Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Med (Lausanne) ; 10: 1251827, 2023.
Article in English | MEDLINE | ID: mdl-38034531

ABSTRACT

Background: Asthma is a chronic respiratory disease and is often associated with multiple comorbidities. The causal relationship between asthma and these comorbidities is still unclear. This study aimed to investigate the association between genetically predicted asthma and common comorbidities. Methods: After searching PubMed and GWAS summary statistics, we identified 26 comorbidities of asthma. The causal relationship between asthma and comorbidities was assessed in two independent GWASs by bidirectional Mendelian randomization, followed by validation of the results using a multivariate Mendelian randomization analysis and several sensitivity analyses. Results: In the bidirectional Mendelian randomization analysis, chronic sinusitis [odds ratio (OR) = 1.54, p = 1.40 × 10-5], atopic dermatitis (OR = 1.36, p = 9.37 × 10-21), allergic conjunctivitis (OR = 2.07, p = 4.32 × 10-6), and allergic rhinitis (OR = 1.53, p = 5.20 × 10-6) were significantly associated with increased asthma risk. Hyperthyroidism (OR = 1.12, p = 0.04) had a potential increased risk for asthma. For the reverse direction, asthma showed significant associations with an increased risk of chronic obstructive pulmonary disease (OR = 1.24, p = 2.25 × 10-9), chronic sinusitis (OR = 1.61, p = 5.25 × 10-21), atopic dermatitis (OR = 2.11, p = 1.24 × 10-24), allergic conjunctivitis (OR = 1.65, p = 6.66 × 10-35), allergic rhinitis (OR = 1.90, p = 2.38 × 10-57), and a potential higher risk of allergic urticaria (OR = 1.25, p = 0.003). Conclusion: This study suggested a significant bidirectional association of chronic sinusitis, atopic dermatitis, allergic conjunctivitis, and allergic rhinitis with asthma. In addition, hyperthyroidism was associated with an increased risk of asthma and asthma increased the risk of chronic obstructive pulmonary disease and allergic urticaria.

2.
Am J Transl Res ; 14(3): 1578-1591, 2022.
Article in English | MEDLINE | ID: mdl-35422917

ABSTRACT

OBJECTIVES: Regulated in development and DNA damage responses 1 (REDD1) is an important transcription factor regulating mitochondria homeostasis, which is the important pathological alteration of pulmonary hypertension (PH). However, it is unclear whether REDD1 regulates the PASMCs mitochondria homeostasis by the similar mechanism in pulmonary arterial remodeling induced by hypoxia. METHODS: The global REDD1-knockout rats (REDD1-KO) on Sprague-Dawley background were used to generate a chronic hypoxia model of PH. Right ventricular hypertrophy and vascular remodeling were detected after exposure to hypoxia. Additionally, proliferation, apoptosis, migration, mitochondria homeostasis, and autophagy were performed in vivo and in vitro. RESULTS: The current research found that in human and experimental rats of PH, REDD1 expression is upregulated in the PASMCs. REDD1 gene knockout alleviated hypoxia PH and hemodynamic changes effectively and reversed hypoxic pulmonary vascular remodeling. In addition, REDD1 knockdown reduces the impairment of mitochondrial function caused by hypoxia in HPASMCs via autophagy inhibition, and this process may be regulated through the Parkin gene. Moreover, REDD1 knockdown can effectively inhibit the proliferation and migration of hypoxic PASMCs, and induce their apoptosis in vivo and in vitro. CONCLUSIONS: Our results suggested that REDD1 might be a potential target for improved pulmonary vascular remodeling in PH.

3.
Front Immunol ; 13: 824188, 2022.
Article in English | MEDLINE | ID: mdl-35444652

ABSTRACT

Exosomes are small extracellular vesicles that are secreted by almost all types of cells and exist in almost all extracellular spaces. As an important mediator of intercellular communication, exosomes encapsulate the miRNA, lncRNA, cirRNA, mRNA, cytokine, enzyme, lipid, and other components from the cytoplasm into its closed single membrane structure and transfer them to recipient units in an autocrine, paracrine, or endocrine manner. Hypoxia is a state of low oxygen tension and is involved in many pathological processes. Hypoxia influences the size, quantity, and expression of exosome cargos. Exosomes derived from hypoxic tumor cells transfer genetics, proteins, and lipids to the recipient units to exert pleiotropic effects. Different donor cells produce different cargo contents, target different recipient units and lead to different biological effects. Hypoxic exosomes derived from tumor cells uptaken by normoxic tumor cells lead to promoted proliferation, migration, and invasion; uptaken by extracellular space or liver lead to promoted metastasis; uptaken by endothelial cells lead to promoted angiogenesis; uptaken by immune cells lead to promoted macrophage polarization and changed tumor immune microenvironment. In addition to various types of tumors, hypoxic exosomes also participate in the development of diseases in the cardiovascular system, neuron system, respiratory system, hematology system, endocrine system, urinary system, reproduction system, and skeletomuscular system. Understanding the special characteristics of hypoxic exosomes provide new insight into elaborating the pathogenesis of hypoxia related disease. This review summarizes hypoxia induced cargo changes and the biological effects of hypoxic exosomes in tumors and non-malignant diseases in different systems.


Subject(s)
Exosomes , Neoplasms , Cell Communication , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , Hypoxia/metabolism , Neoplasms/metabolism , Tumor Microenvironment
4.
Int J Mol Med ; 49(3)2022 Mar.
Article in English | MEDLINE | ID: mdl-35059734

ABSTRACT

Spliced X­box binding protein 1 (XBP1s) has been reported to participate in the pathogenesis of numerous types of cancer; however, whether XBP1s plays a role in lung cancer remains to be elucidated. In the present study, bioinformatics analysis was performed to determine the mRNA expression level of XBP1 in lung cancer and adjacent normal tissues. Gene Ontology terms, pathway enrichment and Pearson's correlation analysis were performed to investigate the possible mechanism involved. Western blot and reverse transcription­quantitative PCR were performed to quantify the protein and mRNA expression level of target proteins, respectively. Small interfering RNA or overexpression plasmid were used to knockdown or overexpress the expression level of XBP1s. EdU staining, colony formation, Cell Counting Kit­8, Transwell and wound healing assays, and flow cytometry were performed to detect the proliferation, colony forming ability, cell viability, migration and invasion ability, and the apoptosis rate. The results showed that the mRNA and protein expression level of XBP1 was higher in tumor tissues compared with that in adjacent normal tissues using data from the TIMER2.0, ONCOMINE and UALCAN online databases. In addition, the mRNA expression level of XBP1 was also associated with clinical features, including age, smoking habit, individual cancer stage and nodal metastasis status. In the in vitro experiments, the mRNA and protein expression level of XBP1s was increased in the A549 cell line compared with that in the human bronchial epithelial (HBE), H1299, PC9 and H460 cell lines. Hypoxia further increased the protein expression level of XBP1s in the A549 cell line. Knockdown of XBP1s expression in the A549 cell line resulted in decreased proliferation, colony formation, cell viability, migration and invasion, and increased apoptosis. By contrast, overexpressing XBP1s in the HBE cell line led to the opposite results. To investigate the mechanism involved, proteins associated with XBP1 were analyzed using the LinkedOmics database. Pathway enrichment revealed the MAPK pathway to be the possible XBP1 downstream target. Furthermore, Pearson's correlation and western blot analyses verified that phosphorylated (p)­JNK rather than p­ERK or p­p38 was the downstream effector of XBP1s. Phosphorylation of JNK was decreased when XBP1s expression was knocked down in the A549 cell line under normoxic and hypoxic conditions. Inhibiting p­JNK with SP600125 reversed the increased prosurvival effects caused by XBP1s overexpression. The results from the present study suggest that XBP1s/p­JNK function as a prosurvival factors in the A549 cell line and could be a potential target for the treatment of lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , X-Box Binding Protein 1 , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , MAP Kinase Signaling System , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
5.
Respir Res ; 23(1): 6, 2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35016680

ABSTRACT

BACKGROUND: Hypoxic pulmonary hypertension (HPH) is a chronic progressive advanced disorder pathologically characterized by pulmonary vascular remodeling. Notch4 as a cell surface receptor is critical for vascular development. However, little is known about the role and mechanism of Notch4 in the development of hypoxic vascular remodeling. METHODS: Lung tissue samples were collected to detect the expression of Notch4 from patients with HPH and matched controls. Human pulmonary artery smooth muscle cells (HPASMCs) were cultured in hypoxic and normoxic conditions. Real-time quantitative PCR and western blotting were used to examine the mRNA and protein levels of Notch4. HPASMCs were transfected with small interference RNA (siRNA) against Notch4 or Notch4 overexpression plasmid, respectively. Cell viability, cell proliferation, apoptosis, and migration were assessed using Cell Counting Kit-8, Edu, Annexin-V/PI, and Transwell assay. The interaction between Notch4 and ERK, JNK, P38 MAPK were analyzed by co-immunoprecipitation. Adeno-associated virus 1-mediated siRNA against Notch4 (AAV1-si-Notch4) was injected into the airways of hypoxic rats. Right ventricular systolic pressure (RVSP), right ventricular hypertrophy and pulmonary vascular remodeling were evaluated. RESULTS: In this study, we demonstrate that Notch4 is highly expressed in the media of pulmonary vascular and is upregulated in lung tissues from patients with HPH and HPH rats compared with control groups. In vitro, hypoxia induces the high expression of Delta-4 and Notch4 in HPASMCs. The increased expression of Notch4 promotes HPASMCs proliferation and migration and inhibits cells apoptosis via ERK, JNK, P38 signaling pathways. Furthermore, co-immunoprecipitation result elucidates the interaction between Notch4 and ERK/JNK/P38. In vivo, silencing Notch4 partly abolished the increase in RVSP and pulmonary vascular remodeling caused by hypoxia in HPH rats. CONCLUSIONS: These findings reveal an important role of the Notch4-ERK/JNK/P38 MAPK axis in hypoxic pulmonary remodeling and provide a potential therapeutic target for patients with HPH.


Subject(s)
Gene Expression Regulation , Hypertension, Pulmonary/genetics , Hypoxia/complications , Myocytes, Smooth Muscle/metabolism , Receptor, Notch4/genetics , Vascular Remodeling/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypoxia/genetics , Hypoxia/metabolism , MAP Kinase Signaling System , Male , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Receptor, Notch4/biosynthesis , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases/biosynthesis
6.
J Cell Physiol ; 237(3): 1948-1963, 2022 03.
Article in English | MEDLINE | ID: mdl-34964131

ABSTRACT

Hypoxic pulmonary hypertension (HPH) is characterized by elevated pulmonary artery resistance and vascular remodeling. Endoplasmic reticulum stress (ERS) is reported to be involved in HPH, but the underlying mechanisms remain uncertain. We found that Xbp1s, a potent transcription factor during ERS, was elevated in hypoxic-cultured rat PASMCs and lung tissues from HPH rats. Our in vitro experiments demonstrated that overexpressing Xbp1s can promote proliferation, cell viability, and migration and inhibit the apoptosis of PASMCs, while silencing Xbp1s led to the opposite. Through data-independent acquisition (DIA) mass spectrometry, we identified extensive proteomic alterations regulated by hypoxia and Xbp1s. Further validation revealed that p-JNK, rather than p-ERK or p-p38, was the downstream effector of Xbp1s. p-JNK inhibition reversed the biological effects of Xbp1s overexpression in vitro. In the animal HPH model, rats were randomly assigned to five groups: normoxia, hypoxia, hypoxia+AAV-CTL (control), hypoxia+AAV-Xbp1s (prevention), and hypoxia+AAV-Xbp1s (therapy). Adeno-associated virus (AAV) serotype 1-mediated Xbp1s knockdown in the prevention and therapy groups significantly reduced right ventricular systolic pressure, total pulmonary resistance, right ventricular hypertrophy, and the medial wall thickness of muscularized distal pulmonary arterioles; AAV-Xbp1s also decreased proliferating cell nuclear antigen expression and increased apoptosis in pulmonary arterioles. Collectively, our findings demonstrated that the Xbp1s-p-JNK pathway is important in hypoxic vascular remodeling and that targeting this pathway could be an effective strategy to prevent and alleviate HPH development.


Subject(s)
Hypertension, Pulmonary , MAP Kinase Signaling System , X-Box Binding Protein 1 , Animals , Cell Proliferation/genetics , Disease Models, Animal , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Myocytes, Smooth Muscle/metabolism , Proteomics , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Vascular Remodeling , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
7.
Respir Res ; 22(1): 312, 2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34906150

ABSTRACT

BACKGROUND: Hypoxic pulmonary hypertension (PH) is a refractory pulmonary vascular remodeling disease, and the efficiency of current PH treatment strategies is unsatisfactory. Tribbles homolog 3 (TRB3), a member of the pseudokinase family, is upregulated in diverse types of cellular stresses and functions as either a pro-proliferative or pro-apoptotic factor depending on the specific microenvironment. The regulatory mechanisms of TRB3 in hypoxic PH are poorly understood. METHODS: We performed studies using TRB3-specific silencing and overexpressing lentiviral vectors to investigate the potential roles of TRB3 on hypoxic pulmonary artery smooth muscle cells (PASMCs). Adeno-associated virus type 1(AVV1) vectors encoding short-hairpin RNAs against rat TRB3 were used to assess the role of TRB3 on hypoxic PH. TRB3 protein expression in PH patients was explored in clinical samples by western blot analysis. RESULTS: The results of whole-rat genome oligo microarrays showed that the expression of TRB3 and endoplasmic reticulum stress (ERS)-related genes was upregulated in hypoxic PASMCs. TRB3 protein expression was significantly upregulated by hypoxia and thapsigargin. In addition, 4-PBA and 4µ8C, both inhibitors of ERS, decreased the expression of TRB3. TRB3 knockdown promoted apoptosis and damaged the proliferative and migratory abilities of hypoxic PASMCs as well as inhibited activation of the MAPK signaling pathway. TRB3 overexpression stimulated the proliferation and migration of PASMCs but decreased the apoptosis of PASMCs, which was partly reversed by specific inhibitors of ERK, JNK and p38 MAPK. The Co-IP results revealed that TRB3 directly interacts with ERK, JNK, and p38 MAPK. Knockdown of TRB3 in rat lung tissue reduced the right ventricular systolic pressure and decreased pulmonary medial wall thickness in hypoxic PH model rats. Further, the expression of TRB3 in lung tissues was higher in patients with PH compared with those who have normal pulmonary artery pressure. CONCLUSIONS: TRB3 was upregulated in hypoxic PASMCs and was affected by ERS. TRB3 plays a key role in the pathogenesis of hypoxia-induced PH by binding and activating the ERK, JNK, and p38 MAPK pathways. Thus, TRB3 might be a promising target for the treatment of hypoxic PH.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Hypertension, Pulmonary/genetics , Hypoxia/complications , MAP Kinase Signaling System/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Vascular Remodeling/genetics , Animals , Apoptosis , Cell Communication , Disease Models, Animal , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Male , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation
8.
Clin Sci (Lond) ; 135(21): 2467-2481, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34676402

ABSTRACT

Pulmonary hypertension (PH) is a life-threatening disease characterized by vascular remodeling. Exploring new therapy target is urgent. The purpose of the present study is to investigate whether and how spliced x-box binding protein 1 (xbp1s), a key component of endoplasmic reticulum stress (ERS), contributes to the pathogenesis of PH. Forty male SD rats were randomly assigned to four groups: Control, Monocrotaline (MCT), MCT+AAV-CTL (control), and MCT+AAV-xbp1s. The xbp1s protein levels were found to be elevated in lung tissues of the MCT group. Intratracheal injection of adeno-associated virus serotype 1 carrying xbp1s shRNA (AAV-xbp1s) to knock down the expression of xbp1s effectively ameliorated the MCT-induced elevation of right ventricular systolic pressure (RVSP), total pulmonary resistance (TPR), right ventricular hypertrophy and medial wall thickness of muscularized distal pulmonary arterioles. The abnormally increased positive staining rates of proliferating cell nuclear antigen (PCNA) and Ki67 and decreased positive staining rates of terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) in pulmonary arterioles were also reversed in the MCT+AAV-xbp1s group. For mechanistic exploration, bioinformatics prediction of the protein network was performed on the STRING database, and further verification was performed by qRT-PCR, Western blots and co-immunoprecipitation (Co-IP). DNA damage-inducible transcript 3 (Ddit3) was identified as a downstream protein that interacted with xbp1s. Overexpression of Ddit3 restored the decreased proliferation, migration and cell viability caused by silencing of xbp1s. The protein level of Ddit3 was also highly consistent with xbp1s in the animal model. Taken together, our study demonstrated that xbp1s-Ddit3 may be a potential target to interfere with vascular remodeling in PH.


Subject(s)
Arterial Pressure , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transcription Factor CHOP/metabolism , Vascular Remodeling , X-Box Binding Protein 1/metabolism , Animals , Apoptosis , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Male , Monocrotaline , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Signal Transduction , Transcription Factor CHOP/genetics , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right , X-Box Binding Protein 1/genetics
9.
Am J Transl Res ; 13(4): 2283-2295, 2021.
Article in English | MEDLINE | ID: mdl-34017389

ABSTRACT

Irreversible pulmonary hypertension (PH) mainly results from vascular remodeling, in which the aberrant growth of pulmonary arterial smooth muscle cells (PASMCs) plays a significant role. Our previous work suggested that KLF5 and HIF1α are closely associated with the pathogenesis of hypoxic PH as they intervene in the growth of PASMCs. MicroRNAs (miRNAs) have been demonstrated to be involved in the control of cell proliferation and apoptosis. In the present study, we detected the expression of six miRNAs connected with KLF5 in hypoxia-exposed rat PH models and PASMCs and then further investigated the role of miR-320-3p in the abnormal proliferation of hypoxic PASMCs and in the progression and treatment outcomes of hypoxia-induced PH. The results indicated that miR-320-3p was downregulated in hypoxia-exposed rat PH models, hypoxia-induced PASMCs and chronic thromboembolic pulmonary hypertension (CTEPH) patients. Moreover, miR-320-3p directly regulated the expression of KLF5 and HIF1α. miR-320-3p mimics inhibited proliferation and migration and promoted apoptosis in hypoxic PASMCs. KLF5 and HIF1α reversed the above effects of miR-320-3p. In conclusion, miR-320-3p plays a certain role in the progression of hypoxic PH via KLF5 and HIF1α and might be a potent therapeutic tool for PH.

10.
Biochem Biophys Res Commun ; 557: 40-47, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33862458

ABSTRACT

Hypoxic pulmonary vascular remodeling is a pathological feature of pulmonary hypertension (PH). Our results showed that centromere-associated protein E (CENPE) expression in PH patients and hypoxia-induced PH rats was significantly higher than that in normal controls. In addition, CENPE deficiency significantly inhibited the development of pulmonary vascular remodeling and right ventricular hypertrophy. Moreover, knocking out CENPE effectively inhibited the proliferation and induced the apoptosis of primary pulmonary artery smooth muscle cells (PASMCs) in vivo. Furthermore, CENPE silencing by small interference significantly inhibited abnormal proliferation, apoptosis resistance, migration, and cell cycle arrest in hypoxia-induced PASMCs. Interestingly, we found that CENPE might exert its biological effect by targeting the transcription of CDK1 proteins.


Subject(s)
CDC2 Protein Kinase/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/pathology , Pulmonary Artery/pathology , Vascular Remodeling/physiology , Animals , CDC2 Protein Kinase/genetics , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Female , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Male , Middle Aged , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
11.
J Int Med Res ; 49(1): 300060520986689, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33478317

ABSTRACT

Pulmonary veno-occlusive disease (PVOD) is a rare type of pulmonary hypertension characterized by capillary damage or arterial pulmonary hypertension. Early lung transplantation is the only effective treatment for PVOD because of the lack of specificity in its clinical manifestations and its rapid progression and poor prognosis. A 28-year-old woman presented with exertional dyspnoea. A chest computed tomography scan revealed diffuse centrilobular ground glass opacities in both lungs, a ratio of the transverse diameter of the main pulmonary trunk to the ascending aorta of >1, and enlargement of the right ventricle and right atrium. A right atrial floating catheter test showed right ventricular pressure of 82/0/4 mmHg, mean pulmonary artery pressure of 83/34/53 mmHg, and pulmonary artery wedge pressure of 15/8/12 mmHg. A mutation was found in the eukaryotic translation initiation factor 2 alpha kinase 4 (EIF2AK4) gene. Thus, the patient was diagnosed with PVOD and subsequently given standard bosentan treatment (62.5 mg twice a day). However, after 6 months of follow-up, there was no significant improvement in the pulmonary artery pressure or activity tolerance (6-minute walking test). Therefore, cardiopulmonary transplantation was performed. Early diagnosis and timely treatment of PVOD may improve the patient's prognosis.


Subject(s)
Hypertension, Pulmonary , Lung Transplantation , Pulmonary Veno-Occlusive Disease , Adult , Dyspnea , Female , Humans , Hypertension, Pulmonary/diagnostic imaging , Hypertension, Pulmonary/drug therapy , Lung , Pulmonary Veno-Occlusive Disease/complications , Pulmonary Veno-Occlusive Disease/diagnostic imaging , Pulmonary Veno-Occlusive Disease/surgery
12.
Front Cell Dev Biol ; 9: 780121, 2021.
Article in English | MEDLINE | ID: mdl-34988077

ABSTRACT

Hypoxia contributes to the progression and metastasis of lung adenocarcinoma (LUAD). However, the specific underlying molecular mechanisms have not been fully elucidated. Here we report that Notch4 is upregulated in lung tissue from lung cancer patients. Functionally, Hypoxia activates the expressions of Delta-like 4 and Notch4, resulting in the excessive proliferation and migration of LUAD cells as well as apoptotic resistance. Notch4 silencing reduced ERK, JNK, and P38 activation. Meanwhile, Notch4 overexpression enhanced ERK, JNK, and P38 activation in LUAD cells. Furthermore, Notch4 exerted pro-proliferation, anti-apoptosis and pro-migration effects on LUAD cells that were partly reversed by the inhibitors of ERK, JNK, and p38. The binding interaction between Notch4 and ERK/JNK/P38 were confirmed by the co-immunoprecipitation assay. In vivo study revealed that Notch4 played a key role in the growth and metastasis of LUAD using two xenograft models. This study demonstrates that hypoxia activates Notch4-ERK/JNK/P38 MAPK signaling pathways to promote LUAD cell progression and metastasis.

13.
Med Sci Monit ; 26: e927212, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32883943

ABSTRACT

BACKGROUND The rapid worldwide spread of the coronavirus disease 2019 (COVID-19) epidemic has placed patients with pre-existing conditions at risk of severe morbidity and mortality. The present study investigated the clinical characteristics and outcomes of patients with severe COVID-19 and chronic obstructive pulmonary disease (COPD). MATERIAL AND METHODS This study enrolled 336 consecutive patients with confirmed severe COVID-19, including 28 diagnosed with COPD, from January 20, 2020, to April 1, 2020. Demographic data, symptoms, laboratory values, comorbidities, and clinical results were measured and compared in survivors and non-survivors. RESULTS Patients with severe COVID-19 and COPD were older than those without COPD. The proportions of men, of patients admitted to the intensive care unit (ICU) and of those requiring invasive ventilation were significantly higher in patients with than without COPD. Leukocyte and neutrophil counts, as well as the concentrations of NT-proBNP, hemoglobin, D-dimer, hsCRP, ferritin, IL-2R, TNF-alpha and procalcitonin were higher, whereas lymphocyte and monocyte counts were lower, in patients with than without COPD. Of the 28 patients with COPD, 22 (78.6%) died, a rate significantly higher than in patients without COPD (36.0%). A comparison of surviving and non-surviving patients with severe COVID-19 and COPD showed that those who died had a longer history of COPD, more fatigue, and a higher ICU occupancy rate, but a shorter average hospital stay, than those who survived. CONCLUSIONS COPD increases the risks of death and negative outcomes in patients with severe COVID-19.


Subject(s)
Betacoronavirus , Coronavirus Infections/epidemiology , Pandemics , Pneumonia, Viral/epidemiology , Pulmonary Disease, Chronic Obstructive/epidemiology , Age Distribution , Aged , Biomarkers , COVID-19 , Cardiovascular Diseases/epidemiology , China/epidemiology , Comorbidity , Coronavirus Infections/blood , Coronavirus Infections/therapy , Critical Care , Diabetes Mellitus/epidemiology , Fatigue/etiology , Female , Humans , Length of Stay/statistics & numerical data , Male , Middle Aged , Neoplasms/epidemiology , Pneumonia, Viral/blood , Pneumonia, Viral/therapy , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/therapy , Renal Insufficiency, Chronic/epidemiology , Respiration, Artificial , SARS-CoV-2 , Sex Distribution , Survivors , Treatment Outcome
14.
Chem Biol Interact ; 317: 108942, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31930969

ABSTRACT

Pulmonary hypertension (PH) is distal pulmonary arterial remodelling and is mainly due to the abnormal proliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs). Apigenin, a natural dietary flavonoid, is a promising PH preventive agent that inhibits PASMC proliferation and induces apoptosis. In this study, we investigated the biological effects of apigenin on PH. PH was induced in male Sprague-Dawley rats by chronic hypoxia exposure. Administration of apigenin prevented the development of PH, hypoxia-induced right ventricular hypertrophy and pulmonary arterial remodelling and prevented the progression of established PH in this model. Moreover, treatment with apigenin induced mitochondria-dependent apoptosis. To explore the underlying mechanisms, the mitochondrial membrane potential (Δψm) and the mitochondria-dependent apoptosis factors cytochrome C, BAX, Bcl-2, cleaved caspase 3, and cleaved caspase 9 were analysed. These results confirmed that apigenin induces mitochondria-dependent apoptosis in hypoxic PASMCs to protect against PH. In addition, treatment with apigenin reversed hypoxia-induced inhibition of KV1.5 expression both in vivo and in vitro. The KV1.5 inhibitor diphenyl phosphine oxide-1 (DPO-1) abrogated apigenin-induced mitochondria-dependent apoptosis in hypoxic PASMCs, suggesting that KV1.5 is implicated in apigenin-induced mitochondria-dependent apoptosis. Furthermore, functional studies revealed that apigenin activated mitochondria-dependent apoptosis by modulation of hypoxia-induced factor 1α (HIF-1α) signalling. Together, our study shows that apigenin attenuates PH via inhibiting the HIF-1α-KV1.5 channel pathway to induce PASMC mitochondria-dependent apoptosis.


Subject(s)
Apigenin/therapeutic use , Apoptosis/drug effects , Hypertension, Pulmonary/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , Myocytes, Smooth Muscle/drug effects , Animals , Cell Proliferation , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Hypertension, Pulmonary/complications , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Male , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/cytology , Rats , Rats, Sprague-Dawley
15.
J Cell Physiol ; 235(1): 538-547, 2020 01.
Article in English | MEDLINE | ID: mdl-31256425

ABSTRACT

Tribbles homolog 3 (TRB3) has been accounted for regulation of a few cell processes through interaction with other significant proteins. The molecular mechanisms underlying TRB3 in tumorigenesis in lung adenocarcinoma have not been entirely elucidated. The present study is aimed at determining the function and fundamental mechanisms of TRB3 in lung adenocarcinoma progression. TRB3 was highly expressed in A549 and H1299 cells and lung adenocarcinoma tissues compared with human bronchial epithelial cells (HBEpC) and adjacent normal lung tissues. Hypoxia significantly upregulated the expression of TRB3 protein in A549 and H1299 cells in a time-dependent way. Gene expression profiling interactive analysis data analysis indicated that patients with lung adenocarcinoma with excessive expression of TRB3 mRNA had fundamentally shorter survival time. TRB3 knockdown in A549 cells can inhibit cell proliferation and migration, and promote cell apoptosis. TRB3 knockdown reduced the expression of p-ERK and p-JNK, but did not affect the expression of p-P38 MAPK. TRB3 overexpression enhances the malignant transformation abilities of HBEpC such as cell proliferation, migration and colony formation, which could be reversed by U0126 and SP600125. TRB3 overexpression promotes the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) but was not affected by U0126 and SP600125. The results of coimmunoprecipitation experiments indicated that TRB3 binds directly to ERK and JNK. This study suggests that TRB3 has a potentially carcinogenic role in lung adenocarcinoma by binding to ERK and JNK and promoting the phosphorylation of ERK and JNK. TRB3 can be a possible therapeutic focus for lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/pathology , Cell Cycle Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/genetics , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , MAP Kinase Signaling System , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Am J Transl Res ; 11(2): 641-654, 2019.
Article in English | MEDLINE | ID: mdl-30899368

ABSTRACT

Hypoxia is a common cause of pulmonary vascular remodeling and endoplasmic reticulum stress (ERS). Upon ER stress, the unfolded protein response (UPR) which activates the IRE1α, PERK and ATF6 signaling pathways is activated to cope with ERS in mammalian cells; however, the role of the three UPR arms in pulmonary vascular remodeling has not been defined. The present study showed that GRP78, a marker of ERS, was upregulated in hypoxic pulmonary artery smooth muscle cells (PASMCs). Among the three arms of the UPR, the IRE1α pathway was noticeably upregulated in hypoxic PASMCs. An inhibitor of IRE1α/XBP1 pathway, 4u8c, inhibited hypoxia-induced cell proliferation and migration and increased cell apoptosis by downregulating PCNA and MMP9 and activating mitochondrial apoptosis by enhancing the expression of BAX, activating caspase-9 and caspase-3, and eventually cleaving PARP. Quercetin affects ERS in many cell types and was shown to relieve hypoxic pulmonary hypertension (HPH) in our previous study. We demonstrated that quercetin evoked excessive GRP78 expression in hypoxic PASMCs compared with hypoxia alone by evaluating the expression of GRP78. The expression of IRE1α and XBP1s, a cleavage form of XBP1u, was upregulated by quercetin in a dose-dependent manner. Pretreatment with 4u8c reversed the apoptosis-promoting effect of quercetin by inhibiting mitochondrial apoptosis. However, 4u8c amplified the effect of quercetin on proliferation and migration in hypoxic PASMCs. In conclusion, the study demonstrated that the IRE1α-XBP1 pathway is involved in the process of hypoxia-induced pulmonary vascular remodeling; 4u8c could restrain hypoxia-induced cell proliferation and migration and reverse the hypoxia-induced apoptosis arrest, while quercetin excited excessive ERS and the IRE1α pathway in hypoxic PASMCs and promoted apoptosis. Our data suggest that intervening the IRE1α-XBP1 pathway may be useful for hypoxia-induced pulmonary arterial hypertension therapy.

17.
Free Radic Biol Med ; 103: 165-176, 2017 02.
Article in English | MEDLINE | ID: mdl-27979659

ABSTRACT

Quercetin, an important dietary flavonoid has been demonstrated to potentially reverse or even prevent pulmonary arterial hypertension (PAH) progression. However, the effects of quercetin on apoptosis and autophagy in pulmonary arterial smooth muscle cells (PASMCs) have not yet been clearly elucidated. The current study found that quercetin significantly induce the apoptotic and autophagic capacities of PASMCs in vitro and in vivo in hypoxia. In addition, we found that quercetin increases FOXO1 (a major mediator in autophagy regulation) expression and transcriptional activity. Moreover, FOXO1 knockdown by siRNAs inhibited the phosphorylation of mTOR and 4E-BPI, which is downstream of P70-S6K, and markedly blocked quercetin-induced autophagy. We also observed that FOXO1-mediated autophagy was achieved via SESN3 not Rictor upregulation and after mTOR suppression. Furthermore, Treatment with autophagy-specific inhibitors could markedly enhance quercetin-induced apoptosis in PASMCs under hypoxia. Finally, quercetin in combination with autophagy inhibition treatment could enhance the therapeutic effects of quercetin in hypoxia-associated PAH in vivo. Taken together, quercetin could enhance hypoxia-induced autophagy through the FOXO1-SENS3-mTOR pathway in PASMCs. Combining quercetin and autophagy inhibitors may be a novel therapeutic strategy for treating hypoxia-associated PAH.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Myocytes, Smooth Muscle/physiology , Nerve Tissue Proteins/metabolism , Quercetin/pharmacology , Animals , Cell Hypoxia , Cells, Cultured , Drug Evaluation, Preclinical , Gene Expression/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Nerve Tissue Proteins/genetics , Oxidative Stress , Pulmonary Artery/cytology , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , Transcriptional Activation/drug effects
18.
Ann Allergy Asthma Immunol ; 117(4): 417-422.e1, 2016 10.
Article in English | MEDLINE | ID: mdl-27590640

ABSTRACT

BACKGROUND: The association between MBL2 gene polymorphisms and the risk of asthma has been evaluated in multiple studies; however, the results are inconsistent. OBJECTIVE: To perform a meta-analysis to explore whether MBL2 gene polymorphisms were associated with the risk of asthma. METHODS: We searched PubMed, Web of Science, and Cochrane Library to find relevant articles published up to March 2016. Nine studies, including 2066 cases and 2183 controls, were included in the meta-analysis. The strength of association was evaluated by odds ratio (OR) with 95% confidence interval (CI). RESULTS: The results reveal that MBL2 gene polymorphisms (codon 54 A/B, -550 H/L or -221 X/Y) were not associated with the risk of asthma (codon 54 A/B: BB+AB vs AA: OR, 1.02; 95% CI, 0.85-1.23; -550 H/L: LL+HL vs HH: OR, 0.81; 95% CI, 0.63-1.03; -221 X/Y: XX+YX vs YY: OR, 0.85; 95% CI, 0.69-1.04). Subgroup analysis by ethnicity implied that the MBL2 codon 54 A/B polymorphism was not significantly associated with the risk of asthma in Asians (BB+AB vs AA: OR, 0.95; 95% CI, 0.70-1.29) or whites (BB+AB vs AA: OR, 1.07; 95% CI, 0.84-1.35). CONCLUSION: The results indicated that MBL2 gene polymorphisms (codon 54 A/B, -550 H/L or -221 X/Y) may be not associated with the risk of asthma.


Subject(s)
Asthma/genetics , Mannose-Binding Lectin/genetics , Asian People/genetics , Genetic Predisposition to Disease , Humans , Odds Ratio , Polymorphism, Genetic , Risk Factors
19.
Am J Physiol Lung Cell Mol Physiol ; 310(4): L299-310, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26702149

ABSTRACT

Hypoxic pulmonary hypertension (HPH) is characterized by active vasoconstriction and profound vascular remodeling. KLF5, a zinc-finger transcription factor, is involved in the excessive proliferation and apoptotic resistance phenotype associated with monocrotaline-induced pulmonary hypertension. However, the molecular mechanisms of KLF5-mediated pathogenesis of HPH are largely undefined. Adult male Sprague-Dawley rats were exposed to normoxia or hypoxia (10% O2) for 4 wk. Hypoxic rats developed pulmonary arterial remodeling and right ventricular hypertrophy with significantly increased right ventricular systolic pressure. The levels of KLF5 and hypoxia-inducible factor-1α (HIF-1α) were upregulated in distal pulmonary arterial smooth muscle from hypoxic rats. The knockdown of KLF5 via short-hairpin RNA attenuated chronic hypoxia-induced hemodynamic and histological changes in rats. The silencing of either KLF5 or HIF-1α prevented hypoxia-induced (5%) proliferation and migration and promoted apoptosis in human pulmonary artery smooth muscle cells. KLF5 was immunoprecipitated with HIF-1α under hypoxia and acted as an upstream regulator of HIF-1α. The cell cycle regulators cyclin B1 and cyclin D1 and apoptosis-related proteins including bax, bcl-2, survivin, caspase-3, and caspase-9, were involved in the regulation of KLF5/HIF-1α-mediated cell survival. This study demonstrated that KLF5 plays a crucial role in hypoxia-induced vascular remodeling in an HIF-1α-dependent manner and provided a better understanding of the pathogenesis of HPH.


Subject(s)
Hypertension, Pulmonary/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Kruppel-Like Transcription Factors/metabolism , Vascular Remodeling/physiology , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Male , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Rats, Sprague-Dawley , Transcriptional Activation
20.
Exp Cell Res ; 339(1): 122-34, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26476374

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by excessive proliferation, resistance to apoptosis, and increased migration of pulmonary artery smooth muscle cells (PASMCs). We hypothesized that quercetin exerts protective effects against this disease; thus, a chronic hypoxia model of PAH was generated using male Sprague-Dawley rats, which were treated with quercetin. In this model, quercetin prevented the development of PAH, right ventricular hypertrophy, and vascular remodeling after exposure to hypoxia. Quercetin inhibited PASMC proliferation and increased the apoptosis of PASMCs in vivo. In vitro, quercetin significantly inhibited hypoxia-induced PASMC proliferation, arrested cells in G1/G0 and inhibited cell migration in a dose-dependent manner. Moreover, our results showed that quercetin increased cyclin D1 protein levels and decreased the protein expression of cyclin B1 and Cdc2. Additionally, quercetin altered the Bax/Bcl-2 ratio and reduced MMP2, MMP9, CXCR4, integrin ß1, and integrin α5 expression. Using genome-wide microarray analysis, we found that factors regulating proliferation, apoptosis, cell cycle, and migration were related to the tyrosine receptor kinase A (TrkA) pathway. In addition, activation of the TrkA/AKT signaling cascade during hypoxia was inhibited by quercetin in a dose-dependent manner. Moreover, quercetin alone inhibited the TrkA/AKT signaling pathway, resulting in decreased PASMC migration, cell cycle arrest and the induction of apoptosis. Our data suggest that quercetin is a potential candidate for the treatment of hypoxia-induced PAH.


Subject(s)
Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/complications , Muscle, Smooth, Vascular/drug effects , Pulmonary Artery/drug effects , Quercetin/pharmacology , Receptor, trkA/metabolism , Animals , Antioxidants/pharmacology , Apoptosis , Blotting, Western , Cell Cycle , Cell Movement , Cell Proliferation , Cells, Cultured , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/metabolism , Hypoxia/physiopathology , Immunoenzyme Techniques , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, trkA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...