Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Transl Res ; 205: 51-63, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30452888

RESUMO

In diabetes, stromal cell-derived factor-1 (SDF-1) expression and progenitor cell recruitment are reduced. Dipeptidyl peptidase-4 (DPP-4) inhibits SDF-1 expression and progenitor cell recruitment. Here we examined the impact of the DPP-4 inhibitor, MK0626, on progenitor cell kinetics in the context of wound healing. Wildtype (WT) murine fibroblasts cultured under high-glucose to reproduce a diabetic microenvironment were exposed to MK0626, glipizide, or no treatment, and SDF-1 expression was measured with ELISA. Diabetic mice received MK0626, glipizide, or no treatment for 6 weeks and then were wounded. Immunohistochemistry was used to quantify neovascularization and SDF-1 expression. Gene expression was measured at the RNA and protein level using quantitative polymerase chain reaction and ELISA, respectively. Flow cytometry was used to characterize bone marrow-derived mesenchymal progenitor cell (BM-MPC) population recruitment to wounds. BM-MPC gene expression was assayed using microfluidic single cell analysis. WT murine fibroblasts exposed to MK0626 demonstrated increased SDF-1 expression. MK0626 treatment significantly accelerated wound healing and increased wound vascularity, SDF-1 expression, and dermal thickness in diabetic wounds. MK0626 treatment increased the number of BM-MPCs present in bone marrow and in diabetic wounds. MK0626 had no effect on BM-MPC population dynamics. BM-MPCs harvested from MK0626-treated mice exhibited increased chemotaxis in response to SDF-1 when compared to diabetic controls. Treatment with a DPP-4 inhibitor significantly improved wound healing, angiogenesis, and endogenous progenitor cell recruitment in the setting of diabetes.


Assuntos
Diabetes Mellitus Experimental/complicações , Dipeptidil Peptidase 4/efeitos dos fármacos , Inibidores da Dipeptidil Peptidase IV/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Neovascularização Patológica , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões/fisiopatologia , Animais , Quimiocina CXCL12/metabolismo , Glipizida/farmacologia , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Triazóis/farmacologia
2.
Soft Matter ; 13(48): 9132-9137, 2017 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-29184951

RESUMO

Granular materials jam when developing a network of contact forces able to resist the applied stresses. Through numerical simulations of the dynamics of the jamming process, we show that the jamming transition does not occur when the kinetic energy vanishes. Rather, as the system jams, the kinetic energy becomes dominated by rattler particles, which scatter within their cages. The relaxation of the kinetic energy in the jammed configuration exhibits a double power-law decay, which we interpret in terms of the interplay between backbone and rattler particles.

3.
Diabetes ; 65(6): 1699-713, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26956489

RESUMO

The mechanisms for the development of diabetic cardiomyopathy remain largely unknown. Methylglyoxal (MG) can accumulate and promote inflammation and vascular damage in diabetes. We examined if overexpression of the MG-metabolizing enzyme glyoxalase 1 (GLO1) in macrophages and the vasculature could reduce MG-induced inflammation and prevent ventricular dysfunction in diabetes. Hyperglycemia increased circulating inflammatory markers in wild-type (WT) but not in GLO1-overexpressing mice. Endothelial cell number was reduced in WT-diabetic hearts compared with nondiabetic controls, whereas GLO1 overexpression preserved capillary density. Neuregulin production, endothelial nitric oxide synthase dimerization, and Bcl-2 expression in endothelial cells was maintained in the hearts of GLO1-diabetic mice and corresponded to less myocardial cell death compared with the WT-diabetic group. Lower receptor for advanced glycation end products and tumor necrosis factor-α (TNF-α) levels were also observed in GLO1-diabetic versus WT-diabetic mice. Over a period of 8 weeks of hyperglycemia, GLO1 overexpression delayed and limited the loss of cardiac function. In vitro, MG and TNF-α were shown to synergize in promoting endothelial cell death, which was associated with increased angiopoietin 2 expression and reduced Bcl-2 expression. These results suggest that MG in diabetes increases inflammation, leading to endothelial cell loss. This contributes to the development of diabetic cardiomyopathy and identifies MG-induced endothelial inflammation as a target for therapy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/etiologia , Células Endoteliais/metabolismo , Lactoilglutationa Liase/metabolismo , Aldeído Pirúvico/metabolismo , Angiopoietina-2/metabolismo , Animais , Estudos de Casos e Controles , Morte Celular , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Genes bcl-2 , Produtos Finais de Glicação Avançada/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/metabolismo , Miocárdio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
Diabetes ; 64(9): 3273-84, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26294429

RESUMO

The assumption underlying current diabetes treatment is that lowering the level of time-averaged glucose concentrations, measured as HbA1c, prevents microvascular complications. However, 89% of variation in risk of retinopathy, microalbuminuria, or albuminuria is due to elements of glycemia not captured by mean HbA1c values. We show that transient exposure to high glucose activates a multicomponent feedback loop that causes a stable left shift of the glucose concentration-reactive oxygen species (ROS) dose-response curve. Feedback loop disruption by the GLP-1 cleavage product GLP-1(9-36)(amide) reverses the persistent left shift, thereby normalizing persistent overproduction of ROS and its pathophysiologic consequences. These data suggest that hyperglycemic spikes high enough to activate persistent ROS production during subsequent periods of normal glycemia but too brief to affect the HbA1c value are a major determinant of the 89% of diabetes complications risk not captured by HbA1c. The phenomenon and mechanism described in this study provide a basis for the development of both new biomarkers to complement HbA1c and novel therapeutic agents, including GLP-1(9-36)(amide), for the prevention and treatment of diabetes complications.


Assuntos
Complicações do Diabetes/metabolismo , Retroalimentação Fisiológica , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Glucose/metabolismo , Hiperglicemia/metabolismo , Mitocôndrias/metabolismo , Peptídeos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular , Células Endoteliais/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hemoglobinas Glicadas/metabolismo , Humanos , Ferro/metabolismo , Potencial da Membrana Mitocondrial
5.
Atherosclerosis ; 239(2): 393-400, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25682038

RESUMO

OBJECTIVE: The pathogenic events responsible for accelerated atherosclerosis in patients with chronic renal failure (CRF) are poorly understood. Here we investigate the hypothesis that concentrations of urea associated with CRF and increased ROS production in adipocytes might also increase ROS production directly in arterial endothelial cells, causing the same pathophysiologic changes seen with hyperglycemia. METHODS: Primary cultures of human aortic endothelial cells (HAEC) were exposed to 20mM urea for 48 h. C57BL/6J wild-type mice underwent 5/6 nephrectomy or a sham operation. Randomized groups of 5/6 nephrectomized mice and their controls were also injected i.p. with a SOD/catalase mimetic (MnTBAP) for 15 days starting immediately after the final surgical procedure. RESULTS: Urea at concentrations seen in CRF induced mitochondrial ROS production in cultured HAEC. Urea-induced ROS caused the activation of endothelial pro-inflammatory pathways through the inhibition of GAPDH, including increased protein kinase C isoforms activity, increased hexosamine pathway activity, and accumulation of intracellular AGEs (advanced glycation end products). Urea-induced ROS directly inactivated the anti-atherosclerosis enzyme PGI2 synthase and also caused ER stress. Normalization of mitochondrial ROS production prevented each of these effects of urea. In uremic mice, treatment with MnTBAP prevented aortic oxidative stress, PGI2 synthase activity reduction and increased expression of the pro-inflammatory proteins TNFα, IL-6, VCAM1, Endoglin, and MCP-1. CONCLUSIONS: Taken together, these data show that urea itself, at levels common in patients with CRF, causes endothelial dysfunction and activation of proatherogenic pathways.


Assuntos
Endotélio Vascular/patologia , Falência Renal Crônica/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ureia/química , Animais , Antígenos CD/metabolismo , Aorta/metabolismo , Aterosclerose/enzimologia , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Catalase/metabolismo , Quimiocina CCL2/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Endoglina , Células Endoteliais/metabolismo , Endotélio/enzimologia , Endotélio Vascular/efeitos dos fármacos , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Humanos , Interleucina-6/metabolismo , Oxirredutases Intramoleculares/metabolismo , Falência Renal Crônica/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Distribuição Aleatória , Receptores de Superfície Celular/metabolismo , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
Diabetes ; 63(1): 291-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24062246

RESUMO

Differences in susceptibility to diabetic nephropathy (DN) between mouse strains with identical levels of hyperglycemia correlate with renal levels of oxidative stress, shown previously to play a central role in the pathogenesis of DN. Susceptibility to DN appears to be genetically determined, but the critical genes have not yet been identified. Overexpression of the enzyme glyoxalase 1 (Glo1), which prevents posttranslational modification of proteins by the glycolysis-derived α-oxoaldehyde, methylglyoxal (MG), prevents hyperglycemia-induced oxidative stress in cultured cells and model organisms. In this study, we show that in nondiabetic mice, knockdown of Glo1 increases to diabetic levels both MG modification of glomerular proteins and oxidative stress, causing alterations in kidney morphology indistinguishable from those caused by diabetes. We also show that in diabetic mice, Glo1 overexpression completely prevents diabetes-induced increases in MG modification of glomerular proteins, increased oxidative stress, and the development of diabetic kidney pathology, despite unchanged levels of diabetic hyperglycemia. Together, these data indicate that Glo1 activity regulates the sensitivity of the kidney to hyperglycemic-induced renal pathology and that alterations in the rate of MG detoxification are sufficient to determine the glycemic set point at which DN occurs.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Hiperglicemia/metabolismo , Rim/metabolismo , Lactoilglutationa Liase/metabolismo , Albuminúria/genética , Albuminúria/metabolismo , Albuminúria/fisiopatologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/fisiopatologia , Hiperglicemia/genética , Hiperglicemia/fisiopatologia , Rim/fisiopatologia , Lactoilglutationa Liase/genética , Camundongos , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo
7.
Biosystems ; 112(3): 258-64, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23542676

RESUMO

We consider a network of leaky integrate and fire neurons, whose learning mechanism is based on the Spike-Timing-Dependent Plasticity. The spontaneous temporal dynamic of the system is studied, including its storage and replay properties, when a Poissonian noise is added to the post-synaptic potential of the units. The temporal patterns stored in the network are periodic spatiotemporal patterns of spikes. We observe that, even in absence of a cue stimulation, the spontaneous dynamics induced by the noise is a sort of intermittent replay of the patterns stored in the connectivity and a phase transition between a replay and non-replay regime exists at a critical value of the spiking threshold. We characterize this transition by measuring the order parameter and its fluctuations.


Assuntos
Potenciais de Ação/fisiologia , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Modelos Neurológicos , Neurônios/fisiologia , Somação de Potenciais Pós-Sinápticos/fisiologia , Processos Estocásticos , Fatores de Tempo
8.
J Comput Neurosci ; 34(2): 319-36, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23053861

RESUMO

We study the collective dynamics of a Leaky Integrate and Fire network in which precise relative phase relationship of spikes among neurons are stored, as attractors of the dynamics, and selectively replayed at different time scales. Using an STDP-based learning process, we store in the connectivity several phase-coded spike patterns, and we find that, depending on the excitability of the network, different working regimes are possible, with transient or persistent replay activity induced by a brief signal. We introduce an order parameter to evaluate the similarity between stored and recalled phase-coded pattern, and measure the storage capacity. Modulation of spiking thresholds during replay changes the frequency of the collective oscillation or the number of spikes per cycle, keeping preserved the phases relationship. This allows a coding scheme in which phase, rate and frequency are dissociable. Robustness with respect to noise and heterogeneity of neurons parameters is studied, showing that, since dynamics is a retrieval process, neurons preserve stable precise phase relationship among units, keeping a unique frequency of oscillation, even in noisy conditions and with heterogeneity of internal parameters of the units.


Assuntos
Potenciais de Ação/fisiologia , Memória/fisiologia , Modelos Neurológicos , Rede Nervosa/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Animais , Humanos , Aprendizagem/fisiologia , Ruído , Dinâmica não Linear , Fatores de Tempo
9.
J Cell Physiol ; 227(4): 1485-92, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21618539

RESUMO

Clinical and experimental evidence indicates that atypical antipsychotics impair glucose metabolism. We investigated whether clozapine may directly affect insulin action by analyzing insulin signaling in vitro and in vivo. Clozapine reduced insulin-stimulated glucose uptake in PC12 and in L6 cells, representative models of neuron and skeletal muscle, respectively. Consistently, clozapine reduced insulin effect on insulin receptor (IR) by 40% and on IR substrate-1 (IRS1) tyrosine phosphorylation by 60%. Insulin-stimulated Akt phosphorylation was also reduced by about 40%. Moreover, insulin-dependent phosphorylation of protein kinase C-ζ (PKC-ζ) was completely blunted in clozapine-treated cells. Interestingly, clozapine treatment was accompanied by an insulin-independent increase of Akt phosphorylation, with no change of IR, IRS1, and PKC-ζ basal phosphorylation. The cellular abundance of Ped/Pea-15, an Akt substrate and inducer of insulin resistance, was also increased following clozapine exposure, both in the absence and in the presence of cyclohexymide, a protein synthesis inhibitor. Similar as in cellular models, in the caudate-putamen and in the tibialis muscle of clozapine-treated C57/BL/KsJ mice, Akt phosphorylation and Ped/Pea-15 protein levels were increased and PKC-ζ phosphorylation was decreased. Thus, in these experimental models, clozapine deranged Akt function and up-regulated Ped/Pea-15, thereby inhibiting insulin stimulation of PKC-ζ and of glucose uptake.


Assuntos
Antipsicóticos/farmacologia , Clozapina/farmacologia , Insulina/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular , Glucose/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células PC12 , Fosforilação , Proteína Quinase C/metabolismo , Ratos , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
10.
J Cell Physiol ; 227(5): 2106-16, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21780113

RESUMO

Cell migration is dependent on the control of signaling events that play significant roles in creating contractile force and in contributing to wound closure. We evaluated wound closure in fibroblasts from mice overexpressing (TgPED) or lacking ped/pea-15 (KO), a gene overexpressed in patients with type 2 diabetes. Cultured skin fibroblasts isolated from TgPED mice showed a significant reduction in the ability to recolonize wounded area during scratch assay, compared to control fibroblasts. This difference was observed both in the absence and in the presence of mytomicin C, an inhibitor of mitosis. In time-lapse experiments, TgPED fibroblasts displayed about twofold lower velocity and diffusion coefficient, as compared to controls. These changes were accompanied by reduced spreading and decreased formation of stress fibers and focal adhesion plaques. At the molecular level, TgPED fibroblasts displayed decreased RhoA activation and increased abundance of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2). Inhibition of ERK1/2 activity by PD98059 restored RhoA activation, cytoskeleton organization and cell motility, and almost completely rescued wound closure of TgPED fibroblasts. Interestingly, skin fibroblasts isolated from KO mice displayed an increased wound closure ability. In vivo, healing of dorsal wounds was delayed in TgPED and accelerated in KO mice. Thus, PED/PEA-15 may affect fibroblast motility by a mechanism, at least in part, mediated by ERK1/2.


Assuntos
Movimento Celular/fisiologia , Fibroblastos/fisiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfoproteínas/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Adesão Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/fisiopatologia , Fibroblastos/citologia , Flavonoides/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Fosfoproteínas/genética , Proteína rhoA de Ligação ao GTP/metabolismo
11.
Circ Res ; 107(9): 1058-70, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-21030723

RESUMO

Oxidative stress plays a pivotal role in the development of diabetes complications, both microvascular and cardiovascular. The metabolic abnormalities of diabetes cause mitochondrial superoxide overproduction in endothelial cells of both large and small vessels, as well as in the myocardium. This increased superoxide production causes the activation of 5 major pathways involved in the pathogenesis of complications: polyol pathway flux, increased formation of AGEs (advanced glycation end products), increased expression of the receptor for AGEs and its activating ligands, activation of protein kinase C isoforms, and overactivity of the hexosamine pathway. It also directly inactivates 2 critical antiatherosclerotic enzymes, endothelial nitric oxide synthase and prostacyclin synthase. Through these pathways, increased intracellular reactive oxygen species (ROS) cause defective angiogenesis in response to ischemia, activate a number of proinflammatory pathways, and cause long-lasting epigenetic changes that drive persistent expression of proinflammatory genes after glycemia is normalized ("hyperglycemic memory"). Atherosclerosis and cardiomyopathy in type 2 diabetes are caused in part by pathway-selective insulin resistance, which increases mitochondrial ROS production from free fatty acids and by inactivation of antiatherosclerosis enzymes by ROS. Overexpression of superoxide dismutase in transgenic diabetic mice prevents diabetic retinopathy, nephropathy, and cardiomyopathy. The aim of this review is to highlight advances in understanding the role of metabolite-generated ROS in the development of diabetic complications.


Assuntos
Complicações do Diabetes/metabolismo , Estresse Oxidativo/fisiologia , Animais , Aterosclerose/etiologia , Aterosclerose/metabolismo , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Complicações do Diabetes/etiologia , Humanos , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Microvasos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia
12.
Artigo em Inglês | MEDLINE | ID: mdl-21423518

RESUMO

We study the storage and retrieval of phase-coded patterns as stable dynamical attractors in recurrent neural networks, for both an analog and a integrate and fire spiking model. The synaptic strength is determined by a learning rule based on spike-time-dependent plasticity, with an asymmetric time window depending on the relative timing between pre and postsynaptic activity. We store multiple patterns and study the network capacity. For the analog model, we find that the network capacity scales linearly with the network size, and that both capacity and the oscillation frequency of the retrieval state depend on the asymmetry of the learning time window. In addition to fully connected networks, we study sparse networks, where each neuron is connected only to a small number z ≪ N of other neurons. Connections can be short range, between neighboring neurons placed on a regular lattice, or long range, between randomly chosen pairs of neurons. We find that a small fraction of long range connections is able to amplify the capacity of the network. This imply that a small-world-network topology is optimal, as a compromise between the cost of long range connections and the capacity increase. Also in the spiking integrate and fire model the crucial result of storing and retrieval of multiple phase-coded patterns is observed. The capacity of the fully-connected spiking network is investigated, together with the relation between oscillation frequency of retrieval state and window asymmetry.

13.
J Biol Chem ; 283(52): 36088-99, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18955497

RESUMO

Chronic hyperglycemia promotes insulin resistance at least in part by increasing the formation of advanced glycation end products (AGEs). We have previously shown that in L6 myotubes human glycated albumin (HGA) induces insulin resistance by activating protein kinase Calpha (PKCalpha). Here we show that HGA-induced PKCalpha activation is mediated by Src. Coprecipitation experiments showed that Src interacts with both the receptor for AGE (RAGE) and PKCalpha in HGA-treated L6 cells. A direct interaction of PKCalpha with Src and insulin receptor substrate-1 (IRS-1) has also been detected. In addition, silencing of IRS-1 expression abolished HGA-induced RAGE-PKCalpha co-precipitation. AGEs were able to induce insulin resistance also in vivo, as insulin tolerance tests revealed a significant impairment of insulin sensitivity in C57/BL6 mice fed a high AGEs diet (HAD). In tibialis muscle of HAD-fed mice, insulin-induced glucose uptake and protein kinase B phosphorylation were reduced. This was paralleled by a 2.5-fold increase in PKCalpha activity. Similarly to in vitro observations, Src phosphorylation was increased in tibialis muscle of HAD-fed mice, and co-precipitation experiments showed that Src interacts with both RAGE and PKCalpha. These results indicate that AGEs impairment of insulin action in the muscle might be mediated by the formation of a multimolecular complex including RAGE/IRS-1/Src and PKCalpha.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Insulina/metabolismo , Animais , Feminino , Teste de Tolerância a Glucose , Humanos , Hiperglicemia/patologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Quinases da Família src/metabolismo
14.
Hum Gene Ther ; 18(2): 106-17, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17328681

RESUMO

Diabetes mellitus (DM) derives from either insulin deficiency (type 1) or resistance (type 2). Insulin regulates glucose metabolism and homeostasis by binding to a specific membrane receptor (IR) with tyrosine kinase activity, expressed by its canonical target tissues. General or tissue-specific IR ablation in mice results in complex metabolic abnormalities, which give partial insights into the role of IR signaling in glucose homeostasis and diabetes development. We generated a chimeric IR (LFv2IRE) inducible on administration of the small molecule drug AP20187. This represents a powerful tool to induce insulin receptor signaling in the hormone target tissues in DM animal models. Here we use adeno-associated viral (AAV) vectors to transduce muscle and liver of nonobese diabetic (NOD) mice with LFv2IRE. Systemic AP20187 administration results in time-dependent LFv2IRE tyrosine phosphorylation and activation of the insulin signaling pathway in both liver and muscle of AAV-treated NOD mice. AP20187 stimulation significantly increases hepatic glycogen content and muscular glucose uptake similarly to insulin. The LFv2IRE-AP20187 system represents a useful tool for regulated and rapid tissue-specific restoration of IR signaling and for dissection of insulin signaling and function in the hormone canonical and noncanonical target tissues.


Assuntos
Insulina/metabolismo , Fígado/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Receptor de Insulina/metabolismo , Tacrolimo/análogos & derivados , Animais , Dependovirus , Vetores Genéticos/metabolismo , Glucose/metabolismo , Glicogênio/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Fosfotirosina/metabolismo , Receptor de Insulina/química , Proteínas Recombinantes de Fusão/metabolismo , Tacrolimo/farmacologia , Transdução Genética
15.
Diabetes ; 56(3): 622-33, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17327429

RESUMO

The phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (ped/pea-15) gene is overexpressed in human diabetes and causes this abnormality in mice. Transgenic mice with beta-cell-specific overexpression of ped/pea-15 (beta-tg) exhibited decreased glucose tolerance but were not insulin resistant. However, they showed impaired insulin response to hyperglycemia. Islets from the beta-tg also exhibited little response to glucose. mRNAs encoding the Sur1 and Kir6.2 potassium channel subunits and their upstream regulator Foxa2 were specifically reduced in these islets. Overexpression of PED/PEA-15 inhibited the induction of the atypical protein kinase C (PKC)-zeta by glucose in mouse islets and in beta-cells of the MIN-6 and INS-1 lines. Rescue of PKC-zeta activity elicited recovery of the expression of the Sur1, Kir6.2, and Foxa2 genes and of glucose-induced insulin secretion in PED/PEA-15-overexpressing beta-cells. Islets from ped/pea-15-null mice exhibited a twofold increased activation of PKC-zeta by glucose; increased abundance of the Sur1, Kir6.2, and Foxa2 mRNAs; and enhanced glucose effect on insulin secretion. In conclusion, PED/PEA-15 is an endogenous regulator of glucose-induced insulin secretion, which restrains potassium channel expression in pancreatic beta-cells. Overexpression of PED/PEA-15 dysregulates beta-cell function and is sufficient to impair glucose tolerance in mice.


Assuntos
Regulação da Expressão Gênica/fisiologia , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fosfoproteínas/metabolismo , Canais de Potássio/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular , Regulação para Baixo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Secreção de Insulina , Masculino , Camundongos , Camundongos Transgênicos , Proteína Quinase C/genética , Proteína Quinase C/metabolismo
16.
FASEB J ; 20(13): 2402-4, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17015410

RESUMO

Platelet components have found successful clinical utilization to initiate or to accelerate tissue-repair mechanisms. However, the molecular pathways by which platelet factors contribute to tissue regeneration have not been fully elucidated. We have studied the effect of thrombin-activated platelets (TAPs) on cell growth in vivo and in cultured cell systems. Application of TAPs to ulcerative skin lesions of diabetic patients induced local activation of ERK1/2 and Akt/PKB. Moreover, when applied to cultured human skin fibroblasts, TAPs promoted cell growth and DNA synthesis and activated platelet-derived growth factor (PDGF) and insulin-like growth factor (IGF)-1 receptor tyrosine kinases. PDGF was released by TAPs and rapidly achieved a plateau. At variance, the release of IGF-1 was mainly provided by the TAPs-stimulated fibroblasts and progressively increased up to 48 h. The PDGF-R blocker Ag1296 reduced the activation of Akt/PKB and, at a lesser extent, of ERK1/2. Conversely, inhibition of IGF-1 signaling by Ag1024 and expression of a dominant-negative IGF-1R mutant selectively reduced the stimulation of ERK1/2 by TAPs and fibroblast-released factors, with minor changes of Akt/PKB activity. Thus, platelet factors promote fibroblast growth by acutely activating Akt/PKB and ERK1/2. Sustained activation of ERK1/2, however, requires autocrine production of IGF-1 by TAPs-stimulated fibroblasts.


Assuntos
Plaquetas/fisiologia , Fibroblastos/fisiologia , Fator de Crescimento Insulin-Like I/biossíntese , Ativação Plaquetária , Trombina/fisiologia , Plaquetas/citologia , Técnicas de Cultura de Células , Divisão Celular , Fibroblastos/citologia , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pele/citologia , Fenômenos Fisiológicos da Pele
17.
Mol Cell Biol ; 23(13): 4511-21, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12808093

RESUMO

The antiapoptotic protein PED/PEA-15 features an Akt phosphorylation motif upstream from Ser(116). In vitro, recombinant PED/PEA-15 was phosphorylated by Akt with a stoichiometry close to 1. Based on Western blotting with specific phospho-Ser(116) PED/PEA-15 antibodies, Akt phosphorylation of PED/PEA-15 occurred mainly at Ser(116). In addition, a mutant of PED/PEA-15 featuring the substitution of Ser(116)-->Gly (PED(S116-->G)) showed 10-fold-decreased phosphorylation by Akt. In intact 293 cells, Akt also induced phosphorylation of PED/PEA-15 at Ser(116). Based on pull-down and coprecipitation assays, PED/PEA-15 specifically bound Akt, independently of Akt activity. Serum activation of Akt as well as BAD phosphorylation by Akt showed no difference in 293 cells transfected with PED/PEA-15 and in untransfected cells (which express no endogenous PED/PEA-15). However, the antiapoptotic action of PED/PEA-15 was almost twofold reduced in PED(S116-->G) compared to that in PED/PEA-15(WT) cells. PED/PEA-15 stability closely paralleled Akt activation by serum in 293 cells. In these cells, the nonphosphorylatable PED(S116-->G) mutant exhibited a degradation rate threefold greater than that observed with wild-type PED/PEA-15. In the U373MG glioma cells, blocking Akt also reduced PED/PEA-15 levels and induced sensitivity to tumor necrosis factor-related apoptosis-inducing ligand apoptosis. Thus, phosphorylation by Akt regulates the antiapoptotic function of PED/PEA-15 at least in part by controlling the stability of PED/PEA-15. In part, Akt survival signaling may be mediated by PED/PEA-15.


Assuntos
Apoptose , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Androstadienos/farmacologia , Proteínas Reguladoras de Apoptose , Sítios de Ligação , Western Blotting , Linhagem Celular , Cicloeximida/farmacologia , DNA Complementar/metabolismo , Inibidores Enzimáticos/farmacologia , Glioma/metabolismo , Glutationa Transferase/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mutação , Peptídeos/química , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes/metabolismo , Serina/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA