Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Regul Integr Comp Physiol ; 299(6): R1610-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20881100

RESUMO

Endothelial death is critical in diabetic vascular diseases, but regulating factors have been only partially elucidated. Phosphatases play important regulatory roles in cell metabolism, but have not previously been implicated in hyperglycemia-induced cell death. We investigated the role of the phosphatase, type 2A protein phosphatase (PP2A), in hyperglycemia-induced changes in signaling and death in bovine aortic endothelial cells (BAEC). We explored also the influence of benfotiamine on this phosphatase. Activation of PP2A was assessed in BAEC by the extent of methylation and measurement of activity, and the enzyme was inhibited using selective pharmacological (okadaic acid, sodium fostriecin) and molecular (small interfering RNA) approaches. BAECs cultured in 30 mM glucose significantly increased PP2A methylation and activity, and PP2A inhibitors blocked these abnormalities. PP2A activity was increased also in aorta and retina from diabetic rats. NF-κB activity and cell death in BAEC were significantly increased in 30 mM glucose and inhibited by PP2A inhibition. NF-κB played a role in the hyperglycemia-induced death of BAEC, since blocking its translocation with SN50 also inhibited cell death. Inhibition of PP2A blocked the hyperglycemia-induced dephosphorylation of NF-κB and Bad, thus favoring cell survival. Incubation of benfotiamine with BAEC inhibited the high glucose-induced activation of PP2A and NF-κB and cell death, as well as several other metabolic defects, which likewise were inhibited by inhibitors of PP2A. Activation of PP2A contributes to endothelial cell death in high glucose, and beneficial actions of benfotiamine are due, at least in part, to inhibition of PP2A activation.


Assuntos
Aorta/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/metabolismo , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Proteína Fosfatase 2/metabolismo , Tiamina/análogos & derivados , Análise de Variância , Animais , Aorta/metabolismo , Aorta/patologia , Western Blotting , Bovinos , Morte Celular/fisiologia , Células Cultivadas , Diabetes Mellitus Experimental/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Ensaio de Imunoadsorção Enzimática , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Endogâmicos Lew , Tiamina/farmacologia , Transfecção
2.
J Cell Mol Med ; 12(5A): 1571-83, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18053091

RESUMO

Histone (de)acetylases control gene transcription via modification of the chromatin structure. Herein, we investigated potential roles for histone deacetylation (or hypoacetylation) in interleukin-1beta (IL-1beta)-mediated inducible nitric oxide synthase (iNOS) and nitric oxide (NO) release in insulin-secreting INS 832/13 (INS) cells. Western blot analysis suggested localization of members of Class 1 and Class 2 families of histone deacetylases (HDACs) in these cells. Trichostatin A (TSA), a known inhibitor of HDACs, markedly reduced IL-1beta-mediated iNOS expression and NO release from these cells in a concentration-dependent manner. TSA also promoted hyperacetylation of histone H4 under conditions in which it inhibited IL-1beta-mediated effects on isolated beta cells. Rottlerin, a known inhibitor of protein kinase Cdelta, also increased histone H4 acetylation, and inhibited IL-1beta-induced iNOS expression and NO release in these cells. It appears that the putative mechanism underlying the stimulatory effects of rottlerin on acetylation status of histone H4 are distinct from the HDAC inhibitory property of TSA, since rottlerin failed to inhibit HDAC activity in nuclear extracts isolated from INS cells. These data are suggestive of potential regulatory effects of rottlerin at the level of increasing the histone acetyltransferase activity in these cells. Together our studies present the first evidence to suggest a PKCdelta-mediated signalling step, which promotes hypoacetylation of candidate histones culminating in IL-1beta-induced metabolic dysfunction of the isolated beta cell.


Assuntos
Histonas/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/farmacologia , Óxido Nítrico/metabolismo , Acetilação/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/farmacologia , Células Secretoras de Insulina/enzimologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Proteína Quinase C-delta/metabolismo , Fatores de Tempo
3.
Apoptosis ; 10(4): 841-50, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16133874

RESUMO

C2-ceramide, a cell permeable analogue of ceramide [CER] markedly reduced mitochondrial membrane potential [MMP] in insulin-secreting INS cells, which was followed by a significant accumulation of cytochrome c [Cyt c] into the cytosolic compartment. In a manner akin to CER, exposure of these cells to interleukin-1beta [IL-1beta] also resulted in reduction in MMP and cytosolic accumulation of Cyt c. Further, long-term exposure of these cells to either CER [but not its inactive analogue] or IL-1beta caused a marked reduction in their metabolic viability. However, unlike IL-1beta, which increased nitric oxide [NO] release, CER-treatment of INS cells had no effects of CER on NO release were demonstrable. Together, these findings suggest that CER-induced mitochondrial effects may not be mediated via iNOS gene expression and NO production. CER also activated an okadaic acid -sensitive protein phosphatase [CAPP] in the purified mitochondrial fraction, suggesting that CAPP might represent one of the target proteins for CER in the beta cell mitochondria. Together, our findings suggest direct detrimental effects of CER on mitochondrial function in beta cells leading to their dysfunction and demise via apoptosis. Moreover, our findings provide evidence for a potential difference in the mechanisms underlying CER- and IL-1beta-induced mitochondrial defects and apoptotic demise of the effete beta cell.


Assuntos
Ceramidas/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Animais , Domínio Catalítico , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocromos c/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Metilação/efeitos dos fármacos , Mitocôndrias/metabolismo , Óxido Nítrico/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Ratos
4.
Apoptosis ; 9(2): 145-8, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15004511

RESUMO

Our recent data implicated small molecular weight G-proteins (e.g., H-Ras) in interleukin 1beta (IL 1beta)-induced metabolic dysfunction and apoptotic demise of the islet beta cell (Tannous et al., Biochem Pharmacol 2001; 62:1459-1468, Kowluru and Morgan, Biochem Pharmacol, 2002; 63:1027-1035, Chen et al. Biochem Pharmacol, 2003; 66:1681-1694). Recently, we have shown that mastoparan, a tetradecapeptide from wasp venom, has been shown to directly activate islet endogenous G-proteins and regulate islet function (Amin et al., Endocrinology 2003; 144: 4508-4518). Herein, we investigated potential contributory roles, if any, of mastoparan (Mas)-sensitive G-proteins in IL-induced nitric oxide (NO) release from insulin-secreting HIT-T15 cells. While, ineffective by itself, Mas significantly potentiated IL-induced NO release from HIT-T15 cells. Interestingly, Mas-17, an inactive analog of Mas, also potentiated IL-induced NO release, suggesting that the potentiating effect of Mas may not involve activation of specific G-proteins. Such potentiating effects on IL-induced NO release were also demonstrable in the presence of another polycationic compound, melittin. Together, these findings suggest that Mas-induced potentiation of IL-induced NO release may in part be due to its amphiphilic and polycationic nature. These data also warrant caution in the use of Mas to study its regulation of cellular function without the use of an appropriate negative control, such as Mas-17.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Interleucina-1/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Óxido Nítrico/metabolismo , Venenos de Vespas/farmacologia , Relação Dose-Resposta a Droga , Humanos , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Ilhotas Pancreáticas/metabolismo , Meliteno/farmacologia , Nitritos/metabolismo , Peptídeos
5.
Artigo em Inglês | MEDLINE | ID: mdl-12476787

RESUMO

It is well established that glucose-induced insulin secretion involves generation of intracellular second messengers. Using specific inhibitors of guanosine triphosphate [GTP] biosynthesis [e.g., mycophenolic acid; MPA], we have identified a permissive role for GTP in glucose-stimulated insulin secretion. While the exact site of action for GTP within the islet beta cell remains to be identified and defined, recent evidence from several laboratories, including our own, indicate that it could involve activation of GTP-binding proteins [G-proteins]. These studies have identified both trimeric and monomeric forms of G-proteins within the pancreatic beta cell. Recent data also indicate that these G-proteins, specifically the monomeric G-proteins and the gamma subunits of trimeric G-proteins undergo a series of posttranslational modifications at their C-terminal cysteine. Such modifications include, isoprenylation, carboxyl methylation and palmitoylation. These modification steps appear to be essential for translocation of these proteins to the membrane sites for interaction with their respective effector proteins. This review primarily focuses on recent findings that clearly support the viewpoint that these posttranslational modification steps not only play obligatory roles in fuel-induced insulin secretion, but also in cytokine-mediated apoptotic demise of the beta cell. In this review, we also attempted to describe those findings involving the use of specific inhibitors for each of these pathways, and it is our hope that these aspects of beta cell metabolism and function generate interest in development of therapeutic intervention modalities to states of perturbed insulin release.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Ilhotas Pancreáticas/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Apoptose , Glucose/fisiologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Interleucinas/metabolismo , Ilhotas Pancreáticas/metabolismo , Metilação , Ácido Palmítico/metabolismo , Prenilação de Proteína
6.
Apoptosis ; 7(3): 241-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11997668

RESUMO

AIMS/HYPOTHESIS: It is well established that long-term exposure of isolated beta cells to cytokines [e.g., IL-1beta] results in increased expression of inducible nitric oxide synthase and subsequent release of nitric oxide, which in turn, has been shown to mediate a wide array of effects, including alterations in cellular high-energy metabolism. In this context, several extant studies have demonstrated significant reduction in adenine and guanine nucleotide triphosphate levels in beta cells exposed to IL-1beta. Herein, we examined the functional status of glyceraldehyde-3-phosphate dehydrogenase [GAPDH] in insulin-secreting cells exposed to IL-1beta, since it represents the first enzyme in the glycolytic pathway that is involved in the generation of ATP. METHODS: GAPDH was assayed spectrophotometrically in the cytosolic fraction derived from control and IL-1beta -treated [300 pM for 24 hrs] insulin-secreting cell lines [HIT-T15 and RINm5F]. RESULTS: IL-treatment resulted in marked attenuation of GAPDH activity in HIT and RIN cells; such a reduction in this activity was not due to inhibition of its expression by IL-1. Instead, we observed that incubation of HIT and RIN lysates with peroxynitrite, a reactive intermediate of nitric oxide with superoxide anion, resulted in significant reduction in the GAPDH activity. CONCLUSION/INTERPRETATION: These results identify a GAPDH as one of the biochemical loci for the effects of IL-derived peroxynitrite in the islet beta cell. The previously reported reduction in high-energy phosphate levels in an IL-treated beta cell may, in part, be due to inhibition of GAPDH activity, and subsequent reduction in the glycolytic efficiency of the beta cell.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/antagonistas & inibidores , Insulina/metabolismo , Interleucina-1/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Fracionamento Celular , Linhagem Celular , Citoplasma/enzimologia , Inibidores Enzimáticos/farmacologia , Etilmaleimida/farmacologia , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Secreção de Insulina , Interleucina-1/metabolismo , Ilhotas Pancreáticas/metabolismo , Ácido Peroxinitroso/farmacologia
7.
Diabetes Metab ; 28(6 Pt 2): 3S78-84; discussion 3S108-12, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12688637

RESUMO

Recently, we have demonstrated regulatory roles for G-proteins (e.g., H-Ras) in IL-1beta induced NO release from HIT-T15 cells. Herein, we report a similar regulatory mechanism for IL-1beta induced NO release from RIN5F and INS-1 cells. Our data indicate that functional inactivation of Ras, either by Clostridial toxins or by specific inhibitors of Ras function, results in a significant inhibition in IL-1beta induced NO release, suggesting that activation of specific G-proteins is essential for IL-1beta induced NO release. In the present study, we report possible loci where IL-1beta treatment might result in functional activation of these G-proteins. For example, IL-1beta treatment resulted in significant reduction in (high-and low-affinity) GTPase activities in lysates derived from normal rat islets; such a scenario might lead to retention of candidate G-proteins in GTP-bound, active conformation. Further, IL-1beta treatment increased the G-protein carboxyl methyl transferase activity as well as carboxyl methylation of endogenous beta-cell proteins; such a modification has been shown to increase the membrane association and interaction of these G-proteins with their respective effector proteins. Also, we report immunologic localization of H-Ras regulatory proteins including its nucleotide exchange factor (GRF-1) and its effector protein (eg., Raf-1) in isolated beta-cells. Together, our data indicate localization, and regulation by IL-1beta, of specific enzymes that are critical to activation of G-proteins. Based on these preliminary findings, we propose a model for the involvement of G-proteins in IL-1beta induced NO release and subsequent demise of the pancreatic beta-cell.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Interleucina-1/farmacologia , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação ao GTP/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Cinética , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas
8.
Biochem Pharmacol ; 62(11): 1459-68, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11728382

RESUMO

In the present study, we have shown that exposure of insulin-secreting clonal beta (HIT-T15) cells to interleukin-1beta (IL-1beta) results in a time- and concentration-dependent increase in nitric oxide (NO) release. These effects by IL-1beta on NO release were mediated by induction of inducible nitric oxide synthase (iNOS) from the cells. Preincubation of HIT cells with Clostridium sordellii lethal toxin-82, which irreversibly glucosylates and inactivates small G-proteins, such as Ras, Rap, Ral, and Rac, but not Cdc42, completely abolished IL-1beta-induced NO release. Pre-exposure of HIT cells to C. sordellii lethal toxin-9048, which monoglucosylates and inhibits Ras, Cdc42, Rac, and Rap, but not Ral, also attenuated IL-1beta-mediated NO release. These data indicate that activation of Ras and/or Rac may be necessary for IL-1beta-mediated NO release. Preincubation of HIT cells with C. difficile toxin-B, which monoglucosylates Rac, Cdc42, and Rho, had no demonstrable effects on IL-mediated NO release, ruling out the possibility that Rac may be involved in this signaling step. Further, two structurally dissimilar inhibitors of Ras function, namely manumycin A and damnacanthal, inhibited, in a concentration-dependent manner, the IL-1beta-mediated NO release from these cells. Together, our data provide evidence, for the first time, that Ras activation is an obligatory step in IL-1beta-mediated NO release and, presumably, the subsequent dysfunction of the pancreatic beta cell. Our data also provide a basis for future investigations to understand the mechanism of cytokine-induced beta cell death leading to the onset of insulin-dependent diabetes mellitus.


Assuntos
Interleucina-1/farmacologia , Óxido Nítrico/biossíntese , Proteínas ras/farmacologia , Animais , Células Cultivadas , Cricetinae , Relação Dose-Resposta a Droga , Interações Medicamentosas , Insulina/metabolismo , Secreção de Insulina , Fatores de Tempo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas ras/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/metabolismo
9.
Diabetes ; 50(7): 1580-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11423479

RESUMO

Acetyl-CoA carboxylase (ACC) catalyzes the formation of malonyl-CoA, a precursor in the biosynthesis of long-chain fatty acids, which have been implicated in physiological insulin secretion. The catalytic function of ACC is regulated by phosphorylation (inactive)-dephosphorylation (active). In this study we investigated whether similar regulatory mechanisms exist for ACC in the pancreatic islet beta-cell. ACC was quantitated in normal rat islets, human islets, and clonal beta-cells (HIT-15 or INS-1) using a [(14)C]bicarbonate fixation assay. In the beta-cell lysates, ACC was stimulated by magnesium in a concentration-dependent manner. Of all the dicarboxylic acids tested, only glutamate, albeit ineffective by itself, significantly potentiated magnesium-activated ACC in a concentration-dependent manner. ACC stimulation by glutamate and magnesium was maximally demonstrable in the cytosolic fraction; it was markedly reduced by okadaic acid (OKA) in concentrations (<50 nmol/l) that inhibited protein phosphatase 2A (PP2A). Furthermore, pretreatment of the cytosolic fraction with anti-PP2A serum attenuated the glutamate- and magnesium-mediated activation of ACC, thereby suggesting that ACC may be regulated by an OKA-sensitive PP2A-like enzyme. Streptavidin-agarose chromatography studies have indicated that glutamate- and magnesium-mediated effects on ACC are attributable to activation of ACC's dephosphorylation; this suggests that the stimulatory effects of glutamate and magnesium on ACC might involve activation of an OKA-sensitive PP2A-like enzyme that dephosphorylates and activates ACC. In our study, 5-amino-imidazolecarboxamide (AICA) riboside, a stimulator of AMP kinase, significantly inhibited glucose-mediated activation of ACC and insulin secretion from isolated beta-cells. Together, our data provide evidence for a unique regulatory mechanism for the activation of ACC in the pancreatic beta-cell, leading to the generation of physiological signals that may be relevant for physiological insulin secretion.


Assuntos
Acetil-CoA Carboxilase/metabolismo , Ácido Glutâmico/metabolismo , Ilhotas Pancreáticas/enzimologia , Magnésio/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Adenilato Quinase/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Células Cultivadas , Glucose/farmacologia , Humanos , Masculino , Proteína Fosfatase 2 , Ratos , Ribonucleosídeos/farmacologia
10.
Diabetologia ; 44(1): 89-94, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11206416

RESUMO

AIMS/HYPOTHESIS: Succinyl-CoA synthetase catalyses the substrate level phosphorylation of ADP or GDP. It also supplies succinyl-CoA for heme synthesis. Recently, two distinct mitochondrial succinyl-CoA synthetase activities, one specific for ATP and the other for GTP, have been characterized in various tissues of pigeon. Because of the relative importance of mitochondrial high-energy phosphate metabolism in physiological insulin secretion and the few data available on mitochondrial succinyl-CoA synthetase in the beta cell, this study examined whether ATP-specific and GTP-specific succinyl-CoA synthetase activities are localized in the clonal beta-cell mitochondria. METHODS: Using the mitochondrial extracts from clonal beta [INS-1 and HIT-T15] cells, we measured the formation of succinyl-CoA from succinate, CoA and ATP or GTP. To confirm the identity of these two enzymes, individual subunits of ATP-specific and GTP-specific to succinyl-CoA synthetase were identified by Western blot analysis. RESULTS: Both ATP-and GTP activities of succinylCoA synthetase were observed in the mitochondrial fractions from these cells. The ratios of GTP to ATP activities of succinyl-CoA synthetase were near unity in both of the cell types studied. Using affinity-purified antisera directed specifically against individual (alpha and beta) subunits of succinyl-CoA synthetase, we also identified both ATP-specific and GTP-specific forms of succinyl-CoA synthetase in HIT and INS cell mitochondria. Furthermore, using [gamma-32P]ATP as a phosphoryl donor, we observed that the alpha subunit of succinyl-CoA synthetase undergoes autophosphorylation at a histidine residue; co-provision of exogenous succinate and CoA resulted in pronounced dephosphorylation of the phosphorylated alpha subunit of succinyl-CoA synthetase. CONCLUSION/INTERPRETATION: We provide evidence for the localization of two distinct activities of succinyl-CoA synthetase in the beta cell mitochondria. Whereas it is well established that ATP is critical for the beta cell mitochondrial metabolism, we propose that GTP generated by the activation of succinylCoA synthetase could promote key functional roles in the mitochondrial metabolism leading to insulin secretion.


Assuntos
Trifosfato de Adenosina/metabolismo , Guanosina Trifosfato/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/enzimologia , Fosfatos/metabolismo , Succinato-CoA Ligases/metabolismo , Linhagem Celular , Células Clonais/enzimologia , Células Clonais/ultraestrutura , Coenzima A/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/ultraestrutura , Mitocôndrias/enzimologia , Fosforilação , Ácido Succínico/metabolismo
11.
Biochem Biophys Res Commun ; 268(2): 249-54, 2000 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-10679189

RESUMO

Lamins are intermediate filament proteins that constitute the main components of the lamina underlying the inner-nuclear membrane and serve to organize chromatin. Lamins (e.g., lamin-B) undergo posttranslational modifications (e.g., isoprenylation and methylation) at their C-terminal cysteine. Such modifications are thought to render optimal association of lamins with the nuclear envelop. Herein, we examined whether nuclear lamin-B undergoes carboxyl methylation in islet beta cells. A 65- to 70-kDa protein was carboxyl methylated in intact rat islets and clonal beta (HIT or INS) cells or in homogenates which could be immunoprecipitated using lamin-B antiserum. Incubation of purified HIT cell-nuclear fraction with [(3)H]S-adenosyl methionine yielded a single carboxyl methylated protein peak (ca. 65-70 kDa); this protein was immunologically identified as lamin-B. Several methylation inhibitors, including acetyl farnesyl cysteine, a competitive inhibitor of protein prenyl cysteine methylation, inhibited the carboxyl methylation of lamin-B, indicating that the carboxyl-methylated amino acid is cysteine. These findings, together with our recent observations demonstrating that inhibition of protein isoprenylation causes apoptotic death of the pancreatic beta cell, raise an interesting possibility that inhibition of C-terminal cysteine modifications of lamin-B might result in disruption of nuclear assembly, leading to further propagation of apoptotic signals, including DNA fragmentation and chromatin condensation.


Assuntos
Ilhotas Pancreáticas/metabolismo , Proteínas Nucleares/metabolismo , Animais , Apoptose/fisiologia , Núcleo Celular/metabolismo , Cisteína/metabolismo , Guanosina Trifosfato/metabolismo , Técnicas In Vitro , Lamina Tipo B , Laminas , Masculino , Metilação , Peso Molecular , Proteínas Nucleares/isolamento & purificação , Testes de Precipitina , Prenilação de Proteína , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley
12.
Diabetes ; 48(9): 1754-62, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10480605

RESUMO

A widely accepted genetically determined rodent model for human type 2 diabetes is the Goto-Kakizaki (GK) rat; however, the lesion(s) in the pancreatic islets of these rats has not been identified. Herein, intact islets from GK rats (aged 8-14 weeks) were studied, both immediately after isolation and after 18 h in tissue culture. Despite intact contents of insulin and protein, GK islets had markedly deficient insulin release in response to glucose, as well as to pure mitochondrial fuels or a non-nutrient membrane-depolarizing stimulus (40 mmol/l K+). In contrast, mastoparan (which activates GTP-binding proteins [GBPs]) completely circumvented any secretory defect. Basal and stimulated levels of adenine and guanine nucleotides, the activation of phospholipase C by Ca2+ or glucose, the secretory response to pertussis toxin, and the activation of selected low-molecular weight GBPs were not impaired. Defects were found, however, in the autophosphorylation and catalytic activity of cytosolic nucleoside diphosphokinase (NDPK), which may provide compartmentalized GTP pools to activate G-proteins; a deficient content of phosphoinositides was also detected. These studies identify novel, heretofore unappreciated, defects late in signal transduction in the islets of our colony of GK rats, possibly occurring at the site of activation by NDPK of a mastoparan-sensitive G-protein-dependent step in exocytosis.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Secreção de Insulina , Nucleotídeos de Purina/metabolismo , Ratos , Ratos Endogâmicos , Ratos Wistar , Taxa Secretória , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/metabolismo
13.
Proc Assoc Am Physicians ; 111(4): 335-46, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10417742

RESUMO

Studies of pancreatic islet function in the pathogenesis of type 2 diabetes mellitus have tended to focus on the short-term control of insulin secretion. However, the long-term control of beta-cell mass is also relevant to diabetes, since this parameter is reduced substantially even in non-insulin-dependent diabetes in humans. In animal models of type 2 diabetes, the normal balance between beta-cell proliferation and programmed cell death is perturbed. We take the perspective in this overview that inosine monophosphate dehydrogenase (IMPDH; EC 1.1.1. 205) may represent a previously neglected molecular integrator or sensor that exerts both functional (secretory) and anatomical (proliferative) effects within beta-cells. These properties reflect the fact that IMPDH is a rate-limiting enzyme in the new synthesis of the purine guanosine triphosphate (GTP), which modulates both exocytotic insulin secretion and DNA synthesis, as well as a number of other critical cellular functions within the beta-cell. Alterations in the expression or activity of IMPDH may be central to beta-cell replication, cell cycle progression, differentiation, and maintenance of adequate islet mass, effects that are probably mediated both by GTP directly, and indirectly via low molecular mass GTPases. If GTP becomes depleted, a hierarchy of beta-cell functions becomes progressively paralyzed, until eventually the effete cell is removed via apoptosis.


Assuntos
Guanosina Trifosfato/fisiologia , IMP Desidrogenase/fisiologia , Ilhotas Pancreáticas/enzimologia , Animais , Apoptose , Divisão Celular , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Indução Enzimática , Inibidores Enzimáticos/farmacologia , GTP Fosfo-Hidrolases/fisiologia , Glucose/metabolismo , Glucose/farmacologia , Humanos , IMP Desidrogenase/antagonistas & inibidores , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Ácido Micofenólico/farmacologia
14.
Endocrinology ; 139(9): 3752-62, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9724027

RESUMO

Inhibitors of IMP dehydrogenase, such as mycophenolic acid (MPA) and mizoribine, which deplete cellular GTP, are used clinically as immunosuppressive drugs. The prolonged effect of such agents on insulin-secreting beta-cells (HIT-T15 and INS-1) was investigated. Both MPA and mizoribine inhibited mitogenesis, as reflected by [3H]thymidine incorporation. Cell number, DNA and protein contents, and cell (metabolic) viability were decreased by about 30%, 60%, and 80% after treatment of HIT cells with clinically relevant concentrations (e.g. 1 microg/ml) of MPA for 1, 2, and 4 days, respectively. Mizoribine (48 h) similarly induced the death of HIT cells. INS-1 cells also were damaged by prolonged MPA treatment. MPA-treated HIT cells displayed a strong and localized staining with a DNA-binding dye (propidium iodide), suggesting condensation and fragmentation of DNA, which were confirmed by detection of DNA laddering in multiples of about 180 bp. DNA fragmentation was observed after 24-h MPA treatment and was dose dependent (29%, 49%, and 70% of cells were affected after 48-h exposure to 1, 3, and 10 microg/ml MPA, respectively). Examination of MPA-treated cells by electron microscopy revealed typical signs of apoptosis: condensed and marginated chromatin, apoptotic bodies, cytosolic vacuolization, and loss of microvilli. MPA-induced cell death was almost totally prevented by supplementation with guanosine, but not with adenosine or deoxyguanosine, indicating a specific effect of GTP depletion. An inhibitor of protein isoprenylation (lovastatin, 10-100 microM for 2-3 days) induced cell death and DNA degradation similar to those induced by sustained GTP depletion, suggesting a mediatory role of posttranslationally modified GTP-binding proteins. Indeed, impeding the function of G proteins of the Rho family (via glucosylation using Clostridium difficile toxin B), although not itself inducing apoptosis, potentiated cell death induced by MPA or lovastatin. These findings indicate that prolonged depletion of GTP induces beta-cell death compatible with apoptosis; this probably involves a direct impairment of GTP-dependent RNA-primed DNA synthesis, but also appears to be modulated by small GTP-binding proteins. Treatment of intact adult rat islets (the beta-cells of which replicate slowly) induced a modest, but definite, death by apoptosis over 1- to 3-day periods. Thus, more prolonged use of the new generation of immunosuppressive agents exemplified by MPA might have deleterious effects on the survival of islet or pancreas grafts.


Assuntos
Apoptose/fisiologia , Proteínas de Bactérias , Guanosina Trifosfato/deficiência , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Animais , Apoptose/efeitos dos fármacos , Toxinas Bacterianas/farmacologia , Proteínas de Ligação ao GTP/fisiologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Lovastatina/farmacologia , Mitose/fisiologia , Ácido Micofenólico/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
15.
Biosci Rep ; 18(4): 171-86, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9877231

RESUMO

Recently, we demonstrated that the 36 kDa catalytic subunit of protein phosphatase 2A (PP2Ac) undergoes methylation at its C-terminal leucine in normal rat islets, human islets and isolated beta cells; this modification increases the catalytic activity of PP2A [Kowluru et al. Endocrinology. 137:2315-2323, 1996]. Previous studies have suggested that adenine and guanine nucleotides or glycolytic intermediates [which are critical mediators in beta cell function] also modulate phosphatase activity in the pancreatic beta cell. Therefore, we examined whether these phosphorylated molecules specifically regulate the carboxyl methylation and the catalytic activity of PP2A in beta cells. Micromolar concentrations of ATP, ADP, GTP or GDP each inhibited the carboxyl methylation of PP2Ac and, to a lesser degree, the catalytic activity of PP2A. Likewise, the carboxyl methylation of PP2Ac and its catalytic activity were inhibited by [mono- or di-] phosphates of glucose or fructose. Additionally, however, the carboxyl methylation of PP2Ac was significantly stimulated by divalent metal ions (Mn2+ > Mg2+ > Ca2+ > control). The nucleotide or sugar phosphate-mediated inhibition of carboxyl methylation of PP2Ac and the catalytic activity of PP2A were completely prevented by Mn2+ or Mg2+. These data indicate that divalent metal ions protect against the inhibition by purine nucleotides or sugar phosphates of the carboxyl methylation of PP2Ac perhaps permitting PP2A to function under physiologic conditions. Therefore, these data warrant caution in interpretation of extant data on the regulation of phosphatase function by purine nucleotides.


Assuntos
Insulina/biossíntese , Ilhotas Pancreáticas/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Nucleotídeos de Purina/farmacologia , Fosfatos Açúcares/farmacologia , Animais , Catálise , Cátions Bivalentes/metabolismo , Inibidores Enzimáticos/farmacologia , Glicólise/fisiologia , Humanos , Técnicas In Vitro , Insulina/metabolismo , Masculino , Manganês/metabolismo , Metilação , Ácido Okadáico/farmacologia , Pâncreas/fisiologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Proteína Fosfatase 2 , Processamento de Proteína Pós-Traducional/fisiologia , Ratos
16.
Biosci Rep ; 18(4): 187-98, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9877232

RESUMO

Nucleoside diphosphate kinase (NDP kinase) catalyzes the transfer of terminal phosphate from nucleotide triphosphates (e.g. ATP) to nucleotide diphosphates (e.g. GDP) to yield nucleotide triphosphates (e.g. GTP). Since guanine nucleotides play critical role(s) in GTP-binding protein (G-protein)-mediated signal transduction mechanisms in retina, we quantitated NDP kinase activity in subcellular fraction-derived from normal rat retina. A greater than 85% of the total specific activity was present in the soluble fraction, which was stimulated (up to 7 fold) by 2 mM magnesium. NDP kinase exhibited saturation kinetics towards di- and tri-phosphate substrates, and was inhibited by known inhibitors of NDP kinase, uridine diphosphate (UDP) or cromoglycate (CRG). We have previously reported significant abnormalities in the activation of G-proteins in streptozotocin (STZ)-diabetic rat retina (Kowluru et al. Diabetologia 35:624 631, 1992). Since NDP kinase has been implicated in direct interaction with and/or activation of various G-proteins, we quantitated both basal and magnesium-stimulated NDP kinase activity in soluble and particulate fractions of retina derived from STZ-diabetic rats to examine whether abnormalities in G-protein function in diabetes are attributable to alterations in retinal NDP kinase. There was no effect of diabetes either on the basal or the magnesium-activated retinal NDP kinase activity. This study represents the first characterization of NDP kinase activity in rat retina, and suggests that in diabetes, this enzyme may not be rate-limiting and/or causal for the observed alterations in retinal G-protein functions.


Assuntos
Diabetes Mellitus Experimental/enzimologia , Núcleosídeo-Difosfato Quinase/análise , Retina/enzimologia , Animais , Membrana Celular/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Membrana/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/ultraestrutura , Difosfato de Uridina/farmacologia
17.
FEBS Lett ; 418(1-2): 179-82, 1997 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-9414122

RESUMO

Okadaic acid (OKA)-sensitive phosphatase (PP2A) activity may modulate nutrient-induced insulin secretion from pancreatic beta cells [Kowluru et al., Endocrinology 137 (1996) 2315-2323]. Ceramides, a new class of lipid second messengers may regulate PP2A [Dobrowsky and Hannun, J. Biol. Chem. (1992) 267, 5048-5051], and might play a role in cytokine-mediated apoptosis in beta cells [Sjöholm, FEBS Lett. 367 (1995) 283-286]. Therefore, we investigated the regulation of PP2A-like activity by ceramides in isolated beta (HIT-T15 or INS-1) cells. Cell-permeable (C2, C6 or C18) ceramides stimulated OKA-sensitive (but not -insensitive) phosphatase activity in a concentration-dependent manner (0-12.5 microM), with maximal stimulation (+50-100%) at < 12.5 microM. C2-dihydroceramide (a biologically inactive analog of C2 ceramide) failed to augment PP2A-like activity. Stimulatory effects of ceramides do not appear to be mediated via activation of the carboxyl methylation of the catalytic subunit of protein phosphatase 2A, since no effects of ceramides (up to 25 microM) were demonstrable on this parameter. These data identify a ceramide-activated protein phosphatase as a possible locus at which ceramides might exert their effects on beta cells leading to altered insulin secretion, and decreased cell viability followed by apoptotic cell demise.


Assuntos
Ceramidas/farmacologia , Ilhotas Pancreáticas/enzimologia , Fosfoproteínas Fosfatases/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Cinética , Masculino , Ácido Okadáico/farmacologia , Proteína Fosfatase 2 , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Células Tumorais Cultivadas
18.
J Clin Invest ; 100(6): 1596-610, 1997 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9294129

RESUMO

The gamma subunits of trimeric G-proteins (gamma1, gamma2, gamma5, and gamma7 isoforms) were found to be methylated at their carboxyl termini in normal rat islets, human islets and pure beta [HIT-T15] cells. Of these, GTPgammaS significantly stimulated the carboxyl methylation selectively of gamma2 and gamma5 isoforms. Exposure of intact HIT cells to either of two receptor-independent agonists--a stimulatory concentration of glucose or a depolarizing concentration of K+--resulted in a rapid (within 30 s) and sustained (at least up to 60 min) stimulation of gamma subunit carboxyl methylation. Mastoparan, which directly activates G-proteins (and insulin secretion from beta cells), also stimulated the carboxyl methylation of gamma subunits in intact HIT cells. Stimulatory effects of glucose or K+ were not demonstrable after removal of extracellular Ca2+ or depletion of intracellular GTP, implying regulatory roles for calcium fluxes and GTP; however, the methyl transferase itself was not directly activated by either. The stimulatory effects of mastoparan were resistant to removal of extracellular Ca2+, implying a mechanism of action that is different from glucose or K+ but also suggesting that dissociation of the alphabetagamma trimer is conducive to gamma subunit carboxyl methylation. Indeed, pertussis toxin also markedly attenuated the stimulatory effects of glucose, K+ or mastoparan without altering the rise in intracellular calcium induced by glucose or K+. Glucose-induced carboxyl methylation of gamma2 and gamma5 isoforms was vitiated by coprovision of any of three structurally different cyclooxygenase inhibitors. Conversely, exogenous PGE2, which activates Gi and Go in HIT cells and which thereby would dissociate alpha from beta(gamma), stimulated the carboxyl methylation of gamma2 and gamma5 isoforms and reversed the inhibition of glucose-stimulated carboxyl methylation of gamma subunits elicited by cyclooxygenase inhibitors. These data indicate that gamma subunits of trimeric G-proteins undergo a glucose- and calcium-regulated methylation-demethylation cycle in insulin-secreting cells, findings that may imply an important role in beta cell function. Furthermore, this is the first example of the regulation of the posttranslational modification of G-protein gamma subunits via nonreceptor-mediated activation mechanisms, which are apparently dependent on calcium influx and the consequent activation of phospholipases releasing arachidonic acid.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Glucose/farmacologia , Ilhotas Pancreáticas/metabolismo , 3-O-Metilglucose/farmacologia , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Cálcio/farmacologia , Cálcio/fisiologia , Células Cultivadas , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Guanosina Trifosfato/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Metilação/efeitos dos fármacos , Ácido Micofenólico/farmacologia , Peptídeos , Toxina Pertussis , Potássio/farmacologia , Proteínas Metiltransferases/metabolismo , Ratos , Ratos Sprague-Dawley , S-Adenosilmetionina/farmacologia , Fatores de Virulência de Bordetella/farmacologia , Venenos de Vespas/farmacologia
19.
Biochem Pharmacol ; 54(10): 1097-108, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9464452

RESUMO

We utilized clostridial toxins (with known specificities for inhibition of GTPases) to ascertain the contribution of candidate GTPases in physiologic insulin secretion from beta cells. Exposure of normal rat islets or isolated beta (HIT-T15) cells to Clostridium difficile toxins A and B catalyzed the glucosylation (and thereby the inactivation) of Rac, Cdc42, and Rho endogenous to beta cells; concomitantly, either toxin reduced glucose- or potassium-induced insulin secretion from rat islets and HIT cells. Treatment of beta cells with Clostridium sordellii lethal toxin (LT; which modified only Ras, Rap, and Rac) also reduced glucose- or potassium-induced secretion. However, clostridial toxin C3-exoenzyme (which ADP-ribosylates and inactivates only Rho) was without any effect on either glucose- or potassium-induced insulin secretion. These data suggest that Cdc42, Rac, Ras, and/or Rap (but not Rho) may be needed for glucose- or potassium-mediated secretion. The effects of these toxins appear to be specific on stimulus-secretion coupling, since no difference in metabolic viability (assessed colorimetrically by quantitating the conversion of the tetrazolium salt into a formazan in a reduction reaction driven by nutrient metabolism) was demonstrable between control and toxin (A or LT)-treated beta cells. Toxin (A or LT) treatment also did not alter glucose- or potassium-mediated rises in cytosolic free calcium concentrations ([Ca2+]i), suggesting that these GTPases are involved in steps distal to elevations in [Ca2+]i. Recent findings indicate that the carboxyl methylation of Cdc42 is stimulated by only glucose, whereas that of Rap (Kowluru et al., J Clin Invest 98: 540-555, 1996) and Rac (present study) are regulated by glucose or potassium. Together, these findings provide direct evidence, for the first time, that the Rho subfamily of GTPases plays a key regulatory role(s) in insulin secretion, and they suggest that Cdc42 may be required for early steps in glucose stimulation of insulin release, whereas Rap and/or Rac may be required for a later step(s) in the stimulus-secretion coupling cascade (i.e. Ca2+-induced exocytosis of insulin).


Assuntos
Proteínas de Bactérias , Toxinas Botulínicas , Cálcio/metabolismo , GTP Fosfo-Hidrolases/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Glucose/farmacologia , Insulina/metabolismo , Proteínas de Membrana/fisiologia , ADP Ribose Transferases/farmacologia , Adenosina Difosfato Ribose/metabolismo , Animais , Toxinas Bacterianas/farmacologia , Enterotoxinas/farmacologia , Glicosilação , Secreção de Insulina , Masculino , Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Proteína rhoB de Ligação ao GTP
20.
J Clin Invest ; 98(2): 540-55, 1996 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8755667

RESUMO

Several GTP-binding proteins (G-proteins) undergo post-translational modifications (isoprenylation and carboxyl methylation) in pancreatic beta cells. Herein, two of these were identified as CDC42 and rap 1, using Western blotting and immunoprecipitation. Confocal microscopic data indicated that CDC42 is localized only in islet endocrine cells but not in acinar cells of the pancreas. CDC42 undergoes a guanine nucleotide-specific membrane association and carboxyl methylation in normal rat islets, human islets, and pure beta (HIT or INS-1) cells. GTPgammaS-dependent carboxyl methylation of a 23-kD protein was also demonstrable in secretory granule fractions from normal islets or beta cells. AFC (a specific inhibitor of prenyl-cysteine carboxyl methyl transferases) blocked the carboxyl methylation of CDC42 in five types of insulin-secreting cells, without blocking GTPgammaS-induced translocation, implying that methylation is a consequence (not a cause) of transfer to membrane sites. High glucose (but not a depolarizing concentration of K+) induced the carboxyl methylation of CDC42 in intact cells, as assessed after specific immunoprecipitation. This effect was abrogated by GTP depletion using mycophenolic acid and was restored upon GTP repletion by coprovision of guanosine. In contrast, although rap 1 was also carboxyl methylated, it was not translocated to the particulate fraction by GTPgammaS; furthermore, its methylation was also stimulated by 40 mM K+ (suggesting a role which is not specific to nutrient stimulation). AFC also impeded nutrient-induced (but not K+-induced) insulin secretion from islets and beta cells under static or perifusion conditions, whereas an inactive structural analogue of AFC failed to inhibit insulin release. These effects were reproduced not only by S-adenosylhomocysteine (another methylation inhibitor), but also by GTP depletion. Thus, the glucose- and GTP-dependent carboxyl methylation of G-proteins such as CDC42 is an obligate step in the stimulus-secretion coupling of nutrient-induced insulin secretion, but not in the exocytotic event itself. Furthermore, AFC blocked glucose-activated phosphoinositide turnover, which may provide a partial biochemical explanation for its effect on secretion, and implies that certain G-proteins must be carboxyl methylated for their interaction with signaling effector molecules, a step which can be regulated by intracellular availability of GTP.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Glucose/farmacologia , Guanosina Trifosfato/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Proteínas Metiltransferases/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Western Blotting , Proteínas de Ciclo Celular/análise , Linhagem Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/análise , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Humanos , Secreção de Insulina , Insulinoma/fisiopatologia , Ilhotas Pancreáticas/efeitos dos fármacos , Cinética , Masculino , Metilação , Neoplasias Pancreáticas/fisiopatologia , Potássio/farmacologia , Proteínas Metiltransferases/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Proteína cdc42 de Ligação ao GTP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...