Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 123(2): 903-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23321668

RESUMO

HIV infection results in gastrointestinal (GI) tract damage, microbial translocation, and immune activation, which are not completely ameliorated with suppression of viremia by antiretroviral (ARV) therapy. Furthermore, increased morbidity and mortality of ARV-treated HIV-infected individuals is associated with these dysfunctions. Thus, to enhance GI tract physiology, we treated SIV-infected pigtail macaques with ARVs, probiotics, and prebiotics or with ARVs alone. This synbiotic treatment resulted in increased frequency and functionality of GI tract APCs, enhanced reconstitution and functionality of CD4+ T cells, and reduced fibrosis of lymphoid follicles in the colon. Thus, ARV synbiotic supplementation in HIV-infected individuals may improve GI tract immunity and thereby mitigate inflammatory sequelae, ultimately improving prognosis.


Assuntos
Antirretrovirais/administração & dosagem , Trato Gastrointestinal/imunologia , Prebióticos , Probióticos/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Adjuvantes Imunológicos/administração & dosagem , Animais , Linfócitos T CD4-Positivos/imunologia , Terapia Combinada , Suplementos Nutricionais , Modelos Animais de Doenças , Infecções por HIV/imunologia , Infecções por HIV/terapia , Humanos , Macaca nemestrina , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Transcriptoma
2.
J Vasc Surg ; 54(4): 1117-1123.e1, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21803524

RESUMO

BACKGROUND: Thrombosis is a major cause of the early failure of vein grafts (VGs) implanted during peripheral and coronary arterial bypass surgeries. Endothelial expression of thrombomodulin (TM), a key constituent of the protein C anticoagulant pathway, is markedly suppressed in VGs after implantation and contributes to local thrombus formation. While stretch-induced paracrine release of transforming growth factor-ß (TGF-ß) is known to negatively regulate TM expression in heart tissue, its role in regulating TM expression in VGs remains unknown. METHODS: Changes in relative mRNA expression of major TGF-ß isoforms were measured by quantitative polymerase chain reaction (qPCR) in cultured human saphenous vein smooth muscle cells (HSVSMCs) subjected to cyclic stretch. To determine the effects of paracrine release of TGF-ß on endothelial TM mRNA expression, human saphenous vein endothelial cells (HSVECs) were co-cultured with stretched HSVSMCs in the presence of 1D11, a pan-neutralizing TGF-ß antibody, or 13C4, an isotype-control antibody. Groups of rabbits were then administered 1D11 or 13C4 and underwent interpositional grafting of jugular vein segments into the carotid circulation. The effect of TGF-ß inhibition on TM gene expression was measured by qPCR; protein C activating capacity and local thrombus formation were measured by in situ chromogenic substrate assays; and VG remodeling was assessed by digital morphometry. RESULTS: Cyclic stretch induced TGF-ß(1) expression in HSVSMCs by 1.9 ± 0.2-fold (P < .001) without significant change in the expressions of TGF-ß(2) and TGF-ß(3). Paracrine release of TGF-ß(1) by stretched HSVSMCs inhibited TM expression in stationary HSVECs placed in co-culture by 57 ± 12% (P = .03), an effect that was abolished in the presence of 1D11. Similarly, TGF-ß(1) was the predominant isoform induced in rabbit VGs 7 days after implantation (3.5 ± 0.4-fold induction; P < .001). TGF-ß(1) protein expression localized predominantly to the developing neointima and coincided with marked suppression of endothelial TM expression (16% ± 2% of vein controls; P < .03), a reduction in situ activated protein C (APC)-generating capacity (53% ± 9% of vein controls; P = .001) and increased local thrombus formation (3.7 ± 0.8-fold increase over vein controls; P < .01). External stenting of VGs to limit vessel distension significantly reduced TGF-ß(1) induction and TM downregulation. Systemic administration of 1D11 also effectively prevented TM downregulation, preserved APC-generating capacity, and reduced local thrombus in rabbit VGs without observable effect on neointima formation and other morphometric parameters 6 weeks after implantation. CONCLUSION: TM downregulation in VGs is mediated by paracrine release of TGF-ß(1) caused by pressure-induced vessel stretch. Systemic administration of an anti-TGF-ß antibody effectively prevented TM downregulation and preserved local thromboresistance without negative effect on VG remodeling.


Assuntos
Anticorpos Neutralizantes/farmacologia , Células Endoteliais/efeitos dos fármacos , Oclusão de Enxerto Vascular/prevenção & controle , Veias Jugulares/efeitos dos fármacos , Mecanotransdução Celular/efeitos dos fármacos , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Trombose Venosa/prevenção & controle , Animais , Artérias Carótidas/cirurgia , Células Cultivadas , Células Endoteliais/metabolismo , Oclusão de Enxerto Vascular/sangue , Oclusão de Enxerto Vascular/metabolismo , Humanos , Veias Jugulares/metabolismo , Veias Jugulares/transplante , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Proteína C/metabolismo , RNA Mensageiro/metabolismo , Coelhos , Veia Safena/efeitos dos fármacos , Veia Safena/metabolismo , Estresse Mecânico , Trombomodulina/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Trombose Venosa/sangue , Trombose Venosa/metabolismo
3.
Hum Mol Genet ; 19(8): 1453-60, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20089530

RESUMO

Two common inflammatory skin disorders with impaired barrier, atopic dermatitis (AD) and psoriasis, share distinct genetic linkage to the Epidermal Differentiation Complex (EDC) locus on 1q21. The EDC is comprised of tandemly arrayed gene families encoding proteins involved in skin cell differentiation. Discovery of semi-dominant mutations in filaggrin (FLG) associated with AD and a copy number variation within the LCE genes associated with psoriasis provide compelling evidence for the role of EDC genes in the pathogenesis of these diseases. To date, little is known about the potentially complex regulatory landscape within the EDC. Here, we report a computational approach to identify conserved non-coding elements (CNEs) in the EDC queried for regulatory function. Coordinate expression of EDC genes during mouse embryonic skin development and a striking degree of synteny and linearity in the EDC locus across a wide range of mammalian (placental and marsupial) genomes suggests an evolutionary conserved regulatory milieu in the EDC. CNEs identified by comparative genomics exhibit dynamic regulatory activity (enhancer or repressor) in differentiating or proliferating conditions. We further demonstrate epidermal-specific, developmental in vivo enhancer activities (DNaseI and transgenic mouse assays) in CNEs, including one within the psoriasis-associated deletion, LCE3C_LCE3B-del. Together, our multidisciplinary study features a network of regulatory elements coordinating developmental EDC gene expression as an unexplored resource for genetic variants in skin diseases.


Assuntos
Dermatite Atópica/genética , Loci Gênicos , Psoríase/genética , Elementos Reguladores de Transcrição , Animais , Dermatite Atópica/embriologia , Epiderme/embriologia , Proteínas Filagrinas , Humanos , Camundongos , Psoríase/embriologia , Pele/embriologia
4.
Arterioscler Thromb Vasc Biol ; 29(10): 1587-93, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19661484

RESUMO

OBJECTIVE: Impairment of the thrombomodulin-protein C anticoagulant pathway has been implicated in pathological thrombosis associated with malignancy. Patients who receive proteasome inhibitors as part of their chemotherapeutic regimen appear to be at decreased risk for thromboembolic events. We investigated the effects of proteasome inhibitors on endothelial thrombomodulin expression and function. METHODS AND RESULTS: Proteasome inhibitors as a class markedly induced the expression of thrombomodulin and enhanced the protein C activating capacity of endothelial cells. Thrombomodulin upregulation was independent of NF-kappaB signaling, a principal target of proteasome inhibitors, but was instead a direct consequence of increased expression of the Krüppel-like transcription factors, KLF2 and KLF4. These effects were confirmed in vivo, where systemic administration of a proteasome inhibitor enhanced thrombomodulin expression that was paralleled by changes in the expression of KLF2 and KLF4. CONCLUSIONS: These findings identify a novel mechanism of action of proteasome inhibitors that may help to explain their clinically observed thromboprotective effects.


Assuntos
Ácidos Borônicos/farmacologia , Células Endoteliais/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/fisiologia , Inibidores de Proteases/farmacologia , Inibidores de Proteassoma , Pirazinas/farmacologia , Trombomodulina/genética , Animais , Bortezomib , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , NF-kappa B/antagonistas & inibidores , Proteína C/fisiologia , Trombomodulina/fisiologia
5.
Science ; 324(5931): 1190-2, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19478181

RESUMO

Human skin is a large, heterogeneous organ that protects the body from pathogens while sustaining microorganisms that influence human health and disease. Our analysis of 16S ribosomal RNA gene sequences obtained from 20 distinct skin sites of healthy humans revealed that physiologically comparable sites harbor similar bacterial communities. The complexity and stability of the microbial community are dependent on the specific characteristics of the skin site. This topographical and temporal survey provides a baseline for studies that examine the role of bacterial communities in disease states and the microbial interdependencies required to maintain healthy skin.


Assuntos
Bactérias/isolamento & purificação , Metagenoma , Pele/microbiologia , Actinobacteria/classificação , Actinobacteria/genética , Actinobacteria/isolamento & purificação , Adulto , Bactérias/classificação , Bactérias/genética , Bacteroidetes/classificação , Bacteroidetes/genética , Bacteroidetes/isolamento & purificação , Biodiversidade , Feminino , Genes de RNAr , Humanos , Masculino , Dados de Sequência Molecular , Filogenia , Proteobactérias/classificação , Proteobactérias/genética , Proteobactérias/isolamento & purificação , RNA Ribossômico 16S , Fatores de Tempo , Adulto Jovem
6.
Circulation ; 115(1): 67-75, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17190863

RESUMO

BACKGROUND: Patients with heart failure are at increased risk for thromboembolic events, including stroke. Historically attributed to blood stasis, little is known about the adverse effects of elevated chamber filling pressure on endocardial function, which could predispose to intracardiac thrombus formation. METHODS AND RESULTS: We investigated changes in the expression of thrombomodulin, a key component of the anticoagulant protein C pathway, in rats subjected to acute atrial pressure overload caused by aortic banding. Acute elevation of left atrial filling pressure, without an associated decline in ventricular systolic function, caused a 70% inhibition of atrial endocardial thrombomodulin expression and resulted in increased local thrombin generation. Targeted restoration of atrial thrombomodulin expression with adenovirus-mediated gene transfer successfully reduced thrombin generation to baseline levels. In vitro co-culture studies revealed that thrombomodulin downregulation is caused by the paracrine release of transforming growth factor-beta from cardiac connective tissue in response to mechanical stretch. This was confirmed in vivo by administration of a neutralizing transforming growth factor-beta antibody, which effectively prevented thrombomodulin downregulation during acute pressure overload. CONCLUSIONS: These findings suggest that increased hemodynamic load adversely affects endocardial function and is a potentially important contributor to thromboembolus formation in heart failure.


Assuntos
Pressão Sanguínea/fisiologia , Endocárdio/metabolismo , Frequência Cardíaca/fisiologia , Trombomodulina/biossíntese , Animais , Modelos Animais de Doenças , Insuficiência Cardíaca/metabolismo , Masculino , Contração Miocárdica/fisiologia , Ratos , Ratos Wistar
7.
Blood ; 105(10): 3910-7, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15677570

RESUMO

Inflammation and thrombosis are increasingly recognized as interrelated biologic processes. Endothelial cell expression of thrombomodulin (TM), a key component of the anticoagulant protein C pathway, is potently inhibited by inflammatory cytokines. Because the mechanism underlying this effect is largely unknown, we investigated a potential role for the inflammatory transcription factor nuclear factor-kappa B (NF-kappaB). Blocking NF-kappaB activation effectively prevented cytokine-induced down-regulation of TM, both in vitro and in a mouse model of tumor necrosis factor-alpha (TNF-alpha)-mediated lung injury. Although the TM promoter lacks a classic NF-kappaB consensus site, it does contain tandem Ets transcription factor binding sites previously shown to be important for both constitutive TM gene expression and cytokine-induced repression. Using electrophoretic mobility shift assay and chromatin immunoprecipitation, we found that multiple Ets species bind to the TNF-alpha response element within the TM promoter. Although cytokine exposure did not alter Ets factor binding, it did reduce binding of p300, a coactivator required by Ets for full transcriptional activity. Overexpression of p300 also prevented TM repression by cytokines. We conclude that NF-kappaB is a critical mediator of TM repression by cytokines. Further evidence suggests a mechanism involving competition by NF-kappaB for limited pools of the transcriptional coactivator p300 necessary for TM gene expression.


Assuntos
Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Mediadores da Inflamação/farmacologia , Interleucina-1/farmacologia , NF-kappa B/metabolismo , Trombomodulina/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Proteína p300 Associada a E1A , Células Endoteliais/metabolismo , Humanos , Masculino , Camundongos , NF-kappa B/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Trombomodulina/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
8.
FASEB J ; 17(15): 2345-6, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14525937

RESUMO

Nitric oxide (NO) is an antiviral effector of the innate immune system, but few of the viral targets of NO have been identified. We now show that NO inhibits adenovirus replication by targeting the adenovirus proteinase (AVP). NO generated from diethylamine NONOate (DEA-NONOate) or spermine NONOate (Sp-NONOate) inhibited the AVP. Inhibition was reversible with dithiothreitol. The equilibrium dissociation constant for reversible binding to the AVP by Sp-NONOate, or Ki, was 0.47 mM, and the first-order rate constant for irreversible inhibition of the AVP by Sp-NONOate, or ki, was 0.0036 s(-1). Two hallmarks of a successful adenovirus infection were abolished by the NO donors: the appearance of E1A protein and the cleavage of cytokeratin 18 by AVP. Treatment of infectious virus by DEA-NONOate dramatically decreased viral infectivity. These data suggest that NO may be a useful antiviral agent against viruses encoding a cysteine proteinase and in particular may be an antiadenovirus agent.


Assuntos
Adenoviridae/efeitos dos fármacos , Adenoviridae/enzimologia , Antivirais/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Doadores de Óxido Nítrico/farmacologia , Infecções por Adenoviridae/tratamento farmacológico , Antivirais/uso terapêutico , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/uso terapêutico , Ditiotreitol/farmacologia , Células HeLa , Humanos , Hidrazinas/farmacologia , Modelos Biológicos , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/uso terapêutico , Óxidos de Nitrogênio , Fragmentos de Peptídeos/metabolismo , Proteínas Virais/metabolismo
9.
Circ Res ; 92(1): 41-7, 2003 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-12522119

RESUMO

Thrombomodulin (TM), a key component of the anticoagulant protein C pathway, is a major contributor to vascular thromboresistance. We previously found that TM protein expression is dramatically reduced in autologous vein grafts during the first two weeks after implantation, coincident to a local inflammatory response, and remains suppressed for at least 6 weeks. To determine the proximate cause of TM loss, in vivo gene expression was quantified by real-time PCR. TM gene expression in vein grafts declined >85% during the first postoperative week and remained suppressed >55% at 6 weeks, accounting for the observed changes in protein expression. The effects of vein graft inflammation were evaluated in animals rendered leukopenic with vinblastine before graft implantation. Abrogating the local inflammatory response affected neither TM protein nor gene expression. To determine how hemodynamic forces might modulate TM expression, the surgical protocol was modified to alter blood flow and pressure-induced vessel distension. TM protein and gene expression did not correlate to changes in shear stress but highly correlated to changes in wall tension, both acutely and over time. We conclude that the primary stimulus for altered TM expression in vein grafts is the exposure to arterial pressure. Furthermore, these data identify strain as a novel and important pathway for in vivo TM gene regulation.


Assuntos
Pressão Sanguínea/fisiologia , Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica , Trombomodulina/genética , Trombomodulina/metabolismo , Animais , Artérias Carótidas/fisiologia , Artérias Carótidas/cirurgia , Progressão da Doença , Oclusão de Enxerto Vascular/patologia , Oclusão de Enxerto Vascular/fisiopatologia , Granulócitos/patologia , Hemodinâmica/fisiologia , Veias Jugulares/transplante , Leucopenia/induzido quimicamente , Leucopenia/fisiopatologia , Masculino , Modelos Animais , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Mecânico , Vasculite/patologia , Vasculite/fisiopatologia , Vimblastina
10.
DNA Seq ; 14(6): 399-405, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15018348

RESUMO

Thrombomodulin (TM), a component of the protein C anticoagulant pathway, is critical for the maintenance of vascular thromboresistance. To facilitate the study of in vivo TM regulation, we cloned and sequenced the cDNA encoding full-length rabbit TM. Translation of the open reading frame predicts a 580 amino acid protein that contains a 19 amino acid signal peptide, one lectin-like and six EGF-like extracellular domains, a 23 amino acid transmembrane domain and a 36 amino acid cytoplasmic domain. In addition, there are three potential N-linked and six O-linked glycosylation sites. Comparison of the predicted rabbit TM protein with those of human, mouse and rat reveals 67-72% primary sequence conservation with identical domain homology. TM gene expression was quantified in rabbit cardiovascular tissue by real-time PCR using primers and probe based on the derived cDNA sequence and found to correlate with protein expression as determined by Western blot analysis.


Assuntos
Clonagem Molecular , Miocárdio/metabolismo , Trombomodulina/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , DNA Complementar/genética , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Veias Jugulares/transplante , Dados de Sequência Molecular , Coelhos , Ratos , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Trombomodulina/metabolismo , Fatores de Tempo
11.
Circ Res ; 90(2): 205-12, 2002 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-11834714

RESUMO

Thrombosis is the major cause of early vein graft failure. Our aim was to determine whether alterations in the expression of the anticoagulant proteins, thrombomodulin (TM) and the endothelial cell protein C receptor (EPCR), impair endothelial thromboresistance that may contribute to vein graft failure. Immunohistochemical staining of autologous rabbit vein graft sections revealed that the expression of TM, but not EPCR, was reduced significantly early after graft implantation. Western blot analysis revealed that TM expression was reduced by >95% during the first 2 weeks after implantation, with gradual but incomplete recovery by 42 days. This resulted in up to a 95% reduction in the capacity of the grafts to activate protein C and was associated with an increase in bound thrombin activity, which peaked on day 7 at 28.7 +/- 3.8 mU/cm(2) and remained elevated for more than 14 days. Restoration of TM expression using adenovirus vector-mediated gene transfer significantly enhanced the capacity of grafts to activate protein C and reduced bound thrombin activity on day 7 to levels comparable to that of normal veins (5.7 +/- 0.4 versus 5.2 +/- 1.1 mU/cm(2), respectively, P=0.74). Surprisingly, neointima formation was not affected by this inhibition of local thrombin activity. These data suggest that the early loss of TM expression significantly impairs vein graft thromboresistance and results in enhanced local thrombin generation. Although enhanced local thrombin generation may predispose to early vein graft failure due to thrombosis, it does not seem to contribute significantly to late vein graft failure due to neointimal hyperplasia.


Assuntos
Fatores de Coagulação Sanguínea , Veias Jugulares/metabolismo , Receptores de Superfície Celular/metabolismo , Trombomodulina/metabolismo , Trombose Venosa/metabolismo , Animais , Western Blotting , Artérias Carótidas/cirurgia , Modelos Animais de Doenças , Fibrinolíticos/farmacologia , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Imuno-Histoquímica , Veias Jugulares/efeitos dos fármacos , Veias Jugulares/patologia , Veias Jugulares/transplante , Masculino , Proteína C/metabolismo , Coelhos , Trombina/antagonistas & inibidores , Trombina/metabolismo , Trombomodulina/deficiência , Trombomodulina/genética , Transdução Genética , Transplante Autólogo/efeitos adversos , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/patologia , Procedimentos Cirúrgicos Vasculares/efeitos adversos , Trombose Venosa/etiologia , Trombose Venosa/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...