Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Chemosphere ; 346: 140480, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37879369

RESUMO

Maternal exposure to particulate matter derived from diesel exhaust has been shown to cause metabolic dysregulation, neurological problems, and increased susceptibility to diabetes in the offspring. Diesel exhaust is a major source of air pollution and the use of biodiesel (BD) and its blends have been progressively increasing throughout the world; however, studies on the health impact of BD vs. petrodiesel combustion-generated exhaust have been controversial in part, due to differences in the chemical and physical nature of the associated particulate matter (PM). To explore the long-term impact of prenatal exposure, pregnant mice were exposed to PM generated by combustion of petrodiesel (B0) and a 20% soy BD blend (B20) by intratracheal instillation during embryonic days 9-17 and allowed to deliver. Offspring were then followed for 52 weeks. We found that mother's exposure to B0 and B20 PM manifested in striking sex-specific phenotypes with respect to metabolic adaptation, maintenance of glucose homeostasis, and medial hypothalamic glial cell makeup in the offspring. The data suggest PM exposure limited to a narrower critical developmental window may be compensated for by the mother and/or the fetus by altered metabolic programming in a marked sex-specific and fuel-derived PM-specific manner, leading to sex-specific risk for diseases related to environmental exposure later in life.


Assuntos
Poluentes Atmosféricos , Poluição do Ar , Masculino , Feminino , Camundongos , Animais , Material Particulado/toxicidade , Material Particulado/análise , Emissões de Veículos/toxicidade , Emissões de Veículos/análise , Biocombustíveis/toxicidade , Biocombustíveis/análise , Exposição Ambiental , Gasolina/análise , Poluentes Atmosféricos/toxicidade
2.
Br J Nutr ; 128(6): 1013-1028, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34605388

RESUMO

Metabolic impairments associated with type 2 diabetes, including insulin resistance and loss of glycaemic control, disproportionately impact the elderly. Lifestyle interventions, such as manipulation of dietary fat quality (i.e. fatty acid (FA) composition), have been shown to favourably modulate metabolic health. Yet, whether or not chronic consumption of beneficial FAs can protect against metabolic derangements and disease risk during ageing is not well defined. We sought to evaluate whether long-term dietary supplementation of fish-, dairy- or echium-derived FAs to the average FA profile in a U.S. American diet may offset metabolic impairments in males and females during ageing. One-month-old CD-1® mice were fed isoenergetic, high-fat (40 %) diets with the fat content composed of either 100 % control fat blend (CO) or 70 % CO with 30 % fish oil, dairy fat or echium oil for 13 months. Every 3 months, parameters of glucose homoeostasis were evaluated via glucose and insulin tolerance tests. Glucose tolerance improved in males consuming a diet supplemented with fish oil or echium oil as ageing progressed, but not in females. Yet, females were more metabolically protected than males regardless of age. Additionally, Spearman correlations were performed between indices of glucose homoeostasis and previously reported measurements of diet-derived FA content in tissues and colonic bacterial composition, which also revealed sex-specific associations. This study provides evidence that long-term dietary fat quality influences risk factors of metabolic diseases during ageing in a sex-dependent manner; thus, sex is a critical factor to be considered in future dietary strategies to mitigate type 2 diabetes risk.


Assuntos
Diabetes Mellitus Tipo 2 , Gorduras na Dieta , Camundongos , Masculino , Feminino , Animais , Óleos de Peixe , Suplementos Nutricionais , Glucose
3.
J Biol Chem ; 297(5): 101250, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34592314

RESUMO

Loss of functional pancreatic ß-cell mass leads to type 2 diabetes (T2D), attributable to modified ß-cell-dependent adaptive gene expression patterns. SetD7 is a histone methyltransferase enriched in pancreatic islets that mono- and dimethylates histone-3-lysine-4 (H3K4), promoting euchromatin modifications, and also maintains the regulation of key ß-cell function and survival genes. However, the transcriptional regulation of this important epigenetic modifier is unresolved. Here we identified the nuclear hormone receptor peroxisome proliferator-activated receptor-gamma (PPARγ) as a major transcriptional regulator of SetD7 and provide evidence for direct binding and functionality of PPARγ in the SetD7 promoter region. Furthermore, constitutive shRNA-mediated PPARγ knockdown in INS-1 ß-cells or pancreas-specific PPARγ deletion in mice led to downregulation of SetD7 expression as well as its nuclear enrichment. The relevance of the SetD7-PPARγ interaction in ß-cell adaptation was tested in normoglycemic 60% partial pancreatectomy (Px) and hyperglycemic 90% Px rat models. Whereas a synergistic increase in islet PPARγ and SetD7 expression was observed upon glycemic adaptation post-60% Px, in hyperglycemic 90% Px rats, islet PPARγ, and PPARγ targets SetD7 and Pdx1 were downregulated. PPARγ agonist pioglitazone treatment in 90% Px rats partially restored glucose homeostasis and ß-cell mass and enhanced expression of SetD7 and Pdx1. Collectively, these data provide evidence that the SetD7-PPARγ interaction serves as an important element of the adaptive ß-cell response.


Assuntos
Regulação Enzimológica da Expressão Gênica , Histona-Lisina N-Metiltransferase/biossíntese , Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , PPAR gama/metabolismo , Elementos de Resposta , Animais , Linhagem Celular , Histona-Lisina N-Metiltransferase/genética , Hiperglicemia/genética , Camundongos , Camundongos Transgênicos , PPAR gama/genética , Ratos
4.
J Nutr ; 151(7): 1779-1790, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33982087

RESUMO

BACKGROUND: There is currently no consensus on which tissues are optimal for assessing specific diet-derived fatty acids (FAs) as biomarkers for long-term dietary studies. OBJECTIVES: This study measured the content of unique diet-derived FAs from dairy, echium, and fish in tissues (adipose, muscle, liver, erythrocyte membranes, and plasma phospholipids, cholesterol esters, triglycerides, and free fatty acids) after long-term feeding in CD-1 mice. METHODS: Beginning at weaning, mice (n = 10-11/sex/diet) were fed 1 of 4 diets (40% kcal/total energy) that only differed in FA composition: control fat blend (CON), reflecting the FA profile of the average US American diet, or CON supplemented with 30% of fish oil (FO), dairy fat (DF), or echium oil (EO). After 13 mo, tissues were collected to determine FAs via gas-liquid chromatography. Tissue FAs were analyzed via 2-factor ANOVA, and relationships between FA intake and tissue content were assessed with Spearman correlations. RESULTS: As anticipated, 20:5n-3 (ω-3) tissue content was ≤32-fold greater in FO- compared with CON-fed mice (P < 0.05). In addition, 20:5n-3 intake strongly correlated with its content in all tissues (ρ = 0.67-0.76; P < 0.05). Echium oil intake also influenced tissue FA content in mice as expected. For example, 18:3n-6 was ≤25-fold greater in adipose, muscle, and liver tissues of EO-fed compared with CON-fed mice (P < 0.05). Tissue content of FAs typically considered biomarkers of dairy fat intake (15:0, 16:1 t9, and 17:0) was often not greater in mice fed DF than other diet groups, although 18:2 c9, t11 content was ≤6-fold greater in tissues from DF-fed compared with CON-fed mice (P < 0.05). The content of dairy-derived FAs in blood fractions of females was up to 2-fold greater compared with males, whereas docosapentaenoic acid content was up to 1-fold greater in all blood fractions and in liver tissue of males compared with females (P < 0.05). In adipose, muscle, and liver tissue, the content of γ-linolenic acid and stearidonic acid was less than 1-fold greater in females than in males (P < 0.05). CONCLUSIONS: Our study indicates that the distribution of dietary FAs is tissue and sex dependent in aged CD-1 mice. Research using FA biomarkers should assess a combination of FA biomarkers to accurately validate patterns of FA intake and source.


Assuntos
Ácidos Graxos , Óleos de Peixe , Animais , Biomarcadores , Dieta , Suplementos Nutricionais , Feminino , Masculino , Camundongos
5.
PLoS One ; 15(9): e0238893, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32956361

RESUMO

Utilization of murine models remains a valuable tool in biomedical research, yet, disease phenotype of mice across studies can vary considerably. With advances in next generation sequencing, it is increasingly recognized that inconsistencies in host phenotype can be attributed, at least in part, to differences in gut bacterial composition. Research with inbred murine strains demonstrates that housing conditions play a significant role in variations of gut bacterial composition, however, few studies have assessed whether observed variation influences host phenotype in response to an intervention. Our study initially sought to examine the effects of a long-term (9-months) dietary intervention (i.e., diets with distinct fatty acid compositions) on the metabolic health, in particular glucose homeostasis, of genetically-outbred male and female CD-1 mice. Yet, mice were shipped from two different husbandry facilities of the same commercial vendor (Cohort A and B, respectively), and we observed throughout the study that diet, sex, and aging differentially influenced the metabolic phenotype of mice depending on their husbandry facility of origin. Examination of the colonic bacteria of mice revealed distinct bacterial compositions, including 23 differentially abundant genera and an enhanced alpha diversity in mice of Cohort B compared to Cohort A. We also observed that a distinct metabolic phenotype was linked with these differentially abundant bacteria and indices of alpha diversity. Our findings support that metabolic phenotypic variation of mice of the same strain but shipped from different husbandry facilities may be influenced by their colonic bacterial community structure. Our work is an important precautionary note for future research of metabolic diseases via mouse models, particularly those that seek to examine factors such diet, sex, and aging.


Assuntos
Bactérias/classificação , Dieta/efeitos adversos , Fezes/microbiologia , Glucose/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Camundongos Endogâmicos/genética , Criação de Animais Domésticos , Animais , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/isolamento & purificação , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Modelos Animais , Fenótipo , Filogenia , Análise de Sequência de DNA
6.
PLoS One ; 14(12): e0226635, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31851713

RESUMO

Evidence suggests that sex influences the effect of diet on the gut bacterial composition, yet, no studies have been performed assessing dietary fatty acid composition (i.e., fat quality) in this context. This study examined the effect of dietary fat quality on colonic bacterial composition in an aged, genetically-diverse mouse population. CD-1 mice were fed isoenergetic diets consisting of (1) control fat (CO; "Western-style" fat blend), (2) CO supplemented with 30% fish oil, (3) CO supplemented with 30% dairy fat, or (4) CO supplemented with 30% echium oil. Fecal samples were collected at mid-life and aged (reproductively senescent) time points. Overall, the abundance of Bacteroidetes was greater in mice fed echium oil compared to mice fed the control fat. Examination of colonic bacterial relative abundance also revealed sex differences, with 73 bacterial taxa being differentially expressed in males and females. Notably, results showed a strong interactive effect among the diet, sex, and age of mice which influenced colonic bacterial relative abundance and alpha diversity. In males, supplementation of the diet with dairy fat or echium oil caused the abundance of Bacteroidetes and Bacteroides to change with age. Additionally, supplementation of the diet with fish oil induced sex-dependent changes in the alpha diversity of aged mice compared to mid-life. This work supports that sex is a critical factor in colonic bacterial composition of an aged, genetically-heterogenous population. Moreover, this study establishes that the effectiveness of dietary interventions for health maintenance and disease prevention via direct or indirect manipulation of the gut microbiota is likely dependent on an individual's sex, age, and genetic background.


Assuntos
Colo/microbiologia , Gorduras na Dieta/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Fatores Etários , Animais , Bacteroides/efeitos dos fármacos , Bacteroides/crescimento & desenvolvimento , Feminino , Óleos de Peixe/farmacologia , Masculino , Camundongos , Óleos de Plantas/farmacologia , Fatores Sexuais
7.
J Biol Chem ; 293(52): 20295-20306, 2018 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-30397183

RESUMO

Although it is well-established how nutrients, growth factors, and hormones impact functional ß-cell mass (BCM), the influence of the central nervous system in this regard, and especially in the context of islet immune modulation, has been understudied. Here we investigated the expression and activity of pancreatic islet α7 nicotinic acetylcholine receptor (α7nAChR) in islet anti-inflammatory and prosurvival signaling. Systemic administration of α7nAChR agonists in mice improved glucose tolerance and curtailed streptozotocin-induced hyperglycemia by retaining BCM, in part through maintaining Pdx1 and MafA expression and reducing apoptosis. α7nAChR activation of mouse islets ex vivo led to reduced inflammatory drive through a JAK2-STAT3 pathway that couples with CREB/Irs2/Akt survival signaling. Because the vagus nerve conveys anti-inflammatory signals to immune cells of the spleen and other nonneural tissues in the viscera by activating α7nAChR agonists, our study suggests a novel role for ß-cell α7nAChR that functions to maintain ß-cell survival and mass homeostasis through modulating islet cytokine and phosphatidylinositol 3-kinase-dependent signaling pathways. Exploiting these pathways may have therapeutic potential for the treatment of autoimmune diabetes.


Assuntos
Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Transdução de Sinais , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hiperglicemia/induzido quimicamente , Hiperglicemia/genética , Hiperglicemia/patologia , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estreptozocina/toxicidade , Receptor Nicotínico de Acetilcolina alfa7/genética
8.
J Biol Chem ; 292(30): 12449-12459, 2017 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-28487366

RESUMO

The onset of type 2 diabetes is characterized by transition from successful to failed insulin secretory compensation to obesity-related insulin resistance and dysmetabolism. Energy-rich diets in rodents are commonly studied models of compensatory increases in both insulin secretion and ß cell mass. However, the mechanisms of these adaptive responses are incompletely understood, and it is also unclear why these responses eventually fail. We measured the temporal trends of glucose homeostasis, insulin secretion, ß cell morphometry, and islet gene expression in C57BL/6NTac mice fed a 60% high-fat diet (HFD) or control diet for up to 16 weeks. A 2-fold increased hyperinsulinemia was maintained for the first 4 weeks of HFD feeding and then further increased through 16 weeks. ß cell mass increased progressively starting at 4 weeks, principally through nonproliferative growth. Insulin sensitivity was not significantly perturbed until 11 weeks of HFD feeding. Over the first 8 weeks, we observed two distinct waves of increased expression of ß cell functional and prodifferentiation genes. This was followed by activation of the unfolded protein response at 8 weeks and overt ß cell endoplasmic reticulum stress at 12-16 weeks. In summary, ß cell adaptation to an HFD in C57BL/6NTac mice entails early insulin hypersecretion and a robust growth phase along with hyperexpression of related genes that begin well before the onset of observed insulin resistance. However, continued HFD exposure results in cessation of gene hyperexpression, ß cell functional failure, and endoplasmic reticulum stress. These data point to a complex but not sustainable integration of ß cell-adaptive responses to nutrient overabundance, obesity development, and insulin resistance.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hiperinsulinismo/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Estresse do Retículo Endoplasmático , Hiperinsulinismo/patologia , Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Fatores de Tempo
9.
Front Physiol ; 4: 184, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23888144

RESUMO

Decreased peroxisome proliferator-activated receptor gamma (PPARγ) activity is thought to have a major role in preeclampsia through abnormal placental development. However, the role of PPARγ in adaptation of the uteroplacental vasculature that may lead to placental hypoperfusion and fetal growth restriction during pregnancy is not known. Here, pregnant Sprague-Dawley rats (n = 11/group) were treated during the second half of pregnancy with the PPARγ inhibitor GW9662 (10 mg/kg/day in food) or vehicle. Pregnancy outcome and PPARγ mRNA, vasodilation and structural remodeling were determined in maternal uterine and mesenteric arteries. PPARγ was expressed in uterine vascular tissue of both non-pregnant and pregnant rats with ~2-fold greater expression in radial vs. main uterine arteries. PPARγ mRNA levels were significantly higher in uterine compared to mesenteric arteries. GW9662 treatment during pregnancy did not affect maternal physiology (body weight, glucose, blood pressure), mesenteric artery vasodilation or structural remodeling of uterine and mesenteric vessels. Inhibition of PPARγ for the last 10 days of gestation caused decreased fetal weights on both day 20 and 21 of gestation that was associated with impaired vasodilation of radial uterine arteries in response to acetylcholine and sodium nitroprusside. These results define an essential role of PPARγ in the control of uteroplacental vasodilatory function during pregnancy, an important determinant of blood flow to the placenta and fetus. Strategies that target PPARγ activation in the uterine circulation could have important therapeutic potential in treatment of pregnancies complicated by hypertension, diabetes or preeclampsia.

10.
J Biol Chem ; 288(35): 25440-25449, 2013 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-23788637

RESUMO

The molecular mechanisms and signaling pathways that drive islet ß-cell compensation and failure are not fully resolved. We have used in vitro and in vivo systems to show that FoxO1, an integrator of metabolic stimuli, inhibits PPARγ expression in ß-cells, thus transcription of its target genes (Pdx1, glucose-dependent insulinotropic polypeptide (GIP) receptor, and pyruvate carboxylase) that are important regulators of ß-cell function, survival, and compensation. FoxO1 inhibition of target gene transcription is normally relieved when upstream activation induces its translocation from the nucleus to the cytoplasm. Attesting to the central importance of this pathway, islet expression of PPARγ and its target genes was enhanced in nondiabetic insulin-resistant rats and markedly reduced with diabetes induction. Insight into the impaired PPARγ signaling with hyperglycemia was obtained with confocal microscopy of pancreas sections that showed an intense nuclear FoxO1 immunostaining pattern in the ß-cells of diabetic rats in contrast to the nuclear and cytoplasmic FoxO1 in nondiabetic rats. These findings suggest a FoxO1/PPARγ-mediated network acting as a core component of ß-cell adaptation to metabolic stress, with failure of this response from impaired FoxO1 activation causing or exacerbating diabetes.


Assuntos
Núcleo Celular/metabolismo , Diabetes Mellitus Experimental/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , PPAR gama/metabolismo , Transcrição Gênica , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/patologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , PPAR gama/genética , Ratos , Ratos Zucker
11.
Endocrinology ; 152(4): 1300-13, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21285317

RESUMO

Circulating levels of matrix metalloproteinases (MMPs) and their endogenous inhibitors, tissue inhibitor of metalloproteinases (TIMPs), are altered in human obesity and may contribute to its pathology. TIMP-2 exerts MMP-dependent (MMP inhibition and pro-MMP-2 activation) and MMP-independent functions. To assess the role of TIMP-2 in a murine model of nutritionally induced obesity, weight gain in wild-type and TIMP-2 deficient [knockout (KO)] mice fed a chow or high-fat diet (HFD) was determined. The effects of diet on glucose tolerance and insulin sensitivity, as well as pancreatic ß-cell and adipocyte physiology, were assessed. Chow-fed TIMP-2 KO mice of both sexes became obese but maintained relatively normal glucose tolerance and insulin sensitivity. Obesity was exacerbated on the HFD. However, HFD-fed male, but not female, TIMP-2 KO mice developed insulin resistance with reduced glucose transporter 2 and pancreatic and duodenal homeobox 1 levels, despite increased ß-cell mass and hyperplasia. Thus, although ß-cell mass was increased, HFD-fed male TIMP-2 KO mice develop diabetes likely due to ß-cell exhaustion and failure. TIMP-2 mRNA, whose expression was greatest in sc adipose tissue, was down-regulated in HFD-fed wild-type males, but not females. Furthermore, HFD increased membrane type 1-MMP (MMP-14) expression and activity in male, but not female, sc adipose tissue. Strikingly, MMP-14 expression increased to a greater extent in TIMP-2 KO males and was associated with decreased adipocyte collagen. Taken together, these findings demonstrate a role for TIMP-2 in maintaining extracellular matrix integrity necessary for normal ß-cell and adipocyte physiology and that loss of extracellular matrix integrity may underlie diabetic and obesogenic phenotypes.


Assuntos
Obesidade/metabolismo , Inibidor Tecidual de Metaloproteinase-2/deficiência , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Western Blotting , Gorduras na Dieta/efeitos adversos , Feminino , Técnicas Imunoenzimáticas , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Leptina/metabolismo , Masculino , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia de Fluorescência , Obesidade/induzido quimicamente , Reação em Cadeia da Polimerase , Fatores Sexuais , Inibidor Tecidual de Metaloproteinase-2/genética , Aumento de Peso/genética
12.
J Biol Chem ; 285(44): 33718-26, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20736162

RESUMO

It remains unclear how α-ketoisocaproate (KIC) and leucine are metabolized to stimulate insulin secretion. Mitochondrial BCATm (branched-chain aminotransferase) catalyzes reversible transamination of leucine and α-ketoglutarate to KIC and glutamate, the first step of leucine catabolism. We investigated the biochemical mechanisms of KIC and leucine-stimulated insulin secretion (KICSIS and LSIS, respectively) using BCATm(-/-) mice. In static incubation, BCATm disruption abolished insulin secretion by KIC, D,L-α-keto-ß-methylvalerate, and α-ketocaproate without altering stimulation by glucose, leucine, or α-ketoglutarate. Similarly, during pancreas perfusions in BCATm(-/-) mice, glucose and arginine stimulated insulin release, whereas KICSIS was largely abolished. During islet perifusions, KIC and 2 mM glutamine caused robust dose-dependent insulin secretion in BCATm(+/+) not BCATm(-/-) islets, whereas LSIS was unaffected. Consistently, in contrast to BCATm(+/+) islets, the increases of the ATP concentration and NADPH/NADP(+) ratio in response to KIC were largely blunted in BCATm(-/-) islets. Compared with nontreated islets, the combination of KIC/glutamine (10/2 mM) did not influence α-ketoglutarate concentrations but caused 120 and 33% increases in malate in BCATm(+/+) and BCATm(-/-) islets, respectively. Although leucine oxidation and KIC transamination were blocked in BCATm(-/-) islets, KIC oxidation was unaltered. These data indicate that KICSIS requires transamination of KIC and glutamate to leucine and α-ketoglutarate, respectively. LSIS does not require leucine catabolism and may be through leucine activation of glutamate dehydrogenase. Thus, KICSIS and LSIS occur by enhancing the metabolism of glutamine/glutamate to α-ketoglutarate, which, in turn, is metabolized to produce the intracellular signals such as ATP and NADPH for insulin secretion.


Assuntos
Cetoácidos/química , Leucina/química , Mitocôndrias/enzimologia , Transaminases/genética , Trifosfato de Adenosina/química , Animais , Feminino , Glucose/química , Glucose/metabolismo , Glutamina/química , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ácidos Cetoglutáricos/química , Camundongos , Camundongos Transgênicos , Oxigênio/química , Transaminases/metabolismo
13.
Am J Physiol Endocrinol Metab ; 299(5): E786-93, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20716695

RESUMO

The physiological mechanisms that preserve pancreatic ß-cell mass (BCM) are not fully understood. Although the regulation of islet function by the autonomic nervous system (ANS) is well established, its potential roles in BCM homeostasis and compensatory growth have not been adequately explored. The parasympathetic vagal branch of the ANS serves to facilitate gastrointestinal function, metabolism, and pancreatic islet regulation of glucose homeostasis, including insulin secretion. Given the functional importance of the vagus nerve and its branches to the liver, gut, and pancreas in control of digestion, motility, feeding behavior, and glucose metabolism, it may also play a role in BCM regulation. We have begun to examine the potential roles of the parasympathetic nervous system in short-term BCM maintenance by performing a selective bilateral celiac branch-vagus nerve transection (CVX) in normal Sprague-Dawley rats. CVX resulted in no detectable effects on basic metabolic parameters or food intake through 1 wk postsurgery. Although there were no differences in BCM or apoptosis in this 1-wk time frame, ß-cell proliferation was reduced 50% in the CVX rats, correlating with a marked reduction in activated protein kinase B/Akt. Unexpectedly, acinar proliferation was increased 50% in these rats. These data suggest that the ANS, via the vagus nerve, contributes to the regulation of BCM maintenance at the level of cell proliferation and may also mediate the drive for enhanced growth under physiological conditions when insulin requirements have increased. Furthermore, the disparate effects of CVX on ß-cell and acinar cells suggest that the endocrine and exocrine pancreas respond to different neural signals in regard to mass homeostasis.


Assuntos
Células Secretoras de Insulina/fisiologia , Nervo Vago/fisiologia , Animais , Apoptose/fisiologia , Glicemia/análise , Peso Corporal/fisiologia , Processos de Crescimento Celular/fisiologia , Ingestão de Líquidos/fisiologia , Ingestão de Alimentos/fisiologia , Peptídeo 1 Semelhante ao Glucagon/sangue , Teste de Tolerância a Glucose , Insulina/sangue , Masculino , Microscopia Confocal , Ratos , Ratos Sprague-Dawley , Nervo Vago/cirurgia , Nervo Vago/ultraestrutura
14.
Diabetes ; 59(9): 2178-87, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20547980

RESUMO

OBJECTIVE: C57Bl/6 mice develop obesity and mild hyperglycemia when fed a high-fat diet (HFD). Although diet-induced obesity (DIO) is a widely studied model of type 2 diabetes, little is known about beta-cell failure in these mice. RESEARCH DESIGN AND METHODS: DIO mice were separated in two groups according to body weight gain: low- and high-HFD responders (LDR and HDR). We examined whether mild hyperglycemia in HDR mice is due to reduced beta-cell mass or function and studied islet metabolism and signaling. RESULTS: HDR mice were more obese, hyperinsulinemic, insulin resistant, and hyperglycemic and showed a more altered plasma lipid profile than LDR. LDR mice largely compensated insulin resistance, whereas HDR showed perturbed glucose homeostasis. Neither LDR nor HDR mice showed reduced beta-cell mass, altered islet glucose metabolism, and triglyceride deposition. Insulin secretion in response to glucose, KCl, and arginine was impaired in LDR and almost abolished in HDR islets. Palmitate partially restored glucose- and KCl-stimulated secretion. The glucose-induced rise in ATP was reduced in both DIO groups, and the glucose-induced rise in Ca(2+) was reduced in HDR islets relatively to LDR. Glucose-stimulated lipolysis was decreased in LDR and HDR islets, whereas fat oxidation was increased in HDR islets only. Fatty acid esterification processes were markedly diminished, and free cholesterol accumulated in HDR islets. CONCLUSIONS: beta-Cell failure in HDR mice is not due to reduced beta-cell mass and glucose metabolism or steatosis but to a secretory dysfunction that is possibly due to altered ATP/Ca(2+) and lipid signaling, as well as free cholesterol deposition.


Assuntos
Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Obesidade/fisiopatologia , Aumento de Peso/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Divisão Celular , Dieta , Gorduras na Dieta/efeitos adversos , Glucose/metabolismo , Técnica Clamp de Glucose , Insulina/sangue , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/patologia , Lipólise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/induzido quimicamente , Obesidade/etiologia , Proinsulina/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Diabetes ; 59(6): 1445-50, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20332343

RESUMO

OBJECTIVE: We previously showed that peroxisome proliferator-activated receptor (PPAR)-gamma in beta-cells regulates pdx-1 transcription through a functional PPAR response element (PPRE). Gene Bank blast for a homologous nucleotide sequence revealed the same PPRE within the rat glucose-dependent insulinotropic polypeptide receptor (GIP-R) promoter sequence. We investigated the role of PPARgamma in GIP-R transcription. RESEARCH DESIGN AND METHODS: Chromatin immunoprecipitation assay, siRNA, and luciferase gene transcription assay in INS-1 cells were performed. Islet GIP-R expression and immunohistochemistry studies were performed in pancreas-specific PPARgamma knockout mice (PANC PPARgamma(-/-)), normoglycemic 60% pancreatectomy rats (Px), normoglycemic and hyperglycemic Zucker fatty (ZF) rats, and mouse islets incubated with troglitazone. RESULTS: In vitro studies of INS-1 cells confirmed that PPAR-gamma binds to the putative PPRE sequence and regulates GIP-R transcription. In vivo verification was shown by a 70% reduction in GIP-R protein expression in islets from PANC PPARgamma(-/-) mice and a twofold increase in islets of 14-day post-60% Px Sprague-Dawley rats that hyperexpress beta-cell PPARgamma. Thiazolidinedione activation (72 h) of this pathway in normal mouse islets caused a threefold increase of GIP-R protein and a doubling of insulin secretion to 16.7 mmol/l glucose/10 nmol/l GIP. Islets from obese normoglycemic ZF rats had twofold increased PPARgamma and GIP-R protein levels versus lean rats, with both lowered by two-thirds in ZF rats made hyperglycemic by 60% Px. CONCLUSIONS: Our studies have shown physiologic and pharmacologic regulation of GIP-R expression in beta-cells by PPARgamma signaling. Also disruption of this signaling pathway may account for the lowered beta-cell GIP-R expression and resulting GIP resistance in type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Células Secretoras de Insulina/fisiologia , PPAR gama/fisiologia , Receptores dos Hormônios Gastrointestinais/genética , Animais , Cromanos/farmacologia , Resistência a Medicamentos , Polipeptídeo Inibidor Gástrico/fisiologia , Glucose/fisiologia , Masculino , Camundongos , PPAR gama/deficiência , PPAR gama/genética , Pancreatectomia , Ratos , Ratos Sprague-Dawley , Receptores dos Hormônios Gastrointestinais/efeitos dos fármacos , Receptores dos Hormônios Gastrointestinais/fisiologia , Tiazolidinedionas/farmacologia , Troglitazona
16.
Diabetes ; 58(11): 2607-15, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19720802

RESUMO

OBJECTIVE: The G-protein-coupled receptor GPR40 mediates fatty acid potentiation of glucose-stimulated insulin secretion, but its contribution to insulin secretion in vivo and mechanisms of action remain uncertain. This study was aimed to ascertain whether GPR40 controls insulin secretion in vivo and modulates intracellular fuel metabolism in islets. RESEARCH DESIGN AND METHODS: Insulin secretion and sensitivity were assessed in GPR40 knockout mice and their wild-type littermates by hyperglycemic clamps and hyperinsulinemic euglycemic clamps, respectively. Transcriptomic analysis, metabolic studies, and lipid profiling were used to ascertain whether GPR40 modulates intracellular fuel metabolism in islets. RESULTS: Both glucose- and arginine-stimulated insulin secretion in vivo were decreased by approximately 60% in GPR40 knockout fasted and fed mice, without changes in insulin sensitivity. Neither gene expression profiles nor intracellular metabolism of glucose and palmitate in isolated islets were affected by GPR40 deletion. Lipid profiling of isolated islets revealed that the increase in triglyceride and decrease in lyso-phosphatidylethanolamine species in response to palmitate in vitro was similar in wild-type and knockout islets. In contrast, the increase in intracellular inositol phosphate levels observed in wild-type islets in response to fatty acids in vitro was absent in knockout islets. CONCLUSIONS: These results indicate that deletion of GPR40 impairs insulin secretion in vivo not only in response to fatty acids but also to glucose and arginine, without altering intracellular fuel metabolism in islets, via a mechanism that may involve the generation of inositol phosphates downstream of GPR40 activation.


Assuntos
Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Tecido Adiposo/anatomia & histologia , Animais , Ácido Araquidônico/metabolismo , Arginina/farmacologia , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Perfilação da Expressão Gênica , Fosfatos de Inositol/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Lipídeos/isolamento & purificação , Lipídeos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
17.
J Biol Chem ; 283(47): 32462-70, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18718916

RESUMO

We reported that peroxisome proliferator-activated receptor gamma (PPARgamma) transcriptionally regulates the beta-cell differentiation factor pancreatic duodenal homeobox (PDX)-1 based on in vitro RNA interference studies. We have now studied mice depleted of PPARgamma within the pancreas (PANC PPARgamma(-/-)) created by a Cre/loxP recombinase system, with Cre driven by the pdx-1 promoter. Male PANC PPARgamma(-/-) mice were hyperglycemic at 8 weeks of age (8.1+/-0.2 mM versus 6.4+/-0.3 mM, p=0.009) with islet cytoarchitecture and pancreatic mass of islet beta-cells that were indistinguishable from the controls. Islet PDX-1 mRNA (p=0.001) and protein levels (p=0.003) were lowered 60 and 40%, respectively, in tandem with impaired glucose-induced insulin secretion and loss of thiazolidinedione-induced increase in PDX-1 expression. We next identified a putative PPAR-response element (PPRE) in the mouse pdx-1 promoter with substantial homology to the corresponding region of the human PDX-1 promoter. Electrophoretic mobility supershift assays with nuclear extracts from beta-cell lines and mouse islets, also in vitro translated PPARgamma and retinoid X receptor, and chromatin immunoprecipitation analysis demonstrated specific binding of PPARgamma and retinoid X receptor to the human and mouse pdx-1 x PPREs. Transient transfection assays of beta-cells with reporter constructs of mutated PPREs showed dramatically reduced pdx-1 promoter activity. In summary, we have presented in vivo and in vitro evidence showing PPARgamma regulation of pdx-1 transcription in beta-cells, plus our results support an important regulatory role for PPARgamma in beta-cell physiology and thiazolidinedione pharmacology of type 2 diabetes.


Assuntos
Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , PPAR gama/metabolismo , Regiões Promotoras Genéticas , Transativadores/genética , Animais , Sequência de Bases , Humanos , Hiperglicemia/genética , Técnicas In Vitro , Células Secretoras de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Elementos de Resposta , Homologia de Sequência do Ácido Nucleico
18.
Am J Physiol Endocrinol Metab ; 294(4): E679-87, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18230696

RESUMO

The physiological mechanisms underlying pancreatic beta-cell mass (BCM) homeostasis are complex and not fully resolved. Here we examined the factors contributing to the increased BCM following a mild glucose infusion (GI) whereby normoglycemia was maintained through 96 h. We used morphometric and immunochemical methods to investigate enhanced beta-cell growth and survival in Sprague-Dawley rats. BCM was elevated >2.5-fold over saline-infused control rats by 48 h and increased modestly thereafter. Unexpectedly, increases in beta-cell proliferation were not observed at any time point through 4 days. Instead, enhanced numbers of insulin(+) cell clusters and small islets (400-12,000 microm(2); approximately 23- to 124-microm diameter), mostly scattered among the acini, were observed in the GI rats by 48 h despite no difference in the numbers of medium to large islets. We previously showed that increased beta-cell growth in rodent models of insulin resistance and pancreatic regeneration involves increased activated Akt/PKB, a key beta-cell signaling intermediate, in both islets and endocrine cell clusters. GI in normal rats also leads to increased Akt activation in islet beta-cells, as well as in insulin(+) and insulin(-) cells in the common duct epithelium and endocrine clusters. This correlated with strong Pdx1 expression in these same cells. These results suggest that mechanisms other than proliferation underlie the rapid beta-cell growth response following a mild GI in the normal rat and involve Akt-regulated enhanced beta-cell survival potential and neogenesis from epithelial precursors.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Regeneração/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Insulina/sangue , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/fisiologia , Masculino , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Cloreto de Sódio/farmacologia
19.
Diabetes ; 57(2): 424-31, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17991758

RESUMO

OBJECTIVE: Prolonged exposure of isolated islets of Langerhans to elevated levels of fatty acids, in the presence of high glucose, impairs insulin gene expression via a transcriptional mechanism involving nuclear exclusion of pancreas-duodenum homeobox-1 (Pdx-1) and loss of MafA expression. Whether such a phenomenon also occurs in vivo is unknown. Our objective was therefore to ascertain whether chronic nutrient oversupply inhibits insulin gene expression in vivo. RESEARCH DESIGN AND METHODS: Wistar rats received alternating 4-h infusions of glucose and Intralipid for a total of 72 h. Control groups received alternating infusions of glucose and saline, saline and Intralipid, or saline only. Insulin and C-peptide secretion were measured under hyperglycemic clamps. Insulin secretion and gene expression were assessed in isolated islets, and beta-cell mass was quantified by morphometric analysis. RESULTS: Neither C-peptide secretion nor insulin sensitivity was different among infusion regimens. Insulin content and insulin mRNA levels were lower in islets isolated from rats infused with glucose plus Intralipid. This was associated with reduced Pdx-1 binding to the endogenous insulin promoter, and an increased proportion of Pdx-1 localized in the cytoplasm versus the nucleus. In contrast, MafA mRNA and protein levels and beta-cell mass and proliferation were unchanged. CONCLUSIONS: Cyclical and alternating infusions of glucose and Intralipid in normal rats inhibit insulin gene expression without affecting insulin secretion or beta-cell mass. We conclude that fatty acid inhibition of insulin gene expression, in the presence of high glucose, is an early functional defect that may contribute to beta-cell failure in type 2 diabetes.


Assuntos
Emulsões Gordurosas Intravenosas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Proteínas de Homeodomínio/metabolismo , Insulina/genética , Transativadores/metabolismo , Animais , Glicemia/metabolismo , Peptídeo C/efeitos dos fármacos , Peptídeo C/metabolismo , Emulsões Gordurosas Intravenosas/administração & dosagem , Ácidos Graxos não Esterificados/sangue , Glucose/administração & dosagem , Teste de Tolerância a Glucose , Proteínas de Homeodomínio/efeitos dos fármacos , Hiperglicemia , Infusões Intravenosas , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , RNA Mensageiro/genética , Ratos , Ratos Wistar , Transativadores/efeitos dos fármacos
20.
Diabetes ; 56(4): 1087-94, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17395749

RESUMO

Long-chain fatty acids amplify insulin secretion from the pancreatic beta-cell. The G-protein-coupled receptor GPR40 is specifically expressed in beta-cells and is activated by fatty acids; however, its role in acute regulation of insulin secretion in vivo remains unclear. To this aim, we generated GPR40 knockout (KO) mice and examined glucose homeostasis, insulin secretion in response to glucose and Intralipid in vivo, and insulin secretion in vitro after short- and long-term exposure to fatty acids. Our results show that GPR40 KO mice have essentially normal glucose tolerance and insulin secretion in response to glucose. Insulin secretion in response to Intralipid was reduced by approximately 50%. In isolated islets, insulin secretion in response to glucose and other secretagogues was unaltered, but fatty acid potentiation of insulin release was markedly reduced. The Galpha(q/11) inhibitor YM-254890 dose-dependently reduced palmitate potentiation of glucose-induced insulin secretion. Islets from GPR40 KO mice were as sensitive to fatty acid inhibition of insulin secretion upon prolonged exposure as islets from wild-type animals. We conclude that GPR40 contributes approximately half of the full acute insulin secretory response to fatty acids in mice but does not play a role in the mechanisms by which fatty acids chronically impair insulin secretion.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Animais , Células Cultivadas , Emulsões Gordurosas Intravenosas/farmacologia , Feminino , Glucose/farmacologia , Heparina/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA