Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Aging (Albany NY) ; 11(1): 209-229, 2019 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-30636723

RESUMO

Atp8b1 (ATPase, aminophospholipid transporter, class I, type 8B, member 1) is a cardiolipin transporter in the apical membrane of lung epithelial cells. While the role of Atp8b1 in pneumonia-induced acute lung injury (ALI) has been well studied, its potential role in oxidative stress-induced ALI is poorly understood. We herein show that Atp8b1G308V/G308V mice under hyperoxic conditions display exacerbated cell apoptosis at alveolar epithelium and aberrant proliferation of club cells at bronchiolar epithelium. This hyperoxia-induced ambivalent response in Atp8b1G308V/G308V lungs was followed by patchy distribution of non-uniform interstitial fibrosis at late recovery phase under normoxia. Since this club cell abnormality is commonly observed between Atp8b1G308V/G308V lungs under hyperoxic conditions and IPF lungs, we characterized this mouse fibrosis model focusing on club cells. Intriguingly, subcellular morphological analysis of IPF lungs, using transmission electron microscopy (TEM), revealed that metaplastic bronchiolar epithelial cells in fibrotic lesions and deformed type II alveolar epithelial cells (AECs) in alveoli with mild fibrosis, have common morphological features including cytoplasmic vacuolation and dysmorphic lamellar bodies. In conclusion, the combination of Atp8b1 mutation and hyperoxic insult serves as a novel platform to study unfocused role of club cells in IPF.


Assuntos
Adenosina Trifosfatases/metabolismo , Oxigênio/toxicidade , Proteínas de Transferência de Fosfolipídeos/metabolismo , Fibrose Pulmonar/etiologia , Adenosina Trifosfatases/genética , Animais , Morte Celular , Proliferação de Células , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Mutação , Estresse Oxidativo , Proteínas de Transferência de Fosfolipídeos/genética , Alvéolos Pulmonares/citologia , Uteroglobina/genética , Uteroglobina/metabolismo
2.
Aging (Albany NY) ; 8(11): 3091-3109, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27899769

RESUMO

Idiopathic pulmonary fibrosis (IPF) is an age-related multifactorial disease featuring non-uniform lung fibrosis. The decisive cellular events at early stages of IPF are poorly understood. While the involvement of club cells in IPF pathogenesis is unclear, their migration has been associated with lung fibrosis. In this study, we labeled club cells immunohistochemically in IPF lungs using a club cell marker Claudin-10 (Cldn10), a unique protein based on the recent report which demonstrated that the appearance of Cldn10 in developing and repairing lungs precedes other club cell markers including club cell secretory protein (CCSP). Cldn10-positive cells in IPF lungs displayed marked pleomorphism and were found in varied arrangements, suggesting their phenoconversion. These results were corroborated by immunogold labeling for Cldn10. Further, immunohistochemical double-labeling for Cldn10 and α-smooth muscle actin (α-SMA) demonstrated that aberrant α-SMA signals are frequently encountered near disorganized Cldn10-positive cells in hyperplastic bronchiolar epithelium and thickened interstitium of IPF lungs. Collectively, these data indicate that club cells actively participate in the initiation and progression of IPF through phenoconversion involving the acquisition of proliferative and migratory abilities. Thus, our new findings open the possibility for club cell-targeted therapy to become a strategic option for the treatment of IPF.


Assuntos
Actinas/metabolismo , Células Epiteliais Alveolares/metabolismo , Movimento Celular , Claudinas/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Células Epiteliais Alveolares/citologia , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia
3.
PLoS One ; 11(1): e0147652, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26807721

RESUMO

Apoptosis signal-regulating kinase 1 (ASK1), a member of the MAPK kinase kinase kinase (MAP3K) family, is activated by various stimuli, which include oxidative stress, endoplasmic reticulum (ER) stress, calcium influx, DNA damage-inducing agents and receptor-mediated signaling through tumor necrosis factor receptor (TNFR). Inspiration of a high concentration of oxygen is a palliative therapy which counteracts hypoxemia caused by acute lung injury (ALI)-induced pulmonary edema. However, animal experiments so far have shown that hyperoxia itself could exacerbate ALI through reactive oxygen species (ROS). Our previous data indicates that ASK1 plays a pivotal role in hyperoxia-induced acute lung injury (HALI). However, it is unclear whether or not deletion of ASK1 in vivo protects against HALI. In this study, we investigated whether ASK1 deletion would lead to attenuation of HALI. Our results show that ASK1 deletion in vivo significantly suppresses hyperoxia-induced elevation of inflammatory cytokines (i.e. IL-1ß and TNF-α), cell apoptosis in the lung, and recruitment of immune cells. In summary, the results from the study suggest that deletion of ASK1 in mice significantly inhibits hyperoxic lung injury.


Assuntos
Lesão Pulmonar Aguda/genética , Hiperóxia/complicações , Pulmão/metabolismo , MAP Quinase Quinase Quinase 5/genética , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Animais , Apoptose/genética , Epitélio/metabolismo , Epitélio/patologia , Hiperóxia/genética , Hiperóxia/patologia , Interleucina-1beta/metabolismo , Pulmão/patologia , MAP Quinase Quinase Quinase 5/metabolismo , Camundongos , Camundongos Knockout , Fator de Necrose Tumoral alfa/metabolismo
4.
PLoS One ; 10(8): e0136755, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26317859

RESUMO

BACKGROUND: Inflammation is a key hallmark of ALI and is mediated through ungoverned cytokine signaling. One such cytokine, interleukin-1beta (IL-1ß) has been demonstrated to be the most bioactive cytokine in ALI patients. Macrophages are the key players responsible for IL-1ß secretion into the alveolar space. Following the binding of IL-1ß to its receptor, "activated" alveolar epithelial cells show enhanced barrier dysfunction, adhesion molecule expression, cytokine secretion, and leukocyte attachment. More importantly, it is an important communication molecule between the macrophage and alveolar epithelium. While the molecular determinants of this inflammatory event have been well documented, endogenous resolution processes that decrease IL-1ß secretion and resolve alveolar epithelial cell activation and tissue inflammation have not been well characterized. Lipid mediator Aspirin-Triggered Resolvin D1 (AT-RvD1) has demonstrated potent pro-resolutionary effects in vivo models of lung injury; however, the contribution of the alveoli to the protective benefits of this molecule has not been well documented. In this study, we demonstrate that AT-RvD1 treatment lead to a significant decrease in oxidant induced macrophage IL-1ß secretion and production, IL-1ß-mediated cytokine secretion, adhesion molecule expression, leukocyte adhesion and inflammatory signaling. METHODS: THP-1 macrophages were treated with hydrogen peroxide and extracellular ATP in the presence or absence of AT-RvD1 (1000-0.1 nM). A549 alveolar-like epithelial cells were treated with IL-1ß (10 ng/mL) in the presence or absence of AT-RvD1 (0.1 µM). Following treatment, cell lysate and cell culture supernatants were collected for Western blot, qPCR and ELISA analysis of pro-inflammatory molecules. Functional consequences of IL-1ß induced alveolar epithelial cell and macrophage activation were also measured following treatment with IL-1ß ± AT-RvD1. RESULTS: Results demonstrate that macrophages exposed to H2O2 and ATP in the presence of resolvins show decreased IL-1ß production and activity. A549 cells treated with IL-1ß in the presence of AT-RvD1 show a reduced level of proinflammatory cytokines IL-6 and IL-8. Further, IL-1ß-mediated adhesion molecule expression was also reduced with AT-RvD1 treatment, which was correlated with decreased leukocyte adhesion. AT-RvD1 treatment demonstrated reduced MAP-Kinase signaling. Taken together, our results demonstrate AT-RvD1 treatment reduced IL-1ß-mediated alveolar epithelial cell activation. This is a key step in unraveling the protective effects of resolvins, especially AT-RvD1, during injury.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Células Epiteliais/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Lesão Pulmonar Aguda/patologia , Linhagem Celular Tumoral , Células Epiteliais/patologia , Humanos , Macrófagos/patologia , Alvéolos Pulmonares , Transdução de Sinais/efeitos dos fármacos
5.
Cell Physiol Biochem ; 36(5): 2012-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26202361

RESUMO

BACKGROUND: Neuregulin (NRG)-1-human epidermal receptor (HER)-2 signaling pathway is a key regulator of IL-1ß-mediated pulmonary inflammation and epithelial permeability. The inflammasome is a newly discovered molecular platform required for caspase-1 activation and maturation of IL-1ß. However, the role of the inflammasome in NRG-1-HER2 signaling-mediated alveolar cell permeability is unknown. METHODS: The inflammasome was activated or inhibited in THP-1 cells; supernatants from these cells were added to A549 cells and human small airway epithelial cells (HSAEC). The protein expression of NRG-1 and phospho-HER2 (pHER2) were measured by Western blot analysis and epithelial permeability was measured using Lucifer yellow dye. RESULTS: Results reveal that alveolar permeability in A549 cells and HSAEC is increased when treated with supernatants of inflammasome-activated THP-1 cells. Alveolar permeability is significantly suppressed when treated with supernatant of inflammasome-inhibited THP-1 cells. Inflammasome-mediated permeability is decreased when A549 cells and HSAEC are pretreated with IL-1ß receptor antagonist (IL-1ßRA). In addition, HER2 kinase inhibitor AG825 or NRG-1 inhibitor TAPI inhibits inflammasome-mediated permeability in A549 cells and HSAEC demonstrating critical roles of IL-1ß, NRG-1, and HER2 in inflammasome-mediated alveolar permeability. CONCLUSION: These findings suggest that inflammasome-induced alveolar cell permeability is mediated by NRG-1/HER2 signaling through IL-1ß regulation.


Assuntos
Inflamassomos , Neuregulina-1/metabolismo , Linhagem Celular Tumoral , Genes erbB-2 , Humanos , Interleucina-1beta/metabolismo , Alvéolos Pulmonares
6.
Am J Respir Cell Mol Biol ; 53(3): 422-35, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25647402

RESUMO

Acute lung injury (ALI), which presents as acute respiratory failure, is a major clinical problem that requires aggressive care, and patients who require prolonged oxygen exposure are at risk of developing this disease. Although molecular determinants of ALI have been reported, the molecules involved in disease catabasis associated with oxygen toxicity have not been well studied. It has been reported that lung mucosa is rich in omega-3 fatty acid dicosahexanoic acid (DHA), which has antiinflammatory properties. Aspirin-triggered resolvin D1 (AT-RvD1) is a potent proresolution metabolite of DHA that can curb the inflammatory effects in various acute injuries, yet the effect of AT-RvD1 on hyperoxic acute lung injury (HALI) or in the oxygen toxicity setting in general has not been investigated. The effects of AT-RvD1 on HALI were determined for the first time in 8- to 10-week-old C57BL/6 mice that were exposed to hyperoxia (≥95% O2) for 48 hours. Mice were given AT-RvD1 (100 ng) in saline or a saline vehicle for 24 hours in normoxic (≈21% O2) conditions after hyperoxia. Lung tissue and bronchoalveolar lavage (BAL) fluid were collected for analysis associated with proinflammatory signaling and lung inflammation. AT-RvD1 treatment resulted in reduced oxidative stress, increased glutathione production, and significantly decreased tissue inflammation. AT-RvD1 treatment also significantly reduced the lung wet/dry ratio, protein in BAL fluid, and decreased apoptotic and NF-κB signaling. These results show that AT-RvD1 curbs oxygen-induced lung edema, permeability, inflammation, and apoptosis and is thus an effective therapy for prolonged hyperoxia exposure in this murine model.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Ácidos Docosa-Hexaenoicos/fisiologia , Hiperóxia/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/metabolismo , Resistência das Vias Respiratórias , Animais , Apoptose , Avaliação Pré-Clínica de Medicamentos , Hiperóxia/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Estresse Oxidativo
7.
Am J Physiol Cell Physiol ; 306(11): C999-C1007, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24696144

RESUMO

Hyperoxic acute lung injury (HALI) is characterized by inflammation and epithelial cell death. CLOCK genes are master regulators of circadian rhythm also implicated in inflammation and lung diseases. However, the relationship of CLOCK genes in hyperoxia-induced lung injury has not been studied. This study will determine if HALI alters CLOCK gene expression. To test this, wild-type and NALP3(-/-) mice were exposed to room air or hyperoxia for 24, 48, or 72 h. In addition, mice were exposed to different concentrations of hyperoxia (50, 75, or 100% O2) or room air for 72 h. The mRNA and protein levels of lung CLOCK genes, based on quantitative PCR and Western blot analysis, respectively, and their target genes are significantly elevated in mice exposed to hyperoxia compared with controls. Alterations in CLOCK genes are associated with increased inflammatory markers in bronchoalveolar lavage fluid of hyperoxic mice compared with controls. Histological examination of mice lungs exposed to hyperoxia show increased inflammation and alveolar congestion compared with controls. Our results indicate sequential increase in CLOCK gene expression in lungs of mice exposed to hyperoxia compared with controls. Additionally, data suggest a dose-dependent increase in CLOCK gene expression with increased oxygen concentrations. To validate if the expression changes related to CLOCK genes are indeed associated with inflammation, NALP3(-/-) was introduced to analyze loss of function in inflammation. Western blot analysis showed significant CLOCK gene downregulation in NALP3(-/-) mice compared with wild-type controls. Together, our results demonstrate that hyperoxia-mediated lung inflammation is associated with alterations in CLOCK gene expression.


Assuntos
Proteínas CLOCK/biossíntese , Regulação da Expressão Gênica , Hiperóxia/metabolismo , Lesão Pulmonar/metabolismo , Animais , Proteínas CLOCK/genética , Hiperóxia/genética , Hiperóxia/patologia , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
8.
Am J Physiol Cell Physiol ; 305(2): C182-9, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23636457

RESUMO

Inspiration of a high concentration of oxygen, a therapy for acute lung injury (ALI), could unexpectedly lead to reactive oxygen species (ROS) production and hyperoxia-induced acute lung injury (HALI). Nucleotide-binding domain and leucine-rich repeat PYD-containing protein 3 (NLRP3) senses the ROS, triggering inflammasome activation and interleukin-1ß (IL-1ß) production and secretion. However, the role of NLRP3 inflammasome in HALI is unclear. The main aim of this study is to determine the effect of NLRP3 gene deletion on inflammatory response and lung epithelial cell death. Wild-type (WT) and NLRP3(-/-) mice were exposed to 100% O2 for 48-72 h. Bronchoalveolar lavage fluid and lung tissues were examined for proinflammatory cytokine production and lung inflammation. Hyperoxia-induced lung pathological score was suppressed in NLRP3(-/-) mice compared with WT mice. Hyperoxia-induced recruitment of inflammatory cells and elevation of IL-1ß, TNFα, macrophage inflammatory protein-2, and monocyte chemoattractant protein-1 were attenuated in NLRP3(-/-) mice. NLRP3 deletion decreased lung epithelial cell death and caspase-3 levels and a suppressed NF-κB levels compared with WT controls. Taken together, this research demonstrates for the first time that NLRP3-deficient mice have suppressed inflammatory response and blunted lung epithelial cell apoptosis to HALI.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/prevenção & controle , Proteínas de Transporte/metabolismo , Hiperóxia/complicações , Oxigênio/efeitos adversos , Lesão Pulmonar Aguda/genética , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Proteínas de Transporte/genética , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Fragmentação do DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Predisposição Genética para Doença , Hiperóxia/metabolismo , Inflamassomos/efeitos adversos , Inflamassomos/metabolismo , Interleucina-1beta/análise , Interleucina-6/análise , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Oxigênio/administração & dosagem , Oxigênio/uso terapêutico , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
10.
PLoS One ; 7(10): e46808, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071643

RESUMO

Pulmonary Arterial Hypertension (PAH) is a progressive devastating disease characterized by excessive proliferation of the Pulmonary Arterial Smooth Muscle Cells (PASMCs). Studies suggest that PAH and cancers share an apoptosis-resistant state featuring excessive cell proliferation. MicroRNA-206 (miR-206) is known to regulate proliferation and is implicated in various types of cancers. However, the role of miR-206 in PAH has not been studied. In this study, it is hypothesized that miR-206 could play a role in the proliferation of PASMCs. In the present study, the expression patterns of miR-206 were investigated in normal and hypertensive mouse PASMCs. The effects of miR-206 in modulating cell proliferation, apoptosis and smooth muscle cell markers in human pulmonary artery smooth muscle cells (hPASMCs) were investigated in vitro. miR-206 expression in mouse PASMCs was correlated with an increase in right ventricular systolic pressure. Reduction of miR-206 levels in hPASMCs causes increased proliferation and reduced apoptosis and these effects were reversed by the overexpression of miR-206. miR-206 over expression also increased the levels of smooth muscle cell differentiation markers α-smooth muscle actin and calponin implicating its importance in the differentiation of SMCs. miR-206 overexpression down regulated Notch-3 expression, which is key a factor in PAH development. These results suggest that miR-206 is a potential regulator of proliferation, apoptosis and differentiation of PASMCs, and that it could be used as a novel treatment strategy in PAH.


Assuntos
Diferenciação Celular/genética , Proliferação de Células , MicroRNAs/genética , Miócitos de Músculo Liso/metabolismo , Animais , Apoptose/genética , Western Blotting , Movimento Celular/genética , Células Cultivadas , Regulação para Baixo , Hipertensão Pulmonar Primária Familiar , Feminino , Perfilação da Expressão Gênica , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Hipóxia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/citologia , Artéria Pulmonar/citologia , Receptor Notch3 , Receptores Notch/genética , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Biochem Biophys Res Commun ; 426(2): 203-8, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22940131

RESUMO

Acute lung injury (ALI) is a devastating disease characterized by pulmonary edema. Removal of edema from the air spaces of lung is a critical function of the epithelial sodium channel (ENaC) in ALI. The molecular mechanisms behind resolution of pulmonary edema are incompletely understood. MicroRNA's (miRNA) are crucial gene regulators and are dysregulated in various diseases including ALI. Recent studies suggest that microRNA-16 (miR-16) targets serotonin transporter (SERT) involved in the serotonin (5-HT) transmitter system. Alterations in serotonin levels have been reported in various pulmonary diseases. However, the role of miR-16 on its target SERT, and ENaC, a key ion channel involved in the resolution of pulmonary edema, have not been studied. In the present study, the expression patterns of miR-16, SERT, ENaC and serotonin were investigated in mice exposed to room air and hyperoxia. The effects of miR-16 overexpression on ENaC, SERT, TGF-ß and Nedd4 in human alveolar epithelial cells were analyzed. miR-16 and ENaC were downregulated in mice exposed to hyperoxia. miR-16 downregulation in mouse lung was correlated with an increase in SERT expression and pulmonary edema. Overexpression of miR-16 in human alveolar epithelial cells (A549) suppressed SERT and increased ENaCß levels when compared to control-vector transfected cells. In addition, miR-16 over expression suppressed TGFß release, a critical inhibitor of ENaC. Interestingly Nedd4, a negative regulator of ENaC remained unaltered in miR-16 over expressed A549 cells when compared to controls. Taken together, our data suggests that miR-16 upregulates ENaC, a major sodium channel involved in resolution of pulmonary edema in ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Canais Epiteliais de Sódio/metabolismo , MicroRNAs/metabolismo , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/metabolismo , Aerobiose , Animais , Linhagem Celular Tumoral , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Canais Epiteliais de Sódio/genética , Humanos , Camundongos , MicroRNAs/genética , Ubiquitina-Proteína Ligases Nedd4 , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
12.
PLoS One ; 7(5): e37712, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22662197

RESUMO

INTRODUCTION: Pulmonary Arterial Hypertension (PAH) is a progressively devastating disease characterized by excessive proliferation of the Pulmonary Arterial Smooth Muscle Cells (PASMCs). Studies suggest that PAH and cancers share an apoptosis-resistant state featuring excessive cell proliferation. The proliferation of cancer cells is mediated by increased expression of Enhancer of Zeste Homolog 2 (EZH2), a mammalian histone methyltransferase that contributes to the epigenetic silencing of target genes. However, the role of EZH2 in PAH has not been studied. In this study, it is hypothesized that EZH2 could play a role in the proliferation of PASMCs. METHODS: In the present study, the expression patterns of EZH2 were investigated in normal and hypertensive mouse PASMCs. The effects of EZH2 overexpression on the proliferation of human PASMCs were tested. PASMCs were transfected with EZH2 or GFP using nucleofector system. After transfection, the cells were incubated for 48 hours at 37°C. Proliferation and cell cycle analysis were performed using flow cytometry. Apoptosis of PASMCs was determined using annexin V staining and cell migration was tested by wound healing assay. RESULTS: EZH2 protein expression in mouse PASMCs were correlated with an increase in right ventricular systolic pressure and Right Ventricular Hypertrophy (RVH). The overexpression of EZH2 in human PASMCs enhances proliferation, migration, and decrease in the rate of apoptosis when compared to GFP-transfected cells. In the G2/M phase of the EZH2 transfected cells, there was a 3.5 fold increase in proliferation, while there was a significant decrease in the rate of apoptosis of PASMCs, when compared to control. CONCLUSION: These findings suggest that EZH2 plays a role in the migration and proliferation of PASMCs, which is a major hallmark in PAH. It also suggests that EZH2 could play a role in the development of PAH and can serve as a potential target for new therapies for PAH.


Assuntos
Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Artéria Pulmonar/metabolismo , Animais , Apoptose/genética , Movimento Celular/genética , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Proteína Potenciadora do Homólogo 2 de Zeste , Expressão Gênica , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipóxia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Complexo Repressor Polycomb 2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA