Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Sci Rep ; 14(1): 11782, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38782996

ABSTRACT

Killer cell lectin-like receptor B1 (KLRB1) is implicated in cancer progression and immunity. In this study, we aimed to evaluate the expression levels of KLRB1 in lung adenocarcinoma (LUAD) and analyze the relationship between KLRB1 expression levels, LUAD progression, and the tumor immune microenvironment. KLRB1 levels in LUAD were analyzed using data from the TCGA and XENA databases. Additionally, the diagnostic values of KLRB1 were analyzed in patients with LUAD. Survival and meta-analyses were employed to investigate the relationship between KLRB1 levels and other prognostic factors in patients with LUAD. Bioinformatics and cellular experiments were used to understand the functions and mechanisms of KLRB1. In addition, correlation analysis was used to investigate the relationship between KLRB1 levels and the immune microenvironment in LUAD. Reduced KLRB1 expression in LUAD was found to positively correlate with tumor size, distant metastasis, pathological stage, age, overall survival, diagnostic value, and disease-specific survival in patients with LUAD (P < 0.05). Conversely, increased KLRB1 expression was found to positively correlate with the overall survival and disease-specific survival in patients with LUAD (P < 0.05). We also found that the overexpression of KLRB1 can inhibit the proliferation, migration, and invasion of LUAD cells and promote apoptosis. KLRB1 was involved in immune cell differentiation, NF-kB, PD-L1, and PD-1 checkpoint pathways and others. Additionally, KLRB1 expression was linked to tumor purity, stromal, immune, and estimate scores, the levels of immune cells including B cells, CD8+ T cells, and CD4+ T cells, and immune cell markers in LUAD. Reduced KLRB1 expression has a significant positive correlation with diagnosis, poor prognosis, and immunity to cancer in patients with LUAD. KLRB1 inhibited cell proliferation and migration in patients with LUAD. These results suggest that KLRB1 may serve as a potential therapeutic target in patients with LUAD.


Subject(s)
Adenocarcinoma of Lung , Cell Proliferation , Lung Neoplasms , Tumor Microenvironment , Female , Humans , Male , Middle Aged , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/mortality , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Neoplasm Metastasis , Prognosis , Tumor Microenvironment/immunology
2.
Aging (Albany NY) ; 16(2): 1605-1619, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38244585

ABSTRACT

BACKGROUND: Overexpression of solute carrier family 7 member 8 (SLC7A8) has been shown to relate to the survival time and tumor progression in cancer patients. However, the role of SLC7A8 in lung adenocarcinoma (LUAD) is still obscure. METHOD: The relationships between SLC7A8 expression in LUAD tissues and clinical values as well as immune infiltration were explored through bioinformatics. The functions and pathways of SLC7A8 in LUAD were investigated using Kyoto Encyclopedia of Genes and Genomes enrichment analysis, Gene Set Enrichment Analysis, Western blotting, and other methods. RESULTS: We found that the expression of SLC7A8 was decreased significantly in LUAD tissues compared with normal tissues, which was related to the dismal survival time and disease progression. Moreover, it carried diagnostic value in LUAD and was a risk factor for dismal prognosis. Receiver operating characteristic curve analysis indicated that the expression level of SLC7A8 carried significant diagnostic value in LUAD. Overexpression of SLC7A8 inhibited the proliferation, invasion, and migration of LUAD cells, likely through a mechanism involving the cell cycle. SLC7A8 expression in LUAD was significantly correlated with the infiltration of immune cells, especially B cells, interstitial dendritic cells, mast cells, CD56 bright cells, natural killer cells, plasmacytoid dendritic cells, T follicular helper cells, T helper 2 and 17 cells, and immune factors. CONCLUSION: The downregulation of SLC7A8 was related to a dismal prognosis and immune cell infiltration in LUAD. Increasing the expression of SLC7A8 inhibited the growth and migration of LUAD cells, thereby improving the prognosis of patients.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Humans , Adenocarcinoma of Lung/genetics , Prognosis , Disease Progression , Lung Neoplasms/genetics , Amino Acid Transport System y+ , Fusion Regulatory Protein 1, Light Chains
3.
J Am Med Dir Assoc ; 25(3): 431-438.e15, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37660722

ABSTRACT

OBJECTIVES: Physical activity (PA) and telomeres both contribute to healthy aging and longevity. To investigate the optimal dosage of various PA for longevity and the role of telomere length in PA and mortality. DESIGN: Prospective cohort study. SETTING AND PARTICIPANTS: A total of 333,865 adults (mean age of 56 years) from the UK Biobank were analyzed. METHODS: Walking, moderate PA (MPA), and vigorous PA (VPA) were self-reported via questionnaire, and leukocyte telomere length (LTL) was measured. Cox proportional hazards regression was used to predict all-cause mortality risk. A flexible parametric Royston-Parmar survival model was used to estimate life expectancy. RESULTS: During a median follow-up of 13.8 years, 19,789 deaths were recorded. Compared with the no-walking group, 90 to 720 minutes/week of walking was similarly associated with 27% to 31% of lower mortality and about 6 years of additional life expectancy. We observed nearly major benefits for mortality and life expectancy among those meeting the PA guidelines [151-300 minutes/wk for MPA: hazard ratio (HR) 0.80, 95% CI 0.75-0.85, 3.40-3.42 additional life years; 76-150 minutes/wk for VPA: HR 0.78, 95% CI 0.75-0.82, 2.61 years (2.33-2.89)] vs the no-PA group. Similar benefits were also observed at 76-150 and 301-375 minutes/wk of MPA (18%-19% lower mortality, 3.20-3.42 gained years) or 151-300 minutes/wk of VPA (20%-26% lower mortality, 2.41-2.61 gained years). The associations between MPA, VPA, and mortality risk were slightly mediated by LTL (≈1% mediation proportion, both P < .001). CONCLUSIONS AND IMPLICATIONS: Our study suggests a more flexible range of PA than the current PA guidelines, which could gain similar benefits and is easier to achieve: 90 to 720 minutes/wk of walking, 75 to 375 minutes/wk of MPA, and 75 to 300 minutes/wk of VPA. Telomeres might be a potential mechanism by which PA promotes longevity.


Subject(s)
Exercise , Life Expectancy , Adult , Humans , Middle Aged , Prospective Studies , Longevity , Telomere
4.
Aging (Albany NY) ; 15(22): 13265-13286, 2023 11 20.
Article in English | MEDLINE | ID: mdl-37988189

ABSTRACT

BACKGROUND: The association between Killer cell lectin like receptor B1 (KLRB1) and cancer has been reported, but the roles of KLRB1 in breast invasive carcinoma (BRCA) has not been fully revealed. METHODS: Our study utilized the Cancer Genome Atlas (TCGA), Kaplan-Meier (K-M) Plotter, and TIMER databases to investigate the expression and clinical relevance of KLRB1 in BRCA and to explore its roles and mechanism in BRCA progression using gene set enrichment analysis, CCK-8, migration, apoptosis, and western blotting. We examined the relationship between KLRB1 expression and the BRCA immune microenvironment, using data from TCGA, and Gene Expression Profiling Interactive Analysis (GEPIA) databases and validated these findings in K-M Plotter databases. RESULTS: A significant decrease of KLRB1 expression was observed in BRCA patients. BRCA patients with low KLRB1 levels were associated with older age, advanced disease stage, HER2-positivity, poor prognosis, and a decreased survival probability compared to the high-expression group. Increased KLRB1 expression levels were correlated with inhibition of breast cancer cell proliferation, migration, and invasion, as well as promotion of cell apoptosis, possible through regulation of the NF-κB, PI3K/AKT, and TNF signaling pathways. Moreover, the study also indicated that decreased KLRB1 expression correlated with tumor purity, immune score, and immune cell infiltration (B cells, CD8+ T cells, CD4+ T cells, neutrophils, dendritic cells, among others), cell markers, and immunotherapy. CONCLUSION: Decreased KLRB1 expression in BRCA is associated with poor prognosis and immune microenvironment. This study also highlights KLRB1 as a potential molecular marker for poor prognosis in BRCA patients, and therefore, it may provide clinical implications for the management of patients with BRCA.


Subject(s)
Breast Neoplasms , Carcinoma , Humans , Female , CD8-Positive T-Lymphocytes , Phosphatidylinositol 3-Kinases , Breast Neoplasms/genetics , Prognosis , Biomarkers, Tumor/genetics , Tumor Microenvironment/genetics , NK Cell Lectin-Like Receptor Subfamily B
5.
Cell Signal ; 109: 110783, 2023 09.
Article in English | MEDLINE | ID: mdl-37356602

ABSTRACT

BACKGROUND: Acute allograft rejection remains a major obstacle after heart transplantation, and CD4+ T cells play a crucial role in allograft rejection. Upregulation of Nr4A1 could regulate CD4+ T-cell function and alleviate allograft rejection. However, the regulatory mechanism of Nr4A1 in allograft rejection remains elusive. METHODS: BALB/c mouse hearts were transplanted into WT C57BL/6 mice, and dynamic detection of the changes in Nr4A1 expression revealed that Nr4A1 was regulated posttranscriptionally after heart transplantation. Potential upstream miRNAs of Nr4A1 were screened, and the transfection of cells with these miRNA mimics/inhibitors and dual-luciferase reporter experiments were performed to clarify the regulatory mechanism of miRNAs on Nr4A1 expression. The miRNA agomiR/antagomiR was applied in vivo to validate the role of the corresponding miRNA in heart transplantation. Finally, Nr4A1 knockout mice and an adoptive T-cell cotransfer model were used to confirm the specific effects of miRNA. RESULTS: The expression of Nr4A1 protein (rather than mRNA) exhibited a trend of initially increasing and then decreasing rapidly, and this phenomenon could not be reversed by lysosomal or proteasomal inhibitors. The miRNA let-7a directly binds to the Nr4A1 3'UTR and posttranscriptionally regulates Nr4A1 expression. The let-7a antagomiR prolonged allograft survival and regulated CD4+ T-cell function by upregulating Nr4A1 protein expression in CD4+ T cells. CONCLUSIONS: This study confirmed that let-7a is a potential target for interfering with Nr4A1 expression in CD4+ T cells and preventing the pathological progression of cardiac allograft rejection.


Subject(s)
MicroRNAs , Mice , Animals , MicroRNAs/metabolism , Antagomirs , Mice, Inbred C57BL , CD4-Positive T-Lymphocytes/metabolism , Allografts/metabolism , Mice, Inbred BALB C
6.
Front Oncol ; 12: 960866, 2022.
Article in English | MEDLINE | ID: mdl-36276113

ABSTRACT

The roles and mechanisms of T-cell receptor (TCR)-associated transmembrane adaptor 1 (TRAT1) in lung adenocarcinoma (LAC) have not yet been reported in the relevant literature. Therefore, this study aimed to understand the roles and mechanisms of TRAT1 in LAC using bioinformatics and in vitro experiments. TRAT1 expression levels in LAC samples were analysed using various databases. TRAT1 co-expressed genes were acquired by the correlation analysis of LAC tissues. The functional mechanisms and protein network of TRAT1 co-expressed genes were analysed using bioinformatics analysis. The expression of TRAT1 was activated in LAC cells, and the roles of TRAT1 overexpression in the growth and migration of cancer cells was investigated using flow cytometry, Cell Counting Kit-8 (CCK-8), and migration and invasion assays. The relationship between TRAT1 overexpression, the immune microenvironment, and RNA modification was evaluated using correlation analysis. TRAT1 expression levels were significantly abnormal at multiple mutation sites and were related to the prognosis of LAC. TRAT1 co-expressed genes were involved in cell proliferation, adhesion, and differentiation, and TRAT1 overexpression significantly inhibited cell viability, migration, and invasion and promoted apoptosis of A549 and H1299 cells, which might be related to the TCR, B cell receptor (BCR), MAPK, and other pathways. TRAT1 expression levels were significantly correlated with the ESTIMATE, immune, and stromal scores in the LAC microenvironment. Additionally, TRAT1 expression levels were significantly correlated with the populations of B cells, CD8 T cells, cytotoxic cells, and other immune cells. TRAT1 overexpression was significantly correlated with the expression of immune cell markers (such as PDCD1, CD2, CD3E) and genes involved in RNA modification (such as ALKBH1, ALKBH3, ALKBH5). In conclusions, TRAT1 overexpression inhibited the growth and migration of LAC cells, thereby delaying cancer progression, and was correlated with the LAC microenvironment and RNA modifications.

7.
Front Cardiovasc Med ; 9: 972449, 2022.
Article in English | MEDLINE | ID: mdl-35990967

ABSTRACT

Background: Hypoxemia is a very common issue in patients undergoing heart valve surgery (HVS), related to poor clinical outcomes. However, studies on severe hypoxemia (SH) after HVS have not been reported. The aims of this study were to identify predictors for SH in patients undergoing HVS and to develop and validate a risk prediction model. Methods: Patients undergoing HVS between 2016 and 2019 in a cardiovascular center were enrolled and were assigned to training and validation sets by a 7:3 ratio. Based on whether patients developed SH, they were divided into two groups. By univariate and multivariate analysis, predictors for SH were identified. Based on the predictors and logistic rule, a nomogram and a risk calculator were generated. The model was evaluated using calibration, discrimination and clinical utility. Results: The incidence rates of SH, moderate hypoxemia and mild hypoxemia were respectively 2.4, 23.9, and 58.2%. By multivariate analysis, seven independent risk factors for SH after HVS were identified, including body mass index, chronic obstructive pulmonary disease, renal insufficiency, white blood cell count, serum globulin, cardiopulmonary bypass time, and surgical types. The logistic model demonstrated satisfactory discrimination, calibration and clinical utility in both the training and validation sets. A nomogram and a risk calculator based on the logistic model were generated for easy application. Risk stratification was performed and three risk intervals were defined according to the nomogram and clinical practice. In addition, compared to patients without SH, patients with SH had significantly poorer clinical outcomes. Conclusions: Postoperative hypoxemia was prevalent after HVS, related to poor clinical outcomes. A logistic model including seven independent predictors for SH after HVS were established and validated, which demonstrated satisfactory discrimination, calibration and clinical utility. The results of this study may provide help to individualized risk assessment, early prevention and perioperative management.

8.
Front Genet ; 13: 823075, 2022.
Article in English | MEDLINE | ID: mdl-35281822

ABSTRACT

The tumor microenvironment (TME) plays an important regulatory role in the progression of non-small cell lung cancer (NSCLC). Mesenchymal stem cells (MSCs) in the TME might contribute to the occurrence and development of cancer. This study evaluates the role of differentially expressed genes (DEGs) of MSCs and the development of NSCLC and develops a prognostic risk model to assess the therapeutic responses. The DEGs in MSCs from lung tissues and from normal tissues were analyzed using GEO2R. The functions and mechanisms of the DEGs were analyzed using the Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Additionally, the Cancer Genome Atlas (TCGA) database was used to determine the expression levels of the DEGs of MSCs in the NSCLC tissues. The prognostic factors of NSCLC related to MSCs were screened by survival analysis, meta-analysis, Cox regression analysis, and a prognostic risk model and nomogram was developed. The signaling mechanisms and immune roles that risk model participate in NSCLC development were determined via Gene Set Enrichment Analysis and CIBERSORT analysis. Compared to the normal tissues, 161 DEGs were identified in the MSCs of the lung tissues. These DEGs were associated with mechanisms, such as DNA replication, nuclear division, and homologous recombination. The overexpression of DDIT4, IL6, ITGA11, MME, MSX2, POSTN, and TRPA1 were associated with dismal prognosis of NSCLC patients. A high-risk score based on the prognostic risk model indicated the dismal prognosis of NSCLC patients. The nomogram showed that the age, clinical stage, and risk score affected the prognosis of NSCLC patients. Further, the high-risk model was associated with signaling mechanisms, such as the ECM-receptor interaction pathways, cytokine-cytokine receptor interaction, and MAPK pathways, involved in the progression of NSCLC and was also related to the components of the immune system, such as macrophages M0, T follicular helper cells, regulatory T cells. Therefore, the risk model and nomogram that was constructed on the basis of MSC-related factors such as POSTN, TRPA1, and DDIT4 could facilitate the discovery of target molecules that participate in the progression of NSCLC, which might also serve as new candidate markers for evaluating the prognosis of NSCLC patients.

9.
Int Immunopharmacol ; 104: 108521, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35026656

ABSTRACT

CD4+T cell-mediated acute rejection remains a major factor that affects the early survival of transplanted organs post-transplantation. Here, we reveal that nuclear receptor subfamily 4 Group A member 1 (Nr4A1) was upregulated during cardiac allograft rejection and that the increased Nr4A1 was primarily localized in intragraft-infiltrating CD4+T cells. Nr4A1 acts as a transcription factor with an important role in CD4+T cell apoptosis, differentiation and T cell dysfunction, which indicates that Nr4A1 may play a critical role in transplant rejection. Cytosporone B (Csn-B) is a naturally occurring agonist of Nr4A1, and the role of Csn-B in the physiological process of cardiac rejection is poorly defined. This study constructed an acute rejection model of abdominal heterotopic cardiac transplantation in mice and investigated whether Csn-B could attenuate acute transplant rejection by modulating the CD4+T lymphocyte response. The results showed that Csn-B prolonged murine cardiac allograft survival and reduced inflammation in allografts. Subsequently, it was confirmed that Csn-B functions by inducing non-Treg apoptosis and promoting Treg cell differentiation. Finally, we also confirmed that Csn-B attenuates acute rejection by directly targeting Nr4A1 in CD4+T cells. Our data suggest that Csn-B is a promising novel therapeutic approach for acute cardiac allograft rejection.


Subject(s)
Graft Rejection/prevention & control , Heart Transplantation , Nuclear Receptor Subfamily 4, Group A, Member 1/agonists , Phenylacetates/therapeutic use , Allografts/immunology , Animals , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cytokines/blood , Cytokines/genetics , Graft Rejection/blood , Graft Rejection/genetics , Graft Rejection/immunology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Phenylacetates/pharmacology
10.
Int J Antimicrob Agents ; 56(3): 106114, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32712334

ABSTRACT

The aim of this study was to compare ribavirin therapy versus supportive therapy only for patients with severe coronavirus disease 2019 (COVID-19). A total of 115 patients with laboratory-confirmed COVID-19 were retrospectively analysed. All patients received supportive care as well as regular laboratory and clinical monitoring. The 115 patients comprised 44 patients who received intravenous ribavirin (treatment group) and 71 who did not (control group). Baseline laboratory and clinical characteristics were similar between the two groups. The negative conversion time for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RT-PCR in the ribavirin group was 12.8 ± 4.1 days compared with 14.1 ± 3.5 days in the control group (P = 0.314). Moreover, 7/41 patients (17.1%) in the ribavirin group died compared with 17/69 (24.6%) in the control group (P = 0.475). Adverse effects were similar between the two groups. In conclusion, in patients with severe COVID-19, ribavirin therapy is not associated with improved negative conversion time for SARS-CoV-2 test and is not associated with an improved mortality rate. Further assessment in designed randomised controlled trials is recommended.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Ribavirin/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Aged , Betacoronavirus/immunology , COVID-19 , Coronavirus Infections/mortality , Coronavirus Infections/virology , Female , Humans , Immunoglobulins/therapeutic use , Male , Middle Aged , Pandemics , Patient Discharge , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , Retrospective Studies , SARS-CoV-2 , Severity of Illness Index , Survival Analysis , Time Factors , Treatment Outcome
11.
J Nanosci Nanotechnol ; 20(8): 5260-5266, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32126728

ABSTRACT

Syngas conversion to higher alcohols remains a very attractive alternative due to the abundance of syngas feedstock, such as renewable carbon and waste-carbon resources. Catalysts suitable for syngas conversion still show low selectivity to alcohols. In this article, we present nanostructured NiMoS2 and CoMoS2 catalysts supported on activated carbon pellets and design strategies to improve its selectivity towards higher alcohols. Activated carbon pellets were treated with concentrated HNO3 to enlarge porous channels and enable better dispersion of NiMoS2 and CoMoS2. These treatment steps lead to a formation of nanostructured NiMoS2 and CoMoS2 catalysts and promoted higher selectivity to ethanol, propanol and butanol. BET surface area of 532 m² g-1 was obtained for NiMoS2/Carbon catalysts from the nitrogen physisorption analysis. In catalytic tests, the highest CO conversion (39.1%) was achieved by the NiMoS2/Carbon, whereas the CoMoS2/Carbon showed the highest alcohol selectivity (74.4%). CoMoS2 catalysts supported on activated carbon pellets proved to be highly active towards undesired by-product "filamentous carbon."

12.
Transplantation ; 104(6): e151-e163, 2020 06.
Article in English | MEDLINE | ID: mdl-32108749

ABSTRACT

BACKGROUND: Obliterative bronchiolitis (OB) remains the major complication limiting long-term survival of patients after lung transplantation. We aimed to explore the effects of the selective NACHT, LRR, and PYD domains-containing protein 3 (Nlrp3) inflammasome inhibitor MCC950 on the pathogenesis of OB. METHODS: Mouse orthotopic tracheal transplants were performed to mimic OB. MCC950 (50 mg/kg) or saline was intraperitoneally injected daily. The luminal occlusion rate and collagen deposition were evaluated by hematoxylin and eosin and Masson's trichrome staining, respectively. Infiltration of CD4+, CD8+ T cells, and neutrophils was detected with immunohistochemical staining. The frequencies of T helper 1 cell (Th1), T helper 17 cell (Th17), and regulatory T cells (Treg) were measured by flow cytometry. Cytokine levels were measured by ELISA kits. RESULTS: MCC950 treatment significantly inhibited Nlrp3 inflammasome activation after allogeneic tracheal transplant and markedly decreased the luminal occlusion rate and collagen deposition in the allograft. The numbers of infiltrating CD4+, CD8+ T cells, and neutrophils in the allograft were also significantly reduced by MCC950 treatment. MCC950 dramatically decreased the frequencies of Th1/Th17 cells and the levels of interferon gamma/interleukin (IL)-17A and increased the Treg cell frequencies and IL-10 level; however, these effects were abolished by the addition of IL-1ß and IL-18 both in vitro and in vivo. OB was also rescued by the addition of IL-1ß and/or IL-18. CONCLUSIONS: Blocking Nlrp3 inflammasome activation with MCC950 ameliorates OB lesions. The mechanistic analysis showed that MCC950 regulated the balance of Th1/Th17 and Treg cells and that this process is partially mediated by inhibition of IL-1ß and IL-18. Therefore, targeting the Nlrp3 inflammasome is a promising strategy for controlling OB after lung transplantation.


Subject(s)
Bronchiolitis Obliterans/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammasomes/antagonists & inhibitors , Lung Transplantation/adverse effects , Postoperative Complications/drug therapy , Sulfones/pharmacology , Animals , Bronchiolitis Obliterans/immunology , Disease Models, Animal , Furans , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Indenes , Inflammasomes/immunology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Postoperative Complications/immunology , Sulfonamides , Sulfones/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Trachea/transplantation
13.
Ann Transl Med ; 8(23): 1582, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33437781

ABSTRACT

BACKGROUND: Transmethylation reactions play an important role on lymphocyte activation and function. S-adenosyl-L-homocysteine hydrolase (SAHH) inhibitors prevent the feedback of transmethylation reactions by S-adenosyl-L-homocysteine (SAH) accumulation, a competitive antagonist of S-adenosylmethionine (SAM)-dependent methyltransferases. However, the role of SAH in solid organ transplantation is currently unclear. METHODS: A murine model of cardiac transplantation (BALB/C to C57B/6) was established to assess allograft survival, histology, and T cell infiltration. The reversible SAHH inhibitor, DZ2002, and irreversible SAHH inhibitor, adenosine dialdehyde (AdOx), were used to assess their immunosuppressive effects in murine cardiac transplantation, compared with mice with DMSO. RESULTS: Both SAHH inhibitors prolonged the survival of cardiac allografts and alleviated alloimmune response. Notably, AdOx and DZ2002 both eliminated frequencies of Th1 and Th17 in CD4+ T cells in cardiac transplantation, and reduced the frequency of active CD4+ T cell (CD44+ CD62L-). The irreversible SAHH inhibitor facilitated the differentiation of regulatory T cells (Tregs) and increased Bim expression. Furthermore, both SAHH inhibitors alleviated infiltration of CD4+ T cells in cardiac allografts. CONCLUSIONS: The SAHH inhibitors (AdOx and DZ2002) alleviates allograft rejection in cardiac transplantation by inhibition of CD4+ T alloimmune response. SAHH inhibitors, especially DZ2002, is a promising complementary therapeutic agent in organ transplantation.

14.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-819335

ABSTRACT

@#Since December 2019, a novel coronavirus (2019-nCoV, SARS-CoV-2) pneumonia (COVID-19) outbreak has occurred in Wuhan, Hubei Province, and the epidemic situation has continued to spread. Such cases have also been found in other parts of the country. The spread of the novel coronavirus pneumonia epidemic has brought great challenges to the clinical practice of thoracic surgery. Outpatient clinics need to strengthen the differential diagnosis of ground glass opacity and pulmonary plaque shadows. During the epidemic, surgical indications are strictly controlled, and selective surgery is postponed. Patients planning to undergo a limited period of surgery should be quarantined for 2 weeks and have a nucleic acid test when necessary before surgery. For patients who are planning to undergo emergency surgery, nucleic acid testing should be carried out before surgery, and three-level protection should be performed during surgery. Patients who are planning to undergo emergency surgery in the epidemic area should be confirmed with or without novel coronavirus pneumonia before operation, and perform nucleic acid test if necessary. Surgical disinfection and isolation measures should be strictly carried out. Among postoperative patients, cases with new coronavirus infection were actively investigated. For the rescue of patients with novel coronavirus infection, attention needs to be paid to prevention and treatment and related complications, including mechanical ventilation-related pneumothorax or mediastinal emphysema, and injury after tracheal intubation.

15.
Cell Rep ; 28(4): 966-978.e4, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31340157

ABSTRACT

Platelet-derived growth factor receptor (PDGFR) signaling is involved in proliferation and survival in a wide array of cell types. The role of PDGFR signaling in heart regeneration is still unknown. We find that PDGFR-ß signaling decreases in myocardium with age and that conditional activation PDGFR-ß in cardiomyocytes promotes heart regeneration. Employing RNA sequencing, we show that the enhancer of zeste homolog 2 (Ezh2) can be upregulated by PDGFR-ß signaling in primary cardiomyocytes. Conditional knockout of Ezh2 blocks cardiomyocyte proliferation and H3K27me3 modification during neonatal heart regeneration with Ink4a/Arf upregulation, even in mice with myocyte-specific conditional activation of PDGFR-ß. We also show that PDGFR-ß controls EZH2 expression via the phosphatidylinositol 3-kinase (PI3K)/p-Akt pathway in cardiomyocytes. Gene therapy with adeno-associated virus serotype 9 (AAV9) encoding activated PDGFR-ß enhances adult heart regeneration and systolic function. Our data demonstrate that the PDGFR-ß/EZH2 pathway is critical for promoting cardiomyocyte proliferation and heart regeneration, providing a potential target for cardiac repair.


Subject(s)
Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Regeneration , Signal Transduction , Animals , Animals, Newborn , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/metabolism , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
16.
J Mol Cell Cardiol ; 127: 105-114, 2019 02.
Article in English | MEDLINE | ID: mdl-30553885

ABSTRACT

Long noncoding RNAs (lncRNAs) play important roles in the regulation of genes involved in cell proliferation. We have previously sought to more globally understand the differences of lncRNA expression between human fetal heart and adult heart to identify some functional lncRNAs which involve in the process of heart repair. We found that a highly conserved long noncoding RNA NR_045363 was mainly expressed in cardiomyocytes and rarely in non-cardiomyocytes. NR_045363 overexpression in 7-day-old mice heart could improve cardiac function and stimulate cardiomyocyte proliferation after myocardial infarction. Furthermore, NR_045363 knockdown inhibited proliferation of primary embryonic cardiomyocytes, while NR_045363 overexpression enhanced DNA synthesis and cytokinesis in neonatal cardiomyocytes in vitro. Mechanistic analysis revealed that NR_045363 promoted cardiomyocyte proliferation through interaction with miR-216a, which regulated the JAK2-STAT3 pathway. Our results showed that NR_045363 is a potent lncRNA modulator essential for cardiomyocyte proliferation.


Subject(s)
Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/metabolism , Wound Healing , Animals , Animals, Newborn , Base Sequence , Cell Proliferation , Conserved Sequence , Heart/embryology , Humans , Janus Kinases/metabolism , Mice , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Regeneration , STAT3 Transcription Factor/metabolism , Signal Transduction
17.
J Am Heart Assoc ; 8(1): e011211, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30587058

ABSTRACT

Background The arteriovenous fistula ( AVF ) is the preferred hemodialysis access for patients with chronic kidney disease. Chronic kidney disease can increase neointima formation, which greatly contributes to AVF failure by an unknown mechanism. Our study aimed to determine the role of nucleotide-binding oligomerization domain-like receptor protein 3 ( NLRP 3) in neointima formation induced by experimental AVF s in the presence of chronic kidney disease. Methods and Results From our findings, NLRP 3 was upregulated in the intimal lesions of AVF s in both uremic mice and patients. Smooth muscle-specific knockout NLRP 3 mice exhibited markedly decreased neointima formation in the outflow vein of AVF s. Compared with primary vascular smooth muscle cells isolated from control mice, those isolated from smooth muscle-specific knockout NLRP 3 mice showed compromised proliferation, migration, phenotypic switching, and a weakened ability to activate mononuclear macrophages. To identify how NLRP 3 functions, several small-molecule inhibitors were used. The results showed that NLRP 3 regulates smooth muscle cell proliferation and migration through Smad2/3 phosphorylation rather than through caspase-1/interleukin-1 signaling. Unexpectedly, the selective NLRP 3-inflammasome inhibitor MCC 950 also repressed Smad2/3 phosphorylation and relieved chronic kidney disease-promoted AVF failure independent of macrophages. Conclusions Our findings suggest that NLRP 3 in vascular smooth muscle cells may play a crucial role in uremia-associated AVF failure and may be a promising therapeutic target for the treatment of AVF failure.


Subject(s)
Arteriovenous Shunt, Surgical/methods , Gene Expression Regulation , Myocytes, Smooth Muscle/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Renal Dialysis/methods , Renal Insufficiency, Chronic/blood , Animals , Blotting, Western , Cells, Cultured , DNA/genetics , Humans , Inflammasomes , Jugular Veins/metabolism , Jugular Veins/pathology , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Renal Insufficiency, Chronic/therapy , Signal Transduction , Treatment Failure
18.
J Am Heart Assoc ; 7(18): e008604, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30371220

ABSTRACT

Background Cardiac hypertrophy has been recognized as an important independent risk factor for the development of heart failure and increases the risk of cardiac morbidity and mortality. A disintegrin and metalloprotease 23 (ADAM23), a member of ADAM family, is involved in cancer and neuronal differentiation. Although ADAM23 is expressed in the heart, the role of ADAM23 in the heart and in cardiac diseases remains unknown. Methods and Results We observed that ADAM23 expression is decreased in both failing human hearts and hypertrophic mice hearts. Cardiac-specific conditional ADAM23-knockout mice significantly exhibited exacerbated cardiac hypertrophy, fibrosis, and dysfunction, whereas transgenic mice overexpressing ADAM23 in the heart exhibited reduced cardiac hypertrophy in response to pressure overload. Consistent results were also observed in angiotensin II -induced neonatal rat cardiomyocyte hypertrophy. Mechanistically, ADAM23 exerts anti-hypertrophic effects by specifically targeting the focal adhesion kinase-protein kinase B (FAK-AKT) signaling cascade. Focal adhesion kinase inactivation by inhibitor ( PF -562271) greatly reversed the detrimental effects in ADAM23-knockout mice subjected to aortic banding. Conclusion Altogether, we identified ADAM23 as a negative regulator of cardiac hypertrophy through inhibiting focal adhesion kinase-protein kinase B signaling pathway, which could be a promising therapeutic target for this malady.


Subject(s)
ADAM Proteins/genetics , Cardiomegaly/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/genetics , ADAM Proteins/biosynthesis , Animals , Blotting, Western , Cardiomegaly/diagnosis , Cardiomegaly/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/biosynthesis , RNA/genetics , Signal Transduction
19.
Arterioscler Thromb Vasc Biol ; 38(5): 1086-1101, 2018 05.
Article in English | MEDLINE | ID: mdl-29519942

ABSTRACT

OBJECTIVE: Thoracic aortic aneurysm and dissection (TAAD) are severe vascular conditions. Dysfunctional transforming growth factor-ß (TGF-ß) signaling in vascular smooth muscle cells and elevated angiotensin II (AngII) levels are implicated in the development of TAAD. In this study, we investigated whether these 2 factors lead to TAAD in a mouse model and explored the possibility of using microRNA-21 (miR-21) for the treatment of TAAD. APPROACH AND RESULTS: TAAD was developed in Smad3 (mothers against decapentaplegic homolog 3) heterozygous (S3+/-) mice infused with AngII. We found that p-ERK (phosphorylated extracellular regulated protein kinases)- and p-JNK (phosphorylated c-Jun N-terminal kinase)-associated miR-21 was higher in TAAD lesions. We hypothesize that downregulation of miR-21 mitigate TAAD formation. However, Smad3+/-:miR-21-/- (S3+/-21-/-) mice exhibited conspicuous TAAD formation after AngII infusion. The vascular wall was dilated, and aortic rupture occurred within 23 days during AngII infusion. We then examined canonical and noncanonical TGF-ß signaling and found that miR-21 knockout in S3+/- mice increased SMAD7 and suppressed canonical TGF-ß signaling. Vascular smooth muscle cells lacking TGF-ß signals tended to switch from a contractile to a synthetic phenotype. The silencing of Smad7 with lentivirus prevented AngII-induced TAAD formation in S3+/-21-/- mice. CONCLUSIONS: Our study demonstrated that miR-21 knockout exacerbated AngII-induced TAAD formation in mice, which was associated with TGF-ß signaling dysfunction. Therapeutic strategies targeting TAAD should consider unexpected side effects associated with alterations in TGF-ß signaling.


Subject(s)
Angiotensin II , Aorta, Thoracic/metabolism , Aortic Aneurysm, Thoracic/prevention & control , Aortic Dissection/prevention & control , Aortic Rupture/prevention & control , MicroRNAs/metabolism , Smad3 Protein/deficiency , Transforming Growth Factor beta/metabolism , Aortic Dissection/chemically induced , Aortic Dissection/genetics , Aortic Dissection/metabolism , Animals , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/chemically induced , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Rupture/chemically induced , Aortic Rupture/genetics , Aortic Rupture/metabolism , Cells, Cultured , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Extracellular Signal-Regulated MAP Kinases , Genetic Predisposition to Disease , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Phosphorylation , Signal Transduction , Smad3 Protein/genetics , Smad7 Protein/genetics , Smad7 Protein/metabolism
20.
Sci Rep ; 7(1): 15799, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29150694

ABSTRACT

Graft arteriosclerosis (GA) is the leading cause of late cardiac allograft dysfunction. The innate immune system plays a major role in GA, paprticularly Toll-like receptor 4 (TLR4) signaling. Here we characterized the role of TLR4 and its antagonist TAK-242 in a mouse model of GA. BALB/c (H-2d) donor aortas were transplanted into C57BL/6 (H-2b) recipients, and the mice received intraperitoneal injection of 3 or 10 mg/kg of TAK-242 or vehicle every other day for 1, 2, 4, 6, 8 and 12 weeks. With TAK-242 administration, intimal hyperplasia initially appeared at 2 weeks after transplantation, and TAK-242 postponed the progression of neointimal formation in allogeneic aortic grafts. TAK-242 treatment reduced CD68+ macrophage accumulation in the allografts, reduced the levels of ly-6Chi monocytes in peripheral blood, bone marrow and spleen, and downregulated proinflammatory cytokine and chemokine levels. Ex vivo we observed that TAK-242 could improve the graft microenvironment by interfering the Tck/Mφ IL12p70 and IFNγ axis, reducing CCL2-mediated migration of vascular smooth cells.


Subject(s)
Aorta/transplantation , Neointima/drug therapy , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Ly/metabolism , Aorta/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Movement/drug effects , Chemokine CCL2/pharmacology , Chemokines/metabolism , Down-Regulation/drug effects , Hyperplasia , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Neointima/blood , Neointima/pathology , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism , Sulfonamides/pharmacology , Toll-Like Receptor 4/metabolism , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...