Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Med Chem Lett ; 12(10): 1568-1577, 2021 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-34676039

RESUMO

Glucocorticoids (GCs) are widely used in medicine for their role in the treatment of autoimmune-mediated conditions, certain cancers, and organ transplantation. The transcriptional activities GCs elicit include transrepression, postulated to be responsible for the anti-inflammatory activity, and transactivation, proposed to underlie the undesirable side effects associated with long-term use. A GC analogue that could elicit only transrepression and beneficial transactivation properties would be of great medicinal value and is highly sought after. In this study, a series of 1-(4-substituted phenyl)pyrazole-based GC analogues were synthesized, biologically screened, and evaluated for SARs leading to the desired activity. Activity observed in compounds bearing an electron deficient arylpyrazole moiety showed promise toward a dissociated steroid, displaying transrepression while having limited transactivation activity. In addition, compounds 11aa and 11ab were found to have anti-inflammatory efficacy comparable to that of dexamethasone at 10 nM, with minimal transactivation activity and no reduction of insulin secretion in cultured rat 832/13 beta cells.

2.
Obesity (Silver Spring) ; 28(9): 1726-1735, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32741148

RESUMO

OBJECTIVE: An ethanolic extract of Artemisia scoparia (SCO) improves adipose tissue function and reduces negative metabolic consequences of high-fat feeding. A. scoparia has a long history of medicinal use across Asia and has anti-inflammatory effects in various cell types and disease models. The objective of the current study was to investigate SCO's effects on inflammation in cells relevant to metabolic health. METHODS: Inflammatory responses were assayed in cultured adipocytes, macrophages, and insulinoma cells by quantitative polymerase chain reaction, immunoblotting, and NF-κB reporter assays. RESULTS: In tumor necrosis factor α-treated adipocytes, SCO mitigated ERK and NF-κB signaling as well as transcriptional responses but had no effect on fatty acid-binding protein 4 secretion. SCO also reduced levels of deleted in breast cancer 1 protein in adipocytes and inhibited inflammatory gene expression in stimulated macrophages. Finally, in pancreatic ß-cells, SCO decreased NF-κB-responsive promoter activity induced by IL-1ß treatment. CONCLUSIONS: SCO's ability to promote adipocyte development and function is thought to mediate its insulin-sensitizing actions in vivo. Our findings that SCO inhibits inflammatory responses through at least two distinct signaling pathways (ERK and NF-κB) in three cell types known to contribute to metabolic disease reveal that SCO may act more broadly than previously thought to improve metabolic health.


Assuntos
Adipócitos/metabolismo , Anti-Inflamatórios/uso terapêutico , Artemisia/química , Inflamação/tratamento farmacológico , Células Secretoras de Insulina/metabolismo , Macrófagos/metabolismo , Scoparia/química , Animais , Anti-Inflamatórios/farmacologia , Modelos Animais de Doenças , Humanos , Camundongos , Transfecção
3.
Mol Metab ; 14: 95-107, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29914854

RESUMO

OBJECTIVE: Pancreatic tissue, and islets in particular, are enriched in expression of the interleukin-1 receptor type I (IL-1R). Because of this enrichment, islet ß-cells are exquisitely sensitive to the IL-1R ligands IL-1α and IL-1ß, suggesting that signaling through this pathway regulates health and function of islet ß-cells. METHODS: Herein, we report a targeted deletion of IL-1R in pancreatic tissue (IL-1RPdx1-/-) in C57BL/6J mice and in db/db mice on the C57 genetic background. Islet morphology, ß-cell transcription factor abundance, and expression of the de-differentiation marker Aldh1a3 were analyzed by immunofluorescent staining. Glucose and insulin tolerance tests were used to examine metabolic status of these genetic manipulations. Glucose-stimulated insulin secretion was evaluated in vivo and in isolated islets ex vivo by perifusion. RESULTS: Pancreatic deletion of IL-1R leads to impaired glucose tolerance, a phenotype that is exacerbated by age. Crossing the IL-1RPdx1-/- with db/db mice worsened glucose tolerance without altering body weight. There were no detectable alterations in insulin tolerance between IL-1RPdx1-/- mice and littermate controls. However, glucose-stimulated insulin secretion was reduced in islets isolated from IL-1RPdx1-/- relative to control islets. Insulin output in vivo after a glucose challenge was also markedly reduced in IL-1RPdx1-/- mice when compared with littermate controls. Pancreatic islets from IL-1RPdx1-/- mice displayed elevations in Aldh1a3, a marker of de-differentiation, and reduction in nuclear abundance of the ß-cell transcription factor MafA. Nkx6.1 abundance was unaltered. CONCLUSIONS: There is an important physiological role for pancreatic IL-1R to promote glucose homeostasis by suppressing expression of Aldh1a3, sustaining MafA abundance, and supporting glucose-stimulated insulin secretion in vivo.


Assuntos
Diferenciação Celular , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores Tipo I de Interleucina-1/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Deleção de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Homeostase , Resistência à Insulina , Células Secretoras de Insulina/citologia , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Retinal Desidrogenase/genética , Retinal Desidrogenase/metabolismo
4.
Redox Biol ; 16: 248-254, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29547847

RESUMO

Early podocyte loss is characteristic of chronic kidney diseases (CKD) in obesity and diabetes. Since treatments for hyperglycemia and hypertension do not prevent podocyte loss, there must be additional factors causing podocyte depletion. The role of oxidative stress has been implicated in CKD but it is not known how exactly free radicals affect podocyte physiology. To assess this relationship, we investigated the effects of lipid radicals on podocytes, as lipid peroxidation is a major form of oxidative stress in diabetes. We found that lipid radicals govern changes in podocyte homeostasis through redox sensitive RhoA signaling: lipid radicals inhibit migration and cause loss of F-actin fibers. These effects were prevented by mutating the redox sensitive cysteines of RhoA. We therefore suggest that in diseases associated with increased lipid peroxidation, lipid radicals can determine podocyte function with potentially pathogenic consequences for kidney physiology.


Assuntos
Peroxidação de Lipídeos/genética , Podócitos/metabolismo , Insuficiência Renal Crônica/genética , Proteína rhoA de Ligação ao GTP/genética , Actinas/genética , Actinas/metabolismo , Movimento Celular/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Complicações do Diabetes/genética , Complicações do Diabetes/patologia , Radicais Livres/metabolismo , Humanos , Mutação , Obesidade/complicações , Obesidade/genética , Obesidade/patologia , Oxirredução , Podócitos/patologia , Insuficiência Renal Crônica/patologia , Transdução de Sinais
5.
Biochimie ; 121: 140-50, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26700141

RESUMO

AIMS: Exposure to human adenovirus Ad36 is causatively and correlatively linked with better glycemic control in animals and humans, respectively. Although the anti-hyperglycemic property of Ad36 may offer some therapeutic potential, it is impractical to use an infectious agent for therapeutic benefit. Cell-based studies identified that Ad36 enhances cellular glucose disposal via its E4orf1 protein. Ability to improve glycemic control in vivo is a critical prerequisite for further investigating the therapeutic potential of E4orf1. Therefore, the aim of this study was to determine the ability of E4orf1 to improve glycemic control independent of insulin despite high fat diet. MATERIALS & METHODS: 8-9wk old male C57BL/6J mice fed a high-fat diet (60% kcal) were injected with a retrovirus plasmid expressing E4orf1, or a null vector (Control). Glycemic control was determined by glucose and insulin tolerance test. Islet cell size, amount of insulin and glucagon were determined in formalin-fixed pancreas. Rat insulinoma cell line (832/13) was infected with E4orf1 or control to determine changes in glucose stimulated insulin secretion. Protein from flash frozen adipose tissue depots, liver and muscle was used to determine molecular signaling by western blotting. RESULTS: In multiple experiments, retrovirus-mediated E4orf1 expression in C57BL/6J mice significantly and reproducibly improved glucose excursion following a glucose load despite a high fat diet (60% energy). Importantly, E4orf1 improved glucose clearance without increasing insulin sensitivity, production or secretion, underscoring its insulin-independent effect. E4orf1 modulated molecular signaling in mice tissue, which included greater protein abundance of adiponectin, p-AKT and Glucose transporter Glu4. CONCLUSIONS: This study provides the proof of concept for translational development of E4orf1 as a potential anti-diabetic agent. High fat intake and impaired insulin signaling are often associated with obesity, diabetes and insulin resistance. Hence, the ability of E4orf1 to improve glycemic control despite high fat diet and independent of insulin, is particularly attractive.


Assuntos
Hipoglicemiantes/uso terapêutico , Adenoviridae/genética , Animais , Glicemia/efeitos dos fármacos , Diabetes Mellitus/sangue , Diabetes Mellitus/tratamento farmacológico , Humanos , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Obesity (Silver Spring) ; 21(4): 737-47, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23712977

RESUMO

OBJECTIVE: Obesity is associated with chronic inflammation. Toll-like receptors (TLR) and NOD-like receptors (NLR) are two families of pattern recognition receptors that play important roles in immune response and inflammation in adipocytes. It has been reported that TLR4 and TLR2 activation induce proinflammatory changes that impair adipocyte differentiation. However, the effects of activation of NOD1 and NOD2, the two prominent members of NLR, on adipocyte differentiation have not been studied. DESIGN AND METHODS: 3T3-L1 and human adipose-derived stem cells were tested for adipocyte differentiation in the presence or absence of NOD ligand. Adipocyte differentiation was evaluated by the adipocyte markers gene expression and Oil Red O staining for lipid accumulation. RESULTS: Activation of NOD1, but not NOD2, by a synthetic ligand dose-dependently suppressed 3T3-L1 adipocyte differentiation as revealed by Oil Red O stained cell morphology, lipid accumulation, and attenuated gene expression of adipocyte markers (PPARγ, C/EBPα, SCD, FABP4, Adiponectin). Activation of NOD1, but not NOD2, induced NF-κB activation, which correlated with their abilities to suppress ligand-induced PPARγ transaction. Moreover, the suppressive effect by NOD1 activation was reversed by IκB super-repressor which blocks NF-κB activation. The suppression by NOD1 ligand C12-iEDAP on adipocyte differentiation was reversed by small RNA interference targeting NOD1, demonstrating the specificity of NOD1 activation. In contrast, activation of NOD1 and NOD2 both significantly suppressed adipocyte differentiation of human adipose-derived adult stem cells, demonstrating the species specific effects of NOD activation. In contrast to enhanced leptin mRNA by LPS and TNFα, NOD1 activation suppressed leptin mRNA in adipocytes, suggesting the differential effects of NOD1 activation in adipocytes. CONCLUSIONS: Overall, our results suggest that NOD1 represents a novel target for adipose inflammation in obesity.


Assuntos
Adipócitos/metabolismo , Diferenciação Celular , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Células 3T3-L1 , Adiponectina/genética , Adiponectina/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Expressão Gênica , Humanos , Leptina/antagonistas & inibidores , Leptina/genética , Leptina/metabolismo , Lipopolissacarídeos/metabolismo , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , PPAR gama/genética , PPAR gama/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
14.
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
15.
Nutr Cancer ; 63(8): 1287-96, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21958043

RESUMO

Prostate cancer is the most commonly diagnosed solid malignancy, and tumor cells eventually transform to castrate resistance through multiple pathways including activation of the androgen receptor via insulin-like growth factor receptor (IGF-1R) signaling involving phospho-AKT (pAKT). In this study, a mixture of herbal extracts, Zyflamend®, was used as a treatment in a model of castrate-resistant prostate cancer using CWR22Rv1 cells. Zyflamend reduced androgen receptor and IGF-1R expression along with a reduction of IGF-1-mediated proliferation of CWR22Rv1 cells. IGF-1 induced downstream AKT phosphorylation; however, the induction of pAKT was not associated with androgen receptor expression. Further, constitutively active form of AKT had no effect on nuclear expression of androgen receptor, indicating that upregulation of pAKT did not promote androgen receptor expression or nuclear translocation in castrate-resistant CWR22Rv1 cells. Conversely, Zyflamend reduced androgen receptor expression following IGF-1 stimulation and in cells overexpressing pAKT. These results demonstrated that Zyflamend inhibited IGF-1-stimulated cell growth, IGF-1R expression, and androgen receptor expression and its nuclear localization, but these effects were not dependent upon phosphatidylinositol 3-kinase/pAKT signaling. In conclusion, Zyflamend decreased cell proliferation and inhibited IGF-1R and androgen receptor expression in a phosphatidylinositol 3-kinase/pAKT independent manner.


Assuntos
Proliferação de Células , Extratos Vegetais/farmacologia , Receptor IGF Tipo 1/metabolismo , Receptores Androgênicos/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Cross-Talk , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/genética , Receptores Androgênicos/genética , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
16.
Cell Immunol ; 271(2): 379-84, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21885043

RESUMO

The pro-inflammatory cytokine IL-1ß leads to losses in functional ß-cell mass in part by inducing the expression of genes that produce soluble mediators of inflammation, such as cyclooxygenase-2 (COX2). In the current study, we sought to understand what factors control the COX2 gene in response to IL-1ß and how prostaglandins downstream of COX2 impact pro-inflammatory gene transcription in pancreatic ß-cells. We analyzed COX2 gene expression in response to different maneuvers impacting NF-κB proteins. Also, we report alterations in the expression of COX2, EP-3 and EP-4 receptor genes by PGD(2) and PGE(2). Moreover, we examined whether PGD(2) and PGE(2) regulated NF-κB and interferon-gamma activated sequence (GAS) reporter gene activity. IL-1ß-mediated induction of the COX2 gene requires the p65 and p50 subunits of NF-κB. In addition, PGD(2) and PGE(2) coordinately alter COX2 and EP receptor gene expression patterns and potentiate the cytokine-mediated transcriptional activity of promoters containing NF-κB or GAS response elements.


Assuntos
Ciclo-Oxigenase 2/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Interleucina-1beta/farmacologia , Prostaglandinas/farmacologia , Animais , Linhagem Celular Tumoral , Dinoprostona/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/farmacologia , Células Secretoras de Insulina/imunologia , Insulinoma/enzimologia , Insulinoma/genética , Insulinoma/imunologia , Interleucina-1beta/imunologia , Subunidade p50 de NF-kappa B/metabolismo , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Regiões Promotoras Genéticas , Prostaglandina D2/farmacologia , Ratos , Receptores de Prostaglandina E Subtipo EP3/genética , Receptores de Prostaglandina E Subtipo EP4/genética , Fator de Transcrição RelA/metabolismo
17.
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
18.
J Mol Biol ; 392(3): 578-88, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19631660

RESUMO

Glucose and cAMP reciprocally regulate expression of the L-type pyruvate kinase (L-PK) gene by controlling the formation of a complex containing the carbohydrate response element binding protein (ChREBP) and the coactivator CREB binding protein (CBP) on the L-PK promoter. However, the role of posttranslational histone modifications on the opposing effects of glucose and cAMP on the L-PK gene is unknown. Using the highly glucose-sensitive 832/13 rat insulinoma cell line, we demonstrated that glucose regulates acetylation and methylation of various histone residues at the L-PK gene promoter. These glucose-dependent histone modifications correlated with an increase in the recruitment and phosphorylation of RNA polymerase II (Pol II) on the L-PK gene promoter. Conversely, the cAMP agonist forskolin prevented glucose-mediated expression of the L-PK gene by decreasing the acetylation of histones H3 and H4 on the promoter, decreasing the methylation of H3-K4 on the coding region, and increasing the methylation of H3-K9 on the coding region. These changes induced by cAMP culminated with a decrease in the glucose-dependent recruitment of phosphorylated Pol II to the L-PK gene promoter. Furthermore, maneuvers that interfere with the glucose-dependent assembly of ChREBP and CBP on the L-PK promoter, such as increasing intracellular cAMP levels, overexpression of a dominant-negative form of ChREBP, and small-interfering-RNA-mediated suppression of CBP abundance, all altered the acetylation and methylation of histones on the L-PK promoter, which decreased Pol II recruitment and subsequently inhibited transcriptional activation of the L-PK gene. We conclude that the effects of glucose and cAMP are mediated in part by epigenetic modulation of histones.


Assuntos
AMP Cíclico/metabolismo , Glucose/metabolismo , Histonas/metabolismo , Holoenzimas/metabolismo , Regiões Promotoras Genéticas , Piruvato Quinase , RNA Polimerase II/metabolismo , Animais , Linhagem Celular , Colforsina/metabolismo , Regulação da Expressão Gênica , Histonas/genética , Holoenzimas/genética , Metilação , Piruvato Quinase/genética , Piruvato Quinase/metabolismo , RNA Polimerase II/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais/fisiologia
19.
FASEB J ; 23(9): 2855-65, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19406844

RESUMO

Glucose-mediated activation of the L-type pyruvate kinase (L-PK) gene is repressed by cAMP, making this an excellent model for studying the mechanism by which these contrary signals regulate gene expression. Using the 832/13 rat insulinoma cell line, we demonstrate using RNA interference and chromatin immunoprecipitation that carbohydrate response element binding protein (ChREBP), hepatic nuclear factor 4alpha (HNF4alpha), and the coactivator CREB binding protein (CBP) are required for the glucose response of the L-PK gene and are recruited to the promoter by glucose. The cAMP agonist forskolin blocked the glucose-mediated induction of the L-PK gene in a PKA-dependent manner and blocked the recruitment of ChREBP, HNF4alpha, and CBP to the L-PK promoter, while simultaneously recruiting CBP to the cAMP-inducible gene, nuclear receptor subfamily 4, group A, member 2 (NR4A2). Overexpression of CBP, but not ChREBP, reversed the cAMP repression of the L-PK gene. In addition, CBP augmented the glucose response of the L-PK promoter. We conclude that cAMP and glucose signaling converge on a complex containing ChREBP, HNF4alpha, and CBP, and that cAMP acts by disrupting this transcriptional complex assembled by glucose-derived signals.


Assuntos
AMP Cíclico/farmacologia , Glucose/farmacologia , Complexos Multiproteicos/metabolismo , Piruvato Quinase/genética , Ativação Transcricional/efeitos dos fármacos , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proteína de Ligação a CREB/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico , Fator 4 Nuclear de Hepatócito/metabolismo , Regiões Promotoras Genéticas , Transporte Proteico , Ratos , Transdução de Sinais , Transcrição Gênica
20.
Biochem Biophys Res Commun ; 372(1): 131-6, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18468514

RESUMO

Glucose has powerful effects on gene expression and participates in the fasted-to-fed transition of the liver. However, the molecular mechanism of glucose-regulated gene expression has not been completely described. In the present study, we performed a detailed analysis of the molecular events of the insulin-independent glucose response of the liver-type pyruvate kinase (L-PK) gene. L-PK mRNA was increased by glucose at the transcriptional level as determined by real-time RT-PCR, mRNA stability measurements, and nuclear run-on assays. LY294002 and LY303511 inhibited the glucose response of the L-PK gene at the transcriptional level. Histones H3 and H4 associated with the L-PK gene promoter were hyperacetylated and HNF4alpha was constitutively bound in low and high glucose. Treatment with 20mM glucose increased recruitment of ChREBP, additional HNF4alpha, and RNA polymerase II. Glucose-stimulated the phosphorylation of the C-terminal domain of RNA polymerase II, with increased Ser5 phosphorylation near the transcription start site and increased Ser2 phosphorylation near the termination signal. LY294002 and LY303511 blocked the recruitment of RNA polymerase II to the L-PK gene, reducing the rate of transcription. The results of these studies demonstrate fundamental details of the molecular mechanism of glucose activated gene expression.


Assuntos
Regulação da Expressão Gênica , Glucose/metabolismo , Fígado/enzimologia , Piruvato Quinase/genética , Acetilação , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Linhagem Celular Tumoral , Cromonas/farmacologia , Glucose/farmacologia , Fator 4 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Piperazinas/farmacologia , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Ratos , Serina/metabolismo , Sítio de Iniciação de Transcrição , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA