Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
2.
Cell Death Discov ; 9(1): 218, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37393392

ABSTRACT

The impact of Tim-3 (T cell immunoglobulin and mucin domain-containing protein 3) on cisplatin-induced acute kidney injury was investigated in this study. Cisplatin-induced Tim-3 expression in mice kidney tissues and proximal tubule-derived BUMPT cells in a time-dependent manner. Compared with wild-type mice, Tim-3 knockout mice have higher levels of serum creatinine and urea nitrogen, enhanced TUNEL staining signals, more severe 8-OHdG (8-hydroxy-2' -deoxyguanosine) accumulation, and increased cleavage of caspase 3. The purified soluble Tim-3 (sTim-3) protein was used to intervene in cisplatin-stimulated BUMPT cells by competitively binding to the Tim-3 ligand. sTim-3 obviously increased the cisplatin-induced cell apoptosis. Under cisplatin treatment conditions, Tim-3 knockout or sTim-3 promoted the expression of TNF-α (tumor necrosis factor-alpha) and IL-1ß (Interleukin-1 beta) and inhibited the expression of IL-10 (interleukin-10). NF-κB (nuclear factor kappa light chain enhancer of activated B cells) P65 inhibitor PDTC or TPCA1 lowed the increased levels of creatinine and BUN (blood urea nitrogen) in cisplatin-treated Tim-3 knockout mice serum and the increased cleavage of caspase 3 in sTim-3 and cisplatin-treated BUMPT cells. Moreover, sTim-3 enhanced mitochondrial oxidative stress in cisplatin-induced BUMPT cells, which can be mitigated by PDTC. These data indicate that Tim-3 may protect against renal injury by inhibiting NF-κB-mediated inflammation and oxidative stress.

3.
Heart Vessels ; 38(3): 448-458, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36344842

ABSTRACT

Acute myocardial infarction (AMI) is associated with high morbidity and mortality. An effective therapeutic strategy is to rescue cardiomyocytes from death. Apoptosis is a key reason of cardiomyocyte death that can be prevented. In this study, we investigated the role of TNF-related apoptosis-inducing ligand (TRAIL) in initiating apoptosis by binding to death receptor 5 (DR5), and this procession is inhibited by soluble DR5 (sDR5) in rats after AMI. First, we found that the level of TRAIL in serum was down-regulated in AMI patients. Then, TRAIL and DR5 expression was analysed in the myocardium of rats after AMI, and their expression was up-regulated. sDR5 treatment reduced the myocardial infarct size and the levels of CK-MB and cTn-I in serum. The expression of caspase 3 and PARP is decreased, but the anti-apoptotic factor Bcl-2 was increased in sDR5 treatment rats after AMI. DR5 expression was also analysed after sDR5 treatment and it was down-regulated, and a low level of DR5 expression seemed to be beneficial for the myocardium. Overall, our findings indicated that sDR5 decreases myocardial damage by inhibiting apoptosis in rat after AMI. We expect to observe the potential therapeutic effects of sDR5 on AMI in the future.


Subject(s)
Myocardial Infarction , Receptors, TNF-Related Apoptosis-Inducing Ligand , Rats , Animals , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/physiology , Myocardium/metabolism , Myocardial Infarction/prevention & control , Myocardial Infarction/metabolism
4.
Mol Immunol ; 152: 224-231, 2022 12.
Article in English | MEDLINE | ID: mdl-36379130

ABSTRACT

At present, the occurrence and development of inflammatory diseases are closely related to the abnormal changes of the content and function of many cytokines. At the same time, targeting related cytokines to prevent and treat diseases has also achieved good results. Therefore, it is very important to explore the role of various cytokines in inflammatory diseases. As an inflammation related protein, Tumor necrosis factor alpha stimulating gene-6 (TSG-6) has attracted more and more attention. In the process of disease, it's like a double-edged sword, showing different responses. It is constitutively expressed in some tissues with high metabolic activity and barrier protection. The diversity of its functions depends on the binding of TSG-6 with a variety of ligands, including matrix molecules, autoimmune regulatory factors and growth factors, participating in extracellular matrix remodeling and regulating protease network. This paper reviews the structure, biological function and research progress of TSG-6 in inflammatory diseases, in order to provide reference for drug development in the future.


Subject(s)
Cell Adhesion Molecules , Tumor Necrosis Factor-alpha , Humans , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Extracellular Matrix/metabolism , Inflammation/genetics , Inflammation/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Biology (Basel) ; 11(8)2022 Aug 09.
Article in English | MEDLINE | ID: mdl-36009821

ABSTRACT

In acute myocardial infarction (AMI), endothelial progenitor cells (EPCs) are essential for the recovery of collateral circulation via angiogenesis. Clinical research has shown that the poor prognosis of the patients with AMI is closely associated with the cell quantity and function of EPCs. Whether there are differences in the biological features of EPCs from AMI patients and healthy subjects is worth exploring. In this study, EPCs were isolated from human peripheral blood and identified as late-stage EPCs by flow cytometry, immunofluorescence, and blood vessel formation assay. Compared to healthy subjects, AMI patients had more EPCs in the peripheral blood compared to healthy subjects. In addition, EPCs from AMI patients exhibited higher migration ability in the transwell assay compared to EPCs from healthy subjects. However, no difference in the angiogenesis of EPCs was observed between AMI patients and healthy subjects. Further studies revealed that soluble vascular endothelial growth factor receptor 1 (sFlt-1) in the serum of AMI patients was involved in the inhibition of EPCs angiogenesis by suppressing the Akt and Erk pathways. In conclusion, this study demonstrated that elevated serum sFlt-1 inhibits angiogenesis of EPC in AMI patients. Our findings uncover a pathogenic role of sFlt-1 in AMI.

6.
Life Sci ; 303: 120670, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35640777

ABSTRACT

AIMS: Myocardial infarction (MI) is a leading cause of death worldwide for which there is no cure. Percutaneous coronary intervention (PCI) can restore blood supply in a timely manner, which greatly reduces the mortality of patients, but ischemia/reperfusion (I/R) injury is inevitable. A number of clinical studies have shown that gut microbiota play an essential role in cardiovascular diseases. This study aims to explore the mechanism of gut microbiota to limit I/R injury. MATERIALS AND METHODS: This study adopted the myocardial I/R model using gut microbiota clearance mice, neutrophil clearance mice and double-scavenging mice, and explored the relationship between gut microbiota and NETs during I/R injury. Neutrophils were isolated in vitro to explore the effect of NETs on myocardial cell injury and its molecular mechanism. KEY FINDINGS: Gut microbiota aggravate cardiac I/R injury via regulating the formation of NETs. The migration of gut microbiota to blood stimulated the formation of NETs after cardiac I/R. NETs, which can directly lead to apoptosis of myocardial cells and myocardial microvascular endothelial cells. The time point of NETs formation in tissue and blood after I/R were determined by experiments. SIGNIFICANCE: It was confirmed that gut microbiota participates in cardiac I/R injury by regulating the formation of NETs, which reveals a new mechanism of I/R injury and provides a new potential target for the treatment of I/R injury.


Subject(s)
Extracellular Traps , Gastrointestinal Microbiome , Percutaneous Coronary Intervention , Reperfusion Injury , Animals , Disease Models, Animal , Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Neutrophils
7.
IEEE Trans Nanobioscience ; 21(1): 37-43, 2022 01.
Article in English | MEDLINE | ID: mdl-34406945

ABSTRACT

Coronavirus disease 2019 is an infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 is highly transmissible. Early and rapid testing is necessary to effectively prevent and control the outbreak. Detection of SARS-CoV-2 antibodies with lateral flow immunoassay can achieve this goal. In this study, SARS-CoV-2 nucleoprotein (NP) was expressed and purified. We used the selenium nanoparticle as the labeling probe coupled with the NP to prepare an antibody (IgM and IgG) detection kit. The detection limit, cross reaction, sensitivity and specificity of the kit is verified. Separate detection of IgM and IgG, such as in this assay, was performed in order to reduce mutual interference and improve the accuracy of the test results.The final purity of NP was 91.83%. Selenium nanoparticle and NP successfully combined with stable effect. The LOD of the kit was 20 ng/mL for anti-NP IgG and 60 ng/mL for anti-NP IgM, respectively. The kit does not cross reaction with RF. The sensitivity of the kit was 94.74% and the specificity was 96.23%. The assay kit does not require any special device for reading the results and the readout is a simple color change that can be evaluated with the naked eye. This kit is suitable for rapid and real-time detection of the SARS-CoV-2 antibody IgG and IgM.


Subject(s)
COVID-19 , Nanoparticles , Selenium , Humans , Immunoassay , Immunoglobulin M , SARS-CoV-2 , Sensitivity and Specificity
8.
Front Microbiol ; 12: 692279, 2021.
Article in English | MEDLINE | ID: mdl-34335518

ABSTRACT

Staphylococcus aureus is a major pathogenic bacterium that causes a variety of clinical infections. The emergence of multi-drug resistant mechanisms requires novel strategies to mitigate S. aureus infection. Alpha-hemolysin (Hla) is a key virulence factor that is believed to play a significant role in the pathogenesis of S. aureus infections. In this study, we screened a naïve human Fab library for identification of monoclonal antibodies targeting Hla by phage display technology. We found that the monoclonal antibody YG1 blocked the Hla-mediated lysis of rabbit red blood cells and inhibited Hla binding to A549 cells in a concentration-dependent manner. YG1 also provided protection against acute peritoneal infection, bacteremia, and pneumonia in murine models. We further characterized its epitope using different Hla variants and found that the amino acids N209 and F210 of Hla were functionally and structurally important for YG1 binding. Overall, these results indicated that targeting Hla with YG1 could serve as a promising protective strategy against S. aureus infection.

9.
Front Immunol ; 12: 656090, 2021.
Article in English | MEDLINE | ID: mdl-33841441

ABSTRACT

Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with multiple autoantibody production and often affects the kidneys, known as lupus nephritis. However, the mechanism underlying lupus nephritis development is unclear. Biofilms that protect bacteria from stress are ubiquitous in almost every environment. Here, we identified that a conserved peptide (HU1) derived from DNABII proteins, one of major bacterial biofilm components, was specifically recognized by sera from about 47% patients with SLE. Moreover, the serum anti-HU1 levels showed a significant positive correlation with lupus nephritis occurrence. Presence of antibodies against HU1 in pristane-induced mice aggravated lupus nephritis, although these antibodies also attenuated bacterial biofilm formation. We further identified that antibodies against HU1 cross-recognized protein disulfide isomerase (P4HB) located on the renal cell surface and inhibited the activities of this enzyme. Our findings reveal a novel mechanism underlying the development of lupus nephritis triggered by bacterial biofilms.


Subject(s)
Autoantibodies/immunology , Bacteria/immunology , Biofilms , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/etiology , Lupus Nephritis/pathology , Amino Acid Sequence , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Autoantibodies/blood , Autoantigens/immunology , Biomarkers , Disease Models, Animal , Disease Progression , Disease Susceptibility , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/blood , Mice , Mice, Transgenic , Peptides/chemistry , Peptides/immunology , Terpenes/adverse effects
10.
Front Immunol ; 12: 654649, 2021.
Article in English | MEDLINE | ID: mdl-33868297

ABSTRACT

Extracellular traps released by neutrophils (NETs) are essential for the clearance of Pseudomonas aeruginosa. Alkaline protease (AprA) secreted by P. aeruginosa negatively correlates with clinical improvement. Moreover, anti-AprA in patients with cystic fibrosis (CF) can help identify patients with aggressive forms of chronic infection. However, the mechanism underlying the clinical outcomes remains unclear. We demonstrated that aprA deficiency in P. aeruginosa decreased the bacterial burden and reduced lung infection. AprA degraded NET components in vitro and in vivo but did not affect NET formation. Importantly, antibodies induced by AprA acted as an agonist and directly enhanced the degrading activities of AprA. Moreover, antisera from patients with P. aeruginosa infection exhibited antibody-dependent enhancement (ADE) similar to that of the antibodies we prepared. Our further investigations showed that the interaction between AprA and the specific antibodies might make the enzyme active sites better exposed, and subsequently enhance the recognition of substrates and accelerate the degradation. Our findings revealed that AprA secreted by P. aeruginosa may aggravate infection by destroying formed NETs, an effect that was further enhanced by its antibodies.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Endopeptidases/immunology , Extracellular Traps/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Animals , Antibody-Dependent Cell Cytotoxicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Disease Models, Animal , Endopeptidases/genetics , Endopeptidases/metabolism , Extracellular Traps/enzymology , Female , Host-Pathogen Interactions/immunology , Humans , Mice , Neutrophils/immunology , Neutrophils/metabolism , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/pathogenicity
11.
Int J Nanomedicine ; 16: 2059-2070, 2021.
Article in English | MEDLINE | ID: mdl-33727813

ABSTRACT

PURPOSE: The purpose of this study was to establish a lateral flow immunoassay using selenium nanoparticles (Se-NPs) as a probe to detect ractopamine (RAC) and salbutamol (SAL) in swine urine. METHODS: SDS and PEG were used as templates to prepare Se-NPs; anti-RAC monoclonal antibodies or anti-SAL monoclonal antibodies were labelled with Se-NPs; and rapid detection kits were prepared. The sensitivity, specificity, and stability were measured, and actual samples were analysed. RESULTS: The Se-NPs were spherical with a diameter of 40.63 ± 5.91 nm, and were conjugated successfully with an anti-RAC antibody to give a total diameter of 82.33 ± 17.91 nm. The detection limit of a RAC kit in swine urine was 1 ng/mL, and that of a SAL kit was 3 ng/mL. Both procedures could be completed within 5 minutes. No cross-reaction occurred with clenbuterol, bambuterol and phenylethanolamine A. Samples were tested consistently across different batches of kits for swine urine. The results of the kits were identical to those of actual clinical samples analysed by ELISA, and the coincidence rate was 100%. CONCLUSION: The assay kit does not require any special device for reading the results, and the readout is a simple colour change that can be evaluated with the naked eye. It is easy to operate, sensitive, specific, and stable This kit is suitable for the rapid and real-time detection of RAC and SAL residues in swine urine samples. CLINICAL TRIAL REGISTRATION: Swine urines samples were used under approval from the Experimental Animal Ethics committee of the Joint National Laboratory for Antibody Drug Engineering, Henan University.


Subject(s)
Albuterol/urine , Chromatography, Affinity/methods , Metal Nanoparticles/chemistry , Phenethylamines/urine , Selenium/chemistry , Animals , Antibodies/metabolism , Hydrogen-Ion Concentration , Swine
12.
Medicine (Baltimore) ; 100(3): e24273, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33546048

ABSTRACT

ABSTRACT: The exact dose of cytarabine still remain controversial for the management of patients with acute myeloid leukemia (AML) after complete remission (CR), but recent studies favor lower doses. This study aimed to investigate the toxic effects of single-intermediate dose (ID) cytarabine in patients with AML after achieving CR, compared with standard-dose cytarabine.In this retrospective study, AML patients who achieved CR after consolidation therapy before enrollment between 07/2008 and 05/2019 were included. All patients were divided into single-ID cytarabine and standard-dose cytarabine. The Kaplan-Meier method was used to compare overall survival (OS) and relapse-free time (RFS). Cox regression models were used to assess factors independently associated with OS and RFS. The toxic side effects of hematology and non-hematology were observed.52 patients were enrolled. There were 33 in ID group, 19 in Standard dose group. The 3-year RFS rate (40.4% vs 22.2%, P = .031) was better in the ID group than in the standard-dose group, while the 3-year OS rate was not different between the 2 groups (50.2% vs 27.8%, P = .074). Treatment stratage of ID cytarabine chemotherapy significantly improve the prognosis of AML regardless of patient age, risk grade, WBC count. There were no significant differences between the 2 groups in grade 3 to 4 bone marrow suppression, gastrointestinal symptoms, blood transfusion, infections.Patients with AML receiving ID cytarabine showed better survival and similar toxicity profiles compared with patients who received standard-dose cytarabine.


Subject(s)
Consolidation Chemotherapy/standards , Cytarabine/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Remission Induction/methods , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Consolidation Chemotherapy/methods , Consolidation Chemotherapy/statistics & numerical data , Cytarabine/pharmacology , Cytarabine/therapeutic use , Female , Humans , Male , Middle Aged , Prognosis , Proportional Hazards Models , Retrospective Studies , Survival Analysis
13.
J Cell Mol Med ; 25(6): 2861-2871, 2021 03.
Article in English | MEDLINE | ID: mdl-33512774

ABSTRACT

Cisplatin nephrotoxicity has been considered as serious side effect caused by cisplatin-based chemotherapy. Recent evidence indicates that renal tubular cell apoptosis and inflammation contribute to the progression of cisplatin-induced acute kidney injury (AKI). Hepatocyte nuclear factor 1ß (HNF1ß) has been reported to regulate the development of kidney cystogenesis, diabetic nephrotoxicity, etc However, the regulatory mechanism of HNF1ß in cisplatin nephrotoxicity is largely unknown. In the present study, we examined the effects of HNF1ß deficiency on the development of cisplatin-induced AKI in vitro and in vivo. HNF1ß down-regulation exacerbated cisplatin-induced RPTC apoptosis by indirectly inducing NF-κB p65 phosphorylation and nuclear translocation. HNF1ß knockdown C57BL/6 mice were constructed by injecting intravenously with HNF1ß-interfering shRNA and PEI. The HNF1ß scramble and knockdown mice were treated with 30 mg/kg cisplatin for 3 days to induce acute kidney injury. Cisplatin treatment caused increased caspase 3 cleavage and p65 phosphorylation, elevated serum urea nitrogen and creatinine, and obvious histological damage of kidney such as fractured tubules in control mice, which were enhanced in HNF1ß knockdown mice. These results suggest that HNF1ß may ameliorate cisplatin nephrotoxicity in vitro and in vivo, probably through regulating NF-κB signalling pathway.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Hepatocyte Nuclear Factor 1-beta/genetics , NF-kappa B/metabolism , Nephrons/drug effects , Signal Transduction/drug effects , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Disease Models, Animal , Hepatocyte Nuclear Factor 1-beta/metabolism , Kidney Tubules/drug effects , Mice , Mice, Knockout , Phosphorylation/drug effects , Rats , Transcription Factor RelA/metabolism
15.
Lab Chip ; 20(22): 4255-4261, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33064114

ABSTRACT

COVID-19 is a widespread and highly contagious disease in the human population. COVID-19 is caused by SARS-CoV-2 infection. There is still a great demand for point-of-care tests for detection, epidemic prevention and epidemiological investigation, both now and after the epidemic. We present a lateral flow immunoassay kit based on a selenium nanoparticle-modified SARS-CoV-2 nucleoprotein, which detects anti-SARS-CoV-2 IgM and anti-SARS-CoV-2 IgG in human serum, and the results can be read by the naked eye in 10 minutes. We expressed and purified the SARS-CoV-2 nucleoprotein in HEK293 cells, with a purity of 98.14% and a concentration of 5 mg mL-1. Selenium nanoparticles were synthesized by l-ascorbic acid reduction of seleninic acid at room temperature. After conjugation with the nucleoprotein, a lateral flow kit was successfully prepared. The IgM and IgG detection limits of the lateral flow kit reached 20 ng mL-1 and 5 ng mL-1, respectively, in human serum. A clinical study sample comprising 90 COVID-19-diagnosed patients and 263 non-infected controls was used to demonstrate a sensitivity and specificity of 93.33% and 97.34%, respectively, based on RT-PCR and clinical results. No cross-reactions with rheumatoid factor and positive serum for anti-nuclear antibodies, influenza A, and influenza B were observed. Moreover, the lateral flow kit remained stable after storage for 30 days at 37 °C. Our results demonstrate that the selenium nanoparticle lateral flow kit can conveniently, rapidly, and sensitively detect anti-SARS-CoV-2 IgM and IgG in human serum and blood; it can also be suitable for the epidemiological investigation of COVID-19.


Subject(s)
Antibodies, Viral/blood , Betacoronavirus/immunology , Clinical Laboratory Techniques , Coronavirus Infections/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Pneumonia, Viral/blood , Antibodies, Viral/immunology , COVID-19 , COVID-19 Testing , Coronavirus Infections/diagnosis , Coronavirus Infections/immunology , Equipment Design , HEK293 Cells , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Lab-On-A-Chip Devices , Limit of Detection , Nanoparticles/chemistry , Pandemics , Pneumonia, Viral/immunology , Point-of-Care Testing , Reagent Strips/analysis , SARS-CoV-2 , Selenium/chemistry
16.
J Cell Mol Med ; 24(15): 8849-8861, 2020 08.
Article in English | MEDLINE | ID: mdl-32558367

ABSTRACT

Uric acid crystal is known to activate the NLRP3 inflammasome and to cause tissue damages, which can result in many diseases, such as gout, chronic renal injury and myocardial damage. Meanwhile, soluble uric acid (sUA), before forming crystals, is also related to these diseases. This study was carried out to investigate whether sUA could also activate NLRP3 inflammasome in cardiomyocytes and to analyse the mechanisms. The cardiomyocyte activity was monitored, along with the levels of mature IL-1ß and caspase-1 from H9c2 cells following sUA stimulus. We found that sUA was able to activate NLRP3 inflammasome, which was responsible for H9c2 cell apoptosis induced by sUA. By elevating TLR6 levels and then activating NF-κB/p65 signal pathway, sUA promoted NLRP3, pro-caspase 1 and pro-IL-1ß production and provided the first signal of NLRP3 inflammasome activation. Meanwhile, ROS production regulated by UCP2 levels also contributed to NLRP3 inflammasome assembly and subsequent caspase 1 activation and mature IL-1ß secretion. In addition, the tlr6 knockdown rats suffering from hyperuricemia showed the lower level of IL-1ß and an ameliorative cardiac function. These findings suggest that sUA activates NLRP3 inflammasome in cardiomyocytes and they may provide one therapeutic strategy for myocardial damage induced by sUA.


Subject(s)
Inflammasomes/metabolism , Myocardium/metabolism , Myocardium/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uric Acid/blood , Animals , Apoptosis/drug effects , Cell Line , Gene Knockdown Techniques , Lysosomes/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , Toll-Like Receptor 6/metabolism , Uric Acid/pharmacology , Ventricular Remodeling/genetics
17.
Sci Transl Med ; 12(540)2020 04 22.
Article in English | MEDLINE | ID: mdl-32321866

ABSTRACT

Myocardial infarction (MI) is a leading cause of death worldwide for which there is no cure. Although cardiac cell death is a well-recognized pathological mechanism of MI, therapeutic blockade of cell death to treat MI is not straightforward. Death receptor 5 (DR5) and its ligand TRAIL [tumor necrosis factor (TNF)-related apoptosis-inducing ligand] are up-regulated in MI, but their roles in pathological remodeling are unknown. Here, we report that blocking TRAIL with a soluble DR5 immunoglobulin fusion protein diminished MI by preventing cardiac cell death and inflammation in rats, pigs, and monkeys. Mechanistically, TRAIL induced the death of cardiomyocytes and recruited and activated leukocytes, directly and indirectly causing cardiac injury. Transcriptome profiling revealed increased expression of inflammatory cytokines in infarcted heart tissue, which was markedly reduced by TRAIL blockade. Together, our findings indicate that TRAIL mediates MI directly by targeting cardiomyocytes and indirectly by affecting myeloid cells, supporting TRAIL blockade as a potential therapeutic strategy for treating MI.


Subject(s)
Myocardial Infarction , Receptors, TNF-Related Apoptosis-Inducing Ligand , Animals , Apoptosis , Cell Line, Tumor , Haplorhini , Myocardial Infarction/drug therapy , Rats , Swine , TNF-Related Apoptosis-Inducing Ligand
18.
J Leukoc Biol ; 108(1): 283-295, 2020 07.
Article in English | MEDLINE | ID: mdl-32237257

ABSTRACT

1,25-dihydroxyvitamin D3 (1,25(OH)2 D3, VitD3) is the major active ingredient of vitamin D and has anti-inflammatory activity; however, the mechanism for this remains poorly understood. In this study, we found that VitD3 was able to abolish NOD-like receptor protein 3 (NLRP3) inflammasome activation and subsequently inhibit caspase-1 activation and IL-1ß secretion via the vitamin D receptor (VDR). Furthermore, VitD3 specifically prevented NLRP3-mediated apoptosis-associated speck-like protein with a caspase-recruitment domain (ASC) oligomerization. In additional to this, NLRP3 binding to NIMA-related kinase 7 (NEK7) was also inhibited. Notably, VitD3 inhibited autophagy, leading to the inhibition of the NLRP3 inflammasome. Uncoupling protein 2-reactive oxygen species signaling may be involved in inflammasome suppression by VitD3. Importantly, VitD3 had both preventive and therapeutic effects on mouse model of ulcerative colitis, via inhibition of NLRP3 inflammasome activation. Our results reveal a mechanism through which VitD3 represses inflammation and prevents the relevant diseases, and suggest a potential clinical use of VitD3 in autoimmune syndromes or other NLRP3 inflammasome-driven inflammatory diseases.


Subject(s)
Calcitriol/therapeutic use , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Autophagy/drug effects , CARD Signaling Adaptor Proteins/metabolism , Calcitriol/pharmacology , Caspase 1/metabolism , Cell Polarity/drug effects , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/immunology , Colon/drug effects , Colon/pathology , Dextran Sulfate , Enzyme Activation/drug effects , Interleukin-1beta/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/pathology , Mice , NIMA-Related Kinases/metabolism , Proteolysis/drug effects , Reactive Oxygen Species/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Ubiquitination/drug effects , Uncoupling Protein 2/metabolism
19.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165724, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32061775

ABSTRACT

Removal of nuclei in lens fiber cells is required for organelle-free zone (OFZ) formation during lens development. Defect in degradation of nuclear DNA leads to cataract formation. DNase2ß degrades nuclear DNA of lens fiber cells during lens differentiation in mouse. Hsf4 is the principal heat shock transcription factor in lens and facilitates the lens differentiation. Knockout of Hsf4 in mouse and zebrafish resulted in lens developmental defect that was characterized by retaining of nuclei in lens fiber cells. In previous in vitro studies, we found that Hsf4 promoted DNase2ß expression in human and mouse lens epithelial cells. In this study, it was found that, instead of DNase2ß, DNase1l1l is uniquely expressed in zebrafish lens and was absent in Hsf4-/- zebrafish lens. Using CRISPR-Cas9 technology, a DNase1l1l knockout zebrafish line was constructed, which developed cataract. Deletion of DNase1l1l totally abrogated lens primary and secondary fiber cell denucleation process, whereas had little effect on the clearance of other organelles. The transcriptional regulation of DNase1l1l was dramatically impaired in Hsf4-/- zebrafish lens. Rescue of DNase1l1l mRNA into Hsf4-/- zebrafish embryos alleviated its defect in lens fiber cell denucleation. Our results in vivo demonstrated that DNase1l1l is the primary DNase responsible for nuclear DNA degradation in lens fiber cells, and Hsf4 can transcriptionally activate DNase1l1l expression in zebrafish.


Subject(s)
Cataract/genetics , Deoxyribonucleases/genetics , Gene Expression Regulation, Developmental , Heat Shock Transcription Factors/metabolism , Lens, Crystalline/embryology , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , CRISPR-Cas Systems/genetics , Cataract/pathology , Cell Nucleus/metabolism , Deoxyribonucleases/metabolism , Disease Models, Animal , Embryo, Nonmammalian , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Knockout Techniques , Heat Shock Transcription Factors/genetics , Humans , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Male , Zebrafish , Zebrafish Proteins/metabolism
20.
Mol Med Rep ; 20(5): 4323-4330, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31545442

ABSTRACT

Cell migration is important for renal recovery from tubular cell injury. Heat shock transcription factor 1 (HSF1) is a well­studied regulatory factor that is active during acute kidney injury. HSF1 is also involved in the migration process during tumor metastasis. Therefore, we hypothesized that HSF1 may promote the recovery of renal function by affecting kidney tubular cell migration. A wound healing assay was used to examine the cell migration rate. The results demonstrated that the migration of rat kidney proximal tubular cells (RPTCs) was increased following knockdown of HSF1. In addition, the invasion ability of HSF1 knockdown RPTCs was also significantly upregulated. The present study also identified that transforming growth factor­ß1 (TGF­ß1) was highly expressed at the edge of the wound in control cells, and its expression was further increased upon knockdown of HSF1. Inhibition of TGF­ß1 signaling prevented RPTC HSF1 knockdown cell migration, suggesting that HSF1­regulated RPTC cell migration was dependent on the TGF­ß1 signaling pathway. Furthermore, phosphorylation of TGF­ß1 and Smad2/3 was induced in HSF1 knockdown cells. Together, these results suggest that HSF1 may suppress RPTC migration by inhibiting the activation of the TGF­ß1­Smad2/3 signaling pathway.


Subject(s)
Epithelial Cells/metabolism , Heat Shock Transcription Factors/metabolism , Kidney Tubules/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Cell Movement/genetics , Gene Knockdown Techniques , Heat Shock Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...