Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Res Nurs ; 28(4): 285-298, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37534263

ABSTRACT

Backgrounds: Nursing is the key group to provide healthcare services, and it is easy for nursing staff to develop mental health problems. Aims: The study aimed to evaluate prevalence of psychological symptoms in nurses working in an intensive care unit (ICU) and the inter-relationship of associations of psychological symptoms using network analysis. Methods: This study is a cross-sectional design study. The Chinese version of the Symptom Check List-90 (SCL-90) was used to measure the psychological status of ICU nurses. The network structure of psychological symptoms was characterised, and indices of 'Expected influence' were used to identify symptoms central to the network. Network stability was examined using a case-dropping bootstrap procedure. Results: Multiple logistic regression analysis found those who had worked more than 15 years were less likely to experience positive psychological symptoms, whereas nurses working in emergency ICU and other ICUs, nurses working in departments with over 16 beds were more likely to develop psychological symptoms. In addition, 'Anxiety', 'Mental degeneration' and 'Depression' were central symptoms in the network. Conclusions: ICU nurses reported a high level of psychological symptoms, which may affect the quality of their work and worsen public health problems.

2.
Neuroscience ; 524: 220-232, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37290684

ABSTRACT

Apolipoprotein E (apoE, protein; APOE, gene), divided into three alleles of E2, E3 and E4 in humans, is associated with the progression of white matter lesion load. However, mechanism evidence has not been reported regarding the APOE genotype in early white matter injury (WMI) under subarachnoid hemorrhage (SAH) conditions. In the present study, we investigated the effects of APOE gene polymorphisms, by constructing microglial APOE3 and APOE4-specific overexpression, on WMI and underlying mechanisms of microglia phagocytosis in a mice model of SAH. A total of 167 male C57BL/6J mice (weight 22-26 g) were used. SAH and bleeding environment were induced by endovascular perforation in vivo and oxyHb in vitro, respectively. Multi-technology approaches, including immunohistochemistry, high throughput sequencing, gene editing for adeno-associated viruses, and several molecular biotechnologies were used to validate the effects of APOE polymorphisms on microglial phagocytosis and WMI after SAH. Our results revealed that APOE4 significantly aggravated the WMI and decreased neurobehavioral function by impairing microglial phagocytosis after SAH. Indicators negatively associated with microglial phagocytosis increased like CD16, CD86 and the ratio of CD16/CD206, while the indicators positively associated with microglial phagocytosis decreased like Arg-1 and CD206. The increased ROS and aggravating mitochondrial damage demonstrated that the damaging effects of APOE4 in SAH may be associated with microglial oxidative stress-dependent mitochondrial damage. Inhibiting mitochondrial oxidative stress by Mitoquinone (mitoQ) can enhance the phagocytic function of microglia. In conclusion, anti-oxidative stress and phagocytosis protection may serve as promising treatments in the management of SAH.


Subject(s)
Brain Injuries , Subarachnoid Hemorrhage , White Matter , Mice , Humans , Animals , Male , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Microglia/metabolism , Subarachnoid Hemorrhage/genetics , Subarachnoid Hemorrhage/metabolism , White Matter/pathology , Mice, Inbred C57BL , Apolipoproteins E/genetics , Brain Injuries/pathology , Apolipoprotein E3/metabolism , Phagocytosis/genetics
3.
J Affect Disord ; 329: 343-349, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36842649

ABSTRACT

BACKGROUND: The outbreak of COVID-19 disarranged lives across mainland China. No study has examined changes in psychological symptoms of healthcare professionals in the intensive care unit (ICU) before and after the outbreak of COVID-19. The aim of this study was to estimate changes in psychological symptoms of ICU healthcare professionals before and after the COVID-19 outbreak, and to analyze factors related to psychological symptoms. METHODS: Two waves' administrations were implemented between December 13 and December 14, 2018, and between April 5 and April 7, 2020, respectively. The symptom checklist-90 (SCL-90) were used to evaluate psychological symptoms. Multiple logistical regression was used to reveal the risk of psychological symptoms. RESULTS: A total of 3902 and 3908 ICU healthcare professionals took part in the first and second surveys. The mean total score of the SCL-90 was 179.27 (70.02) at wave 1 and 147.75 (58.40) at wave 2, respectively. The proportion of psychological symptoms was 55.6 % (95%CI = 54.0-57.1) at wave 1. But rates of psychological symptoms decreased to 36.6 % (95%CI = 35.1-38.2) at wave 2. ICU healthcare professionals with western economic belt and 6-10 years of work were more likely to develop psychological symptoms, while ICU healthcare professionals with the later survey and doctoral degree were less likely to develop psychological symptoms. CONCLUSION: Although COVID-19 period benefited psychological symptoms of ICU healthcare professionals, psychological symptoms still had a related high prevalence. Regular screening and appropriate interventions should still be implemented to decrease the risk for psychological symptoms among Chinese ICU healthcare professionals.


Subject(s)
COVID-19 , Health Personnel , Intensive Care Units , Humans , COVID-19/epidemiology , Cross-Sectional Studies , Delivery of Health Care , Disease Outbreaks , East Asian People , Health Personnel/psychology
4.
Cancer Gene Ther ; 30(5): 683-693, 2023 05.
Article in English | MEDLINE | ID: mdl-36575317

ABSTRACT

Glioma is a primary brain tumor with limited treatment approaches and glioblastoma stem cells (GSCs) are manifested with the self-renewal capability and high tumorigenic capacity. This study was performed to investigate the regulatory effect of the SUMO-specific protease 1 (SENP1)/methyltransferase-like 3 (METTL3)/MYC axis on the self-renewal of GSCs mediated by transcription factor Yin Yang 1 (YY1). Following bioinformatics analysis and clinical and cellular experiments, we found that YY1 was highly expressed in GBM tissues and cells, while silencing its expression reduced the self-renewal ability of GSCs. Functionally, YY1 promoted the transcriptional expression of SENP1 by binding to the promoter region of SENP1, while the deSUMOase SENP1 facilitated the methylase activity of m6A through deSUMOylation of the methylase METTL3, thereby promoting the m6A modification of MYC mRNA via METL3 and promoting the expression of MYC. A nude mouse xenograft model of GBM was also constructed to examine the tumorigenicity of GSCs. The obtained findings demonstrated that YY1 promoted tumorigenicity of GSCs by promoting the expression of MYC in vivo. Conclusively, YY1 can transcriptionally upregulate the SUMOylase SENP1 and enhance the methylase activity of METTL3, resulting in the increased m6A modification level of MYC mRNA, thereby promoting the self-renewal of GSCs.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Mice , Humans , Glioblastoma/pathology , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism , Peptide Hydrolases/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/metabolism , Brain Neoplasms/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Methyltransferases/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism
5.
J Clin Med ; 11(22)2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36431168

ABSTRACT

OBJECTIVE: Venous thromboembolism (VTE) is a life threating complication in intensive care units (ICUs). This study aimed to pool the prevalence of VTE and examined the risk factors of VTE in intensive care patients worldwide. METHODS: A systematic search in PubMed, EMBASE and Web of Science databases was performed. Studies reported that the data on the prevalence of VTE or relevant information were synthesized using a random-effects model. RESULTS: A total of 42 studies reporting on 27,344 patients were included. The pooled prevalence of VTE was 10.0% (95% CI: 7.0-14.0%). Subgroup and metaregression analyses found that thromboprophylaxis strategy, simplified acute physiology score (SAPS II), age, study quality, sample size, malignancy, sex, spinal cord injury and injury severity score (ISS) moderated the prevalence of VTE in intensive care patients. CONCLUSIONS: The present meta-analysis revealed a high prevalence of VTE in critically ill patients. The risk factors of VTE included thromboprophylaxis strategy, SAPS II, age, malignancy, sex, spinal cord injury and ISS. Therefore, we need to pay more attention to high-risk populations of VTE in intensive care patients.

6.
Am J Cancer Res ; 12(6): 2558-2575, 2022.
Article in English | MEDLINE | ID: mdl-35812057

ABSTRACT

Tumor cells exhibit enhanced uptake and processing of nutrients to fulfill the demands of rapid growth of tumor tissues. Tryptophan metabolizing dioxygenases are frequently up-regulated in several tumor types, which has been recognized as a crucial determinant in accelerated tumor progression. In our study, we explored the specific role of tryptophan 2,3-dioxygenase 2 (TDO2) in glioma progression. Analysis of mRNA profiles in 325 glioma patients based on the rich set of CCGA database was performed, which revealed that high TDO2 expression was tightly correlated with poor prognosis in glioma patients. TDO2 increased intracellular levels of tryptophan metabolism in the kynurenine (Kyn) pathway in vitro and in vivo, resulting in sustained glioma cell proliferation. Mechanistically, overexpression of TDO2 promoted the secretion of Kyn, which in turn stimulated the activation of the aryl hydrocarbon receptor (AhR)/AKT signaling pathway, resulting in heightened proliferative properties and tumorigenic potential in glioma cells. Meanwhile, Kyn produced by tumor cells further suppressed the proliferation of functional T cells, thereby resulting in immunosuppression and enhanced tumor growth in glioma. Our study showed that TDO2-induced increase in tryptophan metabolite Kyn played a pivotal role in glioma development via the AhR/AKT pro-survival signals and immunosuppressive effects, suggesting that the use of TDO2 inhibitors in combination with chemotherapy may be a novel strategy to effectively and synergistically eliminate glioma cells.

7.
Pharmacol Res Perspect ; 10(4): e00981, 2022 08.
Article in English | MEDLINE | ID: mdl-35796398

ABSTRACT

Glioblastoma angiogenesis is critical for tumor growth, making it an appealing target for treatment development. BAY1143269 is a novel inhibitor of mitogen-activated protein kinase interacting serine/threonine-protein kinase 1 (MKN1) and has potent anti-cancer activity. We identified BAY1143269 as an angiogenesis inhibitor, by in vitro and in vivo glioblastoma angiogenesis models. BAY1143269 inhibited the capillary network formation of glioblastoma microvascular endothelial cells (GMECs), particularly the early stage of tubular structure formation. It also inhibited migration and proliferation, and induced apoptosis of GMECs isolated from glioblastoma patients. We found that BAY1143269 acted on GMECs by suppressing the eukaryotic translation initiation factor 4E (eIF4E) and eIF4E-mediated expression of oncogenic proteins, including those involved in cell cycle, epithelial-mesenchymal transition (EMT), and pro-survival. In addition, BAY1143269 suppressed eIF4E phosphorylation, inhibited proliferation, and induced apoptosis of glioblastoma cells. Interestingly, it reduced vascular endothelial growth factor (VEGF) level in tumor cells and culturing medium, demonstrating the inhibitory effect of BAY1143269 on tumor proangiogenic microenvironment. We finally challenged BAY1143269 on the glioblastoma xenograft mice model and observed a significant tumor growth reduction without toxicity in mice receiving oral BAY1143269. Immunoblotting analysis demonstrated significantly less phosphorylated-eIF4E (p-eIF4E), cluster of differentiation 31 (CD31) (microvascular endothelial cell marker), and VEGF in tumors from drug-treated mice. In summary, the inhibition of glioblastoma angiogenesis with BAY1143269 may provide an alternative approach for anti-glioblastoma therapy.


Subject(s)
Glioblastoma , Vascular Endothelial Growth Factor A , Animals , Endothelial Cells , Eukaryotic Initiation Factor-4E , Glioblastoma/drug therapy , Humans , Immunotherapy , Mice , Tumor Microenvironment
8.
Int J Biol Sci ; 18(11): 4578-4594, 2022.
Article in English | MEDLINE | ID: mdl-35864966

ABSTRACT

Ischemic stroke is characterized by the presence of both brain ischemic and reperfusion-induced injuries in the brain, leading to neuronal dysfunction and death. Artemisinin, an FDA-approved antimalarial drug, has been reported to have neuroprotective properties. However, the effect of artemisinin on ischemic stroke is not known. In the present study, we investigated the effect of artemisinin on ischemic stroke using an oxygen-glucose deprivation/reperfusion (OGD/RP) cellular model and a mouse middle cerebral artery occlusion (MCAO) animal model and examined the underlying mechanisms. The obtained results revealed that a subclinical antimalarial concentration of artemisinin increased cell viability and decreased LDH release and cell apoptosis. Artemisinin also attenuated the production of reactive oxygen species (ROS) and the loss of mitochondrial membrane potential (Δψm). Importantly, artemisinin attenuated the infarction volume and the brain water content in the MCAO animal model. Artemisinin also improved neurological and behavioural outcomes and restored grasp strength and the recovery of motor function in MCAO animals. Furthermore, artemisinin treatment significantly inhibited the molecular indices of apoptosis, oxidative stress and neuroinflammation and activated the ERK1/2/CREB/BCL-2 signaling pathway. Further validation of the involved signaling pathway by the ERK1/2 inhibitor PD98059 revealed that inhibiting the ERK1/2 signaling pathway or silencing ERK1/2 reversed the neuroprotective effects of artemisinin. These results indicate that artemisinin provides neuroprotection against ischemic stroke via the ERK1/2/CREB/BCL-2 signaling pathway. Our study suggests that artemisinin may play an important role in the prevention and treatment of stroke.


Subject(s)
Artemisinins , Brain Ischemia , Ischemic Stroke , Neuroprotective Agents , Reperfusion Injury , Animals , Apoptosis , Artemisinins/pharmacology , Artemisinins/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Disease Models, Animal , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Stroke/drug therapy , MAP Kinase Signaling System , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Reperfusion Injury/metabolism , Signal Transduction
9.
Cell Death Dis ; 13(2): 160, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35181676

ABSTRACT

Long non-coding RNAs (lncRNAs) have been manifested to manipulate diverse biological processes, including tumor-induced immune tolerance. Thus, we aimed in this study to identify the expression pattern of lncRNA homeobox A cluster antisense RNA 2 (HOXA-AS2) in glioma and decipher its role in immune tolerance and glioma progression. We found aberrant upregulation of lncRNA HOXA-AS2, lysine demethylase 2A (KDM2A), and jagged 1 (JAG1) and a downregulation of microRNA-302a (miR-302a) in glioma specimens. Next, RNA immunoprecipitation, chromatin immunoprecipitation, and dual-luciferase reporter gene assay demonstrated that lncRNA HOXA-AS2 upregulated KDM2A expression by preventing miR-302a from binding to its 3'untranslated region. The functional experiments suggested that lncRNA HOXA-AS2 could promote regulatory T (Treg) cell proliferation and immune tolerance, which might be achieved through inhibition of miR-302a and activation of KDM2A/JAG1 axis. These findings were validated in a tumor xenograft mouse model. To conclude, lncRNA HOXA-AS2 facilitates KDM2A/JAG1 expression to promote Treg cell proliferation and immune tolerance in glioma by binding to miR-302a. These findings may aid in the development of novel antitumor targets.


Subject(s)
F-Box Proteins , Glioma , RNA, Antisense , RNA, Long Noncoding , Animals , Cell Line, Tumor , Cell Proliferation/genetics , F-Box Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Humans , Immune Tolerance , Jagged-1 Protein/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , MicroRNAs/metabolism , RNA, Antisense/metabolism , RNA, Long Noncoding/metabolism
10.
Int J Biol Sci ; 18(2): 459-472, 2022.
Article in English | MEDLINE | ID: mdl-35002503

ABSTRACT

The COVID-19 outbreak is emerging as a significant public health challenge. Excessive production of proinflammatory cytokines, also known as cytokine storm, is a severe clinical syndrome known to develop as a complication of infectious or inflammatory diseases. Clinical evidence suggests that the occurrence of cytokine storm in severe acute respiratory syndrome secondary to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is closely associated with the rapid deterioration and high mortality of severe cases. In this review, we aim to summarize the mechanism of SARS-CoV-2 infection and the subsequent immunological events related to excessive cytokine production and inflammatory responses associated with ACE2-AngII signaling. An overview of the diagnosis and an update on current therapeutic regimens and vaccinations is also provided.


Subject(s)
COVID-19/complications , COVID-19/pathology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/pathology , SARS-CoV-2 , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/therapy , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/immunology , Humans
11.
Interv Neuroradiol ; 28(4): 482-488, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34519241

ABSTRACT

BACKGROUND: The treatment of posterior inferior cerebellar artery aneurysms is controversial. Recently, flow diverters have emerged as an attractive treatment option. Here, we performed a systematic review and meta-analysis of the angiographic and clinical outcomes of flow diverter-treated posterior inferior cerebellar artery aneurysms. METHODS: We searched the PubMed, EMBASE and Web of Science databases for studies published from inception to January 2021. We included studies that described flow diverters procedures for posterior inferior cerebellar artery aneurysms with ≥2 patients. The outcomes were the complete occlusion rate and flow diverter-related complication rate. Meta-analysis was performed using a random or fixed effects model based on heterogeneity. RESULTS: We included six studies involving 46 posterior inferior cerebellar artery aneurysms. There were 14 ruptured aneurysms. All flow diverters were successfully released and the technical success rate was 100%. The complete occlusion rate was 81% (95% confidence interval = 65-93%; I2 = 0.00%; P < 0.01). The flow diverter-related complication rate was 18% (95% confidence interval = 5-36%; I2 = 0.00%; P < 0.01). One patient died of rebleeding. The mortality rate was <1% (95% confidence interval = -1-1%; I2 = 0.00%; P = 0.951). CONCLUSIONS: Treatment of posterior inferior cerebellar artery aneurysms with flow diverters is feasible and carries a high degree of technical success. However, this treatment is underutilized in patients with posterior inferior cerebellar artery aneurysms due to a higher complication rate and lower occlusion rate compared with clipping and traditional endovascular treatment. Further well-designed prospective and randomized studies are required to fully understand the effects of flow diverters especially in posterior inferior cerebellar artery aneurysms patients requiring endovascular treatment.


Subject(s)
Endovascular Procedures , Intracranial Aneurysm , Endovascular Procedures/methods , Humans , Intracranial Aneurysm/diagnostic imaging , Intracranial Aneurysm/surgery , Prospective Studies , Retrospective Studies , Stents , Treatment Outcome , Vertebral Artery
12.
Oxid Med Cell Longev ; 2021: 3823122, 2021.
Article in English | MEDLINE | ID: mdl-34790286

ABSTRACT

BACKGROUND: Circular RNA phosphorylase kinase regulatory subunit alpha 2 (circPHKA2; hsa_circ_0090002) has a significantly, specifically different expression in acute ischemic stroke (AIS) patients' blood. Here, we intended to investigate the role and mechanism of circPHKA2 in oxygen-glucose deprivation- (OGD-) induced stoke model in human brain microvascular endothelial cells (HBMEC). METHODS: Expression of circPHKA2, microRNA- (miR-) 574-5p, and superoxide dismutase-2 (SOD2) was detected by quantitative PCR and western blotting. Cell injury was measured by detecting cell proliferation (EdU assay and CCK-8 assay), migration (transwell assay), neovascularization (tube formation assay), apoptosis (flow cytometry and western blotting), endoplasmic reticulum stress (western blotting), and oxidative stress (assay kits). Direct intermolecular interaction was determined by bioinformatics algorithms, dual-luciferase reporter assay, biotin-labelled miRNA capture, and argonaute 2 RNA immunoprecipitation. RESULTS: circPHKA2 was downregulated in AIS patients' blood in SOD2-correlated manner. Reexpressing circPHKA2 rescued EdU incorporation, cell viability and migration, tube formation, B cell lymphoma-2 (Bcl-2) expression, and SOD activity of OGD-induced HBMEC and alleviate apoptotic rate and levels of Bcl-2-associated protein (Bax), glucose-regulated protein 78 kD (GRP78), C/EBP-homologous protein (CHOP), caspase-12, reactive oxygen species (ROS), and malondialdehyde (MDA). Additionally, blocking SOD2 partially attenuated these roles of circPHKA2 overexpression. Molecularly, circPHKA2 upregulated SOD2 expression via interacting with miR-574-5p, and miR-574-5p could target SOD2. Similarly, allied to neurovascular protection of circPHKA2 was the downregulation of miR-574-5p. CONCLUSION: circPHKA2 could protect HBMEC against OGD-induced cerebral stroke model via the miR-574-5p/SOD2 axis, suggesting circPHKA2 as a novel and promising candidate in ischemic brain injury.


Subject(s)
Endothelium, Vascular/metabolism , Glucose/deficiency , Hypoxia/complications , Ischemic Stroke/therapy , MicroRNAs/genetics , Phosphorylase Kinase/genetics , RNA, Circular/administration & dosage , Superoxide Dismutase/metabolism , Case-Control Studies , Endothelium, Vascular/pathology , Gene Expression Regulation , Humans , Ischemic Stroke/etiology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , RNA, Circular/genetics , Superoxide Dismutase/genetics
13.
Clin Transl Med ; 11(9): e424, 2021 09.
Article in English | MEDLINE | ID: mdl-34586733

ABSTRACT

Glioma is regarded as an aggressive lethal primary brain tumor. Jumonji domain containing 1C (JMJD1C) is a H3K9 demethylase which participates in the progression of various tumors, but its specific function and underlying mechanism in glioma development remain undefined, which is the purpose of our work. We initially assessed JMJD1C expression in glioma tissues and cells using the assays of RT-qPCR and immunohistochemistry. Meanwhile, the H3K9 level at the microRNA (miR)-302a promoter region was measured by chromatin immunoprecipitation assay, while luciferase-based reporter assay was performed for validation of the binding affinity between miR-302a and methyltransferase-like 3 (METTL3). The effect of METTL3 on suppressor of cytokine signaling 2 (SOCS2) was subsequently analyzed by MeRIP-RT-qPCR. Finally, a xenograft tumor model was established in nude mice, followed by measurement of tumor-associated macrophages using flow cytometry. JMJD1C was poorly expressed in glioma tissues. Furthermore, JMJD1C increased miR-302a expression through promoting H3K9me1 demethylation at the miR-302a promoter region. miR-302a was identified to target METTL3, which could inhibit SOCS2 expression via m6A modification. JMJD1C promoted M1 macrophage polarization and suppressed the growth of glioma xenografts through the miR-302a/METTL3/SOCS2 axis both in vivo and in vitro. In conclusion, JMJD1C could enhance M1 macrophage polarization to inhibit the onset of glioma, bringing a new insight into the contribution of JMJD1C to the pathobiology of glioma, with possible implications for targeted therapeutic method.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , MicroRNAs/genetics , Oxidoreductases, N-Demethylating/genetics , Adult , Aged , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Polarity/genetics , Female , Glioma/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Nude , MicroRNAs/metabolism , Middle Aged , Oxidoreductases, N-Demethylating/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Young Adult
14.
Front Public Health ; 9: 572415, 2021.
Article in English | MEDLINE | ID: mdl-34485206

ABSTRACT

Objective: To understand the impact of COVID-19 epidemic on the mental health status of intensive care unit (ICU) practitioners in China, and to explore the relevant factors that may affect the mental health status of front-line medical workers so as to adopt efficient and comprehensive measures in a timely manner to protect the mental health of medical staff. Methods: The study covered most of the provinces in China, and a questionnaire survey was conducted based on the WeChat platform and the Wenjuanxing online survey tool. With the method of anonymous investigation, we chose ICU practitioners to participate in the investigation from April 5, 2020 to April 7, 2020. The respondents were divided into two groups according to strict criteria of inclusion and exclusion, those who participated in the rescue work of COVID-19 (COVID-19 group) and those who did not (non-COVID-19 group). The SCL-90 self-evaluation scale was used for the evaluation of mental health status of the subjects. Results: A total of 3,851 respondents completed the questionnaire. First, the overall mental health status of the targeted population, compared with the Chinese norm (n = 1,388), was reflected in nine related factor groups of the SCL-90 scale, and significant differences were found in every factor in both men and women, except for the interpersonal sensitivity in men. Second, the overall mental health of the non-COVID-19 group was worse than that of the COVID-19 group by the SCL-90 scale (OR = 1.98, 95% CI, 1.682-2.331). Third, we have revealed several influencing factors for their mental health in the COVID-19 group, current working status (P < 0.001), satisfaction of diet and accommodation (P < 0.05), occupational exposure (P = 0.005), views on the risk of infection (P = 0.034), and support of training (P = 0.01). Conclusion: The mental health status of the ICU practitioners in the COVID-19 group is better than that of the non-COVID-19 group, which could be attributed to a strengthened mentality and awareness of risks related to occupational exposure and enforced education on preventive measures for infectious diseases, before being on duty.


Subject(s)
COVID-19 , Epidemics , China/epidemiology , Female , Health Status , Humans , Intensive Care Units , Male , SARS-CoV-2
15.
Redox Biol ; 46: 102069, 2021 10.
Article in English | MEDLINE | ID: mdl-34303216

ABSTRACT

Ischemic stroke is one of the leading causes of death and disability among adults. Despite the economic burden of the disease, available treatment options are still very limited. With the exception of anti-thrombolytics and hypothermia, current therapies fail to reduce neuronal injury, neurological deficits and mortality rates, suggesting that the development of novel and more effective therapies against ischemic stroke is urgent. In the present study, we found that artemether, which has been used in the clinic as an anti-malarial drug, was able to improve the neurological deficits, attenuate the infarction volume and the brain water content in a middle cerebral artery occlusion (MCAO) animal model. Furthermore, artemether treatment significantly suppressed cell apoptosis, stimulated cell proliferation and promoted the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), P90rsk and cAMP responsive element-binding protein (CREB). Artemether protective effect was attenuated by PD98059, an ERK1/2 inhibitor, administration. Similarly, in oxygen-glucose deprivation/reperfusion (OGD/RP) cell models, artemether pre-treatment induced the suppression of the intracellular ROS, the down-regulation of LDH activity, the reduction of caspase 3 activity and of the apoptosis cell rate and reversed the decrease of mitochondrial membrane potential. As with MCAO animal model, artemether promoted the activation of Erk1/2-P90rsk-CREB signaling pathway. This effect was blocked by the inhibition or knock-down of ERK1/2. The present study provides evidences of the neuroprotective effect of artemether unravelling its potential as a new therapeutic candidate for the prevention and treatment of stroke.


Subject(s)
Brain Ischemia , Neuroprotective Agents , Reperfusion Injury , Animals , Apoptosis , Artemether/pharmacology , Brain Ischemia/drug therapy , MAP Kinase Signaling System , Neuroprotection , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/pharmacology , Signal Transduction
16.
Mol Biol Rep ; 48(5): 4137-4151, 2021 May.
Article in English | MEDLINE | ID: mdl-34110576

ABSTRACT

The purpose of this study was to explore the effect of miR-34c on PDGF-BB-induced HAVSMCs phenotypic transformation and proliferation via PDGFR-ß/SIRT1 pathway, so as to find a new method for early diagnosis and treatment of cardiovascular disease. HA-VSMCs were treated with platelet-derived growth factor-BB (PDGF-BB) at 0 h, 12 h, 24 h, 48 h or 36 h to explore the optimal time for phenotypic transformation of VSMCs. And then, PDGF-BB-induced HA-VSMCs were transfected with miR-34c mimics/mimics NC and pcDNA3.1-PDGFR-ß/pcDNA3.1-NC to observe cell biological behaviour. CCK8 was used to detect cell proliferation activity. Transwell chamber assay was used to detect cell invasion. Early apoptosis was analyzed by flow cytometry. The expression of α-SMA and Smemb was detected by immunofluorescence staining. The expressions of PDGFR-ß, IRF9, Acetyl-NF-κB/p65, Acetyl-p53 and CyclinD1 were analyzed by Western blot analysis. The expression of miR-34a, miR-34b and miR-34c was detected by RT-PCR, and the targeting relationship between miR-34c and PDGFR-ß was detected by luciferase reporting assay. The results indicated the proliferation and migration of PDGF-BB-induced HA-VSMCs significantly increased, and apoptosis significantly decreased. Besides, α-SMA decreased significantly, while Smemb increased significantly. Furthermore, expressions of PDGFR-ß, IRF9, Acetyl-NF-κB/p65, Acetyl-p53 and CyclinD1 increased significantly, and SIRT1 decreased significantly. Experimental results showed that, miR-34c mimics significantly inhibited cell proliferation and migration, and promoted cell apoptosis, and miR-34c inhibitor had the opposite effects. MiR-34c mimics significantly increased α-SMA expression and decreased Smemb expression, while the opposite effects were reflected after transfection with miR-34c inhibitor. Moreover, miR-34c mimics significantly decreased the expressions of PDGFR-ß, IRF9, Acetyl-NF-κB/p65, Acetyl-p53 and CyclinD1, and significantly increased the expression of SIRT1, while miR-34c inhibitor had the opposite effects. Luciferase assay confirmed that PDGFR-ß was a potential target of miR-34c. Subsequently, PDGF-BB-induced HA-VSMCs were co-transfected with miR-34c mimics and pcDNA3.1-PDGFR-ß. The results indicated that PDGFR-ß reversed the biological function of miR-34c mimic. The results revealed the potential application value of miR-34c as a marker molecule of phenotypic transformation, providing a potential target for improving phenotypic transformation.


Subject(s)
Becaplermin/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirtuin 1/metabolism , Aorta/cytology , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Humans , Hypertension/genetics , Hypertension/metabolism , MicroRNAs/genetics , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Receptor, Platelet-Derived Growth Factor beta/genetics , Transfection , Vascular Remodeling/genetics
17.
Adv Drug Deliv Rev ; 173: 439-460, 2021 06.
Article in English | MEDLINE | ID: mdl-33857553

ABSTRACT

Diabetic retinopathy is a frequent microvascular complication of diabetes and a major cause of visual impairment. In advanced stages, the abnormal neovascularization can lead to fibrosis and subsequent tractional retinal detachment and blindness. The low bioavailability of the drugs at the target site imposed by the anatomic and physiologic barriers within the eye, requires long term treatments with frequent injections that often compromise patient's compliance and increase the risk of developing more complications. In recent years, much effort has been put towards the development of new drug delivery platforms aiming to enhance their permeation, to prolong their retention time at the target site and to provide a sustained release with reduced toxicity and improved efficacy. This review provides an overview of the etiology and pathophysiology of diabetic retinopathy and current treatments. It addresses the specific challenges associated to the different ocular delivery routes and provides a critical review of the most recent developments made in the drug delivery field.


Subject(s)
Diabetic Retinopathy/drug therapy , Drug Delivery Systems , Fibrosis/drug therapy , Hypoglycemic Agents/therapeutic use , Animals , Humans , Hypoglycemic Agents/chemistry
18.
Aging (Albany NY) ; 13(4): 5284-5296, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33535177

ABSTRACT

Matrix stiffness is a key physical characteristic of the tumor microenvironment and correlates tightly with tumor progression. Here, we explored the association between matrix stiffness and glioma development. Using atomic force microscopy, we observed higher matrix stiffness in highly malignant glioma tissues than in low-grade/innocent tissues. In vitro and in vivo analyses revealed that culturing glioma cells on stiff polyacrylamide hydrogels enhanced their proliferation, tumorigenesis and CD133 expression. Greater matrix stiffness could obviously up-regulated the expression of BCL9L, thereby promoting the activation of Wnt/ß-catenin signaling and ultimately increasing the stemness of glioma cells. Inhibiting Wnt/ß-catenin signaling using gigantol consistently improved the anticancer effects of chemotherapy and radiotherapy in mice with subcutaneous glioma tumors. These findings demonstrate that a stiffer matrix increases the stemness of glioma cells by activating BCL9L/Wnt/ß-catenin signaling. Moreover, we have provided a potential strategy for clinical glioma treatment by demonstrating that gigantol can improve the effectiveness of traditional chemotherapy/radiotherapy by suppressing Wnt/ß-catenin signaling.


Subject(s)
Brain Neoplasms/metabolism , Extracellular Matrix/pathology , Glioma/metabolism , Transcription Factors/metabolism , Tumor Microenvironment , Wnt Signaling Pathway , AC133 Antigen/metabolism , Acrylic Resins , Animals , Antineoplastic Agents , Bibenzyls/pharmacology , Brain Neoplasms/pathology , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Glioma/pathology , Guaiacol/analogs & derivatives , Guaiacol/pharmacology , Humans , Mice , Microscopy, Atomic Force , Neoplasm Grading , Neoplasm Transplantation , Neoplastic Stem Cells , Radiotherapy , Xenograft Model Antitumor Assays
19.
Inflamm Res ; 70(3): 285-296, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33507312

ABSTRACT

OBJECTIVE: microRNAs (miRNAs) play critical roles in embryogenesis, cell differentiation and the pathogenesis of several human diseases, including systemic lupus erythematosus (SLE). Toll-like receptors (TLRs) are also known to exert crucial functions in the immune response activation occurring in the pathogenesis of autoimmune diseases like SLE. Herein, the current study aimed to explore the potential role of miR-152-3p in TLR-mediated inflammatory response in SLE. METHODS: We determined the miR-152-3p expression profiles in CD4+ T cells and peripheral blood mononuclear cells (PBMCs) harvested from patients with SLE and healthy controls, and analyzed the correlation between miR-152-3p expression and clinicopathological parameters. CD70 and CD40L expression patterns in CD4+ T cells were assessed by RT-qPCR and flow cytometry. ChIP was adopted to determine the enrichment of DNA methyltransferase 1 (DNMT1) in the promoter region of myeloid differentiation factor 88 (MyD88). RESULTS: The obtained findings revealed that miR-152-3p was highly-expressed in CD4+ T cells and PBMCs of patients with SLE, and this high expression was associated with facial erythema, joint pain, double-stranded DNA, and IgG antibody. DNMT1 could be enriched in the MyD88 promoter, and miR-152-3p inhibited the methylation of MyD88 by targeting DNMT1. We also found that silencing miR-152-3p inhibited MyD88 expression not only to repress the autoreactivity of CD4+ T cells and but also to restrain their cellular inflammation, which were also validated in vivo. CONCLUSION: Our study suggests that miR-152-3p promotes TLR-mediated inflammatory response in CD4+ T cells by regulating the DNMT1/MyD88 signaling pathway, which highlights novel anti-inflammatory target for SLE treatment.


Subject(s)
Lupus Erythematosus, Systemic/genetics , MicroRNAs , Adolescent , Adult , Aged , Animals , Antibodies, Anti-Idiotypic/blood , Antibodies, Antinuclear/blood , Arthralgia/genetics , Arthralgia/immunology , Child , Cytokines/immunology , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/immunology , Demethylation , Erythema/genetics , Erythema/immunology , Face , Female , Humans , Inflammation/genetics , Inflammation/immunology , Leukocytes, Mononuclear/immunology , Lupus Erythematosus, Systemic/immunology , Male , Mice, Inbred MRL lpr , Middle Aged , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptors/immunology , Young Adult
20.
Theranostics ; 11(4): 1991-2005, 2021.
Article in English | MEDLINE | ID: mdl-33408794

ABSTRACT

Cancer development is a complex set of proliferative progression, which arises in most cases via multistep pathways associated with various factors, including the tumor microenvironment and extracellular matrix. However, the underlying mechanisms of cancer development remain unclear and this study aimed to explore the role of extracellular matrix in glioma progression. Methods: The expression of type I collagen and fibronectin in tumor tissues from glioma patients was examined by immunofluorescence staining. The correlations between collagen/fibronectin and glioma progression were then analyzed. A 3D collagen/fibronectin cultured system was established for tumor cells culture in vitro. Quantitative, real-time PCR and western blot were used to detect PI3K/ATK and CDC42 signals associated proteins expression in glioma. We used in vitro Cell Counting Kit-8, colony formation, and tumorigenesis assays to investigate the function of PI3K/AKT and CDC42 signals associated proteins. A xenograft glioma mice model was also used to study the anticancer effects of integrin inhibitor in vivo. Results: Our study demonstrated that type I collagen and fibronectin collaborate to regulate glioma cell stemness and tumor growth. In a 3D collagen/fibronectin culture model, glioma cells acquired tumorigenic potential and revealed strengthened proliferative characteristics. More significantly, collagen/fibronectin could facilitate the activation of PI3K/AKT/SOX2 and CDC42/YAP-1/NUPR1/Nestin signaling pathways via integrin αvß3, eliciting sustained tumor growth and cancer relapse. Combination of the integrin signaling pathway inhibitor and the chemotherapeutic agent efficiently suppressed glioma cell proliferation and tumorigenic ability. Conclusion: We demonstrated that type I collagen and fibronectin could collaborate to promote glioma progression through PI3K/AKT/SOX2 and CDC42/YAP-1/NUPR1/Nestin signaling pathways. Blockade of the upstream molecular integrin αvß3 revealed improved outcome in glioma therapy, which provide new insights for eradicating tumors and reducing glioma cancer relapse.


Subject(s)
Collagen/metabolism , Fibronectins/metabolism , Glioma/pathology , Neoplastic Stem Cells/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Fibronectins/genetics , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Humans , Mice , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , cdc42 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...