Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 33(4): 804-817.e5, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33321098

RESUMO

Metabolic fuels regulate insulin secretion by generating second messengers that drive insulin granule exocytosis, but the biochemical pathways involved are incompletely understood. Here we demonstrate that stimulation of rat insulinoma cells or primary rat islets with glucose or glutamine + 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (Gln + BCH) induces reductive, "counter-clockwise" tricarboxylic acid (TCA) cycle flux of glutamine to citrate. Molecular or pharmacologic suppression of isocitrate dehydrogenase-2 (IDH2), which catalyzes reductive carboxylation of 2-ketoglutarate to isocitrate, results in impairment of glucose- and Gln + BCH-stimulated reductive TCA cycle flux, lowering of NADPH levels, and inhibition of insulin secretion. Pharmacologic suppression of IDH2 also inhibits insulin secretion in living mice. Reductive TCA cycle flux has been proposed as a mechanism for generation of biomass in cancer cells. Here we demonstrate that reductive TCA cycle flux also produces stimulus-secretion coupling factors that regulate insulin secretion, including in non-dividing cells.


Assuntos
Ciclo do Ácido Cítrico/fisiologia , Glucose/farmacologia , Glutamina/farmacologia , Secreção de Insulina/efeitos dos fármacos , Animais , Células Cultivadas , Glucose/metabolismo , Glutamina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Isocitrato Desidrogenase/antagonistas & inibidores , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Lipogênese/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Compostos de Fenilureia/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Sulfonamidas/farmacologia , Sumoilação/efeitos dos fármacos
2.
PLoS One ; 15(3): e0224344, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32176701

RESUMO

A key event in the development of both major forms of diabetes is the loss of functional pancreatic islet ß-cell mass. Strategies aimed at enhancing ß-cell regeneration have long been pursued, but methods for reliably inducing human ß-cell proliferation with full retention of key functions such as glucose-stimulated insulin secretion (GSIS) are still very limited. We have previously reported that overexpression of the homeobox transcription factor NKX6.1 stimulates ß-cell proliferation, while also enhancing GSIS and providing protection against ß-cell cytotoxicity through induction of the VGF prohormone. We developed an NKX6.1 pathway screen by stably transfecting 832/13 rat insulinoma cells with a VGF promoter-luciferase reporter construct, using the resultant cell line to screen a 630,000 compound chemical library. We isolated three compounds with consistent effects to stimulate human islet cell proliferation, but not expression of NKX6.1 or VGF, suggesting an alternative mechanism of action. Further studies of the most potent of these compounds, GNF-9228, revealed that it selectively activates human ß-cell relative to α-cell proliferation and has no effect on δ-cell replication. In addition, pre-treatment, but not short term exposure of human islets to GNF-9228 enhances GSIS. GNF-9228 also protects 832/13 insulinoma cells against ER stress- and inflammatory cytokine-induced cytotoxicity. GNF-9228 stimulates proliferation via a mechanism distinct from recently emergent DYRK1A inhibitors, as it is unaffected by DYRK1A overexpression and does not activate NFAT translocation. In conclusion, we have identified a small molecule with pleiotropic positive effects on islet biology, including stimulation of human ß-cell proliferation and insulin secretion, and protection against multiple agents of cytotoxic stress.


Assuntos
Proliferação de Células/efeitos dos fármacos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Animais , Linhagem Celular Tumoral , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Glucose/farmacologia , Proteínas de Homeodomínio/metabolismo , Humanos , Células Secretoras de Insulina/patologia , Insulinoma/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Ratos , Quinases Dyrk
3.
Cell Rep ; 13(1): 157-167, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26411681

RESUMO

Pancreatic islet failure, involving loss of glucose-stimulated insulin secretion (GSIS) from islet ß cells, heralds the onset of type 2 diabetes (T2D). To search for mediators of GSIS, we performed metabolomics profiling of the insulinoma cell line 832/13 and uncovered significant glucose-induced changes in purine pathway intermediates, including a decrease in inosine monophosphate (IMP) and an increase in adenylosuccinate (S-AMP), suggesting a regulatory role for the enzyme that links the two metabolites, adenylosuccinate synthase (ADSS). Inhibition of ADSS or a more proximal enzyme in the S-AMP biosynthesis pathway, adenylosuccinate lyase, lowers S-AMP levels and impairs GSIS. Addition of S-AMP to the interior of patch-clamped human ß cells amplifies exocytosis, an effect dependent upon expression of sentrin/SUMO-specific protease 1 (SENP1). S-AMP also overcomes the defect in glucose-induced exocytosis in ß cells from a human donor with T2D. S-AMP is, thus, an insulin secretagogue capable of reversing ß cell dysfunction in T2D.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Diabetes Mellitus Tipo 2/metabolismo , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Monofosfato de Adenosina/metabolismo , Monofosfato de Adenosina/farmacologia , Adenilossuccinato Liase/antagonistas & inibidores , Adenilossuccinato Liase/genética , Adenilossuccinato Liase/metabolismo , Adenilossuccinato Sintase/antagonistas & inibidores , Adenilossuccinato Sintase/genética , Adenilossuccinato Sintase/metabolismo , Animais , Linhagem Celular Tumoral , Cisteína Endopeptidases , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Endopeptidases/genética , Endopeptidases/metabolismo , Inibidores Enzimáticos/farmacologia , Exocitose/efeitos dos fármacos , Regulação da Expressão Gênica , Glucose/metabolismo , Guanina/farmacologia , Humanos , Inosina Monofosfato/metabolismo , Insulina/biossíntese , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Metaboloma/genética , Ácido Micofenólico/farmacologia , Técnicas de Patch-Clamp , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
4.
Am J Physiol Endocrinol Metab ; 309(8): E715-26, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26306596

RESUMO

Proinflammatory cytokines impact islet ß-cell mass and function by altering the transcriptional activity within pancreatic ß-cells, producing increases in intracellular nitric oxide abundance and the synthesis and secretion of immunomodulatory proteins such as chemokines. Herein, we report that IL-1ß, a major mediator of inflammatory responses associated with diabetes development, coordinately and reciprocally regulates chemokine and insulin secretion. We discovered that NF-κB controls the increase in chemokine transcription and secretion as well as the decrease in both insulin secretion and proliferation in response to IL-1ß. Nitric oxide production, which is markedly elevated in pancreatic ß-cells exposed to IL-1ß, is a negative regulator of both glucose-stimulated insulin secretion and glucose-induced increases in intracellular calcium levels. By contrast, the IL-1ß-mediated production of the chemokines CCL2 and CCL20 was not influenced by either nitric oxide levels or glucose concentration. Instead, the synthesis and secretion of CCL2 and CCL20 in response to IL-1ß were dependent on NF-κB transcriptional activity. We conclude that IL-1ß-induced transcriptional reprogramming via NF-κB reciprocally regulates chemokine and insulin secretion while also negatively regulating ß-cell proliferation. These findings are consistent with NF-κB as a major regulatory node controlling inflammation-associated alterations in islet ß-cell function and mass.


Assuntos
Quimiocinas/metabolismo , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Interleucina-1beta/metabolismo , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocinas/genética , Espectroscopia de Ressonância de Spin Eletrônica , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Immunoblotting , Insulina/genética , Secreção de Insulina , Insulinoma , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Consumo de Oxigênio , Neoplasias Pancreáticas , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Ratos Zucker , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína S9 Ribossômica , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Células Tumorais Cultivadas
5.
Islets ; 7(1): e1027854, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26030060

RESUMO

Type 1 and type 2 diabetes are ultimately characterized by depleted ß-cell mass. Characterization of the molecular pathways that control ß-cell proliferation could be harnessed to restore these cells. The homeobox ß-cell transcription factor Nkx6.1 induces ß-cell proliferation by activating the orphan nuclear receptors Nr4a1 and Nr4a3. Here, we demonstrate that Nkx6.1 localizes to the promoter of the mitotic kinase AURKA (Aurora Kinase A) and induces its expression. Adenovirus mediated overexpression of AURKA is sufficient to induce proliferation in primary rat islets while maintaining glucose stimulated insulin secretion. Furthermore, AURKA is necessary for Nkx6.1 mediated ß-cell proliferation as demonstrated by shRNA mediated knock down and pharmacological inhibition of AURKA kinase activity. AURKA preferentially induces DNA replication in ß-cells as measured by BrdU incorporation, and enhances the rate of histone H3 phosphorylation in primary ß-cells, demonstrating that AURKA induces the replicative and mitotic cell cycle phases in rat ß-cells. Finally, overexpression of AURKA results in phosphorylation of the cell cycle regulator p53, which targets p53 for degradation and permits cell cycle progression. These studies define a pathway by which AURKA upregulation by Nkx6.1 results in phosphorylation and degradation of p53, thus removing a key inhibitory factor and permitting engagement of the ß-cell proliferation pathway.


Assuntos
Aurora Quinase A/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Aurora Quinase A/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA , Genes p53/genética , Proteínas de Homeodomínio/genética , Técnicas In Vitro/métodos , Proteínas do Tecido Nervoso , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , RNA/genética , Ratos , Transdução Genética
6.
J Biol Chem ; 290(21): 13401-16, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25851902

RESUMO

Glucocorticoids signal through the glucocorticoid receptor (GR) and are administered clinically for a variety of situations, including inflammatory disorders, specific cancers, rheumatoid arthritis, and organ/tissue transplantation. However, glucocorticoid therapy is also associated with additional complications, including steroid-induced diabetes. We hypothesized that modification of the steroid backbone is one strategy to enhance the therapeutic potential of GR activation. Toward this goal, two commercially unavailable, thiobenzothiazole-containing derivatives of hydrocortisone (termed MS4 and MS6) were examined using 832/13 rat insulinoma cells as well as rodent and human islets. We found that MS4 had transrepression properties but lacked transactivation ability, whereas MS6 retained both transactivation and transrepression activities. In addition, MS4 and MS6 both displayed anti-inflammatory activity. Furthermore, MS4 displayed reduced impact on islet ß-cell function in both rodent and human islets. Similar to dexamethasone, MS6 promoted adipocyte development in vitro, whereas MS4 did not. Moreover, neither MS4 nor MS6 activated the Pck1 (Pepck) gene in primary rat hepatocytes. We conclude that modification of the functional groups attached to the D-ring of the hydrocortisone steroid molecule produces compounds with altered structure-function GR agonist activity with decreased impact on insulin secretion and reduced adipogenic potential but with preservation of anti-inflammatory activity.


Assuntos
Anti-Inflamatórios/farmacologia , Benzimidazóis/farmacologia , Benzotiazóis/farmacologia , Hidrocortisona/análogos & derivados , Hidrocortisona/farmacologia , Inflamação/tratamento farmacológico , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Tiazóis/química , Células 3T3-L1 , Animais , Anti-Inflamatórios/síntese química , Apoptose/efeitos dos fármacos , Benzimidazóis/síntese química , Benzotiazóis/síntese química , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Dexametasona/farmacologia , Perfilação da Expressão Gênica , Hepatócitos/efeitos dos fármacos , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Hidrocortisona/síntese química , Técnicas Imunoenzimáticas , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Consumo de Oxigênio/efeitos dos fármacos , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Biochim Biophys Acta ; 1849(6): 637-52, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25882704

RESUMO

Enhanced leukocytic infiltration into pancreatic islets contributes to inflammation-based diminutions in functional ß-cell mass. Insulitis (aka islet inflammation), which can be present in both T1DM and T2DM, is one factor influencing pancreatic ß-cell death and dysfunction. IL-1ß, an inflammatory mediator in both T1DM and T2DM, acutely (within 1h) induced expression of the CCL20 gene in rat and human islets and clonal ß-cell lines. Transcriptional induction of CCL20 required the p65 subunit of NF-κB to replace the p50 subunit at two functional κB sites within the CCL20 proximal gene promoter. The NF-κB p50 subunit prevents CCL20 gene expression during unstimulated conditions and overexpression of p50 reduces CCL20, but enhances cyclooxygenase-2 (COX-2), transcript accumulation after exposure to IL-1ß. We also identified differential recruitment of specific co-activator molecules to the CCL20 gene promoter, when compared with the CCL2 and COX2 genes, revealing distinct transcriptional requirements for individual NF-κB responsive genes. Moreover, IL-1ß, TNF-α and IFN-γ individually increased the expression of CCR6, the receptor for CCL20, on the surface of human neutrophils. We further found that the chemokine CCL20 is elevated in serum from both genetically obese db/db mice and in C57BL6/J mice fed a high-fat diet. Taken together, these results are consistent with a possible activation of the CCL20-CCR6 axis in diseases with inflammatory components. Thus, interfering with this signaling pathway, either at the level of NF-κB-mediated chemokine production, or downstream receptor activation, could be a potential therapeutic target to offset inflammation-associated tissue dysfunction in obesity and diabetes.


Assuntos
Quimiocina CCL20/genética , Diabetes Mellitus/genética , Inflamação/genética , Obesidade/genética , Fator de Transcrição RelA/genética , Animais , Quimiocina CCL20/biossíntese , Quimiocina CCL20/metabolismo , Diabetes Mellitus/patologia , Humanos , Imunidade Inata/genética , Inflamação/patologia , Resistência à Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Obesos , NF-kappa B/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Ratos , Receptores CCR6/genética , Transdução de Sinais/genética , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/metabolismo
8.
Mol Immunol ; 62(1): 54-62, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24972324

RESUMO

Synthesis and secretion of immunomodulatory proteins, such as cytokines and chemokines, controls the inflammatory response within pancreatic islets. When this inflammation does not resolve, destruction of pancreatic islet ß-cells leads to diabetes mellitus. Production of the soluble mediators of inflammation, such as TNF-α and IL-1ß, from resident and invading immune cells, as well as directly from islet ß-cells, is also associated with suboptimal islet transplantation outcomes. In this study, we found that IL-1ß induces rapid increases in TNF-α mRNA in rat and human islets and the 832/13 clonal ß-cell line. The surge in transcription of the TNF-α gene required the inhibitor of kappa B kinase beta (IκKß), the p65 subunit of the NF-κB and a signal-specific recruitment of RNA polymerase II to the gene promoter. Of note was the increased intracellular production of TNF-α protein in a manner consistent with mRNA accumulation in response to IL-1ß, but no detectable secretion of TNF-α into the media. Additionally, TNF-α specifically induces expression of CD11b, but not CD11c, on neutrophils, which could contribute to the inflammatory milieu and diabetes progression. We conclude that activation of the NF-κB pathway in pancreatic ß-cells leads to rapid intracellular production of the pro-inflammatory TNF-α protein through a combination of specific histone covalent modifications and NF-κB signaling pathways.


Assuntos
Células Secretoras de Insulina/imunologia , Interleucina-1beta/farmacologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Quinase I-kappa B/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
9.
Am J Physiol Endocrinol Metab ; 306(2): E131-49, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24280128

RESUMO

Diabetes mellitus results from immune cell invasion into pancreatic islets of Langerhans, eventually leading to selective destruction of the insulin-producing ß-cells. How this process is initiated is not well understood. In this study, we investigated the regulation of the CXCL1 and CXCL2 genes, which encode proteins that promote migration of CXCR2(+) cells, such as neutrophils, toward secreting tissue. Herein, we found that IL-1ß markedly enhanced the expression of the CXCL1 and CXCL2 genes in rat islets and ß-cell lines, which resulted in increased secretion of each of these proteins. CXCL1 and CXCL2 also stimulated the expression of specific integrin proteins on the surface of human neutrophils. Mutation of a consensus NF-κB genomic sequence present in both gene promoters reduced the ability of IL-1ß to promote transcription. In addition, IL-1ß induced binding of the p65 and p50 subunits of NF-κB to these consensus κB regulatory elements as well as to additional κB sites located near the core promoter regions of each gene. Additionally, serine-phosphorylated STAT1 bound to the promoters of the CXCL1 and CXCL2 genes. We further found that IL-1ß induced specific posttranslational modifications to histone H3 in a time frame congruent with transcription factor binding and transcript accumulation. We conclude that IL-1ß-mediated regulation of the CXCL1 and CXCL2 genes in pancreatic ß-cells requires stimulus-induced changes in histone chemical modifications, recruitment of the NF-κB and STAT1 transcription factors to genomic regulatory sequences within the proximal gene promoters, and increases in phosphorylated forms of RNA polymerase II.


Assuntos
Quimiocina CXCL1/genética , Quimiocina CXCL2/genética , Regulação da Expressão Gênica , Células Secretoras de Insulina/metabolismo , NF-kappa B/metabolismo , Fator de Transcrição STAT1/metabolismo , Animais , Células Cultivadas , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1beta/farmacologia , Ratos , Ratos Wistar , Fator de Transcrição STAT1/genética , Transcrição Gênica/efeitos dos fármacos
10.
Mol Endocrinol ; 27(10): 1724-42, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24014650

RESUMO

The proinflammatory cytokines IL-1ß and IFN-γ decrease functional islet ß-cell mass in part through the increased expression of specific genes, such as inducible nitric oxide synthase (iNOS). Dysregulated iNOS protein accumulation leads to overproduction of nitric oxide, which induces DNA damage, impairs ß-cell function, and ultimately diminishes cellular viability. However, the transcriptional mechanisms underlying cytokine-mediated expression of the iNOS gene are not completely understood. Herein, we demonstrated that individual mutations within the proximal and distal nuclear factor-κB sites impaired cytokine-mediated transcriptional activation. Surprisingly, mutating IFN-γ-activated site (GAS) elements in the iNOS gene promoter, which are classically responsive to IFN-γ, modulated transcriptional sensitivity to IL-1ß. Transcriptional sensitivity to IL-1ß was increased by generation of a consensus GAS element and decreased correspondingly with 1 or 2 nucleotide divergences from the consensus sequence. The nuclear factor-κB subunits p65 and p50 bound to the κB response elements in an IL-1ß-dependent manner. IL-1ß also promoted binding of serine-phosphorylated signal transducer and activator of transcription-1 (STAT1) (Ser727) but not tyrosine-phosphorylated STAT1 (Tyr701) to GAS elements. However, phosphorylation at Tyr701 was required for IFN-γ to potentiate the IL-1ß response. Furthermore, coactivator p300 and coactivator arginine methyltransferase were recruited to the iNOS gene promoter with concomitant displacement of the coactivator CREB-binding protein in cells exposed to IL-1ß. Moreover, these coordinated changes in factor recruitment were associated with alterations in acetylation, methylation, and phosphorylation of histone proteins. We conclude that p65 and STAT1 cooperate to control iNOS gene transcription in response to proinflammatory cytokines by a coactivator exchange mechanism. This increase in transcription is also associated with signal-specific chromatin remodeling that leads to RNA polymerase II recruitment and phosphorylation.


Assuntos
Interferon gama/metabolismo , Interleucina-1beta/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Ativação Transcricional , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Montagem e Desmontagem da Cromatina , Indução Enzimática , Proteínas I-kappa B/metabolismo , Janus Quinase 1/metabolismo , Inibidor de NF-kappaB alfa , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases/metabolismo , Ratos , Ratos Wistar , Elementos de Resposta , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição RelA/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
11.
J Immunol ; 191(1): 323-36, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23740952

RESUMO

The CXCL10 gene encodes a peptide that chemoattracts a variety of leukocytes associated with type 1 and type 2 diabetes. The present study was undertaken to determine the molecular mechanisms required for expression of the CXCL10 gene in response to IL-1ß and IFN-γ using rat islets and ß cell lines. IL-1ß induced the expression of the CXCL10 gene and promoter activity, whereas the combination of IL-1ß plus IFN-γ was synergistic. Small interfering RNA-mediated suppression of NF-κB p65 markedly inhibited the ability of cytokines to induce the expression of the CXCL10 gene, whereas targeting STAT1 only diminished the synergy provided by IFN-γ. Furthermore, we found that a JAK1 inhibitor dose dependently reduced IFN-γ-controlled CXCL10 gene expression and promoter activity, concomitant with a decrease in STAT1 phosphorylation at Tyr(701). We further discovered that, although the Tyr(701) phosphorylation site is inducible (within 15 min of IFN-γ exposure), the Ser(727) site within STAT1 is constitutively phosphorylated. Thus, we generated single-mutant STAT1 Y701F and double-mutant STAT1 Y701F/S727A adenoviruses. Using these recombinant adenoviruses, we determined that overexpression of either the single- or double-mutant STAT1 decreased the IFN-γ-mediated potentiation of CXCL10 gene expression, promoter activity, and secretion of protein. Moreover, the Ser(727) phosphorylation was neither contingent on a functional Y701 site in ß cells nor was it required for cytokine-mediated expression of the CXCL10 gene. We conclude that the synergism of IL-1ß and IFN-γ to induce expression of the CXCL10 gene requires NF-κB, STAT1 phosphorylated at Tyr(701), recruitment of coactivators, and acetylation of histones H3 and H4.


Assuntos
Quimiocina CXCL10/genética , Regulação da Expressão Gênica/imunologia , Histonas/metabolismo , Interferon gama/fisiologia , Interleucina-1beta/fisiologia , NF-kappa B/fisiologia , Fator de Transcrição STAT1/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Células Cultivadas , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/metabolismo , Histonas/genética , Humanos , Interferon gama/antagonistas & inibidores , Mutagênese Sítio-Dirigida , Fosforilação/genética , Fosforilação/imunologia , Ratos , Ratos Wistar , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Tirosina/genética , Tirosina/metabolismo
12.
J Biol Chem ; 288(32): 23128-40, 2013 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-23788641

RESUMO

Recent studies have shown that the pyruvate-isocitrate cycling pathway, involving the mitochondrial citrate/isocitrate carrier and the cytosolic NADP-dependent isocitrate dehydrogenase (ICDc), is involved in control of glucose-stimulated insulin secretion (GSIS). Here we demonstrate that pyruvate-isocitrate cycling regulates expression of the voltage-gated potassium channel family member Kv2.2 in islet ß-cells. siRNA-mediated suppression of ICDc, citrate/isocitrate carrier, or Kv2.2 expression impaired GSIS, and the effect of ICDc knockdown was rescued by re-expression of Kv2.2. Moreover, chronic exposure of ß-cells to elevated fatty acids, which impairs GSIS, resulted in decreased expression of Kv2.2. Surprisingly, knockdown of ICDc or Kv2.2 increased rather than decreased outward K(+) current in the 832/13 ß-cell line. Immunoprecipitation studies demonstrated interaction of Kv2.1 and Kv2.2, and co-overexpression of the two channels reduced outward K(+) current compared with overexpression of Kv2.1 alone. Also, siRNA-mediated knockdown of ICDc enhanced the suppressive effect of the Kv2.1-selective inhibitor stromatoxin1 on K(+) currents. Our data support a model in which a key function of the pyruvate-isocitrate cycle is to maintain levels of Kv2.2 expression sufficient to allow it to serve as a negative regulator of Kv channel activity.


Assuntos
Regulação da Expressão Gênica/fisiologia , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Isocitratos/metabolismo , Ácido Pirúvico/metabolismo , Canais de Potássio Shab/biossíntese , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Masculino , Modelos Biológicos , Peptídeos/farmacologia , Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shab/antagonistas & inibidores , Canais de Potássio Shab/genética , Venenos de Aranha/farmacologia
13.
PLoS One ; 7(10): e46986, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056550

RESUMO

Release of pro-inflammatory cytokines from both resident and invading leukocytes within the pancreatic islets impacts the development of Type 1 diabetes mellitus. Synthesis and secretion of the chemokine CCL2 from pancreatic ß-cells in response to pro-inflammatory signaling pathways influences immune cell recruitment into the pancreatic islets. Therefore, we investigated the positive and negative regulatory components controlling expression of the CCL2 gene using isolated rat islets and INS-1-derived ß-cell lines. We discovered that activation of the CCL2 gene by IL-1ß required the p65 subunit of NF-κB and was dependent on genomic response elements located in the -3.6 kb region of the proximal gene promoter. CCL2 gene transcription in response to IL-1ß was blocked by pharmacological inhibition of the IKKß and p38 MAPK pathways. The IL-1ß-mediated increase in CCL2 secretion was also impaired by p38 MAPK inhibition and by glucocorticoids. Moreover, multiple synthetic glucocorticoids inhibited the IL-1ß-stimulated induction of the CCL2 gene. Induction of the MAP Kinase Phosphatase-1 (MKP-1) gene by glucocorticoids or by adenoviral-mediated overexpression decreased p38 MAPK phosphorylation, which diminished CCL2 gene expression, promoter activity, and release of CCL2 protein. We conclude that glucocorticoid-mediated repression of IL-1ß-induced CCL2 gene transcription and protein secretion occurs in part through the upregulation of the MKP-1 gene and subsequent deactivation of the p38 MAPK. Furthermore, the anti-inflammatory actions observed with MKP-1 overexpression were obtained without suppressing glucose-stimulated insulin secretion. Thus, MKP-1 is a possible target for anti-inflammatory therapeutic intervention with preservation of ß-cell function.


Assuntos
Quimiocina CCL2/genética , Fosfatase 1 de Especificidade Dupla/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/farmacologia , Animais , Linhagem Celular Tumoral , Fosfatase 1 de Especificidade Dupla/genética , Humanos , Quinase I-kappa B/metabolismo , Células Secretoras de Insulina/citologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores de Glucocorticoides/metabolismo , Fator de Transcrição RelA/metabolismo , Ativação Transcricional/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
PLoS One ; 6(7): e22485, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21829464

RESUMO

A reduction in functional ß-cell mass leads to both major forms of diabetes; pro-inflammatory cytokines, such as interleukin-1beta (IL-1ß) and gamma-interferon (γ-IFN), activate signaling pathways that direct pancreatic ß-cell death and dysfunction. However, the molecular mechanism of ß-cell death in this context is not well understood. In this report, we tested the hypothesis that individual cellular death pathways display characteristic phenotypes that allow them to be distinguished by the precise biochemical and metabolic responses that occur during stimulus-specific initiation. Using 832/13 and INS-1E rat insulinoma cells and isolated rat islets, we provide evidence that apoptosis is unlikely to be the primary pathway underlying ß-cell death in response to IL-1ß+γ-IFN. This conclusion was reached via the experimental results of several different interdisciplinary strategies, which included: 1) tandem mass spectrometry to delineate the metabolic differences between IL-1ß+γ-IFN exposure versus apoptotic induction by camptothecin and 2) pharmacological and molecular interference with either NF-κB activity or apoptosome formation. These approaches provided clear distinctions in cell death pathways initiated by pro-inflammatory cytokines and bona fide inducers of apoptosis. Collectively, the results reported herein demonstrate that pancreatic ß-cells undergo apoptosis in response to camptothecin or staurosporine, but not pro-inflammatory cytokines.


Assuntos
Apoptose/efeitos dos fármacos , Citocinas/farmacologia , Células Secretoras de Insulina/patologia , Insulinoma/patologia , Neoplasias Pancreáticas/patologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Caspases/metabolismo , Morte Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Inflamação , Insulina/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/imunologia , Insulinoma/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Metabolômica , NF-kappa B/genética , NF-kappa B/metabolismo , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Ratos , Transdução de Sinais , Estaurosporina/farmacologia , Espectrometria de Massas em Tandem
15.
Endocrinology ; 151(7): 3074-83, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20484462

RESUMO

Prolactin (PRL) induces beta-cell proliferation and glucose-stimulated insulin secretion (GSIS) and counteracts the effects of glucocorticoids on insulin production. The mechanisms by which PRL up-regulates GSIS are unknown. We used rat islets and insulinoma (INS-1) cells to explore the interactions of PRL, glucose, and dexamethasone (DEX) in the regulation of beta-cell pyruvate carboxylase (PC), pyruvate dehydrogenase (PDH), and the pyruvate dehydrogenase kinases (PDKs), which catalyze the phosphorylation and inactivation of PDH. PRL increased GSIS by 37% (P < 0.001) in rat islets. Glucose at supraphysiological concentrations (11 mm) increased PC mRNA in islets; in contrast, PRL suppressed PC mRNA levels in islets and INS-1 cells, whereas DEX was without effect. Neither PRL nor DEX altered PC protein or activity levels. In INS-1 cells, PRL increased PDH activity 1.4- to 2-fold (P < 0.05-0.001) at glucose concentrations ranging from 2.5-11 mm. DEX reduced PDH activity; this effect was reversed by PRL. PDK1, -2, -3, and -4 mRNAs were detected in both islets and insulinoma cells, but the latter expressed trivial amounts of PDK4. PRL reduced PDK2 mRNA and protein levels in rat islets and INS-1 cells and PDK4 mRNA in islets; DEX increased PDK2 mRNA in islets and INS-1 cells; this effect was reversed by PRL. Our findings suggest that PRL induction of GSIS is mediated by increases in beta-cell PDH activity; this is facilitated by suppression of PDKs. PRL counteracts the effects of DEX on PDH and PDK expression, suggesting novel roles for the lactogens in the defense against diabetes.


Assuntos
Dexametasona/farmacologia , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Prolactina/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Técnicas In Vitro , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Proteínas Serina-Treonina Quinases/genética , Piruvato Carboxilase/genética , Piruvato Descarboxilase/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
J Biol Chem ; 285(22): 16530-7, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20356834

RESUMO

Glucose-stimulated insulin secretion from pancreatic islet beta-cells is dependent in part on pyruvate cycling through the pyruvate/isocitrate pathway, which generates cytosolic alpha-ketoglutarate, also known as 2-oxoglutarate (2OG). Here, we have investigated if mitochondrial transport of 2OG through the 2-oxoglutarate carrier (OGC) participates in control of nutrient-stimulated insulin secretion. Suppression of OGC in clonal pancreatic beta-cells (832/13 cells) and isolated rat islets by adenovirus-mediated delivery of small interfering RNA significantly decreased glucose-stimulated insulin secretion. OGC suppression also reduced insulin secretion in response to glutamine plus the glutamate dehydrogenase activator 2-amino-2-norbornane carboxylic acid. Nutrient-stimulated increases in glucose usage, glucose oxidation, glutamine oxidation, or ATP:ADP ratio were not affected by OGC knockdown, whereas suppression of OGC resulted in a significant decrease in the NADPH:NADP(+) ratio during stimulation with glucose but not glutamine + 2-amino-2-norbornane carboxylic acid. Finally, OGC suppression reduced insulin secretion in response to a membrane-permeant 2OG analog, dimethyl-2OG. These data reveal that the OGC is part of a mechanism of fuel-stimulated insulin secretion that is common to glucose, amino acid, and organic acid secretagogues, involving flux through the pyruvate/isocitrate cycling pathway. Although the components of this pathway must remain intact for appropriate stimulus-secretion coupling, production of NADPH does not appear to be the universal second messenger signal generated by these reactions.


Assuntos
Glucose/metabolismo , Glutamina/metabolismo , Insulina/metabolismo , Ácidos Cetoglutáricos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Mitocôndrias/metabolismo , Animais , Citosol/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Modelos Biológicos , NADP/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Biol Chem ; 283(43): 28909-17, 2008 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-18755687

RESUMO

We have previously demonstrated a role for pyruvate cycling in glucose-stimulated insulin secretion (GSIS). Some of the possible pyruvate cycling pathways are completed by conversion of malate to pyruvate by malic enzyme. Using INS-1-derived 832/13 cells, it has recently been shown by other laboratories that NADP-dependent cytosolic malic enzyme (MEc), but not NAD-dependent mitochondrial malic enzyme (MEm), regulates GSIS. In the current study, we show that small interfering RNA-mediated suppression of either MEm or MEc results in decreased GSIS in both 832/13 cells and a new and more glucose- and incretin-responsive INS-1-derived cell line, 832/3. The effect of MEm to suppress GSIS in these cell lines was linked to a substantial decrease in cell growth, whereas MEc suppression resulted in decreased NADPH, shown previously to be correlated with GSIS. However, adenovirus-mediated delivery of small interfering RNAs specific to MEc and MEm to isolated rat islets, while leading to effective suppression of the targets transcripts, had no effect on GSIS. Furthermore, islets isolated from MEc-null MOD1(-/-) mice exhibit normal glucose- and potassium-stimulated insulin secretion. These results indicate that pyruvate-malate cycling does not control GSIS in primary rodent islets.


Assuntos
Citosol/metabolismo , Glucose/química , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Malato Desidrogenase/química , Mitocôndrias/metabolismo , Animais , Inativação Gênica , Secreção de Insulina , Masculino , Camundongos , Modelos Biológicos , Isoformas de Proteínas , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley
18.
J Biol Chem ; 283(21): 14248-56, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18381287

RESUMO

Acetyl-CoA carboxylase 1 (ACC1) currently is being investigated as a target for treatment of obesity-associated dyslipidemia and insulin resistance. To investigate the effects of ACC1 inhibition on insulin secretion, three small interfering RNA (siRNA) duplexes targeting ACC1 (siACC1) were transfected into the INS-1-derived cell line, 832/13; the most efficacious duplex was also cloned into an adenovirus and used to transduce isolated rat islets. Delivery of the siACC1 duplexes decreased ACC1 mRNA by 60-80% in 832/13 cells and islets and enzyme activity by 46% compared with cells treated with a non-targeted siRNA. Delivery of siACC1 decreased glucose-stimulated insulin secretion (GSIS) by 70% in 832/13 cells and by 33% in islets. Surprisingly, siACC1 treatment decreased glucose oxidation by 49%, and the ATP:ADP ratio by 52%, accompanied by clear decreases in pyruvate cycling activity and tricarboxylic acid cycle intermediates. Exposure of siACC1-treated cells to the pyruvate cycling substrate dimethylmalate restored GSIS to normal without recovery of the depressed ATP:ADP ratio. In siACC1-treated cells, glucokinase protein levels were decreased by 25%, which correlated with a 36% decrease in glycogen synthesis and a 33% decrease in glycolytic flux. Furthermore, acute addition of the ACC1 inhibitor 5-(tetradecyloxy)-2-furoic acid (TOFA) to beta-cells suppressed [(14)C]glucose incorporation into lipids but had no effect on GSIS, whereas chronic TOFA administration suppressed GSIS and glucose metabolism. In sum, chronic, but not acute, suppression of ACC1 activity impairs GSIS via inhibition of glucose rather than lipid metabolism. These findings raise concerns about the use of ACC inhibitors for diabetes therapy.


Assuntos
Acetil-CoA Carboxilase/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Metabolismo dos Lipídeos , Acetil-CoA Carboxilase/classificação , Acetil-CoA Carboxilase/genética , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Carnitina/análogos & derivados , Carnitina/metabolismo , Linhagem Celular , Ácidos Graxos/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Isoenzimas/classificação , Isoenzimas/genética , Isoenzimas/metabolismo , Oxirredução , Ácido Pirúvico/metabolismo , RNA Interferente Pequeno/genética , Ratos , Fatores de Tempo
19.
Mol Cell Biol ; 28(10): 3465-76, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18347054

RESUMO

The homeodomain transcription factor Nkx6.1 plays an important role in pancreatic islet beta-cell development, but its effects on adult beta-cell function, survival, and proliferation are not well understood. In the present study, we demonstrated that treatment of primary rat pancreatic islets with a cytomegalovirus promoter-driven recombinant adenovirus containing the Nkx6.1 cDNA (AdCMV-Nkx6.1) causes dramatic increases in [methyl-(3)H] thymidine and 5-bromo-2'-deoxyuridine (BrdU) incorporation and in the number of cells per islet relative to islets treated with a control adenovirus (AdCMV-betaGAL), whereas suppression of Nkx6.1 expression reduces thymidine incorporation. Immunocytochemical studies reveal that >80% of BrdU-positive cells in AdCMV-Nkx6.1-treated islets are beta cells. Microarray, real-time PCR, and immunoblot analyses reveal that overexpression of Nkx6.1 in rat islets causes concerted upregulation of a cadre of cell cycle control genes, including those encoding cyclins A, B, and E, and several regulatory kinases. Cyclin E is upregulated earlier than the other cyclins, and adenovirus-mediated overexpression of cyclin E is shown to be sufficient to activate islet cell proliferation. Moreover, chromatin immunoprecipitation assays demonstrate direct interaction of Nkx6.1 with the cyclin A2 and B1 genes. Overexpression of Nkx6.1 in rat islets caused a clear enhancement of glucose-stimulated insulin secretion (GSIS), whereas overexpression of Nkx6.1 in human islets caused an increase in the level of [(3)H]thymidine incorporation that was twice the control level, along with complete retention of GSIS. We conclude that Nkx6.1 is among the very rare factors capable of stimulating beta-cell replication with retention or enhancement of function, properties that may be exploitable for expansion of beta-cell mass in treatment of both major forms of diabetes.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Adenoviridae/genética , Animais , Sequência de Bases , Ciclo Celular/genética , Proliferação de Células , Ciclina A/genética , Ciclina A2 , Ciclina B/genética , Ciclina B1 , Ciclinas/metabolismo , Primers do DNA/genética , Expressão Gênica , Glucose/farmacologia , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Cinética , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Técnicas de Cultura de Tecidos
20.
Mol Endocrinol ; 22(5): 1251-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18258687

RESUMO

Both major forms of diabetes involve a decline in beta-cell mass, mediated by autoimmune destruction of insulin-producing cells in type 1 diabetes and by increased rates of apoptosis secondary to metabolic stress in type 2 diabetes. Methods for controlled expansion of beta-cell mass are currently not available but would have great potential utility for treatment of these diseases. In the current study, we demonstrate that overexpression of trefoil factor 3 (TFF3) in rat pancreatic islets results in a 4- to 5-fold increase in [(3)H]thymidine incorporation, with full retention of glucose-stimulated insulin secretion. This increase was almost exclusively due to stimulation of beta-cell replication, as demonstrated by studies of bromodeoxyuridine incorporation and co-immunofluorescence analysis with anti-bromodeoxyuridine and antiinsulin or antiglucagon antibodies. The proliferative effect of TFF3 required the presence of serum or 0.5 ng/ml epidermal growth factor. The ability of TFF3 overexpression to stimulate proliferation of rat islets in serum was abolished by the addition of epidermal growth factor receptor antagonist AG1478. Furthermore, TFF3-induced increases in [3H]thymidine incorporation in rat islets cultured in serum was blocked by overexpression of a dominant-negative Akt protein or treatment with triciribine, an Akt inhibitor. Finally, overexpression of TFF3 also caused a doubling of [3H]thymidine incorporation in human islets. In summary, our findings reveal a novel TFF3-mediated pathway for stimulation of beta-cell replication that could ultimately be exploited for expansion or preservation of islet beta-cell mass.


Assuntos
Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Neuropeptídeos/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fator de Crescimento Epidérmico/farmacologia , Humanos , Immunoblotting , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Masculino , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Proteína Oncogênica v-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina/metabolismo , Transfecção , Fator Trefoil-3
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA