Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Transl Oncol ; 45: 101964, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38657441

BACKGROUND: Although there is evidence of the association between RA and NSCLC, little is known about their interaction mechanisms. The aim of this study is to identify potential hub genes and biological mechanism in RA and NSCLC via integrated bioinformatics analysis. METHODS: The gene expression datasets of RA and NSCLC were downloaded to discover and validate hub genes. After identifying DEGs, we performed enrichment analysis, PPI network construction and module analysis, selection and validation of hub genes. Moreover, we selected the hub gene PTPRC for expression and prognosis analysis, immune analysis, mutation and methylation analysis in NSCLC. Finally, we performed real-time PCR, colony formation assay, wound healing assay, transwell invasion assay, sphere formation assay and western blotting to validate the role of PTPRC in A549 cells. RESULTS: We obtained 320 DEGs for subsequent analysis. Enrichment results showed that the DEGs were mainly involved in Th1, Th2 and Th17 cell differentiation. In addition, four hub genes, BIRC5, PTPRC, PLEK, and FYN, were identified after selection and validation. These hub genes were subsequently shown to be closely associated with immune cells and related pathways. In NSCLC, PTPRC was downregulated, positively correlated with immune infiltration and immune cells. Experiments showed that PTPRC could promote the proliferation, migration and invasion, and the ability to form spheroids of A549 cells. In addition, PTPRC could regulate the increased expression of CD45, ß-catenin, c-Myc and LEF1 proteins. CONCLUSIONS: This study explored the hub genes and related mechanisms of RA and NSCLC, demonstrated the central role of the inflammatory response and the adaptive immune system, and identified PTPRC as an immune-related biomarker and potential therapeutic target for RA and NSCLC patients. In addition, PTPRC can significantly promote the proliferation, migration and invasion of A549 cells, and its mechanism may be to promote the EMT process by regulating the Wnt signaling pathway and promote cell stemness, which in turn has a promoting effect on A549 cells.

2.
Cancer Med ; 13(3): e7014, 2024 Feb.
Article En | MEDLINE | ID: mdl-38426625

BACKGROUND: Numerous of models have been developed to predict the bone metastasis (BM) risk; however, due to the variety of cancer types, it is difficult for clinicians to use these models efficiently. We aimed to perform the pan-cancer analysis to create the cancer classification system for BM, and construct the nomogram for predicting the BM risk. METHODS: Cancer patients diagnosed between 2010 and 2018 in the Surveillance, Epidemiology, and End Results (SEER) database were included. Unsupervised hierarchical clustering analysis was performed to create the BM prevalence-based cancer classification system (BM-CCS). Multivariable logistic regression was applied to investigate the possible associated factors for BM and construct a nomogram for BM risk prediction. The patients diagnosed between 2017 and 2018 were selected for validating the performance of the BM-CCS and the nomogram, respectively. RESULTS: A total of 50 cancer types with 2,438,680 patients were included in the construction model. Unsupervised hierarchical clustering analysis classified the 50 cancer types into three main phenotypes, namely, categories A, B, and C. The pooled BM prevalence in category A (17.7%; 95% CI: 17.5%-17.8%) was significantly higher than that in category B (5.0%; 95% CI: 4.5%-5.6%), and category C (1.2%; 95% CI: 1.1%-1.4%) (p < 0.001). Advanced age, male gender, race, poorly differentiated grade, higher T, N stage, and brain, lung, liver metastasis were significantly associated with BM risk, but the results were not consistent across all cancers. Based on these factors and BM-CCS, we constructed a nomogram for predicting the BM risk. The nomogram showed good calibration and discrimination ability (AUC in validation cohort = 88%,95% CI: 87.4%-88.5%; AUC in construction cohort = 86.9%,95% CI: 86.8%-87.1%). The decision curve analysis also demonstrated the clinical usefulness. CONCLUSION: The classification system and prediction nomogram may guide the cancer management and individualized BM screening, thus allocating the medical resources to cancer patients. Moreover, it may also have important implications for studying the etiology of BM.


Bone Neoplasms , Nomograms , Humans , Male , Bone Neoplasms/epidemiology , Bone Neoplasms/secondary , Logistic Models , SEER Program
3.
Eur J Pharmacol ; 968: 176354, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38316248

Sepsis is a life-threatening condition involving dysfunctional organ responses stemming from dysregulated host immune reactions to various infections. The lungs are most prone to failure during sepsis, resulting in acute lung injury (ALI). ALI is associated with oxidative stress and inflammation, and current therapeutic strategies are limited. To develop a more specific treatment, this study aimed to synthesise Prussian blue nanozyme (PBzyme), which can reduce oxidative stress and inflammation, to alleviate ALI. PBzyme with good biosafety was synthesised using a modified hydrothermal method. PBzyme was revealed to be an activator of haem oxygenase-1 (HO-1), improving survival rate and ameliorating lung injury in mice. Zinc protoporphyrin, an inhibitor of HO-1, inhibited the prophylactic therapeutic efficacy of PBzyme on ALI, and affected the nuclear factor-κB signaling pathway and activity of HO-1. This study demonstrates that PBzyme can alleviate oxidative stress and inflammation through HO-1 and has a prophylactic therapeutic effect on ALI. This provides a new strategy and direction for the clinical treatment of sepsis-induced ALI.


Acute Lung Injury , Ferrocyanides , Sepsis , Mice , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/etiology , Acute Lung Injury/prevention & control , Heme Oxygenase-1/metabolism , Lung , Inflammation/complications , Inflammation/drug therapy , Sepsis/complications , Sepsis/drug therapy , Lipopolysaccharides/pharmacology , NF-E2-Related Factor 2/metabolism
4.
Article En | MEDLINE | ID: mdl-38294742

Objective: To explore intestinal flora differences in species diversity, community structure, and abundance of breast cancer and non-breast cancer populations with anxiety and depression and the corresponding group without anxiety and depression by 16S rRNA high-throughput sequencing technology. Method: Breast cancer and non-breast cancer participants were recruited based on the inclusion and exclusion criteria as the research subjects. The study employed the anxiety self-assessment scale and the depression self-rating scale in the questionnaire survey to collect data. Results: The scores of anxiety and depression of the four groups are as follows: In the breast cancer with anxiety and/or depression (BCAD) group, the anxiety score is 58.80 ± 5.27 and the depression score is 59.60 ± 4.94. In the breast cancer without anxiety and/or depression (BCWAD) group, the anxiety score is 36.53 ± 4.52 and the depression score is 38.20 ± 3.78. In the non-breast cancer group with anxiety and/or depression (HAD) group, the anxiety score is 57.87 ± 4.53 and the depression score is 59.13 ± 5.24. In the non-breast cancer group without anxiety and depression (HWAD) group, the anxiety score is 35.13 ± 5.28 and the depression score is 32.33 ± 4.37. Conclusion: The intestinal flora of the breast cancer patients is significantly different from those of non-breast cancer patients, suggesting that there is an internal relationship between the changes in the intestinal flora and the occurrence and development of breast cancer. People with anxiety and depression without breast cancer show changes in their intestinal flora, suggesting that the changes of the intestinal flora can indeed trigger anxiety and depression. For the breast cancer patients with anxiety and depression, the intestinal flora shows a decrease in diversity and abundance, suggesting that the intestinal flora of the breast cancer patients with anxiety and depression undergo further changes. Thus the intestinal flora can become a new tool for monitoring, preventing, and treating the breast cancer and negative emotions.

5.
Clin Exp Hypertens ; 46(1): 2297642, 2024 Dec 31.
Article En | MEDLINE | ID: mdl-38147409

Pulmonary arterial hypertension (PAH) is characterized by progressive vascular remodeling caused by the excessive proliferation and survival of pulmonary artery smooth muscle cells (PASMCs). Dual-specificity tyrosine regulated kinase 1A (DYRK1A) is a pleiotropic kinase involved in the regulation of multiple biological functions, including cell proliferation and survival. However, the role and underlying mechanisms of DYRK1A in PAH pathogenesis remain unclear. We found that DYRK1A was upregulated in PASMCs in response to hypoxia, both in vivo and in vitro. Inhibition of DYRK1A by harmine significantly attenuated hypoxia-induced pulmonary hypertension and pulmonary artery remodeling. Mechanistically, we found that DYRK1A promoted pulmonary arterial remodeling by enhancing the proliferation and survival of PASMCs through activating the STAT3/Pim-1/NFAT pathway, because STAT3 gain-of-function via adeno-associated virus serotype 2 (AAV2) carrying the constitutively active form of STAT3 (STAT3C) nearly abolished the protective effect of harmine on PAH. Collectively, our results reveal a significant role for DYRK1A in pulmonary arterial remodeling and suggest it as a drug target with translational potential for the treatment of PAH.


Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/metabolism , Vascular Remodeling , Harmine/adverse effects , Harmine/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Pulmonary Artery , Hypoxia , Myocytes, Smooth Muscle/metabolism , Cell Proliferation , Cells, Cultured , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
6.
Neuropsychiatr Dis Treat ; 19: 2573-2583, 2023.
Article En | MEDLINE | ID: mdl-38046832

Purpose: We aimed to explore whether anxiety is a risk factor for acute mountain sickness [AMS] in a young Chinese male population. Patients and Methods: A total of 143 young Chinese men with a median age of 23 years (IQR, 21-25) were employed in the present study, and they were divided into the AMS+ and AMS- groups according to the Lake Louise AMS score [AMS-S] after exposure at 3800 m for two days. Participants' pulse oximeter saturation [SpO2] and heart rate [HR] were measured. AMS was evaluated using the AMS-S. The anxiety and sleep quality of the subjects were assessed using the Zung Self-Rating Anxiety Scale [SAS] and the Athens Insomnia Scale [AIS], respectively. Outcomes were analysed using Spearman's partial correlation and logistic regression analysis. Results: After two days of exposure at 3800 m, the overall prevalence of AMS was 54% in the whole group. The HR was significantly higher in the AMS+ group than in the AMS- group, as well as the SAS score and AIS score. A converse pattern was observed for SpO2. A significant difference was observed for the change in SAS and AIS score between the AMS+ and AMS- groups. Correlation analysis showed that AMS-S was positively correlated with SAS score, AIS score, HR, ΔSAS score, ΔAIS score, and ΔHR but negatively correlated with SpO2. AIS score was positively correlated with SAS score. After logistic regression analysis was adjusted for HR, SpO2, ΔAIS and ΔHR, SAS score (OR=1.446, 95% CI 1.200-1.744, p<0.001), AIS score (OR=1.216, 95% CI 1.033-1.432) and ΔSAS score (OR=1.158, 95% CI 1.012-1.327) were identified as independent risk factors for AMS. Conclusion: The present study suggests that anxiety is a risk factor for AMS among young Chinese men, and poor sleep quality may partially mediate the association.

7.
Clin Exp Hypertens ; 45(1): 2278205, 2023 Dec 31.
Article En | MEDLINE | ID: mdl-37970663

Pulmonary arterial hypertension (PAH) is a disease with a high mortality and few treatment options to prevent the development of pulmonary vessel remodeling, pulmonary vascular resistance, and right ventricular failure. Canagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, is originally used in diabetes patients which could assist the glucose excretion and decrease blood glucose. Recently, a few studies have reported the protective effect of SGLT2 inhibitor on monocrotaline-induced PAH. However, the effects of canagliflozin on hypobaric hypoxia-induced PAH as well as its mechanism still unclear. In this study, we used hypobaric hypoxia-induced PAH mice model to demonstrate if canagliflozin could alleviate PAH and prevent pulmonary vessel remodeling. We found that daily canagliflozin administration significantly improved survival in mice with hypobaric hypoxia-induced PAH compared to vehicle control. Canagliflozin treatment significantly reduced right ventricular systolic pressure and increased pulmonary acceleration time determined by hemodynamic assessments. Canagliflozin significantly reduced medial wall thickening and decreased muscularization of pulmonary arterioles compared to vehicle treated mice. In addition, canagliflozin inhibited the proliferation and migration of pulmonary arterial smooth muscle cells through suppressing glycolysis and reactivating AMP-activated protein kinase signaling pathway under hypoxia condition. In summary, our findings suggest that canagliflozin is sufficient to inhibit pulmonary arterial remodeling which is a potential therapeutic strategy for PAH treatment.


Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Mice , Animals , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Canagliflozin/adverse effects , Pulmonary Artery , Hypoxia/complications , Hypoxia/metabolism , Myocytes, Smooth Muscle/metabolism , Cell Proliferation , Glucose/pharmacology , Vascular Remodeling , Monocrotaline/pharmacology
8.
Phytother Res ; 37(12): 5854-5870, 2023 Dec.
Article En | MEDLINE | ID: mdl-37655750

Doxorubicin (DOX) has aroused contradiction between its potent anti-tumor capacity and severe cardiotoxicity. Galangin (Gal) possesses antioxidant, anti-inflammatory, and antiapoptotic activities. We aimed to explore the role and underlying mechanisms of Gal on DOX-induced cardiotoxicity. Mice were intraperitoneally injected with DOX (3 mg/kg, every 2 days for 2 weeks) to generate cardiotoxicity model and Gal (15 mg/kg, 2 weeks) was co-administered via gavage daily. Nuclear factor erythroid 2-related factor 2 (Nrf2) specific inhibitor, ML385, was employed to explore the underlying mechanisms. Compared to DOX-insulted mice, Gal effectively improved cardiac dysfunction and ameliorated myocardial damage. DOX-induced increase of reactive oxygen species, malondialdehyde, and NADPH oxidase activity and downregulation of superoxide dismutase (SOD) activity were blunted by Gal. Gal also markedly blocked increase of IL-1ß, IL-6, and TNF-α in DOX-insulted heart. Mechanistically, Gal reversed DOX-induced downregulation of Nrf2, HO-1, and promoted nuclear translocation of Nrf2. ML385 markedly blunted the cardioprotective effects of Gal, as well as inhibitive effects on oxidative stress and inflammation. Gal ameliorates DOX-induced cardiotoxicity by suppressing oxidative stress and inflammation via activating Nrf2/HO-1 signaling pathway. Gal may serve as a promising cardioprotective agent for DOX-induced cardiotoxicity.


Cardiotoxicity , Heme Oxygenase-1 , Mice , Animals , Cardiotoxicity/drug therapy , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Apoptosis , Oxidative Stress , Doxorubicin/adverse effects , Signal Transduction , Inflammation/metabolism , Myocytes, Cardiac
9.
Phytomedicine ; 117: 154922, 2023 Aug.
Article En | MEDLINE | ID: mdl-37321078

BACKGROUND: Doxorubicin (DOX) is a potent anticancer chemotherapeutic agent whose clinical application is substantially constrained by its cardiotoxicity. The pathophysiology of DOX-induced cardiotoxicity manifests as cardiomyocyte pyroptosis and inflammation. Amentoflavone (AMF) is a naturally occurring biflavone possessing anti-pyroptotic and anti-inflammatory properties. However, the mechanism through which AMF alleviates DOX-induced cardiotoxicity remains undetermined. PURPOSE: This study aimed at investigating the role of AMF in alleviating DOX-induced cardiotoxicity. STUDY DESIGN AND METHODS: To assess the in vivo effect of AMF, DOX was intraperitoneally administered into a mouse model to induce cardiotoxicity. To elucidate the underlying mechanisms, the activities of STING/NLRP3 were quantified using the NLRP3 agonist nigericin and the STING agonist amidobenzimidazole (ABZI). Primary cardiomyocytes isolated from neonatal Sprague-Dawley rats were treated with saline (vehicle) or DOX with or without AMF and/or ABZI. The echocardiogram, haemodynamics, cardiac injury markers, heart/body weight ratio, and pathological alterations were monitored; the STING/NLRP3 pathway-associated proteins were detected by western blot and cardiomyocyte pyroptosis was analysed by immunofluorescence staining of cleaved N-terminal GSDMD and scanning electron microscopy. Furthermore, we evaluated the potential of AMF in compromising the anticancer effects of DOX in human breast cancer cell lines. RESULTS: AMF substantially alleviated cardiac dysfunction and reduced heart/body weight ratio and myocardial damage in mice models of DOX-induced cardiotoxicity. AMF effectively suppressed DOX-mediated upregulation of IL-1ß, IL-18, TNF-α, and pyroptosis-related proteins, including NLRP3, cleaved caspase-1, and cleaved N-terminal GSDMD. The levels of apoptosis-related proteins, namely Bax, cleaved caspase-3, and BCL-2 were not affected. In addition, AMF inhibited STING phosphorylation in DOX-affected hearts. Intriguingly, the administration of nigericin or ABZI dampened the cardioprotective effects of AMF. The in vitro anti-pyroptotic effect of AMF was demonstrated in attenuating the DOX-induced reduction in cardiomyocyte cell viability, upregulation of cleaved N-terminal GSDMD, and pyroptotic morphology alteration at the microstructural level. AMF exhibited a synergistic effect with DOX to reduce the viability of human breast cancer cells. CONCLUSION: AMF alleviates DOX-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation via inhibition of the STING/NLRP3 signalling pathway, thereby validating its efficacy as a cardioprotective agent.


Breast Neoplasms , Myocytes, Cardiac , Rats , Mice , Animals , Humans , Female , Pyroptosis , Cardiotoxicity/drug therapy , Cardiotoxicity/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nigericin/adverse effects , Nigericin/metabolism , Rats, Sprague-Dawley , Doxorubicin/pharmacology , Apoptosis Regulatory Proteins/metabolism , Inflammation/metabolism , Breast Neoplasms/pathology , Body Weight
10.
Chinese Journal of School Health ; (12): 1008-1011, 2023.
Article Zh | WPRIM | ID: wpr-984485

Objective@#To investigate oral health related life quality and associated factors of senior primary school students in Bengbu City, so as to provide scientific basis for targeted oral health education for children.@*Methods@#A stratified cluster random sampling method was adopted to select 1 760 senior students (grade 5 to 6) from 12 primary schools in 4 districts of Bengbu City from September to November 2022. A cross sectional survey was conducted on the oral health of students through questionnaires related to oral health. The effect of oral problems on quality of life was assessed by the Child Daily Life Oral Influence Scale (Child-OIDP). Chi square test, non parametric test and binary Logistic regression model were used to analyze the influencing factors of oral health related quality of life in senior primary school students.@*Results@#The prevalence of oral problems affecting the quality of daily life was 70.00%, and the severe impact rate was 15.06%. Oral feeding was the most affected (57.95%). The Child-OIDP score was (7.49±8.57). Binary Logistic regression analysis showed that mother s education level, self rated teeth and oral conditions, bruised tooth, toothache in the past year, and gingival bleeding in the past 2 weeks were significantly associated with the incidence of Child-OIDP ( OR=1.86-5.00, P <0.05).@*Conclusion@#It is common that oral problems affect the quality of daily life of senior primary school students in Bengbu. Families and schools should strengthen oral health knowledge education and behavior guidance, so as to reduce the impact of oral problems on daily life among senior primary school students.

11.
Mitochondrion ; 65: 150-160, 2022 07.
Article En | MEDLINE | ID: mdl-35779798

Microvascular protection is the main mechanism of metformin against diabetic complications. Cardiac microvascular endothelial cells (CMECs) are the basic component of cardiac microvessels, and they suffer from oxidative stress and mitochondrial dysfunction under type 2 diabetes mellitus (T2DM). Translocase of the outer mitochondrial membrane 70 (Tom70) improves mitochondrial dysfunction, but its role in the hearts of T2DM patients remains unclear. The purpose of this study was to demonstrate the protective effect of metformin on diabetic cardiac microvascular injury and to identify the role of Tom70 in this effect. T2DM mice were established by multiple intraperitoneal injections of low-dose streptozotocin and 12-week high-fat feeding. CMECs were isolated and cultured with normal glucose (NG), high glucose (HG), and HG plus high fat (HG-HF) media. The results indicated that long-term metformin treatment partly reversed cardiovascular complication and mitigated cardiac microvascular injury in T2DM. In addition, exposure to HG-HF led to CMEC damage, aggravated oxidative stress, aggravated mitochondrial dysfunction, and reduced mitochondrial Tom70 expression, whereas upregulation of Tom70 significantly ameliorated these injuries. Furthermore, metformin treatment promoted Tom70 expression and effectively reversed CMEC injury induced by HG-HF. However, all of these effects were interrupted after Tom70 was knocked down. In conclusion, T2DM damages microvascular integrity by activating a cycle of decreased Tom70 expression, mitochondrial dysfunction, and reactive oxygen species (ROS) overload in CMECs. However, metformin suppresses oxidative stress, relieves mitochondrial dysfunction, and promotes the expression of Tom70, ultimately ameliorating diabetic microvascular injury and heart complications.


Diabetes Mellitus, Type 2 , Metformin , Animals , Mice , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Metformin/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Oxidative Stress
12.
J Cardiovasc Pharmacol ; 80(3): 430-441, 2022 09 01.
Article En | MEDLINE | ID: mdl-35881904

ABSTRACT: Transient receptor potential vanilloid type 1 (TRPV1) is a nonselective cation channel that mediates the relationship between mitochondrial function and pathological myocardial hypertrophy. However, its underlying mechanisms remain unclear. This study aimed to investigate whether TRPV1 activation improves the morphology and function of intracellular mitochondria to protect cardiomyocytes after pressure overload-induced myocardial hypertrophy. The myocardial hypertrophy model was established by performing transverse aortic constriction surgery in C57BL/6 J male mice. The data revealed that TRPV1 activation significantly reduced myocardial hypertrophy, promoted ejection fraction% and fractional shortening%, and decreased the left ventricular internal diameter in end-diastole and left ventricular internal diameter in end-systole after transverse aortic constriction. Moreover, in vitro experiments revealed that TRPV1 reduces cardiomyocyte area and improves mitochondrial function by promoting mitochondria-associated endoplasmic reticulum membranes (MAMs) formation in a phenylephrine-treated cardiomyocyte hypertrophy model. TRPV1 up-regulates the phosphorylation levels of AMP-activated protein kinase and expression of mitofusin2 (MFN2). TRPV1 function is blocked by single-stranded RNA interfering with silent interfering MFN2. Activation of TRPV1 reduced mitochondrial reactive oxygen species caused by phenylephrine, whereas disruption of MAMs by siMFN2 abolished TRPV1-mediated mitochondrial protection. Our findings suggest that TRPV1 effectively protects against pressure overload-induced cardiac hypertrophy by promoting MAM formation and conserved mitochondrial function via the AMP-activated protein kinase/MFN2 pathway in cardiomyocytes.


AMP-Activated Protein Kinases , Cardiomegaly , TRPV Cation Channels/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cardiomegaly/pathology , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Phenylephrine
13.
J Nurs Manag ; 29(2): 240-249, 2021 Mar.
Article En | MEDLINE | ID: mdl-32890453

AIMS: To evaluate psychological well-being and factors associated with post-traumatic stress disorder (PTSD) among front-line nurses during the coronavirus disease-2019 (COVID-19) pandemic. BACKGROUND: Coronavirus disease-2019 is a pandemic that has posed a public health emergency of international concern. Psychological well-being of front-line nurses is a big concern during the COVID-19 pandemic. METHODS: With a predictive study design, a same survey was sent separately at two time points (i.e. before and after nurses worked at COVID-19 units) between January and March 2020 among 356 front-line nurses in First Affiliated Hospital of Bengbu Medical College, Anhui, China. RESULTS: Of a total 356 front-line nurses, stress level and the prevalence of PTSD were significantly increased after they worked at COVID-19 units. Nurses who had work experience less than 2 years were significantly associated with a high risk of developing PTSD. Nurses who worked in COVID-19 inpatients wards had significantly higher odds of being PTSD (odds ratio [OR] = 21.9, 95% confidence interval [CI]: 5.08; 94.5) than those who worked in other COVID-19-related units. Resilience was negatively associated with PTSD (OR = 0.96, 95% CI: 0.93; 0.99). CONCLUSION: Nurses had significantly increased risk to develop PTSD during COVID-19 pandemic. IMPLICATIONS FOR NURSING MANAGEMENT: Clinical and policy strategies to support front-line nurses' psychological well-being, particularly young nurses, in response to COVID-19 crisis are urgently needed.


COVID-19/nursing , Nursing Staff, Hospital/psychology , Occupational Stress/psychology , Stress Disorders, Post-Traumatic/epidemiology , Adult , COVID-19/epidemiology , China/epidemiology , Female , Humans , Male , Nursing Staff, Hospital/statistics & numerical data , Prevalence , Risk Factors , Surveys and Questionnaires
14.
Front Psychiatry ; 11: 580827, 2020.
Article En | MEDLINE | ID: mdl-33192723

Background: As a public health emergency of international concern, the COVID-19 outbreak has had a tremendous impact on patients' psychological health. However, studies on psychological interventions in patients with COVID-19 are relatively rare. Objectives: This study examined the effectiveness of Cognitive Behavioral Therapy (CBT) in relieving patients' psychological distress during the COVID-19 epidemic. Methods: Ninety-three eligible participants selected by cluster sampling were randomized to an intervention group (N = 47) and a control group (N = 46). Participants in the control group received routine treatment according to the Chinese Management Guidelines for COVID-19, while participants in the intervention group received routine treatment with additional CBT. The Chinese Version of Depression Anxiety and Stress Scale-21 (DASS-21) was used to evaluate depression, anxiety, and stress for all participants at baseline and post-intervention. Two-sided t-test, and proportion tests were used to examine the differences between the intervention and control group for each DASS-21 indicator. Univariate linear regression was used to examine the association between chronic disease status and change in each DASS-21 indicator after intervention. Two-way scatter plots were generated to show the association of the length of hospital stay and the changes of each DASS-21 indicator by intervention and control groups. Results: Significant decreases in means were found for scales of depression, anxiety, stress and total DASS-21 in both intervention (p < 0.001) and control group (p = 0.001), with participants in the intervention group having a bigger reduction in means. After the intervention, more participants in the intervention group had no depression or anxiety symptoms than in the control group, but no statistical differences were found (p > 0.05). Compared with participants with chronic disease, participants with no chronic disease had a significantly larger reduction of total DASS-21 scale (coefficient = -4.74, 95% CI: -9.31; -0.17).The length of hospital stay was significantly associated with a greater increase in anxiety scale in the intervention group (p = 0.005), whilst no significant association was found in the control group (p = 0.29). Conclusions: The patients with COVID-19 experienced high levels of anxiety, depression and stress. Our study result highlights the effectiveness of CBT in improving the psychological health among patients with COVID-19, also suggests that CBT should be focused on patients with chronic disease and those who have longer hospital stays. These results have important implications in clinical practice in improving psychological health in the context of COVID-19 pandemic. Trial Registration: ISRCTN68675756. Available at: http://www.isrctn.com/ISRCTN68675756.

15.
J Cardiovasc Pharmacol ; 75(6): 556-563, 2020 06.
Article En | MEDLINE | ID: mdl-32195756

Ischemia and anoxia-induced mitochondrial impairment may be a key factor leading to heart injury during myocardial infarction (MI). Calpain 1 and 2 are involved in the MI-induced mitochondria injury. G protein-coupled receptor 35 (GPR35) could be triggered by hypoxia. Whether or not GPR35 regulates calpain 1/2 in the pathogenesis of MI is still unclear. In this study, we determined that MI increases GPR35 expression in myocardial tissue. Suppression of GPR35 protects heart from MI injury in mice through reduction of reactive oxygen species activity and mitochondria-dependent apoptosis. Further studies show that GPR35 regulates calpain 1/2. Suppression of GPR35 reduces the expression and activity of calpain 1/2, and alleviates calpain 1/2-associated mitochondrial injury to preserve cardiac function. Based on these data, we conclude that a functional inhibition of GPR35 downregulates calpain 1/2 and contributes to maintenance of cardiac function under pathologic conditions with mitochondrial disorder. In conclusion, our study showed that the identified regulation by GPR35 of calpain 1/2 has important implications for the pathogenesis of MI. Targeting the action of GPR35 and calpain 1/2 in mitochondria presents a potential therapeutic intervention for MI.


Calpain/metabolism , Mitochondria, Heart/enzymology , Myocardial Infarction/therapy , Myocytes, Cardiac/enzymology , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Receptors, G-Protein-Coupled/metabolism , Animals , Apoptosis , Calpain/genetics , Cells, Cultured , Disease Models, Animal , Male , Mice, Inbred C57BL , Mitochondria, Heart/pathology , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction
16.
J Hypertens ; 37(11): 2256-2268, 2019 11.
Article En | MEDLINE | ID: mdl-31136458

OBJECTIVES: Vascular smooth muscle cell (VSMC) proliferation is a crucial cause of vascular neointima hyperplasia and restenosis, thus limiting the long-term efficacy of percutaneous vascular intervention. We explored the role of wild-type p53-induced phosphatase 1 (Wip1), a potent regulator of tumorigenesis and atherosclerosis, in VSMC proliferation and neointima hyperplasia. METHODS AND RESULTS: Animal model of vascular restenosis was established in wild type C57BL/6J and VSMC-specific Tuberous Sclerosis 1 (TSC1)-knockdown mice by wire injury. We observed increased protein levels of Wip1, phospho (p)-S6 Ribosomal Protein (S6), p-4EBP1 but decreased p-adenosine 5'-monophosphate-activated protein kinase (AMPK)α both in carotid artery at day 28 after injury and in VSMCs after 48 h of platelet derived growth factor-BB (PDGF-BB) treatment. By using hematoxylin-eosin staining, Ki-67 immunohistochemical staining, cell counting kit-8 assay and Ki-67 immunofluorescence staining, we found Wip1 antagonist GSK2830371 (GSK) or mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin both obviously reversed the neointima formation and VSMC proliferation induced by wire injury and PDGF-BB, respectively. GSK also reversed the increase in mRNA level of Collagen I after wire injury. However, GSK had no obvious effects on VSMC migration induced by PDGF-BB. Simultaneously, TSC1 knockdown as well as AMPK inhibition by Compound C abolished the vascular protective and anti-proliferative effects of Wip1 inhibition. Additionally, suppression of AMPK also reversed the declined mTORC1 activity by GSK. CONCLUSION: Wip1 promotes VSMC proliferation and neointima hyperplasia after wire injury via affecting AMPK/mTORC1 pathway.


Aminopyridines/therapeutic use , Dipeptides/therapeutic use , Myocytes, Smooth Muscle/drug effects , Neointima/prevention & control , Protein Phosphatase 2C/metabolism , Vascular System Injuries/enzymology , AMP-Activated Protein Kinases/metabolism , Aminopyridines/pharmacology , Animals , Becaplermin , Carotid Artery, Common/pathology , Cell Proliferation/drug effects , Dipeptides/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Hyperplasia , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular , Neointima/etiology , Protein Phosphatase 2C/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Vascular System Injuries/complications
17.
Hypertension ; 72(1): 116-127, 2018 07.
Article En | MEDLINE | ID: mdl-29735636

Cardiac microvascular injury often occurs in patients with type 2 diabetes mellitus (T2DM) who develop hyperglycemia and hyperlipidemia. However, besides reported contradictory roles in cardiac diseases, the function of TRPV1 (transient receptor potential vanilloid 1) in cardiac microvessels is not well defined. This study was performed to determine the detailed role of TRPV1 in cardiac microvascular endothelial cells (CMECs) in T2DM. T2DM mice were established by multiple injections of low-dose streptozotocin and high-fat feeding. CMECs were cultured separately in mediums of normal glucose, high glucose (HG), high fatty acid (HF), and HG plus HF (HG-HF). HG-HF inhibited TRPV1 expression in CMECs, reducing cellular Ca2+ content ([Ca2+]i). T2DM impaired cardiac function, disturbed glucose uptake, and damaged microvascular barrier, which were further aggravated by TRPV1-/- Exposure to HG-HF, particularly in TRPV1-/- CMECs, led to a higher level of apoptosis and a lower level of nitric oxide production in viable CMECs. HG-HF markedly enhanced generation of reactive oxygen species and nitrotyrosine, especially in the absence of TRPV1. H2O2 administration reduced TRPV1 expression in CMECs. HG-HF significantly depressed expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator-1α) and OPA1 (optic atrophy 1) by reducing [Ca2+]i, whereas OPA1 supplementation partly reversed those detrimental effects induced by TRPV1-/- Furthermore, capsaicin treatment not only attenuated CMECs injury induced by HG-HF but also mitigated cardiac microvascular injury induced by T2DM. Collectively, T2DM leads to cardiac microvascular injury by exacerbating the vicious circle of TRPV1 blockage and reactive oxygen species overload. Long-term capsaicin can protect cardiac microvessels against T2DM via suppressing oxidative/nitrative stress mediated by TRPV1/Ca2+/PGC-1α/OPA1 pathway in CMECs.


Coronary Vessels/pathology , Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies/pathology , Endothelium, Vascular/pathology , Microvessels/pathology , Oxidative Stress , TRPV Cation Channels/metabolism , Animals , Apoptosis , Coronary Vessels/metabolism , Diabetic Cardiomyopathies/metabolism , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism
18.
J Int Med Res ; 45(6): 1629-1635, 2017 Dec.
Article En | MEDLINE | ID: mdl-27565745

The pathogenesis of myocardial ischaemia/reperfusion injury is multifactorial. Understanding the mechanisms of myocardial ischaemia/reperfusion will benefit patients with ischaemic heart disease. Growth differentiation factor 11 (GDF11), a member of the secreted transforming growth factor-ß superfamily, has been found to reverse age-related hypertrophy, revealing the important role of GDF11 in cardiovascular disease. However, the functions of GDF11 in myocardial ischaemia/reperfusion have not been elucidated yet. A number of signalling molecules are known to occur downstream of GDF11, including mothers against decapentaplegic homolog 3 (SMAD3) and forkhead box O3a (FOXO3a). A hypothesis is presented that GDF11 has protective effects in acute myocardial ischaemia/reperfusion injury through suppression of oxidative stress, prevention of calcium ion overload and promotion of the elimination of abnormal mitochondria via both canonical (SMAD3) and non-canonical (FOXO3a) pathways. Since circulating GDF11 may mainly derive from the spleen, the lack of a spleen may make the myocardium susceptible to damaging insults. Administration of GDF11 may be an efficacious therapy to protect against cardiovascular diseases in splenectomized patients.


Cardiotonic Agents/therapeutic use , Growth Differentiation Factors/therapeutic use , Models, Biological , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Animals , Humans , Spleen/metabolism
19.
J Pineal Res ; 62(1)2017 Jan.
Article En | MEDLINE | ID: mdl-27706848

Mitochondrial dysfunction leads to reactive oxygen species (ROS) overload, exacerbating injury in myocardial infarction (MI). As a receptor for translocases in the outer mitochondrial membrane (Tom) complex, Tom70 has an unknown function in MI, including melatonin-induced protection against MI injury. We delivered specific small interfering RNAs against Tom70 or lentivirus vectors carrying Tom70a sequences into the left ventricles of mice or to cultured neonatal murine ventricular myocytes (NMVMs). At 48 h post-transfection, the left anterior descending coronary arteries of mice were permanently ligated, while the NMVMs underwent continuous hypoxia. At 24 h after ischemia/hypoxia, oxidative stress was assessed by dihydroethidium and lucigenin-enhanced luminescence, mitochondrial damage by transmission electron microscopy and ATP content, and cell apoptosis by terminal deoxynucleotidyl transferase dUTP nick-end labeling and caspase-3 assay. At 4 weeks after ischemia, cardiac function and fibrosis were evaluated in mice by echocardiography and Masson's trichrome staining, respectively. Ischemic/hypoxic insult reduced Tom70 expression in cardiomyocytes. Tom70 downregulation aggravated post-MI injury, with increased mitochondrial fragmentation and ROS overload. In contrast, Tom70 upregulation alleviated post-MI injury, with improved mitochondrial integrity and decreased ROS production. PGC-1α/Tom70 expression in ischemic myocardium was increased with melatonin alone, but not when combined with luzindole. Melatonin attenuated post-MI injury in control but not in Tom70-deficient mice. N-acetylcysteine (NAC) reversed the adverse effects of Tom70 deficiency in mitochondria and cardiomyocytes, but at a much higher concentration than melatonin. Our findings showed that Tom70 is essential for melatonin-induced protection against post-MI injury, by breaking the cycle of mitochondrial impairment and ROS generation.


Melatonin/pharmacology , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Infarction/pathology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Disease Models, Animal , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Mitochondrial Precursor Protein Import Complex Proteins , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
20.
Free Radic Biol Med ; 97: 408-417, 2016 08.
Article En | MEDLINE | ID: mdl-27387769

Mitochondrial dysfunction is linked with myocardial infarction (MI), a disorder in which Notch1 has attracted increasing attention. However, the involvement of Notch1 in mitochondrial impairment after an MI is poorly understood, as is the role of mitochondrial fusion-associated protein 2 (Mfn2). Moreover, whether melatonin potentiates the Notch1/Mfn2 pathway in post-MI cardiac damage remains unclear. In our study, small interfering RNAs against Notch1 or Mfn2 and Jagged1 peptide were delivered via intramyocardial injection. At 3 days after these treatments, MI was induced by ligation of the anterior descending branch. We found that this ablation of Notch1 or Mfn2 aggravated post-MI injury, including worsened mitochondrial damage and increased generation of reactive oxygen species (ROS). In contrast, Jagged1 improved mitochondrial structure and function, decreased ROS production and attenuated post-MI injury. Interestingly, though Mfn2 expression was mildly regulated by Notch1 signaling in myocardium, Mfn2 deficiency nearly eliminated the cardioprotection by Jagged1, as evidenced by suppressed cardiac function, aggravated myocardial fibrosis, increased cell apoptosis, worsened mitochondrial impairment and enhanced oxidative stress. These observations revealed that Mfn2 plays an indispensable role in protection against MI-induced injury by Notch1. The mechanism might involve disrupting a damaging cycle of mitochondrial damage and ROS generation. Furthermore, melatonin activated Notch1 signaling and increased Mfn2 expression were reversed by luzindole, a nonselective antagonist of the melatonin receptor. Notably, melatonin attenuated post-MI injury in normal mice, but not in mice deficient in Notch1 or Mfn2. These results demonstrate that melatonin attenuates post-MI injury via the Notch1/Mfn2 pathway in a receptor-dependent manner.


Cardiotonic Agents/pharmacology , GTP Phosphohydrolases/metabolism , Melatonin/pharmacology , Myocardial Infarction/drug therapy , Receptor, Notch1/metabolism , Animals , Cardiotonic Agents/therapeutic use , Drug Evaluation, Preclinical , GTP Phosphohydrolases/genetics , Gene Expression , Jagged-1 Protein/metabolism , Male , Melatonin/therapeutic use , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Signal Transduction , Ventricular Remodeling/drug effects
...