Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 514
Filtrar
1.
Mol Metab ; 55: 101416, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34896640

RESUMO

OBJECTIVE: Adipogenesis plays an essential role in maintaining energy and hormonal balance. Cavin-2, one of the caveolae-related proteins, is abundant in adipocytes, the leading site of adipogenesis. However, the details of the roles of Cavin-2 in adipogenesis remain unknown. Here, we demonstrate the requirement of Cavin-2 for the expression and stability of IRß in adequate adipocyte differentiation. METHODS: Cavin-2 knockout (Cavin-2 KO) and wild-type (WT) mice were fed with a high-fat diet (HFD) for 8 weeks. We evaluated body weight, food intake, and several tissues. Glucose homeostasis was assessed by glucose and insulin tolerance tests. Insulin signaling in epididymal white adipose tissue (eWAT) was determined by Akt phosphorylation. In vitro study, we evaluated adipocyte differentiation, adipogenesis-related genes, and insulin signaling to clarify the relationship between Cavin-2 and adipogenesis under the manipulation of Cavin-2 expression. RESULTS: Caveolae structure decreased in eWAT of Cavin-2 KO mice and Cavin-2 knockdown 3T3-L1 cells. Cavin-2 enhanced the stability of insulin receptor (IR) through direct association at the plasma membrane in adipocytes, resulting in accelerated insulin/IR/Akt signaling-induced adipogenic gene expression in insulin-containing solution-stimulated 3T3-L1 adipocytes. IR-mediated Akt activation also enhanced Cavin-2 and IR expression. Cavin-2 knockout mice showed insulin resistance with dyslipidemia and pathological hypertrophic adipocytes after a HFD. CONCLUSIONS: Cavin-2 enhances IR stability through binding IR and regulates insulin signaling, promoting adequate adipocyte differentiation. Our findings highlight the pivotal role of Cavin-2 in adipogenesis and lipid metabolism, which may help to develop novel therapies for pathological obesity and adipogenic disorders.


Assuntos
Adipócitos/metabolismo , Proteínas de Membrana/metabolismo , Receptor de Insulina/metabolismo , Células 3T3-L1 , Adipócitos/fisiologia , Adipogenia/genética , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal , Diferenciação Celular , Dieta Hiperlipídica , Glucose/metabolismo , Insulina/metabolismo , Resistência à Insulina , Metabolismo dos Lipídeos , Proteínas de Membrana/fisiologia , Camundongos , Obesidade/metabolismo , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/fisiologia , Transdução de Sinais
2.
J Clin Invest ; 131(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34428182

RESUMO

Insulin and IGF-1 are essential for adipocyte differentiation and function. Mice lacking insulin and IGF-1 receptors in fat (FIGIR-KO, fat-specific IGF-1 receptor and insulin receptor-KO) exhibit complete loss of white and brown adipose tissue (WAT and BAT), glucose intolerance, insulin resistance, hepatosteatosis, and cold intolerance. To determine the role of FOXO transcription factors in the altered adipose phenotype, we generated FIGIR-KO mice with fat-specific KO of fat-expressed Foxos [Foxo1, Foxo3, Foxo4] (F-Quint-KO). Unlike FIGIR-KO mice, F-Quint-KO mice had normal BAT, glucose tolerance, insulin-regulated hepatic glucose production, and cold tolerance. However, loss of FOXOs only partially rescued subcutaneous WAT and hepatosteatosis, did not rescue perigonadal WAT or systemic insulin resistance, and led to even more marked hyperinsulinemia. Thus, FOXOs play different roles in insulin/IGF-1 action in different adipose depots, being most important in BAT, followed by subcutaneous WAT and then by visceral WAT. Disruption of FOXOs in fat also led to a reversal of insulin resistance in liver, but not in skeletal muscle, and an exacerbation of hyperinsulinemia. Thus, adipose FOXOs play a unique role in regulating crosstalk between adipose depots, liver, and ß cells.


Assuntos
Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Branco/efeitos dos fármacos , Proteína Forkhead Box O1/fisiologia , Insulina/farmacologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Metabolismo Energético , Glucose/metabolismo , Insulina/sangue , Células Secretoras de Insulina/patologia , Lipídeos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Receptor IGF Tipo 1/fisiologia , Receptor de Insulina/fisiologia
3.
Acta Trop ; 209: 105552, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32485167

RESUMO

Insulin signaling pathway is an ancient and highly conserved pathway known to play critical roles in cell growth, control and metabolic regulation. In this study, we identified and characterized two insulin receptor genes (TsIR-1316 and TsIR-4810) from Taenia solium. TsIR-1316 was grouped with E. multilocularis insulin receptor (EmIR-1) and TsIR-4810 was closer to Taenia pisiformis insulin-like growth factor receptor (TpIR) on the same branch with a very high bootstrap value. TsIR-1316 was located on the integument of larvae and adult worms, as well as the ovary of adults and eggs. Alternatively, TsIR-4810 was located in the parenchyma and reproductive organs of the adult worms. By using in vitro cultivation systems with Cysticercus pisiformis as a model, we demonstrated that anti-TsIRs-LBD antibodies could effectively block the insulin signaling pathway, resulting in reduced phosphorylation of the insulin receptor as well as lower levels of glucose uptake and glycogen synthesis. The rabbits immunized with TsIR-1316-LBD, TsIR-4810-LBD and TsIR-1316-LBD + TsIR-4810-LBD produced protection against infection of T. pisiformis as demonstrated by a 94.6%, 96% and 80% reduction of establishment of larvae, respectively. These data suggested that TsIR-1316-LBD and TsIR-4810-LBD are promising vaccine candidates or novel drug targets against swine cysticercosis.


Assuntos
Cisticercose/prevenção & controle , Receptor de Insulina/imunologia , Taenia solium/imunologia , Vacinas/imunologia , Animais , Cisticercose/tratamento farmacológico , Feminino , Coelhos , Receptor de Insulina/antagonistas & inibidores , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Suínos , Doenças dos Suínos/prevenção & controle
4.
Proc Natl Acad Sci U S A ; 117(12): 6733-6740, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32156724

RESUMO

Insulin action in the liver is critical for glucose homeostasis through regulation of glycogen synthesis and glucose output. Arrestin domain-containing 3 (Arrdc3) is a member of the α-arrestin family previously linked to human obesity. Here, we show that Arrdc3 is differentially regulated by insulin in vivo in mice undergoing euglycemic-hyperinsulinemic clamps, being highly up-regulated in liver and down-regulated in muscle and fat. Mice with liver-specific knockout (KO) of the insulin receptor (IR) have a 50% reduction in Arrdc3 messenger RNA, while, conversely, mice with liver-specific KO of Arrdc3 (L-Arrdc3 KO) have increased IR protein in plasma membrane. This leads to increased hepatic insulin sensitivity with increased phosphorylation of FOXO1, reduced expression of PEPCK, and increased glucokinase expression resulting in reduced hepatic glucose production and increased hepatic glycogen accumulation. These effects are due to interaction of ARRDC3 with IR resulting in phosphorylation of ARRDC3 on a conserved tyrosine (Y382) in the carboxyl-terminal domain. Thus, Arrdc3 is an insulin target gene, and ARRDC3 protein directly interacts with IR to serve as a feedback regulator of insulin action in control of liver metabolism.


Assuntos
Arrestinas/fisiologia , Glucose/metabolismo , Resistência à Insulina , Insulina/farmacologia , Fígado/metabolismo , Receptor de Insulina/fisiologia , Animais , Membrana Celular/metabolismo , Proteína Forkhead Box O1/metabolismo , Hipoglicemiantes/farmacologia , Fígado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação
5.
F1000Res ; 9: 598, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33552476

RESUMO

Since the discovery of insulin and insulin receptors (IR) in the brain in 1978, numerous studies have revealed a fundamental role of IR in the central nervous system and its implication in regulating synaptic plasticity, long-term potentiation and depression, neuroprotection, learning and memory, and energy balance. Central insulin resistance has been found in diverse brain disorders including Alzheimer's disease (AD). Impaired insulin signaling in AD is evident in the activation states of IR and downstream signaling molecules. This is mediated by Aß oligomer-evoked Ca 2+ influx by activating N-methyl-D-aspartate receptors (NMDARs) with Aß oligomers directly, or indirectly through Aß-induced release of glutamate, an endogenous NMDAR ligand. In the present opinion article, we highlight evidence that IR and free intracellular Ca 2+ concentration [Ca 2+] i form a double-negative regulatory feedback loop controlling insulin sensitivity, in which mitochondria play a key role, being involved in adenosine triphosphate (ATP) synthesis and IR activation. We found recently that the glutamate-evoked rise in [Ca 2+] i inhibits activation of IR and, vice versa, insulin-induced activation of IR inhibits the glutamate-evoked rise in [Ca 2+] i . In theory, such a double-negative feedback loop generates bistability. Thus, a stable steady state could exist with high [Ca 2+] i and nonactive IR, or with active IR and low [Ca 2+] i, but no stable steady state is possible with both high [Ca 2+] i and active IR. Such a circuit could toggle between a high [Ca 2+] i state and an active IR state in response to glutamate and insulin, respectively. This model predicts that any condition leading to an increase of [Ca 2+] i may trigger central insulin resistance and explains why central insulin resistance is implicated in the pathogenesis of AD, with which glutamate excitotoxicity is a comorbid condition. The model also predicts that any intervention aiming to maintain low [Ca 2+] i may be useful for treating central insulin resistance.


Assuntos
Cálcio/fisiologia , Retroalimentação Fisiológica , Resistência à Insulina , Receptor de Insulina/fisiologia , Doença de Alzheimer , Ácido Glutâmico/fisiologia , Humanos , Receptores de N-Metil-D-Aspartato/fisiologia
6.
Gac Med Mex ; 155(5): 541-545, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695236

RESUMO

The metabolic syndrome describes a group of signs that increase the likelihood for developing type 2 diabetes mellitus, cardiovascular diseases and some types of cancer. The action of insulin depends on its binding to membrane receptors on its target cells. We wonder if blood insulin could travel bound to proteins and if, in the presence of hyperinsulinemia, a soluble insulin receptor might be generated. We used young adult Wistar rats (which have no predisposition to obesity or diabetes), whose drinking water was added 20 % of sugar and that were fed a standard diet ad libitum for two and six months. They were compared with control rats under the same conditions, but that had running water for consumption. At two months, the rats developed central obesity, moderate hypertension, high triglyceride levels, hyperinsulinemia, glucose intolerance and insulin resistance, i.e. metabolic syndrome. Electrophoresis of the rats' plasma proteins was performed, followed by Western Blot (WB) for insulin and for the outer portion of the insulin receptor. The bands corresponding to insulin and to the receptor external part were at the same molecular weight level, 25-fold higher than that of free insulin. We demonstrated that insulin, both in control animals and in those with hyperinsulinemia, travels bound to the receptor outer portion (ectodomain), which we called soluble insulin receptor, and that is released al higher amounts in response to plasma insulin increase; in rats with metabolic syndrome and hyperinsulinemia, plasma levels are much higher than in controls. Soluble insulin receptor increase in blood might be an early sign of metabolic syndrome.


El síndrome metabólico es un conjunto de signos que aumentan la probabilidad de desarrollar diabetes mellitus tipo 2, enfermedades cardiovasculares y algunos tipos de cáncer. La acción de la insulina depende de su unión a los receptores en la membrana de sus células diana. Para responder a la pregunta de si la insulina en la sangre podría viajar unida a proteínas y si en presencia de hiperinsulinemia podría generarse un receptor soluble de insulina, utilizamos ratas wistar (no tienen predisposición a la obesidad ni a la diabetes), adultas jóvenes, a cuya agua de consumo se adicionó 20 % de azúcar y a las que se les administró dieta estándar ad libitum, durante dos y seis meses; fueron comparadas con ratas control que tuvieron las mismas condiciones, pero con agua corriente para consumo. A los dos meses, las ratas desarrollaron obesidad central, hipertensión moderada, triglicéridos altos, hiperinsulinemia, intolerancia a la glucosa y resistencia a la insulina, es decir, síndrome metabólico. Se realizó electroforesis de las proteínas del plasma de las ratas, seguida de Western Blot para insulina y para la porción externa del receptor de insulina. Las bandas correspondientes a la insulina y la parte externa del receptor estaban al mismo nivel de peso molecular, 25 veces mayor que el de la insulina libre. Demostramos que la insulina, tanto en animales testigo como en aquellos con hiperinsulinemia, viaja unida a la porción externa del receptor (ectodominio), al cual denominamos receptor soluble de insulina, que se libera en mayor cantidad en respuesta al incremento en la insulina plasmática; en las ratas con síndrome metabólico e hiperinsulinemia, los niveles en plasma son mucho mayores que en los controles. El incremento del receptor soluble de insulina en sangre podría ser un dato temprano de síndrome metabólico.


Assuntos
Antígenos CD/sangue , Insulina/sangue , Síndrome Metabólica/sangue , Receptor de Insulina/sangue , Animais , Antígenos CD/fisiologia , Western Blotting , Diabetes Mellitus Tipo 2/etiologia , Modelos Animais de Doenças , Eletroforese , Hiperinsulinismo/sangue , Insulina/fisiologia , Resistência à Insulina , Síndrome Metabólica/etiologia , Ratos , Ratos Wistar , Receptor de Insulina/fisiologia
7.
Exp Gerontol ; 128: 110753, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31648012

RESUMO

Buckwheat trypsin inhibitor (BTI) is a low molecular weight polypeptide that can help to prevent metabolic diseases such as obesity, hyperglycemia and hyperlipidemia. Herein, the effects of recombinant BTI (rBTI) on fat accumulation in Caenorhabditis elegans were studied. rBTI prevented fat accumulation under normal and high glucose conditions, and led to significantly shorter body widths without affecting C. elegans feeding behavior. Results also indicate that rBTI altered fat breakdown, synthesis, and accumulation by altering the transcription, expression and activity of key enzymes in lipolysis and fat synthesis. In daf-2 and daf-16 mutants, rBTI did not prevent fat accumulation, indicating that rBTI activity relies on the insulin/insulin-like growth factor (IIS) pathway. Overall rBTI may regulate changes in lipolysis and fat synthesis by down-regulating the IIS pathway, which can affect fat accumulation. These findings support the application of rBTI in preventing obesity, hyperglycemia and hyperlipemia.


Assuntos
Tecido Adiposo/metabolismo , Fagopyrum/química , Insulina/fisiologia , Somatomedinas/fisiologia , Inibidores da Tripsina/farmacologia , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiologia , Restrição Calórica , Fatores de Transcrição Forkhead/fisiologia , Lipólise/efeitos dos fármacos , Receptor de Insulina/fisiologia , Proteínas Recombinantes/farmacologia , Reprodução/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
J Clin Endocrinol Metab ; 104(6): 2216-2228, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30657911

RESUMO

CONTEXT: Insulin and leptin may increase growth and proliferation of thyroid cells, underlying an association between type 2 diabetes and papillary thyroid cancer (PTC). Patients with extreme insulin resistance due to lipodystrophy or insulin receptor mutations (INSR) are treated with high-dose insulin and recombinant leptin (metreleptin), which may increase the risk of thyroid neoplasia. OBJECTIVE: The aim of this study was to analyze thyroid structural abnormalities in patients with lipodystrophy and INSR mutations and to assess whether insulin, IGF-1, and metreleptin therapy contribute to the thyroid growth and neoplasia in this population. DESIGN: Thyroid ultrasound characteristics were analyzed in 81 patients with lipodystrophy and 11 with INSR (5 homozygous; 6 heterozygous). Sixty patients were taking metreleptin. RESULTS: The prevalence of thyroid nodules in children with extreme insulin resistance (5 of 30, 16.7%) was significantly higher than published prevalence for children (64 of 3202; 2%), with no difference between lipodystrophy and INSR. Body surface area-adjusted thyroid volume was larger in INSR homozygotes vs heterozygotes or lipodystrophy (10.4 ± 5.1, 3.9 ± 1.5, and 6.2 ± 3.4 cm2, respectively. Three patients with lipodystrophy and one INSR heterozygote had PTC. There were no differences in thyroid ultrasound features in patients treated vs not treated with metreleptin. CONCLUSION: Children with extreme insulin resistance had a high prevalence of thyroid nodules, which were not associated with metreleptin treatment. Patients with homozygous INSR mutation had thyromegaly, which may be a novel phenotypic feature of this disease. Further studies are needed to determine the etiology of thyroid abnormalities in patients with extreme insulin resistance.


Assuntos
Resistência à Insulina , Lipodistrofia/patologia , Mutação , Receptor de Insulina/genética , Glândula Tireoide/patologia , Adolescente , Adulto , Idoso , Criança , Cistos/patologia , Feminino , Humanos , Fator de Crescimento Insulin-Like I/análise , Fator de Crescimento Insulin-Like II/análise , Leptina/análogos & derivados , Leptina/farmacologia , Leptina/uso terapêutico , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Pessoa de Meia-Idade , Receptor de Insulina/fisiologia , Síndrome , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/fisiologia , Adulto Jovem
9.
FASEB J ; 33(2): 2241-2251, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30332298

RESUMO

Diabetes mellitus prevalence is increasing rapidly and is a major cause of mortality and morbidity worldwide. In addition to the known severe complications associated with the disease, in recent years diabetes has been recognized as a major risk factor for cancer. Patients with diabetes experience significantly higher incidence of and higher mortality rates from many types of cancer. However, to date there are no conclusive data on the pathophysiology underlying the association between these two diseases. We previously reported that insulin regulates skin proliferation and differentiation, while IGF1 had different sometimes contrasting effects to those of insulin, suggesting direct involvement of insulin in transformation. To this end, we developed an epidermal skin-specific insulin receptor knockout (SIRKO) mouse, in which the insulin receptor (IR) is inactivated only in skin, with no other metabolic consequences. We found that IR inactivation by itself resulted in a marked decrease in skin tumorigenesis. In the control group 100% of the mice developed tumors, but in the SIRKO group tumor incidence was over 60% lower, and 25% of the SIRKO mice did not develop tumors at all, and the tumors that did develop were smaller and benign in their appearance. Furthermore, IR inactivation in vitro not only prevented cell transformation but also reversed the keratinocyte-transformed phenotype. We found that IR inactivation led to a striking abnormality in the major keratin cytoskeleton filaments structure in both in vivo and in vitro, a change that we were able to link to the decreased transformation potential in IR-null cells. In summary, we identified a unique pathway in which IR regulates cytoskeletal assembly, thus affecting skin transformation, opening a new potential target for cancer treatment and prevention.-Weingarten, G., Ben Yaakov, A., Dror, E., Russ, J., Magin, T. M., Kahn, C. R., Wertheimer, E. Insulin receptor plays a central role in skin carcinogenesis by regulating cytoskeleton assembly.


Assuntos
Citoesqueleto/fisiologia , Receptor de Insulina/fisiologia , Fenômenos Fisiológicos da Pele , Pele/fisiopatologia , Animais , Humanos , Queratinas/genética , Camundongos
10.
Diabetes Obes Metab ; 20 Suppl 2: 51-63, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230175

RESUMO

The classical crystal structure of insulin was determined in 1969 by D.C. Hodgkin et al. following a 35-year program of research. This structure depicted a hexamer remarkable for its self-assembly as a zinc-coordinated trimer of dimer. Prominent at the dimer interface was an "aromatic triplet" of conserved residues at consecutive positions in the B chain: PheB24 , PheB25 and TyrB26 . The elegance of this interface inspired the Oxford team to poetry: "A thing of beauty is a joy forever" (John Keats as quoted by Blundell, T.L., et al. Advances in Protein Chemistry 26:279-286 [1972]). Here, we revisit this aromatic triplet in light of recent advances in the structural biology of insulin bound as a monomer to fragments of the insulin receptor. Such co-crystal structures have defined how these side chains pack at the primary hormone-binding surface of the receptor ectodomain. On receptor binding, the B-chain ß-strand (residues B24-B28) containing the aromatic triplet detaches from the α-helical core of the hormone. Whereas TyrB26 lies at the periphery of the receptor interface and may functionally be replaced by a diverse set of substitutions, PheB24 and PheB25 engage invariant elements of receptor domains L1 and αCT. These critical contacts were anticipated by the discovery of diabetes-associated mutations at these positions by Donald Steiner et al. at the University of Chicago. Conservation of PheB24 , PheB25 and TyrB26 among vertebrate insulins reflects the striking confluence of structure-based evolutionary constraints: foldability, protective self-assembly and hormonal activity.


Assuntos
Insulina/química , Humanos , Hidrocarbonetos Aromáticos/química , Insulina/metabolismo , Ligação Proteica/fisiologia , Dobramento de Proteína , Elementos Estruturais de Proteínas , Receptor de Insulina/química , Receptor de Insulina/fisiologia
11.
J Physiol ; 596(19): 4753-4765, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30044494

RESUMO

KEY POINTS: Insulin enters the brain from the blood via a saturable transport system. It is unclear how insulin is transported across the blood-brain barrier (BBB). Using two models of the signalling-related insulin receptor loss or inhibition, we show insulin transport can occur in vivo without the signalling-related insulin receptor. Insulin in the brain has multiple roles including acting as a metabolic regulator and improving memory. Understanding how insulin is transported across the BBB will aid in developing therapeutics to further increase CNS concentrations. ABSTRACT: A saturable system transports insulin from blood across the blood-brain barrier (BBB) and into the central nervous system. Whether or not the classic or signalling-related insulin receptor plays a role in mediating this transport in vivo is controversial. Here, we employed kinetics methods that distinguish between transport across the brain endothelial cell and reversible luminal surface receptor binding. Using a previously established line of mice with endothelial-specific loss of the signalling-related insulin receptor (EndoIRKO) or inhibiting the insulin receptor with the selective antagonist S961, we show insulin transport across the BBB is maintained. Rates of insulin transport were similar in all groups and transport was still saturable. Unlike transport, binding of insulin to the brain endothelial cell was decreased with the loss or inhibition of the signalling-related insulin receptor. These findings demonstrate that the signalling-related insulin receptor is not required for insulin transport across the BBB.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/fisiologia , Células Endoteliais/metabolismo , Insulina/metabolismo , Receptor de Insulina/fisiologia , Animais , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Permeabilidade da Membrana Celular , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Peptídeos/farmacologia , Receptor de Insulina/antagonistas & inibidores
12.
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
13.
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
14.
J Am Soc Hypertens ; 12(2): 135-145, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29289466

RESUMO

Both renin-angiotensin systems and insulin participate in kidney-involved blood pressure regulation. Activation of angiotensin II type 2 receptor (AT2R) decreases sodium reabsorption in renal proximal tubule (RPT) cells, whereas insulin produces the opposite effect. We presume that AT2R has an inhibitory effect on insulin receptor expression in RPT cells, which may affect renal sodium transport and therefore be of physiological or pathological significance. Our present study found that activation of AT2R inhibited insulin receptor expression in a concentration and time-dependent manner in RPT cells from Wistar-Kyoto (WKY) rats. In the presence of a protein kinase C (PKC) inhibitor (PKC inhibitor peptide 19-31, 10-6 mol/L) or a phosphatidylinositol 3 kinase inhibitor (wortmannin, 10-6 mol/L), the inhibitory effect of AT2R on insulin receptor was blocked, indicating that both PKC and phosphatidylinositol 3 kinase were involved in the signaling pathway. There was a linkage between AT2R and insulin receptor which was determined by both laser confocal microscopy and coimmunoprecipitation. However, the effect of AT2R activation on insulin receptor expression was different in RPT cells from spontaneously hypertensive rats (SHRs). Being contrary to the effect in WKY RPT cells, AT2R stimulation increased insulin receptor in SHR RPT cells. Insulin (10-7 mol/L, 15 minutes) enhanced Na+-K+-ATPase activity in both WKY and SHR RPT cells. Pretreatment with CGP42112 decreased the stimulatory effect of insulin on Na+-K+-ATPase activity in WKY RPT cells, whereas pretreatment with CGP42112 increased it in SHR RPT cells. It is suggested that activation of AT2R inhibits insulin receptor expression and function in RPT cells. The lost inhibitory effect of AT2R on insulin receptor expression may contribute to the pathophysiology of hypertension.


Assuntos
Pressão Sanguínea/fisiologia , Túbulos Renais Proximais/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Receptor de Insulina/metabolismo , Sistema Renina-Angiotensina/fisiologia , Animais , Linhagem Celular , Regulação da Expressão Gênica/fisiologia , Hipertensão/patologia , Túbulos Renais Proximais/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptor de Insulina/fisiologia , Eliminação Renal , Sistema Renina-Angiotensina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
15.
Endocr Rev ; 38(5): 379-431, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973479

RESUMO

The insulin receptor (IR) gene undergoes differential splicing that generates two IR isoforms, IR-A and IR-B. The physiological roles of IR isoforms are incompletely understood and appear to be determined by their different binding affinities for insulin-like growth factors (IGFs), particularly for IGF-2. Predominant roles of IR-A in prenatal growth and development and of IR-B in metabolic regulation are well established. However, emerging evidence indicates that the differential expression of IR isoforms may also help explain the diversification of insulin and IGF signaling and actions in various organs and tissues by involving not only different ligand-binding affinities but also different membrane partitioning and trafficking and possibly different abilities to interact with a variety of molecular partners. Of note, dysregulation of the IR-A/IR-B ratio is associated with insulin resistance, aging, and increased proliferative activity of normal and neoplastic tissues and appears to sustain detrimental effects. This review discusses novel information that has generated remarkable progress in our understanding of the physiology of IR isoforms and their role in disease. We also focus on novel IR ligands and modulators that should now be considered as an important strategy for better and safer treatment of diabetes and cancer and possibly other IR-related diseases.


Assuntos
Receptor de Insulina/fisiologia , Envelhecimento , Processamento Alternativo/fisiologia , Proliferação de Células , Diabetes Mellitus , Humanos , Insulina/metabolismo , Resistência à Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , MicroRNAs/fisiologia , Estrutura Molecular , Neoplasias/patologia , Proinsulina , Isoformas de Proteínas/fisiologia , Receptor de Insulina/química , Receptor de Insulina/genética , Transdução de Sinais , Somatomedinas/metabolismo
16.
FASEB J ; 31(8): 3555-3573, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28442548

RESUMO

Keratins (Ks) are epithelial cell intermediate filament (IF) proteins that are expressed as pairs in a differentiation-regulated manner. Hepatocyte IFs are made only of K8/K18 pairs, which means that a K8 loss in K8-null mice leads to degradation of K18. Functionally, there is accumulating evidence that IFs contribute to signaling platforms. Here, we investigate the role of K8/K18 IFs in the regulation of insulin receptor (IR) signaling and trafficking in hepatocytes. We find that the IR substrate 1 (IRS1)/PI3K/Akt signaling cascade-downstream of IR-displays prolonged activation in K8-null compared with wild-type hepatocytes. Assessment of the Akt/mammalian target of rapamycin complex 1-mediated feedback loop to IRS1/PI3K, in the absence or presence of drug inhibitors, further supports a preferential K8/K18 IF intervention at the surface membrane. In K8-null hepatocytes, IR trafficking vesicles that are labeled by Rab5/EEA1/phosphatidylinositol 3-phosphate accumulate at a juxtanuclear region via a microtubule-dependent process. Moreover, interference with phosphatidylinositol 4,5-biphosphate signaling aggravates IR/Rab5 accumulation. Overall, results uncover K8/K18 IF regulation of IR signaling via a concerted modulation of phosphatidylinositol 4,5-biphosphate-dependent IRS1/PI3K/Akt signaling and Rab5/phosphatidylinositol 3-phosphate/microtubule trafficking in hepatocytes.-Roux, A., Loranger, A., Lavoie, J. N., Marceau, N. Keratin 8/18 regulation of insulin receptor signaling and trafficking in hepatocytes through a concerted phosphoinositide-dependent Akt and Rab5 modulation.


Assuntos
Hepatócitos/fisiologia , Queratina-18/metabolismo , Queratina-8/metabolismo , Fosfatidilinositóis/metabolismo , Receptor de Insulina/fisiologia , Transdução de Sinais/fisiologia , Animais , Queratina-18/genética , Queratina-8/genética , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/metabolismo
17.
Compr Physiol ; 7(2): 741-764, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28333388

RESUMO

The ability of the brain to directly control glucose levels in the blood independently of its effects on food intake and body weight has been known ever since 1854 when Claude Bernard, a French physiologist, discovered that lesioning the floor of the fourth ventricle in rabbits led to a rise of sugar in the blood. Despite this outstanding discovery at that time, it took more than 140 years before progress started to be made in identifying the underlying mechanisms of brain-mediated control of glucose homeostasis. Technological advances including the generation of brain insulin receptor null mice revealed that insulin action specifically in the central nervous system is required for the regulation of glucose metabolism, particularly in the modulation of hepatic glucose production. Furthermore, it was established that the hormone leptin, known for its role in regulating food intake and body weight, actually exerts its most potent effects on glucose metabolism, and that this function of leptin is mediated centrally. Under certain circumstances, high levels of leptin can replicate the actions of insulin, thus challenging the idea that life without insulin is impossible. Disruptions of central insulin signaling and glucose metabolism not only lead to impairments in whole body glucose homeostasis, they also have other serious consequences, including the development of Alzheimer's disease which is sometimes referred to as type 3 diabetes reflecting its common etiology with type 2 diabetes. © 2017 American Physiological Society. Compr Physiol 7:471-764, 2017.


Assuntos
Glicemia/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/efeitos adversos , Homeostase/fisiologia , Humanos , Doenças Hipotalâmicas/etiologia , Inflamação/etiologia , Insulina/fisiologia , Leptina/metabolismo , Leptina/fisiologia , Receptor de Insulina/fisiologia , Via de Sinalização Wnt/fisiologia
18.
J Neuroendocrinol ; 27(12): 899-910, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26485112

RESUMO

Insulin signalling in the brain plays an important role in the central regulation of energy homeostasis and fertility, such that mice exhibiting widespread deletion of insulin receptors (InsR) throughout the brain and peripheral nervous system display diet sensitive obesity and hypothalamic hypogonadism. However, the specific cell types mediating the central effects of insulin on fertility remain largely unidentified. To date, the targeted deletion of InsR from individual neuronal populations implicated in the metabolic control of fertility has failed to recapitulate the hypogonadic and subfertile phenotype observed in brain-specific InsR knockout mice. Because insulin and leptin share similar roles as centrally-acting metabolic regulators of fertility, we used the Cre-loxP system to generate mice with a selective inactivation of the Insr gene from the same widespread neuronal population previously shown to mediate the central effects of leptin on fertility by crossing Insr-flox mice with calcium/calmodulin-dependent protein kinase type IIα (CamkIIα)-Cre mice. Multiple reproductive and metabolic parameters were then compared between male and female Insr-flox/Cre-positive (CamK-IRKO) and Insr-flox/Cre-negative control mice. Consistent with brain-specific InsR knockout mice, CamK-IRKO mice exhibited a mild but significant obesogenic phenotype. Unexpectedly, CamK-IRKO mice exhibited normal reproductive maturation and function compared to controls. No differences in the age of puberty onset, oestrous cyclicity or fecundity were observed between CamK-IRKO and control mice. We conclude that the central effects of insulin on the neuroendocrine reproductive axis are not critically mediated via the same neuronal populations targeted by leptin.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Insulina/fisiologia , Neurônios/metabolismo , Reprodução/fisiologia , Animais , Feminino , Insulina/farmacologia , Leptina/farmacologia , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/biossíntese , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Reprodução/genética
19.
Metabolism ; 64(12): 1682-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26455966

RESUMO

OBJECTIVE: Evidence shows that both macrophage migration inhibitory factor (MIF) and GLUT4 glucose transporter are involved in diabetic cardiomyopathy (DCM), but it remains largely unknown whether and how MIF regulates GLUT4 expression in cardiomyocytes. The present study aims to investigate the mechanism underlying the modulation of GLUT4 by MIF in cardiomyocytes. MATERIAL AND METHODS: Activations of AKT and AMPK signaling, and expressions of MIF, GLUT4 and the candidate GLUT4 regulation associated transcription factors in the diabetic mouse myocardium were determined. The screened transcription factors mediating MIF-promoted GLUT4 expression were verified by RNA interference (RNAi) and electrophoretic mobility shift assay (EMSA), respectively. RESULTS: MIF was increased, but GLUT4 was decreased in the diabetic mouse myocardium. MIF could enhance glucose uptake and up-regulate GLUT4 expression in NMVCs. Expressions of transcription factor MEF2A, -2C, -2D and Zac1 were significantly up-regulated in MIF-treated neonatal mouse ventricular cardiomyocytes (NMVCs), and markedly reduced in the diabetic myocardium. Knockdown of MEF2A, -2C, -2D and Zac1 could significantly inhibit glucose uptake and GLUT4 expression in cardiomyocytes. Moreover, EMSA results revealed that transcriptional activities of MEF2 and Zac1 were significantly increased in MIF-treated NMVCs. AMPK signaling was activated in MIF-stimulated NMVCs, and AMPK activator AICAR could enhance MEF2A, -2C, -2D, Zac1 and GLUT4 expression. Additionally, MIF effects were inhibited by an AMPK inhibitor compound C and siRNA targeting MIF receptor CD74, suggesting the involvement of CD74-dependent AMPK activation. CONCLUSIONS: Transcription factor MEF2 and Zac1 mediate MIF-induced GLUT4 expression through CD74-dependent AMPK activation in cardiomyocytes.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Genes Supressores de Tumor/fisiologia , Transportador de Glucose Tipo 4/genética , Oxirredutases Intramoleculares/fisiologia , Fatores de Transcrição MEF2/fisiologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Antígenos de Diferenciação de Linfócitos B/fisiologia , Células Cultivadas , Cardiomiopatias Diabéticas/fisiopatologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Insulina/fisiologia , Função Ventricular Esquerda
20.
Nefrologia ; 35(2): 131-8, 2015.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-26300505

RESUMO

The prevalence of diabetes mellitus increased during the last century and it is estimated that 45% of the patients are not diagnosed. In South America the prevalence of diabetes and chronic kidney disease (CKD) increased, with a great disparity among the countries with respect to access to dialysis. In Ecuador it is one of the main causes of mortality, principally in the provinces located on the coast of the Pacific Ocean. The greatest single cause of beginning dialysis is diabetic nephropathy (DN). Even using the best therapeutic options for DN, the residual risk of proteinuria and of terminal CKD remains high. In this review we indicate the importance of the problem globally and in our region. We analyse relevant cellular and molecular studies that illustrate the crucial significance of glomerular events in DN development and evolution and in insulin resistance. We include basic anatomical, pathophysiological and clinical concepts, with special attention to the role of angiogenic factors such as the vascular endothelial growth factor (VEGF-A) and their relationship to the insulin receptor, endothelial isoform of nitric oxide synthase (eNOS) and angiopoietins. We also propose various pathways that have therapeutic potential in our opinion. Greater in-depth study of VEGF-A and angiopoietins, the state of glomerular VEGF resistance, the relationship of VEGF receptor 2/nephrin, VEGF/insulin receptors/nephrin and the relationship of VEGF/eNOS-NO at glomerular level could provide solutions to the pressing world problem of DN and generate new treatment alternatives.


Assuntos
Nefropatias Diabéticas/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Angiopoietinas/fisiologia , Nefropatias Diabéticas/epidemiologia , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Saúde Global , Humanos , Resistência à Insulina , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Proteínas de Membrana/fisiologia , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Receptor de Insulina/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA