Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Biol Macromol ; 268(Pt 2): 130853, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38570000

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) is a complex vascular disorder, characterized by pulmonary vessel remodeling and perivascular inflammation. Pulmonary arterial smooth muscle cells (PASMCs) pyroptosis is a novel pathological mechanism implicated of pulmonary vessel remodeling. However, the involvement of circRNAs in the process of pyroptosis and the underlying regulatory mechanisms remain inadequately understood. METHODS: Western blotting, PI staining and LDH release were used to explore the role of circLrch3 in PASMCs pyroptosis. Moreover, S9.6 dot blot and DRIP-PCR were used to assess the formation of R-loop between circLrch3 and its host gene Lrch3. Chip-qPCR were used to evaluate the mechanism of super enhancer-associated circLrh3, which is transcriptionally activated by the transcription factor Tbx2. RESULTS: CircLrch3 was markedly upregulated in hypoxic PASMCs. CircLrch3 knockdown inhibited hypoxia induced PASMCs pyroptosis in vivo and in vitro. Mechanistically, circLrch3 can form R-loop with host gene to upregulate the protein and mRNA expression of Lrch3. Furthermore, super enhancer interacted with the Tbx2 at the Lrch3 promoter locus, mediating the augmented transcription of circLrch3. CONCLUSION: Our findings clarify the role of a super enhancer-associated circLrch3 in the formation of R-loop with the host gene Lrch3 to modulate pyroptosis in PASMCs, ultimately promoting the development of PH.


Asunto(s)
Miocitos del Músculo Liso , Arteria Pulmonar , Piroptosis , ARN Circular , Piroptosis/genética , ARN Circular/genética , ARN Circular/metabolismo , Animales , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Miocitos del Músculo Liso/metabolismo , Ratas , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Hipoxia de la Célula/genética , Músculo Liso Vascular/metabolismo , Masculino , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Regulación de la Expresión Génica , Elementos de Facilitación Genéticos/genética , Hipoxia/genética , Hipoxia/metabolismo , Súper Potenciadores
2.
Arterioscler Thromb Vasc Biol ; 43(7): 1179-1198, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37139839

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) have been implicated in pulmonary hypertension progression through largely unknown mechanisms. Pulmonary artery endothelial cell (PAEC) dysfunction is a hallmark in the pathogenesis of pulmonary hypertension. However, the specific role of circular RNAs in PAEC injury caused by hypoxia remains unclear. METHODS: In this study, using the Western blotting, RNA pull down, Dual-luciferase reporter assay, immunohistochemistry, and immunofluorescence, we identified a novel circular RNA derived from alternative splicing of the keratin 4 gene (circKrt4). RESULTS: CircKrt4 was upregulated in lung tissues and plasma and specifically in PAECs under hypoxic conditions. In the nucleus, circKrt4 induces endothelial-to-mesenchymal transition by interacting with the Pura (transcriptional activator protein Pur-alpha) to promote N-cadherin gene activation. In the cytoplasm, increased circKrt4 leads to mitochondrial dysfunction by inhibiting cytoplasmic-mitochondrial shuttling of mitochondrial-bound Glpk (glycerol kinase). Intriguingly, circKrt4 was identified as a super enhancer-associated circular RNA that is transcriptionally activated by a transcription factor, CEBPA (CCAAT enhancer binding protein alpha). Furthermore, RBM25 (RNA-binding-motif protein 25) was found to regulate circKrt4 cyclization by increase the back-splicing of Krt4 gene. CONCLUSIONS: These findings demonstrate that a super enhancer-associated circular RNA-circKrt4 modulates PAEC injury to promote pulmonary hypertension by targeting Pura and Glpk.


Asunto(s)
Hipertensión Pulmonar , Arteria Pulmonar , Ratones , Animales , Arteria Pulmonar/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Proliferación Celular , Hipoxia/metabolismo , ARN/genética , Células Endoteliales/metabolismo
3.
Mol Med ; 28(1): 126, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36284300

RESUMEN

BACKGROUND: Pyroptosis is a form of programmed cell death involved in the pathophysiological progression of hypoxic pulmonary hypertension (HPH). Emerging evidence suggests that N6-methyladenosine (m6A)-modified transcripts of long noncoding RNAs (lncRNAs) are important regulators that participate in many diseases. However, whether m6A modified transcripts of lncRNAs can regulate pyroptosis in HPH progression remains unexplored. METHODS: The expression levels of FENDRR in hypoxic pulmonary artery endothelial cells (HPAECs) were detected by using quantitative real-time polymerase chain reaction (qRT-PCR) and fluorescence in situ hybridization (FISH). Western blot, Lactate dehydrogenase (LDH) release assay, Annexin V-FITC/PI double staining, Hoechst 33342/PI fluorescence staining and Caspase-1 activity assay were used to detect the role of FENDRR in HPAEC pyroptosis. The relationship between FENDRR and dynamin-related protein 1 (DRP1) was explored using bioinformatics analysis, Chromatin Isolation by RNA Purification (CHIRP), Electrophoretic mobility shift assay (EMSA) and Methylation-Specific PCR (MSP) assays. RNA immunoprecipitation (RIP) and m6A dot blot were used to detect the m6A modification levels of FENDRR. A hypoxia-induced mouse model of pulmonary hypertension (PH) was used to test preventive effect of conserved fragment TFO2 of FENDRR. RESULTS: We found that FENDRR was significantly downregulated in the nucleus of hypoxic HPAECs. FENDRR overexpression inhibited hypoxia-induced HPAEC pyroptosis. Additionally, DRP1 is a downstream target gene of FENDRR, and FENDRR formed an RNA-DNA triplex with the promoter of DRP1, which led to an increase in DRP1 promoter methylation that decreased the transcriptional level of DRP1. Notably, we illustrated that the m6A reader YTHDC1 plays an important role in m6A-modified FENDRR degradation. Additionally, conserved fragment TFO2 of FENDEE overexpression prevented HPH in vivo. CONCLUSION: In summary, our results demonstrated that m6A-induced decay of FENDRR promotes HPAEC pyroptosis by regulating DRP1 promoter methylation and thereby provides a novel potential target for HPH therapy.


Asunto(s)
Hipertensión Pulmonar , ARN Largo no Codificante , Ratones , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Metilación de ADN , Células Endoteliales/metabolismo , Piroptosis , Arteria Pulmonar , Hipertensión Pulmonar/genética , Hibridación Fluorescente in Situ , Hipoxia/genética , Dinaminas/genética , Dinaminas/metabolismo , Cromatina , Lactato Deshidrogenasas/genética , Lactato Deshidrogenasas/metabolismo , Caspasas
4.
Cell Biosci ; 12(1): 9, 2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35090552

RESUMEN

BACKGROUND: Excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) is the main cause of hypoxic pulmonary hypertension (PH), and mitochondrial homeostasis plays a crucial role. However, the specific molecular regulatory mechanism of mitochondrial function in PASMCs remains unclear. METHODS: In this study, using the CCK8 assay, EdU incorporation, flow cytometry, Western blotting, co-IP, mass spectrometry, electron microscopy, immunofluorescence, Seahorse extracellular flux analysis and echocardiography, we investigated the specific involvement of apoptosis-inducing factor (AIF), a mitochondrial oxidoreductase in regulating mitochondrial energy metabolism and mitophagy in PASMCs. RESULTS: In vitro, AIF deficiency in hypoxia leads to impaired oxidative phosphorylation and increased glycolysis and ROS release because of the loss of mitochondrial complex I activity. AIF was also downregulated and ubiquitinated under hypoxia leading to the abnormal occurrence of mitophagy and autophagy through its interaction with ubiquitin protein UBA52. In vivo, treatment with the adeno-associated virus vector to overexpress AIF protected pulmonary vascular remodeling from dysfunctional and abnormal proliferation. CONCLUSIONS: Taken together, our results identify AIF as a potential therapeutic target for PH and reveal a novel posttranscriptional regulatory mechanism in hypoxia-induced mitochondrial dysfunction.

5.
Mol Ther Nucleic Acids ; 22: 530-541, 2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33230455

RESUMEN

Emerging evidence has suggested that circular RNAs (circRNAs) are involved in multiple physiological processes and participate in a variety of human diseases. However, the underlying biological function of circRNAs in pulmonary hypertension (PH) is still ambiguous. Herein, we investigated the implication and regulatory effect of a typical circRNA, CDR1as, in the pathological process of vascular calcification in PH. Human pulmonary artery smooth muscle cell (HPASMC) calcification was analyzed by western blotting, immunofluorescence, alizarin red S staining, alkaline phosphatase activity analysis, and calcium deposition quantification. CDR1as targets were identified by bioinformatics analysis and validated by dual-luciferase reporter and RNA antisense purification assays. We identified that CDR1as was upregulated in hypoxic conditions and promoted a phenotypic switch of HPASMCs from a contractile to an osteogenic phenotype. Moreover, microRNA (miR)-7-5p was shown to be a target of CDR1as, and calcium/calmodulin-dependent kinase II-delta (CAMK2D) and calponin 3 (CNN3) were suggested to be the putative target genes and regulated by CDR1as/miR-7-5p. The results showed that the CDR1as/miR-7-5p/CNN3 and CAMK2D regulatory axis mediates HPASMC osteoblastic differentiation and calcification induced by hypoxia. This evidence reveals an approach to the treatment of PH.

6.
Eur J Pharmacol ; 888: 173485, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-32805255

RESUMEN

Autophagy is a major cause of pathological vascular remodeling under hypoxic pulmonary hypertension (PH). Sirtuin 3 (Sirt 3) has recently been reported to be involved in the regulation of autophagy, however, its role as an autophagy regulator during hypoxic PH, particularly the molecular mechanism, remains poorly understood. In the present study, Western blot, immunohistochemistry, immunofluorescence, bromodeoxyuridine incorporation and cell cycle analyses were performed to elucidate the underlying mechanism of hypoxia-induced autophagy and cell proliferation with respect to Sirt 3. We observed that the Sirt 3 expression was decreased under hypoxia and that Sirt 3 overexpression significantly inhibited the effects of hypoxia on autophagy. Next, we investigated the mechanistic role of microRNAs in Sirt 3-associated autophagy under hypoxic conditions, with luciferase reporter, microscale thermophoresis and RNA immunoprecipitation assays, results confirming that Sirt 3 is a direct target of miR-874-5p. Furthermore, miR-874-5p was upregulated following hypoxia, and miR-874-5p depletion in turn inhibited autophagy and consequently suppressed abnormal smooth muscle cell proliferation. These findings provide insight into the contribution of the miR-874-5p/Sirt 3 cascade with regard to changes in autophagy and proliferation associated with PH.


Asunto(s)
Autofagia/fisiología , Proliferación Celular/fisiología , MicroARNs/biosíntesis , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Sirtuinas/biosíntesis , Animales , Células Cultivadas , Hipoxia/genética , Hipoxia/metabolismo , Masculino , MicroARNs/genética , Ratas , Ratas Sprague-Dawley , Sirtuinas/genética
7.
J Cell Mol Med ; 24(9): 5260-5273, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32227582

RESUMEN

Piwi-interacting RNAs (piRNAs) are thought to be germline-specific and to be involved in maintaining genome stability during development. Recently, piRNA expression has been identified in somatic cells in diverse organisms. However, the roles of piRNAs in pulmonary arterial smooth muscle cell (PASMC) proliferation and the molecular mechanism underlying the hypoxia-regulated pathological process of pulmonary hypertension are not well understood. Using hypoxic animal models, cell and molecular biology, we obtained the first evidence that the expression of piRNA-63076 was up-regulated in hypoxia and was positively correlated with cell proliferation. Subsequently, we showed that acyl-CoA dehydrogenase (Acadm), which is negatively regulated by piRNA-63076 and interacts with Piwi proteins, was involved in hypoxic PASMC proliferation. Finally, Acadm inhibition under hypoxia was partly attributed to DNA methylation of the Acadm promoter region mediated by piRNA-63076. Overall, these findings represent invaluable resources for better understanding the role of epigenetics in pulmonary hypertension associated with piRNAs.


Asunto(s)
Acil-CoA Deshidrogenasa/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , ARN Interferente Pequeño/metabolismo , Acil-CoA Deshidrogenasa/genética , Animales , Secuencia de Bases , Ciclo Celular/genética , Hipoxia de la Célula/genética , Proliferación Celular/genética , Metilación de ADN/genética , Regulación de la Expresión Génica , Masculino , Miocitos del Músculo Liso/citología , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/genética , Ratas Wistar
8.
Am J Physiol Lung Cell Mol Physiol ; 318(3): L472-L482, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31868509

RESUMEN

Pulmonary hypertension (PH) is a clinically common malignant cardiovascular disease. Pyroptosis is a new form of inflammatory cell death that is involved in many disease processes. Glioma-associated oncogene family zinc finger 1 (GLI1) is a transcriptional activator that participates in many diseases, but its role has never been explored in inducing pyroptosis and the progress of PH. In this study, we used an animal model and cell molecular biology to determine the effect of GLI1 on chronic hypoxia-mediated PH progression and pulmonary artery smooth muscle cell (PASMC) pyroptosis. The major findings of the present study are as follows: Hypoxia induced aberrant expression of GLI1. The inhibition of GLI1 attenuated hypoxia-induced PH and PASMC pyroptosis. Meanwhile, GLI1 enhanced apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) expression by binding with its promoter. GLI1 may promote PASMC pyroptosis through ASC to affect the progression of PH. These findings may identify novel targets for molecular therapy of PH.


Asunto(s)
Hipertensión Pulmonar/patología , Hipoxia/fisiopatología , Músculo Liso Vascular/patología , Arteria Pulmonar/patología , Piroptosis , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Masculino , Músculo Liso Vascular/metabolismo , Arteria Pulmonar/metabolismo , Ratas , Ratas Wistar , Proteína con Dedos de Zinc GLI1/genética
9.
J Pharmacol Sci ; 141(2): 97-105, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31640920

RESUMEN

Pulmonary arterial hypertension (PAH) is defined as elevation of mean pulmonary arterial pressure to ≥25 mmHg within the low pressure pulmonary circulatory system. PAH is characterized by obstructive vascular remodeling, partially due to excessive pulmonary arterial smooth muscle cell (PASMC) proliferation. Puerarin is a natural flavonoid isolated from the herb Radix puerariae, which has been widely used for the treatment of cardiovascular and cerebrovascular disorders and diabetes. However, how puerarin mediates autophagy in the progression of pulmonary vascular remodeling is unclear. In this study, we explored the effects of puerarin in a hypoxic pulmonary hypertension (PH) rat model using immunohistochemistry, and morphometric analyses of right ventricle. In addition, cell counting kit 8 assay, western blotting and flow cytometry were employed to test cell proliferation in PASMCs, and then autophagy was tested with mRFP-GFP-LC3 fluorescence microscopy and Western blot. We found that puerarin could alleviate hypoxia-induced PH in rats and improved pulmonary histopathology, and also reduced the expression of autophagy markers in vivo and in vitro. Moreover, puerarin also ameliorated hypoxia-induced PASMC proliferation in an autophagy-dependent manner. Overall, these findings demonstrated that puerarin could prevent hypoxia-induced PH in rats, possibly via reducing autophagy and suppressing cell proliferation.


Asunto(s)
Autofagia/efectos de los fármacos , Hipertensión Pulmonar/prevención & control , Isoflavonas/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Isoflavonas/metabolismo , Pulmón/metabolismo , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/patología , Ratas Wistar , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...