Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Respir Crit Care Med ; 201(2): 198-211, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31738079

RESUMO

Rationale: Alveolar epithelial cell (AEC) injury and dysregulated repair are implicated in the pathogenesis of pulmonary fibrosis. Endoplasmic reticulum (ER) stress in AEC has been observed in idiopathic pulmonary fibrosis (IPF), a disease of aging.Objectives: To investigate a causal role for ER stress in the pathogenesis of pulmonary fibrosis (PF) and therapeutic potential of ER stress inhibition in PF.Methods: The role of ER stress in AEC dysfunction and fibrosis was studied in mice with tamoxifen (Tmx)-inducible deletion of ER chaperone Grp78, a key regulator of ER homeostasis, in alveolar type II (AT2) cells, progenitors of distal lung epithelium, and in IPF lung slice cultures.Measurements and Main Results:Grp78 deletion caused weight loss, mortality, lung inflammation, and spatially heterogeneous fibrosis characterized by fibroblastic foci, hyperplastic AT2 cells, and increased susceptibility of old and male mice, all features of IPF. Fibrosis was more persistent in more severely injured Grp78 knockout (KO) mice. Grp78 KO AT2 cells showed evidence of ER stress, apoptosis, senescence, impaired progenitor capacity, and activation of TGF-ß (transforming growth factor-ß)/SMAD signaling. Glucose-regulated protein 78 is reduced in AT2 cells from old mice and patients with IPF, and ER stress inhibitor tauroursodeoxycholic acid ameliorates ER stress and fibrosis in Grp78 KO mouse and IPF lung slice cultures.Conclusions: These results support a causal role for ER stress and resulting epithelial dysfunction in PF and suggest ER stress as a potential mechanism linking aging to IPF. Modulation of ER stress and chaperone function may offer a promising therapeutic approach for pulmonary fibrosis.


Assuntos
Células Epiteliais Alveolares/metabolismo , Estresse do Retículo Endoplasmático/genética , Proteínas de Choque Térmico/genética , Fibrose Pulmonar/genética , Células-Tronco/metabolismo , Fatores Etários , Células Epiteliais Alveolares/patologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Antioxidantes/farmacologia , Apoptose/genética , Senescência Celular/genética , Dasatinibe/farmacologia , Chaperona BiP do Retículo Endoplasmático , Técnicas de Inativação de Genes , Proteínas de Choque Térmico/metabolismo , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/efeitos dos fármacos , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Inibidores de Proteínas Quinases/farmacologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Quercetina/farmacologia , Quinolinas/farmacologia , Proteínas Smad/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Fator de Transcrição CHOP/efeitos dos fármacos , Fator de Transcrição CHOP/metabolismo , Fator de Crescimento Transformador beta/metabolismo
2.
Int J Cancer ; 143(12): 3169-3180, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30325015

RESUMO

Claudins are a family of transmembrane proteins integral to the structure and function of tight junctions (TJ). Disruption of TJ and alterations in claudin expression are important features of invasive and metastatic cancer cells. Expression of CLDN18.1, the lung-specific isoform of CLDN18, is markedly decreased in lung adenocarcinoma (LuAd). Furthermore, we recently observed that aged Cldn18 -/- mice have increased propensity to develop LuAd. We now demonstrate that CLDN18.1 expression correlates inversely with promoter methylation and with LuAd patient mortality. In addition, when restored in LuAd cells that have lost expression, CLDN18.1 markedly attenuates malignant properties including xenograft tumor growth in vivo as well as cell proliferation, migration, invasion and anchorage-independent colony formation in vitro. Based on high throughput analyses of Cldn18 -/- murine lung alveolar epithelial type II cells, as well as CLDN18.1-repleted human LuAd cells, we hypothesized and subsequently confirmed by Western analysis that CLDN18.1 inhibits insulin-like growth factor-1 receptor (IGF-1R) and AKT phosphorylation. Consistent with recent data in Cldn18 -/- knockout mice, expression of CLDN18.1 in human LuAd cells also decreased expression of transcriptional co-activator with PDZ-binding motif (TAZ) and Yes-associated protein (YAP) and their target genes, contributing to its tumor suppressor activity. Moreover, analysis of LuAd cells in which YAP and/or TAZ are silenced with siRNA suggests that inhibition of TAZ, and possibly YAP, is also involved in CLDN18.1-mediated AKT inactivation. Taken together, these data indicate a tumor suppressor role for CLDN18.1 in LuAd mediated by a regulatory network that encompasses YAP/TAZ, IGF-1R and AKT signaling.


Assuntos
Adenocarcinoma de Pulmão/metabolismo , Claudinas/fisiologia , Neoplasias Pulmonares/metabolismo , Transdução de Sinais/fisiologia , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Animais , Western Blotting , Proliferação de Células , Claudinas/genética , Metilação de DNA , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Invasividade Neoplásica , Metástase Neoplásica , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-yes/metabolismo , Receptor IGF Tipo 1/metabolismo , Transativadores , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional
3.
Cell Stem Cell ; 21(6): 712-714, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29220663

RESUMO

Recently in Stem Cell Reports, Guo et al. (2017) adopted an intermittent reprogramming strategy to generate "induced Progenitor-Like (iPL)" cells, which can maintain lineage commitment while undergoing controlled expansion. The iPLCs were successfully engrafted into a damaged airway, highlighting this significant advancement for regenerative medicine strategies in the lung.


Assuntos
Reprogramação Celular , Medicina Regenerativa , Células Epiteliais , Regeneração , Células-Tronco
4.
Vascul Pharmacol ; 84: 38-46, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27435474

RESUMO

We investigated the vasorelaxant effect of repaglinide and its related signaling pathways using phenylephrine (Phe)-induced pre-contracted aortic rings. Repaglinide induced vasorelaxation in a concentration-dependent manner. The repaglinide-induced vasorelaxation was not affected by removal of the endothelium. In addition, application of a nitric oxide synthase inhibitor (L-NAME) and a small-conductance Ca(2+)-activated K(+) (SKCa) channel inhibitor (apamin) did not alter the vasorelaxant effect of repaglinide on endothelium-intact arteries. Pretreatment with an adenylyl cyclase inhibitor (SQ 22536) or a PKA inhibitor (KT 5720) effectively reduced repaglinide-induced vasorelaxation. Also, pretreatment with a guanylyl cyclase inhibitor (ODQ) or a PKG inhibitor (KT 5823) inhibited repaglinide-induced vasorelaxation. However, pretreatment with a voltage-dependent K(+) (Kv) channel inhibitor (4-AP), ATP-sensitive K(+) (KATP) channel inhibitor (glibenclamide), large-conductance Ca(2+)-activated K(+) (BKCa) channel inhibitor (paxilline), or the inwardly rectifying K(+) (Kir) channel inhibitor (Ba(2+)) did not affect the vasorelaxant effect of repaglinide. Furthermore, pretreatment with a Ca(2+) inhibitor (nifedipine) and a sarco-endoplasmic reticulum Ca(2+)-ATPase (SERCA) inhibitor (thapsigargin) did not affect the vasorelaxant effect of repaglinide. The vasorelaxant effect of repaglinide was not affected by elevated glucose (50mM). Based on these results, we conclude that repaglinide induces vasorelaxation via activation of adenylyl cyclase/PKA and guanylyl cyclase/PKG signaling pathways independently of the endothelium, K(+) channels, Ca(2+) channels, and intracellular Ca(2+) ([Ca(2+)]i).


Assuntos
Carbamatos/farmacologia , Hipoglicemiantes/farmacologia , Piperidinas/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Carbamatos/administração & dosagem , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Glucose/metabolismo , Hipoglicemiantes/administração & dosagem , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fenilefrina/farmacologia , Piperidinas/administração & dosagem , Coelhos , Transdução de Sinais/efeitos dos fármacos , Vasodilatadores/administração & dosagem
5.
Cell Rep ; 12(9): 1385-90, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26299960

RESUMO

Lung disease is a major cause of death in the United States, with current therapeutic approaches serving only to manage symptoms. The most common chronic and life-threatening genetic disease of the lung is cystic fibrosis (CF) caused by mutations in the cystic fibrosis transmembrane regulator (CFTR). We have generated induced pluripotent stem cells (iPSCs) from CF patients carrying a homozygous deletion of F508 in the CFTR gene, which results in defective processing of CFTR to the cell membrane. This mutation was precisely corrected using CRISPR to target corrective sequences to the endogenous CFTR genomic locus, in combination with a completely excisable selection system, which significantly improved the efficiency of this correction. The corrected iPSCs were subsequently differentiated to mature airway epithelial cells where recovery of normal CFTR expression and function was demonstrated. This isogenic iPSC-based model system for CF could be adapted for the development of new therapeutic approaches.


Assuntos
Diferenciação Celular , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Reparo Gênico Alvo-Dirigido/métodos , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Pulmão/citologia , Mutação
6.
Cell Stem Cell ; 16(4): 367-72, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25772073

RESUMO

X-linked Severe Combined Immunodeficiency (SCID-X1) is a genetic disease that leaves newborns at high risk of serious infection and a predicted life span of less than 1 year in the absence of a matched bone marrow donor. The disease pathogenesis is due to mutations in the gene encoding the Interleukin-2 receptor gamma chain (IL-2Rγ), leading to a lack of functional lymphocytes. With the leukemogenic concerns of viral gene therapy there is a need to explore alternative therapeutic options. We have utilized induced pluripotent stem cell (iPSC) technology and genome editing mediated by TALENs to generate isogenic subject-specific mutant and gene-corrected iPSC lines. While the subject-derived mutant iPSCs have the capacity to generate hematopoietic precursors and myeloid cells, only wild-type and gene-corrected iPSCs can additionally generate mature NK cells and T cell precursors expressing the correctly spliced IL-2Rγ. This study highlights the potential for the development of autologous cell therapy for SCID-X1 subjects.


Assuntos
Terapia Genética/métodos , Imunoterapia Adotiva , Células-Tronco Pluripotentes Induzidas/fisiologia , Células Matadoras Naturais/fisiologia , Células Precursoras de Linfócitos T/fisiologia , Regeneração , Medicina Regenerativa , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/terapia , Antígenos CD/metabolismo , Proteínas de Bactérias/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Lactente , Subunidade gama Comum de Receptores de Interleucina/genética , Células Matadoras Naturais/transplante , Mutação/genética , Células Precursoras de Linfócitos T/transplante , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética
7.
Proc Natl Acad Sci U S A ; 111(17): E1723-30, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24706852

RESUMO

Despite therapeutic advancement, pulmonary disease still remains a major cause of morbidity and mortality around the world. Opportunities to study human lung disease either in vivo or in vitro are currently limited. Using induced pluripotent stem cells (iPSCs), we generated mature multiciliated cells in a functional airway epithelium. Robust multiciliogenesis occurred when notch signaling was inhibited and was confirmed by (i) the assembly of multiple pericentrin-stained centrioles at the apical surface, (ii) expression of transcription factor forkhead box protein J1, and (iii) presence of multiple acetylated tubulin-labeled cilia projections in individual cells. Clara, goblet, and basal cells were all present, confirming the generation of a complete polarized epithelial-cell layer. Additionally, cAMP-activated and cystic fibrosis transmembrane regulator inhibitor 172-sensitive cystic fibrosis transmembrane regulator currents were recorded in isolated epithelial cells. Our report demonstrating the generation of mature multiciliated cells in respiratory epithelium from iPSCs is a significant advance toward modeling a number of human respiratory diseases in vitro.


Assuntos
Cílios/metabolismo , Células Epiteliais/citologia , Epitélio/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Pulmão/citologia , Diferenciação Celular , Membrana Celular/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Endoderma/citologia , Células Epiteliais/metabolismo , Humanos , Receptores Notch/metabolismo , Transdução de Sinais
8.
J Biosci ; 38(5): 937-49, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24296897

RESUMO

Protein kinases are one of the largest gene families and have regulatory roles in all aspects of eukaryotic cell function. Modulation of protein kinase activity is a desirable therapeutic approach for a number of human diseases associated with aberrant kinase activity, including cancers, arthritis and cardiovascular disorders. Several strategies have been used to develop specific and selective protein kinase modulators, primarily via inhibition of phosphorylation and down-regulation of kinase gene expression. These strategies are effective at regulating intracellular signalling pathways, but are unfortunately associated with several undesirable effects, particularly those that modulate ion channel function. In fact, the side-effects have precluded these inhibitors from being both useful experimental tools and therapeutically viable. This review focuses on the ion channel side-effects of several protein kinase inhibitors and specifically on those modulating K+, Na+ and Ca2+ ion channels. It is hoped that the information provided with a detailed summary in this review will assist the future development of novel specific and selective compounds targeting protein kinases both for experimental tools and for therapeutic approaches.


Assuntos
Antineoplásicos/efeitos adversos , Doenças Cardiovasculares/induzido quimicamente , Canais Iônicos/metabolismo , Moduladores de Transporte de Membrana/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Animais , Antineoplásicos/uso terapêutico , Relação Dose-Resposta a Droga , Humanos , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico
11.
Am J Physiol Cell Physiol ; 302(2): C405-11, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22031597

RESUMO

Platelet-derived growth factor (PDGF) and its receptor are known to be substantially elevated in lung tissues and pulmonary arterial smooth muscle cells (PASMC) isolated from patients and animals with pulmonary arterial hypertension. PDGF has been shown to phosphorylate and activate Akt and mammalian target of rapamycin (mTOR) in PASMC. In this study, we investigated the role of PDGF-mediated activation of Akt signaling in the regulation of cytosolic Ca(2+) concentration and cell proliferation. PDGF activated the Akt/mTOR pathway and, subsequently, enhanced store-operated Ca(2+) entry (SOCE) and cell proliferation in human PASMC. Inhibition of Akt attenuated the increase in cytosolic Ca(2+) concentration due to both SOCE and PASMC proliferation. This effect correlated with a significant downregulation of stromal interacting molecule (STIM) and Orai, proposed molecular correlates for SOCE in many cell types. The data from this study present a novel pathway for the regulation of Ca(2+) signaling and PASMC proliferation involving activation of Akt in response to upregulated expression of PDGF. Targeting this pathway may lead to the development of a novel therapeutic option for the treatment of pulmonary arterial hypertension.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/citologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Células Cultivadas , Humanos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Proteína ORAI1 , Fosforilação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Molécula 1 de Interação Estromal , Serina-Treonina Quinases TOR/antagonistas & inibidores
12.
Stem Cells ; 29(7): 1158-64, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21544903

RESUMO

By mimicking embryonic development of the hematopoietic system, we have developed an optimized in vitro differentiation protocol for the generation of precursors of hematopoietic lineages and primitive hematopoietic cells from human embryonic stem cells (ESC) and induced pluripotent stem cells (iPSCs). Factors such as cytokines, extra cellular matrix components, and small molecules as well as the temporal association and concentration of these factors were tested on seven different human ESC and iPSC lines. We report the differentiation of up to 84% human CD45+ cells (average 41% ± 16%, from seven pluripotent lines) from the differentiation culture, including significant numbers of primitive CD45+/CD34+ and CD45+/CD34+/CD38- hematopoietic progenitors. Moreover, the numbers of hematopoietic progenitor cells generated, as measured by colony forming unit assays, were comparable to numbers obtained from fresh umbilical cord blood mononuclear cell isolates on a per CD45+ cell basis. Our approach demonstrates highly efficient generation of multipotent hematopoietic progenitors with among the highest efficiencies reported to date (CD45+/CD34+) using a single standardized differentiation protocol on several human ESC and iPSC lines. Our data add to the cumulating evidence for the existence of an in vitro derived precursor to the hematopoietic stem cell (HSC) with limited engrafting ability in transplanted mice but with multipotent hematopoietic potential. Because this protocol efficiently expands the preblood precursors and hematopoietic progenitors, it is ideal for testing novel factors for the generation and expansion of definitive HSCs with long-term repopulating ability.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Diferenciação Celular/fisiologia , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia
13.
Mol Pharmacol ; 79(3): 381-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21127130

RESUMO

Cardiac-directed expression of AC6 has pronounced favorable effects on cardiac function possibly not linked with cAMP production. To determine rigorously whether cAMP generation is required for the beneficial effects of increased AC6 expression, we generated a catalytically inactive AC6 mutant (AC6mut) that has markedly diminished cAMP generating capacity by replacing aspartic acid with alanine at position 426 in the C1 domain (catalytic region) of AC6. Gene transfer of AC6 or AC6mut (adenovirus-mediated) in adult rat cardiac myocytes resulted in similar expression levels and intracellular distribution, but AC6mut expression was associated with marked reduction in cAMP production. Despite marked reduction in cAMP generation, AC6mut influenced intracellular signaling events similarly to that observed after expression of catalytically intact AC6. For example, both AC6 and AC6mut reduced phenylephrine-induced cardiac myocyte hypertrophy and apoptosis (p < 0.001), expression of cardiac ankyrin repeat protein (p < 0.01), and phospholamban (p < 0.05). AC6mut expression, similar to its catalytically intact cohort, was associated with increased Ca2+ transients in cardiac myocytes after isoproterenol stimulation. Many of the biological effects of AC6 expression are replicated by a catalytically inactive AC6 mutant, indicating that the mechanisms for these effects do not require increased cAMP generation.


Assuntos
Adenilil Ciclases/fisiologia , Miócitos Cardíacos/enzimologia , Mutação Puntual/fisiologia , Adenilil Ciclases/biossíntese , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Calcineurina/metabolismo , Cálcio/metabolismo , Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/metabolismo , AMP Cíclico/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipertrofia/enzimologia , Hipertrofia/fisiopatologia , Proteínas Musculares , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Proteínas Nucleares/metabolismo , Fenilefrina/farmacologia , Fosforilação , Mutação Puntual/genética , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/metabolismo , Troponina I/biossíntese , Troponina I/metabolismo
14.
Am J Physiol Cell Physiol ; 298(5): C1217-25, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20181931

RESUMO

Factors contributing to the development of a fibrotic vascular scar and pulmonary vascular remodeling leading to chronic thromboembolic pulmonary hypertension (CTEPH) are still unknown. This study investigates the potential contribution of multipotent progenitor cells and myofibroblasts to the development and progression of CTEPH. Histological examination of endarterectomized tissues from patients with CTEPH identified significant neointimal formation. Morphological heterogeneity was observed in cells isolated from these tissues, including a network-like growth pattern and the formation of colony-forming unit-fibroblast-like colonies (CFU-F). Cells typically coexpressed intermediate filaments vimentin and smooth muscle alpha-actin. Cells were characterized by immunofluorescence and quantitated by fluorescent-activated cell sorting (FACS) for the presence of cell surface markers typical of mesenchymal progenitor cells; cells were >99% CD44(+) CD73(+), CD90(+), CD166(+); >80% CD29(+); 45-99% CD105(+); CD34(-) and CD45(-). Cells were capable of adipogenic and osteogenic differentiation, determined by Oil Red O and Alizarin Red staining, respectively. Additionally, a population of Stro-1(+) cells, a marker of bone marrow-derived stromal cells (4.2%), was sorted by FACS and also capable of adipogenic and osteogenic differentiation. In conclusion, this study is the first to identify a myofibroblast cell phenotype to be predominant within endarterectomized tissues, contributing extensively to the vascular lesion/clot. This cell may arise from transdifferentiation of adventitial fibroblasts or differentiation of mesenchymal progenitor cells. The unique microenvironment created by the stabilized clot is likely a factor in stimulating such cellular changes. These findings will be critical in establishing future studies in the development of novel and much needed therapeutic approaches for pulmonary hypertension.


Assuntos
Hipertensão Pulmonar/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Multipotentes/citologia , Músculo Liso Vascular/citologia , Embolia Pulmonar/patologia , Animais , Diferenciação Celular , Células Cultivadas , Endarterectomia , Humanos , Hipertensão Pulmonar/metabolismo , Masculino , Miócitos de Músculo Liso , Artéria Pulmonar/citologia , Embolia Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley
15.
Am J Physiol Lung Cell Mol Physiol ; 298(4): L548-57, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20139178

RESUMO

Oct-4 is a transcription factor considered to be one of the defining pluripotency markers in embryonic stem cells. Its expression has also been demonstrated in adult stem cells, tumorigenic cells, and, most recently and controversially, in somatic cells. Oct-4 pseudogenes also contribute to carcinogenesis. Oct-4 may be involved in the excessive proliferation of pulmonary arterial smooth muscle cells (PASMC) in patients with idiopathic pulmonary arterial hypertension (IPAH), contributing to the pathogenesis of IPAH. In this study, we show that Oct-4 isoforms are upregulated in IPAH-PASMC. Human embryonic stem cells (H9 line) and human PASMC from normotensive subjects were used throughout the investigation as positive and negative controls. In addition to significant upregulation of Oct-4 in a population of IPAH-PASMC, HIF-2alpha, a hypoxia-inducible transcription factor that has been shown to bind to the Oct-4 promoter and induces its expression and transcriptional activity, was also increased. Interestingly, a substantial upregulation of Oct-4 isoforms and HIF-2alpha was also observed in normal PASMC exposed to chronic hypoxia. In conclusion, the data suggest that both Oct-4 isoforms are upregulated and potentially have a significant role in the development of vascular abnormalities associated with the pathogenesis of IPAH and in pulmonary hypertension triggered by chronic hypoxia.


Assuntos
Hipertensão Pulmonar/genética , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fator 3 de Transcrição de Octâmero/genética , Artéria Pulmonar/patologia , Regulação para Cima/genética , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipóxia Celular/genética , Regulação para Baixo/genética , Humanos , Hipertensão Pulmonar/patologia , Dados de Sequência Molecular , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pseudogenes/genética , Artéria Pulmonar/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L666-76, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19633069

RESUMO

Pulmonary vascular remodeling occurs in patients with chronic thromboembolic pulmonary hypertension (CTEPH). One factor contributing to this vascular wall thickening is the proliferation of pulmonary artery smooth muscle cells (PASMC). Store-operated Ca(2+) entry (SOCE) and cytosolic free Ca(2+) concentration ([Ca(2+)](cyt)) in PASMC are known to be important in cell proliferation and vascular remodeling in pulmonary hypertension. Rapamycin is widely known for its antiproliferative effects in injured coronary arteries. Although several reports have suggested favorable effects of rapamycin in animal models of pulmonary hypertension, no reports have been published to date in human tissues. Here we report that rapamycin has an inhibitory effect on SOCE and an antiproliferative effect on PASMC derived from endarterectomized tissues of CTEPH patients. Cells were isolated from endarterectomized tissues obtained from patients undergoing pulmonary thromboendarterectomy (PTE). Immunohistochemical analysis indicated high deposition of platelet-derived growth factor (PDGF) in tissue sections from PTE tissues and increased PDGF receptor expression. PDGF transiently phosphorylated Akt, mammalian target of rapamycin (mTOR), and p70S6 kinase in CTEPH cells from CTEPH patients. Acute treatment (30 min) with rapamycin (10 nM) slightly increased cyclopiazonic acid (10 microM)-induced Ca(2+) mobilization and significantly reduced SOCE. Chronic treatment (24 h) with rapamycin reduced Ca(2+) mobilization and markedly inhibited SOCE. The inhibitory effects of rapamycin on SOCE were less prominent in control cells. Rapamycin also significantly reduced PDGF-stimulated cell proliferation. In conclusion, the data from this study indicate the importance of the mTOR pathway in the development of pulmonary vascular remodeling in CTEPH and suggest a potential therapeutic benefit of rapamycin (or inhibition of mTOR) in these patients.


Assuntos
Cálcio/metabolismo , Endarterectomia , Hipertensão Pulmonar/metabolismo , Proteínas Quinases/fisiologia , Artéria Pulmonar/cirurgia , Tromboembolia/metabolismo , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citosol/efeitos dos fármacos , Citosol/metabolismo , Imunofluorescência , Humanos , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/cirurgia , Técnicas Imunoenzimáticas , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Tromboembolia/patologia , Tromboembolia/cirurgia
17.
Am J Physiol Lung Cell Mol Physiol ; 296(6): L870-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19286928

RESUMO

Chronic thromboembolic pulmonary hypertension (CTEPH) is characterized by a fibrotic thrombus persisting and obliterating the lumen of pulmonary arteries; its pathogenesis remains poorly defined. This study investigates a potential contribution for progenitor cell types in the development of vascular obliteration and remodeling in CTEPH patients. Endarterectomized tissue from patients undergoing pulmonary thromboendarterectomy was collected and examined for the structure and cellular composition. Our data show an organized fibrin network structure in unresolved thromboemboli and intimal remodeling in vascular wall tissues, characterized by smooth muscle alpha-actin (SM-alphaA)-positive cell proliferation in proximal regions (adjacent to thromboemboli) and neoangiogenesis/recanalization in distal regions (downstream from thromboemboli). Cells that are positively stained with CD34 and fetal liver kinase-1 (Flk-1) (CD34(+)Flk-1(+)) were identified in both the proximal and distal vascular tissues; a subpopulation of CD34(+)Flk-1(+)CD133(+) cells were further identified by immunostaining. Triple-positive cells are indicative of a population of putative endothelial progenitor cells or potential colony-forming units of endothelial cells. In addition, inflammatory cells (CD45(+)) and collagen-secreting cells (procollagen-1(+)) were detected in the proximal vascular wall. Some of the CD34(+) cells in CTEPH cells isolated from proximal regions were also positive for SM-alphaA. Our data indicate that putative progenitor cell types are present in the neointima of occluded vessels of CTEPH patients. It is possible that the microenvironment provided by thromboemboli may promote these putative progenitor cells to differentiate and enhance intimal remodeling.


Assuntos
Endotélio Vascular/citologia , Hipertensão Pulmonar/patologia , Células-Tronco Mesenquimais/citologia , Artéria Pulmonar/patologia , Embolia Pulmonar/patologia , Antígeno AC133 , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos CD34/genética , Antígenos CD34/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Doença Crônica , Endarterectomia , Células Endoteliais/citologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/cirurgia , Células-Tronco Mesenquimais/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/cirurgia , Embolia Pulmonar/metabolismo , Embolia Pulmonar/cirurgia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 296(3): L347-60, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19098127

RESUMO

Mitochondria are proposed to be a major oxygen sensor in hypoxic pulmonary vasoconstriction (HPV), a unique response of the pulmonary circulation to low oxygen tension. Mitochondrial factors including reactive oxygen species, cytochrome c, ATP, and magnesium are potent modulators of voltage-gated K(+) (K(v)) channels in the plasmalemmal membrane of pulmonary arterial (PA) smooth muscle cells (PASMCs). Mitochondria have also been found close to the plasmalemmal membrane in rabbit main PA smooth muscle sections. Therefore, we hypothesized that differences in mitochondria localization in rat PASMCs and systemic mesenteric arterial smooth muscle cells (MASMCs) may contribute to the divergent oxygen sensitivity in the two different circulations. Cellular localization of mitochondria was compared with immunofluorescent labeling, and differences in functional coupling between mitochondria and K(v) channels was evaluated with the patch-clamp technique and specific mitochondrial inhibitors antimycin A (acting at complex III of the mitochondrial electron transport chain) and oligomycin A (which inhibits the ATP synthase). It was found that mitochondria were located significantly closer to the plasmalemmal membrane in PASMCs compared with MASMCs. Consistent with these findings, the effects of the mitochondrial inhibitors on K(v) current (I(Kv)) were significantly more potent in PASMCs than in MASMCs. The cytoskeletal disruptor cytochalasin B (10 microM) also altered mitochondrial distribution in PASMCs and significantly attenuated the effect of antimycin A on the voltage-dependent parameters of I(Kv). These findings suggest a greater structural and functional coupling between mitochondria and K(v) channels specifically in PASMCs, which could contribute to the regulation of PA excitability in HPV.


Assuntos
Artérias Mesentéricas/metabolismo , Artérias Mesentéricas/ultraestrutura , Mitocôndrias/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/ultraestrutura , Animais , Antimicina A/farmacologia , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Magnésio/metabolismo , Masculino , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/ultraestrutura , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/ultraestrutura , Oligomicinas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Circulação Pulmonar/fisiologia , Ratos , Ratos Wistar , Retículo Sarcoplasmático/ultraestrutura , Circulação Esplâncnica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA