior whole-body glucose tolerance and ß-cell glucose responsiveness than NOD-Ctrl mice. Higher ß-cell mass and reduced ß-cell apoptosis were also detected in NOD-ißSTX4 pancreata compared with pancreata of NOD-Ctrl mice. Single-cell RNA sequencing revealed that islets from NOD-ißSTX4 had markedly reduced interferon-γ signaling and tumor necrosis factor-α signaling via nuclear factor-κB in islet ß-cells, including reduced expression of the chemokine CCL5; CD4+ regulatory T cells were also enriched in NOD-ißSTX4 islets. These results provide a deeper mechanistic understanding of STX4 function in ß-cell protection and warrant further investigation of STX4 enrichment as a strategy to reverse or prevent T1D in humans or protect ß-cell grafts.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Células Secretoras de Insulina/metabolismo , Estado Pré-Diabético/genética , Proteínas Qa-SNARE/genética , Animais , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Especificidade de Órgãos/genética , Estado Pré-Diabético/imunologia , Proteínas Qa-SNARE/metabolismo
11.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673206

RESUMO

Type 2 diabetes (T2D) is one of the prominent causes of morbidity and mortality in the United States and beyond, reaching global pandemic proportions. One hallmark of T2D is dysfunctional glucose-stimulated insulin secretion from the pancreatic ß-cell. Insulin is secreted via the recruitment of insulin secretory granules to the plasma membrane, where the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and SNARE regulators work together to dock the secretory granules and release insulin into the circulation. SNARE proteins and their regulators include the Syntaxins, SNAPs, Sec1/Munc18, VAMPs, and double C2-domain proteins. Recent studies using genomics, proteomics, and biochemical approaches have linked deficiencies of exocytosis proteins with the onset and progression of T2D. Promising results are also emerging wherein restoration or enhancement of certain exocytosis proteins to ß-cells improves whole-body glucose homeostasis, enhances ß-cell function, and surprisingly, protection of ß-cell mass. Intriguingly, overexpression and knockout studies have revealed novel functions of certain exocytosis proteins, like Syntaxin 4, suggesting that exocytosis proteins can impact a variety of pathways, including inflammatory signaling and aging. In this review, we present the conventional and unconventional functions of ß-cell exocytosis proteins in normal physiology and T2D and describe how these insights might improve clinical care for T2D.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Exocitose , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas SNARE/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Humanos , Insulina/genética , Células Secretoras de Insulina/patologia , Proteínas SNARE/genética , Transdução de Sinais
12.
Diabetes ; 70(4): 889-902, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33526588

RESUMO

Enrichment of human islets with syntaxin 4 (STX4) improves functional ß-cell mass through a nuclear factor-κB (NF-κB)-dependent mechanism. However, the detailed mechanisms underlying the protective effect of STX4 are unknown. For determination of the signaling events linking STX4 enrichment and downregulation of NF-κB activity, STX4 was overexpressed in human islets, EndoC-ßH1 and INS-1 832/13 cells in culture, and the cells were challenged with the proinflammatory cytokines interleukin-1ß, tumor necrosis factor-α, and interferon-γ individually and in combination. STX4 expression suppressed cytokine-induced proteasomal degradation of IκBß but not IκBα. Inhibition of IKKß prevented IκBß degradation, suggesting that IKKß phosphorylates IκBß. Moreover, the IKKß inhibitor, as well as a proteosomal degradation inhibitor, prevented the loss of STX4 caused by cytokines. This suggests that STX4 may be phosphorylated by IKKß in response to cytokines, targeting STX4 for proteosomal degradation. Expression of a stabilized form of STX4 further protected IκBß from proteasomal degradation, and like wild-type STX4, stabilized STX4 coimmunoprecipitated with IκBß and the p50-NF-κB. This work proposes a novel pathway wherein STX4 regulates cytokine-induced NF-κB signaling in ß-cells via associating with and preventing IκBß degradation, suppressing chemokine expression, and protecting islet ß-cells from cytokine-mediated dysfunction and demise.


Assuntos
Proteínas Qa-SNARE/metabolismo , Western Blotting , Quimiocinas/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Imunoprecipitação , NF-kappa B/metabolismo , Oligopeptídeos/farmacologia , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
13.
Metabolism ; 115: 154431, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33181191

RESUMO

OBJECTIVE: p21 (Cdc42/Rac1) activated Kinase 1 (PAK1) is a candidate susceptibility factor for type 2 diabetes (T2D). PAK1 is depleted in the islets from T2D donors, compared to control individuals. In addition, whole-body PAK1 knock out (PAK1-KO) in mice worsens the T2D-like effects of high-fat diet. The current study tested the effects of modulating PAK1 levels only in ß-cells. MATERIALS/METHODS: ß-cell-specific inducible PAK1 KO (ßPAK1-iKO) mice were generated and used with human ß-cells and T2D islets to evaluate ß-cell function. RESULTS: ßPAK1-iKO mice exhibited glucose intolerance and elevated ß-cell apoptosis, but without peripheral insulin resistance. ß-cells from ßPAK-iKO mice also contained fewer mitochondria per cell. At the cellular level, human PAK1-deficient ß-cells showed blunted glucose-stimulated insulin secretion and reduced mitochondrial function. Mitochondria from human PAK1-deficient ß-cells were deficient in the electron transport chain (ETC) subunits CI, CIII, and CIV; NDUFA12, a CI complex protein, was identified as a novel PAK1 binding partner, and was significantly reduced with PAK1 knockdown. PAK1 knockdown disrupted the NAD+/NADH and NADP+/NADPH ratios, and elevated ROS. An imbalance of the redox state due to mitochondrial dysfunction leads to ER stress in ß-cells. PAK1 replenishment in the ß-cells of T2D human islets ameliorated levels of ER stress markers. CONCLUSIONS: These findings support a protective function for PAK1 in ß-cells. The results support a new model whereby the PAK1 in the ß-cell plays a required role upstream of mitochondrial function, via maintaining ETC protein levels and averting stress-induced ß-cell apoptosis to retain healthy functional ß-cell mass.


Assuntos
Apoptose/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Humanos , Resistência à Insulina/fisiologia , Secreção de Insulina/fisiologia , Camundongos , Camundongos Knockout , Oxirredução , Quinases Ativadas por p21/genética
14.
Diabetologia ; 62(5): 845-859, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30707251

RESUMO

AIMS/HYPOTHESIS: Skeletal muscle accounts for >80% of insulin-stimulated glucose uptake; dysfunction of this process underlies insulin resistance and type 2 diabetes. Insulin sensitivity is impaired in mice deficient in the double C2 domain ß (DOC2B) protein, while whole-body overexpression of DOC2B enhances insulin sensitivity. Whether insulin sensitivity in the skeletal muscle is affected directly by DOC2B or is secondary to an effect on other tissues is unknown; the underlying molecular mechanisms also remain unclear. METHODS: Human skeletal muscle samples from non-diabetic or type 2 diabetic donors were evaluated for loss of DOC2B during diabetes development. For in vivo analysis, new doxycycline-inducible skeletal-muscle-specific Doc2b-overexpressing mice fed standard or high-fat diets were evaluated for insulin and glucose tolerance, and insulin-stimulated GLUT4 accumulation at the plasma membrane (PM). For in vitro analyses, a DOC2B-overexpressing L6-GLUT4-myc myoblast/myotube culture system was coupled with an insulin resistance paradigm. Biochemical and molecular biology methods such as site-directed mutagenesis, co-immunoprecipitation and mass spectrometry were used to identify the molecular mechanisms linking insulin stimulation to DOC2B. RESULTS: We identified loss of DOC2B (55% reduction in RNA and 40% reduction in protein) in the skeletal muscle of human donors with type 2 diabetes. Furthermore, inducible enrichment of DOC2B in skeletal muscle of transgenic mice enhanced whole-body glucose tolerance (AUC decreased by 25% for female mice) and peripheral insulin sensitivity (area over the curve increased by 20% and 26% for female and male mice, respectively) in vivo, underpinned by enhanced insulin-stimulated GLUT4 accumulation at the PM. Moreover, DOC2B enrichment in skeletal muscle protected mice from high-fat-diet-induced peripheral insulin resistance, despite the persistence of obesity. In L6-GLUT4-myc myoblasts, DOC2B enrichment was sufficient to preserve normal insulin-stimulated GLUT4 accumulation at the PM in cells exposed to diabetogenic stimuli. We further identified that DOC2B is phosphorylated on insulin stimulation, enhancing its interaction with a microtubule motor protein, kinesin light chain 1 (KLC1). Mutation of Y301 in DOC2B blocked the insulin-stimulated phosphorylation of DOC2B and interaction with KLC1, and it blunted the ability of DOC2B to enhance insulin-stimulated GLUT4 accumulation at the PM. CONCLUSIONS/INTERPRETATION: These results suggest that DOC2B collaborates with KLC1 to regulate insulin-stimulated GLUT4 accumulation at the PM and regulates insulin sensitivity. Our observation provides a basis for pursuing DOC2B as a novel drug target in the muscle to prevent/treat type 2 diabetes.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Glucose/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Idoso , Animais , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica , Feminino , Regulação da Expressão Gênica , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 4/metabolismo , Humanos , Insulina/metabolismo , Resistência à Insulina , Cinesinas , Masculino , Camundongos , Pessoa de Meia-Idade , Ligação Proteica , Domínios Proteicos
15.
Diabetes ; 67(12): 2626-2639, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30305365

RESUMO

Syntaxin 4 (Stx4) enrichment in human and mouse islet grafts improves the success of transplants in reversing streptozotocin (STZ)-induced diabetes in mice, although the underlying molecular mechanisms remain elusive. Toward a further understanding of this, human islets and inducible transgenic mice that selectively overexpress Stx4 in islet ß-cells (ßTG-Stx4) were challenged with proinflammatory stressors in vitro and in vivo. Remarkably, ßTG-Stx4 mice resisted the loss of ß-cell mass and the glucose intolerance that multiple low doses of STZ induce. Under standard conditions, glucose tolerance was enhanced and mice maintained normal fasting glycemia and insulinemia. Conversely, Stx4 heterozygous knockout mice succumbed rapidly to STZ-induced glucose intolerance compared with their wild-type littermates. Human islet ß-cells overexpressing Stx4 exhibited enhanced insulin secretory capability; resilience against proinflammatory cytokine-induced apoptosis; and reduced expression of the CXCL9, CXCL10, and CXCL11 genes coordinate with decreased activation/nuclear localization of nuclear factor-κB. Finding ways to boost Stx4 expression presents a novel potential therapeutic avenue for promoting islet function and preserving ß-cell mass.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Intolerância à Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Qa-SNARE/metabolismo , Animais , Apoptose/fisiologia , Citocinas/metabolismo , Diabetes Mellitus Experimental/genética , Intolerância à Glucose/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas Qa-SNARE/genética
16.
Diabetes ; 67(7): 1332-1344, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29661782

RESUMO

Loss of functional ß-cell mass is an early feature of type 1 diabetes. To release insulin, ß-cells require soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes, as well as SNARE complex regulatory proteins like double C2 domain-containing protein ß (Doc2b). We hypothesized that Doc2b deficiency or overabundance may confer susceptibility or protection, respectively, to the functional ß-cell mass. Indeed, Doc2b+/- knockout mice show an unusually severe response to multiple-low-dose streptozotocin (MLD-STZ), resulting in more apoptotic ß-cells and a smaller ß-cell mass. In addition, inducible ß-cell-specific Doc2b-overexpressing transgenic (ßDoc2b-dTg) mice show improved glucose tolerance and resist MLD-STZ-induced disruption of glucose tolerance, fasting hyperglycemia, ß-cell apoptosis, and loss of ß-cell mass. Mechanistically, Doc2b enrichment enhances glucose-stimulated insulin secretion (GSIS) and SNARE activation and prevents the appearance of apoptotic markers in response to cytokine stress and thapsigargin. Furthermore, expression of a peptide containing the Doc2b tandem C2A and C2B domains is sufficient to confer the beneficial effects of Doc2b enrichment on GSIS, SNARE activation, and apoptosis. These studies demonstrate that Doc2b enrichment in the ß-cell protects against diabetogenic and proapoptotic stress. Furthermore, they identify a Doc2b peptide that confers the beneficial effects of Doc2b and may be a therapeutic candidate for protecting functional ß-cell mass.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Citoproteção/genética , Inflamação/genética , Células Secretoras de Insulina/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Pancreatite/genética , Animais , Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Feminino , Predisposição Genética para Doença , Inflamação/patologia , Inflamação/prevenção & controle , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Pancreatite/patologia , Pancreatite/prevenção & controle , Estreptozocina
17.
J Clin Endocrinol Metab ; 103(5): 1966-1976, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29506054

RESUMO

Context: Efforts to preserve ß-cell mass in the preclinical stages of type 1 diabetes (T1D) are limited by few blood-derived biomarkers of ß-cell destruction. Objective: Platelets are proposed sources of blood-derived biomarkers for a variety of diseases, and they show distinct proteomic changes in T1D. Thus, we investigated changes in the exocytosis protein, double C2 domain protein-ß (DOC2B) in platelets and islets from T1D humans, and prediabetic nonobese diabetic (NOD) mice. Design, Patients, and Main Outcome Measure: Protein levels of DOC2B were assessed in platelets and islets from prediabetic NOD mice and humans, with and without T1D. Seventeen new-onset T1D human subjects (10.3 ± 3.8 years) were recruited immediately following diagnosis, and platelet DOC2B levels were compared with 14 matched nondiabetic subjects (11.4 ± 2.9 years). Furthermore, DOC2B levels were assessed in T1D human pancreatic tissue samples, cytokine-stimulated human islets ex vivo, and platelets from T1D subjects before and after islet transplantation. Results: DOC2B protein abundance was substantially reduced in prediabetic NOD mouse platelets, and these changes were mirrored in the pancreatic islets from the same mice. Likewise, human DOC2B levels were reduced over twofold in platelets from new-onset T1D human subjects, and this reduction was mirrored in T1D human islets. Cytokine stimulation of normal islets reduced DOC2B expression ex vivo. Remarkably, platelet DOC2B levels increased after islet transplantation in patients with T1D. Conclusions: Reduction of DOC2B is an early feature of T1D, and DOC2B abundance may serve as a valuable in vivo indicator of ß-cell mass and an early biomarker of T1D.


Assuntos
Biomarcadores/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Diabetes Mellitus Tipo 1/diagnóstico , Proteínas do Tecido Nervoso/metabolismo , Adolescente , Animais , Plaquetas/metabolismo , Proteínas de Ligação ao Cálcio/fisiologia , Estudos de Casos e Controles , Contagem de Células , Criança , Diabetes Mellitus Tipo 1/metabolismo , Exocitose , Feminino , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Proteínas do Tecido Nervoso/fisiologia
18.
Diabetologia ; 59(10): 2145-55, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27394663

RESUMO

AIMS/HYPOTHESIS: Human islets from type 2 diabetic donors are reportedly 80% deficient in the p21 (Cdc42/Rac)-activated kinase, PAK1. PAK1 is implicated in beta cell function and maintenance of beta cell mass. We questioned the mechanism(s) by which PAK1 deficiency potentially contributes to increased susceptibility to type 2 diabetes. METHODS: Non-diabetic human islets and INS 832/13 beta cells cultured under diabetogenic conditions (i.e. with specific cytokines or under glucolipotoxic [GLT] conditions) were evaluated for changes to PAK1 signalling. Combined effects of PAK1 deficiency with GLT stress were assessed using classic knockout (Pak1 (-/-) ) mice fed a 45% energy from fat/palmitate-based, 'western' diet (WD). INS 832/13 cells overexpressing or depleted of PAK1 were also assessed for apoptosis and signalling changes. RESULTS: Exposure of non-diabetic human islets to diabetic stressors attenuated PAK1 protein levels, concurrent with increased caspase 3 cleavage. WD-fed Pak1 knockout mice exhibited fasting hyperglycaemia and severe glucose intolerance. These mice also failed to mount an insulin secretory response following acute glucose challenge, coinciding with a 43% loss of beta cell mass when compared with WD-fed wild-type mice. Pak1 knockout mice had fewer total beta cells per islet, coincident with decreased beta cell proliferation. In INS 832/13 beta cells, PAK1 deficiency combined with GLT exposure heightened beta cell death relative to either condition alone; PAK1 deficiency resulted in decreased extracellular signal-related kinase (ERK) and B cell lymphoma 2 (Bcl2) phosphorylation levels. Conversely, PAK1 overexpression prevented GLT-induced cell death. CONCLUSIONS/INTERPRETATION: These findings suggest that PAK1 deficiency may underlie an increased diabetic susceptibility. Discovery of ways to remediate glycaemic dysregulation via altering PAK1 or its downstream effectors offers promising opportunities for disease intervention.


Assuntos
Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Glicemia/metabolismo , Morte Celular/genética , Morte Celular/fisiologia , Linhagem Celular , Proliferação de Células/genética , Proliferação de Células/fisiologia , Diabetes Mellitus Tipo 2/genética , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Humanos , Hiperglicemia/genética , Hiperglicemia/metabolismo , Immunoblotting , Técnicas In Vitro , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Fosforilação/genética , Fosforilação/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Quinases Ativadas por p21/genética
20.
Cell Metab ; 22(3): 499-507, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26331606

RESUMO

Indirect evidence suggests that improved insulin sensitivity may contribute to improved lifespan of mice in which aging has been slowed by mutations, drugs, or dietary means, even in stocks of mice that do not show signs of late-life diabetes. Peripheral responses to insulin can be augmented by overexpression of Syntaxin 4 (Syn4), a plasma-membrane-localized SNARE protein. We show here that Syn4 transgenic (Tg) mice with high level expression of Syn4 had a significant extension of lifespan (33% increase in median) and showed increased peripheral insulin sensitivity, even at ages where controls exhibited age-related insulin resistance. Moreover, skeletal muscle GLUT4 and islet insulin granule exocytosis processes were fully protected in Syn4 Tg mice challenged with a high-fat diet. Hence, high-level expressing Syn4 Tg mice may exert better glycemic control, which slows multiple aspects of aging and extends lifespan, even in non-diabetic mice.


Assuntos
Envelhecimento , Glucose/metabolismo , Proteínas Qa-SNARE/genética , Regulação para Cima , Animais , Dieta Hiperlipídica , Feminino , Teste de Tolerância a Glucose , Insulina/metabolismo , Longevidade , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/genética , Obesidade/metabolismo , Proteínas Qa-SNARE/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...