Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Aging Neurosci ; 13: 665348, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34393754

RESUMO

AIM: Population based studies indicate a positive association between type 2 diabetes (T2D) and Parkinson's disease (PD) where there is an increased risk of developing PD in patients with T2D. PD is characterized by the abnormal accumulation of intraneuronal aggregated α-synuclein (α-syn) in Lewy bodies, which negatively impact neuronal viability. α-syn is also expressed in both pancreatic islets and skeletal muscle, key players in glucose regulation. Therefore, we examined the functional role of α-syn in these tissues. METHODS: Using mice lacking, overexpressing or transiently injected with α-syn, effects on glucose and insulin tolerance and insulin secretion were determined, with further characterization of the effects on GLUT4 translocation using GLUT4myc myotubes. RESULTS: Mice genetically ablated for α-syn became glucose intolerant and insulin resistant with hyperinsulinemia and reduced glucose-stimulated insulin secretion (GSIS). Mice overexpressing human α-syn are more insulin senstive and glucose tolerant compared to controls with increased GSIS. Injection of purified α-syn monomers also led to improved glucose tolerance and insulin sensitivity with hightened GSIS. α-syn monomer treatments increased surface GLUT4 levels in myotubes but without any significant change in Akt phosphorylation. The increase in cell surface GLUT4 was largely due to a large reduction in GLUT4 endocytosis, however, with a compensatory reduction in GLUT4 exocytosis. CONCLUSION: Cumulatively, this data suggests that α-syn modulates both pancreatic beta cell function and glucose transport in peripheral tissues, thereby playing a pivitol role in the maintenance of normal glucose homeostasis.

2.
JCI Insight ; 6(10)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34027899

RESUMO

GWAS have shown that the common R325W variant of SLC30A8 (ZnT8) increases the risk of type 2 diabetes (T2D). However, ZnT8 haploinsufficiency is protective against T2D in humans, counterintuitive to earlier work in humans and mouse models. Therefore, whether decreasing ZnT8 activity is beneficial or detrimental to ß cell function, especially under conditions of metabolic stress, remains unknown. In order to examine whether the existence of human islet amyloid polypeptide (hIAPP), a coresident of the insulin granule, affects the role of ZnT8 in regulating ß cell function, hIAPP-expressing transgenics were generated with reduced ZnT8 (ZnT8B+/- hIAPP) or null ZnT8 (ZnT8B-/- hIAPP) expression specifically in ß cells. We showed that ZnT8B-/- hIAPP mice on a high-fat diet had intensified amyloid deposition and further impaired glucose tolerance and insulin secretion compared with control, ZnT8B-/-, and hIAPP mice. This can in part be attributed to impaired glucose sensing and islet cell synchronicity. Importantly, ZnT8B+/- hIAPP mice were also glucose intolerant and had reduced insulin secretion and increased amyloid aggregation compared with controls. These data suggest that loss of or reduced ZnT8 activity in ß cells heightened the toxicity induced by hIAPP, leading to impaired ß cell function and glucose homeostasis associated with metabolic stress.


Assuntos
Amiloidose/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Transportador 8 de Zinco , Animais , Modelos Animais de Doenças , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Masculino , Camundongos , Camundongos Transgênicos , Transportador 8 de Zinco/genética , Transportador 8 de Zinco/metabolismo
3.
J Pathol ; 254(3): 244-253, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33797777

RESUMO

Amyloid plaques and neurofibrillary tangles composed of hyperphosphorylated tau are important contributors to Alzheimer's disease (AD). Tau also impacts pancreatic beta cell function and glucose homeostasis. Amyloid deposits composed of islet amyloid polypeptide (IAPP) are a pathological feature of type 2 diabetes (T2D). The current study investigates the role of human tau (hTau) in combination with human IAPP (hIAPP) as a potential mechanism connecting AD and T2D. Transgenic mice expressing hTau and hIAPP in the absence of murine tau were generated to determine the impact of these pathological factors on glucose metabolism. Co-expression of hIAPP and hTau resulted in mice with increased hyperglycaemia, insulin resistance, and glucose intolerance. The hTau-hIAPP mice also exhibited reduced beta cell area, increased amyloid deposition, impaired insulin processing, and reduced insulin content in islets. Tau phosphorylation also increased after stimulation with high glucose. In addition, brain insulin content and signalling were reduced, and tau phosphorylation was increased in these animals. These data support a link between tau and IAPP amyloid, which seems to act co-ordinately to impair beta cell function and glucose homeostasis, and suggest that the combined pathological actions of these proteins may be a potential mechanism connecting AD and T2D. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Proteínas tau/metabolismo , Animais , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/patologia , Intolerância à Glucose/metabolismo , Humanos , Hiperglicemia/metabolismo , Resistência à Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Transgênicos
4.
Artigo em Inglês | MEDLINE | ID: mdl-32226410

RESUMO

Microtubule-associated protein tau assists in stabilizing microtubules and has been particularly implicated in Alzheimer's disease (AD). Given the importance of tau to AD pathogenesis and therapies, it is important to understand non-classic physiological functions for this protein inside and outside the central nervous system (CNS). Our group has previously shown that tau ablation triggers glucose intolerance and pancreatic dysfunction in mice, suggesting that tau plays a role in peripheral metabolic regulation. Little is known about the role of tau in anxiety. Moreover, inconsistent results have been generated regarding the effects of tau deletion in memory. Here, we characterize systemic insulin resistance, anxiety-related behavior and memory in 15 to 20 weeks old Wild-Type (WT), Tau knockout (TauKO) and a distinct hTau mouse model consisting of tau knockout expressing the longest isoform (2N4R) of a non-mutant WT human Tau protein under the prion promoter (hTau). Our findings demonstrate that tau deletion leads to anxiety-related behavior, impaired contextual and cued fear memory. The presence of a human Tau transgene did not ameliorate the phenotypes observed in animals lacking the mouse tau protein and it elicited impairments in learning, memory, and peripheral insulin sensitivity. Our results suggest that tau protein plays a role in memory and anxiety-related behavior. Our findings also indicate that previously unrecognized functions for tau protein may be a complicating factor in using animal models on the TauKO background. Understanding the link between tau pathophysiology and cognitive and metabolic alterations is of great importance to establish the complete contribution of tau protein to AD pathogenesis.


Assuntos
Doença de Alzheimer , Comportamento Animal , Modelos Animais de Doenças , Camundongos Transgênicos , Proteínas tau/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Animais , Ansiedade/genética , Ansiedade/patologia , Ansiedade/fisiopatologia , Medo/fisiologia , Medo/psicologia , Feminino , Humanos , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Front Cell Neurosci ; 13: 17, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30804755

RESUMO

The microtubule-associated protein tau (MAPT) is mainly identified as a tubulin binding protein essential for microtubule dynamics and assembly and for neurite outgrowth. However, several other possible functions for Tau remains to be investigated. Insulin signaling is important for synaptic plasticity and memory formation and therefore is essential for proper brain function. Tau has recently been characterized as an important regulator of insulin signaling, with evidence linking Tau to brain and peripheral insulin resistance and beta cell dysfunction. In line with this notion, the hypothesis of Tau pathology as a key trigger of impaired insulin sensitivity and secretion has emerged. Conversely, insulin resistance can also favor Tau dysfunction, resulting in a vicious cycle of these events. In this review article, we discuss recent evidence linking Tau pathology, insulin resistance and insulin deficiency. We further highlight the deleterious consequences of Tau pathology-induced insulin resistance to the brain and/or peripheral tissues, suggesting that these are key events mediating cognitive decline in Alzheimer's disease (AD) and other tauopathies.

6.
FASEB J ; 32(6): 3166-3173, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29401605

RESUMO

The microtubule-associated protein tau is highly expressed in pancreatic islets. Abnormally phosphorylated tau aggregates assemble into neurofibrillary tangles linked to Alzheimer's disease pathology and has also been found in islets of patients with type 2 diabetes. However, the significance of tau in islet function remains relatively unexplored. Therefore, we investigated the role of tau on ß cell function and glucose homeostasis using tau knockout (tauKO) mice. TauKO mice were hyperglycemic and glucose intolerant at an early age. Islet insulin content was reduced and proinsulin levels were significantly elevated in tauKO mice, resulting in impaired glucose-stimulated insulin secretion. Loss of tau also resulted in increased epididymal fat mass and leptin levels, reduced glucose production, and insulin resistance at later ages, leading to complete onset of diabetes. Transgenic expression of human tau in islets was unable to rescue those defects in glucose regulation, indicating structural and/or functional differences between mouse and human tau. Cumulatively, these results suggest an important role for tau in the proper maintenance of pancreatic ß cell function and glucose homeostasis.-Wijesekara, N., Gonçalves, R. A., Ahrens, R., De Felice, F. G., Fraser, P. E. Tau ablation in mice leads to pancreatic ß cell dysfunction and glucose intolerance.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Intolerância à Glucose/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas tau/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Glucose/genética , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Humanos , Células Secretoras de Insulina/patologia , Leptina/genética , Leptina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proinsulina/genética , Proinsulina/metabolismo , Especificidade da Espécie , Proteínas tau/genética
7.
Neuropharmacology ; 136(Pt B): 172-181, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29169962

RESUMO

Alzheimer's disease (AD) is the most common type of dementia. Recent studies suggest that metabolic disturbances, particularly type 2 diabetes (T2D) increase the risk of cognitive decline and AD. AD is also a risk factor for T2D, and a growing body of evidence indicates that these diseases are connected both at clinical and molecular levels. In T2D, peripheral insulin resistance, hyperglycemia and eventually insulin deficiency develops, leading to an overall decline in tissue health. More recently, brain insulin resistance has been shown to be a key feature of AD that is linked to neuronal dysfunction and cognitive impairment. Furthermore, both AD and T2D are amyloidogenic diseases, with abnormal aggregation of amyloid-ß peptide (Aß) and islet amyloid polypeptide (IAPP) respectively contributing to cellular death and disease pathogenesis. Emerging data suggests that Aß may have peripheral effects including its co-deposition in the pancreas. In this review, we discuss how peripheral effects of Aß and metabolic disturbances may impact AD pathogenesis. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'


Assuntos
Doença de Alzheimer/metabolismo , Glucose/metabolismo , Homeostase/fisiologia , Animais , Humanos
8.
Sci Rep ; 7(1): 9766, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28852028

RESUMO

Growing evidence supports the hypothesis that type 2 diabetes (T2D) increases the risk of developing dementia. Experimental evidence from mouse models demonstrates that the induction of T2D/insulin resistance (IR) can promote the accumulation of Alzheimer's disease (AD) pathological features. However, the association of T2D with pathological and clinical phenotypes in humans is unclear. Here we investigate the relationship of indices of IR (HOMA-IR) and pancreatic ß-cell function (HOMA-B) with cognitive performance across several domains (Verbal/Visual Episodic Memory, Executive Function, Language and a measure of Global cognition) and AD biomarkers (CSF Aß42, T-tau/P-tau, hippocampal volume and neocortical Aß-amyloid burden). We reveal that HOMA-IR (p < 0.001) incrementally increases across diagnostic groups, becoming significantly elevated in the AD group compared with cognitively normal (CN) adults. In CN adults, higher HOMA-IR was associated with poorer performance on measures of verbal episodic memory (p = 0.010), executive function (p = 0.046) and global cognition (p = 0.007), as well as with higher CSF T-tau (p = 0.008) and P-tau (p = 0.014) levels. No association was observed with CSF Aß or imaging modalities. Together our data suggest that IR may contribute to reduced cognitive performance and the accumulation of CSF tau biomarkers in cognitively normal adults.


Assuntos
Cognição , Resistência à Insulina , Proteínas tau/líquido cefalorraquidiano , Adulto , Idoso , Idoso de 80 Anos ou mais , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Biomarcadores , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Masculino
9.
FASEB J ; 31(12): 5409-5418, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28808140

RESUMO

Alzheimer's disease (AD) and type 2 diabetes (T2D) present a significant risk to each other. AD and T2D are characterized by deposition of cerebral amyloid-ß (Aß) and pancreatic human islet amyloid polypeptide (hIAPP), respectively. We investigated the role of amyloidogenic proteins in the interplay between these diseases. A novel double transgenic mouse model combining T2D and AD was generated and characterized. AD-related amyloid transgenic mice coexpressing hIAPP displayed peripheral insulin resistance, hyperglycemia, and glucose intolerance. Aß and IAPP amyloid co-deposition increased tau phosphorylation, and a reduction in pancreatic ß-cell mass was detected in islets. Increased brain Aß deposition and tau phosphorylation and reduced insulin levels and signaling were accompanied by extensive synaptic loss and decreased neuronal counts. Aß immunization rescued the peripheral insulin resistance and hyperglycemia, suggesting a role for Aß in T2D pathogenesis for individuals predisposed to AD. These findings demonstrate that Aß and IAPP are key factors in the overlapping pathologies of AD and T2D.-Wijesekara, N., Ahrens, R., Sabale, M., Wu, L., Ha, K., Verdile, G., Fraser, P. E. Amyloid-ß and islet amyloid pathologies link Alzheimer's disease and type 2 diabetes in a transgenic model.


Assuntos
Doença de Alzheimer/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Western Blotting , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Feminino , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas tau/metabolismo
10.
J Alzheimers Dis ; 59(2): 421-432, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28269785

RESUMO

A wealth of evidence indicates a strong link between type 2 diabetes (T2D) and neurodegenerative diseases such as Alzheimer's disease (AD). Although the precise mechanism remains unclear, T2D can exacerbate neurodegenerative processes. Brain atrophy, reduced cerebral glucose metabolism, and central nervous system insulin resistance are features of both AD and T2D. The T2D phenotype (glucose dyshomeostasis, insulin resistance, impaired insulin signaling) also promotes AD pathology, namely accumulation of amyloid-ß (Aß) and hyperphosphorylated tau and can induce other aspects of neuronal degeneration including inflammatory and oxidative processes. Aß and hyperphosphorylated tau may also have roles in pancreatic ß-cell dysfunction and in reducing insulin sensitivity and glucose uptake by peripheral tissues such as liver, skeletal muscle, and adipose tissue. This suggests a role for these AD-related proteins in promoting T2D. The accumulation of the islet amyloid polypeptide (IAPP, or amylin) within islet ß-cells is a major pathological feature of the pancreas in patients with chronic T2D. Co-secreted with insulin, amylin accumulates over time and contributes to ß-cell toxicity, ultimately leading to reduced insulin secretion and onset of overt (insulin dependent) diabetes. Recent evidence also suggests that this protein accumulates in the brain of AD patients and may interact with Aß to exacerbate the neurodegenerative process. In this review, we highlight evidence indicating T2D in promoting Aß and tau mediated neurodegeneration and the potential contributions of Aß and tau in promoting a diabetic phenotype that could further exacerbate neurodegeneration. We also discuss underlying mechanisms by which amylin can contribute to the neurodegenerative processes.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Proteínas tau/metabolismo , Animais , Humanos , Proteínas tau/genética
11.
Diabetologia ; 59(6): 1242-6, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26970755

RESUMO

AIMS/HYPOTHESIS: Islet amyloid, a pathological feature of type 2 diabetes, forms from the aggregation of islet amyloid polypeptide (IAPP), a beta cell peptide that is produced and co-secreted with insulin. Cholesterol regulates amyloid-ß processing, deposition and clearance, promoting amyloidogenesis in the brain. ATP-binding cassette transporter 1 (ABCA1) is a cholesterol efflux transporter that when absent increases and when overexpressed reduces brain amyloid-ß deposition in mouse models of Alzheimer's disease. We examined whether alterations in ABCA1 expression and islet cholesterol content could also modulate islet amyloidogenesis. METHODS: Thioflavin S staining for amyloid was performed in islets isolated from mice with beta cell expression of human IAPP (hIAPP (Tg/o)) and cultured for 8 days following cholesterol loading, microRNA-33 overexpression (to reduce ABCA1 expression) or palmitate treatment in the presence or absence of ABCA1 overexpression or mevastatin treatment (to reduce cholesterol synthesis). hIAPP (Tg/o) mice were crossed with beta cell-specific Abca1-knockout mice (hIAPP (Tg/o) Abca1 (ßKO)) and glucose tolerance and amyloid formation were assessed. RESULTS: Cholesterol loading and microRNA-33-induced reduction in islet ABCA1 expression increased Thioflavin S-positive amyloid in hIAPP (Tg/o) islets. Palmitate treatment also increased amyloid formation and this was reduced by both ABCA1 overexpression and mevastatin treatment. hIAPP (Tg/o) Abca1 (ßKO) mice had increased islet cholesterol, accompanied by fasting hyperglycaemia, glucose intolerance, impaired in vivo insulin secretion and an increased islet proinsulin:insulin ratio. Amyloid area was increased in cultured hIAPP (Tg/o) Abca1 (ßKO) islets compared with hIAPP (Tg/o) controls. CONCLUSIONS/INTERPRETATION: These data suggest that elevations in islet cholesterol may lead to increases in IAPP aggregation and islet amyloid formation, further worsening beta cell function and glucose homeostasis.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/deficiência , Amiloidose/metabolismo , Amiloidose/patologia , Colesterol/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Amiloide/metabolismo , Animais , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Lovastatina/análogos & derivados , Lovastatina/farmacologia , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Palmitatos/farmacologia , Ratos
12.
Mediators Inflamm ; 2015: 105828, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26693205

RESUMO

Type 2 diabetes (T2DM), Alzheimer's disease (AD), and insulin resistance are age-related conditions and increased prevalence is of public concern. Recent research has provided evidence that insulin resistance and impaired insulin signalling may be a contributory factor to the progression of diabetes, dementia, and other neurological disorders. Alzheimer's disease (AD) is the most common subtype of dementia. Reduced release (for T2DM) and decreased action of insulin are central to the development and progression of both T2DM and AD. A literature search was conducted to identify molecular commonalities between obesity, diabetes, and AD. Insulin resistance affects many tissues and organs, either through impaired insulin signalling or through aberrant changes in both glucose and lipid (cholesterol and triacylglycerol) metabolism and concentrations in the blood. Although epidemiological and biological evidence has highlighted an increased incidence of cognitive decline and AD in patients with T2DM, the common molecular basis of cell and tissue dysfunction is rapidly gaining recognition. As a cause or consequence, the chronic inflammatory response and oxidative stress associated with T2DM, amyloid-ß (Aß) protein accumulation, and mitochondrial dysfunction link T2DM and AD.


Assuntos
Doença de Alzheimer/etiologia , Diabetes Mellitus Tipo 2/etiologia , Inflamação/complicações , Resistência à Insulina , Obesidade/etiologia , Estresse Oxidativo , Peptídeos beta-Amiloides/metabolismo , Animais , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Heme Oxigenase-1/análise , Humanos , NADP/metabolismo , eIF-2 Quinase/fisiologia
13.
Diabetes ; 64(4): 1168-79, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25338812

RESUMO

Besides their role in facilitating lipid absorption, bile acids are increasingly being recognized as signaling molecules that activate cell-signaling receptors. Targeted disruption of the sterol 12α-hydroxylase gene (Cyp8b1) results in complete absence of cholic acid (CA) and its derivatives. Here we investigate the effect of Cyp8b1 deletion on glucose homeostasis. Absence of Cyp8b1 results in improved glucose tolerance, insulin sensitivity, and ß-cell function, mediated by absence of CA in Cyp8b1(-/-) mice. In addition, we show that reduced intestinal fat absorption in the absence of biliary CA leads to increased free fatty acids reaching the ileal L cells. This correlates with increased secretion of the incretin hormone GLP-1. GLP-1, in turn, increases the biosynthesis and secretion of insulin from ß-cells, leading to the improved glucose tolerance observed in the Cyp8b1(-/-) mice. Thus, our data elucidate the importance of Cyp8b1 inhibition on the regulation of glucose metabolism.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/metabolismo , Homeostase/fisiologia , Resistência à Insulina/fisiologia , Insulina/metabolismo , Esteroide 12-alfa-Hidroxilase/metabolismo , Animais , Ácido Cólico/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/genética , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Esteroide 12-alfa-Hidroxilase/genética
14.
Arterioscler Thromb Vasc Biol ; 33(12): 2724-32, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24135019

RESUMO

OBJECTIVE: The ATP-binding cassette transporter A1 (ABCA1) protein maintains cellular cholesterol homeostasis in several different tissues. In the liver, ABCA1 is crucial for high-density lipoprotein biogenesis, and in the pancreas ABCA1 can regulate insulin secretion. In this study, our aim was to identify novel microRNAs that regulate ABCA1 expression in these tissues. APPROACH AND RESULTS: We combined multiple microRNA prediction programs to identify 8 microRNAs that potentially regulate ABCA1. A luciferase reporter assay demonstrated that 5 of these microRNAs (miR-148, miR-27, miR-144, miR-145, and miR-33a/33b) significantly repressed ABCA1 3'-untranslated region activity with miR-145 resulting in one of the larger decreases. In hepatic HepG2 cells, miR-145 can regulate both ABCA1 protein expression levels and cholesterol efflux function. In murine islets, an increase in miR-145 expression decreased ABCA1 protein expression, increased total islet cholesterol levels, and decreased glucose-stimulated insulin secretion. Inhibiting miR-145 produced the opposite effect of increasing ABCA1 protein levels and improving glucose-stimulated insulin secretion. Finally, increased glucose levels in media significantly decreased miR-145 levels in cultured pancreatic beta cells. These findings suggest that miR-145 is involved in glucose homeostasis and is regulated by glucose concentration. CONCLUSIONS: Our studies demonstrate that miR-145 regulates ABCA1 expression and function, and inhibiting this microRNA represents a novel strategy for increasing ABCA1 expression, promoting high-density lipoprotein biogenesis in the liver, and improving glucose-stimulated insulin secretion in islets.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , Hepatócitos/metabolismo , Ilhotas Pancreáticas/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Transportador 1 de Cassete de Ligação de ATP/genética , Animais , Sítios de Ligação , Colesterol/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Glucose/metabolismo , Células Hep G2 , Homeostase , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Lipoproteínas HDL/metabolismo , Camundongos , Transfecção
15.
Diabetes ; 61(3): 659-64, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22315310

RESUMO

Cellular cholesterol homeostasis is important for normal ß-cell function. Disruption of cholesterol transport by decreased function of the ATP-binding cassette (ABC) transporter ABCA1 results in impaired insulin secretion. Mice lacking ß-cell ABCA1 have increased islet expression of ABCG1, another cholesterol transporter implicated in ß-cell function. To determine whether ABCA1 and ABCG1 have complementary roles in ß-cells, mice lacking ABCG1 and ß-cell ABCA1 were generated and glucose tolerance, islet sterol levels, and ß-cell function were assessed. Lack of both ABCG1 and ß-cell ABCA1 resulted in increased fasting glucose levels and a greater impairment in glucose tolerance compared with either ABCG1 deletion or loss of ABCA1 in ß-cells alone. In addition, glucose-stimulated insulin secretion was decreased and sterol accumulation increased in islets lacking both transporters compared with those isolated from knockout mice with each gene alone. Combined deficiency of ABCA1 and ABCG1 also resulted in significant islet inflammation as indicated by increased expression of interleukin-1ß and macrophage infiltration. Thus, lack of both ABCA1 and ABCG1 induces greater defects in ß-cell function than deficiency of either transporter individually. These data suggest that ABCA1 and ABCG1 each make complimentary and important contributions to ß-cell function by maintaining islet cholesterol homeostasis in vivo.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Colesterol/metabolismo , Homeostase , Inflamação/etiologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Lipoproteínas/fisiologia , Transportador 1 de Cassete de Ligação de ATP , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Intolerância à Glucose/etiologia , Interleucina-1beta/genética , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição CHOP/fisiologia
16.
Diabetes ; 61(3): 653-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22315319

RESUMO

Changes in cellular cholesterol affect insulin secretion, and ß-cell-specific deletion or loss-of-function mutations in the cholesterol efflux transporter ATP-binding cassette transporter A1 (ABCA1) result in impaired glucose tolerance and ß-cell dysfunction. Upregulation of ABCA1 expression may therefore be beneficial for the maintenance of normal islet function in diabetes. Studies suggest that microRNA-33a (miR-33a) expression inversely correlates with ABCA1 expression in hepatocytes and macrophages. We examined whether miR-33a regulates ABCA1 expression in pancreatic islets, thereby affecting cholesterol accumulation and insulin secretion. Adenoviral miR-33a overexpression in human or mouse islets reduced ABCA1 expression, decreased glucose-stimulated insulin secretion, and increased cholesterol levels. The miR-33a-induced reduction in insulin secretion was rescued by cholesterol depletion by methyl-ß-cyclodextrin or mevastatin. Inhibition of miR-33a expression in apolipoprotein E knockout islets and ABCA1 overexpression in ß-cell-specific ABCA1 knockout islets rescued normal insulin secretion and reduced islet cholesterol. These findings confirm the critical role of ß-cell ABCA1 in islet cholesterol homeostasis and ß-cell function and highlight modulation of ß-cell miR-33a expression as a means to influence insulin secretion.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Colesterol/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , MicroRNAs/fisiologia , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/análise , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Glucose/farmacologia , Humanos , Secreção de Insulina , Camundongos , beta-Ciclodextrinas/farmacologia
17.
Diabetes ; 60(12): 3186-96, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21998401

RESUMO

OBJECTIVE: The ATP-binding cassette transporter A1 (ABCA1) is essential for normal insulin secretion from ß-cells. The aim of this study was to elucidate the mechanisms underlying the impaired insulin secretion in islets lacking ß-cell ABCA1. RESEARCH DESIGN AND METHODS: Calcium imaging, patch clamp, and membrane capacitance were used to assess the effect of ABCA1 deficiency on calcium flux, ion channel function, and exocytosis in islet cells. Electron microscopy was used to analyze ß-cell ultrastructure. The quantity and distribution of proteins involved in insulin-granule exocytosis were also investigated. RESULTS: We show that a lack of ß-cell ABCA1 results in impaired depolarization-induced exocytotic fusion of insulin granules. We observed disturbances in membrane microdomain organization and Golgi and insulin granule morphology in ß-cells as well as elevated fasting plasma proinsulin levels in mice in the absence of ß-cell ABCA1. Acute cholesterol depletion rescued the exocytotic defect in ß-cells lacking ABCA1, indicating that elevated islet cholesterol accumulation directly impairs granule fusion and insulin secretion. CONCLUSIONS: Our data highlight a crucial role of ABCA1 and cellular cholesterol in ß-cells that is necessary for regulated insulin granule fusion events. These data suggest that abnormalities of cholesterol metabolism may contribute to the impaired ß-cell function in diabetes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol/metabolismo , Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Western Blotting , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Eletrofisiologia , Exocitose/genética , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão
18.
J Biol Chem ; 285(44): 33623-31, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20709750

RESUMO

The functional impact of adiponectin on pancreatic beta cells is so far poorly understood. Although adiponectin receptors (AdipoR1/2) were identified, their involvement in adiponectin-induced signaling and other molecules involved is not clearly defined. Therefore, we investigated the role of adiponectin in beta cells and the signaling mediators involved. MIN6 beta cells and mouse islets were stimulated with globular (2.5 µg/ml) or full-length (5 µg/ml) adiponectin under serum starvation, and cell viability, proliferation, apoptosis, insulin gene expression, and secretion were measured. Lysates were subjected to Western blot analysis to determine phosphorylation of AMP-activated protein kinase (AMPK), Akt, or ERK. Functional significance of signaling was confirmed using dominant negative mutants or pharmacological inhibitors. Participation of AdipoRs was assessed by overexpression or siRNA. Adiponectin failed to activate AMPK after 10 min or 1- and 24-h stimulation. ERK was significantly phosphorylated after 24-h treatment with adiponectin, whereas Akt was activated at all time points examined. 24-h stimulation with adiponectin significantly increased cell viability by decreasing cellular apoptosis, and this was prevented by dominant negative Akt, wortmannin (PI3K inhibitor), and U0126 (MEK inhibitor). Moreover, adiponectin regulated insulin gene expression and glucose-stimulated insulin secretion, which was also prevented by wortmannin and U0126 treatment. Interestingly, the data also suggest adiponectin-induced changes in Akt and ERK phosphorylation and caspase-3 may occur independent of the level of AdipoR expression. This study demonstrates a lack of AMPK involvement and implicates Akt and ERK in adiponectin signaling, leading to protection against apoptosis and stimulation of insulin gene expression and secretion in pancreatic beta cells.


Assuntos
Adiponectina/metabolismo , Apoptose , Regulação da Expressão Gênica , Células Secretoras de Insulina/citologia , Insulina/biossíntese , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/patologia , Camundongos , Fosforilação , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Tempo
19.
Cell Metab ; 10(4): 285-95, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19808021

RESUMO

The Lkb1 tumor suppressor exerts its biological effects through phosphorylation and consequent activation of the AMP kinase (AMPK) family. Extensive genetic and biochemical evidence supports a role for Lkb1 in cell cycle arrest, establishment of cell polarity, and cellular energy metabolism. However, the role of Lkb1 and the AMPK family in beta cell function in vivo has not been established. We generated conditional knockout mice with a deletion of the Lkb1 gene in the beta cell compartment of pancreatic islets; these mice display improved glucose tolerance and protection against diet-induced hyperglycemia. Lkb1(-/-) beta cells are hypertrophic because of elevated mTOR activity; they also proliferate more and secrete more insulin in response to glucose. These data indicate that inhibiting Lkb1 activity in beta cells may facilitate beta cell expansion and glucose tolerance in vivo.


Assuntos
Glucose/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Cultivadas , Gorduras na Dieta , Antagonistas de Estrogênios/farmacologia , Teste de Tolerância a Glucose , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Tamoxifeno/farmacologia , Transgenes
20.
J Biol Chem ; 284(44): 30441-52, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19690348

RESUMO

Voltage-gated eag-related gene (Erg) K(+) channels regulate the electrical activity of many cell types. Data regarding Erg channel expression and function in electrically excitable glucagon and insulin producing cells of the pancreas is limited. In the present study Erg1 mRNA and protein were shown to be highly expressed in human and mouse islets and in alpha-TC6 and Min6 cells alpha- and beta-cell lines, respectively. Whole cell patch clamp recordings demonstrated the functional expression of Erg1 in alpha- and beta-cells, with rBeKm1, an Erg1 antagonist, blocking inward tail currents elicited by a double pulse protocol. Additionally, a small interference RNA approach targeting the kcnh2 gene (Erg1) induced a significant decrease of Erg1 inward tail current in Min6 cells. To investigate further the role of Erg channels in mouse and human islets, ratiometric Fura-2 AM Ca(2+)-imaging experiments were performed on isolated alpha- and beta-cells. Blocking Erg channels with rBeKm1 induced a transient cytoplasmic Ca(2+) increase in both alpha- and beta-cells. This resulted in an increased glucose-dependent insulin secretion, but conversely impaired glucagon secretion under low glucose conditions. Together, these data present Erg1 channels as new mediators of alpha- and beta-cell repolarization. However, antagonism of Erg1 has divergent effects in these cells; to augment glucose-dependent insulin secretion and inhibit low glucose stimulated glucagon secretion.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Células Secretoras de Glucagon/química , Células Secretoras de Insulina/química , Ilhotas Pancreáticas/citologia , Animais , Cálcio/metabolismo , Glucagon/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Potenciais da Membrana , Camundongos , Técnicas de Patch-Clamp
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA