Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Respir J ; 18(5): e13771, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38747117

RESUMO

BACKGROUND: Hypertension is a main contributing factor of cardiovascular diseases; deregulated circular RNAs are involved in the pathogenesis of pulmonary arterial hypertension (PAH). Herein, we evaluated the function and mechanism of circST6GAL1 in PAH process. METHODS: Human pulmonary artery smooth muscle cells (HPASMCs) were cultured in hypoxic environment for functional analysis. The cell counting kit-8, 5-ethynyl-2'-deoxyuridine, wound healing, and flow cytometry assays were used to investigate cell proliferation, migration, and apoptosis. qRT-PCR and Western blotting analyses were used for level measurement of genes and proteins. The binding between miR-509-5p and circST6GAL1 or multiple C2 and transmembrane domain containing 2 (MCTP2) was analyzed by dual-luciferase reporter, RNA immunoprecipitation, and pull-down assays. The monocrotaline (MCT)-induced PAH mouse models were established for in vivo assay. RESULTS: CircST6GAL1 was highly expressed in PAH patients and hypoxia-induced HPASMCs. Functionally, circST6GAL1 deficiency reversed hypoxia-induced proliferation and migration, as well as apoptosis arrest in HPASMCs. Mechanistically, circST6GAL1 directly targeted miR-509-5p, and MCTP2 was a target of miR-509-5p. Rescue assays showed that the regulatory effects of circST6GAL1 deficiency on hypoxia-induced HPASMCs were abolished. Moreover, forced expression of miR-509-5p suppressed HPASMC proliferation and migration and induced cell apoptosis under hypoxia stimulation, while these effects were abolished by MCTP2 overexpression. Moreover, circST6GAL1 silencing improved MCT-induced pulmonary vascular remodeling and PAH. CONCLUSION: CircST6GAL1 deficiency reversed hypoxia-induced proliferation and migration, as well as apoptosis arrest in HPASMCs, and alleviated pulmonary vascular remodeling in MCT-induced PAH mouse models through the miR-509-5p/MCTP2 axis, indicating a potential therapeutic target for PAH.


Assuntos
Apoptose , Proliferação de Células , MicroRNAs , Hipertensão Arterial Pulmonar , RNA Circular , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Camundongos , Animais , RNA Circular/genética , RNA Circular/metabolismo , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/genética , Hipertensão Arterial Pulmonar/patologia , Modelos Animais de Doenças , Miócitos de Músculo Liso/metabolismo , Masculino , Movimento Celular/genética , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Células Cultivadas , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia
2.
Life Sci ; 239: 116928, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31682848

RESUMO

AIM: The dysfunction of human pulmonary arterial smooth muscle cells (HPASMCs) has been suggested to participate in the pathophysiology of pulmonary arterial hypertension (PAH). This study determined miR-19a expression in hypoxia-induced HPASMCs and explored the mechanistic actions of miR-19a in hypoxia-induced HPASMC proliferation and migration. METHODS: QRT-PCR and western blot assays respectively determined the mRNA and protein expression of miR-19a, phosphatase and tensin homolog (PTEN) and hypoxia-inducible factor-1 alpha (HIF-1α). In vitro functional assays determined HPASMC proliferation and migration, respectively. Luciferase reporter assay determined interaction between miR-19a and PTEN. The knockdown effects of miR-19a on PAH were confirmed in in vivo mice model. RESULTS: Hypoxia treatment time-dependently up-regulated miR-19a expression and enhanced cell proliferation in HPASMCs. MiR-19a overexpression increased cell proliferation and migration of HPASMCs, while repression of miR-19a reduced cell proliferative and migratory potentials of hypoxia-treated HPASMCs. Bioinformatics analysis and luciferase reporter assay showed that PTEN 3' untranslated region was targeted by miR-19a, and miR-19a repressed the mRNA and protein expression of PTEN in HPASMCs. Further rescue studies revealed that miR-19a regulated proliferative and migratory potentials of hypoxia-treated HPASMCs via suppressing PTEN expression. In addition, HIF-1α was identified as one of the mediators for the hypoxia-induced aberrant expression levels of miR-19a and PTEN. MiR-19a overexpression enhanced PI3K/AKT signaling, which was attenuated by enforced expression of PTEN in HPASMCs. More importantly, knockdown of miR-19 attenuated the chronic hypoxia-induced PAH in in vivo mice model. CONCLUSION: This study presented a novel mechanistic action of miR-19a-mediated cell proliferation and migration of HPASMCs.


Assuntos
MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Artéria Pulmonar/metabolismo , Animais , Apoptose , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Linhagem Celular , Movimento Celular/genética , Proliferação de Células/genética , Humanos , Hipertensão Pulmonar/fisiopatologia , Hipóxia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Músculo Liso/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais
3.
Pflugers Arch ; 471(2): 347-355, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30353369

RESUMO

Our study explored the effects of lncRNA UCA1 on the proliferation and apoptosis in hypoxic human pulmonary artery smooth muscle cells (HPASMCs) and highlighted the endogenous relationship between UCA1, ING5, and hnRNP I in cell proliferation. Hypoxia-induced HPASMCs were used to simulate pulmonary arterial hypertension in vitro. Microarray assay was adopted to screen the dysregulated expressed lncRNAs in HPASMCs to find out the target gene of our study. And RT-qPCR was performed to detect the expression of lncRNA UCA1 under hypoxia and normoxia. After transfection, the relationship between UCA1 and cell proliferation in HPASMCs under hypoxia were determined by cell proliferation assay and relative expression of PCNA. Next, ELISA assays were conducted to measure the protein levels of PCNA and ING5. What's more, flow cytometry was employed to measure the apoptosis rate in differentially UCA1-expressed HPASMCs. RIP assays were conducted to further clarify the endogenous relationship between UCA1 and ING5 in hypoxic HPASMCs. Finally, the effects of ING5 to HPASMCs were detected after transfection of ING5 and UCA1 to figure out the role of ING5 in HPASMCs. Hypoxia was revealed to induce proliferation and inhibited apoptosis in HPASMCs. Besides, UCA1 was confirmed to be highly expressed under hypoxia compared with normoxia. UCA1 boosted cell proliferation under hypoxia in HPASMCs. However, the apoptosis was suppressed in the hypoxic HPASMCs transfected with pcDNA3.1-UCA1. Further, mechanism studies found that UCA1 competed with ING5 for hnRNP I, so that upregulating UCA1 inhibited the protein levels of ING5. And finally we found that ING5 restrained cell viability, but promoted cell apoptosis in hypoxic HPASMCs, which was reversed by UCA1 over-expression. In summary, our findings manifested that UCA1 promoted proliferation and restrained apoptosis by competing with ING5 for hnRNP I in HPASMCs induced by hypoxia, indicating their potential roles for the cure of hypoxic pulmonary hypertension.


Assuntos
Proliferação de Células/genética , Hipóxia/genética , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/metabolismo , RNA Longo não Codificante/genética , Apoptose/genética , Sobrevivência Celular/genética , Células Cultivadas , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Pulmão/metabolismo , Músculo Liso Vascular/metabolismo , Fatores de Transcrição/genética , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA