Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Exp Lung Res ; 50(1): 160-171, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287558

RESUMEN

Background: Hypoxic pulmonary hypertension (HPH) is one of the important pathophysiological changes in chronic pulmonary heart disease. Hypoxia promotes the phenotypic transformation of pulmonary artery smooth muscle cells (PASMCs). Extracellular exosomes regulate vascular smooth muscle cell (VSMC) phenotypic switch. Aim: Given the importance of exosomes and alveolar epithelial cells (AECs) in HPH, the present study aimed to address the issue of whether AEC-derived exosomes promote HPH by triggering PASMC phenotypic switch. Methods: Cell Counting Kit-8 (CCK-8), TRITC-phalloidin staining, and Western blotting were used to examine the effects of AEC-derived exosomes on cell proliferation, intracellular actin backbone distribution, and expression of phenotypic marker proteins in PASMCs. Transcriptomics sequencing was used to analyze differentially expressed genes (DEGs) between groups. Results: Hypoxia-induced exosomes (H-exos) could promote the proliferation of PASMCs, cause the reduction of cellular actin microfilaments, promote the expression of synthetic marker proteins (ELN and OPN), reduce the expression of contractile phenotypic marker proteins (SM22-α and α-SMA), and induce the phenotypic transformation of PASMCs. Transcriptomics sequencing analysis showed that the Rap1 signaling pathway was involved in the phenotypic transformation of PASMCs induced by H-exos. Conclusion: The present study identified that hypoxia-induced AEC-derived exosomes promote the phenotypic transformation of PASMCs and its mechanism is related to the Rap1 signaling pathway.


Asunto(s)
Proliferación Celular , Exosomas , Miocitos del Músculo Liso , Fenotipo , Arteria Pulmonar , Transducción de Señal , Exosomas/metabolismo , Arteria Pulmonar/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Células Epiteliales Alveolares/metabolismo , Ratas , Proteínas de Unión al GTP rap1/metabolismo , Proteínas de Unión al GTP rap1/genética , Músculo Liso Vascular/metabolismo , Hipertensión Pulmonar/metabolismo , Ratas Sprague-Dawley , Células Cultivadas , Hipoxia/metabolismo , Hipoxia de la Célula/fisiología
2.
Respir Res ; 25(1): 270, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987833

RESUMEN

BACKGROUND: Hypoxic pulmonary hypertension (HPH) is a challenging lung arterial disorder with remarkably high incidence and mortality rates, and the efficiency of current HPH treatment strategies is unsatisfactory. Endothelial-to-mesenchymal transition (EndMT) in the pulmonary artery plays a crucial role in HPH. Previous studies have shown that lncRNA-H19 (H19) is involved in many cardiovascular diseases by regulating cell proliferation and differentiation but the role of H19 in EndMT in HPH has not been defined. METHODS: In this research, the expression of H19 was investigated in PAH human patients and rat models. Then, we established a hypoxia-induced HPH rat model to evaluate H19 function in HPH by Echocardiography and hemodynamic measurements. Moreover, luciferase reporter gene detection, and western blotting were used to explore the mechanism of H19. RESULTS: Here, we first found that the expression of H19 was significantly increased in the endodermis of pulmonary arteries and that H19 deficiency obviously ameliorated pulmonary vascular remodelling and right heart failure in HPH rats, and these effects were associated with inhibition of EndMT. Moreover, an analysis of luciferase activity indicated that microRNA-let-7 g (let-7 g) was a direct target of H19. H19 deficiency or let-7 g overexpression can markedly downregulate the expression of TGFßR1, a novel target gene of let-7 g. Furthermore, inhibition of TGFßR1 induced similar effects to H19 deficiency. CONCLUSIONS: In summary, our findings demonstrate that the H19/let-7 g/TGFßR1 axis is crucial in the pathogenesis of HPH by stimulating EndMT. Our study may provide new ideas for further research on HPH therapy in the near future.


Asunto(s)
Transición Epitelial-Mesenquimal , Hipertensión Pulmonar , MicroARNs , ARN Endógeno Competitivo , ARN Largo no Codificante , Transducción de Señal , Factor de Crecimiento Transformador beta , Animales , Femenino , Humanos , Masculino , Ratas , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/fisiología , Transición Epitelial-Mesenquimal/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipoxia/metabolismo , Hipoxia/genética , MicroARNs/metabolismo , MicroARNs/genética , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Ratas Sprague-Dawley , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , ARN Endógeno Competitivo/genética , ARN Endógeno Competitivo/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
3.
J Ginseng Res ; 48(4): 405-416, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39036731

RESUMEN

Background: Hypoxic pulmonary hypertension (HPH) is the main pathological change in vascular remodeling, a complex cardiopulmonary disease caused by hypoxia. Some research results have shown that ginsenoside Rg1 (Rg1) can improve vascular remodeling, but the effect and mechanism of Rg1 on hypoxia-induced pulmonary hypertension are not clear. The purpose of this study was to discuss the potential mechanism of action of Rg1 on HPH. Methods: C57BL/6 mice, calpain-1 knockout mice and Pulmonary artery smooth muscle cells (PASMCs) were exposed to a low oxygen environment with or without different treatments. The effect of Rg1 and calpain-1 silencing on inflammation, fibrosis, proliferation and the protein expression levels of calpain-1, STAT3 and p-STAT3 were determined at the animal and cellular levels. Results: At the mouse and cellular levels, hypoxia promotes inflammation, fibrosis, and cell proliferation, and the expression of calpain-1 and p-STAT3 is also increased. Ginsenoside Rg1 administration and calpain-1 knockdown, MDL-28170, and HY-13818 treatment showed protective effects on hypoxia-induced inflammation, fibrosis, and cell proliferation, which may be associated with the downregulation of calpain-1 and p-STAT3 expression in mice and cells. In addition, overexpression of calpain 1 increased p-STAT3 expression, accelerating the onset of inflammation, fibrosis and cell proliferation in hypoxic PASMCs. Conclusion: Ginsenoside Rg1 may ameliorate hypoxia-induced pulmonary vascular remodeling by suppressing the calpain-1/STAT3 signaling pathway.

4.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(7): 757-764, 2024 Jul 15.
Artículo en Chino | MEDLINE | ID: mdl-39014954

RESUMEN

OBJECTIVES: To investigate the protective effects of 2-methoxyestradiol (2ME) against hypoxic pulmonary hypertension (HPH) in neonatal rats. METHODS: Ninety-six Wistar neonatal rats were randomly divided into a normoxia group, a hypoxia group, and a hypoxia + 2ME group, with each group further subdivided into 3-day, 7-day, 14-day, and 21-day subgroups, containing eight rats each. The hypoxia and hypoxia + 2ME groups received daily subcutaneous injections of saline and 2ME (240 µg/kg), respectively, while the normoxia group was raised in a normoxic environment with daily saline injections. Right ventricular systolic pressure (RVSP) was measured using the direct pressure method. Pulmonary vascular morphology was assessed using hematoxylin and eosin staining, with metrics including the percentage of medial thickness of small pulmonary arteries relative to the external diameter (MT%) and the cross-sectional area of the media of small pulmonary arteries relative to the total cross-sectional area (MA%). Immunohistochemistry was used to detect the expression levels of hypoxia-inducible factor-1α (HIF-1α) and proliferating cell nuclear antigen (PCNA) proteins, while real-time quantitative PCR was used to to assess HIF-1α and PCNA mRNA levels. RESULTS: Compared to the normoxia group, the hypoxia and hypoxia + 2ME groups showed increased RVSP and upregulated HIF-1α and PCNA protein and mRNA expression levels at 3, 7, 14, and 21 days after hypoxia (P<0.05). Furthermore, at 7, 14, and 21 days after hypoxia, the hypoxia group showed increased MT% and MA% (P<0.05). In comparison to the hypoxia group, the hypoxia + 2ME group exhibited reduced RVSP and downregulated HIF-1α and PCNA protein and mRNA expression levels, along with decreased MT% and MA% at 7, 14, and 21 days after hypoxia (P<0.05). CONCLUSIONS: 2ME may protect against HPH in neonatal rats by inhibiting the expression of HIF-1α and PCNA and reducing pulmonary vascular remodeling. Citation:Chinese Journal of Contemporary Pediatrics, 2024, 26(7): 757-764.


Asunto(s)
2-Metoxiestradiol , Animales Recién Nacidos , Hipertensión Pulmonar , Subunidad alfa del Factor 1 Inducible por Hipoxia , Hipoxia , Antígeno Nuclear de Célula en Proliferación , Arteria Pulmonar , Ratas Wistar , Animales , 2-Metoxiestradiol/farmacología , Ratas , Hipertensión Pulmonar/prevención & control , Hipertensión Pulmonar/tratamiento farmacológico , Antígeno Nuclear de Célula en Proliferación/análisis , Antígeno Nuclear de Célula en Proliferación/genética , Hipoxia/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Arteria Pulmonar/efectos de los fármacos , Masculino , Femenino , Estradiol/farmacología , Estradiol/análogos & derivados , ARN Mensajero/análisis
5.
Biochem Pharmacol ; 226: 116382, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38909785

RESUMEN

Hypoxic pulmonary hypertension (HPH) is a serious and life-threatening chronic cardiopulmonary disease characterized by progressive elevation of pulmonary artery pressure and pulmonary vascular remodeling. Mesenchymal stem cell- derived exosomes (MSC-Exos) can relieve HPH by reversing pulmonary vascular remodeling. The HPH model was established in healthy male Sprague-Dawley (SD) rats aged 6 to 8 weeks. The rats were placed in a room with oxygen concentration of (10 ± 1) % for 8 hours a day over 28 days, were then injected intravenously with MSC-Exos (100 ug protein/kg) or equal-volume phosphate buffer saline (PBS) once a day over 1 week. Right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI) and pulmonary vascular remodeling were observed after anesthesia. In addition, platelet-derived growth factor BB (PDGF-BB) was used to stimulate rat pulmonary artery smooth muscle cells (PASMCs) to construct HPH pathological cell models. The results showed that MSC-Exos could not only reduce the elevation of RVSP, right ventricular hypertrophy and the degree of pulmonary vascular remodeling in HPH rats, but also reduce the proliferation, migration and apoptosis resistance of PASMCs. Finally, GSE53408 and GSE113439 datasets were analyzed and showed that the expression of Hsp90aa1 and pERK/ERK were significantly increased in HPH, also could be inhibited by MSC-Exos. Meanwhile, inhibition of Hsp90aa1 also reduced PASMCs migration and pERK/ERK protein level. In conclusion, MSC-Exos alleviated HPH by suppressing PASMCs proliferation, migration and apoptosis resistance through inhibiting the Hsp90aa1/ERK/pERK pathway.


Asunto(s)
Exosomas , Proteínas HSP90 de Choque Térmico , Hipertensión Pulmonar , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas , Ratas Sprague-Dawley , Animales , Masculino , Ratas , Exosomas/metabolismo , Exosomas/trasplante , Proteínas HSP90 de Choque Térmico/metabolismo , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/terapia , Hipoxia/metabolismo , Hipoxia/terapia , Sistema de Señalización de MAP Quinasas/fisiología , Células Madre Mesenquimatosas/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología
6.
Curr Pharm Des ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38867532

RESUMEN

BACKGROUND: Hypoxic Pulmonary Hypertension (HPH), a prevalent disease in highland areas, is a crucial factor in various complex highland diseases with high mortality rates. Zhishi-Xiebai-Guizhi Decoction (ZXGD), traditional Chinese medicine with a long history of use in treating heart and lung diseases, lacks a clear understanding of its pharmacological mechanism. OBJECTIVE: This study aimed to investigate the pharmacological effects and mechanisms of ZXGD on HPH. METHODS: We conducted a network pharmacological prediction analysis and molecular docking to predict the effects, which were verified through in vivo experiments. RESULTS: Network pharmacological analysis revealed 51 active compounds of ZXGD and 701 corresponding target genes. Additionally, there are 2,116 target genes for HPH, 311 drug-disease co-target genes, and 17 core target genes. GO functional annotation analysis revealed that the core target genes primarily participate in biological processes such as apoptosis and cellular response to hypoxia. Furthermore, KEGG pathway enrichment analysis demonstrated that the core targets are involved in several pathways, including the phosphatidylinositol- 3 kinase/protein kinase B (PI3K/Akt) signaling pathway and Hypoxia Inducible Factor 1 (HIF1) signaling pathway. In vivo experiments, the continuous administration of ZXGD demonstrated a significant improvement in pulmonary artery pressure, right heart function, pulmonary vascular remodeling, and pulmonary vascular fibrosis in HPH rats. Furthermore, ZXGD was found to inhibit the expression of PI3K, Akt, and HIF1α proteins in rat lung tissue. CONCLUSION: In summary, this study confirmed the beneficial effects and mechanism of ZXGD on HPH through a combination of network pharmacology and in vivo experiments. These findings provided a new insight for further research on HPH in the field of traditional Chinese medicine.

7.
J Mol Cell Cardiol ; 194: 16-31, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38821243

RESUMEN

BACKGROUND: Hypoxia-induced pulmonary artery hypertension (HPH) is a complication of chronic hypoxic lung disease and the third most common type of pulmonary artery hypertension (PAH). Epigenetic mechanisms play essential roles in the pathogenesis of HPH. N6-methyladenosine (m6A) is an important modified RNA nucleotide involved in a variety of biological processes and an important regulator of epigenetic processes. To date, the precise role of m6A and regulatory molecules in HPH remains unclear. METHODS: HPH model and pulmonary artery smooth muscle cells (PASMCs) were constructed from which m6A changes were observed and screened for AlkB homolog 5 (Alkbh5). Alkbh5 knock-in (KI) and knock-out (KO) mice were constructed to observe the effects on m6A and evaluate right ventricular systolic pressure (RVSP), left ventricular and septal weight [RV/(LV + S)], and pulmonary vascular remodeling in the context of HPH. Additionally, the effects of Alkbh5 knockdown using adenovirus were examined in vitro on m6A, specifically in PASMCs with regard to proliferation, migration and cytochrome P450 1A1 (Cyp1a1) mRNA stability. RESULTS: In both HPH mice lung tissues and hypoxic PASMCs, a decrease in m6A was observed, accompanied by a significant up-regulation of Alkbh5 expression. Loss of Alkbh5 attenuated the proliferation and migration of hypoxic PASMCs in vitro, with an associated increase in m6A modification. Furthermore, Alkbh5 KO mice exhibited reduced RVSP, RV/(LV + S), and attenuated vascular remodeling in HPH mice. Mechanistically, loss of Alkbh5 inhibited Cyp1a1 mRNA decay and increased its expression through an m6A-dependent post-transcriptional mechanism, which hindered the proliferation and migration of hypoxic PASMCs. CONCLUSION: The current study highlights the loss of Alkbh5 impedes the proliferation and migration of PASMCs by inhibiting post-transcriptional Cyp1a1 mRNA decay in an m6A-dependent manner.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB , Citocromo P-450 CYP1A1 , Hipoxia , Ratones Noqueados , Arteria Pulmonar , Estabilidad del ARN , Animales , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Hipoxia/genética , Hipoxia/metabolismo , Ratones , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/metabolismo , Miocitos del Músculo Liso/metabolismo , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/etiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Masculino , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Adenosina/análogos & derivados , Adenosina/metabolismo , Remodelación Vascular/genética , Ratones Endogámicos C57BL
8.
Apoptosis ; 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38635022

RESUMEN

Hypoxic pulmonary hypertension (HPH) is a pathophysiological syndrome in which pulmonary vascular pressure increases under hypoxic stimulation and there is an urgent need to develop emerging therapies for the treatment of HPH. LncRNA MIR210HG is a long non-coding RNA closely related to hypoxia and has been widely reported in a variety of tumor diseases. But its mechanism in hypoxic pulmonary hypertension is not clear. In this study, we identified for the first time the potential effect of MIR210HG on disease progression in HPH. Furthermore, we investigated the underlying mechanism through which elevated levels of MIR210HG promotes the transition from a contractile phenotype to a synthetic phenotype in PASMCs under hypoxia via activation of autophagy-dependent ferroptosis pathway. While overexpression of HIF-2α in PASMCs under hypoxia significantly reversed the phenotypic changes induced by MIR210HG knockdown. We further investigated the potential positive regulatory relationship between STAT3 and the transcription of MIR210HG in PASMCs under hypoxic conditions. In addition, we established both in vivo and in vitro models of HPH to validate the differential expression of specific markers associated with hypoxia. Our findings suggest a potential mechanism of LncRNA MIR210HG in the progression of HPH and offer potential targets for disease intervention and treatment.

9.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1020927

RESUMEN

Objective To investigate the effects of polydatin(PD)on the proliferation and apoptosis of pulmonary artery smooth muscle cells(PASMCs)in hypoxic pulmonary hypertension(HPH)neonatal rats,and its mechanism of action.Methods Neonatal rats were randomly separated into six groups:control group,model group,low dose PD group,medium dose PD group,high dose PD group,and high dose PD+Hippo pathway inhibitor(high dose PD+XMU-MP-1)group,with 10 rats in each group.After 2 weeks of hypoxia treatment,the right ventricular systolic blood pressure(RVSP)and right ventricular hypertro-phy index(RVHI)of rats in each group were measured.Hematoxylin-eosin(HE)staining was applied to observe pathological changes in lung tissue,and the percentage of pulmonary artery wall thickness to total thickness(WT)and the percentage of wall area to total area(WA)were calculated.Neonatal rat PASMCs were separated from each group,which were divided into NC group,hypoxia group,low dose PD group,medium dose PD group,high dose PD group,and high dose PD+XMU-MP-1 group.Cell counting kit 8(CCK-8)and 5-ethynyl-2'-deoxyuridine(EdU)were applied to detect cell proliferation.Flow cytometry was applied to detect cell apoptosis.Western blot was applied to detect the expression of Yes-associated protein 1(YAP1),tran-scriptional coactivator with PDZ-binding motif(TAZ),mammalian sterile 20-like kinase 1(MST1),B-cell lymphoma 2(Bcl-2),and Bcl-2 associated protein(Bax)in lung tissue and PASMCs.Results Compared with the control group,the pulmonary artery wall in the model group was significantly thickened,lumen was narrowed,and protein expressions of RVSP,RVHI,WT%,WA%,YAP1,MST1 and TAZ were significantly increased(all P<0.05).Compared with the model group,pulmonary artery thickening and lumen enlargement were observed in the low,medium and high dose PD groups,and the protein expressions of RVSP,RVHI,WT%,WA%,YAP1,MST1 and TAZ were significantly decreased,which showed a dose-dependent relationship(all P<0.05).The effect could be reversed by XMU-MP-1.Compared with the NC group,the cell A450nm value,EdU positive rate,the protein expression of YAP1,MST1,TAZ and Bcl-2 in the hydropoxia group were significantly increased.The apoptosis rate and the expression of Bax protein were obviously reduced(all P<0.05).Compared with the hypoxia group,the cell A450nm value,EdU positive rate,the protein expression of YAP1,MST1,TAZ and Bcl-2 in the low,medium and high dose PD groups were obviously reduced.The apoptosis rate and the expression of Bax were significantly increased,which showed a dose-depend-ent relationship(all P<0.05).The effect could be reversed by XMU-MP-1.Conclusion PD may inhibit the proliferation of PASMCs in HPH neonatal rats and promote apoptosis by inhibiting YAP1/TAZ signaling pathway.

10.
Herald of Medicine ; (12): 545-549, 2024.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1023746

RESUMEN

Hypoxic pulmonary hypertension(HPH)is a kind of chronic high-altitude sickness disease,which is also the initiation link of high-altitude sickness such as pneumocardial disease and high-altitude pulmonary edema.Its main features are persistent vasoconstriction and irreversible remodeling of blood vessels.HPH has greatly impacted the health of people in the plat-eau section.As a typical representative of ethnic medicine,Zang medicine embodies the wisdom of the Zang people in their long-term struggle against diseases.Adapting to local conditions for a long time,Zang medicine has unique advantages in the treatment of HPH.By reviewing the pathogenesis of HPH and the research of single Zang medicine and the complex prescription of Zang medicine in the treatment of HPH,this article aims to provide a theoretical basis for the physiological and pathological research of HPH and to provide a reference for the clinical application of Zang medicine.

11.
Herald of Medicine ; (12): 550-560, 2024.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1023747

RESUMEN

Objective To explore and verify the protective and therapeutic effects and possible mechanisms of Zukamu granules on hypoxia alone and hypoxia+Su5416-induced hypoxic pulmonary hypertension(HPH)in mice.Methods Multiple databases and related literature were used to collect the active ingredients data in Zukamu granules and the HPH-related targets were predicted and obtained.The network construction and enrichment analysis were performed.The HPH mouse models were es-tablished by two-week hypoxia and four-week hypoxia+Su5416 induction,and the relevant indicators and the main pharmacodyna-mic indexes such as right ventricular pressure were tested.Masson staining was used to observe the pathological changes in lung tissues,and Western blotting was used to detect the expression levels of bax,bcl-2,PI3K,p-PI3K,eNOS,and HIF-1α in lung tis-sues.Results A total of 167 active ingredients of Zukamu granules were screened,with 179 intersecting targets with HPH,in-cluding targets like PIK3CA and HIF-1.The validation experimental results showed that Zukamu granules could significantly re-duce right ventricular systolic pressure and right ventricular hypertrophy in HPH mice,and down-regulate the expression of bcl-2 and HIF-1α and up-regulate the expression of bax,PI3K,p-PI3K and eNOS in mice lung tissues.Conclusion Zukamu gran-ules may act against HPH by modulating bax/bcl and PI3K-eNOS/HIF-1α signaling pathways.

12.
Chinese Pharmacological Bulletin ; (12): 565-573, 2024.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1013655

RESUMEN

Aim To explore the efficacy of levosimendan on hypoxia pulmonary hypertension through animal experiments, and to further explore the potential mechanism of action using network pharmacological methods and molecular docking technique. Methods The rat model of hypoxia pulmonary hypertension was constructed to detect right heart systolic pressure and right heart remodeling index. HE , Masson, and VG staining were core targets were screened out. GO and KEGG pathway enrichment analysis were performed using the DAVID database. Molecular docking of the core targets was performed with the AutoDock software. Results The results of animal experiments showed that levosimendan had obvious therapeutic effect on hypoxia pulmonary hypertension. The network pharmacology results showed that SRC, HSP90AA1, MAPK1, PIK3R1, AKT1, HRAS, MAPK14, LCK, EGFR and ESR1 used to analyze the changes of rat lung histopathology. Search the Swiss Target Prediction, DrugBank Online, BatMan, Targetnet, SEA, and PharmMapper databases were used to screen for drug targets. Disease targets were retrieved from the GeneCards, OMIM databases. The "drug-target-disease" network was constructed after identification of the two intersection targets. The protein interaction network was constructed and the were the key targets to play a therapeutic role. Molecular docking showed good docking of levosimendan with all the top five core targets with degree values. Conclusions Levosimendan may exert a therapeutic effect on hypoxia-induced pulmonary hypertension through multiple targets.

13.
Stem Cells ; 42(4): 329-345, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38153856

RESUMEN

Pulmonary hypertension (PH) is an intractable, severe, and progressive cardiopulmonary disease. Recent findings suggest that human umbilical cord mesenchymal stromal cells (HUCMSCs) and HUCMSC-derived exosomes (HUCMSC-Exos) possess potential therapeutic value for PH. However, whether they have beneficial effects on hypoxic pulmonary hypertension (HPH) is unclear. Exos are released into the extracellular environment by the fusion of intracellular multivesicular bodies with the cell membrane, and they play an important role in cellular communication. Exos ameliorate immune inflammation levels, alter macrophage phenotypes, regulate mitochondrial metabolic function, and inhibit pulmonary vascular remodeling, thereby improving PH. Macrophages are important sources of cytokines and other transmitters and can promote the release of cytokines, vasoactive molecules, and reactive oxygen species, all of which are associated with pulmonary vascular remodeling. Therefore, the aim of this study was to investigate whether HUCMSC-Exos could improve the lung inflammatory microenvironment and inhibit pulmonary vascular remodeling by targeting macrophages and identifying the underlying mechanisms. The results showed that HUCMSC-Exos promoted M2 macrophage polarization, decreased pro-inflammatory factors, increased IL-10 levels, and inhibited IL-33/ST2 axis expression, thereby inhibiting hypoxia-induced proliferation of pulmonary artery smooth muscle cells and ameliorating HPH.


Asunto(s)
Exosomas , Hipertensión Pulmonar , Células Madre Mesenquimatosas , Hipertensión Arterial Pulmonar , Humanos , Ratones , Animales , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/terapia , Hipertensión Pulmonar/metabolismo , Exosomas/metabolismo , Remodelación Vascular , Cordón Umbilical/metabolismo , Hipoxia/complicaciones , Hipoxia/metabolismo , Macrófagos/metabolismo , Citocinas/metabolismo , Células Madre Mesenquimatosas/metabolismo
14.
Heliyon ; 9(11): e22044, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38074866

RESUMEN

Hypoxic pulmonary hypertension (HPH) is caused by chronic persistent hypoxia, which leads to the continuous increase of pulmonary artery pressure and pulmonary vascular resistance. In recent years, there has been a substantial increase in research on HPH. To study the trends of HPH research over the last decade, we used WOSCC to search for relevant research on this topic, and dealt with the relevant information using VOSviewer, CiteSpace, and R-tool. Our results show that the number of publications on HPH has generally increased in the last decade, albeit not significantly, while the average number of citations has been declining year by year. Researchers from the USA top the list with 5498 publications, who widely cooperate with researchers from other countries, followed by those from China. Kurt R. Stenmark has an authoritative position in this field, ranking first with 635 citations. American Journal of Physiology Lung Cellular and Molecular Physiology and Pulmonary Circulation have published 151 articles on HPH in the last 10 years, but the former has higher impact factor and article quality. Circulation proved its leadership in this field with 8812 citations. Our findings reveal the trends in HPH research and should provide researchers with plenty of useful information.

15.
Respir Res ; 24(1): 310, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38093274

RESUMEN

BACKGROUND: Hypoxic pulmonary hypertension (HPH) is a common type of pulmonary hypertension and characterized by pulmonary vascular remodeling and constriction. A large number of studies have shown that pulmonary vascular endothelial cells (PVECs) dysfunction plays an important role in the initiation and development stages of HPH, but the mechanism of PVECs dysfunction after hypoxia remains unclear. In this study, we explored the exact mechanism of PVECs dysfunction after hypoxia. METHODS: In vitro, we used primary cultured PVECs hypoxia model to mimic HPH injury. We detected the expressions of mitochondrial biogenesis markers, mitochondrial transcription factor A (TFAM) level inside mitochondria, mitochondrial quantity and function, and the components expressions of translocase of outer mitochondrial membrane (TOM) at 24 h after hypoxia. To explore the effects of Tom70 on mitochondrial biogenesis and functions of PVECs after hypoxia, Tom70 overexpression adenovirus was constructed, and the expressions of mitochondrial biogenesis markers, TFAM level inside mitochondria, mitochondrial quantity and function, and the functions of PVECs were detected. And in vivo, we used cre-dependent overexpression adenovirus of Tom70 in the Cdh5-CreERT2 mouse model of HPH to verify the role of upregulating PVECs Tom70 in improving HPH. RESULTS: Hypoxia obviously increased the expressions of mitochondrial biogenesis markers for PGC-1α, NRF-1 and TFAM, but reduced the content of TFAM in mitochondria and the quantity and functions of mitochondria. In addition, only Tom70 expression among the TOM components was significantly decreased after hypoxia, and up-regulation of Tom70 significantly increased the content of TFAM in mitochondria of PVECs by transporting TFAM into mitochondria after hypoxia, enhanced the quantity and functions of mitochondria, improved the functions of PVECs, and ultimately alleviated HPH. CONCLUSION: The findings of present study demonstrated that hypoxia induced the decreased expression of Tom70 in PVECs, reduced the mitochondrial biogenesis-associated TFAM protein transporting into mitochondria, inhibited mitochondrial biogenesis, caused PVECs injury, and prompted the formation of HPH. However, up-regulation of Tom70 abolished the hypoxia-induced injurious effects on PVECs and alleviated HPH.


Asunto(s)
Hipertensión Pulmonar , Animales , Ratones , Células Endoteliales/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipoxia/complicaciones , Pulmón/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Biogénesis de Organelos
16.
Biomed Pharmacother ; 168: 115816, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37918254

RESUMEN

OBJECTIVE: Hypoxic pulmonary hypertension (HPH) is a progressive and life-threatening disease characterized by perivascular inflammation, pulmonary vascular remodeling, and occlusion. Mesenchymal stromal cell-derived exosomes (MSC-exo) have emerged as potential therapeutic agents due to their role in cell communication and the transportation of bioactive molecules. In this study, we aimed to investigate the therapeutic effects of MSC-exo against HPH and elucidate the underlying molecular mechanism. METHODS: Exosomes were isolated from conditioned media of human bone mesenchymal stromal cells using ultracentrifugation and characterized through western blotting, transmission electron microscopy (TEM), and nanoparticle tracking analysis (NTA). An HPH animal model was established in male SD rats, and MSC-exo or phosphate-buffered saline (PBS) were administered via the tail vein for three weeks. Subsequently, right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), and pulmonary vascular remodeling were evaluated. Lung tissues from HPH rats and normal rats underwent high-throughput sequencing and transcriptomic analysis. Gene Ontology (GO) analysis was employed to identify upregulated differentially expressed genes. Additionally, rat pulmonary artery smooth muscle cells (PASMC) exposed to platelet-derived growth factor-BB (PDGF-BB) were used to simulate HPH-related pathological behavior. In vitro cellular models were established to examine the molecular mechanism of MSC-exo in HPH. RESULTS: MSC-exo administration protected rats from hypoxia-induced increases in RVSP, RVHI, and pulmonary vascular remodeling. Additionally, MSC-exo alleviated PDGF-BB-induced proliferation and migration of PASMC. Transcriptomic analysis revealed 267 upregulated genes in lung tissues of HPH rats compared to control rats. Gene Ontology analysis indicated significant differences in pathways associated with Yes Associated Protein 1 (YAP1), a key regulator of cell proliferation and organ size. RT-qPCR and western blot analysis confirmed significantly increased expression of YAP1 in HPH lung tissues and PASMC, which was inhibited by MSC-exo treatment. Furthermore, analysis of datasets demonstrated that Secreted Phosphoprotein 1 (SPP1), also known as Osteopontin (OPN), is a downstream binding protein of YAP1 and can be upregulated by PDGF-BB. MSC-exo treatment reduced the expression of both YAP1 and SPP1. Lentivirus-mediated knockdown of YAP1 inhibited PDGF-BB-induced PASMC proliferation, migration, and SPP1 protein levels. CONCLUSION: Our findings demonstrate that MSC-exo exert a therapeutic effect against hypoxia-induced pulmonary hypertension by modulating the YAP1/SPP1 signaling pathway. The inhibition of YAP1 and downstream SPP1 expression by MSC-exo may contribute to the attenuation of pulmonary vascular remodeling and PASMC proliferation and migration. These results suggest that MSC-exo could serve as a potential therapeutic strategy for the treatment of HPH. Further investigations are warranted to explore the clinical applicability of MSC-exo-based therapies in HPH patients.


Asunto(s)
Exosomas , Hipertensión Pulmonar , Células Madre Mesenquimatosas , Humanos , Ratas , Masculino , Animales , Hipertensión Pulmonar/metabolismo , Osteopontina/metabolismo , Exosomas/metabolismo , Becaplermina/farmacología , Remodelación Vascular , Ratas Sprague-Dawley , Hipoxia/metabolismo , Transducción de Señal , Arteria Pulmonar/metabolismo , Células Madre Mesenquimatosas/metabolismo , Miocitos del Músculo Liso/metabolismo , Proliferación Celular , Células Cultivadas
17.
Immunol Lett ; 263: 113-122, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37875238

RESUMEN

The role of inflammation in pulmonary hypertension is gradually gaining increasing research attention. However, no previous study has evaluated the characteristics of inflammation during chronic hypoxia-induced pulmonary hypertension. Therefore, the aim of this study was to investigate the characteristics of the inflammatory process involved in hypoxia-induced pulmonary hypertension in mice. The current study evaluated from day 4 to day 28 of hypoxia, the PAAT and PAAT/PET decreased, accompanied by pulmonary vascular remodeling and right ventricular hypertrophy, as well as increased numbers of CD68 macrophages. The expression of the pro-inflammatory factors IL-1ß and IL-33 increased, but decreased on day 28. The expression of IL-12 increased from day 4 to day 28, whereas that of the anti-inflammatory factor IL-10 in lung tissue decreased. Furthermore, the expression of the IL-33/ST2 signaling pathway also increased over time under hypoxic conditions. In conclusion, pulmonary artery remodeling in HPH mice worsens progressively in a time-dependent manner, with inflammatory cell infiltration predominating in the early stage and pulmonary vascular remodeling occurring in the later stage.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Ratones , Animales , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/complicaciones , Interleucina-33 , Remodelación Vascular , Inflamación/complicaciones , Macrófagos/metabolismo , Hipoxia
18.
Mol Med ; 29(1): 119, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37670258

RESUMEN

Small ubiquitin-like modifier mediated modification (SUMOylation) is a critical post-translational modification that has a broad spectrum of biological functions, including genome replication and repair, transcriptional regulation, protein stability, and cell cycle progression. Perturbation or deregulation of a SUMOylation and deSUMOylation status has emerged as a new pathophysiological feature of lung diseases. In this review, we highlighted the link between SUMO pathway and lung diseases, especially the sumoylated substrate such as C/EBPα in bronchopulmonary dysplasia (BDP), PPARγ in pneumonia, TFII-I in asthma, HDAC2 in chronic obstructive pulmonary disease (COPD), KLF15 in hypoxic pulmonary hypertension (HPH), SMAD3 in idiopathic pulmonary fibrosis (IPF), and YTHDF2 in cancer. By exploring the impact of SUMOylation in pulmonary diseases, we intend to shed light on its potential to inspire the development of innovative diagnostic and therapeutic strategies, holding promise for improving patient outcomes and overall respiratory health.


Asunto(s)
Asma , Displasia Broncopulmonar , Enfermedad Pulmonar Obstructiva Crónica , Recién Nacido , Humanos , Sumoilación , Hipoxia
19.
Front Physiol ; 14: 1239643, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37645564

RESUMEN

Mitochondria are the centrol hub for cellular energy metabolisms. They regulate fuel metabolism by oxygen levels, participate in physiological signaling pathways, and act as oxygen sensors. Once oxygen deprived, the fuel utilizations can be switched from mitochondrial oxidative phosphorylation to glycolysis for ATP production. Notably, mitochondria can also adapt to hypoxia by making various functional and phenotypes changes to meet the demanding of oxygen levels. Hypoxic pulmonary hypertension is a life-threatening disease, but its exact pathgenesis mechanism is still unclear and there is no effective treatment available until now. Ample of evidence indicated that mitochondria play key factor in the development of hypoxic pulmonary hypertension. By hypoxia-inducible factors, multiple cells sense and transmit hypoxia signals, which then control the expression of various metabolic genes. This activation of hypoxia-inducible factors considered associations with crosstalk between hypoxia and altered mitochondrial metabolism, which plays an important role in the development of hypoxic pulmonary hypertension. Here, we review the molecular mechanisms of how hypoxia affects mitochondrial function, including mitochondrial biosynthesis, reactive oxygen homeostasis, and mitochondrial dynamics, to explore the potential of improving mitochondrial function as a strategy for treating hypoxic pulmonary hypertension.

20.
Exp Cell Res ; 431(2): 113755, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37586455

RESUMEN

Hypoxia-induced pulmonary hypertension is a subgroup of type 3 pulmonary hypertension (PH) with the recommended treatment limited to oxygen therapy and lacks potential therapeutic targets. To investigate the role of NLRC3 in hypoxia-induced PH and its potential mechanism, we first collected lung tissues of high-altitude pulmonary hypertension (HAPH) patients. Immunohistochemistry and immunofluorescence showed that NLRC3 was downregulated and was mainly co-localized with the smooth muscle cells of the pulmonary vessels in HAPH patients. Besides, we found that NLRC3 was also expressed in endothelial cells in HAPH patients for the first time. Then, wild type (WT) and NLRC3 knockout (NLRC3-/-) mice were used to construct hypoxia models and primary pulmonary arterial smooth muscle cells (PASMCs) of rats and endothelial cells were cultured for verification. Right heart catheterization and echocardiography suggested that NLRC3 knockout promoted right ventricular systolic pressure (RVSP) up-regulation, right ventricular hypertrophy and fibrosis in hypoxia-induced mice. This study first demonstrated that NLRC3 deficiency promoted hypoxia-stimulated PASMCs proliferation, Human umbilical vein endothelial cells (HUVECs) apoptosis, migration and inflammation through IKK/NF-κB p65/HIF-1α pathway in vitro and in vivo, further promoted vascular remodeling and PH progression, which provided a new target for the treatment of hypoxia-induced PH.


Asunto(s)
Hipertensión Pulmonar , Animales , Humanos , Ratones , Ratas , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Arteria Pulmonar/metabolismo , Remodelación Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA