Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 844
Filtrar
1.
Eur J Pharmacol ; 972: 176547, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38561103

RESUMO

Idiopathic pulmonary fibrosis (IPF) associated to pulmonary hypertension (PH) portends a poor prognosis, characterized by lung parenchyma fibrosis and pulmonary artery remodeling. Serum and parenchyma levels of Interleukin 11 (IL-11) are elevated in IPF-PH patients and contributes to pulmonary artery remodeling and PH. However, the effect of current approved therapies against IPF in pulmonary artery remodeling induced by IL-11 is unknown. The aim of this study is to analyze the effects of nintedanib and pirfenidone on pulmonary artery endothelial and smooth muscle cell remodeling induced by IL-11 in vitro. Our results show that nintedanib (NTD) and pirfenidone (PFD) ameliorates endothelial to mesenchymal transition (EnMT), pulmonary artery smooth muscle cell to myofibroblast-like transformation and pulmonary remodeling in precision lung cut slices. This study provided also evidence of the inhibitory effect of PFD and NTD on IL-11-induced endothelial and muscle cells proliferation and senescence. The inhibitory effect of these drugs on monocyte arrest and angiogenesis was also studied. Finally, we observed that IL-11 induced canonical signal transducer and activator of transcription 3 (STAT3) and non-canonical mitogen-activated protein kinase 1/2 (ERK1/2) phosphorylation, but, PFD and NTD only inhibited ERK1/2 phosphorylation. Therefore, this study provided evidence of the inhibitory effect of NTD and PFD on markers of pulmonary artery remodeling induced by IL-11.


Assuntos
Proliferação de Células , Células Endoteliais , Indóis , Interleucina-11 , Miócitos de Músculo Liso , Artéria Pulmonar , Piridonas , Fator de Transcrição STAT3 , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/citologia , Interleucina-11/metabolismo , Indóis/farmacologia , Animais , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fator de Transcrição STAT3/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Piridonas/farmacologia , Proliferação de Células/efeitos dos fármacos , Ratos , Humanos , Masculino , Senescência Celular/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/patologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Remodelação Vascular/efeitos dos fármacos
2.
Int Immunopharmacol ; 102: 108379, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34865992

RESUMO

BACKGROUND: Sex hormone paradox is a crucial but unresolved issue in the field of pulmonary artery hypertension (PAH), and is thought to be related to different pathogenic factors. Inflammation is one of pathological mechanisms of PAH development. However, effects of sex hormones on the pulmonary vasculature under the condition of inflammation are still elusive. METHODS: Interleukin-6 (IL-6) was used as a representative inflammatory stimulator. Effects of 17ß-estradiol or progesterone on human pulmonary artery smooth muscle cells (PASMCs) were measured under the condition of IL-6. Cell functions of proliferation and migration were measured by Alarmar Blue, EdU assay, wound-healing assay and transwell chambers. We explored further mechanisms using western blot, immunofluorescence, co-immunoprecipitation, qPCR and chromatin immunoprecipitation. RESULTS: Our results revealed that IL-6 promoted the proliferation of PASMCs, but progesterone could reverse the adverse effect of IL-6. The protective effect was dependent on progesterone receptor (PGR). By interacting with signal transducer and activator of transcription 3 (STAT3), activated PGR could reduce the IL-6-induced nuclear translocation of STAT3 and prevent STAT3-chromatin binding in PASMCs, leading to the decreased transcription of downstream CCND1 and BCL2. Alternatively, progesterone slightly decreased the phosphorylation of pro-proliferative Erk1/2 and Akt kinases and upregulated the anti-proliferative pSmad1-Id1/2 axis in IL-6-incubated PASMCs. CONCLUSIONS: Progesterone played a protective role on PASMCs in the context of IL-6, by blocking the functions of STAT3. Our findings might assist in explaining the clinical phenomenon of better prognosis for women with PAH.


Assuntos
Miócitos de Músculo Liso/efeitos dos fármacos , Progesterona/farmacologia , Substâncias Protetoras/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Estradiol/farmacologia , Humanos , Interleucina-6/imunologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Artéria Pulmonar/citologia , Fator de Transcrição STAT3/metabolismo
3.
J Biochem Mol Toxicol ; 35(10): e22879, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34369038

RESUMO

Growth hormone-releasing hormone (GHRH) is a hypothalamic hormone, which regulates the secretion of growth hormone (GH) from the anterior pituitary gland. The effects of GHRH extend beyond the GH-insulin-like growth factor I axis, and that neuropeptide has been involved in the potentiation of several malignancies and other inflammatory disorders. The development of GHRH antagonists (GHRHAnt) delivers an exciting possibility to counteract the pathogenesis of the GHRH-related effects in human pathophysiology, especially when considered that GHRHAnt support endothelial barrier integrity. Those GHRHAnt-mediated effects are exerted at least in part due to the suppression of major inflammatory pathways, and the modulation of major cytoskeletal components. In the present study, we measured the production of reactive oxygen species (ROS) in bovine pulmonary artery endothelial cells, human cerebral microvascular endothelial cells, and human lung microvascular endothelial cells exposed to GHRH or a commercially available GHRHAnt. Our findings reveal the antioxidative effects of GHRHAnt in all three cell lines, which express GHRH receptors. The redox status of NIH/3T3 cells, which do not produce GHRH receptors, was not significantly affected by GHRH or GHRHAnt. Hence, the application of GHRHAnt in pathologies related to increased ROS production should be further investigated.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Hormônio Liberador de Hormônio do Crescimento/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Bovinos , Linhagem Celular Transformada , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos , Células NIH 3T3 , Artéria Pulmonar/citologia , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo
4.
J Mol Cell Cardiol ; 161: 9-22, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34339758

RESUMO

OBJECTIVE: Pulmonary vascular remodeling due to excessive growth factor production and pulmonary artery smooth muscle cells (PASMCs) proliferation is the hallmark feature of pulmonary arterial hypertension (PAH). Recent studies suggest that miR-663 is a potent modulator for tumorigenesis and atherosclerosis. However, whether miR-663 involves in pulmonary vascular remodeling is still unclear. METHODS AND RESULTS: By using quantitative RT-PCR, we found that miR-663 was highly expressed in normal human PASMCs. In contrast, circulating level of miR-663 dramatically reduced in PAH patients. In addition, in situ hybridization showed that expression of miR-663 was decreased in pulmonary vasculature of PAH patients. Furthermore, MTT and cell scratch-wound assay showed that transfection of miR-663 mimics significantly inhibited platelet derived growth factor (PDGF)-induced PASMCs proliferation and migration, while knockdown of miR-663 expression enhanced these effects. Mechanistically, dual-luciferase reporter assay revealed that miR-663 directly targets the 3'UTR of TGF-ß1. Moreover, western blots and ELISA results showed that miR-663 decreased PDGF-induced TGF-ß1 expression and secretion, which in turn suppressed the downstream smad2/3 phosphorylation and collagen I expression. Finally, intratracheal instillation of adeno-miR-663 efficiently inhibited the development of pulmonary vascular remodeling and right ventricular hypertrophy in monocrotaline (MCT)-induced PAH rat models. CONCLUSION: These results indicate that miR-663 is a potential biomarker for PAH. MiR-663 decreases PDGF-BB-induced PASMCs proliferation and prevents pulmonary vascular remodeling and right ventricular hypertrophy in MCT-PAH by targeting TGF-ß1/smad2/3 signaling. These findings suggest that miR-663 may represent as an attractive approach for the diagnosis and treatment for PAH.


Assuntos
MicroRNAs/sangue , Hipertensão Arterial Pulmonar/genética , Fator de Crescimento Transformador beta1/metabolismo , Remodelação Vascular/genética , Idoso , Animais , Becaplermina/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Monocrotalina/toxicidade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Hipertensão Arterial Pulmonar/induzido quimicamente , Hipertensão Arterial Pulmonar/metabolismo , Artéria Pulmonar/citologia , Ratos Sprague-Dawley , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/genética , Remodelação Vascular/efeitos dos fármacos
5.
Artigo em Inglês | MEDLINE | ID: mdl-34403986

RESUMO

INTRODUCTION: Prostacyclin (PGI2) is synthetized by PGI2 synthase (PGIS) and induces vasorelaxation via activation of cyclic AMP (cAMP) generating IP-receptor. Several components of the PGI2 signaling pathway are reduced in patients with pulmonary hypertension (PH). AIM: To study the effect of 17ß-estradiol (E2) on the PGI2 signaling pathway in human pulmonary arteries (HPA) and in their smooth muscle cells (hPASMC) derived from Group-3 PH and non-PH patients. METHODS: Following E2-treatments of isolated HPA and cultured hPASMC, we measured: 6-keto-Prostaglandin F1α (PGI2 stable metabolite) by ELISA, PGIS and IP protein levels by Western blot and HPA vasorelaxations with an organ bath system. RESULTS: Incubation with E2 (24/48 h, doses ≥ 10 nM) significantly increased the expression of PGIS in hPASMC derived from both PH (65-98%) and non-PH (21-33%) patients, whereas incubation with E2 (2 h, 0.1 and 1 µM) increased 6-keto-PGF1α production in HPA from Group-3 PH patients only, and did not affect 6-keto-PGF1α production in hPASMC from either non-PH or Group-3 PH patients. Increases in IP receptor expression were observed following 10 mM E2-treatment of hPASMC from non-PH (33% after 48 h) and Group-3 PH (23% after 24 h) patient lungs. Finally, preincubation with 100 nM E2 significantly increased arachidonic acid-induced vasorelaxation of HPA from non-PH patient lungs but not of HPA from Group-3 PH patient lungs. CONCLUSION: E2-treatment may help to restore the PGI2-pathway in Group-3 PH.


Assuntos
6-Cetoprostaglandina F1 alfa/metabolismo , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/farmacologia , Hipertensão Pulmonar/metabolismo , Oxirredutases Intramoleculares/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Artéria Pulmonar/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Anti-Hipertensivos/farmacologia , Ácido Araquidônico/farmacologia , Estudos de Casos e Controles , Sistema Enzimático do Citocromo P-450/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Feminino , Humanos , Hipertensão Pulmonar/fisiopatologia , Oxirredutases Intramoleculares/metabolismo , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia
6.
Am J Physiol Heart Circ Physiol ; 321(3): H542-H557, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34296965

RESUMO

Pulmonary arterial hypertension (PAH) is a fatal cardiopulmonary disease characterized by increased vascular cell proliferation with apoptosis resistance and occlusive remodeling of the small pulmonary arteries. The Notch family of proteins subserves proximal signaling of an evolutionarily conserved pathway that effects cell proliferation, fate determination, and development. In endothelial cells (ECs), Notch receptor 2 (Notch2) was shown to promote endothelial apoptosis. However, a pro- or antiproliferative role for Notch2 in pulmonary endothelial proliferation and ensuing PAH is unknown. We postulated that suppressed Notch2 signaling drives pulmonary endothelial proliferation in the context of PAH. We observed that levels of Notch2 are ablated in lungs from PAH subjects compared with non-PAH controls. Notch2 expression was attenuated in human pulmonary artery endothelial cells (hPAECs) exposed to vasoactive stimuli including hypoxia, TGF-ß, ET-1, and IGF-1. Notch2-deficient hPAECs activated Akt, Erk1/2, and antiapoptotic protein Bcl-2 and reduced levels of p21cip and Bax associated with increased EC proliferation and reduced apoptosis. In addition, Notch2 suppression elicited a paradoxical activation of Notch1 and canonical Notch target gene Hes1, Hey1, and Hey2 transcription. Furthermore, reduction in Rb and increased E2F1 binding to the Notch1 promoter appear to explain the Notch1 upregulation. Yet, when Notch1 was decreased in Notch2-suppressed cells, the wound injury response was augmented. In aggregate, our results demonstrate that loss of Notch2 in hPAECs derepresses Notch1 and elicits EC hallmarks of PAH. Augmented EC proliferation upon Notch1 knockdown points to a context-dependent role for Notch1 and 2 in endothelial cell homeostasis.NEW & NOTEWORTHY This study demonstrates a previously unidentified role for Notch2 in the maintenance of lung vascular endothelial cell quiescence and pulmonary artery hypertension (PAH). A key novel finding is that Notch2 suppression activates Notch1 via Rb-E2F1-mediated signaling and induces proliferation and apoptosis resistance in human pulmonary artery endothelial cells. Notably, PAH patients show reduced levels of endothelial Notch2 in their pulmonary arteries, supporting Notch2 as a fundamental driver of PAH pathogenesis.


Assuntos
Proliferação de Células , Células Endoteliais/metabolismo , Hipertensão Pulmonar/metabolismo , Receptor Notch2/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , Receptor Notch1/metabolismo , Receptor Notch2/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição HES-1/metabolismo
7.
Drug Des Devel Ther ; 15: 2025-2033, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34012255

RESUMO

PURPOSE: Pulmonary hypertension (PH) is a pathological process mainly characterized by the progressive increase in pulmonary vascular resistance. The degradation of pulmonary artery smooth muscle cells (PASMCs) from contractile/differentiated phenotype to synthetic/dedifferentiated phenotype is a key factor for hypoxic pulmonary hypertension. MATERIALS AND METHODS: In this study, qPCR was performed to evaluate the gene expression of mRNAs. Western blot, immunofluorescence and RNA pull down were used to detect gene expression levels. RESULTS: We found that the gene expression of polypyrimidine tract-binding protein1 (PTBP1) was increased significantly in a time-dependent manner in rats PA tissues and PASMCs after hypoxia. PTBP1 knockdown can inhibit the phenotypic transition of PASMCs. PTBP1 inhibits the phenotypic transition of PASMCs. In addition, PTBP1 inhibits the integrin-linked kinase (ILK) expression under hypoxic conditions, thereby down-regulating the expression of downstream proteins. It inhibits the phenotypic transition of PASMCs and alleviates pulmonary hypertension. CONCLUSION: In conclusion, PTBP1/ILK axis promotes the development of PH via inducing phenotypic transition of PASMCs. This may provide a novel therapy for PH.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/genética , Hipertensão Pulmonar/fisiopatologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteínas Serina-Treonina Quinases/genética , Artéria Pulmonar/patologia , Animais , Hipóxia Celular , Regulação para Baixo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Hipertensão Pulmonar/genética , Masculino , Miócitos de Músculo Liso/patologia , Fenótipo , Artéria Pulmonar/citologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
8.
Biomolecules ; 11(4)2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921883

RESUMO

Parvovirus B19 (B19V) is a widespread human pathogen possessing a high tropism for erythroid precursor cells. However, the persistence or active replication of B19V in endothelial cells (EC) has been detected in diverse human pathologies. The VP1 unique region (VP1u) of the viral capsid has been reported to act as a major determinant of viral tropism for erythroid precursor cells. Nevertheless, the interaction of VP1u with EC has not been studied. We demonstrate that recombinant VP1u is efficiently internalized by rats' pulmonary trunk blood vessel-derived EC in vitro compared to the human umbilical vein EC line. The exposure to VP1u was not acutely cytotoxic to either human- or rat-derived ECs, but led to the upregulation of cellular stress signaling-related pathways. Our data suggest that high levels of circulating B19V during acute infection can cause endothelial damage, even without active replication or direct internalization into the cells.


Assuntos
Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Parvovirus B19 Humano/química , Proteínas Virais de Fusão/farmacologia , Animais , Sobrevivência Celular , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Artéria Pulmonar/citologia , Ratos , Ratos Wistar , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Virais de Fusão/química
9.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805714

RESUMO

Trifluoperazine (TFP), an antipsychotic drug approved by the Food and Drug Administration, has been show to exhibit anti-cancer effects. Pulmonary arterial hypertension (PAH) is a devastating disease characterized by a progressive obliteration of small pulmonary arteries (PAs) due to exaggerated proliferation and resistance to apoptosis of PA smooth muscle cells (PASMCs). However, the therapeutic potential of TFP for correcting the cancer-like phenotype of PAH-PASMCs and improving PAH in animal models remains unknown. PASMCs isolated from PAH patients were exposed to different concentrations of TFP before assessments of cell proliferation and apoptosis. The in vivo therapeutic potential of TFP was tested in two preclinical models with established PAH, namely the monocrotaline and sugen/hypoxia-induced rat models. Assessments of hemodynamics by right heart catheterization and histopathology were conducted. TFP showed strong anti-survival and anti-proliferative effects on cultured PAH-PASMCs. Exposure to TFP was associated with downregulation of AKT activity and nuclear translocation of forkhead box protein O3 (FOXO3). In both preclinical models, TFP significantly lowered the right ventricular systolic pressure and total pulmonary resistance and improved cardiac function. Consistently, TFP reduced the medial wall thickness of distal PAs. Overall, our data indicate that TFP could have beneficial effects in PAH and support the view that seeking new uses for old drugs may represent a fruitful approach.


Assuntos
Fármacos Cardiovasculares/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipertensão Pulmonar/tratamento farmacológico , Hipóxia/prevenção & controle , Miócitos de Músculo Liso/efeitos dos fármacos , Trifluoperazina/farmacologia , Animais , Antipsicóticos/farmacologia , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Feminino , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Hemodinâmica/efeitos dos fármacos , Humanos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/fisiopatologia , Hipóxia/induzido quimicamente , Hipóxia/genética , Hipóxia/fisiopatologia , Indóis/administração & dosagem , Monocrotalina/administração & dosagem , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Pirróis/administração & dosagem , Ratos , Ratos Sprague-Dawley , Survivina/genética , Survivina/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L688-L704, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33502939

RESUMO

Early therapeutic effect of intratracheally (IT)-administered extracellular vesicles secreted by mesenchymal stem cells (MSC-EVs) has been demonstrated in a rat model of bronchopulmonary dysplasia (BPD) involving hyperoxia exposure in the first 2 postnatal weeks. The aim of this study was to evaluate the protective effects of IT-administered MSC-EVs in the long term. EVs were produced from MSCs following GMP standards. At birth, rats were distributed in three groups: (a) animals raised in ambient air for 6 weeks (n = 10); and animals exposed to 60% hyperoxia for 2 weeks and to room air for additional 4 weeks and treated with (b) IT-administered saline solution (n = 10), or (c) MSC-EVs (n = 10) on postnatal days 3, 7, 10, and 21. Hyperoxia exposure produced significant decreases in total number of alveoli, total surface area of alveolar air spaces, and proliferation index, together with increases in mean alveolar volume, mean linear intercept and fibrosis percentage; all these morphometric changes were prevented by MSC-EVs treatment. The medial thickness index for <100 µm vessels was higher for hyperoxia-exposed/sham-treated than for normoxia-exposed rats; MSC-EV treatment significantly reduced this index. There were no significant differences in interstitial/alveolar and perivascular F4/8-positive and CD86-positive macrophages. Conversely, hyperoxia exposure reduced CD163-positive macrophages both in interstitial/alveolar and perivascular populations and MSC-EV prevented these hyperoxia-induced reductions. These findings further support that IT-administered EVs could be an effective approach to prevent/treat BPD, ameliorating the impaired alveolarization and pulmonary artery remodeling also in a long-term model. M2 macrophage polarization could play a role through anti-inflammatory and proliferative mechanisms.


Assuntos
Displasia Broncopulmonar/complicações , Modelos Animais de Doenças , Vesículas Extracelulares/fisiologia , Lesão Pulmonar/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Administração por Inalação , Animais , Animais Recém-Nascidos , Feminino , Hiperóxia/fisiopatologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/patologia , Masculino , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , Traqueia
12.
Pharmacol Res ; 163: 105287, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33157235

RESUMO

Resveratrol, trans 3,5,4'-trihydroxystilbene, is a stilbenoid polyphenol with a wide range of properties including antioxidant, neuroprotective, cardioprotective, anti-inflammatory and anticancer activities. It is found in the skins of grape (50-100 µg/mL), red wine, peanuts, bilberries, blueberries and cranberries. The most important effects of resveratrol have been found in cardiovascular disease, with pulmonary arterial hypertension (PAH) being a major severe and progressive component. Many factors are involved in the pathogenesis of PAH, including enzymes, transcription factors, proteins, chemokines, cytokines, hypoxia, oxidative stress and others. Resveratrol treats PAH through its actions on various signaling pathways. These signaling pathways are mainly suppressed SphK1-mediated NF-κB activation, BMP/SMAD signaling pathway, miR-638 and NR4A3/cyclin D1 pathway, SIRT1 pathway, Nrf-2, HIF-1 α expression, MAPK/ERK1 and PI3K/AKT pathways, and RhoA-ROCK signaling pathway. Resveratrol efficiently inhibits the proliferation of pulmonary arterial smooth muscle cells and right ventricular remodeling, which are underlying processes leading to enhanced PAH. While supportive evidence from randomized controlled trials is yet to be available, current in vitro and in vivo studies seem to be convincing and suggest a therapeutic promise for the use of resveratrol in PAH.


Assuntos
Hipertensão Arterial Pulmonar/tratamento farmacológico , Estilbenos/uso terapêutico , Animais , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/citologia , Estilbenos/farmacologia , Remodelação Vascular/efeitos dos fármacos
13.
Eur J Pharmacol ; 889: 173618, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33010302

RESUMO

Pulmonary hypertension (PH) is a progressive and potentially serious lung disease, defined by an abnormal elevation of pulmonary arterial pressure. PH occurs for many reasons, and hypoxia is considered as an important stimulus for the disease. Proliferation and migration of pulmonary artery smooth muscular cells (PASMCs) in the small peripheral pulmonary arteries are common characteristic features in hypoxia-induced PH (HPH). However, the mechanisms involved in the hypoxia-induced cell proliferation and migration are not clear. The aim of the present study was to investigate the role of lncRNA Gas5 in the hypoxia-stimulated proliferation and migration of human PASMCs (hPASMCs). We found that the expression of Gas5 was down-regulated in a rat model with hypoxia and in cultured hypoxic hPASMCs, and silence of Gas5 significantly promoted hPASMCs proliferation and migration in both normal and hypoxia condition. Subsequent studies revealed that miR-23b-3p interacted with Gas5 by directly targeting the miRNA-binding site in the Gas5 sequence, and qRT-PCR results showed miR-23b-3p and Gas5 could affect each other's expression, respectively. Further study demonstrated that Gas5 acted as a competing endogenous RNA (ceRNA) for miR-23b-3p to modulate the KCNK3 expression, and these interactions led to promotion of hPASMCs proliferation and migration. This study identified that Gas5/miR-23b-3p/KCNK3 axis may be a mechanism that hypoxia-induced PASMCs proliferation and migration, providing a strategy for clinical treatment of HPH in the future.


Assuntos
Regulação para Baixo/fisiologia , Miócitos de Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Canais de Potássio de Domínios Poros em Tandem/biossíntese , Artéria Pulmonar/metabolismo , RNA Longo não Codificante/biossíntese , Animais , Hipóxia Celular/fisiologia , Células Cultivadas , Expressão Gênica , Masculino , Proteínas do Tecido Nervoso/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Artéria Pulmonar/citologia , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , Ratos , Ratos Sprague-Dawley
14.
Life Sci ; 262: 118420, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32926932

RESUMO

BACKGROUND: Circular RNAs (circRNAs) function as important modulators in the progression of pulmonary arterial hypertension (PAH). Here, we aimed to discover the role and working mechanism of circATP2B4 in hypoxia-induced proliferation and migration of PASMCs. METHODS: The proliferation, migration and apoptosis of pulmonary arterial smooth muscle cells (PASMCs) were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), wound healing assay and flow cytometry. The expression of circATP2B4, ATPase plasma membrane Ca2+ transporting 4 (ATP2B4), microRNA-223 (miR-223) and ATR serine/threonine kinase (ATR) was quantified by quantitative real time polymerase chain reaction (qRT-PCR). Circular RNA Interactome and microT-CDS were used to search the targets of circATP2B4 and miR-223, respectively. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were used to validate the above predictions. Western blot assay was performed to detect the protein expression of ATR. RESULTS: Hypoxia treatment promoted the proliferation and migration and impeded the apoptosis of PASMCs. A significant increase of circATP2B4 expression was observed in the serum of PAH patients and hypoxia-induced PASMCs compared with healthy volunteers and PASMCs under normoxia condition. MiR-223 is a target of circATP2B4, and the effects of circATP2B4 silencing on PASMCs were overturned by the transfection of anti-miR-223. ATR is a functional target of miR-223, and miR-223 inhibited the proliferation and migration while accelerated the apoptosis of PASMCs through targeting and down-regulating ATR. CircATP2B4 could up-regulate the level of ATR through sponging miR-223 in PSAMCs. CONCLUSION: CircATP2B4 potentiated hypoxia-induced proliferation and migration of PASMCs through the miR-223/ATR axis. Restoration of the level of miR-223 might be an effective therapeutic method for the treatment of PAH.


Assuntos
Movimento Celular/genética , Proliferação de Células/genética , Hipertensão Arterial Pulmonar/genética , Artéria Pulmonar/citologia , Idoso , Apoptose/genética , Estudos de Casos e Controles , Hipóxia Celular/fisiologia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Miócitos de Músculo Liso/citologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Hipertensão Arterial Pulmonar/fisiopatologia
15.
Physiol Rep ; 8(17): e14559, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32914566

RESUMO

Phosphodiesterase 3 (PDE3), of which there are two isoforms, PDE3A and PDE3B, hydrolyzes cAMP and cGMP-cyclic nucleotides important in the regulation of pulmonary vascular tone. PDE3 has been implicated in pulmonary hypertension unresponsive to nitric oxide (NO); however, contributions of the two isoforms are not known. Furthermore, adenosine monophosphate-activated protein kinase (AMPK), a critical regulator of cellular energy homeostasis, has been shown to be modulated by PDE3 in varying cell types. While AMPK has recently been implicated in pulmonary hypertension pathogenesis, its role and regulation in the pulmonary vasculature remain to be elucidated. Therefore, we utilized human pulmonary artery smooth muscle cells (hPASMC) to test the hypothesis that NO increases PDE3 expression in an isoform-specific manner, thereby activating AMPK and inhibiting hPASMC proliferation. We found that in hPASMC, NO treatment increased PDE3A protein expression and PDE3 activity with a concomitant decrease in cAMP concentrations and increase in AMPK phosphorylation. Knockdown of PDE3A using siRNA transfection blunted the NO-induced AMPK activation, indicating that PDE3A plays an important role in AMPK regulation in hPASMC. Treatment with a soluble guanylate cyclase (sGC) stimulator increased PDE3A expression and AMPK activation similar to that seen with NO treatment, whereas treatment with a sGC inhibitor blunted the NO-induced increase in PDE3A and AMPK activation. These results suggest that NO increases PDE3A expression, decreases cAMP, and activates AMPK via the sGC-cGMP pathway. We speculate that NO-induced increases in PDE3A and AMPK may have implications in the pathogenesis and the response to therapies in pulmonary hypertensive disorders.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Miócitos de Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Proteínas Quinases/metabolismo , Artéria Pulmonar/citologia , Quinases Proteína-Quinases Ativadas por AMP , Células Cultivadas , AMP Cíclico/metabolismo , Humanos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Óxido Nítrico/farmacologia
16.
Circ Res ; 127(9): 1138-1152, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32752980

RESUMO

RATIONALE: POSTN (Periostin) is an ECM (extracellular matrix) protein involved in tissue remodeling in response to injury and a contributing factor in tumorigenesis, suggesting that POSTN plays a role in the pathogenesis of pulmonary hypertension (PH). OBJECTIVE: We aimed to gain insight into the mechanistic contribution of POSTN in experimental mouse models of PH and correlate these findings with PH in humans. METHODS AND RESULTS: We used genetic epistasis approaches in human pulmonary artery endothelial cells (hPAECs), human pulmonary artery smooth muscle cells, and experimental mouse models of PH (Sugen 5416/hypoxia or chronic hypoxia) to discern the role of POSTN and its relationship to HIF (hypoxia-inducible factor)-1α signaling. We found that POSTN expression was correlated with the extent of PH in mouse models and in humans. Decreasing POSTN improved hemodynamic and cardiac responses in PH mice, blunted the release of growth factors and HIF-1α, and reversed the downregulated BMPR (bone morphogenetic protein receptor)-2 expression in hPAECs from patients with PH, whereas increasing POSTIN had the opposite effects and induced a hyperproliferative and promigratory phenotype in both hPAECs and human pulmonary artery smooth muscle cells. Overexpression of POSTN-induced activation of HIFs and increased the production of ET (endothelin)-1 and VEGF (vascular endothelial growth factor) in hPAECs. SiRNA-mediated knockdown of HIF-1α abolished the proangiogenic effect of POSTN. Blockade of TrkB (tyrosine kinase receptor B) attenuated the effect of POSTN on HIF-1α expression, while inhibition of HIF-1α reduced the expression of POSTN and TrkB. These results suggest that hPAECs produce POSTN via a HIF-1α-dependent mechanism. CONCLUSIONS: Our study reveals that POSTN expression is increased in human and animal models of PH and fosters PH development via a positive feedback loop between HIF-1α and POSTN during hypoxia. We propose that manipulating POSTIN expression may be an efficacious therapeutic target in the treatment of PH. Our results also suggest that POSTN may serve as a biomarker to estimate the severity of PH.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hipertensão Pulmonar/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Animais , Biomarcadores/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Hipóxia Celular , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Endotelina-1/metabolismo , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/terapia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Indóis , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Artéria Pulmonar/citologia , Pirróis , Receptor trkB/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Life Sci ; 256: 118009, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32603819

RESUMO

AIMS: Abnormal mitochondrial metabolism is an essential factor for excessive proliferation of pulmonary artery smooth muscle cells (PASMCs), which drives the pathological process of pulmonary arterial hypertension (PAH). 3-Bromopyruvate (3-BrPA) is an effective glycolytic inhibitor that improves mitochondrial metabolism, thereby repressing anomalous cell proliferation. MAIN METHODS: An experimental PAH model was established by injection of monocrotaline (MCT) in male Sprague Dawley rats, following which rats were assigned to three groups: control, MCT, and 3-BrPA groups. Three days post injection of MCT, rats were treated with 3-BrPA or vehicle for 4 weeks. At the end of the study, hemodynamic data were measured to confirm PAH condition. Indicators of pulmonary arterial and right ventricular (RV) remodeling as well as the proliferative ability of PASMCs were assayed. Additionally, mitochondrial morphology and function, and antiglycolytic and antiproliferative pathways and genes were analyzed. KEY FINDINGS: Treatment with 3-BrPA effectively improved pulmonary vascular remodeling and right ventricular function, inhibited PASMC proliferation, and preserved mitochondrial morphology and function. Besides, 3-BrPA treatment inhibited the PI3K/AKT/mTOR pathway and regulated the expression of antiproliferative genes in PASMCs. However, bloody ascites, bloating, and cirrhosis of organs were observed in some 3-BrPA treated rats. SIGNIFICANCE: 3-BrPA acts as an important glycolytic inhibitor to improve energy metabolism and reverse the course of PAH. However, 3-BrPA is associated with side effects in MCT-induced rats, indicating that it should be caution in drug delivery dosage, and further studies are needed to evaluate this toxicological mechanism.


Assuntos
Mitocôndrias/efeitos dos fármacos , Hipertensão Arterial Pulmonar/tratamento farmacológico , Piruvatos/farmacologia , Animais , Modelos Animais de Doenças , Masculino , Mitocôndrias/metabolismo , Monocrotalina , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Piruvatos/toxicidade , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/metabolismo
18.
Can J Physiol Pharmacol ; 98(9): 629-636, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32615041

RESUMO

The increase in thickening of the arterial wall of pulmonary arterial hypertension (PAH) includes cellular proliferation as well as matrix deposition and interrupted internal elastic lamina (IEL) consisting of a thick homogeneous sheet of elastin. Little is, although, known about the detail of IEL formation in PAH. Endothelin-1 is overexpressed in pulmonary arterioles of PAH. We aimed to examine the expression of genes contributing to IEL formation in pulmonary artery smooth muscle cells (PASMCs) especially focused on lysyl oxidase (LOx), an exreacellular matrix enzyme that catalyzes the cross-linking of collagens or elastin. We quantified mRNA expressions of genes contributing to IEL formation including LOx in PASMCs using real-time quantitative polymerase chain reaction. We stimulated human PASMCs with endothelin-1 with prostacyclin or trapidil. Endothelin-1 significantly increased LOx expression. Prostacyclin and trapidil restored endothelin-1-induced LOx expression to the basal level. Endothelin-1 increased LOx expression strongly in PASMCs from PAH patients compared to those from controls. Trapidil reduced LOx expression only in PASMCs from PAH patients. Overexpressed endothelin-1 in PAH patients can increase expression of LOx and agitate cross-linking of elastin and collagen, resulting in ectopic deposition of these in the vascular media.


Assuntos
Endotelina-1/metabolismo , Miócitos de Músculo Liso/patologia , Proteína-Lisina 6-Oxidase/metabolismo , Hipertensão Arterial Pulmonar/patologia , Artéria Pulmonar/patologia , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Elastina/metabolismo , Epoprostenol/farmacologia , Perfilação da Expressão Gênica , Humanos , Pulmão/irrigação sanguínea , Pulmão/cirurgia , Transplante de Pulmão , Pneumonectomia , Cultura Primária de Células , Hipertensão Arterial Pulmonar/cirurgia , Artéria Pulmonar/citologia , Trapidil/farmacologia , Regulação para Cima/efeitos dos fármacos
19.
J Physiol Biochem ; 76(3): 445-456, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32592088

RESUMO

Increased evidence indicates that adenosine monophosphate-activated protein kinase (AMPK) plays a vital role in vascular homeostasis, especially under hypoxia, and protects against the progression of pulmonary hypertension (PH). However, the role of AMPK in the pathogenesis of PH remains to be clarified. In the present study, we confirmed that a loss of AMPKα2 exacerbated the development of PH by using hypoxia-induced PH model in AMPKα2 -/- mice. After a 4-week period of hypoxic exposure, AMPKα2 -/- mice exhibited more severe pulmonary vascular remodeling and pulmonary vascular smooth muscle cell (SMC) proliferation when compared with wild type (WT) mice. In vitro, AMPKα2 knockdown promoted the proliferation of pulmonary arterial smooth muscle cells (PASMCs) under hypoxia. This phenomenon was accompanied by upregulated Skp2 and downregulated p27kip1 expression and was abolished by rapamycin, an inhibitor of mTOR. These results indicate that AMPKα2 deficiency exacerbates hypoxia-induced PH by promoting PASMC proliferation via the mTOR/Skp2/p27kip1 signaling axis. Therefore, enhanced AMPKα2 activity might underlie a novel therapeutic strategy for the management of PH.


Assuntos
Proteínas Quinases Ativadas por AMP/deficiência , Hipóxia/patologia , Miócitos de Músculo Liso/citologia , Hipertensão Arterial Pulmonar/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Hipóxia Celular , Linhagem Celular , Proliferação de Células , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Artéria Pulmonar/citologia , Ratos
20.
Sci Rep ; 10(1): 10345, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587339

RESUMO

Pulmonary arterial hypertension (PAH) is a progressive fatal disease caused by pulmonary arterial remodeling. Midkine regulates cell proliferation and migration, and it is induced by hypoxia, but its roles in pulmonary arterial remodeling remain unclear. Serum midkine levels were significantly increased in PAH patients compared with control patients. Midkine expression was increased in lungs and sera of hypoxia-induced PAH mice. Hypoxia-induced pulmonary arterial remodeling and right ventricular hypertrophy were attenuated in midkine-knockout mice. Midkine-induced proliferation and migration of pulmonary arterial smooth muscle cells (PASMC) and epidermal growth factor receptor (EGFR) signaling were significantly increased under hypoxia, which also induced cell-surface translocation of nucleolin. Nucleolin siRNA treatment suppressed midkine-induced EGFR activation in vitro, and nucleolin inhibitor AS1411 suppressed proliferation and migration of PASMC induced by midkine. Furthermore, AS1411 significantly prevented the development of PAH in Sugen hypoxia rat model. Midkine plays a crucial role in PAH development through interaction with surface nucleolin. These data define a role for midkine in PAH development and suggest midkine-nucleolin-EGFR axis as a novel therapeutic target for PAH.


Assuntos
Hipóxia/complicações , Midkina/metabolismo , Fosfoproteínas/metabolismo , Hipertensão Arterial Pulmonar/patologia , Proteínas de Ligação a RNA/metabolismo , Remodelação Vascular/fisiologia , Idoso , Animais , Aptâmeros de Nucleotídeos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Feminino , Humanos , Hipóxia/fisiopatologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Midkina/sangue , Midkina/genética , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/patologia , Oligodesoxirribonucleotídeos/farmacologia , Oligodesoxirribonucleotídeos/uso terapêutico , Fosfoproteínas/antagonistas & inibidores , Cultura Primária de Células , Hipertensão Arterial Pulmonar/sangue , Hipertensão Arterial Pulmonar/prevenção & controle , Artéria Pulmonar/citologia , Artéria Pulmonar/patologia , Proteínas de Ligação a RNA/antagonistas & inibidores , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Nucleolina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA