Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(9)2023 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-37175791

RESUMO

Pancreatic beta cell function is an important component of glucose homeostasis. Here, we investigated the function of PIMT (PRIP-interacting protein with methyl transferase domain), a transcriptional co-activator binding protein, in the pancreatic beta cells. We observed that the protein levels of PIMT, along with key beta cell markers such as PDX1 (pancreatic and duodenal homeobox 1) and MafA (MAF bZIP transcription factor A), were reduced in the beta cells exposed to hyperglycemic and hyperlipidemic conditions. Consistently, PIMT levels were reduced in the pancreatic islets isolated from high fat diet (HFD)-fed mice. The RNA sequencing analysis of PIMT knockdown beta cells identified that the expression of key genes involved in insulin secretory pathway, Ins1 (insulin 1), Ins2 (insulin 2), Kcnj11 (potassium inwardly-rectifying channel, subfamily J, member 11), Kcnn1 (potassium calcium-activated channel subfamily N member 1), Rab3a (member RAS oncogene family), Gnas (GNAS complex locus), Syt13 (synaptotagmin 13), Pax6 (paired box 6), Klf11 (Kruppel-Like Factor 11), and Nr4a1 (nuclear receptor subfamily 4, group A, member 1) was attenuated due to PIMT depletion. PIMT ablation in the pancreatic beta cells and in the rat pancreatic islets led to decreased protein levels of PDX1 and MafA, resulting in the reduction in glucose-stimulated insulin secretion (GSIS). The results from the immunoprecipitation and ChIP experiments revealed the interaction of PIMT with PDX1 and MafA, and its recruitment to the insulin promoter, respectively. Importantly, PIMT ablation in beta cells resulted in the nuclear translocation of insulin. Surprisingly, forced expression of PIMT in beta cells abrogated GSIS, while Ins1 and Ins2 transcript levels were subtly enhanced. On the other hand, the expression of genes, PRIP/Asc2/Ncoa6 (nuclear receptor coactivator 6), Pax6, Kcnj11, Syt13, Stxbp1 (syntaxin binding protein 1), and Snap25 (synaptosome associated protein 25) associated with insulin secretion, was significantly reduced, providing an explanation for the decreased GSIS upon PIMT overexpression. Our findings highlight the importance of PIMT in the regulation of insulin synthesis and secretion in beta cells.


Assuntos
Células Secretoras de Insulina , Insulina , Animais , Camundongos , Ratos , Genes Homeobox , Glucose/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Insulina/metabolismo , Insulina Regular Humana , Células Secretoras de Insulina/metabolismo , Potássio/metabolismo , Transativadores/metabolismo , Histonas
2.
Drug Discov Today ; 27(10): 103317, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35835313

RESUMO

That reversible protein phosphorylation by kinases and phosphatases occurs in metabolic disorders is well known. Various studies have revealed that a multi-faceted and tightly regulated phosphatase, pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP)-1/2 displays robust effects in cardioprotection, ischaemia/reperfusion (I/R), and vascular remodelling. PHLPP1 promotes foamy macrophage development through ChREBP/AMPK-dependent pathways. Adipocyte-specific loss of PHLPP2 reduces adiposity, improves glucose tolerance,and attenuates fatty liver via the PHLPP2-HSL-PPARα axis. Discoveries of PHLPP1-mediated insulin resistance and pancreatic ß cell death via the PHLPP1/2-Mst1-mTORC1 triangular loop have shed light on its significance in diabetology. PHLPP1 downregulation attenuates diabetic cardiomyopathy (DCM) by restoring PI3K-Akt-mTOR signalling. In this review, we summarise the functional role of, and cellular signalling mediated by, PHLPPs in metabolic tissues and discuss their potential as therapeutic targets.


Assuntos
Resistência à Insulina , Fosfoproteínas Fosfatases , Proteínas Quinases Ativadas por AMP , Glucose , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Nucleares/metabolismo , PPAR alfa , Fosfatidilinositol 3-Quinases , Fosfoproteínas Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR
3.
Sci Rep ; 8(1): 9599, 2018 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-29942003

RESUMO

AMPK is considered as a potential high value target for metabolic disorders. Here, we present the molecular modeling, in vitro and in vivo characterization of Activator-3, 2-[2-(4-(trifluoromethyl)phenylamino)thiazol-4-yl]acetic acid, an AMP mimetic and a potent pan-AMPK activator. Activator-3 and AMP likely share common activation mode for AMPK activation. Activator-3 enhanced AMPK phosphorylation by upstream kinase LKB1 and protected AMPK complex against dephosphorylation by PP2C. Molecular modeling analyses followed by in vitro mutant AMPK enzyme assays demonstrate that Activator-3 interacts with R70 and R152 of the CBS1 domain on AMPK γ subunit near AMP binding site. Activator-3 and C2, a recently described AMPK mimetic, bind differently in the γ subunit of AMPK. Activator-3 unlike C2 does not show cooperativity of AMPK activity in the presence of physiological concentration of ATP (2 mM). Activator-3 displays good pharmacokinetic profile in rat blood plasma with minimal brain penetration property. Oral treatment of High Sucrose Diet (HSD) fed diabetic rats with 10 mg/kg dose of Activator-3 once in a day for 30 days significantly enhanced glucose utilization, improved lipid profiles and reduced body weight, demonstrating that Activator-3 is a potent AMPK activator that can alleviate the negative metabolic impact of high sucrose diet in rat model.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Acetatos/farmacologia , Tiazóis/farmacologia , Proteínas Quinases Ativadas por AMP/química , Acetatos/metabolismo , Acetatos/farmacocinética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células Hep G2 , Humanos , Simulação de Acoplamento Molecular , Domínios Proteicos , Ratos , Tiazóis/metabolismo , Tiazóis/farmacocinética
4.
Biochem Biophys Res Commun ; 486(2): 533-538, 2017 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-28322791

RESUMO

We have previously reported that bacterial endotoxin LPS attenuates expression of PHLPP, a ser/thr phosphatase, at both transcript and protein levels in different immune cells, however the underlying molecular mechanism is unknown and is of significant interest. Here, in line with the decreased transcript levels upon LPS treatment, we observed that LPS caused significant reduction in PHLPP promoter activity. We observed that SP1, a transcription factor frequently associated with inflammation, was recruited to the PHLPP promoter region. Ectopic expression of SP1 enhanced both transcript and protein levels of PHLPP while knockdown of SP1 or pharmacological inhibition of SP1 DNA binding by mithramycin reduced PHLPP expression. Moreover, over-expression of SP1 co-activators CBP/p300 augmented SP1 driven PHLPP promoter activity. Of note, LPS treatment depleted SP1 and CBP protein levels due to which recruitment of SP1 to PHLPP promoter was reduced. Further, we found that re-introduction of SP1 restored promoter activity and transcript levels of PHLPP in LPS stimulated cells. Collectively, our data revealed the molecular mechanism underlying the regulation of PHLPP expression during LPS induced macrophage inflammatory response.


Assuntos
Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , RNA Mensageiro/genética , Fator de Transcrição Sp1/genética , Animais , Linhagem Celular , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Luciferases/genética , Luciferases/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/imunologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/imunologia , Plicamicina/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Transcrição Sp1/imunologia , Transcrição Gênica , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/imunologia
5.
Biochem Biophys Res Commun ; 483(1): 69-74, 2017 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-28063927

RESUMO

Post translational modifications, ubiquitination and its reversal by deubiquitination play an important role in regulating innate immune system. USP12 is a poorly studied deubiquitinase reported to regulate T-cell receptor signalling however the functional role of USP12 in macrophages, the principal architects of inflammation, is unknown. Thus, in this study we probed the involvement of USP12 in macrophage mediated inflammatory responses using bacterial endotoxin, LPS, as the model system. Here, we observed that the expression of USP12 was altered in time dependent manner in LPS stimulated RAW 264.7 macrophages at both mRNA and protein levels as revealed by qPCR and western blot analysis, respectively. Further analysis showed that LPS reduced the levels of Sp1 which enhanced the transcriptional levels of USP12. We observed that siRNA mediated ablation of USP12 expression in mouse macrophages suppressed the induction of LPS-induced iNOS and IL-6 expression but failed to alter IFN-ß synthesis, oxidative stress and phagocytic ability of macrophages. Mechanistic analysis suggest that USP12 may be required for the activation of NFκB pathway as knockdown of USP12 reduced the inhibitory phosphorylation of IκBα, a well characterized inhibitor of NFκB nuclear translocation. Further, USP12 was observed to be required for LPS elicited phosphorylation of ERK1/2 and p38. Collectively, our data suggest that USP12 may be a key mediator of LPS stimulated macrophage responses.


Assuntos
Endopeptidases/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Inibidor de NF-kappaB alfa/antagonistas & inibidores , Animais , Endopeptidases/deficiência , Endopeptidases/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Inflamação/metabolismo , Interferon gama/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Ativação de Macrófagos/fisiologia , Macrófagos/imunologia , Camundongos , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Células RAW 264.7 , RNA Interferente Pequeno/genética , Ubiquitina Tiolesterase
6.
Mol Immunol ; 82: 1-9, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27992764

RESUMO

Chronic inflammatory diseases such as insulin resistance, Type 2 diabetes, neurodegenerative diseases etc., are shown to be caused due to imbalanced activation states of macrophages. MicroRNAs which are transcriptional/post-transcriptional regulators of gene expression drive several pathophysiological processes including macrophage polarization. However the functional role of microRNAs in regulating inflammation induced insulin resistance is ill defined. In our current study we observed that the expression of miR-712 was reduced in macrophages exposed to LPS and IFN-γ. Ectopic expression of miR-712 in RAW 264.7 mouse macrophages impaired the expression of iNOS protein and secretion of pro-inflammatory cytokines such as TNF-α, IL-6 and IFN-ß which in turn led to improved insulin stimulated glucose uptake in co-cultured L6 myoblasts. Mechanistically, we identified that miR-712 targets the 3'UTR of a potent inflammatory gene LRRK2 and dampens the phosphorylation of p38 and ERK1/2 kinases. Taken together, our data underscore the regulatory role of miR-712 in restoring insulin stimulated glucose uptake by myoblasts through down-regulating macrophage mediated inflammatory responses.


Assuntos
Resistência à Insulina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/imunologia , Ativação de Macrófagos/genética , MicroRNAs/imunologia , Mioblastos/metabolismo , Animais , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica/imunologia , Glucose/metabolismo , Immunoblotting , Inflamação/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Camundongos , Mioblastos/imunologia , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real
7.
Biochimie ; 119: 16-26, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26453808

RESUMO

Uncontrolled inflammation leads to several diseases such as insulin resistance, T2D and several types of cancers. The functional role of microRNAs in inflammation induced insulin resistance is poorly studied. MicroRNAs are post-transcriptional regulatory molecules which mediate diverse biological processes. We here show that miR-16 expression levels are down-regulated in different inflammatory conditions such as LPS/IFNγ or palmitate treated macrophages, palmitate exposed myoblasts and insulin responsive tissues of high sucrose diet induced insulin resistant rats. Importantly, forced expression of miR-16 in macrophages impaired the production of TNF-α, IL-6 and IFN-ß leading to enhanced insulin stimulated glucose uptake in co-cultured skeletal myoblasts. Further, ectopic expression of miR-16 enhanced insulin stimulated glucose uptake in skeletal myoblasts via the up-regulation of GLUT4 and MEF2A, two key players involved in insulin stimulated glucose uptake. Collectively, our data highlight the important role of miR-16 in ameliorating inflammation induced insulin resistance.


Assuntos
Regulação para Baixo , Resistência à Insulina , Ativação de Macrófagos , Macrófagos/metabolismo , MicroRNAs/metabolismo , Mioblastos/metabolismo , Animais , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Polaridade Celular/efeitos dos fármacos , Técnicas de Cocultura , Sacarose Alimentar/efeitos adversos , Regulação para Baixo/efeitos dos fármacos , Endotoxinas/toxicidade , Ácidos Graxos não Esterificados/efeitos adversos , Interferon gama/genética , Interferon gama/metabolismo , Interferon gama/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , MicroRNAs/antagonistas & inibidores , Músculo Esquelético/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/imunologia , Células RAW 264.7 , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
8.
Sci Rep ; 5: 15197, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26468734

RESUMO

The mechanisms underlying inflammation induced insulin resistance are poorly understood. Here, we report that the expression of PIMT, a transcriptional co-activator binding protein, was up-regulated in the soleus muscle of high sucrose diet (HSD) induced insulin resistant rats and TNF-α exposed cultured myoblasts. Moreover, TNF-α induced phosphorylation of PIMT at the ERK1/2 target site Ser(298). Wild type (WT) PIMT or phospho-mimic Ser298Asp mutant but not phospho-deficient Ser298Ala PIMT mutant abrogated insulin stimulated glucose uptake by L6 myotubes and neonatal rat skeletal myoblasts. Whereas, PIMT knock down relieved TNF-α inhibited insulin signaling. Mechanistic analysis revealed that PIMT differentially regulated the expression of GLUT4, MEF2A, PGC-1α and HDAC5 in cultured cells and skeletal muscle of Wistar rats. Further characterization showed that PIMT was recruited to GLUT4, MEF2A and HDAC5 promoters and overexpression of PIMT abolished the activity of WT but not MEF2A binding defective mutant GLUT4 promoter. Collectively, we conclude that PIMT mediates TNF-α induced insulin resistance at the skeletal muscle via the transcriptional modulation of GLUT4, MEF2A, PGC-1α and HDAC5 genes.


Assuntos
Transportador de Glucose Tipo 4/metabolismo , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/metabolismo , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Glicemia/análise , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Transportador de Glucose Tipo 4/genética , Células HEK293 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Resistência à Insulina , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosforilação/efeitos dos fármacos , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/genética , Ratos , Ratos Wistar , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/sangue
9.
J Leukoc Biol ; 95(5): 775-783, 2014 05.
Artigo em Inglês | MEDLINE | ID: mdl-24443556

RESUMO

PHLPP1 is a novel tumor suppressor, but its role in the regulation of innate immune responses, which are frequently dysregulated in cancer, is unexplored. Here, we report that LPS attenuated PHLPP1 expression at mRNA and protein levels in immune cells, suggesting its involvement in immune responses. To test this, we overexpressed PHLPP1 in RAW 264.7 macrophages and observed a dramatic reduction in LPS/IFN-γ-induced iNOS expression. Conversely, silencing of PHLPP1 by siRNA or by shRNA robustly augmented LPS/IFN-γ-induced iNOS expression. qPCR and iNOS promoter reporter experiments showed that PHLPP1 inhibited iNOS transcription. Mechanistic analysis revealed that PHLPP1 suppressed LPS/IFN-γ-induced phosphorylation of ser727 STAT1; however, the underlying mechanisms differed. PHLPP1 reduced IFN-γ-stimulated but not LPS-induced ERK1/2 phosphorylation, and inhibition of ERK1/2 abolished IFN-γ-induced ser727 STAT1 phosphorylation and iNOS expression. In contrast, PHLPP1 knockdown augmented LPS-induced but not IFN-γ-elicited p38 phosphorylation. Blockade of p38 abolished LPS-stimulated phosphorylation of ser727 STAT1 and iNOS expression. Furthermore, PHLPP1 suppressed LPS-induced phosphorylation of tyr701 STAT1 by dampening p38-dependent IFN-ß feedback. Collectively, our data demonstrate for the first time that PHLPP1 plays a vital role in restricting innate immune responses of macrophages, and further studies may show it to be a potential therapeutic target within the context of dysregulated macrophage activity.


Assuntos
Imunomodulação , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Proteínas Nucleares/imunologia , Fosfoproteínas Fosfatases/imunologia , Fator de Transcrição STAT1/imunologia , Animais , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/imunologia , Interferon gama , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Óxido Nítrico Sintase Tipo II/genética , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fosforilação/imunologia , Células RAW 264.7 , Fator de Transcrição STAT1/genética , Serina/imunologia
10.
PLoS One ; 8(12): e83787, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24358311

RESUMO

PRIP-Interacting protein with methyl transferase domain (PIMT) serves as a molecular bridge between CREB-binding protein (CBP)/ E1A binding protein p300 (Ep300) -anchored histone acetyl transferase and the Mediator complex sub-unit1 (Med1) and modulates nuclear receptor transcription. Here, we report that ERK2 phosphorylates PIMT at Ser(298) and enhances its ability to activate PEPCK promoter. We observed that PIMT is recruited to PEPCK promoter and adenoviral-mediated over-expression of PIMT in rat primary hepatocytes up-regulated expression of gluconeogenic genes including PEPCK. Reporter experiments with phosphomimetic PIMT mutant (PIMT(S298D)) suggested that conformational change may play an important role in PIMT-dependent PEPCK promoter activity. Overexpression of PIMT and Med1 together augmented hepatic glucose output in an additive manner. Importantly, expression of gluconeogenic genes and hepatic glucose output were suppressed in isolated liver specific PIMT knockout mouse hepatocytes. Furthermore, consistent with reporter experiments, PIMT(S298D) but not PIMT(S298A) augmented hepatic glucose output via up-regulating the expression of gluconeogenic genes. Pharmacological blockade of MAPK/ERK pathway using U0126, abolished PIMT/Med1-dependent gluconeogenic program leading to reduced hepatic glucose output. Further, systemic administration of T4 hormone to rats activated ERK1/2 resulting in enhanced PIMT ser(298) phosphorylation. Phosphorylation of PIMT led to its increased binding to the PEPCK promoter, increased PEPCK expression and induction of gluconeogenesis in liver. Thus, ERK2-mediated phosphorylation of PIMT at Ser(298) is essential in hepatic gluconeogenesis, demonstrating an important role of PIMT in the pathogenesis of hyperglycemia.


Assuntos
Gluconeogênese/fisiologia , Hepatócitos/metabolismo , Fígado/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/metabolismo , Animais , Linhagem Celular , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Gluconeogênese/efeitos dos fármacos , Glucose/biossíntese , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Knockout , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , Ligação Proteica , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/genética , Proteínas Serina-Treonina Quinases/genética , Ratos , Especificidade por Substrato , Hormônios Tireóideos/farmacologia , Transcrição Gênica , Ativação Transcricional
11.
Org Biomol Chem ; 11(39): 6680-5, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-23986357

RESUMO

A series of 1,3-disubstituted pyrrolo[2,3-b]quinoxalines has been designed for the potential inhibition of PDE4 without inhibiting luciferase. A ligand/PTC (phase transfer catalyst) free intramolecular Heck cyclization strategy was used to prepare these compounds, some of which showed significant inhibition of PDE4B (IC50≈ 5-14 µM) and growth inhibition of oral cancer cells (CAL 27) but not inhibition of luciferase in vitro. They also showed acceptable safety profiles but no apoptosis in zebrafish embryos.


Assuntos
Neoplasias Bucais/tratamento farmacológico , Inibidores da Fosfodiesterase 4/síntese química , Inibidores da Fosfodiesterase 4/farmacologia , Quinoxalinas/síntese química , Quinoxalinas/farmacologia , Animais , Sítios de Ligação , Catálise , Proliferação de Células/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Ligantes , Luciferases/metabolismo , Estrutura Molecular , Inibidores da Fosfodiesterase 4/química , Quinoxalinas/química , Peixe-Zebra/embriologia
12.
Org Biomol Chem ; 11(13): 2075-9, 2013 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-23436140

RESUMO

A new strategy for converting antipsychotic drug olanzapine into PDE4 inhibitors is described via the design and Pd/C mediated synthesis of novel N-indolylmethyl olanzapine derivatives. One compound showed good inhibition (IC50 1.1 µM) and >10 fold selectivity towards PDE4B over D that was supported by docking studies. This compound also showed significant inhibition of TNF-α and no major toxicities in cell lines and a zebrafish embryo model except the teratogenic effects to be re-assessed in rodents.


Assuntos
Antipsicóticos/farmacologia , Benzodiazepinas/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Desenho de Fármacos , Inibidores da Fosfodiesterase 4/síntese química , Inibidores da Fosfodiesterase 4/farmacologia , Antipsicóticos/síntese química , Antipsicóticos/química , Benzodiazepinas/síntese química , Benzodiazepinas/química , Relação Dose-Resposta a Droga , Humanos , Olanzapina , Inibidores da Fosfodiesterase 4/química , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/antagonistas & inibidores
13.
Org Biomol Chem ; 10(29): 5554-69, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22710638

RESUMO

Novel thieno[2,3-d]pyrimidines containing a cyclohexane ring fused with a six- or five-membered heterocyclic moiety along with a benzylic nitrile were designed as potential inhibitors of PDE4. Expeditious synthesis of these compounds was carried out via a multi-step sequence consisting of a few key steps such as Gewald reaction, Dieckmann type cyclisation and Krapcho decarboxylation. This newly developed strategy involved construction of the thienopyrimidine ring followed by the cyclohexanone moiety and subsequently the fused heterocyclic ring. A number of thieno[2,3-d]pyrimidine based derivatives were synthesized using this method some of which showed promising PDE4B inhibitory properties. One of them was tested for PDE4D inhibition in vitro and dose dependent inhibition of TNF-α. A few selected molecules were docked into the PE4B protein the results of which showed good overall correlations to their observed PDE4B inhibitory properties in vitro. The crystal structure analysis of representative compounds along with hydrogen bonding patterns and molecular arrangement present within the molecule is described.


Assuntos
Pirimidinas/química , Tiofenos/química , Animais , Linhagem Celular , Cristalografia por Raios X , Cicloexanonas/química , Humanos , Ligação de Hidrogênio , Camundongos , Modelos Moleculares , Inibidores da Fosfodiesterase 4/síntese química , Inibidores da Fosfodiesterase 4/química , Inibidores da Fosfodiesterase 4/farmacologia , Pirimidinas/síntese química , Pirimidinas/farmacologia , Tiofenos/síntese química , Tiofenos/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/metabolismo
15.
Mol Cell Biochem ; 351(1-2): 217-32, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21253851

RESUMO

Bcl-2 is an anti-apoptotic member of the Bcl-2 family of proteins that protects cells from apoptosis induced by a large variety of stimuli. The protein BMRP (MRPL41) was identified as a Bcl-2 binding partner and shown to have pro-apoptotic activity. We have performed deletion mutational analyses to identify the domain(s) of Bcl-2 and BMRP that are involved in the Bcl-2/BMRP interaction, and the region(s) of BMRP that mediate its pro-apoptotic activity. The results of these studies indicate that both the BH4 domain of Bcl-2 and its central region encompassing its BH1, BH2, and BH3 domains are required for its interaction with BMRP. The loop region and the transmembrane domain of Bcl-2 were found to be dispensable for this interaction. The Bcl-2 deletion mutants that do not interact with BMRP were previously shown to be functionally inactive. Deletion analyses of the BMRP protein delimited the region of BMRP needed for its interaction with Bcl-2 to the amino-terminal two-thirds of the protein (amino acid residues 1-92). Further deletions at either end of the BMRP(1-92) truncated protein resulted in lack of binding to Bcl-2. Functional studies performed with BMRP deletion mutants suggest that the cell death-inducing domains of the protein reside mainly within its amino-terminal two-thirds. The region of BMRP required for the interaction with Bcl-2 is very relevant for the cell death-inducing activity of the protein, suggesting that one possible mechanism by which BMRP induces cell death is by binding to and blocking the anti-apoptotic activity of Bcl-2.


Assuntos
Apoptose , Proteínas Mitocondriais/metabolismo , Mutação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Ribossômicas/metabolismo , Deleção de Sequência , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Humanos , Camundongos , Proteínas Mitocondriais/genética , Células NIH 3T3 , Ligação Proteica , Proteínas Ribossômicas/genética , Técnicas do Sistema de Duplo-Híbrido
16.
PLoS One ; 4(11): e7919, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19936232

RESUMO

Francisella tularensis infects macrophages and escapes phago-lysosomal fusion to replicate within the host cytosol, resulting in host cell apoptosis. Here we show that the Fas-mediated death pathway is activated in infected cells and correlates with escape of the bacterium from the phagosome and the bacterial burden. Our studies also demonstrate that constitutive activation of Akt, or deletion of SHIP, promotes phago-lysosomal fusion and limits bacterial burden in the host cytosol, and the subsequent induction of Fas expression and cell death. Finally, we show that phagosomal escape/intracellular bacterial burden regulate activation of the transcription factors sp1/sp3, leading to Fas expression and cell death. These data identify for the first time host cell signaling pathways that regulate the phagosomal escape of Francisella, leading to the induction of Fas and subsequent host cell death.


Assuntos
Francisella/metabolismo , Fagossomos/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor fas/metabolismo , Animais , Células da Medula Óssea/citologia , Morte Celular , Linhagem Celular , Citosol/metabolismo , Inositol Polifosfato 5-Fosfatases , Lisossomos/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose
17.
Curr Drug Targets ; 10(1): 71-87, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19149538

RESUMO

Dipeptidyl peptidase IV (DPP IV) is a key regulator of insulin-stimulating hormones, glucagon-like peptide (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), thus it is a promising target for the treatment of Type 2 Diabetes mellitus (T2DM). Inhibition of plasma DPP IV enzyme leads to enhanced endogenous GLP-1 and GIP activity, which ultimately results in the potentiation of insulin secretion by pancreatic beta-cells and subsequent lowering of blood glucose levels, HbA[1(c)], glucagon secretion and liver glucose production. Various classes of structurally different DPP IV inhibitors are currently being explored and few of them such as Sitagliptin and Vildagliptin were successfully launched. These drugs have been approved as a once-daily oral monotherapy or as a combination therapy with current anti-diabetic agents like pioglitazone, glibenclamide, metformin etc. for the treatment of T2DM. Several other novel DPP IV inhibitors are in pipeline. The present review summarizes the latest preclinical and clinical trial data of different DPP IV inhibitors with a special emphasis on their DPP8/9 fold selectivity and therapeutic advantages over GLP-1 based approach.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipeptidil Peptidase 4/efeitos adversos , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Drogas em Investigação/uso terapêutico , Hipoglicemiantes/uso terapêutico , Adamantano/administração & dosagem , Adamantano/efeitos adversos , Adamantano/análogos & derivados , Adamantano/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 2/enzimologia , Inibidores da Dipeptidil Peptidase IV/administração & dosagem , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Drogas em Investigação/administração & dosagem , Drogas em Investigação/efeitos adversos , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/efeitos adversos , Nitrilas/administração & dosagem , Nitrilas/efeitos adversos , Nitrilas/uso terapêutico , Pirazinas/administração & dosagem , Pirazinas/efeitos adversos , Pirazinas/uso terapêutico , Pirrolidinas/administração & dosagem , Pirrolidinas/efeitos adversos , Pirrolidinas/uso terapêutico , Fosfato de Sitagliptina , Especificidade por Substrato , Resultado do Tratamento , Triazóis/administração & dosagem , Triazóis/efeitos adversos , Triazóis/uso terapêutico , Vildagliptina
18.
J Immunol ; 180(12): 7847-58, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18523248

RESUMO

The pulmonary innate immune system responds to various airborne microbes. Although its specificity is broad and based on the recognition of pathogen-associated molecular patterns, it is uniquely regulated to limit inflammation and thereby prevent damage to the gas-exchanging alveoli. Macrophages, critical cell determinants of this system, recognize microbes through pattern recognition receptors such as TLRs, which typically mediate proinflammatory responses. The lung collectin, surfactant protein A (SP-A), has emerged as an important innate immune determinant that regulates microbe-macrophage interactions in this environment. In this study, we report the basal and SP-A-induced transcriptional and posttranslational regulation of TLR2 and TLR4 expression during the differentiation of primary human monocytes into macrophages. Despite SP-A's ability to up-regulate TLR2 expression on human macrophages, it dampens TLR2 and TLR4 signaling in these cells. SP-A decreases the phosphorylation of IkappaBalpha, a key regulator of NF-kappaB activity, and nuclear translocation of p65 which result in diminished TNF-alpha secretion in response to TLR ligands. SP-A also reduces the phosphorylation of TLR signaling proteins upstream of NF-kappaB, including members of the MAPK family. Finally, we report for the first time that SP-A decreases the phosphorylation of Akt, a major cell regulator of NF-kappaB and potentially MAPKs. These data identify a critical role for SP-A in modulating the lung inflammatory response by regulating macrophage TLR activity.


Assuntos
Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Proteína A Associada a Surfactante Pulmonar/fisiologia , Receptores Toll-Like/biossíntese , Receptores Toll-Like/genética , Adulto , Diferenciação Celular/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Macrófagos Alveolares/citologia , Modelos Imunológicos , Monócitos/citologia , Monócitos/imunologia , Monócitos/metabolismo , Ligação Proteica/imunologia , Proteína A Associada a Surfactante Pulmonar/biossíntese , Proteína A Associada a Surfactante Pulmonar/genética , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/biossíntese , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/fisiologia , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/imunologia
19.
Mol Immunol ; 45(12): 3428-37, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18514317

RESUMO

Tularemia is a zoonotic disease caused by the Gram-negative intracellular pathogen Francisella tularensis. These bacteria evade phagolysosomal fusion, escape from the phagosome and replicate in the host cell cytoplasm. IFNgamma has been shown to suppress the intra-macrophage growth of Francisella through both nitric oxide-dependent and -independent pathways. Since Francisella is known to subvert host immune responses, we hypothesized that this pathogen could interfere with IFNgamma signaling. Here, we report that infection with Francisella suppresses IFNgamma-induced STAT1 expression and phosphorylation in both human and murine mononuclear phagocytes. This suppressive effect of Francisella is independent of phagosomal escape or replication and is mediated by a heat-stable and constitutively expressed bacterial factor. An analysis of the molecular mechanism of STAT1 inhibition indicated that expression of SOCS3, an established negative regulator of IFNgamma signaling, is highly up-regulated during infection and suppresses STAT1 phosphorylation. Functional analyses revealed that this interference with IFNgamma signaling is accompanied by the suppression of IP-10 production and iNOS induction resulting in increased intracellular bacterial survival. Importantly, the suppressive effect on IFNgamma-mediated host cell protection is most effective when IFNgamma is added post infection, suggesting that the bacteria establish a permissive environment within the host cell.


Assuntos
Francisella tularensis/citologia , Interferon gama/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/microbiologia , Viabilidade Microbiana/imunologia , Fagócitos/imunologia , Fagócitos/microbiologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Francisella tularensis/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/microbiologia , Camundongos , Viabilidade Microbiana/efeitos dos fármacos , Modelos Imunológicos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/microbiologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Fagócitos/efeitos dos fármacos , Fagossomos/efeitos dos fármacos , Fagossomos/microbiologia , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Tularemia/imunologia , Tularemia/microbiologia
20.
Mol Immunol ; 45(10): 3012-21, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18295889

RESUMO

Francisella tularensis is a highly infectious, Gram-negative intra-cellular pathogen that can cause the zoonotic disease tularemia. Although the receptors critical for internalization of Francisella by macrophages are beginning to be defined, the identity of the downstream signaling pathways essential for the engulfment are not yet identified. In this study we have tested the role of Syk in the phagocytosis of Francisella. We report that Syk is activated during Francisella infection and is critical for the uptake of the organisms. Pharmacologic inhibition of Syk almost completely abrogated uptake, whereas the overexpression of Syk significantly enhanced uptake. However, Syk appears to be dispensable during initial host-pathogen contact. Further analyses of the molecular mechanism of Syk influence on Francisella uptake revealed that the MAPK Erk but not the phosphatidylinositol 3 kinase (PI3K)/Akt pathway is the downstream effector of Syk. Thus, the inhibition of Erk in Syk-overexpressing cells or the inhibition of Syk in Erk-overexpressing cells led to a significant attenuation of uptake. Collectively, these data identify Syk and Erk as key players in the phagocytosis of Francisella.


Assuntos
Francisella/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fagocitose , Proteínas Tirosina Quinases/metabolismo , Animais , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Francisella/efeitos dos fármacos , Infecções por Bactérias Gram-Negativas/enzimologia , Infecções por Bactérias Gram-Negativas/microbiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/microbiologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Fagocitose/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinase Syk
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA