Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Intervalo de ano de publicação
2.
Cancer Prev Res (Phila) ; 5(8): 1061-71, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22700853

RESUMO

Patients with chronic obstructive pulmonary disease (COPD) are at an increased risk for the development of lung cancer, the mechanisms for which are incompletely understood. We hypothesized that the hypoxic pulmonary microenvironment present in COPD would augment lung carcinogenesis. Mice were subjected to chemical carcinogenesis protocols and placed in either hypoxia or normoxia. Mice exposed to chronic hypoxia developed tumors with increased volume compared with normoxic controls. Both lungs and tumors from hypoxic mice showed a preferential stabilization of HIF-2α and increased expression of VEGF-A, FGF2, and their receptors as well as other survival, proliferation, and angiogenic signaling pathways regulated by HIF-2α. We showed that tumors arising in hypoxic animals have increased sensitivity to VEGFR-2/EGFR inhibition, as chemoprevention with vandetanib showed markedly increased activity in hypoxic mice. These studies showed that lung tumors arising in a hypoxic microenvironment express increased growth, angiogenic, and survival signaling that could contribute to the increased lung cancer risk in COPD. Furthermore, the differential sensitivity of tumors arising in hypoxia to VEGFR-2/EGFR inhibition suggests that the altered signaling present in tumors arising in hypoxic lung might be therapeutically exploited in patients with underlying COPD.


Assuntos
Carcinógenos/toxicidade , Receptores ErbB/metabolismo , Hipóxia/fisiopatologia , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/patologia , Alvéolos Pulmonares/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Antioxidantes/toxicidade , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores Tumorais/metabolismo , Western Blotting , Hidroxitolueno Butilado/toxicidade , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Transição Epitelial-Mesenquimal , Receptores ErbB/antagonistas & inibidores , Feminino , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/metabolismo , Metilcolantreno/análogos & derivados , Metilcolantreno/toxicidade , Camundongos , Neovascularização Patológica , Piperidinas/uso terapêutico , Proteínas Proto-Oncogênicas c-myc/metabolismo , Quinazolinas/uso terapêutico , Síndromes da Apneia do Sono/fisiopatologia , Uretana/toxicidade , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
3.
Circulation ; 121(25): 2747-54, 2010 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-20547927

RESUMO

BACKGROUND: The plexiform lesion is the hallmark of severe pulmonary arterial hypertension. However, its genesis and hemodynamic effects are largely unknown because of the limited availability of lung tissue samples from patients with pulmonary arterial hypertension and the lack of appropriate animal models. This study investigated whether rats with severe progressive pulmonary hypertension developed plexiform lesions. METHODS AND RESULTS: After a single subcutaneous injection of the vascular endothelial growth factor receptor blocker Sugen 5416, rats were exposed to hypoxia for 3 weeks. They were then returned to normoxia for an additional 10 to 11 weeks. Hemodynamic and histological examinations were performed at 13 to 14 weeks after the Sugen 5416 injection. All rats developed pulmonary hypertension (right ventricular systolic pressure approximately 100 mm Hg) and severe pulmonary arteriopathy, including concentric neointimal and complex plexiform-like lesions. There were 2 patterns of complex lesion formation: a lesion forming within the vessel lumen (stalk-like) and another that projected outside the vessel (aneurysm-like). Immunohistochemical analyses showed that these structures had cellular and molecular features closely resembling human plexiform lesions. CONCLUSIONS: Severe, sustained pulmonary hypertension in a very late stage of the Sugen 5416/hypoxia/normoxia-exposed rat is accompanied by the formation of lesions that are indistinguishable from the pulmonary arteriopathy of human pulmonary arterial hypertension. This unique model provides a new and rigorous approach for investigating the genesis, hemodynamic effects, and reversibility of plexiform and other occlusive lesions in pulmonary arterial hypertension.


Assuntos
Modelos Animais de Doenças , Hipertensão Pulmonar/patologia , Animais , Artérias/patologia , Hipertensão Pulmonar/etiologia , Hipóxia , Pulmão/irrigação sanguínea , Ratos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
4.
J Am Coll Cardiol ; 54(1 Suppl): S108-S117, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19555854

RESUMO

Over the past 2 decades, pulmonary arterial hypertension has evolved from a uniformly fatal condition to a chronic, manageable disease in many cases, the result of unparalleled development of new therapies and advances in early diagnosis. However, none of the currently available therapies is curative, so the search for new treatment strategies continues. With a deeper understanding of the genetics and the molecular mechanisms of pulmonary vascular disorders, we are now at the threshold of entering a new therapeutic era. Our working group addressed what can be expected in the near future. The topics span the understanding of genetic variations, novel antiproliferative treatments, the role of stem cells, the right ventricle as a therapeutic target, and strategies and challenges for the translation of novel experimental findings into clinical practice.


Assuntos
Hipertensão Pulmonar/terapia , Doença Crônica , Endotelinas/fisiologia , Endotélio Vascular/citologia , Proteínas de Ligação ao GTP/fisiologia , Insuficiência Cardíaca/epidemiologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/epidemiologia , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/fisiopatologia , NADP/fisiologia , Peptídeos Natriuréticos/fisiologia , Neovascularização Fisiológica , Estresse Oxidativo , Fator de Crescimento Derivado de Plaquetas/fisiologia , Polimorfismo Genético , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/fisiologia , Remodelação Ventricular/fisiologia
5.
Am J Pathol ; 174(3): 782-96, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19234135

RESUMO

Neprilysin is a transmembrane metalloendopeptidase that degrades neuropeptides that are important for both growth and contraction. In addition to promoting carcinogenesis, decreased levels of neprilysin increases inflammation and neuroendocrine cell hyperplasia, which may predispose to vascular remodeling. Early pharmacological studies showed a decrease in chronic hypoxic pulmonary hypertension with neprilysin inhibition. We used a genetic approach to test the alternate hypothesis that neprilysin depletion increases chronic hypoxic pulmonary hypertension. Loss of neprilysin had no effect on baseline airway or alveolar wall architecture, vessel density, cardiac function, hematocrit, or other relevant peptidases. Only lung neuroendocrine cell hyperplasia and a subtle neuropeptide imbalance were found. After chronic hypoxia, neprilysin-null mice exhibited exaggerated pulmonary hypertension and striking increases in muscularization of distal vessels. Subtle thickening of proximal media/adventitia not typically seen in mice was also detected. In contrast, adaptive right ventricular hypertrophy was less than anticipated. Hypoxic wild-type pulmonary vessels displayed close temporal and spatial relationships between decreased neprilysin and increased cell growth. Smooth muscle cells from neprilysin-null pulmonary arteries had increased proliferation compared with controls, which was decreased by neprilysin replacement. These data suggest that neprilysin may be protective against chronic hypoxic pulmonary hypertension in the lung, at least in part by attenuating the growth of smooth muscle cells. Lung-targeted strategies to increase neprilysin levels could have therapeutic benefits in the treatment of this disorder.


Assuntos
Hipertensão Pulmonar/patologia , Hipóxia/genética , Camundongos Knockout , Neprilisina/deficiência , Artéria Pulmonar/patologia , Circulação Pulmonar/fisiologia , Animais , Divisão Celular , Doença Crônica , Primers do DNA , Predisposição Genética para Doença , Genótipo , Hemodinâmica , Hipertensão Pulmonar/genética , Hipóxia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/patologia , Neprilisina/genética
6.
Am J Physiol Lung Cell Mol Physiol ; 295(1): L71-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18469113

RESUMO

RhoA/Rho kinase (ROCK) signaling plays a key role in the pathogenesis of experimental pulmonary hypertension (PH). Dehydroepiandrosterone (DHEA), a naturally occurring steroid hormone, effectively inhibits chronic hypoxic PH, but the responsible mechanisms are unclear. This study tested whether DHEA was also effective in treating monocrotaline (MCT)-induced PH in left pneumonectomized rats and whether inhibition of RhoA/ROCK signaling was involved in the protective effect of DHEA. Three weeks after MCT injection, pneumonectomized rats developed PH with severe vascular remodeling, including occlusive neointimal lesions in pulmonary arterioles. In lungs from these animals, we detected cleaved (constitutively active) ROCK I as well as increases in activities of RhoA and ROCK and increases in ROCK II protein expression. Chronic DHEA treatment (1%, by food for 3 wk) markedly inhibited the MCT-induced PH (mean pulmonary artery pressures after treatment with 0% and 1% DHEA were 33+/-5 and 16+/-1 mmHg, respectively) and severe pulmonary vascular remodeling in pneumonectomized rats. The MCT-induced changes in RhoA/ROCK-related protein expression were nearly normalized by DHEA. A 3-wk DHEA treatment (1%) started 3 wk after MCT injection completely inhibited the progression of PH (mean pulmonary artery pressures after treatment with 0% and 1% DHEA were 47+/-3 and 30+/-3 mmHg, respectively), and this treatment also resulted in 100% survival in contrast to 30% in DHEA-untreated rats. These results suggest that inhibition of RhoA/ROCK signaling, including the cleavage and constitutive activation of ROCK I, is an important component of the impressive protection of DHEA against MCT-induced PH in pneumonectomized rats.


Assuntos
Adjuvantes Imunológicos/farmacologia , Desidroepiandrosterona/farmacologia , Hipertensão Pulmonar/enzimologia , Monocrotalina/toxicidade , Quinases Associadas a rho/biossíntese , Proteína rhoA de Ligação ao GTP/biossíntese , Animais , Pressão Sanguínea/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/patologia , Pneumonectomia , Artéria Pulmonar/enzimologia , Artéria Pulmonar/patologia , Ratos , Fatores de Tempo
7.
Cardiovasc Res ; 74(3): 377-87, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17346686

RESUMO

OBJECTIVE: It has been reported that dehydroepiandrosterone is a pulmonary vasodilator and inhibits chronic hypoxia-induced pulmonary hypertension. Additionally, dehydroepiandrosterone has been shown to improve systemic vascular endothelial function. Thus, we hypothesized that chronic treatment with dehydroepiandrosterone would attenuate hypoxic pulmonary hypertension by enhancing pulmonary artery endothelial function. METHODS AND RESULTS: Rats were randomly assigned to five groups. Three groups received food containing 0, 0.3, or 1% dehydroepiandrosterone during a 3-wk-exposure to simulated high altitude (HA). The other 2 groups were kept at Denver's low altitude (LA) and received food containing 0 or 1% dehydroepiandrosterone. Dehydroepiandrosterone dose-dependently inhibited hypoxic pulmonary hypertension (mean pulmonary artery pressures after treatment with 0, 0.3, and 1% dehydroepiandrosterone=45+/-5, 33+/-2*, and 25+/-1*# mmHg, respectively. *P<0.05 vs. 0% and # vs. 0.3%). Dehydroepiandrosterone (1%, 3 wks) treatment started after rats had been exposed to 3-wk hypoxia also effectively reversed established hypoxic pulmonary hypertension. Pulmonary artery rings isolated from both LA and HA rats treated with 1% dehydroepiandrosterone showed enhanced relaxations to acetylcholine and sodium nitroprusside, but not to 8-bromo-cGMP. In the pulmonary artery tissue from dehydroepiandrosterone-treated LA and HA rats, soluble guanylate cyclase, but not endothelial nitric oxide synthase, protein levels were increased. CONCLUSION: These results indicate that the protective effect of dehydroepiandrosterone against hypoxic pulmonary hypertension may involve upregulation of pulmonary artery soluble guanylate cyclase protein expression and augmented pulmonary artery vasodilator responsiveness to nitric oxide.


Assuntos
Desidroepiandrosterona/uso terapêutico , Guanilato Ciclase/metabolismo , Hipertensão Pulmonar/tratamento farmacológico , Artéria Pulmonar/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Regulação para Cima , Acetilcolina/farmacologia , Animais , Western Blotting , GMP Cíclico/farmacologia , Desidroepiandrosterona/metabolismo , Sulfato de Desidroepiandrosterona/sangue , Sulfato de Desidroepiandrosterona/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Estradiol/sangue , Guanilato Ciclase/análise , Hipertensão Pulmonar/sangue , Hipertensão Pulmonar/metabolismo , Hipóxia/metabolismo , Técnicas In Vitro , Pulmão/enzimologia , Masculino , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/análise , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Nitroprussiato/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/análise , Guanilil Ciclase Solúvel , Testosterona/sangue , Vasodilatadores/farmacologia
10.
Rev. bras. reumatol ; 43(5): 294-301, set.-out. 2003. tab
Artigo em Inglês | LILACS | ID: lil-386625

RESUMO

Pacientes com esclerodermia têm risco aumentado para desenvolver hipertensão pulmonar. O aparecimento de dispnéia e/ou a diminuição da capacidade de difusão devem levar à suspeita imediata dessa complicação. A ecodopplercardiografia é importante para o diagnóstico e o seguimento desses casos. Os casos não tratados de hipertensão pulmonar em esclerodermia têm mau prognóstico, daí a necessidade em manter sob vigilância estes pacientes. Na última década surgiram avanços para o tratamento da hipertensão arterial pulmonar, incluindo os medicamentos epoprostenol EV, bosentan VO e treprostinil SC. À medida que novas terapias vão sendo desnvolvidas, torna-se necessário a realização de estudos clínicos de maior validade


Assuntos
Humanos , Dispneia , Hipertensão Pulmonar , Escleroderma Sistêmico
11.
Am J Physiol Heart Circ Physiol ; 284(4): H1321-31, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12505875

RESUMO

PKC contributes to regulation of pulmonary vascular reactivity in response to hypoxia. The role of individual PKC isozymes is less clear. We used a knockout (null, -/-) mouse to test the hypothesis that PKC-epsilon is important in acute hypoxic pulmonary vasoconstriction (HPV). We asked whether deletion of PKC-epsilon would decrease acute HPV in adult C57BL6xSV129 mice. In isolated, salt solution-perfused lung, reactivity to acute hypoxic challenges (0% and 3% O(2)) was compared with responses to angiotensin II (ANG II) and KCl. PKC-epsilon -/- mice had decreased HPV, whereas responses to ANG II and KCl were preserved. Inhibition of nitric oxide synthase (NOS) with nitro-l-arginine augmented HPV in PKC-epsilon +/+ but not -/- mice. Inhibition of Ca(2+)-gated K(+) channels (K(Ca)) with charybdotoxin and apamin did not enhance HPV in -/- mice relative to wild-type (+/+) controls. In contrast, the voltage-gated K(+) channel (K(V)) antagonist 4-aminopyridine increased the response of -/- mice beyond that of +/+ mice. This suggested that increased K(V) channel expression could contribute to blunted HPV in PKC-epsilon -/- mice. Therefore, expression of the O(2)-sensitive K(V) channel subunit Kv3.1b (100-kDa glycosylated form and 70-kDa core protein) was compared in whole lung and pulmonary artery smooth muscle cell (PASMC) lysates from +/+ and -/- mice. A subtle increase in Kv3.1b was detected in -/- vs. +/+ whole lung lysates. A much greater rise in Kv3.1b expression was found in -/- vs. +/+ PASMC. Thus deletion of PKC-epsilon blunts murine HPV. The decreased response could not be attributed to a general loss in vasoreactivity or derangements in NOS or K(Ca) channel activity. Instead, the absence of PKC-epsilon allows increased expression of K(V) channels (like Kv3.1b) to occur in PASMC, which likely contributes to decreased HPV.


Assuntos
Hipóxia/fisiopatologia , Pulmão/irrigação sanguínea , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Proteína Quinase C/deficiência , Vasoconstrição , Angiotensina II/farmacologia , Animais , Cálcio/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Deleção de Genes , Pulmão/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/química , Óxido Nítrico Sintase/antagonistas & inibidores , Oxigênio/administração & dosagem , Oxigênio/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/análise , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Cloreto de Potássio/farmacologia , Proteína Quinase C/genética , Proteína Quinase C/fisiologia , Proteína Quinase C-épsilon , Artéria Pulmonar/química , Artéria Pulmonar/fisiopatologia , Canais de Potássio Shaw
12.
Adv Exp Med Biol ; 543: 127-37, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14713118

RESUMO

Intracellular signaling via the small GTP-binding protein RhoA and its downstream effector Rho-kinase plays a role in regulating diverse cellular functions, including cell contraction, migration, gene expression, proliferation, and differentiation. Rho/Rho-kinase signaling has an obligatory role in embryonic cardiac development, and low-level chemical activation of Rho promotes branching morphogenesis in fetal lung explants. Gebb has found that hypoxia markedly augments branching morphogenesis in fetal rat lung explants, and our preliminary results suggest this is associated with activation of RhoA. Whereas hypoxia-induced activation of Rho/Rho-kinase may promote fetal lung development, other evidence indicates it has adverse effects in the lungs of neonates and adults. When exposed at birth to the mild hypoxia of Denver's altitude (5,280 ft), the neonatal fawn-hooded rat (FHR) develops severe pulmonary hypertension (PH) associated with impaired lung alveolarization and vascularization. We have observed that administration via the drinking water of the Rho-kinase inhibitor fasudil to the nursing, Denver FHR mother for the first 2 to 3 weeks, and then directly to the Denver FHR pups for the next 7 to 8 weeks, ameliorates the lung dysplasia and PH. The adult Sprague-Dawley rat develops PH when exposed for 3 to 4 wk to a simulated altitude of 17,000 ft. We have found that this hypoxic PH is associated with activation of pulmonary artery Rho/Rho-kinase and is almost completely reversed by acute intravenous administration of the Rho-kinase inhibitor Y-27632. In addition, chronic in vivo treatment with Y-27632 reduces development of the hypoxic PH. In summary, hypoxic activation of Rho/Rho-kinase signaling may be important for fetal lung morphogenesis, but continued activation of this pathway in the neonate impairs postnatal lung development and re-activation in the adult contributes to development of PH.


Assuntos
Hipertensão Pulmonar/etiologia , Hipóxia/metabolismo , Pulmão/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Altitude , Animais , Animais Recém-Nascidos , Feto/metabolismo , Hipóxia/complicações , Hipóxia/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Pulmão/embriologia , Pulmão/metabolismo , Ratos , Transdução de Sinais , Quinases Associadas a rho
13.
J Appl Physiol (1985) ; 93(6): 2162-70, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12391106

RESUMO

Tumor necrosis factor-alpha (TNF-alpha) transgenic mice have previously been found to have characteristics consistent with emphysema and severe pulmonary hypertension. Lungs demonstrated alveolar enlargement as well as interstitial thickening due to chronic inflammation and perivascular fibrosis. In the present report, we sought to determine potential mechanisms leading to development of pulmonary hypertension in TNF-alpha transgenic mice. To determine whether sustained vasoconstriction was an important component of this pulmonary hypertension, nitric oxide was administered and hemodynamics were measured. Nitric oxide (25 ppm) failed to normalize right ventricular pressure in transgene-positive mice, suggesting that the pulmonary hypertension was not due to sustained vasoconstriction. Structural analysis of the pulmonary arteries found adventitial thickening and a trend toward medial hypertrophy in pulmonary arteries of transgene-positive mice, suggesting that vascular remodeling had occurred. Echocardiographic measurement of the percent fractional shortening of the left ventricle as a measurement of ventricular function in vivo revealed that left ventricular dysfunction was not contributing to pulmonary hypertension. We examined expression of genes known to be important in regulation of vascular tone and structure. Messenger RNA expression of vascular endothelial growth factor and its receptor flk-1 was reduced compared with transgene-negative littermates at all ages. Endothelial and inducible nitric oxide synthase mRNA levels were similar in both groups. Endothelin-1 mRNA was also decreased in TNF-alpha transgenic mice. Interestingly, female transgenic mice had decreased survival rate compared with male transgenic mice. We conclude that chronic overexpression of TNF-alpha is associated with decreased vascular endothelial growth factor and flk-1 gene expression, pulmonary vascular remodeling, and severe pulmonary hypertension, although the precise mechanism is unknown.


Assuntos
Fatores de Crescimento Endotelial/genética , Hipertensão Pulmonar/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linfocinas/genética , Fator de Necrose Tumoral alfa/genética , Animais , Ecocardiografia , Feminino , Expressão Gênica/fisiologia , Hipertensão Pulmonar/mortalidade , Hipertensão Pulmonar/patologia , Pulmão/irrigação sanguínea , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Óxido Nítrico/farmacologia , RNA Mensageiro/análise , Taxa de Sobrevida , Fator A de Crescimento do Endotélio Vascular , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA