Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Publication year range
1.
Eur J Clin Microbiol Infect Dis ; 43(3): 403-416, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38153660

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) infection has been identified to serve as the primary cause of acute lower respiratory infectious diseases in children under the age of one and a significant risk factor for the emergence and development of pediatric recurrent wheezing and asthma, though the exact mechanism is still unknown. METHODS AND RESULTS: In this study, we discuss the key routes that lead to recurrent wheezing and bronchial asthma following RSV infection. It is interesting to note that following the coronavirus disease 2019 (COVID-19) epidemic, the prevalence of RSV changes significantly. This presents us with a rare opportunity to better understand the associated mechanism for RSV infection, its effects on the respiratory system, and the immunological response to RSV following the COVID-19 epidemic. To better understand the associated mechanisms in the occurrence and progression of pediatric asthma, we thoroughly described how the RSV infection directly destroys the physical barrier of airway epithelial tissue, promotes inflammatory responses, enhances airway hyper-responsiveness, and ultimately causes the airway remodeling. More critically, extensive discussion was also conducted regarding the potential impact of RSV infection on host pulmonary immune response. CONCLUSION: In conclusion, this study offers a comprehensive perspective to better understand how the RSV infection interacts in the control of the host's pulmonary immune system, causing recurrent wheezing and the development of asthma, and it sheds fresh light on potential avenues for pharmaceutical therapy in the future.


Subject(s)
Asthma , COVID-19 , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Child , Humans , Infant , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Sounds/etiology , COVID-19/complications , Asthma/complications , Asthma/epidemiology
2.
Expert Rev Mol Med ; 25: e17, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37132248

ABSTRACT

Timothy syndrome (TS), characterised by multiple system malfunction especially the prolonged corrected QT interval and synchronised appearance of hand/foot syndactyly, is an extremely rare disease affecting early life with devastating arrhythmia. In this work, firstly, the various mutations in causative gene CACNA1C encoding cardiac L-type voltage-gated calcium channel (LTCC), regard with the genetic pathogeny and nomenclature of TS are reviewed. Secondly, the expression profile and function of CACNA1C gene encoding Cav1.2 proteins, and its gain-of-function mutation in TS leading to multiple organ disease phenotypes especially arrhythmia are discussed. More importantly, we focus on the altered molecular mechanism underlying arrhythmia in TS, and discuss about how LTCC malfunction in TS can cause disorganised calcium handling with excessive intracellular calcium and its triggered dysregulated excitation-transcription coupling. In addition, current therapeutics for TS cardiac phenotypes including LTCC blockers, beta-adrenergic blocking agents, sodium channel blocker, multichannel inhibitors and pacemakers are summarised. Eventually, the research strategy using patient-specific induced pluripotent stem cells is recommended as one of the promising future directions for developing therapeutic approaches. This review updates our understanding on the research progress and future avenues to study the genetics and molecular mechanism underlying the pathogenesis of devastating arrhythmia within TS, and provides novel insights for developing therapeutic measures.


Subject(s)
Long QT Syndrome , Syndactyly , Humans , Long QT Syndrome/therapy , Long QT Syndrome/drug therapy , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/therapy , Syndactyly/therapy , Syndactyly/drug therapy , Mutation
3.
Eur J Immunol ; 51(8): 2062-2073, 2021 08.
Article in English | MEDLINE | ID: mdl-33864383

ABSTRACT

MicroRNA-147 (miR-147) had been previously found induced in synoviocytes by inflammatory stimuli derived from T cells in experimental arthritis. This study was designed to verify whether loss of its function might alleviate inflammatory events in joints of experimental and rheumatoid arthritis (RA). Dark Agouti (DA) rats were injected intradermally with pristane to induce arthritis, and rno-miR-147 antagomir was locally administrated into individual ankle compared with negative control or rno-miR-155-5p antagomir (potential positive control). Arthritis onset, macroscopic severity, and pathological changes were monitored. While in vitro, gain or loss function of hsa-miR-147b-3p/hsa-miR-155-5p and ZNF148 was achieved in human synovial fibroblast cell line SW982 and RA synovial fibroblasts (RASF). The expression of miRNAs and mRNAs was detected by using RT-quantitative PCR, and protein expression was detected by using Western blotting. Anti-miR-147 therapy could alleviate the severity, especially for the synovitis and joint destruction in experimental arthritis. Gain of hsa-miR-147b-3p/hsa-miR-155-5p function in TNF-α stimulated SW982 and RASF cells could upregulate, in contrast, loss of hsa-miR-147b-3p/hsa-miR-155-5p function could downregulate the gene expression of TNF-α, IL-6, MMP3, and MMP13. Hence, such alteration could participate in synovial inflammation and joint destruction. RNAi of ZNF148, a miR-147's target, increased gene expression of TNF-α, IL-6, MMP3, and MMP13 in SW982 and RASF cells. Also, mRNA sequencing data showed that hsa-miR-147b-3p mimic and ZNF148 siRNA commonly regulated the gene expression of CCL3 and DEPTOR as well as some arthritis and inflammation-related pathways. Taken together, miR-147b-3p contributes to synovial inflammation through repressing ZNF148 in RA and experimental arthritis.


Subject(s)
Arthritis, Rheumatoid/immunology , DNA-Binding Proteins/immunology , Gene Expression Regulation/immunology , MicroRNAs/immunology , Synovial Membrane/pathology , Transcription Factors/immunology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , DNA-Binding Proteins/metabolism , Female , Humans , Inflammation , Male , MicroRNAs/metabolism , Middle Aged , Rats , Transcription Factors/metabolism
4.
Bioorg Med Chem ; 66: 116803, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35561631

ABSTRACT

MicroRNA-21 is a carcinogenic microRNA, whose overexpression arises in a variety of tumor tissues. Hence, microRNA-21 a prospective target for cancer treatment, and regulation of microRNA-21 by small molecule inhibitors is deemed as a promising approach for tumor therapy. In this work, to discover potent microRNA-21 inhibitor, series of 4-(N-norfloxacin-acyl)aminobenzamides were designed and synthesized, and their inhibitory effects were appraised by utilizing dual luciferase reporter assays. The results indicated that compound A7 was the most efficient microRNA-21 small molecule inhibitor. What's more, A7 suppressed the migration of Hela cells and the colony formation of Hela and HCT-116 cells as well as promoted apoptosis of Hela cells. In the mechanism study, results of RT-qPCR certified that A7 could reduce the level of mature microRNA-21 via disrupting its expression at the transcriptional level of its primary form "pri-miR-21", which was distinct from most previous inhibitors directly binding with pre-miR-21. Noticeably, Western blotting and RT-qPCR uncovered A7 could upregulate the expression PTEN, EGR1 and SLIT2, which are the downstream functional targets of microRNA-21. These findings demonstrated that A7 was a promising microRNA-21 small molecule inhibitor and 4-(N-norfloxacin-acyl) aminobenzamide can serve as a new scaffold for discovery of potent microRNA-21 inhibitor.


Subject(s)
Antineoplastic Agents , Benzamides , MicroRNAs , Norfloxacin , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Proliferation , HCT116 Cells , HeLa Cells , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Norfloxacin/pharmacology
5.
J Immunol ; 205(1): 181-192, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32503893

ABSTRACT

Recent studies indicate that glucose metabolism is altered in rheumatoid arthritis. We hypothesize that Pkm2, as a key regulatory enzyme of glycolysis pathway, triggers the activation of macrophages (Mφ), which results in proinflammatory cytokine production during the arthritis progress. In this study, Pkm2 was found to be overexpressed in ED1-positive Mφ in spleens and synovial tissues from arthritic rats via immunofluorescence, Western blotting, and quantitative RT-PCR. To reveal the role of Pkm2, Dark Agouti rats were treated with either Pkm2 enzyme inhibitor shikonin or the RNA interference plasmids of Pkm2 and negative control plasmids, respectively, via i.p. injection. Pkm2 intervention could alleviate the severity of pristane-induced arthritis in aspects of the macroscopic arthritis score, perimeter changes of midpaw, and the synovitis and destruction of the bone and cartilage as well as reduce the ED1 and p-Stat1-positive cell population in rat synovial tissues. Silencing Pkm2 by RNA interference in classical activated rat and mouse Mφ resulted in less Tnf-α, Il-1ß production via Stat1 signaling. Collectively, Pkm2 is highly expressed in ED1-positive Mφ of spleens and synovial tissues from arthritic rats and promotes Mφ activation via Stat1 signaling. Pkm2 might be a promising selective metabolic target molecule for rheumatoid arthritis treatment.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Macrophages/immunology , Pyruvate Kinase/metabolism , STAT1 Transcription Factor/metabolism , Animals , Arthritis, Experimental/diagnosis , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/pathology , Gene Knockdown Techniques , Humans , Macrophages/metabolism , Mice , Naphthoquinones/administration & dosage , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Rats , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Synovial Membrane/immunology , Synovial Membrane/pathology
6.
Exp Cell Res ; 398(1): 112404, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33245891

ABSTRACT

Pristane-induced arthritis (PIA) could be adoptively transferred by splenic T cells in rats, and innate immunity should play critical roles in T cell activation. However, in pre-clinical stage, the activation mechanism of innate cells like macrophages remains unclear. Here we found that PIA was dependent on macrophages since cell depletion alleviated disease severity. Splenic macrophages of PIA rats showed M1 phenotypic shifting. The quantitative proteomics analysis suggested that macrophages initiated metabolic reprogramming with the conversion of aerobic oxidation to glycolysis in response to pristane in vivo. Notably, macrophages treated with pristane showed mitochondrial dysregulation and increased glycolysis flux and enzyme activity. Additionally, TNFα production, strongly associating with the glycolysis enzyme Ldha/Ldhb, could be reduced as glycolysis was inhibited or be enhanced as citrate cycle was blocked. This work provides detailed insights into the molecular mechanisms of pristane-mediated metabolic reprogramming in macrophages and suggests a new therapeutic strategy for arthritic disorders.


Subject(s)
Arthritis, Experimental/chemically induced , Inflammation/chemically induced , Macrophages/drug effects , Terpenes/pharmacology , Anaerobiosis/drug effects , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Cells, Cultured , Deoxyglucose/pharmacology , Glycolysis/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Macrophages/metabolism , Malonates/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Nitro Compounds/pharmacology , Propionates/pharmacology , Rats , Terpenes/antagonists & inhibitors , Wortmannin/pharmacology
7.
Int J Mol Sci ; 22(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573239

ABSTRACT

Sevoflurane, one of the most commonly used pediatric anesthetics, was found to cause developmental neurotoxicity. To understand specific risk groups and develop countermeasures, a better understanding of its mechanisms is needed. We hypothesize that, as in many other brain degeneration pathways, long non-coding RNAs (lncRNAs) are involved in the sevoflurane-induced neurotoxicity. Postnatal day 7 (PD7) mice were exposed to 3% sevoflurane for 6 h. To quantify neurotoxicity in these mice, we (1) detected neural apoptosis through analysis of caspase 3 expression level and activity and (2) assessed long-term learning ability via the Morris water maze at PD60. To elucidate specific mechanisms, profiles of 27,427 lncRNAs and 18,855 messenger RNAs (mRNAs) in mouse hippocampi were analyzed using microarray assays. Sevoflurane-induced abnormal lncRNA and mRNA expression-associated function pathways were predicted by bioinformatic analysis. We found that sevoflurane induced significant neurotoxicity, causing acute neuroapoptosis and abnormal expression of 148 mRNAs and 301 lncRNAs on PD7 in mouse hippocampus. Additionally, exposed mice exhibited impaired memory on PD60. Bioinformatic analysis predicted that the dysregulated mRNAs, which are highly correlated with their co-expressed dysregulated lncRNAs, might be involved in 34 neurodegenerative signaling pathways (e.g., brain cell apoptosis and intellectual developmental disorder). Our study reveals for the first time that neonatal exposure to 3% sevoflurane induces abnormal lncRNA and mRNA expression profiles. These dysregulated lncRNAs/mRNAs form wide molecular networks that might contribute to various functional neurological disease pathways in the hippocampus, resulting in the observed acute apoptosis and impaired long-term memory.


Subject(s)
Anesthetics, Inhalation/toxicity , Neurotoxicity Syndromes/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , Sevoflurane/toxicity , Anesthetics, Inhalation/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis/genetics , Child Development/drug effects , Computational Biology , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/pathology , Humans , Infant , Male , Memory/drug effects , Mice , Neurotoxicity Syndromes/pathology , Sevoflurane/administration & dosage , Signal Transduction/drug effects , Signal Transduction/genetics , Toxicity Tests, Acute
8.
Clin Immunol ; 220: 108579, 2020 11.
Article in English | MEDLINE | ID: mdl-32866644

ABSTRACT

Endoplasmic reticulum (ER) stress associated proteins contribute to the pathogenesis of rheumatoid arthritis (RA) through affecting synoviocyte proliferation and proinflammatory cytokine production. The role of DERL3, an ER-associated degradation component, in joint inflammation of RA was explored. Synovial tissues from RA and osteoarthritis (OA) patients were collected, and in RA synovial tissue, DERL3 showed up-regulation and significantly positive correlation with the expression of tumor necrosis factor alpha (TNF-α), interleukin (IL)-6 and matrix metalloproteinase (MMP)-1. Immunofluorescence result suggested DERL3 was located in fibroblast-like synoviocytes (FLS). Among different inflammatory stimuli, DERL3 could be up-regulated by TNF-α stimulation in FLS. Under TNF-α stimulation, knocking down DERL3, the expression of IL-6, IL-8, MMP-1, MMP-13 was reduced and the activation of nuclear factor kappa B (NF-κB) signaling pathway was inhibited. In pristane-induced arthritis (PIA) rat model, Derl3 was up-regulated in synovial tissue and disease was attenuated after intraarticular injection of siDerl3. Overall, we conclude that TNF-α inducing DERL3 expression promotes the inflammation of FLS through activation of NF-κB signaling pathway, suggesting DERL3 plays important roles in the pathogenesis of RA and is a promising therapeutic target.


Subject(s)
Arthritis, Rheumatoid/immunology , Membrane Proteins/immunology , Synoviocytes/immunology , Aged , Animals , Arthritis, Experimental/immunology , Cells, Cultured , Cytokines/immunology , Female , Humans , Male , Matrix Metalloproteinase 1/immunology , Matrix Metalloproteinase 13/immunology , Mice , Middle Aged , NF-kappa B/immunology , Osteoarthritis/immunology , Rats , Signal Transduction
9.
BMC Med Genet ; 20(1): 96, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31151434

ABSTRACT

BACKGROUND: The highly conservative miR-15/107 family (also named as miR-15/107 gene group) including ten miRNA members is currently recognized strongly implicated in multiple human disorders. Some studies focus on the entire family rather than individual miRNA for a bigger picture, while there is also certain signature dysregulation for some of the individual miRNA implicated even in the same disorder. METHODS: Faced with the exponential growth of experimental evidence, our study tries to analyze their function and target interactions using various bioinformatics tools. RESULTS: Firstly, the evolutionary conservative "AGCAGC" sequence and possible clustered transcriptional pattern were described. Secondly, both the experimentally validated and bioinformatically predicted miRNA-target gene relationship of the entire family was analyzed to understand the mechanism of underlying collective effects for target regulation from the miR-15/107 family. Moreover, pathway analysis among miR-15/107 family was performed and displayed in detail, while its impact on cell proliferation is experimentally validated. Eventually, the dysregulation of miR-15/107 in diseases was discussed. CONCLUSIONS: In summary, our study proposes that the collective functions and implication of miR-15/107 family in various human diseases are achieved relying on the massive overlapping target genes. While the minor differences within target gene interaction among family members could also explain the signature behavior for some of the individual miRNA in aspects such as its disease-specific dysregulation and various participation in pathways.


Subject(s)
Epistasis, Genetic , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks , MicroRNAs/genetics , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Cluster Analysis , Computational Biology/methods , Genetic Predisposition to Disease/genetics , Humans , Multigene Family , Signal Transduction/genetics
10.
Biotechnol Appl Biochem ; 66(5): 755-762, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31021480

ABSTRACT

The therapeutic potential of microRNA-21 (miR-21) small-molecule inhibitors has been of particular interest to medicinal chemists. Moreover, the development of more facile screening methods is lacking. In the present study, two potential screening strategies for miR-21 small-molecule inhibitor including the stem-loop reverse transcription-quantitative PCR and dual luciferase reporter assay system were demonstrated and discussed in detail. A pmirGLO-miR21cswt plasmid and its two different mutants were constructed for dual luciferase reporter assay system. In addition, the sensitivity and specificity of these two methods were validated. Our results demonstrated that both strategies are decent choices for the screening of small-molecule inhibitors for miR-21 and possibly other miRNAs. Eventually, we applied our optimized strategy to discover and characterize several promising compounds such as azobenzene derivate A, enoxacin, and norfloxacin for their potential impact on intracellular miR-21 concentration.


Subject(s)
Genes, Reporter/drug effects , Luciferases, Firefly/antagonists & inhibitors , MicroRNAs/pharmacology , Real-Time Polymerase Chain Reaction , Small Molecule Libraries/pharmacology , Drug Evaluation, Preclinical , Genes, Reporter/genetics , HeLa Cells , High-Throughput Screening Assays , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Tumor Cells, Cultured
11.
Mediators Inflamm ; 2019: 6768504, 2019.
Article in English | MEDLINE | ID: mdl-31275058

ABSTRACT

Dysregulation of multiple microRNAs widely takes place during rheumatoid arthritis (RA) and experimental arthritides. This study is performed to explore the possible mechanism underlying DICER1 deficiency-mediated inflammation in human synoviocytes SW982. Firstly, RNAi of DICER1 led to increased COX2, MMP3, and MMP13 protein production, while DICER1 overexpression could reduce MMP13 expression. Secondly, the increase of IL-8 and decrease of TGF-ß1 and TIMP1 were determined in the supernatant derived from DICER1 siRNA-treated cells, while DICER1 overexpression was found capable to reverse this effect. Ingenuity pathway analysis (IPA) software predicted that the Dicer1 deficiency-induced dysregulated cytokines in synoviocytes could possibly lead to the inflammatory disorders in the synovial tissue. Moreover, DICER1 deficiency could also reduce apoptosis, while DICER1 overexpression was found to decrease the proliferation and enhance apoptosis. In addition, DICER1 deficiency could lower the expression of multiple RA-related miRNAs such as miR-155. Meanwhile, DICER1 overexpression could rescue their low expression levels. And then, gain or loss of miR-155 function could regulate the protein levels of MMP3 and MMP13. These results indicated that DICER1 might play its role through regulating its downstream RA-related miRNAs. Our data demonstrated that DICER1 deficiency could cause multiple proinflammatory events in human synoviocytes SW982. This mechanism study might provide the possible target molecule to modify the inflammatory destruction and overproliferation in synoviocytes.


Subject(s)
DEAD-box RNA Helicases/metabolism , Inflammation/metabolism , Ribonuclease III/metabolism , Synoviocytes/metabolism , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line , Cell Proliferation/genetics , Cell Proliferation/physiology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DEAD-box RNA Helicases/genetics , Humans , Inflammation/genetics , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , RNA Interference , Ribonuclease III/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/metabolism
12.
J Cell Mol Med ; 22(1): 241-250, 2018 01.
Article in English | MEDLINE | ID: mdl-28782180

ABSTRACT

MicroRNAs are considered to play critical roles in the pathogenesis of human inflammatory arthritis, including rheumatoid arthritis (RA). The purpose of this study was to determine the relationship between miR-10a-5p and TBX5 in synoviocytes and evaluate their contribution to joint inflammation. The expression of miR-10a-5p and TBX5 in the synovium of RA and human synovial sarcoma cell line SW982 stimulated by IL-1ß was determined by RT-qPCR and Western blotting. The direct interaction between miR-10a-5p and TBX5 3'UTR was determined by dual-luciferase reporter assay in HeLa cells. Mimics and inhibitors of miR-10a-5p were transfected into SW982 cells. TBX5 was overexpressed by plasmid transfection or knocked down by RNAi. Proinflammatory cytokines and TLR3 and MMP13 expressions were determined by RT-qPCR and Western blotting. Down-regulated expression of miR-10a-5p and up-regulation of TBX5 in human patients with RA were found compared to patients with OA. IL-1ß could reduce miR-10a-5p and increase TBX5 expression in SW982 cells in vitro. The direct target relationship between miR-10a-5p and 3'UTR of TBX5 was confirmed by luciferase reporter assay. Alterations of miR-10-5p after transfection with its mimic and inhibitor caused the related depression and re-expression of TBX5 and inflammatory factors in SW982 cells. Overexpression of TBX5 after pCMV3-TBX5 plasmid transfection significantly promoted the production of TLR3, MMP13 and various inflammatory cytokines, while this effect was rescued after knocking down of TBX5 with its specific siRNA. We conclude that miR-10a-5p in a relation with TBX5 regulates joint inflammation in arthritis, which would serve as a diagnostic and therapeutic target for RA treatment.


Subject(s)
Down-Regulation/genetics , Inflammation/genetics , Inflammation/pathology , Joints/pathology , MicroRNAs/genetics , Synoviocytes/metabolism , T-Box Domain Proteins/metabolism , Adult , Aged , Arthritis, Rheumatoid/pathology , Base Sequence , Cell Line, Tumor , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Male , MicroRNAs/metabolism , Middle Aged , Synovial Membrane/pathology , Synoviocytes/pathology , T-Box Domain Proteins/genetics , Up-Regulation/genetics
13.
Cell Physiol Biochem ; 49(6): 2496-2510, 2018.
Article in English | MEDLINE | ID: mdl-30261491

ABSTRACT

BACKGROUND: Propofol induces acute neurotoxicity (e.g., neuroapoptosis) followed by impairment of long-term memory and learning in animals. However, underlying mechanisms remain largely unknown. Long non-coding RNAs (lncRNAs) are found to participate in various pathological processes. We hypothesized that lncRNA profile and the associated signaling pathways were altered, and these changes might be related to the neurotoxicity observed in the neonatal mouse hippocampus following propofol exposure. METHODS: In this laboratory experiment, 7-day-old mice were exposed to a subanesthetic dose of propofol for 3 hours, with 4 animals per group. Hippocampal tissues were harvested 3 hours after propofol administration. Neuroapoptosis was analyzed based on caspase 3 activity using a colorimetric assay. A microarray was performed to investigate the profiles of 35,923 lncRNAs and 24,881 messenger RNAs (mRNAs). Representative differentially expressed lncRNAs and mRNAs were validated using reverse transcription quantitative polymerase chain reaction. All mRNAs dysregulated by propofol and the 50 top-ranked, significantly dysregulated lncRNAs were subject to bioinformatics analysis for exploring the potential mechanisms and signaling network of propofol-induced neurotoxicity. RESULTS: Propofol induced neuroapoptosis in the hippocampus, with differential expression of 159 lncRNAs and 100 mRNAs (fold change ± 2.0, P< 0.05). Bioinformatics analysis demonstrated that these lncRNAs and their associated mRNAs might participate in neurodegenerative pathways (e.g., calcium handling, apoptosis, autophagy, and synaptogenesis). CONCLUSION: This novel report emphasizes that propofol alters profiles of lncRNAs, mRNAs, and their cooperative signaling network, which provides novel insights into molecular mechanisms of anesthetic-induced developmental neurodegeneration and preventive targets against the neurotoxicity.


Subject(s)
Anesthetics/pharmacology , Apoptosis/drug effects , Hippocampus/metabolism , Propofol/pharmacology , RNA, Long Noncoding/metabolism , Transcriptome/drug effects , Animals , Computational Biology , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Clin Immunol ; 175: 56-68, 2017 02.
Article in English | MEDLINE | ID: mdl-27940139

ABSTRACT

Autophagy is involved in both innate and adaptive immune regulation. We propose that autophagy regulates activation of TLR3 in macrophages and is thereby essential for development of pristane-induced arthritis. We found that pristane treatment induced autophagy in macrophages in vitro and in vivo, in spleen cells from pristane injected rats. The induced autophagy was associated with STAT1 phosphorylation and expression of IRF1 and TLR3. Blocking the pristane activated autophagy by Wortmannin and Bafilomycin A1 or by RNAi of Becn1 led to a downregulation of the associated STAT1-IRF1-TLR3 pathway. Most importantly, the development of arthritis was alleviated by suppressing either autophagy or TLR3. We conclude that pristane enhanced autophagy, leading to a STAT1-IRF1 controlled upregulation of TLR3 expression in macrophages, is a pathogenic mechanism in the development of arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Autophagy/drug effects , Interferon Regulatory Factor-1/metabolism , Macrophages/drug effects , STAT1 Transcription Factor/metabolism , Terpenes/pharmacology , Toll-Like Receptor 3/metabolism , Animals , Arthritis, Experimental/metabolism , Down-Regulation/drug effects , Macrophages/metabolism , Phosphorylation/drug effects , Rats , Signal Transduction/drug effects , Up-Regulation/drug effects
15.
J Immunol ; 195(1): 298-306, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26026059

ABSTRACT

Protein arginine methyltransferase (PRMT)1, methylating both histones and key cellular proteins, has emerged as a key regulator of various cellular processes. This study aimed to identify the mechanism that regulates PRMT1 in chronic Ag-induced pulmonary inflammation (AIPI) in the E3 rat asthma model. E3 rats were challenged with OVA for 1 or 8 wk to induce acute or chronic AIPI. Expression of mRNAs was detected by real-time quantitative PCR. PRMT1, TGF-ß, COX2, and vascular endothelial growth factor protein expression in lung tissues was determined by immunohistochemistry staining and Western blotting. In the in vitro study, IL-4-stimulated lung epithelial cell (A549) medium (ISEM) with or without anti-TGF-ß Ab was applied to human fibroblasts from lung (HFL1). The proliferation of HFL1 was determined by MTT. AMI-1 (pan-PRMT inhibitor) was administered intranasally to chronic AIPI rats to determine PRMT effects on asthmatic parameters. In lung tissue sections, PRMT1 expression was significantly upregulated, mainly in epithelial cells, in acute AIPI lungs, whereas it was significantly upregulated mainly in fibroblasts in chronic AIPI lungs. The in vitro study revealed that ISEM elevates PRMT1, COX2, and vascular endothelial growth factor expressions, and it promoted fibroblast proliferation. The application of anti-TGF-ß Ab suppressed COX2 upregulation by ISEM. AMI-1 inhibited the expression of COX2 in TGF-ß-stimulated cells. In the in vivo experiment, AMI-1 administered to AIPI rats reduced COX2 production and humoral immune response, and it abrogated mucus secretion and collagen generation. These findings suggested that TGF-ß-induced PRMT1 expression participates in fibroblast proliferation and chronic airway inflammation in AIPI.


Subject(s)
Asthma/immunology , Cyclooxygenase 2/immunology , Epithelial Cells/immunology , Fibroblasts/immunology , Protein-Arginine N-Methyltransferases/immunology , Transforming Growth Factor beta/immunology , Acute Disease , Animals , Antibodies/pharmacology , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Cell Proliferation , Chronic Disease , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2/genetics , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation , Humans , Interleukin-4/pharmacology , Lung/drug effects , Lung/immunology , Lung/pathology , Naphthalenesulfonates/pharmacology , Ovalbumin , Pneumonia , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Rats , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Urea/analogs & derivatives , Urea/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology
16.
Exp Cell Res ; 344(1): 67-75, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27107686

ABSTRACT

Thioredoxin reductase 2 (TrxR2) is a selenium (Se) containing protein. Se deficiency is associated with an endemic osteoarthropathy characterized by impaired cartilage formation. It is unclear whether TrxR2 have roles in cartilage function. We examined the effects of TrxR2 on chondrogenic ATDC5 cells through shRNA-mediated gene silencing of TrxR2. We demonstrated TrxR2 deficiencies could enhance chondrogenic differentiation and apoptosis of ATDC5 cells. TrxR2 deficiencies increased accumulation of cartilage glycosaminoglycans (GAGs) and mineralization. TrxR2 deficiencies also stimulated expression of extracellular (ECM) gene including Collagen II and Aggrecan. The enhanced chondrogenic properties were further confirmed by activation of Akt signaling which are required for chondrogenesis. In addition, TrxR2 deficiencies promoted chondrocyte proliferation through acceleration of cell cycle progression by increase in both S and G2/M phase cell distribution accompanied with induction of parathyroid hormone-related protein (PTHrP). Moreover, TrxR2 deficiencies induced chondrocyte death via apoptosis and increased cell sensitivity to exogenous oxidative stress. Furthermore, TrxR2 deficiencies induced emission of mitochondrial reactive oxygen species (ROS) without alteration of mitochondrial membrane potential and intracellular ATP content. Finally, treatment of TrxR2 deficiency cells with N-acetylcysteine (NAC) inhibited mitochondrial ROS production and chondrocyte apoptosis. NAC also prevented chondrogenic differentiation of TrxR2 deficiency cells by suppression of ECM gene expression, GAGs accumulation and mineralization, as well as attenuation of Akt signaling. Thus, TrxR2-mediated mitochondrial integrity is indispensable for chondrogenic differentiation of ATDC5 cells. TrxR2 deficiency-induced impaired proliferation and death of chondrocytes may be the pathological mechanism of the osteoarthropathy due to Se deficiency. Notably, this study also uncover the roles of mitochondrial ROS which could stimulate cartilage ECM synthesis that offer novel insights for development of therapeutic agent to prevent cartilage degeneration in human disease.


Subject(s)
Apoptosis , Cell Differentiation , Chondrocytes/cytology , Chondrogenesis , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 2/deficiency , Acetylcysteine/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Humans , Mitochondria/drug effects , Thioredoxin Reductase 2/metabolism
17.
BMC Pulm Med ; 16: 50, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27075111

ABSTRACT

BACKGROUND: This study aims to find out whether extracellular miRNAs is implicated in recurrent childhood wheezing with asthmatic risk. METHODS: One hundred and forty children of Chinese Han population were recruited for this study. Plasma and intracellular miRNAs from children with recurrent wheezing and rats with antigen induced pulmonary inflammation (AIPI) were detected by using reverse transcription-quantitative PCR. Differential leukocytes in blood were automatically counted. Total IgE was detected by enzyme-linked immunosorbent assay. Clinical implication in diagnosis was evaluated using receiver operating characteristic curves. RESULTS: The increase of plasma miR-21 and miR-26a was screened out from 11 candidate miRNAs and validated in wheezing children. The level of expression for both miRNAs were comparable in different age and gender. Plasma miR-21 was more preferable to miR-26a and total IgE for diagnosis. Plasma miR-21 and miR-26a levels were not significantly correlated with various leukocyte counts or miRNA expression in blood cells. In acute and chronic AIPI rats, miR-21 levels increased in both plasma and lavaged lung compared with control. Moreover, circulating miR-21 and miR-26a levels were highly positively correlated with infiltrated cell counts in bronchoalveolar lavage fluid of AIPI rats. CONCLUSIONS: Circulating miR-21 and miR-26a increase in wheezing children and AIPI rats. This not only manifests their strong clinical implication in recurrent childhood wheezing with asthma risk, but also provides novel insights into the role of extracellular miRNAs during development of airway inflammation and recurrent wheezing.


Subject(s)
MicroRNAs/genetics , Pneumonia/genetics , Respiratory Sounds/genetics , Animals , Antigens/immunology , Antigens/toxicity , Asian People , Child , Child, Preschool , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Forced Expiratory Volume , Humans , Immunoglobulin E/immunology , Infant , Male , MicroRNAs/blood , Ovalbumin/immunology , Ovalbumin/toxicity , Pneumonia/chemically induced , Pneumonia/immunology , Rats , Recurrence , Respiratory Sounds/immunology , Respiratory Sounds/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Vital Capacity
18.
Clin Immunol ; 156(2): 141-53, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25533241

ABSTRACT

Based on pristane-induced arthritis (PIA), we found that T cells mediate TLR3 overexpression in fibroblast-like synoviocytes (FLS). The aim of this study is to determine key factors by which T cells induce TLR3 expression. Rat FLS were co-cultured with pristane primed T cell conditioned medium (PPT medium), and TLR3 expression of FLS was significantly induced. TNF-α, IFN-γ and IL-17 were dominantly expressed in PIA T cells. The overexpression of TLR3 and its related genes in FLS co-cultured with PPT medium could be reduced through blocking TNF-α pathway. CD4(+) T cells from spleen of PIA rats showed increase of TNF-α secretion. P38 MAPK and NF-κB were activated in FLS by PPT medium, and their inhibitors decreased TLR3 upregulation significantly. Finally, TNF-α induced TLR3 expression was confirmed in human synovial cells. Summarily, TNF-α derived from pristane primed T cells induced TLR3 expression of FLS through activating p38 MAPK and NF-κB pathways.


Subject(s)
Arthritis, Experimental/immunology , NF-kappa B/immunology , Toll-Like Receptor 3/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Arthritis, Experimental/chemically induced , CD4-Positive T-Lymphocytes/immunology , Cell Line , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Immunosuppressive Agents/pharmacology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-17/biosynthesis , Interleukin-6/biosynthesis , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Pyridines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Sulfones/pharmacology , Synovial Membrane/cytology , Terpenes/pharmacology , Toll-Like Receptor 3/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Up-Regulation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
Bioorg Med Chem ; 23(19): 6510-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26344589

ABSTRACT

MicroRNA-21, as an oncogenic miRNA, has caught great attention for medicinal chemists to develop its novel inhibitors for cancer therapy. In the present study, we designed 4-benzoylamino-N-(prop-2-yn-1-yl)benzamides as miR-21 inhibitor candidates on the basis of scaffold hopping. Eighteen compounds were synthesized. The inhibitory activities of synthesized compounds against the expression of miR-21 were evaluated using stem loop RT-qPCR and compound 1j was discovered as the most potent compound, which displayed a time and concentration dependent inhibition manner. In addition, various functional assays such as the expression of miR-21 target gene detected by Western blotting and the cell growth and apoptosis detected by flow cytometric analysis were checked in Hela (human epithelioid cervix carcinoma) and U-87 MG (human glioblastoma) cells to confirm its activity. The results indicate that compound 1j can enhance apoptosis, retard proliferation, and up-regulate PDCD4, a target protein of miR-21. In addition, the compound 1j does not influence the expression of multiple miRNAs and the genes that participate in miRNA universal biosynthesis pathway. These results strongly support the assumption that title compounds can serve as a small molecule inhibitor of miR-21.


Subject(s)
Benzamides/chemistry , MicroRNAs/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Benzamides/metabolism , Benzamides/toxicity , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , HeLa Cells , Humans , MicroRNAs/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction
20.
Sci Prog ; 107(2): 368504241247402, 2024.
Article in English | MEDLINE | ID: mdl-38651330

ABSTRACT

Idiopathic pulmonary fibrosis is a chronic and progressive interstitial lung disease with a poor prognosis. Idiopathic pulmonary fibrosis is characterized by repeated alveolar epithelial damage leading to abnormal repair. The intercellular microenvironment is disturbed, leading to continuous activation of fibroblasts and myofibroblasts, deposition of extracellular matrix, and ultimately fibrosis. Moreover, pulmonary fibrosis was also found as a COVID-19 complication. Currently, two drugs, pirfenidone and nintedanib, are approved for clinical therapy worldwide. However, they can merely slow the disease's progression rather than rescue it. These two drugs have other limitations, such as lack of efficacy, adverse effects, and poor pharmacokinetics. Consequently, a growing number of molecular therapies have been actively developed. Treatment options for IPF are becoming increasingly available. This article reviews the research platform, including cell and animal models involved in molecular therapy studies of idiopathic pulmonary fibrosis as well as the promising therapeutic targets and their development progress during clinical trials. The former includes patient case/control studies, cell models, and animal models. The latter includes transforming growth factor-beta, vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factor, lysophosphatidic acid, interleukin-13, Rho-associated coiled-coil forming protein kinase family, and Janus kinases/signal transducers and activators of transcription pathway. We mainly focused on the therapeutic targets that have not only entered clinical trials but were publicly published with their clinical outcomes. Moreover, this work provides an outlook on some promising targets for further validation of their possibilities to cure the disease.


Subject(s)
Idiopathic Pulmonary Fibrosis , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Humans , Animals , Molecular Targeted Therapy/methods , Pyridones/therapeutic use , Indoles/therapeutic use , Indoles/pharmacology , COVID-19 , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL