Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
PLoS One ; 10(9): e0137515, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26407006

RESUMO

The heart hosts tissue resident macrophages which are capable of modulating cardiac inflammation and function by multiple mechanisms. At present, the consequences of phenotypic diversity in macrophages in the heart are incompletely understood. The contribution of cardiac M2-polarized macrophages to the resolution of inflammation and repair response following myocardial infarction remains to be fully defined. In this study, the role of M2 macrophages was investigated utilising a specific CSF-1 receptor signalling inhibition strategy to achieve their depletion. In mice, oral administration of GW2580, a CSF-1R kinase inhibitor, induced significant decreases in Gr1lo and F4/80hi monocyte populations in the circulation and the spleen. GW2580 administration also induced a significant depletion of M2 macrophages in the heart after 1 week treatment as well as a reduction of cardiac arginase1 and CD206 gene expression indicative of M2 macrophage activity. In a murine myocardial infarction model, reduced M2 macrophage content was associated with increased M1-related gene expression (IL-6 and IL-1ß), and decreased M2-related gene expression (Arginase1 and CD206) in the heart of GW2580-treated animals versus vehicle-treated controls. M2 depletion was also associated with a loss in left ventricular contractile function, infarct enlargement, decreased collagen staining and increased inflammatory cell infiltration into the infarct zone, specifically neutrophils and M1 macrophages. Taken together, these data indicate that CSF-1R signalling is critical for maintaining cardiac tissue resident M2-polarized macrophage population, which is required for the resolution of inflammation post myocardial infarction and, in turn, for preservation of ventricular function.


Assuntos
Macrófagos/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Transdução de Sinais , Animais , Anisóis/farmacologia , Arginase/genética , Arginase/metabolismo , Regulação da Expressão Gênica , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos/patologia , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Pirimidinas/farmacologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética
4.
Biomaterials ; 69: 22-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26275859

RESUMO

We evaluated the therapeutic efficacy of a novel drug eluting stent (DES) inhibiting inflammation and smooth muscle cell (SMC) proliferation. We identified CX3CR1 as a targetable receptor for prevention of monocyte adhesion and inflammation and in-stent neointimal hyperplasia without interfering with stent re-endothelization. Efficacy of AZ12201182 (AZ1220), a CX3CR1 antagonist was evaluated in inhibition of monocyte attachment in vitro. A prototype AZ1220 eluting PLGA-based polymer coated stent developed with an optimal elution profile and dose of 1 µM/stent was tested over 4 weeks in a porcine model of coronary artery stenting. Polymer coated stents without AZ1220 and bare metal stents were used as controls. AZ1220 inhibited monocyte attachment to CX3CL1 in a dose dependent manner. AZ1220 eluted from polymer coated stents in an ex vivo flow system retained bioactivity in inhibiting monocyte attachment to CX3CL1. At 4 weeks following deployment, AZ1220 eluting stents significantly reduced (∼60%) in-stent stenosis compared to both bare metal and polymer only coated stents and markedly reduced peri-stent inflammation and monocyte/macrophage accumulation without affecting re-endothelization. Anti-CX3CR1 drug eluting stents potently inhibited in-stent stenosis and may offer an alternative to mTOR targeting by current DES, specifically inhibiting polymer-induced inflammatory response and SMC proliferation, while retaining an equivalent re-endothelization response to bare metal stents.


Assuntos
Anti-Inflamatórios/administração & dosagem , Estenose Coronária/prevenção & controle , Vasos Coronários/efeitos dos fármacos , Stents Farmacológicos/efeitos adversos , Inflamação/prevenção & controle , Receptores de Quimiocinas/antagonistas & inibidores , Animais , Anti-Inflamatórios/uso terapêutico , Receptor 1 de Quimiocina CX3C , Proliferação de Células/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Estenose Coronária/etiologia , Estenose Coronária/patologia , Vasos Coronários/patologia , Vasos Coronários/cirurgia , Feminino , Inflamação/etiologia , Inflamação/patologia , Ácido Láctico/química , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Suínos
5.
Mol Pharm ; 12(3): 991-6, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25588055

RESUMO

To extend the temporal window for cytoprotection in cardiomyocytes undergoing apoptosis after hypoxia and myocardial infarction (MI), a synthetic chemically modified mRNA (modRNA) was used to drive delivery of insulin-like growth factor-1 (IGF1) within the area at risk in an in vivo murine model of MI. Delivery of IGF1 modRNA, with a polyethylenimine-based nanoparticle, augmented secreted and cell-associated IGF1, promoting cardiomyocyte survival and abrogating cell apoptosis under hypoxia-induced apoptosis conditions. Translation of modRNA-IGF1 was sufficient to induce downstream increases in the levels of Akt and Erk phosphorylation. Downregulation of IGF1 specific miRNA-1 and -133 but not miR-145 expression was also confirmed. As a proof of concept, intramyocardial delivery of modRNA-IGF1 but not control modRNA-GFP significantly decreased the level of TUNEL positive cells, augmented Akt phosphorylation, and decreased caspase-9 activity within the infarct border zone 24 h post-MI. These findings demonstrate the potential for an extended cytoprotective effect of transient IGF1 driven by synthetic modRNA delivery.


Assuntos
Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , RNA Mensageiro/administração & dosagem , RNA Mensageiro/genética , Animais , Biofarmácia , Linhagem Celular , Sobrevivência Celular , Citoproteção/genética , Sistemas de Liberação de Medicamentos , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Fator de Crescimento Insulin-Like I/genética , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Nanopartículas/química , Polietilenoimina/química , Transfecção
6.
Biochim Biophys Acta ; 1829(11): 1191-201, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24060351

RESUMO

Phenotypic switching of smooth muscle cells (SMCs) plays a central role in the development of vascular diseases such as atherosclerosis and restenosis. However, the factors regulating expression of the human myocardin (Myocd) gene, the master gene regulator of SMC differentiation, have yet to be identified. In this study, we sought to identify the critical factors regulating Myocd expression in human SMCs. Using deletion/genetic reporter analyses, an upstream repressor region (URR) was localised within the Myocd promoter, herein termed PrmM. Bioinformatic analysis revealed three evolutionary conserved Klf4 sites within the URR and disruption of those elements led to substantial increases in PrmM-directed gene expression. Furthermore, ectopic expression established that Klf4 significantly decreased Myocd mRNA levels and PrmM-directed gene expression while electrophoretic mobility shift assays and chromatin immunoprecipitation (ChIP) assays confirmed specific binding of endogenous Klf4, and not Klf5 or Klf2, to the URR of PrmM. Platelet-derived growth factor BB (PDGF-BB), a potent inhibitor of SMC differentiation, reduced Myocd mRNA levels and PrmM-directed gene expression in SMCs. A PDGF-BB-responsive region (PRR) was also identified within PrmM, overlapping with the previously identified URR, where either siRNA knockdown of Klf4 or the combined disruption of the Klf4 elements completely abolished PDGF-BB-mediated repression of PrmM-directed gene expression in SMCs. Moreover, ChIP analysis established that PDGF-BB-induced repression of Myocd gene expression is most likely regulated by enhanced binding of Klf4 and Klf5 to a lesser extent, to the PRR of PrmM. Taken together, these data provide critical insights into the transcriptional regulation of the Myocd gene in vascular SMCs, including during SMC differentiation.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Nucleares/genética , Transativadores/genética , Transcrição Gênica , Sequência de Bases , Becaplermina , Células Cultivadas , Primers do DNA , Humanos , Fator 4 Semelhante a Kruppel , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-sis/farmacologia , RNA Mensageiro/genética
7.
PLoS One ; 8(2): e57230, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437346

RESUMO

Monocyte/macrophages are implicated in initiation of angiogenesis, tissue/organ perfusion and atherosclerosis biology. We recently showed that chemokine receptor CX(3)CR1 is an essential regulator of monocyte/macrophage derived smooth muscle cell differentiation in the vessel wall after injury. Here we hypothesised the contribution of CX(3)CR1- CX(3)CL1 interaction to in vivo neovascularization and studied the functional consequences of genetic and pharmacologic targeting of CX(3)CR1 in formation, maturation and maintenance of microvascular integrity. Cells functionally deficient in CX(3)CR1 lacked matrix tunnelling and tubulation capacity in a 3D Matrigel assay. These morphogenic and cytokinetic responses were driven by CX(3)CL1-CX(3)CR1 interaction and totally abrogated by a Rho antagonist. To evaluate the role of CX(3)CR1 system in vivo, Matrigel plugs were implanted in competent CX(3)CR1(+/gfp) and functionally deficient CX(3)CR1(gfp/gfp) mice. Leaky microvessels (MV) were formed in the Matrigel implanted in CX(3)CR1(gfp/gfp) but not in CX(3)CR1(+/gfp) mice. In experimental plaque neovascularization immature MV phenotype was observed in CX(3)CR1(gfp/gfp) mice, lacking CX(3)CR1 positive smooth muscle-like cells, extracellular collagen and basement membrane (BM) laminin compared to competent CX(3)CR1(+/gfp) mice. This was associated with increased extravasation of platelets into the intima of CX(3)CR1(gfp/gfp) but not functionally competent CX(3)CR1 mice. Pharmacologic targeting using CX(3)CR1 receptor antagonist in wild type mice resulted in formation of plaque MV with poor BM coverage and a leaky phenotype. Our data indicate a hitherto unrecognised role for functional CX(3)CR1 in Matrigel and experimental plaque neovascularization in vivo, which may buttress MV collectively in favour of a more stable non-leaky phenotype.


Assuntos
Endotélio Vascular/metabolismo , Macrófagos/metabolismo , Microvasos/metabolismo , Monócitos/metabolismo , Receptores de Quimiocinas/genética , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/patologia , Receptor 1 de Quimiocina CX3C , Permeabilidade Capilar , Movimento Celular/efeitos dos fármacos , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Colágeno/genética , Colágeno/metabolismo , Combinação de Medicamentos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Expressão Gênica/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde , Laminina/genética , Laminina/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microvasos/efeitos dos fármacos , Microvasos/patologia , Monócitos/efeitos dos fármacos , Monócitos/patologia , Neovascularização Patológica , Peptídeos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteoglicanas , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/metabolismo
8.
J Lipid Res ; 53(11): 2390-404, 2012 11.
Artigo em Inglês | MEDLINE | ID: mdl-22969152

RESUMO

Prostacyclin and its prostacyclin receptor, the I Prostanoid (IP), play essential roles in regulating hemostasis and vascular tone and have been implicated in a range cardio-protective effects but through largely unknown mechanisms. In this study, the influence of cholesterol on human IP [(h)IP] gene expression was investigated in cultured vascular endothelial and platelet-progenitor megakaryocytic cells. Cholesterol depletion increased human prostacyclin receptor (hIP) mRNA, hIP promoter-directed reporter gene expression, and hIP-induced cAMP generation in all cell types. Furthermore, the constitutively active sterol-response element binding protein (SREBP)1a, but not SREBP2, increased hIP mRNA and promoter-directed gene expression, and deletional and mutational analysis uncovered an evolutionary conserved sterol-response element (SRE), adjacent to a known functional Sp1 element, within the core hIP promoter. Moreover, chromatin immunoprecipitation assays confirmed direct cholesterol-regulated binding of SREBP1a to this hIP promoter region in vivo, and immunofluorescence microscopy corroborated that cholesterol depletion significantly increases hIP expression levels. In conclusion, the hIP gene is directly regulated by cholesterol depletion, which occurs through binding of SREBP1a to a functional SRE within its core promoter. Mechanistically, these data establish that cholesterol can regulate hIP expression, which may, at least in part, account for the combined cardio-protective actions of low serum cholesterol through its regulation of IP expression within the human vasculature.


Assuntos
Colesterol/metabolismo , Receptores de Epoprostenol/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Humanos , Microscopia de Fluorescência , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
9.
Biochim Biophys Acta ; 1823(10): 1998-2012, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22884631

RESUMO

Prostacyclin and its I prostanoid receptor, the IP, play central roles in hemostasis and in re-endothelialization in response to vascular injury. Herein, intestinal and kidney enriched PDZ protein (IKEPP) was identified as an interactant of the human (h) IP mediated through binding of PDZ domain 1 (PDZ(D1)) and, to a lesser extent, PDZ(D2) of IKEPP to a carboxyl-terminal Class I 'PDZ ligand' within the hIP. While the interaction is constitutive, agonist-activation of the hIP leads to cAMP-dependent protein kinase (PK) A and PKC-phosphorylation of IKEPP, coinciding with its increased interaction with the hIP. Ectopic expression of IKEPP increases functional expression of the hIP, enhancing its ligand binding and agonist-induced cAMP generation. Originally thought to be restricted to renal and gastrointestinal tissues, herein, IKEPP was also found to be expressed in vascular endothelial cells where it co-localizes and complexes with the hIP. Furthermore, siRNA-disruption of IKEPP expression impaired hIP-induced endothelial cell migration and in vitro angiogenesis, revealing the functional importance of the IKEPP:IP interaction within the vascular endothelium. Identification of IKEPP as a functional interactant of the IP reveals novel mechanistic insights into the role of these proteins within the vasculature and, potentially, in other systems where they are co-expressed.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Receptores de Prostaglandina/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Moléculas de Adesão Celular , Movimento Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Rim/citologia , Ligantes , Modelos Biológicos , Proteínas de Neoplasias/química , Neovascularização Fisiológica , Fosfoproteínas/química , Ligação Proteica , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Receptores de Epoprostenol , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/química , Transdução de Sinais , Trocadores de Sódio-Hidrogênio/química
10.
Biochim Biophys Acta ; 1819(5): 428-45, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22381139

RESUMO

The prostanoid prostacyclin plays a central role in haemostasis and vascular repair. Recent studies investigating the regulation of the human prostacyclin receptor (hIP) gene identified an upstream repressor region (URR) within its regulatory promoter, herein termed the PrmIP. This study aimed to identify the main trans-acting factors that bind within the URR to transcriptionally repress PrmIP-directed gene expression in the megakaryoblastic human erythroleukemia (HEL) 92.1.7 cell line. Of the putative cis-acting elements examined, disruption of C/EBP and PU.1 elements within the URR substantially increased PrmIP-directed gene expression. Chromatin immunoprecipitation (ChIP) confirmed that C/EBPδ and PU.1, but not C/EBPß, bind to the URR in vivo, while ectopic expression of C/EBPδ substantially reduced hIP mRNA levels and PrmIP-directed gene expression. Phorbol 12-myristate 13-acetate (PMA)-induced megakaryocytic differentiation increased hIP mRNA and PrmIP-directed reporter gene expression and hIP-mediated cAMP generation in HEL cells. Two PMA-responsive regions, termed PRR1 and PRR2, were identified within PrmIP. Disruption of C/EBPδ and PU.1 cis-elements within the overlapping PRR1/URR and of Sp1, PU.1 and Oct-1 cis-elements within the overlapping PRR2/core PrmIP, revealed that both PRR1 and PRR2 contribute to the PMA- induction of hIP mRNA and gene expression in HEL cells. Furthermore, ChIP analysis established that induction of PrmIP-directed gene expression during megakaryocytic differentiation is largely regulated by PMA-induced dissociation of C/EBPδ and enhanced binding of PU.1 to PRR1 in addition to increased binding of Sp1, PU.1 and Oct-1 to elements within the core promoter/PRR2 in vivo. Taken together, these data provide critical insights into the transcriptional regulation of the hIP gene within the vasculature, including during megakaryocytic differentiation.


Assuntos
Leucemia Eritroblástica Aguda , Megacariócitos/metabolismo , Proteínas Proto-Oncogênicas , Receptores de Prostaglandina , Elementos de Resposta/genética , Transativadores , Doenças Cardiovasculares/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Megacariócitos/citologia , Megacariócitos/patologia , Nectinas , Fator 1 de Transcrição de Octâmero/genética , Fator 1 de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Epoprostenol , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Elementos de Resposta/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Transativadores/genética , Transativadores/metabolismo
11.
Mol Biol Cell ; 22(15): 2664-79, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21653824

RESUMO

Prostacyclin is increasingly implicated in re-endothelialization and angiogenesis but through largely unknown mechanisms. Herein the high-density lipoprotein (HDL) scavenger receptor class B, type 1 (SR-B1) adapter protein PDZ domain-containing protein 1 (PDZK1) was identified as an interactant of the human prostacyclin receptor (hIP) involving a Class I PDZ ligand at its carboxyl terminus and PDZ domains 1, 3, and 4 of PDZK1. Although the interaction is constitutive, it may be dynamically regulated following cicaprost activation of the hIP through a mechanism involving cAMP-dependent protein kinase (PK)A-phosphorylation of PDZK1 at Ser-505. Although PDZK1 did not increase overall levels of the hIP, it increased its functional expression at the cell surface, enhancing ligand binding and cicaprost-induced cAMP generation. Consistent with its role in re-endothelialization and angiogenesis, cicaprost activation of the hIP increased endothelial cell migration and tube formation/in vitro angiogenesis, effects completely abrogated by the specific IP antagonist RO1138452. Furthermore, similar to HDL/SR-B1, small interfering RNA (siRNA)-targeted disruption of PDZK1 abolished cicaprost-mediated endothelial responses but did not affect VEGF responses. Considering the essential role played by prostacyclin throughout the cardiovascular system, identification of PDZK1 as a functional interactant of the hIP sheds significant mechanistic insights into the protective roles of these key players, and potentially HDL/SR-B1, within the vascular endothelium.


Assuntos
Proteínas de Transporte/metabolismo , AMP Cíclico/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Lipoproteínas HDL/metabolismo , Receptores de Prostaglandina/metabolismo , Transdução de Sinais , Animais , Antineoplásicos/farmacologia , Compostos de Benzil/farmacologia , Proteínas de Transporte/genética , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Endoteliais/citologia , Endotélio Vascular/citologia , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Imidazóis/farmacologia , Proteínas de Membrana , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Domínios PDZ , Fosforilação , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Receptores de Epoprostenol , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/genética
12.
J Biol Chem ; 286(17): 15440-57, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21357687

RESUMO

In humans, thromboxane (TX) A(2) signals through the TPα and TPß isoforms of the TXA(2) receptor or TP. Here, the RhoA effector protein kinase C-related kinase (PRK) 1 was identified as an interactant of both TPα and ΤPß involving common and unique sequences within their respective C-terminal (C)-tail domains and the kinase domain of PRK1 (PRK1(640-942)). Although the interaction with PRK1 is constitutive, agonist activation of TPα/TPß did not regulate the complex per se but enhanced PRK1 activation leading to phosphorylation of its general substrate histone H1 in vitro. Altered PRK1 and TP expression and signaling are increasingly implicated in certain neoplasms, particularly in androgen-associated prostate carcinomas. Agonist activation of TPα/TPß led to phosphorylation of histone H3 at Thr(11) (H3 Thr(11)), a previously recognized specific marker of androgen-induced chromatin remodeling, in the prostate LNCaP and PC-3 cell lines but not in primary vascular smooth muscle or endothelial cells. Moreover, this effect was augmented by dihydrotestosterone in androgen-responsive LNCaP but not in nonresponsive PC-3 cells. Furthermore, PRK1 was confirmed to constitutively interact with TPα/TPß in both LNCaP and PC-3 cells, and targeted disruption of PRK1 impaired TPα/TPß-mediated H3 Thr(11) phosphorylation in, and cell migration of, both prostate cell types. Collectively, considering the role of TXA(2) as a potent mediator of RhoA signaling, the identification of PRK1 as a bona fide interactant of TPα/TPß, and leading to H3 Thr(11) phosphorylation to regulate cell migration, has broad functional significance such as within the vasculature and in neoplasms in which both PRK1 and the TPs are increasingly implicated.


Assuntos
Neoplasias da Próstata/patologia , Proteína Quinase C/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Histonas/metabolismo , Humanos , Masculino , Fosforilação , Neoplasias da Próstata/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo
13.
J Biol Chem ; 285(24): 18709-26, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20395296

RESUMO

The human prostacyclin receptor (hIP) undergoes agonist-induced internalization and subsequent recyclization in slowly recycling endosomes involving its direct physical interaction with Rab11a. Moreover, interaction with Rab11a localizes to a 22-residue putative Rab11 binding domain (RBD) within the carboxyl-terminal tail of the hIP, proximal to the transmembrane 7 (TM7) domain. Because the proposed RBD contains Cys(308) and Cys(311), in addition to Cys(309), that are known to undergo palmitoylation, we sought to identify the structure/function determinants of the RBD, including the influence of palmitoylation, on agonist-induced trafficking of the hIP. Through complementary approaches in yeast and mammalian cells along with computational structural studies, the RBD was localized to a 14-residue domain, between Val(299) and Leu(312), and proposed to be organized into an eighth alpha-helical domain (alpha-helix 8), comprising Val(299)-Val(307), adjacent to the palmitoylated residues at Cys(308)-Cys(311). From mutational and [(3)H]palmitate metabolic labeling studies, it is proposed that palmitoylation at Cys(311) in addition to agonist-regulated deacylation at Cys(309) > Cys(308) may dynamically position alpha-helix 8 in proximity to Rab11a, to regulate agonist-induced intracellular trafficking of the hIP. Moreover, Ala-scanning mutagenesis identified several hydrophobic residues within alpha-helix 8 as necessary for the interaction with Rab11a. Given the diverse membership of the G protein-coupled receptor superfamily, of which many members are also predicted to contain an alpha-helical 8 domain proximal to TM7 and, often, adjacent to palmitoylable cysteine(s), the identification of a functional role for alpha-helix 8, as exemplified as an RBD for the hIP, is likely to have broader significance for certain members of the superfamily.


Assuntos
Ácidos Palmíticos/química , Receptores de Epoprostenol/química , Proteínas rab de Ligação ao GTP/química , Biologia Computacional/métodos , Cisteína/química , Humanos , Leucina/química , Ácido Palmítico/química , Prostaglandinas/química , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico , Receptores Acoplados a Proteínas G/metabolismo , Valina/química
14.
J Mol Biol ; 396(3): 473-86, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20070947

RESUMO

Prostacyclin and the prostacyclin receptor (IP) are implicated in mediating many of the atheroprotective effects of estrogen in both humans and in animal models but through unknown mechanisms. Hence, herein the influence of estrogen on IP gene expression in endothelial EA.hy926, human erythroleukemia 92.1.7 and primary human aortic smooth muscle cells was investigated. Estrogen increased hIP mRNA levels, promoter (PrmIP)-directed reporter gene expression and cicaprost-dependent cAMP generation in all cell types, effects that were abrogated by actinomycin D and the general estrogen receptor (ER)-alpha/ERbeta antagonist ICI 182,780. Furthermore, the ERalpha-selective agonist 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT), but not the ERbeta-agonist 2,3-bis(4-hydroxyphenyl)-propionitrile, significantly increased hIP mRNA and PrmIP-directed gene expression. Deletional and mutational analysis of PrmIP uncovered an evolutionary conserved estrogen-response element, while electrophoretic mobility shift, antibody-supershift and chromatin immunoprecipitations assays confirmed the direct binding of ERalpha, but not ERbeta, to PrmIP both in vitro and in vivo. Moreover, immunofluorescence microscopy corroborated that estrogen and PPT increased hIP expression in primary human aortic smooth muscle cells. In conclusion, the hIP gene is directly regulated by estrogen that largely occurs through an ERalpha-dependent transcriptional mechanism and thereby provides critical insights into the role of prostacyclin/hIP in mediating the atheroprotective effects of estrogen within the human vasculature.


Assuntos
Vasos Sanguíneos/fisiologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Expressão Gênica , Receptores de Prostaglandina/biossíntese , Linhagem Celular , Imunoprecipitação da Cromatina , DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Células Endoteliais/fisiologia , Genes Reporter , Humanos , Microscopia de Fluorescência , Miócitos de Músculo Liso/fisiologia , Mutação Puntual , Ligação Proteica , Receptores de Epoprostenol , Elementos de Resposta , Deleção de Sequência
15.
J Mol Biol ; 394(1): 29-45, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19747485

RESUMO

Thromboxane plays an essential role in hemostasis, regulating platelet aggregation and vessel tone. In humans, it signals through the TPalpha and TPbeta isoforms that are transcriptionally regulated by distinct promoters Prm1 and Prm3, respectively. Herein, the consequence of megakaryocytic differentiation on Prm1-directed TPalpha expression was investigated. Phorbol 12-myristate 13-acetate (PMA) treatment substantially increased TPalpha mRNA and Prm1-directed gene expression in human erythroleukemia and K562 cells. Deletional analyses localized the major responsive element(s) to the upstream -8500 to -7504 region while mutation of four WT1/Egr1/Sp1 cis elements therein established that each contributes to the induction. Moreover, PMA increased Egr1, but not WT1 or Sp1, expression while the NGFI-A-binding protein 1 co-repressor impaired PMA induction of Egr1- and Prm1-directed gene expression. Chromatin immunoprecipitations established that WT1 is predominantly bound in vivo to the 5' Prm1 region in non-differentiated human erythroleukemia cells. In response to PMA, there was initial induction in Egr1 and associated reduction in WT1 binding to Prm1 in vivo, which was displaced by Sp1 following sustained treatment. Collectively, data establish that regulated WT1 followed by sequential Egr1 and Sp1 binding to elements within Prm1 mediate repression and subsequent induction of TPalpha during differentiation into the megakaryocytic phenotype, shedding significant insights into factors regulating TPalpha expression therein.


Assuntos
Diferenciação Celular , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Megacariócitos/citologia , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Fator de Transcrição Sp1/metabolismo , Proteínas WT1/metabolismo , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Análise Mutacional de DNA , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sequência Rica em GC , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células K562 , Luciferases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Modelos Genéticos , Dados de Sequência Molecular , Protaminas/genética , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Proteínas Repressoras/metabolismo , Deleção de Sequência , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo
16.
J Mol Biol ; 386(3): 579-97, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19118563

RESUMO

Prostacyclin plays a central role in hemostasis, inflammation and nociception. However, the factors regulating expression of the prostacyclin receptor (IP) gene in humans and in other species have not been identified. In this study, we sought to identify the key trans-acting factors and cis-acting elements regulating IP expression in the megakaryoblastic human erythroleukemia (HEL) 92.1.7 and vascular endothelial EA.hy 926 cell lines. With the use of deletion and genetic reporter analyses, the essential core promoter, termed PrmIP, was localized to the -1022 to -895 region proximal to the transcription initiation site, while an upstream repressor region, localized to -1502 to -1271, was also identified. Bioinformatic analysis revealed evolutionarily conserved Sp1, PU.1 and Oct-1 sites within the core PrmIP, and disruption of these elements each led to substantial reductions in PrmIP-directed gene expression in both HEL and EA.hy 926 cells. Electrophoretic mobility shift and supershift assays established that Sp1, PU.1 and Oct-1 can bind to elements within the core promoter in vitro, while chromatin immunoprecipitation assays confirmed their specific binding to chromatin in vivo. Furthermore, combination mutations of the Sp1, PU.1 and Oct-1 elements revealed that they act independently to co-regulate basal transcription of the IP gene, while ectopic expression of each of the trans-acting factors led to substantial increases in PrmIP-directed gene expression and IP mRNA expression in both HEL and EA.hy 926 cells. While electrophoretic mobility shift and antibody supershift assays established that the Ets family member Fli1, but not Ets-1, is capable of binding to the PU.1 element within PrmIP in vitro, chromatin immunoprecipitation analysis established that neither Fli1 nor Ets-1 binds to that element in vivo. Collectively, these data provide critical insights into the transcriptional regulation of the IP gene in human megakaryocytic and endothelial cells, identifying Sp1, PU.1 and Oct-1 as the critical factors involved in its basal regulation in humans.


Assuntos
Células Endoteliais/fisiologia , Regulação da Expressão Gênica , Megacariócitos/fisiologia , Fator 1 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Epoprostenol/biossíntese , Fator de Transcrição Sp1/metabolismo , Transativadores/metabolismo , Sítios de Ligação , Linhagem Celular , Imunoprecipitação da Cromatina , DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Genes Reporter , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Receptores de Epoprostenol/genética , Deleção de Sequência
17.
Mol Microbiol ; 67(1): 171-87, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18034794

RESUMO

Phase-variable expression of type 1 fimbriae in Escherichia coli K-12 involves inversion by site-specific recombination of a 314 bp sequence containing the promoter for fim structural gene expression. The invertible sequence is flanked by 9 bp inverted repeats, and each repeat is in turn flanked by non-identical recombinase-binding elements (RBEs) to which the FimB or FimE site-specific recombinases bind. These proteins have distinct DNA inversion preferences: FimB inverts the switch in the ON-to-OFF and OFF-to-ON directions with similar efficiencies, whereas FimE inverts it predominantly in the ON-to-OFF direction. We have found that FimB and FimE invert the switch through a common mechanism. A genetic investigation involving base-by-base substitution combined with a biochemical study shows that the same DNA cleavage and religation sites are used within the 9 bp inverted repeats, and that each recombination involves a common 3 bp spacer region. A comprehensive programme of RBE exchanges and replacements reveals that FimB is much more tolerant of RBE sequence variation than FimE. The asymmetric location of conserved 5'-CA motifs at either side of each spacer region allows the inside and outside of the switch to be differentiated while the RBE sequence heterogeneity permits its ON and OFF forms to be distinguished by the recombinases.


Assuntos
Proteínas de Ligação a DNA/química , Escherichia coli K12/genética , Proteínas de Escherichia coli/química , Integrases/química , Recombinases/química , Tirosina/metabolismo , Sequência de Bases , Inversão Cromossômica , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli K12/enzimologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Integrases/genética , Integrases/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Nucleotídeos/química , Plasmídeos/genética , Recombinases/genética , Recombinases/metabolismo , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico , Especificidade por Substrato
18.
J Bacteriol ; 189(9): 3403-13, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17307842

RESUMO

The VirB protein of Shigella flexneri is a positive regulator of the major virulence operons of this enteroinvasive intracellular pathogen. VirB resembles no other transcription factor but is strongly homologous to plasmid partition proteins. We found that the binding of the VirB protein to the promoter region of the icsB virulence gene induced hypersensitivity to cleavage by DNase I over a region to which the H-NS repressor protein binds and completely abolished the protection of this sequence from DNase I by H-NS. In the absence of H-NS, the VirB protein had no additive effect on the ability of the icsB promoter to form an open transcription complex, indicating that VirB is not involved in the recruitment of RNA polymerase to the promoter or in open complex formation. Similarly, VirB did not stimulate promoter function in an in vitro transcription assay but acted as an antagonist of H-NS-mediated repression. A sequence located upstream of the icsB promoter and related to cis-acting elements involved in plasmid partitioning was required for promoter derepression by VirB. Alterations to one heptameric motif within this DNA sequence attenuated VirB binding and derepression of icsB transcription.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Regulação Bacteriana da Expressão Gênica , Shigella flexneri/fisiologia , Fatores de Transcrição/fisiologia , Sítios de Ligação/genética , Pegada de DNA , DNA Bacteriano/metabolismo , Desoxirribonuclease I/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Regiões Promotoras Genéticas , Ligação Proteica , Shigella flexneri/genética , Shigella flexneri/patogenicidade , Transcrição Gênica
19.
Chem Biol ; 11(1): 69-77, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15112996

RESUMO

Reversible control of the conformation of proteins was employed to probe the relationship between flexibility and specificity of the basic helix-loop-helix protein MyoD. A fusion protein (apaMyoD) was designed where the basic DNA binding helix of MyoD was stablized by an amino-terminal extension with a sequence derived from the bee venom peptide apamin. The disulfide-stabilized helix from apamin served as a nucleus for a helix that extended for a further ten residues, thereby holding apaMyoD's DNA recognition helix in a predominantly alpha-helical conformation. The thermal stability of the DNA complexes of apaMyoD was increased by 13 degrees C relative to MyoD-bHLH. Measurements of the fluorescence anisotropy change on DNA binding indicated that apaMyoD bound to E-box-containing DNA sequences with enhanced affinity relative to MyoD-bHLH. Consequently, the DNA binding specificity of apaMyoD was increased 10-fold.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Anisotropia , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Sítios de Ligação , Dicroísmo Circular , DNA/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Espectroscopia de Ressonância Magnética , Microscopia de Fluorescência/instrumentação , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato , Temperatura , Fatores de Transcrição/química , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...