Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cell Biochem ; 120(6): 10688-10696, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30644120

RESUMO

The contribution of proximal tubules (PT) to albumin uptake is now well recognized, however, its regulation is understudied area. There are reports suggesting that insulin resistance is associated with the development of albuminuria in nondiabetic individuals. We have previously reported reduced insulin receptor (IR) expression in renal-tubular-epithelial cells, including PT in various models of insulin resistance. However, the effect of a physiological fall in insulin levels and the role for IR in PT in tubular albumin uptake is not clear. To address these gaps in our understanding, we estimated urine excretion and renal uptake of albumin in fasted and fed C57Bl/6 mice injected with fluorescein isothiocyanate (FITC)-albumin (5 µg/mL/kg body weight, intraperitoneal, n = 6 per group). In addition, we compared spot urine analysis from 33 clinically healthy humans after overnight fasting (when insulin levels are lower than in the fed state) and then at 2 hours after 75 g oral glucose challenge (postprandial). Fasted mice had attenuated renal uptake of FITC-albumin and higher excretion in urine, relative to fed mice ( P = 0.04). Moreover, a significant drop in urine albumin-to-creatinine ratio (ACR) and urine albumin concentration (UAC) was observed in the postprandial state in these subjects ( P = 0.001 and P = 0.017, for ACR and UAC, respectively). The drop was negatively associated with postprandial blood glucose levels (ρ = -0.36, P = 0.03 for ΔUAC and ρ = -0.34, P = 0.05 for ΔACR). To test the role of IR in PT, we analyzed 24-hour urine albumin excretion in male mice with targeted deletion of IR from PT (insulin receptor knockout [IRKO]) and their wild-type (WT) littermates ( n = 7 per group). IRKO mice had significantly higher 24-hour urine albumin excretion relative to WT. Moreover, kidneys from KO mice revealed reduced expression of megalin and cubulin proteins in the PT relative to the WT. We also demonstrated insulin (100 nM) induced albumin internalization in human proximal tubule cells (hPT) and this effect of insulin was attenuated in hydroxy-2-naphthalenylmethylphosphonic acid (100 µM), a tyrosine kinase inhibitor, pretreated hPT. Our findings revealed albumin excretion was attenuated by glucose administration to fasting individuals implying a regulatory role for insulin in PT albumin reabsorption. Thus albuminuria associated with insulin resistance/diabetes may relate not only to glomerular dysfunction but also to impairment in insulin-mediated reabsorption.


Assuntos
Albuminúria/genética , Células Epiteliais/metabolismo , Insulina/metabolismo , Túbulos Renais Proximais/metabolismo , Receptor de Insulina/genética , Albuminúria/metabolismo , Albuminúria/fisiopatologia , Animais , Creatinina/urina , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Jejum/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Glucose/farmacologia , Teste de Tolerância a Glucose , Humanos , Insulina/farmacologia , Resistência à Insulina , Túbulos Renais Proximais/fisiopatologia , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Naftalenos/farmacologia , Organofosfonatos/farmacologia , Cultura Primária de Células , Receptor de Insulina/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Albumina Sérica/metabolismo
2.
Int J Cancer ; 143(10): 2458-2469, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30070361

RESUMO

Insulin and insulin-like growth factor (IGF)-1 signaling in the thyroid are thought to be permissive for the coordinated regulation by thyroid-stimulating hormone (TSH) of thyrocyte proliferation and hormone production. However, the integrated role of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in thyroid development and function has not been explored. Here, we generated thyrocyte-specific IR and IGF-1R double knockout (DTIRKO) mice to precisely evaluate the coordinated functions of these receptors in the thyroid of neonates and adults. Neonatal DTIRKO mice displayed smaller thyroids, paralleling defective folliculogenesis associated with repression of the thyroid-specific transcription factor Foxe1. By contrast, at postnatal day 14, absence of IR and IGF-1R paradoxically induced thyrocyte proliferation, which was mediated by mTOR-dependent signaling pathways. Furthermore, we found elevated production of TSH during the development of follicular hyperplasia at 8 weeks of age. By 50 weeks, all DTIRKO mice developed papillary thyroid carcinoma (PTC)-like lesions that correlated with induction of the ErbB pathway. Taken together, these data define a critical role for IR and IGF-1R in neonatal thyroid folliculogenesis. They also reveal an important reciprocal relationship between IR/IGF-1R and TSH/ErbB signaling in the pathogenesis of thyroid follicular hyperplasia and, possibly, of papillary carcinoma.


Assuntos
Receptores ErbB/metabolismo , Receptor IGF Tipo 1/deficiência , Receptor de Insulina/deficiência , Câncer Papilífero da Tireoide/metabolismo , Células Epiteliais da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais , Câncer Papilífero da Tireoide/patologia , Células Epiteliais da Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Tireotropina/biossíntese , Tireotropina/metabolismo
3.
J Clin Invest ; 128(7): 2914-2926, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29664737

RESUMO

Complications of diabetes affect tissues throughout the body, including the central nervous system. Epidemiological studies show that diabetic patients have an increased risk of depression, anxiety, age-related cognitive decline, and Alzheimer's disease. Mice lacking insulin receptor (IR) in the brain or on hypothalamic neurons display an array of metabolic abnormalities; however, the role of insulin action on astrocytes and neurobehaviors remains less well studied. Here, we demonstrate that astrocytes are a direct insulin target in the brain and that knockout of IR on astrocytes causes increased anxiety- and depressive-like behaviors in mice. This can be reproduced in part by deletion of IR on astrocytes in the nucleus accumbens. At a molecular level, loss of insulin signaling in astrocytes impaired tyrosine phosphorylation of Munc18c. This led to decreased exocytosis of ATP from astrocytes, resulting in decreased purinergic signaling on dopaminergic neurons. These reductions contributed to decreased dopamine release from brain slices. Central administration of ATP analogs could reverse depressive-like behaviors in mice with astrocyte IR knockout. Thus, astrocytic insulin signaling plays an important role in dopaminergic signaling, providing a potential mechanism by which astrocytic insulin action may contribute to increased rates of depression in people with diabetes, obesity, and other insulin-resistant states.


Assuntos
Astrócitos/fisiologia , Comportamento Animal/fisiologia , Insulina/fisiologia , Transmissão Sináptica/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Ansiedade/etiologia , Ansiedade/fisiopatologia , Encéfalo/fisiologia , Depressão/etiologia , Depressão/fisiopatologia , Diabetes Mellitus/fisiopatologia , Diabetes Mellitus/psicologia , Modelos Animais de Doenças , Dopamina/fisiologia , Exocitose , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Modelos Neurológicos , Proteínas Munc18/metabolismo , Núcleo Accumbens/fisiopatologia , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Receptor de Insulina/fisiologia
4.
Endocrinology ; 159(3): 1253-1263, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29300910

RESUMO

Insulin coordinates the complex response to feeding, affecting numerous metabolic and hormonal pathways. Forkhead box protein O1 (FoxO1) is one of several signaling molecules downstream of insulin; FoxO1 drives gluconeogenesis and is suppressed by insulin. To determine the role of FoxO1 in mediating other actions of insulin, we studied mice with hepatic deletion of the insulin receptor, FoxO1, or both. We found that mice with deletion of the insulin receptor alone showed not only hyperglycemia but also a 70% decrease in plasma insulin-like growth factor 1 and delayed growth during the first 2 months of life, a 24-fold increase in the soluble leptin receptor and a 19-fold increase in plasma leptin levels. Deletion of the insulin receptor also produced derangements in fatty acid metabolism, with a decrease in the expression of the lipogenic enzymes, hepatic diglycerides, and plasma triglycerides; in parallel, it increased expression of the fatty acid oxidation enzymes. Mice with deletion of both insulin receptor and FoxO1 showed a much more modest phenotype, with normal or near-normal glucose levels, growth, leptin levels, hepatic diglycerides, and fatty acid oxidation gene expression; however, lipogenic gene expression remained low. Taken together, these data reveal the pervasive role of FoxO1 in mediating the effects of insulin on not only glucose metabolism but also other hormonal signaling pathways and even some aspects of lipid metabolism.


Assuntos
Proteína Forkhead Box O1/fisiologia , Fígado/química , Receptor de Insulina/deficiência , Receptor de Insulina/fisiologia , Animais , Glicemia/análise , Ácidos Graxos/metabolismo , Proteína Forkhead Box O1/deficiência , Proteína Forkhead Box O1/genética , Expressão Gênica , Gluconeogênese/genética , Insulina/sangue , Insulina/farmacologia , Insulina/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Leptina/sangue , Leptina/metabolismo , Lipídeos/análise , Lipogênese/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Receptores para Leptina/sangue , Triglicerídeos/sangue
5.
Clin Sci (Lond) ; 131(12): 1301-1315, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28468951

RESUMO

In patients with non-alcoholic fatty liver disease (NAFLD), insulin resistance (IR) associates with fibrosis progression independently of the hepatic inflammation, but the mechanisms are still unclear. We modeled the independent contribution of inflammation (non-alcoholic steatohepatitis: NASH) by exploiting the methionine-choline deficient (MCD) diet, and that of IR by insulin receptor (InsR) haploinsufficiency (InsR+/-) in the pathogenesis of liver fibrosis in C57BL/6 mice. We confirmed the study findings in 96 patients with NAFLD. InsR+/- enhanced hepatic fat content and impaired hepatic insulin signaling leading to Forkhead box protein O1 (FoxO1) accumulation in MCD-fed mice. Remarkably, despite reduced inflammation and hampered transdifferentiation of hepatic stellate cells (HSCs), InsR+/- promoted hepatic fibrosis accumulation, which correlated with the induction of the Lysyl Oxidase Like 2 (Loxl2), involved in matrix stabilization. Loxl2 up-regulation was not a cell autonomous property of insulin resistant HSCs, but was dependent on microparticles (MPs) released specifically by insulin resistant hepatocytes (HEPs) exposed to fatty acids. The mechanism entailed FoxO1 up-regulation, as FoxO1 silencing normalized Loxl2 expression reversing fibrosis in InsR+/- MCD-fed mice. Loxl2 up-regulation was similarly detected during IR induced by obesity, but not by lipogenic stimuli (fructose feeding). Most importantly, LOXL2 up-regulation was observed in NAFLD patients with type 2 diabetes (T2D) and LOXL2 hepatic and circulating levels correlated with histological fibrosis progression. IR favors fibrosis deposition independently of the classic 'inflammation - HSC transdifferentiation' pathway. The mechanism entails a cross-talk between enhanced lipotoxicity in insulin resistant HEPs and Loxl2 production by HSCs, which was confirmed in patients with diabetes, thereby facilitating extracellular matrix (ECM) stabilization.


Assuntos
Aminoácido Oxirredutases/biossíntese , Resistência à Insulina , Cirrose Hepática/enzimologia , Fígado/enzimologia , Hepatopatia Gordurosa não Alcoólica/enzimologia , Animais , Proliferação de Células , Transdiferenciação Celular , Células Cultivadas , Deficiência de Colina/complicações , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Indução Enzimática , Matriz Extracelular/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Predisposição Genética para Doença , Células Estreladas do Fígado/enzimologia , Células Estreladas do Fígado/patologia , Hepatócitos/enzimologia , Hepatócitos/patologia , Humanos , Fígado/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Metionina/deficiência , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Fenótipo , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Transdução de Sinais
6.
Diabetologia ; 60(8): 1442-1453, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28526921

RESUMO

AIMS/HYPOTHESIS: We aimed to investigate potential interactions between insulin and glucagon-like peptide (GLP)-1 signalling pathways in the regulation of beta cell-cycle dynamics in vivo, in the context of the therapeutic potential of GLP-1 to modulate impaired beta cell function. METHODS: Beta cell-specific insulin receptor knockout (ßIRKO) mice, which exhibit beta cell dysfunction and an age-dependent decrease in beta cell mass, were treated with the dipeptidyl peptidase-4 inhibitor vildagliptin. Following this, glucose homeostasis and beta cell proliferation were evaluated and underlying molecular mechanisms were investigated. RESULTS: The sustained elevation in circulating GLP-1 levels, caused by treatment of the knockout mice with vildagliptin for 6 weeks, significantly improved glucose tolerance secondary to enhanced insulin secretion and proliferation of beta cells. Treating ßIRKO beta cell lines with the GLP-1 analogue, exendin-4, promoted Akt phosphorylation and protein expression of cyclins A, D1 and E two- to threefold, in addition to cyclin D2. Pancreases from the vildagliptin-treated ßIRKO mice exhibited increased cyclin D1 expression, while cyclin D2 expression was impaired. CONCLUSIONS/INTERPRETATION: Activation of GLP-1 signalling compensates for impaired growth factor (insulin) signalling and enhances expression of cyclins to promote beta cell proliferation. Together, these data indicate the potential of GLP-1-related therapies to enhance beta cell proliferation and promote beneficial outcomes in models with dysfunctional beta cells.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptor de Insulina/metabolismo , Adamantano/análogos & derivados , Adamantano/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Ciclina A/metabolismo , Ciclina D/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Exenatida , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Knockout , Nitrilas/farmacologia , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Pirrolidinas/farmacologia , Receptor de Insulina/deficiência , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Peçonhas/farmacologia , Vildagliptina
7.
Arterioscler Thromb Vasc Biol ; 36(7): 1356-66, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27150392

RESUMO

OBJECTIVE: Plasma levels of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (ApoA-I) are reduced in individuals with defective insulin signaling. Initial studies using liver-specific insulin receptor (InsR) knockout mice identified reduced expression of type 1 deiodinase (Dio1) as a potentially novel link between defective hepatic insulin signaling and reduced expression of the ApoA-I gene. Our objective was to examine the regulation of ApoA-I expression by Dio1. APPROACH AND RESULTS: Acute inactivation of InsR by adenoviral delivery of Cre recombinase to InsR floxed mice reduced HDL-C and expression of both ApoA-I and Dio1. Overexpression of Dio1 in InsR knockout mice restored HDL-C and ApoA-I levels and increased the expression of ApoA-I. Dio1 knockout mice had low expression of ApoA-I and reduced serum levels of HDL-C and ApoA-I. Treatment of C57BL/6J mice with antisense to Dio1 reduced ApoA-I mRNA, HDL-C, and serum ApoA-I. Hepatic 3,5,3'-triiodothyronine content was normal or elevated in InsR knockout mice or Dio1 knockout mice. Knockdown of either InsR or Dio1 by siRNA in HepG2 cells decreased the expression of ApoA-I and ApoA-I synthesis and secretion. siRNA knockdown of InsR or Dio1 decreased activity of a region of the ApoA-I promoter lacking thyroid hormone response elements (region B). Electrophoretic mobility shift assay demonstrated that reduced Dio1 expression decreased the binding of nuclear proteins to region B. CONCLUSIONS: Reductions in Dio1 expression reduce the expression of ApoA-I in a 3,5,3'-triiodothyronine-/thyroid hormone response element-independent manner.


Assuntos
Apolipoproteína A-I/metabolismo , Iodeto Peroxidase/metabolismo , Fígado/enzimologia , Transdução de Sinais , Tri-Iodotironina/metabolismo , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-I/deficiência , Apolipoproteína A-I/genética , HDL-Colesterol/sangue , Regulação da Expressão Gênica , Genótipo , Células Hep G2 , Humanos , Iodeto Peroxidase/deficiência , Iodeto Peroxidase/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Elementos de Resposta , Transfecção
8.
Nature ; 519(7544): 464-7, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25799997

RESUMO

Wing polyphenism is an evolutionarily successful feature found in a wide range of insects. Long-winged morphs can fly, which allows them to escape adverse habitats and track changing resources, whereas short-winged morphs are flightless, but usually possess higher fecundity than the winged morphs. Studies on aphids, crickets and planthoppers have revealed that alternative wing morphs develop in response to various environmental cues, and that the response to these cues may be mediated by developmental hormones, although research in this area has yielded equivocal and conflicting results about exactly which hormones are involved. As it stands, the molecular mechanism underlying wing morph determination in insects has remained elusive. Here we show that two insulin receptors in the migratory brown planthopper Nilaparvata lugens, InR1 and InR2, have opposing roles in controlling long wing versus short wing development by regulating the activity of the forkhead transcription factor Foxo. InR1, acting via the phosphatidylinositol-3-OH kinase (PI(3)K)-protein kinase B (Akt) signalling cascade, leads to the long-winged morph if active and the short-winged morph if inactive. InR2, by contrast, functions as a negative regulator of the InR1-PI(3)K-Akt pathway: suppression of InR2 results in development of the long-winged morph. The brain-secreted ligand Ilp3 triggers development of long-winged morphs. Our findings provide the first evidence of a molecular basis for the regulation of wing polyphenism in insects, and they are also the first demonstration--to our knowledge--of binary control over alternative developmental outcomes, and thus deepen our understanding of the development and evolution of phenotypic plasticity.


Assuntos
Hemípteros/anatomia & histologia , Hemípteros/metabolismo , Receptor de Insulina/metabolismo , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo , Animais , Feminino , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/metabolismo , Hemípteros/enzimologia , Hemípteros/genética , Insulina/metabolismo , Masculino , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/deficiência , Transdução de Sinais , Asas de Animais/anatomia & histologia , Asas de Animais/enzimologia
9.
Nat Commun ; 6: 6496, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25754093

RESUMO

Endoplasmic reticulum (ER) stress is associated with diabetic nephropathy (DN), but its pathophysiological relevance and the mechanisms that compromise adaptive ER signalling in podocytes remain unknown. Here we show that nuclear translocation of the transcription factor spliced X-box binding protein-1 (sXBP1) is selectively impaired in DN, inducing activating transcription factor-6 (ATF6) and C/EBP homology protein (CHOP). Podocyte-specific genetic ablation of XBP1 or inducible expression of ATF6 in mice aggravates DN. sXBP1 lies downstream of insulin signalling and attenuating podocyte insulin signalling by genetic ablation of the insulin receptor or the regulatory subunits phosphatidylinositol 3-kinase (PI3K) p85α or p85ß impairs sXBP1 nuclear translocation and exacerbates DN. Corroborating our findings from murine DN, the interaction of sXBP1 with p85α and p85ß is markedly impaired in the glomerular compartment of human DN. Thus, signalling via the insulin receptor, p85, and XBP1 maintains podocyte homeostasis, while disruption of this pathway impairs podocyte function in DN.


Assuntos
Fator 6 Ativador da Transcrição/genética , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Proteínas de Ligação a DNA/genética , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/genética , Estresse do Retículo Endoplasmático/genética , Podócitos/metabolismo , Fatores de Transcrição/genética , Fator 6 Ativador da Transcrição/deficiência , Animais , Classe Ia de Fosfatidilinositol 3-Quinase/deficiência , Proteínas de Ligação a DNA/deficiência , Bases de Dados Factuais , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Regulação da Expressão Gênica , Humanos , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Podócitos/patologia , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Fatores de Transcrição de Fator Regulador X , Transdução de Sinais , Estreptozocina , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/deficiência , Proteína 1 de Ligação a X-Box
10.
Diabetes ; 63(4): 1270-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24379345

RESUMO

Women with polycystic ovary syndrome (PCOS) exhibit elevated androgen levels, oligoanovulation, infertility, and insulin resistance in metabolic tissues. The aims of these studies were to determine the role of insulin signaling in the development and function of ovarian theca cells and the pathophysiologic effects of hyperinsulinism on ovarian function in obesity. We disrupted the insulin receptor (IR) gene specifically in the theca-interstitial (TI) cells of the ovaries (Cyp17IRKO). No changes in reproductive development or function were observed in lean Cyp17IRKO female mice, suggesting that insulin signaling in TI cell is not essential for reproduction. However, when females were fed a high-fat diet, diet-induced obesity (DIO) wild-type (DIO-WT) mice were infertile and experienced increased circulating testosterone levels, whereas DIO-Cyp17IRKO mice exhibited improved fertility and testosterone levels comparable to those found in lean mice. The levels of phosphorylated IRS1 and CYP17 protein were higher in the ovary of DIO-WT compared with DIO-Cyp17IRKO or lean mice. Ex vivo studies using a whole ovary culture model demonstrated that insulin acts independently or additively with human chorionic gonadotropin to enhance androstenedione secretion. These studies reveal the causal pathway linking hyperinsulinism with ovarian hyperandrogenism and the infertility of obesity.


Assuntos
Hiperandrogenismo/terapia , Infertilidade Feminina/etiologia , Obesidade/complicações , Ovário/fisiologia , Receptor de Insulina/deficiência , Células Tecais/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Hiperandrogenismo/etiologia , Hiperinsulinismo/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Camundongos , Camundongos Knockout , Obesidade/fisiopatologia , Síndrome do Ovário Policístico/fisiopatologia , Transdução de Sinais , Esteroide 17-alfa-Hidroxilase/genética , Esteroide 17-alfa-Hidroxilase/metabolismo , Testosterona/sangue , Técnicas de Cultura de Tecidos
11.
Cardiovasc J Afr ; 24(2): 10-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23612947

RESUMO

AIM: Obesity and type 2 diabetes present with two debilitating complications, namely, hypertension and heart disease. The dried and ground pods of Prosopis glandulosa (commonly known as the Honey mesquite tree) which is part of the Fabaceae (or legume) family are currently marketed in South Africa as a food supplement with blood glucose-stabilising and anti-hypertensive properties. We previously determined its hypoglycaemic effects, and in the current study we determined the efficacy of P glandulosa as anti-hypertensive agent and its myocardial protective ability. METHODS: Male Wistar rats were rendered either pre-diabetic (diet-induced obesity: DIO) or hypertensive (high-fat diet: HFD). DIO animals were treated with P glandulosa (100 mg/kg/day for the last eight weeks of a 16-week period) and compared to age-matched controls. Hearts were perfused ex vivo to determine infarct size. Biometric parameters were determined at the time of sacrifice. Cardiac-specific insulin receptor knock-out (CIRKO) mice were similarly treated with P glandulosa and infarct size was determined. HFD animals were treated with P glandulosa from the onset of the diet or from weeks 12-16, using captopril (50 mg/kg/day) as the positive control. Blood pressure was monitored weekly. RESULTS: DIO rats and CIRKO mice: P glandulosa ingestion significantly reduced infarct size after ischaemia-reperfusion. Proteins of the PI-3-kinase/PKB/Akt survival pathway were affected in a manner supporting cardioprotection. HFD model: P glandulosa treatment both prevented and corrected the development of hypertension, which was also reflected in alleviation of water retention. CONCLUSION: P glandulosa was cardioprotective and infarct sparing as well as anti-hypertensive without affecting the body weight or the intra-peritoneal fat depots of the animals. Changes in the PI-3-kinase/PKB/Akt pathway may be causal to protection. Results indicated water retention, possibly coupled to vasoconstriction in the HFD animals, while ingestion of P glandulosa alleviated both. We concluded that treatment of pre-diabetes, type 2 diabetes or hypertension with P glandulosa poses possible beneficial health effects.


Assuntos
Anti-Hipertensivos/farmacologia , Fabaceae , Hipertensão/prevenção & controle , Infarto do Miocárdio/prevenção & controle , Extratos Vegetais/farmacologia , Estado Pré-Diabético/tratamento farmacológico , Animais , Anti-Hipertensivos/isolamento & purificação , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Fabaceae/química , Frutas , Hipertensão/sangue , Hipertensão/genética , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/sangue , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Fitoterapia , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Estado Pré-Diabético/sangue , Estado Pré-Diabético/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Fatores de Tempo , Micção/efeitos dos fármacos
12.
Endocrinology ; 154(3): 1337-48, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23392256

RESUMO

Pubertal onset only occurs in a favorable, anabolic hormonal environment. The neuropeptide kisspeptin, encoded by the Kiss1 gene, modifies GnRH neuronal activity to initiate puberty and maintain fertility, but the factors that regulate Kiss1 neurons and permit pubertal maturation remain to be clarified. The anabolic factor insulin may signal nutritional status to these neurons. To determine whether insulin sensing plays an important role in Kiss1 neuron function, we generated mice lacking insulin receptors in Kiss1 neurons (IR(ΔKiss) mice). IR(ΔKiss) females showed a delay in vaginal opening and in first estrus, whereas IR(ΔKiss) males also exhibited late sexual maturation. Correspondingly, LH levels in IR(ΔKiss) mice were reduced in early puberty in both sexes. Adult reproductive capacity, body weight, fat composition, food intake, and glucose regulation were comparable between the 2 groups. These data suggest that impaired insulin sensing by Kiss1 neurons delays the initiation of puberty but does not affect adult fertility. These studies provide insight into the mechanisms regulating pubertal timing in anabolic states.


Assuntos
Fertilidade/fisiologia , Kisspeptinas/fisiologia , Puberdade Tardia/fisiopatologia , Receptor de Insulina/fisiologia , Animais , Estradiol/análogos & derivados , Estradiol/farmacologia , Retroalimentação Fisiológica , Feminino , Fertilidade/genética , Hormônio Liberador de Gonadotropina/fisiologia , Insulina/fisiologia , Kisspeptinas/deficiência , Kisspeptinas/genética , Hormônio Luteinizante/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios/fisiologia , Puberdade Tardia/genética , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Maturidade Sexual/genética , Maturidade Sexual/fisiologia
13.
Neuromolecular Med ; 14(4): 262-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22661254

RESUMO

The insulin receptor (IR) is a protein tyrosine kinase playing a pivotal role in the regulation of peripheral glucose metabolism and energy homoeostasis. IRs are also abundantly distributed in the cerebral cortex and hippocampus, where they regulate synaptic activity required for learning and memory. As the major anabolic hormone in mammals, insulin stimulates protein synthesis partially through the activation of the PI3K/Akt/mTOR pathway, playing fundamental roles in neuronal development, synaptic plasticity and memory. Here, by means of a multidisciplinary approach, we report that long-term synaptic plasticity and recognition memory are impaired in IR ß-subunit heterozygous mice. Since IR expression is diminished in type-2 diabetes as well as in Alzheimer's disease (AD) patients, these data may provide a mechanistic link between insulin resistance, impaired synaptic transmission and cognitive decline in humans with metabolic disorders.


Assuntos
Hipocampo/fisiopatologia , Deficiências da Aprendizagem/genética , Potenciação de Longa Duração/genética , Transtornos da Memória/genética , Proteínas do Tecido Nervoso/deficiência , Receptor de Insulina/deficiência , Reconhecimento Psicológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/psicologia , Feminino , Heterozigoto , Humanos , Resistência à Insulina , Deficiências da Aprendizagem/fisiopatologia , Transtornos da Memória/fisiopatologia , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Densidade Pós-Sináptica/ultraestrutura , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Transdução de Sinais/fisiologia , Transmissão Sináptica/genética , Serina-Treonina Quinases TOR/fisiologia
14.
Endocrinology ; 152(12): 4825-37, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21990313

RESUMO

A unique mouse model was developed with elevated endogenous GH (2- to 3-fold) and IGF-I (1.2- to 1.4-fold), due to somatotrope-specific Cre-mediated inactivation of IGF-I receptor (IgfIr) and insulin receptor (Insr) genes (IgfIr,Insr(rGHpCre), referred to as HiGH mice). We demonstrate that the metabolic phenotype of HiGH mice is diet dependent and differs from that observed in other mouse models of GH excess due to ectopic heterologous transgene expression or pituitary tumor formation. Elevated endogenous GH promotes lean mass and whole-body lipid oxidation but has minimal effects on adiposity, even in response to diet-induced obesity. When caloric intake is moderated, elevated GH improves glucose clearance, despite low/normal insulin sensitivity, which may be explained in part by enhanced IGF-I and insulin output. However, when caloric intake is in excess, elevated GH promotes hepatic lipid accumulation, insulin resistance, hyperglycemia, and ketosis. The HiGH mouse model represents a useful tool to study the role endogenous circulating GH levels play in regulating health and disease.


Assuntos
Glucose/metabolismo , Hormônio do Crescimento/metabolismo , Homeostase , Resistência à Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Adiposidade , Animais , Dieta , Hormônio do Crescimento/sangue , Hormônio do Crescimento/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Metabolismo dos Lipídeos , Camundongos , Especificidade de Órgãos , Receptor IGF Tipo 1/deficiência , Receptor de Insulina/deficiência
15.
Eur J Pharmacol ; 672(1-3): 62-9, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22004610

RESUMO

For the development of efficient pulmonary delivery systems for protein and peptide drugs, it is important to understand their transport mechanisms in alveolar epithelial cells. In this study, the uptake mechanism for FITC-insulin in cultured alveolar epithelial cell line RLE-6TN was elucidated. FITC-insulin uptake by RLE-6TN cells was time-dependent, temperature-sensitive, and concentration-dependent. The uptake was inhibited by metabolic inhibitors, cytochalasin D, clathrin-mediated endocytosis inhibitors, and dynasore, an inhibitor of dynamin GTPase. On the other hand, no inhibitory effect was observed with caveolae-mediated endocytosis inhibitors and a macropinocytosis inhibitor. Intracellular FITC-insulin was found to be partly transported to the basal side of the epithelial cell monolayers. In addition, colocalization of FITC-insulin and LysoTracker Red was observed on confocal laser scanning microscopy, indicating that FITC-insulin was partly targeted to lysosomes. In accordance with these findings, SDS-PAGE/fluoroimage analysis showed that intact FITC-insulin in the cells was eliminated with time. The possible receptor involved in FITC-insulin uptake by RLE-6TN cells was examined by using siRNA. Transfection of the cells with megalin or insulin receptor siRNA successfully reduced the corresponding mRNA expression. FITC-insulin uptake decreased on the transfection with insulin receptor siRNA, but not that with megalin siRNA. These results suggest that insulin is taken up through endocytosis in RLE-6TN cells, and after the endocytosis, the intracellular insulin is partly degraded in lysosomes and partly transported to the basal side. Insulin receptor, but not megalin, may be involved at least partly in insulin endocytosis in RLE-6TN cells.


Assuntos
Células Epiteliais/metabolismo , Insulina/metabolismo , Alvéolos Pulmonares/citologia , Animais , Linhagem Celular , Polaridade Celular , Endocitose , Células Epiteliais/citologia , Fluoresceína-5-Isotiocianato/metabolismo , Técnicas de Silenciamento de Genes , Espaço Intracelular/metabolismo , Transporte Proteico , Proteólise , RNA Interferente Pequeno/genética , Ratos , Receptor de Insulina/deficiência , Receptor de Insulina/genética
16.
Diabetes ; 59(9): 2271-80, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20547974

RESUMO

OBJECTIVE: An impaired ability to sense and appropriately respond to insulin-induced hypoglycemia is a common and serious complication faced by insulin-treated diabetic patients. This study tests the hypothesis that insulin acts directly in the brain to regulate critical glucose-sensing neurons in the hypothalamus to mediate the counterregulatory response to hypoglycemia. RESEARCH DESIGN AND METHODS: To delineate insulin actions in the brain, neuron-specific insulin receptor knockout (NIRKO) mice and littermate controls were subjected to graded hypoglycemic (100, 70, 50, and 30 mg/dl) hyperinsulinemic (20 mU/kg/min) clamps and nonhypoglycemic stressors (e.g., restraint, heat). Subsequently, counterregulatory responses, hypothalamic neuronal activation (with transcriptional marker c-fos), and regional brain glucose uptake (via (14)C-2deoxyglucose autoradiography) were measured. Additionally, electrophysiological activity of individual glucose-inhibited neurons and hypothalamic glucose sensing protein expression (GLUTs, glucokinase) were measured. RESULTS: NIRKO mice revealed a glycemia-dependent impairment in the sympathoadrenal response to hypoglycemia and demonstrated markedly reduced (3-fold) hypothalamic c-fos activation in response to hypoglycemia but not other stressors. Glucose-inhibited neurons in the ventromedial hypothalamus of NIRKO mice displayed significantly blunted glucose responsiveness (membrane potential and input resistance responses were blunted 66 and 80%, respectively). Further, hypothalamic expression of the insulin-responsive GLUT 4, but not glucokinase, was reduced by 30% in NIRKO mice while regional brain glucose uptake remained unaltered. CONCLUSIONS: Chronically, insulin acts in the brain to regulate the counterregulatory response to hypoglycemia by directly altering glucose sensing in hypothalamic neurons and shifting the glycemic levels necessary to elicit a normal sympathoadrenal response.


Assuntos
Glicemia/metabolismo , Encéfalo/fisiologia , Hipoglicemia/metabolismo , Hipotálamo/fisiologia , Insulina/fisiologia , Receptor de Insulina/deficiência , Animais , Cruzamentos Genéticos , Regulação da Expressão Gênica , Glucoquinase/genética , Homozigoto , Temperatura Alta , Hipoglicemia/genética , Hipoglicemia/fisiopatologia , Integrases/genética , Proteínas de Filamentos Intermediários/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Nestina , Neurônios/fisiologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/fisiologia , Ratos , Receptor de Insulina/genética , Restrição Física , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Psicológico/sangue
17.
Proc Natl Acad Sci U S A ; 107(24): 10791-8, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20457905

RESUMO

The type 1 insulin-like growth factor receptor (IGF-1R) tyrosine kinase is an important mediator of the protumorigenic effects of IGF-I/II, and inhibitors of IGF-1R signaling are currently being tested in clinical cancer trials aiming to assess the utility of this receptor as a therapeutic target. Despite mounting evidence that the highly homologous insulin receptor (IR) can also convey protumorigenic signals, its direct role in cancer progression has not been genetically defined in vivo, and it remains unclear whether such a role for IR signaling could compromise the efficacy of selective IGF-1R targeting strategies. A transgenic mouse model of pancreatic neuroendocrine carcinogenesis engages the IGF signaling pathway, as revealed by its dependence on IGF-II and by accelerated malignant progression upon IGF-1R overexpression. Surprisingly, preclinical trials with an inhibitory monoclonal antibody to IGF-1R did not significantly impact tumor growth, prompting us to investigate the involvement of IR. The levels of IR were found to be significantly up-regulated during multistep progression from hyperplastic lesions to islet tumors. Its functional involvement was revealed by genetic disruption of the IR gene in the oncogene-expressing pancreatic beta cells, which resulted in reduced tumor burden accompanied by increased apoptosis. Notably, the IR knockout tumors now exhibited sensitivity to anti-IGF-1R therapy; similarly, high IR to IGF-1R ratios demonstrably conveyed resistance to IGF-1R inhibition in human breast cancer cells. The results predict that elevated IR signaling before and after treatment will respectively manifest intrinsic and adaptive resistance to anti-IGF-1R therapies.


Assuntos
Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/terapia , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor de Insulina/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/terapia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Receptor IGF Tipo 2/metabolismo , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Transdução de Sinais
18.
Diabetes ; 59(4): 987-96, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20103709

RESUMO

OBJECTIVE: A major determinant of the progression from insulin resistance to the development of overt type 2 diabetes is a failure to mount an appropriate compensatory beta-cell hyperplastic response to maintain normoglycemia. We undertook the present study to directly explore the significance of the cell cycle protein cyclin D2 in the expansion of beta-cell mass in two different models of insulin resistance. RESEARCH DESIGN AND METHODS: We created compound knockouts by crossing mice deficient in cyclin D2 (D2KO) with either the insulin receptor substrate 1 knockout (IRS1KO) mice or the insulin receptor liver-specific knockout mice (LIRKO), neither of which develops overt diabetes on its own because of robust compensatory beta-cell hyperplasia. We phenotyped the double knockouts and used RT-qPCR and immunohistochemistry to examine beta-cell mass. RESULTS: Both compound knockouts, D2KO/LIRKO and D2KO/IRS1KO, exhibited insulin resistance and hyperinsulinemia and an absence of compensatory beta-cell hyperplasia. However, the diabetic D2KO/LIRKO group rapidly succumbed early compared with a relatively normal lifespan in the glucose-intolerant D2KO/IRS1KO mice. CONCLUSIONS: This study provides direct genetic evidence that cyclin D2 is essential for the expansion of beta-cell mass in response to a spectrum of insulin resistance and points to the cell-cycle protein as a potential therapeutic target that can be harnessed for preventing and curing type 2 diabetes.


Assuntos
Ciclina D2/farmacologia , Diabetes Mellitus Experimental/genética , Hiperglicemia/genética , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/patologia , Receptor de Insulina/deficiência , Animais , Ciclina D1/genética , Ciclina D2/deficiência , Ciclina D2/genética , Genótipo , Homozigoto , Hiperplasia/fisiopatologia , Fígado/fisiologia , Camundongos , Camundongos Knockout , Receptor de Insulina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Circ Res ; 104(9): 1085-94, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19342603

RESUMO

Impaired insulin signaling via phosphatidylinositol 3-kinase/Akt to endothelial nitric oxide synthase (eNOS) in the vasculature has been postulated to lead to arterial dysfunction and hypertension in obesity and other insulin resistant states. To investigate this, we compared insulin signaling in the vasculature, endothelial function, and systemic blood pressure in mice fed a high-fat (HF) diet to mice with genetic ablation of insulin receptors in all vascular tissues (TTr-IR(-/-)) or mice with genetic ablation of Akt1 (Akt1-/-). HF mice developed obesity, impaired glucose tolerance, and elevated free fatty acids that was associated with endothelial dysfunction and hypertension. Basal and insulin-mediated phosphorylation of extracellular signal-regulated kinase 1/2 and Akt in the vasculature was preserved, but basal and insulin-stimulated eNOS phosphorylation was abolished in vessels from HF versus lean mice. In contrast, basal vascular eNOS phosphorylation, endothelial function, and blood pressure were normal despite absent insulin-mediated eNOS phosphorylation in TTr-IR(-/-) mice and absent insulin-mediated eNOS phosphorylation via Akt1 in Akt1-/- mice. In cultured endothelial cells, 6 hours of incubation with palmitate attenuated basal and insulin-stimulated eNOS phosphorylation and NO production despite normal activation of extracellular signal-regulated kinase 1/2 and Akt. Moreover, incubation of isolated arteries with palmitate impaired endothelium-dependent but not vascular smooth muscle function. Collectively, these results indicate that lower arterial eNOS phosphorylation, hypertension, and vascular dysfunction following HF feeding do not result from defective upstream signaling via Akt, but from free fatty acid-mediated impairment of eNOS phosphorylation.


Assuntos
Pressão Sanguínea , Endotélio Vascular/enzimologia , Hipertensão/enzimologia , Resistência à Insulina , Insulina/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Gorduras na Dieta , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/enzimologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Inibidores Enzimáticos/farmacologia , Ácidos Graxos não Esterificados/metabolismo , Intolerância à Glucose/enzimologia , Intolerância à Glucose/fisiopatologia , Hipertensão/etiologia , Hipertensão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Obesidade/enzimologia , Obesidade/fisiopatologia , Ácido Palmítico/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Vasoconstrição , Vasoconstritores/farmacologia , Vasodilatação , Vasodilatadores/farmacologia
20.
J Clin Invest ; 119(4): 1029-41, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19273907

RESUMO

Type 2 diabetes is associated with accelerated atherogenesis, which may result from a combination of factors, including dyslipidemia characterized by increased VLDL secretion, and insulin resistance. To assess the hypothesis that both hepatic and peripheral insulin resistance contribute to atherogenesis, we crossed mice deficient for the LDL receptor (Ldlr-/- mice) with mice that express low levels of IR in the liver and lack IR in peripheral tissues (the L1B6 mouse strain). Unexpectedly, compared with Ldlr-/- controls, L1B6Ldlr-/- mice fed a Western diet showed reduced VLDL and LDL levels, reduced atherosclerosis, decreased hepatic AKT signaling, decreased expression of genes associated with lipogenesis, and diminished VLDL apoB and lipid secretion. Adenovirus-mediated hepatic expression of either constitutively active AKT or dominant negative glycogen synthase kinase (GSK) markedly increased VLDL and LDL levels such that they were similar in both Ldlr-/- and L1B6Ldlr-/- mice. Knocking down expression of hepatic IR by adenovirus-mediated shRNA decreased VLDL triglyceride and apoB secretion in Ldlr-/- mice. Furthermore, knocking down hepatic IR expression in either WT or ob/ob mice reduced VLDL secretion but also resulted in decreased hepatic Ldlr protein. These findings suggest a dual action of hepatic IR on lipoprotein levels, in which the ability to increase VLDL apoB and lipid secretion via AKT/GSK is offset by upregulation of Ldlr.


Assuntos
Aterosclerose/etiologia , Insulina/metabolismo , Lipoproteínas VLDL/metabolismo , Fígado/metabolismo , Animais , Apolipoproteínas B/sangue , Apolipoproteínas B/metabolismo , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/metabolismo , Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Lipídeos/sangue , Lipogênese , Lipoproteínas VLDL/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Obesos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA