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
2.
J Heart Lung Transplant ; 36(5): 509-519, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28190787

RESUMO

BACKGROUND: Detailed hemodynamic data from the phase III PATENT-1 study of riociguat in patients with pulmonary arterial hypertension (PAH) were investigated. METHODS: Patients with PAH who were treatment naïve or pre-treated with endothelin receptor antagonists or non-intravenous prostanoids were randomly assigned to riociguat up to 2.5 mg 3 times a day or placebo. Hemodynamic parameters were assessed at baseline and week 12. RESULTS: Riociguat significantly decreased pulmonary vascular resistance in treatment-naïve (n = 221; least squares [LS] mean difference -266 dyne∙sec∙cm-5 [95% confidence interval (CI) -357 to -175; p < 0.0001]) and pre-treated (n = 222; LS mean difference -186 dyne∙sec ∙cm-5 [95% CI -252 to -120; p < 0.0001]) patients and significantly increased cardiac index (LS mean difference +0.7 [95% CI 0.5 to 0.8] and +0.5 [95% CI 0.3 to 0.7], respectively [both p < 0.0001]). Mean pulmonary artery pressure (p = 0.0056 and p = 0.0019 for treatment-naïve and pre-treated patients, respectively), mean arterial pressure (both p < 0.0001), and systemic vascular resistance (both p < 0.0001) were significantly reduced, and there was an increase in mixed venous oxygen saturation (p < 0.0001 and p = 0.0004, respectively). Results were similar in patients pre-treated with endothelin receptor antagonists and patients pre-treated with non-intravenous prostanoids. Improvements in 6-minute walking distance correlated very weakly with improvements in pulmonary vascular resistance (r = -0.21 [95% CI -0.30 to -0.11; p < 0.0001]) and cardiac index (r = 0.16 [95% CI 0.06 to 0.25; p < 0.0016]). CONCLUSIONS: Riociguat significantly improved hemodynamic parameters in pre-treated and treatment-naïve patients with PAH.


Assuntos
Hemodinâmica/efeitos dos fármacos , Hipertensão Pulmonar/tratamento farmacológico , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Resistência Vascular/efeitos dos fármacos , Administração Oral , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Antagonistas dos Receptores de Endotelina/administração & dosagem , Feminino , Humanos , Hipertensão Pulmonar/diagnóstico , Internacionalidade , Masculino , Dose Máxima Tolerável , Estudos Prospectivos , Prostaglandinas/administração & dosagem , Pirazóis/efeitos adversos , Pirimidinas/efeitos adversos , Valores de Referência , Índice de Gravidade de Doença , Resultado do Tratamento
3.
Heart ; 103(8): 599-606, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28011757

RESUMO

OBJECTIVE: We compared the haemodynamic effects of riociguat in patients with inoperable chronic thromboembolic pulmonary hypertension (CTEPH) or persistent/recurrent CTEPH after pulmonary endarterectomy in the Chronic Thromboembolic Pulmonary Hypertension Soluble Guanylate Cyclase-Stimulator Trial 1 study. METHODS: Patients with inoperable or persistent/recurrent CTEPH (n=261; mean± SD age 59±14 years; 66% women) were randomised to riociguat (up to 2.5 mg three times daily) or placebo. Haemodynamic parameters were assessed at baseline and week 16. RESULTS: Riociguat decreased pulmonary vascular resistance (PVR) in inoperable (n=189; least-squares mean difference: -285 dyn s/cm5 (95% CI -357 to -213); p<0.0001) and persistent/recurrent (n=72; -131 dyn s/cm5 (95% CI -214 to -48); p=0.0025) patients. Cardiac index improved in inoperable patients by a least-squares mean difference of +0.6 L/min/m2 (95% CI 0.4 to 0.7; p<0.0001), while in persistent/recurrent patients the change was +0.2 L/min/m2 (95% CI -0.1 to 0.5; p=0.17). Mean pulmonary artery pressure decreased in inoperable and persistent/recurrent patients(-4.7 mm Hg (95% CI -6.9 to -2.6; p<0.0001 and -4.8 mm Hg (-8.2 to -1.5; p=0.0055), respectively). For all patients, changes in 6 min walk distance correlated with changes in PVR (r=-0.29 (95% CI -0.41 to -0.17); p<0.0001) and cardiac index (r=0.23 (95% CI 0.10 to 0.35); p=0.0004). CONCLUSIONS: Riociguat improved haemodynamics in patients with inoperable CTEPH or persistent/recurrent CTEPH. TRIAL REGISTRATION NUMBER: NCT00855465.


Assuntos
Anti-Hipertensivos/uso terapêutico , Pressão Arterial/efeitos dos fármacos , Hipertensão Pulmonar/tratamento farmacológico , Artéria Pulmonar/efeitos dos fármacos , Embolia Pulmonar/complicações , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Resistência Vascular/efeitos dos fármacos , Idoso , Anti-Hipertensivos/efeitos adversos , Doença Crônica , Método Duplo-Cego , Endarterectomia , Tolerância ao Exercício/efeitos dos fármacos , Feminino , Humanos , Hipertensão Pulmonar/diagnóstico , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Análise dos Mínimos Quadrados , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Artéria Pulmonar/fisiopatologia , Embolia Pulmonar/diagnóstico , Pirazóis/efeitos adversos , Pirimidinas/efeitos adversos , Recuperação de Função Fisiológica , Recidiva , Fatores de Tempo , Resultado do Tratamento , Teste de Caminhada
4.
J Heart Lung Transplant ; 34(3): 348-55, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25813765

RESUMO

BACKGROUND: In the Chronic Thromboembolic Pulmonary Hypertension Soluble Guanylate Cyclase - Stimulator Trial 1 (CHEST-1) study, riociguat improved 6-minute walking distance (6MWD) vs placebo in patients with inoperable chronic thromboembolic pulmonary hypertension or persistent/recurrent pulmonary hypertension after pulmonary endarterectomy. In this study, the proportion of patients who achieved responder thresholds that correlate with improved outcome in patients with pulmonary arterial hypertension was determined at baseline and at the end of CHEST-1. METHODS: Patients received placebo or riociguat individually adjusted up to 2.5 mg 3 times a day for 16 weeks. Response criteria were defined as follows: 6MWD increase ≥40 m, 6MWD ≥380 m, cardiac index ≥2.5 liters/min/m(2), pulmonary vascular resistance <500 dyn∙sec∙cm(-5), mixed venous oxygen saturation ≥65%, World Health Organization functional class I/II, N-terminal pro-brain natriuretic peptide <1,800 pg/ml, and right atrial pressure <8 mm Hg. RESULTS: Riociguat increased the proportion of patients with 6MWD ≥380 m, World Health Organization functional class I/II, and pulmonary vascular resistance <500 dyn∙sec∙cm(-5) from 37%, 34%, and 25% at baseline to 58%, 57%, and 50% at Week 16, whereas there was little change in placebo-treated patients (6MWD ≥380 m, 43% vs 44%; World Health Organization functional class I/II, 29% vs 38%; pulmonary vascular resistance <500 dyn∙sec∙cm(-5), 27% vs 26%). Similar changes were observed for thresholds for cardiac index, mixed venous oxygen saturation, N-terminal pro-brain natriuretic peptide, and right atrial pressure. CONCLUSIONS: In this exploratory analysis, riociguat increased the proportion of patients with inoperable chronic thromboembolic pulmonary hypertension or persistent/recurrent pulmonary hypertension after pulmonary endarterectomy achieving criteria defining a positive response to therapy.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Função Ventricular Direita/fisiologia , Pressão Ventricular/efeitos dos fármacos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Hipertensão Pulmonar/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento , Resistência Vascular , Função Ventricular Direita/efeitos dos fármacos , Pressão Ventricular/fisiologia , Adulto Jovem
5.
J Heart Lung Transplant ; 34(3): 338-47, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25703961

RESUMO

BACKGROUND: In PATENT-1, riociguat significantly improved 6-minute walking distance (6MWD) and a range of secondary end-points in patients with pulmonary arterial hypertension (PAH). We investigated whether riociguat increased the proportion of patients achieving clinically relevant responder thresholds compared with placebo during PATENT-1. METHODS: In PATENT-1, a randomized, double-blind study, treatment-naïve patients or patients on background PAH-targeted therapy with symptomatic PAH received 12 weeks of treatment with placebo, riociguat up to 2.5 mg 3 times daily, or riociguat up to 1.5 mg 3 times daily. Increases in 6MWD ≥40 m, 6MWD ≥380 m, cardiac index ≥2.5 liter/min/m(2), mixed venous oxygen saturation ≥65%, World Health Organization functional class I/II, N-terminal pro-brain natriuretic peptide <1,800 pg/ml, and right atrial pressure <8 mm Hg were chosen as threshold criteria of a positive response. RESULTS: Riociguat increased the proportion of treatment-naïve patients and patients on background PAH-targeted therapy with 6MWD ≥380 m at Week 12 (+21% and +15%, respectively), whereas there was a small reduction in 6MWD in placebo-treated patients for both sub-groups. Riociguat also increased the proportion of treatment-naïve patients and patients on background PAH-targeted therapy achieving World Health Organization functional class I/II (+12% and +19%, respectively) and cardiac index ≥2.5 liter/min/m(2) (+30% and +33%, respectively) at Week 12, whereas there was little change in the respective placebo groups. CONCLUSIONS: Compared with placebo, riociguat increased the proportion of treatment-naïve patients and patients on background PAH-targeted therapy who fulfilled criteria defining a positive response to therapy.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Função Ventricular Direita/fisiologia , Pressão Ventricular/efeitos dos fármacos , Caminhada/fisiologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Teste de Esforço/efeitos dos fármacos , Feminino , Seguimentos , Humanos , Hipertensão Pulmonar/fisiopatologia , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Função Ventricular Direita/efeitos dos fármacos
6.
Eur Respir J ; 45(5): 1303-13, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25614164

RESUMO

Riociguat is a soluble, guanylate cyclase stimulator, approved for pulmonary arterial hypertension. In the 12-week PATENT-1 study, riociguat was well tolerated and improved several clinically relevant end-points in patients with pulmonary arterial hypertension who were treatment naïve or had been pretreated with endothelin-receptor antagonists or prostanoids. The PATENT-2 open-label extension evaluated the long-term safety and efficacy of riociguat. Eligible patients from the PATENT-1 study received riociguat individually adjusted up to a maximum dose of 2.5 mg three times daily. The primary objective was to assess the safety and tolerability of riociguat; exploratory efficacy assessments included 6-min walking distance and World Health Organization (WHO) functional class. Overall, 396 patients entered the PATENT-2 study and 324 (82%) were ongoing at this interim analysis (March 2013). The safety profile of riociguat in PATENT-2 was similar to that observed in PATENT-1, with cases of haemoptysis and pulmonary haemorrhage also being observed in PATENT-2. Improvements in the patients', 6-min walking distance and WHO functional class observed in PATENT-1 persisted for up to 1 year in PATENT-2. In the observed population at the 1-year time point, mean±sd 6-min walking distance had changed by 51±74 m and WHO functional class had improved in 33%, stabilised in 61% and worsened in 6% of the patients versus the PATENT-1 baseline. Long-term riociguat was well tolerated in patients with pulmonary arterial hypertension, and led to sustained improvements in exercise capacity and functional capacity for up to 1 year.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Adulto , Idoso , Anti-Hipertensivos/uso terapêutico , Método Duplo-Cego , Antagonistas dos Receptores de Endotelina/uso terapêutico , Teste de Esforço , Feminino , Seguimentos , Guanilato Ciclase/metabolismo , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Segurança do Paciente , Prostaglandinas/química , Qualidade de Vida , Fatores de Tempo , Resultado do Tratamento
7.
Eur Respir J ; 45(5): 1293-302, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25395036

RESUMO

Riociguat is a soluble guanylate cyclase stimulator approved for the treatment of inoperable and persistent/recurrent chronic thromboembolic pulmonary hypertension (CTEPH). In the 16-week CHEST-1 study, riociguat showed a favourable benefit-risk profile and improved several clinically relevant end-points in patients with CTEPH. The CHEST-2 open-label extension evaluated the long-term safety and efficacy of riociguat. Eligible patients from CHEST-1 received riociguat individually adjusted up to a maximum dose of 2.5 mg three times daily. The primary objective was the safety and tolerability of riociguat; exploratory efficacy end-points included 6-min walking distance (6MWD) and World Health Organization (WHO) functional class (FC). Overall, 237 patients entered CHEST-2 and 211 (89%) were ongoing at this interim analysis (March 2013). The safety profile of riociguat in CHEST-2 was similar to CHEST-1, with no new safety signals. Improvements in 6MWD and WHO FC observed in CHEST-1 persisted for up to 1 year in CHEST-2. In the observed population at 1 year, mean±sd 6MWD had changed by +51±62 m (n=172) versus CHEST-1 baseline (n=237), and WHO FC had improved/stabilised/worsened in 47/50/3% of patients (n=176) versus CHEST-1 baseline (n=236). Long-term riociguat had a favourable benefit-risk profile and apparently showed sustained benefits in exercise and functional capacity for up to 1 year.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Tromboembolia/tratamento farmacológico , Idoso , Anti-Hipertensivos/uso terapêutico , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Teste de Esforço , Feminino , Seguimentos , Guanilato Ciclase/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Função Respiratória , Risco , Fatores de Tempo , Caminhada
8.
Int J Cardiol ; 167(6): 2630-7, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-22854298

RESUMO

BACKGROUND: Little is known about the effects of current PAH therapies and receptor tyrosine kinase inhibitors on heart remodeling. We sought to investigate the effects of the multikinase inhibitors sunitinib (PDGFR-, VEGFR- and KIT-inhibitor) and sorafenib (raf1/b-, VEGFR-, PDGFR-inhibitor) on pressure overload induced right ventricular (RV) remodeling. METHODS: We investigated the effects of the kinase inhibitors on hemodynamics and remodeling in rats subjected either to monocrotaline (MCT)-induced PH or to surgical pulmonary artery banding (PAB). MCT rats were treated from days 21 to 35 with either vehicle, sunitinib (1mg/kg, 5mg/kg and 10mg/kg/day) or sorafenib (10mg/kg/day). PAB rats were treated with vehicle, sunitinib (10mg/kg/day) or sorafenib (10mg/kg/day) from days 7 to 21. RV function and remodeling were determined using echocardiography, invasive hemodynamic measurement and histomorphometry. RESULTS: Treatment with both sorafenib and sunitinib decreased right ventricular systolic pressure, pulmonary vascular remodeling, RV hypertrophy and fibrosis in MCT rats. This was associated with an improvement of RV function. Importantly, after PAB, both compounds reversed RV chamber and cellular hypertrophy, reduced RV interstitial and perivascular fibrosis, and improved RV function. CONCLUSION: We demonstrated that sunitinib and sorafenib reversed RV remodeling and significantly improved RV function measured via a range of invasive and non-invasive cardiopulmonary endpoints in experimental models of RV hypertrophy.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/enzimologia , Inibidores de Proteínas Quinases/uso terapêutico , Função Ventricular Direita/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Hipertensão Pulmonar Primária Familiar , Indóis/farmacologia , Indóis/uso terapêutico , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Pirróis/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Sorafenibe , Sunitinibe , Função Ventricular Direita/fisiologia , Remodelação Ventricular/fisiologia
9.
Life Sci ; 91(13-14): 716-22, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22365955

RESUMO

AIMS: Endothelin (ET) antagonists show promise in animal models of cirrhosis and portal hypertension. The aim was to pharmacologically characterise the expression of endothelin receptors in human liver, hepatic artery and portal vein. MAIN METHODS: Immunofluorescence staining, receptor autoradiography and competition binding assays were used to localise and quantify ET receptors on hepatic parenchyma, hepatic artery and portal vein in human cirrhotic or normal liver. Additional experiments were performed to determine the affinity and selectivity of ET antagonists for liver ET endothelin receptors. An endothelial cell ET(B) knockout murine model was used to examine the function of sinusoid endothelial ET(B) receptors. KEY FINDINGS: ET(B) receptors predominated in normal human liver and displayed the highest ratio (ET(B):ET(A) 63:47) compared with other peripheral tissues. In two patients examined, liver ET(B) expression was up-regulated in cirrhosis (ET(B):ET(A) 83:17). Both sub-types localised to the media of normal portal vein but ET(B) receptors were downregulated fivefold in the media of cirrhotic portal vein. Sinusoid diameter was fourfold smaller in endothelial cell ET(B) knockout mice. The liver morphology of ET(B) knockout mice was markedly different to normal murine liver, with loss of the wide spread sinusoidal pattern. In the knockout mice, sinusoids were reduced in both number and absolute diameter, while large intrahepatic veins were congested with red blood cells. SIGNIFICANCE: These data support a role for the ET system in cirrhosis of the liver and suggest that endothelial ET(B) blockade may cause sinusoidal constriction which may contribute to hepatotoxicity associated with some endothelin antagonists.


Assuntos
Cirrose Hepática/patologia , Fígado/metabolismo , Receptor de Endotelina A/genética , Receptor de Endotelina B/genética , Animais , Autorradiografia , Ligação Competitiva , Artéria Hepática/metabolismo , Humanos , Isoxazóis/farmacologia , Fígado/patologia , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Fenilpropionatos/farmacologia , Veia Porta/metabolismo , Veia Porta/patologia , Piridazinas/farmacologia , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Tiofenos/farmacologia , Regulação para Cima
10.
Respir Res ; 12: 87, 2011 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-21699729

RESUMO

BACKGROUND: Endothelin-1 signalling plays an important role in pathogenesis of pulmonary hypertension. Although different endothelin-A receptor antagonists are developed, a novel therapeutic option to cure the disease is still needed. This study aims to investigate the therapeutic efficacy of the selective endothelin-A receptor antagonist TBC3711 in monocrotaline-induced pulmonary hypertension in rats. METHODS: Monocrotaline-injected male Sprague-Dawley rats were randomized and treated orally from day 21 to 35 either with TBC3711 (Dose: 30 mg/kg body weight/day) or placebo. Echocardiographic measurements of different hemodynamic and right-heart hypertrophy parameters were performed. After day 35, rats were sacrificed for invasive hemodynamic and right-heart hypertrophy measurements. Additionally, histologic assessment of pulmonary vascular and right-heart remodelling was performed. RESULTS: The novel endothelin-A receptor antagonist TBC3711 significantly attenuated monocrotaline-induced pulmonary hypertension, as evident from improved hemodynamics and right-heart hypertrophy in comparison with placebo group. In addition, muscularization and medial wall thickness of distal pulmonary vessels were ameliorated. The histologic evaluation of the right ventricle showed a significant reduction in fibrosis and cardiomyocyte size, suggesting an improvement in right-heart remodelling. CONCLUSION: The results of this study suggest that the selective endothelin-A receptor antagonist TBC3711 demonstrates therapeutic benefit in rats with established pulmonary hypertension, thus representing a useful therapeutic approach for treatment of pulmonary hypertension.


Assuntos
Anti-Hipertensivos/farmacologia , Hipertensão Pulmonar/tratamento farmacológico , Hipertrofia Ventricular Direita/tratamento farmacológico , Isoxazóis/farmacologia , Monocrotalina , Sulfonas/farmacologia , Administração Oral , Animais , Anti-Hipertensivos/administração & dosagem , Modelos Animais de Doenças , Ecocardiografia Doppler , Antagonistas do Receptor de Endotelina A , Fibrose , Hemodinâmica/efeitos dos fármacos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/diagnóstico por imagem , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/diagnóstico por imagem , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/fisiopatologia , Isoxazóis/administração & dosagem , Masculino , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/metabolismo , Sulfonas/administração & dosagem , Fatores de Tempo , Função Ventricular Direita/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
11.
Am J Pathol ; 168(6): 1793-807, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16723696

RESUMO

The precise cellular and molecular mechanisms regulating adventitial vasa vasorum neovascularization, which occurs in the pulmonary arterial circulation in response to hypoxia, remain unknown. Here, using a technique to isolate and culture adventitial fibroblasts (AdvFBs) and vasa vasorum endothelial cells (VVECs) from the adventitia of pulmonary arteries, we report that hypoxia-activated pulmonary artery AdvFBs exhibited pro-angiogenic properties and influenced the angiogenic phenotype of VVEC, in a process of cell-cell communication involving endothelin-1 (ET-1). We demonstrated that AdvFBs, either via co-culture or conditioned media, stimulated VVEC proliferation and augmented the self-assembly and integrity of cord-like networks that formed when VVECs where cultured on Matrigel. In addition, hypoxia-activated AdvFBs produced ET-1, suggesting a paracrine role for this pro-angiogenic molecule in these processes. When co-cultured on Matrigel, AdvFBs and VVECs self-assembled into heterotypic cord-like networks, a process augmented by hypoxia but attenuated by either selective endothelin receptor antagonists or oligonucleotides targeting prepro-ET-1 mRNA. From these observations, we propose that hypoxia-activated AdvFBs exhibit pro-angiogenic properties and, as such, communicate with VVECs, in a process involving ET-1, to regulate vasa vasorum neovascularization occurring in the adventitia of pulmonary arteries in response to chronic hypoxia.


Assuntos
Endotelina-1/metabolismo , Endotélio Vascular/metabolismo , Fibroblastos/patologia , Hipóxia , Artéria Pulmonar/metabolismo , Animais , Bovinos , Técnicas de Cultura de Células/métodos , Proliferação de Células , Células Cultivadas/metabolismo , Modelos Animais de Doenças , Hipertensão/patologia , Microscopia de Fluorescência
12.
Physiology (Bethesda) ; 21: 134-45, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16565479

RESUMO

An increasing volume of experimental data indicates that the adventitial fibroblast, in both the pulmonary and systemic circulations, is a critical regulator of vascular wall function in health and disease. A rapidly emerging concept is that the vascular adventitia acts as biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. In response to stress or injury, resident adventitial cells can be activated and reprogrammed to exhibit different functional and structural behaviors. In fact, under certain conditions, the adventitial compartment may be considered the principal injury-sensing tissue of the vessel wall. In response to vascular stresses such as overdistension and hypoxia, the adventitial fibroblast is activated and undergoes phenotypic changes, which include proliferation, differentiation, upregulation of contractile and extracellular matrix proteins, and release of factors that directly affect medial smooth muscle cell tone and growth and that stimulate recruitment of inflammatory and progenitor cells to the vessel wall. Each of these changes in fibroblast phenotype modulates either directly or indirectly changes in overall vascular function and structure. The purpose of this review is to present the current evidence demonstrating that the adventitial fibroblast acts as a key regulator of pulmonary vascular function and structure from the "outside-in."


Assuntos
Tecido Conjuntivo/fisiologia , Endotélio Vascular/fisiologia , Fibroblastos/fisiologia , Artéria Pulmonar/fisiologia , Veias Pulmonares/fisiologia , Animais , Comunicação Celular , Diferenciação Celular , Proliferação de Células , Tecido Conjuntivo/patologia , Endotélio Vascular/patologia , Matriz Extracelular/fisiologia , Fibroblastos/patologia , Humanos , Hipóxia/etiologia , Hipóxia/patologia , Hipóxia/fisiopatologia , Pneumopatias/etiologia , Pneumopatias/patologia , Pneumopatias/fisiopatologia , Artéria Pulmonar/patologia , Circulação Pulmonar/fisiologia , Veias Pulmonares/patologia , Células-Tronco/patologia , Células-Tronco/fisiologia
13.
Am J Pathol ; 168(2): 659-69, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16436679

RESUMO

Vascular remodeling in chronic hypoxic pulmonary hypertension includes marked fibroproliferative changes in the pulmonary artery (PA) adventitia. Although resident PA fibroblasts have long been considered the primary contributors to these processes, we tested the hypothesis that hypoxia-induced pulmonary vascular remodeling requires recruitment of circulating mesenchymal precursors of a monocyte/macrophage lineage, termed fibrocytes. Using two neonatal animal models (rats and calves) of chronic hypoxic pulmonary hypertension, we demonstrated a dramatic perivascular accumulation of mononuclear cells of a monocyte/macrophage lineage (expressing CD45, CD11b, CD14, CD68, ED1, ED2). Many of these cells produced type I collagen, expressed alpha-smooth muscle actin, and proliferated, thus exhibiting mesenchymal cell characteristics attributed to fibrocytes. The blood-borne origin of these cells was confirmed in experiments wherein circulating monocytes/macrophages of chronically hypoxic rats were in vivo-labeled with DiI fluorochrome via liposome delivery and subsequently identified in the remodeled pulmonary, but not systemic, arterial adventitia. The DiI-labeled cells that appeared in the vessel wall expressed monocyte/macrophage markers and procollagen. Selective depletion of this monocytic cell population, using either clodronate-liposomes or gadolinium chloride, prevented pulmonary adventitial remodeling (ie, production of collagen, fibronectin, and tenascin-C and accumulation of myofibroblasts). We conclude that circulating mesenchymal precursors of a monocyte/macrophage lineage, including fibrocytes, are essential contributors to hypoxia-induced pulmonary vascular remodeling.


Assuntos
Hipertensão Pulmonar/metabolismo , Hipóxia/metabolismo , Macrófagos/fisiologia , Monócitos/fisiologia , Circulação Pulmonar/fisiologia , Células-Tronco/fisiologia , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Bovinos , Linhagem da Célula , Proliferação de Células , Colágeno Tipo I/metabolismo , Fibroblastos/patologia , Fibroblastos/fisiologia , Fibronectinas/metabolismo , Hipertensão Pulmonar/patologia , Lipossomos , Masculino , Músculo Liso Vascular/metabolismo , Artéria Pulmonar/patologia , Ratos , Ratos Endogâmicos WKY , Tenascina/metabolismo
16.
J Appl Physiol (1985) ; 98(2): 715-21, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15649883

RESUMO

Data are rapidly accumulating in support of the idea that circulating monocytes and/or mononuclear fibrocytes are recruited to the pulmonary circulation of chronically hypoxic animals and that these cells play an important role in the pulmonary hypertensive process. Hypoxic induction of monocyte chemoattractant protein-1, stromal cell-derived factor-1, vascular endothelial growth factor-A, endothelin-1, and tumor growth factor-beta(1) in pulmonary vessel wall cells, either directly or indirectly via signals from hypoxic lung epithelial cells, may be a critical first step in the recruitment of circulating leukocytes to the pulmonary circulation. In addition, hypoxic stress appears to induce release of increased numbers of monocytic progenitor cells from the bone marrow, and these cells may have upregulated expression of receptors for the chemokines produced by the lung circulation, which thus facilitates their specific recruitment to the pulmonary site. Once present, macrophages/fibrocytes may exert paracrine effects on resident pulmonary vessel wall cells stimulating proliferation, phenotypic modulation, and migration of resident fibroblasts and smooth muscle cells. They may also contribute directly to the remodeling process through increased production of collagen and/or differentiation into myofibroblasts. In addition, they could play a critical role in initiating and/or supporting neovascularization of the pulmonary artery vasa vasorum. The expanded vasa network may then act as a conduit for further delivery of circulating mononuclear cells to the pulmonary arterial wall, creating a feedforward loop of pathological remodeling. Future studies will need to determine the mechanisms that selectively induce leukocyte/fibrocyte recruitment to the lung circulation under hypoxic conditions, their direct role in the remodeling process via production of extracellular matrix and/or differentiation into myofibroblasts, their impact on the phenotype of resident smooth muscle cells and adventitial fibroblasts, and their role in the neovascularization observed in hypoxic pulmonary hypertension.


Assuntos
Hipertensão Pulmonar/imunologia , Hipóxia/imunologia , Leucócitos/imunologia , Pulmão/imunologia , Ativação Linfocitária/imunologia , Circulação Pulmonar/imunologia , Animais , Humanos , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Modelos Imunológicos
17.
J Neurosci Res ; 76(1): 64-75, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15048930

RESUMO

Progressive "dying back" neurodegenerative diseases are debilitating due to loss of connectivity after nerve terminal and axonal withdrawal, which impairs peripheral nerve function and leads ultimately to neuronal cell death. The mutant mouse (Wallerian degeneration slow; Wld(s)) provides an accessible model system to understand orthograde and retrograde degeneration, because in these mice axotomy induces slow, progressive withdrawal of nerve terminals from motor endplates. Axon degeneration itself is about 10 times slower than in wild-type mice. We describe an organ culture paradigm that permits direct observation of the progressive changes in morphology of neuromuscular junctions in Wld(s) mutant mice. Normal nerve terminal and motor endplate morphology were maintained at most Wld(s) neuromuscular junctions for up to 72 hr in vitro. At others, synaptic boutons were removed from postsynaptic junctional folds in piecemeal fashion, as observed in adults in vivo. By contrast, nerve terminals degenerated rapidly and synchronously in wild-type muscle cultures, resembling Wallerian degeneration in vivo. These observations confirm that in Wld(s) mice, axotomy triggers a mechanism of nerve-terminal withdrawal that seems qualitatively different from that in wild-type animals. The piecemeal dismantling of presynaptic terminals resembles that occurring during neonatal synapse elimination. Organ cultures of neonatal Wld(s) muscle maintained for 1-2 days in vitro also showed no evidence of synaptic terminal degeneration, but elimination of polyneuronal innervation progressed in vitro at approximately the same rate as in vivo. Taken together, the data suggest that both natural and axotomy-induced forms of synapse withdrawal may be accessible to continuous observation and analysis, in organ-cultures of Wld(S) mouse muscles. This offers several advantages over repeated visualization of synaptic remodeling that has thus far been possible only in vivo.


Assuntos
Axônios/fisiologia , Axotomia , Músculo Esquelético/inervação , Músculo Esquelético/fisiologia , Sinapses/fisiologia , Degeneração Walleriana/genética , Animais , Animais Recém-Nascidos , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Placa Motora/fisiologia , Junção Neuromuscular/fisiologia , Técnicas de Cultura de Órgãos , Terminações Pré-Sinápticas/fisiologia , Receptores Colinérgicos/fisiologia
18.
Am J Physiol Lung Cell Mol Physiol ; 286(4): L668-78, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12754186

RESUMO

Information is rapidly emerging regarding the important role of the arterial vasa vasorum in a variety of systemic vascular diseases. In addition, increasing evidence suggests that progenitor cells of bone marrow (BM) origin may contribute to postnatal neovascularization and/or vascular wall thickening that is characteristic in some forms of systemic vascular disease. Little is known regarding postnatal vasa formation and the role of BM-derived progenitor cells in the setting of pulmonary hypertension (PH). We sought to determine the effects of chronic hypoxia on the density of vasa vasorum in the pulmonary artery and to evaluate if BM-derived progenitor cells contribute to the increased vessel wall mass in a bovine model of hypoxia-induced PH. Quantitative morphometric analyses of lung tissue from normoxic and hypoxic calves revealed that hypoxia results in a dramatic expansion of the pulmonary artery adventitial vasa vasorum. Flow cytometric analysis demonstrated that cells expressing the transmembrane tyrosine kinase receptor for stem cell factor, c-kit, are mobilized from the BM in the circulation in response to hypoxia. Immunohistochemistry revealed an increase in the expression of c-kit+ cells together with vascular endothelial growth factor, fibronectin, and thrombin in the hypoxia-induced remodeled pulmonary artery vessel wall. Circulating mononuclear cells isolated from neonatal calves exposed to hypoxia were found to differentiate into endothelial and smooth muscle cell phenotypes depending on culture conditions. From these observations, we suggest that the vasa vasorum and circulating progenitor cells could be involved in vessel wall thickening in the setting of hypoxia-induced PH.


Assuntos
Hipóxia/patologia , Neovascularização Patológica/patologia , Artéria Pulmonar/patologia , Células-Tronco/patologia , Animais , Animais Recém-Nascidos , Bovinos , Diferenciação Celular , Células Cultivadas , Eritrócitos , Hipertensão Pulmonar/patologia , Masculino , Proteínas Proto-Oncogênicas c-kit/análise , Artéria Pulmonar/química , Vasa Vasorum/química , Vasa Vasorum/patologia
19.
Am J Respir Crit Care Med ; 165(3): 398-405, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11818328

RESUMO

We determined the distribution of ET(A) and ET(B) receptors in pulmonary arteries from pulmonary hypertensive patients and control subjects, using in vitro autoradiography, and investigated their role in mediating the proliferative effects of endothelin-1 (ET-1) on distal human pulmonary artery smooth muscle cells (PASMCs). Distal arteries possessed more medial [(125)I]-ET-1 binding sites (105 +/- 10 versus 45 +/- 6 amol/mm(2); p < 0.001) and a greater proportion of ET(B) receptors than proximal arteries (36 +/- 3% versus 3 +/- 1%; p < 0.001). Receptor density in distal arteries and lung parenchyma was twofold greater (p < 0.05) in pulmonary hypertensive patients than in control subjects. ET-1 (10(-9)-10(-7) mol/L) stimulated DNA synthesis (147 +/- 10% of control subjects; p < 0.05) and attenuated the antiproliferative action of cicaprost and forskolin on PASMCs, these effects being mediated via ET(A) and ET(B) receptors. Serum-stimulated proliferation was attenuated by inhibiting either endogenous ET-1 release with phosphoramidon (10(-5) mol/L) or its action with PD145065 (10(-5) mol/L). Cicaprost (10(-10)-10(-7) mol/L) inhibited ET-1 release from PASMCs (49 +/- 16% of control after 24 h; p < 0.001) and increased intracellular cAMP levels, whereas ET(B) receptor stimulation selectively reduced cAMP levels. In conclusion, ET(A) and ET(B) receptors are differentially distributed in human pulmonary arteries. Both receptors promote the proliferation of PASMCs in vitro and may contribute to vascular remodeling in pulmonary hypertension.


Assuntos
Músculo Liso Vascular/citologia , Artéria Pulmonar/citologia , Receptores de Endotelina/fisiologia , Adulto , Divisão Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/química , Artéria Pulmonar/química , Receptores de Endotelina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...