1
|
Chen L, Zhu MY, Wang GX, Lu LL, Lin L, Lei L, Wu T. Ruxolitinib ameliorated coxsackievirus B3-induced acute viral myocarditis by suppressing the JAK-STAT pathway. Int Immunopharmacol 2023; 124:110797. [PMID: 37634445 DOI: 10.1016/j.intimp.2023.110797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Accumulating evidences have demonstrated that overwhelming inflammation occurs in the process of Coxsackievirus B3 (CVB3)-induced acute viral myocarditis (AVM). No specific therapy is available. More than an effective Janus-associated kinase (JAK) inhibiter, ruxolitinib exerts a critical role in the inflammatory diseases. In this study, we investigated the potential effect of ruxolitinib on CVB3-induced acute viral myocarditis. METHOD In vivo, BALB/c mice were intraperitoneally injected of CVB3, treated of a successive gavage of ruxolitinib for seven days, and subjected to a series of analysis. In vitro, primary bone marrow-derived macrophages (BMDMs) and cardiac fibroblasts were isolated, cultured, treated, harvested and finally detected. RESULTS In vivo, acute viral myocarditis was successfully induced by the injection of CVB3 characterized by impaired cardiac function, predominant infiltration of inflammatory cells, necroptosis of myocardium, great increase of cardiac troponin I (cTnI) and cytokine levels, replication of CVB3, and excessive activation of JAK-STAT pathways. Oral administration of ruxolitinib suppressed the activation of JAK-STAT pathway in a dosage-dependent way, lessened the infiltration of inflammatory cells and necroptosis of myocardium, reduced the levels of cTnI and cytokines, and finally alleviated CVB3-induced cardiac dysfunction, with the reduced production of type I interferon and no promising effect on the replication of CVB3. In vitro, the treatment of ruxolitinib inhibited the activation of JAK-STAT pathway and increase of multiple cytokines mRNA levels in BMDMs and had no protective effect against CVB3 replication in cardiac fibroblasts. CONCLUSIONS Our study suggested that ruxolitinib ameliorated CVB3-induced AVM by inhibiting the activation of JAK-STAT pathway, infiltration of inflammatory cells and necroptosis of myocardium, which may provide a novel strategy for AVM therapy.
Collapse
Affiliation(s)
- Liang Chen
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meng-Ying Zhu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gao-Xiang Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei 430030, China
| | - Li-Li Lu
- Institute of Pharmaceutical Innovation, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Lei Lei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Ting Wu
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
2
|
Thanikachalam PV, Ramamurthy S, Mallapu P, Varma SR, Narayanan J, Abourehab MA, Kesharwani P. Modulation of IL-33/ST2 signaling as a potential new therapeutic target for cardiovascular diseases. Cytokine Growth Factor Rev 2023; 71-72:94-104. [PMID: 37422366 DOI: 10.1016/j.cytogfr.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
IL-33 belongs to the IL-1 family of cytokines, which function as inducers of Th2 cytokine production by binding with ST2L and IL-1RAcP. This, in turn, activates various signaling pathways, including the mitogen-activated protein kinase (MAPK), the inhibitor of Kappa-B kinase (IKK) pathway, and the phospholipase D-sphingosine kinase pathway. IL-33 has demonstrated protective effects against various cardiovascular diseases (CVDs) by inducing Th2 cytokines and promoting alternative activating M2 polarization. However, the soluble decoy form of ST2 (sST2) mitigates the biological effects of IL-33, exacerbating CVDs. Furthermore, IL-33 also plays a significant role in the development of asthma, arthritis, atopic dermatitis, and anaphylaxis through the activation of Th2 cells and mast cells. In this review, we aim to demonstrate the protective role of IL-33 against CVDs from 2005 to the present and explore the potential of serum soluble ST2 (sST2) as a diagnostic biomarker for CVDs. Therefore, IL-33 holds promise as a potential therapeutic target for the treatment of CVDs.
Collapse
Affiliation(s)
- Punniyakoti Veeraveedu Thanikachalam
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India.
| | - Srinivasan Ramamurthy
- College of Pharmacy and Health Sciences, University of Science and Technology of Fujairah, Fujairah, United Arab Emirates
| | - Poojitha Mallapu
- Department of Pharmacology, GRT Institute of Pharmaceutical Education and Research, Tiruttani, India
| | - Sudhir Rama Varma
- Department of Clinical Sciences, Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Jayaraj Narayanan
- Department of Basic Sciences, Center of Medical and Bio-allied Health Sciences Research, Ajman university, Ajman, United Arab Emirates
| | - Mohammed As Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
3
|
Xu J, Zhou Z, Zheng Y, Yang S, Huang K, Li H. Roles of inflammasomes in viral myocarditis. Front Cell Infect Microbiol 2023; 13:1149911. [PMID: 37256114 PMCID: PMC10225676 DOI: 10.3389/fcimb.2023.1149911] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/28/2023] [Indexed: 06/01/2023] Open
Abstract
Viral myocarditis (VMC), characterized by viral infection-induced inflammation, is a life-threatening disease associated with dilated cardiomyopathy or heart failure. Innate immunity plays a crucial role in the progression of inflammation, in which inflammasomes provide a platform for the secretion of cytokines and mediate pyroptosis. Inflammasomes are rising stars gaining increasing attention. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, the caspase recruitment domain-containing protein 8 (CARD8) inflammasome, and the caspase-11 inflammasome are three inflammasomes that were reported to affect the process and prognosis of VMC. These inflammasomes can be activated by a wide range of cellular events. Accumulating evidence has suggested that inflammasomes are involved in different stages of VMC, including the trigger and progression of myocardial injury and remodeling after infection. In this review, we summarized the pathways involving inflammasomes in VMC and discussed the potential therapies targeting inflammasomes and related pathways.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yidan Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sai Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huili Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Li M, Chen L, Zhao Y, Sun H, Zhao L. Research on the Mechanism of HRP Relieving IPEC-J2 Cells Immunological Stress Based on Transcriptome Sequencing Analysis. Front Nutr 2022; 9:944390. [PMID: 35911118 PMCID: PMC9336541 DOI: 10.3389/fnut.2022.944390] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/13/2022] [Indexed: 11/27/2022] Open
Abstract
Early weaning increased the economic benefits of piglets. However, early weaning damages the intestinal barrier of piglets and causes immunological stress. The mechanism by which Hippophae rhamnoides polysaccharide (HRP) alleviates lipopolysaccharide (LPS)-induced intestinal porcine epithelial cells (IPEC-J2) inflammatory damage was investigated using proteomics in our previous studies. In this study we employed RNA-sequencing (RNA-seq) to determine the level and function of differentially expressed genes (DEGs) and further explore the mechanism of the HRP anti-inflammatory and immune process. The differential expression analysis indicated that 3622, 1216, and 2100 DEGs in the IPEC-J2 cells were identified in C vs. L, L vs. H6-L, and C vs. H6-L, respectively. The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis foundsix identified pathways related to the immune system. Additionally, we used the Science, Technology, Engineering, and Math (STEM) program to categorize the 3,134 DEGs that were differentially expressed in H2-L, H4-L and H6-L into eight possible expression profiles, in which 612 were clustered into two profiles. The accuracy and consistency of RNA-seq data were validated by the results of qRT-PCR of the nuclear factor of kappa light polypeptide gene enhancer in B-cells 2 (NFKB2), MAP kinase interacting serine/threonine kinase 2 (MKNK2), mitogen-activated protein kinase kinase 1 (MAP2K1), mitogen-activated protein kinase kinase kinase 8 (MAP3K8), Ras-related protein R-Ras (RRAS), TNF receptor-associated factor 1 (TRAF1), NF-kappa-B inhibitor alpha (NFKBIA), interleukin 8 (IL8), tumor necrosis factor, alpha-induced protein 3 (TNFAIP3), and transforming growth factor beta-1 (TGFB1). Transcriptome sequencing also indicated that HRP reduced the expression levels of related DEGs and inhibited the activation of the mitogen-activated protein kinase (MAPK)/nuclear factor kappa-B (NF-κB) signaling pathway. Our findings indicate that the application of HRP in piglet diets during the early weaning period can improve intestinal epithelial function and integrity, and relieve intestinal damage, and improve piglet health.
Collapse
Affiliation(s)
- Muyang Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Lu Chen
- Shanxi Animal Husbandry and Veterinary School, Taiyuan, China
| | - Yiran Zhao
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- *Correspondence: Lei Zhao
| | - Lei Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Hui Sun
| |
Collapse
|
5
|
Li B, Xie X. A20 (TNFAIP3) alleviates viral myocarditis through ADAR1/miR-1a-3p-dependent regulation. BMC Cardiovasc Disord 2022; 22:10. [PMID: 35034631 PMCID: PMC8762865 DOI: 10.1186/s12872-021-02438-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 12/09/2021] [Indexed: 11/29/2022] Open
Abstract
Objective To investigate the effect of A20 and how A20 is regulated in viral myocarditis (VMC). Methods BABL/C mice, primary neonatal rat cardiomyocytes and H9c2 cells were infected with Coxsackie virus B3 (CVB3) to establish animal and cellular models of VMC. H&E staining revealed the pathologic condition of myocardium. ELISA measured the serum levels of creatine kinase, creatine kinase isoenzyme and cardiac troponin I. The effects of A20, miR-1a-3p and ADAR1 were investigated using gain and loss of function approaches. ELISA measured the levels of IL-6, IL-18 and TNF-α in serum or cell culture supernatant. TUNEL staining and flow cytometry assessed the apoptosis of myocardium and cardiomyocytes, respectively. RNA-binding protein immunoprecipitation and dual-luciferase reporter assays verified the binding between A20 and miR-1a-3p. Co-immunoprecipitation assay verified the binding between ADAR1 and Dicer. Results A20 was underexpressed and miR-1a-3p was overexpressed in the myocardium of VMC mice as well as in CVB3-infected cardiomyocytes. Overexpression of A20 suppressed cardiomyocyte inflammation and apoptosis in vivo and in vitro. miR-1a-3p promoted CVB3-induced inflammation and apoptosis in cardiomyocytes by binding to A20. The expression of miR-1a-3p was regulated by ADAR1. ADAR1 promoted the slicing of miR-1a-3p precursor by binding to Dicer. Conclusion A20, regulated by ADAR1/miR-1a-3p, suppresses inflammation and cardiomyocyte apoptosis in VMC. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02438-z.
Collapse
Affiliation(s)
- Bin Li
- Department of Cardiovascular Medicine, Affiliated Hospital of Xiangnan University, No. 25, West Renmin Road, Chenzhou, 423000, Hunan, People's Republic of China
| | - Xing Xie
- Department of Cardiovascular Medicine, Affiliated Hospital of Xiangnan University, No. 25, West Renmin Road, Chenzhou, 423000, Hunan, People's Republic of China.
| |
Collapse
|
6
|
El-Far YM, El-Mesery M. Pevonedistat attenuates cisplatin-induced nephrotoxicity in mice by downregulating the release of inflammatory mediators. J Biochem Mol Toxicol 2021; 35:e22908. [PMID: 34476871 DOI: 10.1002/jbt.22908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 07/09/2021] [Accepted: 08/20/2021] [Indexed: 01/11/2023]
Abstract
Pevonedistat (MLN4924) is a specific NEDD8-activating enzyme inhibitor that inactivates cullin-RING ligases involved in ubiquitylation and turnover of different signaling molecules. In the current study, we evaluated the effect of pevonedistat on cisplatin (CIS)-induced nephrotoxicity in mice. Serum creatinine and urea levels were analyzed in different groups. Histopathological examination of renal tissue was done using hematoxylin and eosin staining. In addition, renal IL-6 and TNF-α expressions were analyzed using the enzyme-linked immunosorbent assay technique, and IL-1β and NF-κB expressions were analyzed by immunohistochemical staining of renal tissue. Caspase-3, A20, β-catenin, and Nrf2 gene expressions in renal tissue were analyzed using the reverse-transcription polymerase chain reaction technique. Western blot analysis was adopted to assess cleaved caspase-3 and β-catenin expressions in renal tissue. Pevonedistat coadministration with CIS improved kidney functions and attenuated CIS-induced nephrotoxicity as indicated by the significant decrease in serum creatinine and urea levels. In addition, pevonedistat coadministration with CIS showed a significant decrease in caspase-3 and a significant increase in A20, β-catenin, and Nrf2 gene expressions. Also, pevonedistat decreased caspase-3 cleavage to p19 in mice treated with CIS. Moreover, pevonedistat decreased CIS-induced upregulation of IL-6, TNF-α, IL-1β, and NF-κB protein expressions in renal tissue. Thus, pevonedistat alleviated CIS-induced nephrotoxicity that might be attributed to suppression of the inflammation induced in renal tissue.
Collapse
Affiliation(s)
- Yousra M El-Far
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Cui SB, Wang TX, Liu ZW, Yan JY, Zhang K. Zinc finger protein A20 regulates the development and progression of osteoarthritis by affecting the activity of NF-κB p65. Immunopharmacol Immunotoxicol 2021; 43:713-723. [PMID: 34463587 DOI: 10.1080/08923973.2021.1970764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
OBJECTIVE To investigate the role of Zinc finger protein A20 in osteoarthritis (OA) by regulating NF-κB p65. METHODS A20, MMP1, MMP13 and IL-1β expressions in human OA cartilage samples were detected by qRT-PCR. IL-1β-induced chondrocyte was treated with A20 lentivirus activation particle, pyrrolidine dithiocarbamate (PDTC, a NF-κB inhibitor) with/without A20 siRNA. IL-6, TNF-α, and PGE2 levels were measured by ELISA, and NO production by Greiss reaction. Destabilization of the medial meniscus (DMM) surgery was used to construct the OA models, followed by injection of A20 adenovirus. MMP1 and MMP13 expression was measured by immunohistochemistry. The mRNA and protein expression were performed by qRT-PCR and western blotting, respectively. RESULTS A20 was down-regulated in human OA cartilage samples, and negatively correlated with the expressions of MMP1, MMP13 and IL-1β. The IL-1β-induced chondrocyte manifested decreased A20 with increased NF-κB p65 activity. A20 overexpression suppressed the NF-κB p65 activity in IL-1β-induced chondrocyte. Furthermore, PDTC decreased IL-1β-induced chondrocyte apoptosis with the upregulated COL1A1, COL2A1, COL10A1 and ACAN, as well as the down-regulated MMP1, MMP13, COX2, iNOS, IL-6, TNF-α, NO and PGE2, which was reversed by A20 siRNA. In vivo, OA mice gained higher OARSI score and Mankin's score, exhibited up-regulations of MMP1 and MMP13, and decreased NF-κB p65 activity, which was improved after injection of A20 adenovirus. CONCLUSION A20 was reduced in OA cartilage samples, and its overexpression, by suppressing the activity of NF-κB p65, could improve IL-1β-induced chondrocyte degradation and apoptosis in vitro, as well as mitigate the inflammation in OA mice.
Collapse
Affiliation(s)
- Shu-Bei Cui
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Tao-Xia Wang
- Department of Nephrology, Affiliated Hospital of Hebei University of Technology, Handan, China
| | - Zhen-Wu Liu
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Ji-Ying Yan
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Kai Zhang
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| |
Collapse
|
8
|
Long C, Xie N, Shu Y, Wu Y, He P, Zhou Y, Xiang Y, Gu J, Yang L, Wang Y. Knockout of the Cannabinoid Receptor 2 Gene Promotes Inflammation and Hepatic Stellate Cell Activation by Promoting A20/Nuclear Factor-κB (NF-κB) Expression in Mice with Carbon Tetrachloride-Induced Liver Fibrosis. Med Sci Monit 2021; 27:e931236. [PMID: 34413280 PMCID: PMC8409143 DOI: 10.12659/msm.931236] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background This study aimed to investigate the effect of deleting the cannabinoid receptor 2 (CB2) gene on the development of hepatic fibrosis induced by carbon tetrachloride (CCl4) in mice via regulating inflammation. Material/Methods The DNA was extracted from the tails of mice to identify whether the cannabinoid receptor 2 gene was successfully knocked out. A liver fibrosis model was established by an intraperitoneal injection of CCl4 into mice. Hepatic damage and hepatic fibrosis were evaluated by detecting serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and staining paraffin sections of liver tissue with hematoxylin-eosin (HE). The secretion and distribution of collagen in liver tissue were observed by Masson staining. Western blot analysis was performed to detect the expression of α-smooth muscle actin (α-SMA), transforming growth factor-β1 (TGF-β1), tumor necrosis factor alpha-induced protein 3 (A20), phosphorylated nuclear factor-κB p65 (p-NF-κB p65), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6) in liver tissue. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect the expression of IL-6 and TNF-α mRNA in liver tissue. Results Compared with the control mice, the mice with CB2 knockout that were exposed to CCl4 exhibited increased liver damage, liver fibrosis, and upregulated α-SMA, TGF-β1, A20, and p-NF-κB p65 protein levels. IL-6 and TNF-α protein levels and mRNA levels were upregulated. Conclusions The deletion of the CB2 gene promoted the activation of hepatic stellate cells in mice with liver fibrosis and aggravated liver fibrosis by up-regulating the protein expression of A20 and p-NF-κB p65 and inducing inflammatory response, potentially providing new insight into the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Cuizhen Long
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,Medical Laboratory Department, The Second Nanning People's Hospital, Nanning, Guangxi, China (mainland)
| | - Na Xie
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Yuanhui Shu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Yafeng Wu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,Department of Clinical Laboratory, The Fourth People's Hospital of Ya'an City, Ya'an, Sichuan, China (mainland)
| | - Ping He
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Yan Zhou
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Yining Xiang
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Junying Gu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Lei Yang
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Yuping Wang
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| |
Collapse
|
9
|
Khanna M, Gautam A, Rajput R, Sharma L. Natural Products as a Paradigm for the Treatment of Coxsackievirus - induced Myocarditis. Curr Top Med Chem 2020; 20:607-616. [PMID: 31995007 DOI: 10.2174/1568026620666200129094516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/22/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
Coxsackievirus B3 (CVB3), a member of the Picornaviridae family, is considered to be one of the most important infectious agents to cause virus-induced myocarditis. Despite improvements in studying viral pathology, structure and molecular biology, as well as diagnosis of this disease, there is still no virus-specific drug in clinical use. Structural and nonstructural proteins produced during the coxsackievirus life cycle have been identified as potential targets for blocking viral replication at the step of attachment, entry, uncoating, RNA and protein synthesis by synthetic or natural compounds. Moreover, WIN (for Winthrop) compounds and application of nucleic-acid based strategies were shown to target viral capsid, entry and viral proteases, but have not reached to the clinical trials as a successful antiviral agent. There is an urgent need for diverse molecular libraries for phenotype-selective and high-throughput screening.
Collapse
Affiliation(s)
- Madhu Khanna
- Department of Microbiology (Virology Unit), Vallabhbhai Patel Chest Institute, University of Delhi, Delhi-110007, India
| | - Anju Gautam
- Department of Microbiology (Virology Unit), Vallabhbhai Patel Chest Institute, University of Delhi, Delhi-110007, India
| | - Roopali Rajput
- Department of Microbiology (Virology Unit), Vallabhbhai Patel Chest Institute, University of Delhi, Delhi-110007, India
| | - Latika Sharma
- Department of Microbiology (Virology Unit), Vallabhbhai Patel Chest Institute, University of Delhi, Delhi-110007, India
| |
Collapse
|
10
|
Xue YL, Zhang SX, Zheng CF, Li YF, Zhang LH, Su QY, Hao YF, Wang S, Li XW. Long non-coding RNA MEG3 inhibits M2 macrophage polarization by activating TRAF6 via microRNA-223 down-regulation in viral myocarditis. J Cell Mol Med 2020; 24:12341-12354. [PMID: 33047847 PMCID: PMC7686963 DOI: 10.1111/jcmm.15720] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 06/24/2020] [Accepted: 07/18/2020] [Indexed: 12/01/2022] Open
Abstract
Viral myocarditis (VMC) commonly triggers heart failure, for which no specific treatments are available. This study aims to explore the specific role of long non‐coding RNA (lncRNA) maternally expressed 3 (MEG3) in VMC. A VMC mouse model was induced by Coxsackievirus B3 (CVB3). Then, MEG3 and TNF receptor‐associated factor 6 (TRAF6) were silenced and microRNA‐223 (miR‐223) was over‐expressed in the VMC mice, followed by determination of ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS). Dual‐luciferase reporter assay was introduced to test the interaction among MEG3, TRAF6 and miR‐223. Macrophages were isolated from cardiac tissues and bone marrow, and polarization of M1 or M2 macrophages was induced. Then, the expressions of components of NLRP3 inflammatory body (NLRP3, ASC, Caspase‐1), M1 markers (CD86, iNOS and TNF‐α) and M2 markers (CD206, Arginase‐1 and Fizz‐1) were measured following MEG3 silencing. In the VMC mouse model, MEG3 and TRAF6 levels were obviously increased, while miR‐223 expression was significantly reduced. Down‐regulation of MEG3 resulted in the inhibition of TRAF6 by promoting miR‐223. TRAF6 was negatively correlated with miR‐223, but positively correlated with MEG3 expression. Down‐regulations of MEG3 or TRAF6 or up‐regulation of miR‐223 was observed to increase mouse weight, survival rate, LVEF and LVFS, while inhibiting myocarditis and inflammation via the NF‐κB pathway inactivation in VMC mice. Down‐regulation of MEG3 decreased M1 macrophage polarization and elevated M2 macrophage polarization by up‐regulating miR‐223. Collectively, down‐regulation of MEG3 leads to the inhibition of inflammation and induces M2 macrophage polarization via miR‐223/TRAF6/NF‐κB axis, thus alleviating VMC.
Collapse
Affiliation(s)
- Yu-Long Xue
- Department of Cardiovascular Medicine, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chao-Feng Zheng
- Department of Genetics Laboratory, Linfen Maternity & Child Healthcare Hospital, Linfen, China
| | - Yu-Feng Li
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li-Hui Zhang
- Department of Cardiovascular Medicine, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Qin-Yi Su
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yu-Fei Hao
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shu Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xue-Wen Li
- Department of Cardiovascular Medicine, Shanxi Dayi Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
11
|
Wang D, Wang J. Antiviral immune mechanism of Toll-like receptor 4-mediated human alveolar epithelial cells type Ⅱ. Exp Ther Med 2020; 20:2561-2568. [PMID: 32765749 PMCID: PMC7401722 DOI: 10.3892/etm.2020.8963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 12/18/2019] [Indexed: 11/06/2022] Open
Abstract
Expression of Toll-like receptor (TLR)4 and its downstream substances, myeloid differentiation factor 88 (MyD88), NF-κB p65, tumor necrosis factor-α (TNF-α) and GR in human alveolar epithelial cells type Ⅱ (AEC Ⅱ) infected with respiratory syncytial virus (RSV) were investigated, and the antiviral immune mechanism mediated by TLR4 was explored. Human AEC Ⅱ were divided into TLR4-/- group, normal group and TLR4+ group, and also into control group, RSV group and RSV+MP (methylprednisolone) group. MTT assay was used to measure the survival of cells after TLR4 knockout and overexpression, and the survival of normal cells after treatment with MP. The concentration of TLR4, MyD88, NF-κB p65, TNF-α, and GR was measured by ELISA after TLR4 knockout and overexpression. Reverse transcription-quantitative PCR (RT-qPCR) was used to measure the mRNA expression of the gene knockout and overexpression groups. RT-qPCR and western blot analysis were used to determine the expression of TLR4, MyD88, NF-κB p65 and GR in RSV and RSV+MP groups. The concentration of the detected substances in the TLR4-/- group was significantly lower than that in the normal group (P<0.01 and <0.001), and in the TLR4+ group was significantly higher than that in the normal group (P<0.05, <0.01 and <0.001); the expression of RSV in the TLR4-/- group was significantly higher than that in the normal group (P<0.001), and in the TLR4+ group was significantly lower than that in the normal group (P<0.05). The expression levels of TLR4, MyD88 and NF-κB p65 in the RSV and RSV+MP groups were significantly higher than those in the control group (P<0.05, <0.01 and <0.001), and the increase presented in the RSV+MP group was significantly lower than that in the RSV group (P<0.05 and <0.01). TLR4-mediated antiviral immunity of human AEC Ⅱ can reduce the levels of TLR4, MyD88, NF-κB p65 and TNF-α and increase the level of GR, participating in the immune defense and reducing the damage of the viral epithelial cells of human type Ⅱ alveoli, thus improving human immunity.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Infectious Disease, Xuzhou Children's Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Jie Wang
- Department of Infectious Disease, Xuzhou Children's Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| |
Collapse
|
12
|
Daba TM, Zhao Y, Pan Z. Advancement of Mechanisms of Coxsackie Virus B3-Induced Myocarditis Pathogenesis and the Potential Therapeutic Targets. Curr Drug Targets 2020; 20:1461-1473. [PMID: 31215390 DOI: 10.2174/1389450120666190618124722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/21/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Viral myocarditis is a cardiac disease caused by Group B Coxsackie virus of Enterovirus genus in the Picorna viridae family. It causes heart failure in children, young and adults. Ten Percent (10%) of acute heart failure and 12% of sudden deaths in young and adults who are less than 40 years is due to this viral myocarditis. If treatment action is not taken earlier, the viral disease can develop into chronic myocarditis and Dilated Cardiomyopathy which lead to congestive heart failure. And these eventually result in a reduced cardiac function which finally brings the victim to death. The only treatment option of the disease is heart transplantation once the acute stage of disease develops to chronic and Dilated Cardiomyopathy. Currently, there is a limitation in daily clinical treatments and even some available treatment options are ineffective. Therefore, focusing on search for treatment options through investigation is imperative. Recent studies have reported that biological molecules show a promising role. But their mechanism of pathogenesis is still unclear. A detailed study on identifying the role of biological molecules involved in Coxsackie B3 virus induced myocarditis and their mechanisms of pathogenesis; compiling and disseminating the findings of the investigation to the scientific communities contribute one step forward to the solution. Therefore, this review is aimed at compiling information from findings of current studies on the potential therapeutic role of micro RNA, cytokines and chemokines on the mechanism of pathogenesis of Coxsackie virus B3- induced myocarditis to give brief information for scholars to conduct a detailed study in the area.
Collapse
Affiliation(s)
- Tolessa Muleta Daba
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, China.,Department of Biology, College of Natural and Computational Sciences, Bule Hora University, Bule Hora, Ethiopia
| | - Yue Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhenwei Pan
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Wu S, Wang HQ, Guo TT, Li YH. Luteolin inhibits CVB3 replication through inhibiting inflammation. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2020; 22:762-773. [PMID: 31321999 DOI: 10.1080/10286020.2019.1642329] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/30/2019] [Accepted: 07/08/2019] [Indexed: 06/10/2023]
Abstract
Coxsackievirus B3 (CVB3) infection causes many inflammation-related diseases, such as viral myocarditis and aseptic meningitis. However, no vaccines or drugs have been approved for prevention or therapy of CVB3-induced diseases. In this study, luteolin (3,4,5,7-tetrahydroxyflavone) had been found that could dose-dependently reduce the production of viral progeny and synthesis of CVB3 RNA and protein. The luteolin-mediated inhibition of CVB3 was found to be mechanistically possible, at least in part, through depressing the phosphorylation of p38 MAPK and JNK MAPK, and inhibiting NF-κB nuclear translocation and subsequently attenuated the expression of inflammatory cytokines in CVB3-infected cells. Luteolin may be a potential agent or supplement against CVB3 infection by inhibiting inflammation.
Collapse
Affiliation(s)
- Shuo Wu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Hui-Qiang Wang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| | - Ting-Ting Guo
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-Huan Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing 100050, China
| |
Collapse
|
14
|
Programmed Cell Death-1: Programmed Cell Death-Ligand 1 Interaction Protects Human Cardiomyocytes Against T-Cell Mediated Inflammation and Apoptosis Response In Vitro. Int J Mol Sci 2020; 21:ijms21072399. [PMID: 32244307 PMCID: PMC7177768 DOI: 10.3390/ijms21072399] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 12/14/2022] Open
Abstract
Aim: Immunological checkpoint therapy is considered a powerful method for cancer therapy and acts by re-activating autologous T cells to kill the cancer cell. Myocarditis cases have been reported in cancer patients after immunological therapy; for example, nivolumab treatment is a monoclonal antibody that blocks programmed cell death-1/programmed cell death ligand-1 ligand interaction. This project provided insight into the inflammatory response as a benchmark to investigate the potential cardiotoxic effect of T cell response to the programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis in regulating cardiomyocyte injury in vitro. Methods and Results: We investigated cardiomyopathy resulted from the PD-1/PD-L1 axis blockade using the anti-PD-1 antibody in Rockefeller University embryonic stem cells-derived cardiomyocytes (RUES2-CMs) and a melanoma tumor-bearing murine model. We found that nivolumab alone did not induce inflammatory-related proteins, including PD-L1 expression, and did not induce apoptosis, which was contrary to doxorubicin, a cardiotoxic chemotherapy drug. However, nivolumab was able to exacerbate the immune response by increasing cytokine and inflammatory gene expression in RUES2-CMs when co-cultured with CD4+ T lymphocytes and induced apoptosis. This effect was not observed when RUES2-CMs were co-cultured with CD8+ T lymphocytes. The in vivo model showed that the heart function of tumor-bearing mice was decreased after treatment with anti-PD-1 antibody and demonstrated a dilated left ventricle histological examination. The dilated left ventricle was associated with an infiltration of CD4+ and CD8+ T lymphocytes into the myocardium. PD-L1 and inflammatory-associated gene expression were significantly increased in anti-PD-1-treated tumor-bearing mice. Cleaved caspase-3 and mouse plasma cardiac troponin I expressions were increased significantly. Conclusion: PD-L1 expression on cardiomyocytes suppressed T-cell function. Blockade of PD-1 by nivolumab enhanced cardiomyocyte inflammation and apoptosis through the enhancement of T-cell response towards cardiomyocytes.
Collapse
|
15
|
Fei Y, Chaulagain A, Wang T, Chen Y, Liu J, Yi M, Wang Y, Huang Y, Lin L, Chen S, Xu W, Tong L, Wu X, Zhao D, Zhang F, Zhao W, Zhong Z. MiR-146a down-regulates inflammatory response by targeting TLR3 and TRAF6 in Coxsackievirus B infection. RNA (NEW YORK, N.Y.) 2020; 26:91-100. [PMID: 31676570 PMCID: PMC6913124 DOI: 10.1261/rna.071985.119] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/28/2019] [Indexed: 05/08/2023]
Abstract
Coxsackievirus B (CVB) is the major cause of human myocarditis and dilated cardiomyopathy. Toll-like receptor 3 (TLR3) is an intracellular sensor to detect pathogen's dsRNA. TLR3, along with TRAF6, triggers an inflammatory response through NF-κB signaling pathway. In the cells infected with CVB type 3 (CVB3), the abundance of miR-146a was significantly increased. The role of miR-146a in CVB infection is unclear. In this study, TLR3 and TRAF6 were identified as the targets of miR-146a. The elevated miR-146a inhibited NF-κB translocation and subsequently down-regulated proinflammatory cytokine expression in the CVB3-infected cells. Therefore, the NF-κB pathway can be doubly blocked by miR-146a through targeting of TLR3 and TRAF6. MiR-146a may be a negative regulator on inflammatory response and an intrinsic protective factor in CVB infection.
Collapse
Affiliation(s)
- Yanru Fei
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Anita Chaulagain
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Tianying Wang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Yang Chen
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Jinchang Liu
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Ming Yi
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Ying Wang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Yike Huang
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Lexun Lin
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Sijia Chen
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Weizhen Xu
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Lei Tong
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Xiaoyu Wu
- Department of Cardiology, The First Hospital of Harbin Medical University, Harbin 150001, China
| | - Dechao Zhao
- Department of Cardiology, The First Hospital of Harbin Medical University, Harbin 150001, China
| | - Fengmin Zhang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Wenran Zhao
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Zhaohua Zhong
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
16
|
Li S, Wang Y, Zhao C, Zhang M, Wang W, Yu X, Huang J, Wang Z, Zhu B, Yin C, Cai H. Akt inhibitor deguelin aggravates inflammation and fibrosis in myocarditis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1275-1282. [PMID: 32128091 PMCID: PMC7038425 DOI: 10.22038/ijbms.2019.35518.8473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 05/12/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Myocarditis is characterized by inflammatory cell infiltration in myocardial stroma. Attenuation of tumor necrosis factor (TNF)-α and interleukin (IL)-1β is a reliable mark for improving the prognosis. Protein kinase B (Akt) plays an important role in the development and progression of myocarditis. The specific role of the natural inhibitor of Akt, Deguelin, on myocarditis has not been reported. In this study, we used deguelin to investigate the effects of natural Akt inhibitor on myocarditis in experimental autoimmune myocarditis (EAM) rats. MATERIALS AND METHODS EAM rat models were made by using Lewis rats and Deguelin was injected intraperitoneally on day 3, 6, 9, 12 and 15 after successful modeling. On day 18, rats were sacrificed and the heart weight (HW)/ body weight (BW) ratio were measured. The pathological changes, pathological scores and fibrosis area were evaluated after H.&E. and Masson's trichrome staining. The mRNA levels of TNF-α and IL-1β were measured by RT-qPCR, while the protein expressions of TNF-α and IL-1β were detected by immunohistochemical staining and Western bolt. The protein expressions of Akt, Akt1, phosphorylated (p-) Akt and nuclear factor (NF)-κB were detected by Western bolt. RESULTS We found that the TNF-α and IL-1β levels, inflammatory scores and fibrosis areas were markedly increased after 18 days deguelin administration. CONCLUSION Akt inhibition with deguelin may aggravate myocarditis of EAM rats.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Yue Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Chunming Zhao
- Human anatomy and Histology and Embryology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Meixiang Zhang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Wei Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaowei Yu
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jiao Huang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zhao Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Bo Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Chengqian Yin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Hongxing Cai
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
17
|
Al-Huseini I, Harada M, Nishi K, Nguyen-Tien D, Kimura T, Ashida N. Improvement of insulin signalling rescues inflammatory cardiac dysfunction. Sci Rep 2019; 9:14801. [PMID: 31616027 PMCID: PMC6794250 DOI: 10.1038/s41598-019-51304-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 09/27/2019] [Indexed: 12/26/2022] Open
Abstract
Inflammation resulting from virus infection is the cause of myocarditis; however, the precise mechanism by which inflammation induces cardiac dysfunction is still unclear. In this study, we investigated the contribution of insulin signalling to inflammatory cardiac dysfunction induced by the activation of signalling by NF-κB, a major transcriptional factor regulating inflammation. We generated mice constitutively overexpressing kinase-active IKK-β, an essential kinase for NF-κB activation, in cardiomyocytes (KA mice). KA mice demonstrated poor survival and significant cardiac dysfunction with remarkable dilation. Histologically, KA hearts revealed increased cardiac apoptosis and fibrosis and the enhanced recruitment of immune cells. By molecular analysis, we observed the increased phosphorylation of IRS-1, indicating the suppression of insulin signalling in KA hearts. To evaluate the contribution of insulin signalling to cardiac dysfunction in KA hearts, we generated mice with cardiac-specific suppression of phosphatase and tensin homologue 10 (PTEN), a negative regulator of insulin signalling, in the KA mouse background (KA-PTEN). The suppression of PTEN successfully improved insulin signalling in KA-PTEN hearts, and interestingly, KA-PTEN mice showed significantly improved cardiac function and survival. These results indicated that impaired insulin signalling underlies the mechanism involved in inflammation-induced cardiac dysfunction, which suggests that it may be a target for the treatment of myocarditis.
Collapse
Affiliation(s)
- Isehaq Al-Huseini
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Masayuki Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiyoto Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA98109, USA
| | - Dat Nguyen-Tien
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Molecular Immunology and Inflammation, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Noboru Ashida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
18
|
Blanter M, Sork H, Tuomela S, Flodström-Tullberg M. Genetic and Environmental Interaction in Type 1 Diabetes: a Relationship Between Genetic Risk Alleles and Molecular Traits of Enterovirus Infection? Curr Diab Rep 2019; 19:82. [PMID: 31401790 PMCID: PMC6689284 DOI: 10.1007/s11892-019-1192-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We provide an overview of the current knowledge regarding the natural history of human type 1 diabetes (T1D) and the documented associations between virus infections (in particular the enteroviruses) and disease development. We review studies that examine whether T1D-specific risk alleles in genes involved in the function of the immune system can alter susceptibility to virus infections or affect the magnitude of the host antiviral response. We also highlight where the major gaps in our knowledge exist and consider possible implications that new insights gained from the discussed gene-environment interaction studies may bring. RECENT FINDINGS A commonality between several of the studied T1D risk variants studied is their role in modulating the host immune response to viral infection. Generally, little support exists indicating that the risk variants increase susceptibility to infection and moreover, they usually appear to predispose the immune system towards a hyper-reactive state, decrease the risk of infection, and/or favor the establishment of viral persistence. In conclusion, although the current number of studies is limited, this type of research can provide important insights into the mechanisms that are central to disease pathogenesis and further describe how genetic and environmental factors jointly influence the risk of T1D development. The latter may provide genetic markers that could be used for patient stratification and for the selection of method(s) for disease prevention.
Collapse
Affiliation(s)
- Marfa Blanter
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- 0000 0001 0668 7884grid.5596.fLaboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, EU Belgium
| | - Helena Sork
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Soile Tuomela
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Flodström-Tullberg
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Hu T, Hu W, Ma L, Zeng X, Liu J, Cheng B, Yang P, Qiu S, Yang G, Chen D, Liu Z. pVAX1-A20 alleviates colitis in mice by promoting regulatory T cells. Dig Liver Dis 2019; 51:790-797. [PMID: 30528569 DOI: 10.1016/j.dld.2018.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/02/2018] [Accepted: 11/04/2018] [Indexed: 12/11/2022]
Abstract
AIM To investigate whether the intrarectal administration of the ubiquitin E3 ligase A20 (A20) attenuates intestinal inflammation and influences regulatory T cells in experimental colitis. METHODS A dextran sulfate sodium induced chronic colitis mouse model was established. The symptoms and manifestations of colitis and the severity of colonic mucosal inflammation were evaluated. The protective role of A20 expression in the intestine was analyzed after the administration of a pVAX1-A20 recombinant eukaryotic vector, which was encapsulated into poly(L-lactide-co-glycolide) as a nanoparticle. RESULTS pVAX1-A20 administration markedly ameliorated colonic tissue damage and reduced intestinal inflammation via the suppression of the mucosal mitogen-activated protein kinase and nuclear factor (NF)-κB signaling cascade. Furthermore, pVAX1-A20 promoted the splenic regulatory T cell population and forkhead box P3 expression in colonic tissue. CONCLUSION A20 plays a key role in the regulation of intestinal inflammation and that the overexpression of A20 in the intestine protects mice from dextran sulfate sodium induced chronic colitis.
Collapse
Affiliation(s)
- Tianyong Hu
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | | | - Li Ma
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Xianhai Zeng
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Jiangqi Liu
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Baohui Cheng
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Pingchang Yang
- Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen, China
| | - Shuqi Qiu
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Gui Yang
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Donghui Chen
- Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China.
| | - Zhiqiang Liu
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China.
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The aim of this study is to summarize the literature describing the pathogenesis, diagnosis and management of cardiomyopathy related to myocarditis. RECENT FINDINGS Myocarditis has a variety of causes and a heterogeneous clinical presentation with potentially life-threatening complications. About one-third of patients will develop a dilated cardiomyopathy and the pathogenesis is a multiphase, mutlicompartment process that involves immune activation, including innate immune system triggered proinflammatory cytokines and autoantibodies. In recent years, diagnosis has been aided by advancements in cardiac MRI, and in particular T1 and T2 mapping sequences. In certain clinical situations, endomyocardial biopsy (EMB) should be performed, with consideration of left ventricular sampling, for an accurate diagnosis that may aid treatment and prognostication. SUMMARY Although overall myocarditis accounts for a minority of cardiomyopathy and heart failure presentations, the clinical presentation is variable and the pathophysiology of myocardial damage is unique. Cardiac MRI has significantly improved diagnostic abilities, but endomyocardial biopsy remains the gold standard. However, current treatment strategies are still focused on routine heart failure pharmacotherapies and supportive care or cardiac transplantation/mechanical support for those with end-stage heart failure.
Collapse
|
21
|
Zhang X, Gao X, Hu J, Xie Y, Zuo Y, Xu H, Zhu S. ADAR1p150 Forms a Complex with Dicer to Promote miRNA-222 Activity and Regulate PTEN Expression in CVB3-Induced Viral Myocarditis. Int J Mol Sci 2019; 20:ijms20020407. [PMID: 30669342 PMCID: PMC6359435 DOI: 10.3390/ijms20020407] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 02/06/2023] Open
Abstract
Adenosine deaminases acting on RNA (ADAR) are enzymes that regulate RNA metabolism through post-transcriptional mechanisms. ADAR1 is involved in a variety of pathological conditions including inflammation, cancer, and the host defense against viral infections. However, the role of ADAR1p150 in vascular disease remains unclear. In this study, we examined the expression of ADAR1p150 and its role in viral myocarditis (VMC) in a mouse model. VMC mouse cardiomyocytes showed significantly higher expression of ADAR1p150 compared to the control samples. Coimmunoprecipitation verified that ADAR1p150 forms a complex with Dicer in VMC. miRNA-222, which is involved in many cardiac diseases, is highly expressed in cardiomyocytes in VMC. In addition, the expression of miRNA-222 was promoted by ADAR1p150/Dicer. Among the target genes of miRNA-222, the expression of phosphatase-and-tensin (PTEN) protein was significantly reduced in VMC. By using a bioinformatics tool, we found a potential binding site of miRNA-222 on the PTEN gene’s 3′-UTR, suggesting that miRNA-222 might play a regulatory role. In cultured cells, miR-222 suppressed PTEN expression. Our findings suggest that ADAR1p150 plays a key role in complexing with Dicer and promoting the expression of miRNA-222, the latter of which suppresses the expression of the target gene PTEN during VMC. Our work reveals a previously unknown role of ADAR1p150 in gene expression in VMC.
Collapse
Affiliation(s)
- Xincai Zhang
- Institute of Forensic Medicine, Soochow University, Suzhou 215021, China.
| | - Xiangting Gao
- Department of Pathology, School of Medicine, Shihezi University, Shihezi 215021, China.
| | - Jun Hu
- Institute of Forensic Medicine, Soochow University, Suzhou 215021, China.
| | - Yuxin Xie
- Institute of Forensic Medicine, Soochow University, Suzhou 215021, China.
| | - Yuanyi Zuo
- Institute of Forensic Medicine, Soochow University, Suzhou 215021, China.
| | - Hongfei Xu
- Institute of Forensic Medicine, Soochow University, Suzhou 215021, China.
| | - Shaohua Zhu
- Institute of Forensic Medicine, Soochow University, Suzhou 215021, China.
| |
Collapse
|
22
|
Chen L, Lei L, Li T, Yan J, Jiang J. A20 alleviates the vascular remodeling induced by homocysteine. Am J Transl Res 2018; 10:3991-4003. [PMID: 30662645 PMCID: PMC6325502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 10/22/2018] [Indexed: 06/09/2023]
Abstract
Hyperhomocysteinemia is an independent risk factor for multiple cardiovascular diseases. The pathogenesis of homocysteine-induced vascular remodeling has not yet been elucidated. In vivo, we established hyperhomocysteinemia model with high L-methionine diet and found that the accumulation of macrophages, proliferation of VSMCs and decreased expression of A20 in the aorta of mice fed with high methionine diet. In vitro, we found that the overexpression of A20 suppressed the nuclear translocation of NF-kappaB p65 and attenuated homocysteine-induced proliferation and migration of VSMCs. However, down-regulation of A20 reversed the protective effects above. Moreover, A20 attenuated homocysteine-induced vascular remodeling by alleviating the activation of inflammation and suppressing the proliferation and migration of VSMCs through enhanced nuclear translocation of IRF3 and binding to PPAR-γ.
Collapse
Affiliation(s)
- Liang Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Lei Lei
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Tianyu Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| | - Jiangang Jiang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and TechnologyWuhan 430030, Hubei Province, China
| |
Collapse
|
23
|
Zhang B, Chen X, Yue H, Ruan W, Qin S, Tang C. Transcriptomic analysis reveals that enterovirus F strain SWUN-AB001 infection activates JNK/SAPK and p38 MAPK signaling pathways in MDBK cells. BMC Vet Res 2018; 14:395. [PMID: 30545363 PMCID: PMC6293526 DOI: 10.1186/s12917-018-1721-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Background Enteroviruses (Picornaviridae family) have been widely detected in the feces from cattle with diarrhea. However, the mechanisms responsible for the pathogenicity of enteroviruses in cattle remain unclear. Recently, we isolated a novel EV-F7 strain called SWUN-AB001 from diarrheal yak (Bos grunniens) feces. To explore the pathogenic mechanisms of this novel virus, we used a transcriptomics approach to find genes with differential expression patterns in Madin-Darby bovine kidney (MDBK) cells during infection with SWUN-AB001 over time. Results MDBK cells were sampled at 12 and 24 h post-infection (hpi) to represent the early and late stages of a SWUN-AB001 infection. Compared with the non-infected cells, 19 and 1050 differentially expressed genes (DEGs) were identified at 12 and 24 hpi, respectively. These DEGs were associated with disease, signal transduction, cellular process and cytokine signaling categories. At 24 hpi, the pathway enrichment analysis revealed that signal pathways such as c-Jun NH2-terminal kinase/ stress-activated protein kinase (JNK/SAPK) and mitogen-activated protein kinase (MAPK) pathways and cytokine-cytokine receptor interactions were associated with the interactions occurring between EV-F7 and MDBK cells. Our additional western blot analysis showed that the phosphorylation levels of JNK/SAPK and p38 MAPK proteins increased significantly in the MDBK cells at 24 hpi. The result indicated that infection with EV-F7 could activate JNK/SAPK and p38 MAPK pathways in MDBK cells, and possibly trigger large-scale cytokine production. Conclusion Our transcriptome analysis provides useful initial data towards better understanding of the infection mechanisms used by EV-F7, while highlighting the potential molecular relationships occurring between the virus and the host’s cellular components. Electronic supplementary material The online version of this article (10.1186/s12917-018-1721-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bin Zhang
- College of Life Science and Technology, Southwest Minzu University, No.16, South 4th Section 1st Ring Road, Chengdu, 610041, China. .,Key laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China. .,Animal Disease Prevention and Control Innovation Team in the Qinghai-Tibetan Plateau of State Ethnic Affairs Commission, Chengdu, 610041, China.
| | - Xinnuo Chen
- College of Life Science and Technology, Southwest Minzu University, No.16, South 4th Section 1st Ring Road, Chengdu, 610041, China
| | - Hua Yue
- College of Life Science and Technology, Southwest Minzu University, No.16, South 4th Section 1st Ring Road, Chengdu, 610041, China.,Key laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China.,Animal Disease Prevention and Control Innovation Team in the Qinghai-Tibetan Plateau of State Ethnic Affairs Commission, Chengdu, 610041, China
| | - Wenqiang Ruan
- College of Life Science and Technology, Southwest Minzu University, No.16, South 4th Section 1st Ring Road, Chengdu, 610041, China
| | - Sinan Qin
- College of Life Science and Technology, Southwest Minzu University, No.16, South 4th Section 1st Ring Road, Chengdu, 610041, China
| | - Cheng Tang
- College of Life Science and Technology, Southwest Minzu University, No.16, South 4th Section 1st Ring Road, Chengdu, 610041, China. .,Key laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China. .,Animal Disease Prevention and Control Innovation Team in the Qinghai-Tibetan Plateau of State Ethnic Affairs Commission, Chengdu, 610041, China.
| |
Collapse
|
24
|
Remels AHV, Derks WJA, Cillero-Pastor B, Verhees KJP, Kelders MC, Heggermont W, Carai P, Summer G, Ellis SR, de Theije CC, Heeren RMA, Heymans S, Papageorgiou AP, van Bilsen M. NF-κB-mediated metabolic remodelling in the inflamed heart in acute viral myocarditis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2579-2589. [PMID: 29730342 DOI: 10.1016/j.bbadis.2018.04.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/29/2018] [Accepted: 04/28/2018] [Indexed: 11/28/2022]
Abstract
Acute viral myocarditis (VM), characterised by leukocyte infiltration and dysfunction of the heart, is an important cause of sudden cardiac death in young adults. Unfortunately, to date, the pathological mechanisms underlying cardiac failure in VM remain incompletely understood. In the current study, we investigated if acute VM leads to cardiac metabolic rewiring and if this process is driven by local inflammation. Transcriptomic analysis of cardiac biopsies from myocarditis patients and a mouse model of VM revealed prominent reductions in the expression of a multitude of genes involved in mitochondrial oxidative energy metabolism. In mice, this coincided with reductions in high-energy phosphate and NAD levels, as determined by Imaging Mass Spectrometry, as well as marked decreases in the activity, protein abundance and mRNA levels of various enzymes and key regulators of cardiac oxidative metabolism. Indicative of fulminant cardiac inflammation, NF-κB signalling and inflammatory cytokine expression were potently induced in the heart during human and mouse VM. In cultured cardiomyocytes, cytokine-mediated NF-κB activation impaired cardiomyocyte oxidative gene expression, likely by interfering with the PGC-1 (peroxisome proliferator-activated receptor (PPAR)-γ co-activator) signalling network, the key regulatory pathway controlling cardiomyocyte oxidative metabolism. In conclusion, we provide evidence that acute VM is associated with extensive cardiac metabolic remodelling and our data support a mechanism whereby cytokines secreted primarily from infiltrating leukocytes activate NF-κB signalling in cardiomyocytes thereby inhibiting the transcriptional activity of the PGC-1 network and consequently modulating myocardial energy metabolism.
Collapse
Affiliation(s)
- Alexander H V Remels
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands.
| | - Wouter J A Derks
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Berta Cillero-Pastor
- The Maastricht Multimodal Molecular Imaging institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | - Koen J P Verhees
- Department of Respiratory Medicine, NUTRIM, Maastricht University, Maastricht, The Netherlands
| | - Marco C Kelders
- Department of Respiratory Medicine, NUTRIM, Maastricht University, Maastricht, The Netherlands
| | - Ward Heggermont
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Paolo Carai
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Georg Summer
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; TNO, Microbiology & Systems Biology, Zeist, The Netherlands
| | - Shane R Ellis
- The Maastricht Multimodal Molecular Imaging institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | - Chiel C de Theije
- Department of Respiratory Medicine, NUTRIM, Maastricht University, Maastricht, The Netherlands
| | - Ron M A Heeren
- The Maastricht Multimodal Molecular Imaging institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Ana P Papageorgiou
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Marc van Bilsen
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Department of Physiology, CARIM, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
25
|
Abdullah M, Berthiaume JM, Willis MS. Tumor necrosis factor receptor-associated factor 6 as a nuclear factor kappa B-modulating therapeutic target in cardiovascular diseases: at the heart of it all. Transl Res 2018; 195:48-61. [PMID: 29175266 PMCID: PMC5898986 DOI: 10.1016/j.trsl.2017.10.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/17/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023]
Abstract
Inflammatory and immune signaling has been documented as a root cause of many cardiovascular pathologies. In this review, we explore the emerging role of tumor necrosis factor receptor-associated factor 6 (TRAF6)-nuclear factor kappa B (NF-κB) signaling axis in atherosclerosis, ischemic heart disease, pathologic cardiac hypertrophy or heart failure, myocarditis, and sepsis-induced cardiomyopathy. We discuss the current understanding of cardiac inflammation in heart disease, present the TRAF6 signaling axis in the heart, then summarize what is known about TRAF6 in pathophysiology of heart disease including proof-of-concept studies that identify the utility of blocking TRAF6 to attenuate cardiac dysfunction, which suggests that TRAF6 is a novel, druggable target in treating cardiovascular disease incurred by inflammatory processes.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Department of Biochemistry, QuaidiAzam University, Islamabad, Pakistan; Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC
| | - Jessica M Berthiaume
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - Monte S Willis
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC; Department of Pharmacology, University of North Carolina, Chapel Hill, NC.
| |
Collapse
|
26
|
Wang X, Ai L, Xu Q, Wu C, Chen Z, Su D, Jiang X, Fan Z. A20 Attenuates Liver Fibrosis in NAFLD and Inhibits Inflammation Responses. Inflammation 2018; 40:840-848. [PMID: 28251449 DOI: 10.1007/s10753-017-0528-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We previously reported A20 was able to inhibit lipid accumulation in nonalcoholic steatohepatitis. We want to investigate whether A20 influences liver fibrosis in this study. Liver tissues from patients with hepatic fibrosis (n = 9) and healthy individuals (n = 7) were studied for A20 protein level by immunohistochemistry. A20 messenger RNA (mRNA) and protein level were also analyzed in two murine hepatic fibrosis models: methionine- and choline-deficient (MCD) diet and extrahepatic bile duct ligation (BDL) operation by real-time PCR and western blot. In vitro, the LX-2 human hepatic stellate cell line was treated by LPS at 0, 0.001, 0.01, 0.1, and 1 μg/mL for 6 h or at the concentration of 0.1 μg/mL for 0, 6, 12, and 24 h, then A20 expression levels were detected by western blot and PCR. The mRNA level of α-SMA, collagen I, collagen III, TGF-β, IL-6, MCP-1, and TLR4 was also examined by PCR. We then overexpressed A20 in LX-2 cells using adenovirus technique. Levels of α-SMA, collagen I, collagen III, TGF-β, IL-6, MCP-1, and TLR4 were examined in A20-overexpression LX-2 cells. Patients with hepatic fibrosis showed significantly higher A20 protein level compared with healthy controls. A20 mRNA and protein levels were also increased in livers from MCD feeding or BDL operation mice in comparison to normal controls. In LX-2 cells, LPS induced A20 protein in a concentration-dependent manner. The mRNA levels of α-SMA, collagen I, collagen III, TGF-β, IL-6, MCP-1, and TLR4 were increased after LPS treatment. Overexpression of A20 in LX-2 cells inhibited α-SMA deposition and collagen I, collagen III secretion. TGF-β, IL-6, MCP-1, and TLR4 mRNA levels were also reduced in A20-overexpression LX-2 cells in response to LPS stimulation. A20 overexpression inhibits hepatic stellate cell activation, which could be the mechanism for high A20 expression protected livers from fibrosis. Enhancement of A20 expression seems to be rational therapeutic strategies for liver fibrosis.
Collapse
Affiliation(s)
- Xiaohan Wang
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.,Department of Gastroenterology and Hepatology, The First Hospital of Jiaxing, 1882 Central-South Road, Jiaxing, Zhejiang Province, China
| | - Luoyan Ai
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.,Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Qingqing Xu
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.,Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Changwei Wu
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.,Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Zhiwei Chen
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.,Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Dazhi Su
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.,Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Xiaoke Jiang
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.,Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai, 200001, China
| | - Zhuping Fan
- Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160, Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
27
|
Green Tea Polyphenol Epigallocatechin-3-gallate-Alleviated Coxsackievirus B3-induced Myocarditis Through Inhibiting Viral Replication but Not Through Inhibiting Inflammatory Responses. J Cardiovasc Pharmacol 2017; 69:41-47. [PMID: 27753702 DOI: 10.1097/fjc.0000000000000439] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Viral myocarditis, which is mainly caused by coxsackievirus B3 (CVB3), affects about 5%-20% of the world population and still lacks efficient treatments. Green tea, a tonic and healthful beverage that was originated in ancient China, has been receiving considerable attention for its protective effect on cardiovascular diseases in recent years. In the present investigation, we aimed to explore the effect of green tea polyphenol epigallocatechin-3-gallate (EGCG) on CVB3-induced myocarditis and its underlying mechanism. Our study showed that EGCG could alleviate CVB3-induced myocarditis as evidenced by less cardiac injury and higher survival rate. Furthermore, we found that EGCG failed to downregulate the expression of inflammatory cytokines but could significantly inhibit the replication of CVB3. Furthermore, we found that EGCG treatment could downregulate the protein expression level of coxsackievirus and adenovirus receptor, the major receptor for CVB3 to infect cardiac myocytes. In conclusion, our data indicated that EGCG could ameliorate CVB3-induced myocarditis through inhibiting viral replication, which might provide a potential novel therapeutic strategy for viral myocarditis.
Collapse
|
28
|
Genome-Wide Analysis of mRNA and Long Noncoding RNA Profiles in Chronic Actinic Dermatitis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7479523. [PMID: 29359156 PMCID: PMC5735319 DOI: 10.1155/2017/7479523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/11/2017] [Accepted: 09/28/2017] [Indexed: 01/15/2023]
Abstract
Chronic actinic dermatitis (CAD), a photosensitive dermatosis, is characterized by inflammatory lesions, especially on sun-exposed skin. However, its pathogenesis remains unclear. In this study, second-generation RNA sequencing and comprehensive bioinformatics analyses of mRNAs and long noncoding RNAs (lncRNAs) were performed to determine the transcriptome profiles of patients with CAD. A total 6889 annotated lncRNAs, 341 novel lncRNAs, and 65091 mRNAs were identified. Interestingly, patients with CAD and healthy controls showed distinct transcriptome profiles. Indeed, 198 annotated (81.48%) and 45 novel (18.52%) lncRNAs were differentially expressed between the two groups. GO, KEGG, and RGSEA analyses of lncRNAs showed that inflammatory and immune response related pathways played crucial roles in the pathogenetic mechanism of CAD. In addition, we unveiled key differentially expressed lncRNAs, including lncRNA RP11-356I2.4 which plays a role probably by regulating TNFAIP3 and inflammation. qRT-PCR data validated the differentially expressed genes. The newly identified lncRNAs may have potential roles in the development of CAD; these findings lay a solid foundation for subsequent functional exploration of lncRNAs and mRNAs as therapeutic targets for CAD.
Collapse
|
29
|
Ursu ON, Beyer T, Sauter M, Fragasso A, Bundschuh S, Klingel K, Munz B. TRAF6: A player in CVB3-induced myocarditis? Cytokine 2017; 122:154143. [PMID: 28886971 DOI: 10.1016/j.cyto.2017.08.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/11/2017] [Accepted: 08/28/2017] [Indexed: 11/26/2022]
Abstract
Coxsackievirus B3 (CVB3) is an important inducer of myocarditis, which, in susceptible individuals, can chronify and eventually lead to the development of dilated cardiomyopathy and heart failure. The respective mechanisms are not completely understood. Here, we analyzed expression of the TRAF6 gene, encoding TNF receptor-associated factor 6 (TRAF6), a signal transduction scaffold protein that acts downstream of cytokine receptors, in heart tissue of susceptible and non-susceptible mouse strains. We found that after infection, TRAF6 expression was upregulated in both non-susceptible C57BL/6 wildtype and susceptible A.BY/SnJ and C57BL/6-TLR3 (-/-) mice, however, to different degrees. In infected HeLa cells, we also found moderately elevated TRAF6 levels after infection, in addition, activity of the transcription factor nuclear factor kappa B (NFκB), which can be activated downstream of TRAF6, was strongly enhanced in infected cells. To functionally analyze the role of TRAF6 with regard to infection progression, TRAF6 expression was knocked down in cultured HeLa cells using specific siRNAs. We found that reduction of TRAF6 expression had no effect on NFκB activation in response to infection. Taken together, our data suggest that CVB3 infection enhances TRAF6 levels, however, this induction might not be necessary for infection-induced NFκB activation.
Collapse
Affiliation(s)
- Oana N Ursu
- University Hospital Tübingen Medical Clinic, Department of Sports Medicine, Hoppe-Seyler-Str. 6, D-72076 Tübingen, Germany; University Hospital Tübingen, Department of Molecular Pathology, Institute for Pathology and Neuropathology, Liebermeisterstr. 8, D-72076 Tübingen, Germany
| | - Tina Beyer
- University Hospital Tübingen Medical Clinic, Department of Sports Medicine, Hoppe-Seyler-Str. 6, D-72076 Tübingen, Germany; University Hospital Tübingen, Department of Molecular Pathology, Institute for Pathology and Neuropathology, Liebermeisterstr. 8, D-72076 Tübingen, Germany
| | - Martina Sauter
- University Hospital Tübingen, Department of Molecular Pathology, Institute for Pathology and Neuropathology, Liebermeisterstr. 8, D-72076 Tübingen, Germany
| | - Annunziata Fragasso
- University Hospital Tübingen Medical Clinic, Department of Sports Medicine, Hoppe-Seyler-Str. 6, D-72076 Tübingen, Germany
| | - Sandra Bundschuh
- University Hospital Tübingen, Department of Molecular Pathology, Institute for Pathology and Neuropathology, Liebermeisterstr. 8, D-72076 Tübingen, Germany
| | - Karin Klingel
- University Hospital Tübingen, Department of Molecular Pathology, Institute for Pathology and Neuropathology, Liebermeisterstr. 8, D-72076 Tübingen, Germany
| | - Barbara Munz
- University Hospital Tübingen Medical Clinic, Department of Sports Medicine, Hoppe-Seyler-Str. 6, D-72076 Tübingen, Germany.
| |
Collapse
|
30
|
Müller I, Pappritz K, Savvatis K, Puhl K, Dong F, El-Shafeey M, Hamdani N, Hamann I, Noutsias M, Infante-Duarte C, Linke WA, Van Linthout S, Tschöpe C. CX3CR1 knockout aggravates Coxsackievirus B3-induced myocarditis. PLoS One 2017; 12:e0182643. [PMID: 28800592 PMCID: PMC5553786 DOI: 10.1371/journal.pone.0182643] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/22/2017] [Indexed: 11/19/2022] Open
Abstract
Studies on inflammatory disorders elucidated the pivotal role of the CX3CL1/CX3CR1 axis with respect to the pathophysiology and diseases progression. Coxsackievirus B3 (CVB3)-induced myocarditis is associated with severe cardiac inflammation, which may progress to heart failure. We therefore investigated the influence of CX3CR1 ablation in the model of acute myocarditis, which was induced by inoculation with 5x105 plaque forming units of CVB3 (Nancy strain) in either CX3CR1-/- or C57BL6/j (WT) mice. Seven days after infection, myocardial inflammation, remodeling, and titin expression and phosphorylation were examined by immunohistochemistry, real-time PCR and Pro-Q diamond stain. Cardiac function was assessed by tip catheter. Compared to WT CVB3 mice, CX3CR1-/- CVB3 mice exhibited enhanced left ventricular expression of inflammatory cytokines and chemokines, which was associated with an increase of immune cell infiltration/presence. This shift towards a pro-inflammatory immune response further resulted in increased cardiac fibrosis and cardiomyocyte apoptosis, which was reflected by an impaired cardiac function in CX3CR1-/- CVB3 compared to WT CVB3 mice. These findings demonstrate a cardioprotective role of CX3CR1 in CVB3-infected mice and indicate the relevance of the CX3CL1/CX3CR1 system in CVB3-induced myocarditis.
Collapse
MESH Headings
- Animals
- Apoptosis
- CX3C Chemokine Receptor 1
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Chemokine CX3CL1/genetics
- Chemokine CX3CL1/immunology
- Coxsackievirus Infections/genetics
- Coxsackievirus Infections/immunology
- Coxsackievirus Infections/pathology
- Coxsackievirus Infections/virology
- Disease Models, Animal
- Enterovirus B, Human/growth & development
- Enterovirus B, Human/pathogenicity
- Gene Expression Regulation
- Heart Function Tests
- Host-Pathogen Interactions/immunology
- Humans
- Interleukins/genetics
- Interleukins/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocarditis/genetics
- Myocarditis/immunology
- Myocarditis/pathology
- Myocarditis/virology
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/pathology
- Phosphorylation
- Protein Kinases/genetics
- Protein Kinases/immunology
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/immunology
Collapse
Affiliation(s)
- Irene Müller
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Klinikum, Berlin, Germany
| | - Kathleen Pappritz
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Klinikum, Berlin, Germany
| | - Konstantinos Savvatis
- Inherited Cardiovascular Diseases Unit, Barts Health NHS Trust, Barts Heart Centre, London, United Kingdom
- William Harvey Research Institute, Queen Mary University London, London, United Kingdom
| | - Kerstin Puhl
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Berlin, Germany
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Klinikum, Berlin, Germany
| | - Fengquan Dong
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Berlin, Germany
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Klinikum, Berlin, Germany
| | - Muhammad El-Shafeey
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Berlin, Germany
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Klinikum, Berlin, Germany
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Isabell Hamann
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Berlin, Germany
| | - Michel Noutsias
- Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Halle (Saale), Germany
| | - Carmen Infante-Duarte
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Berlin, Germany
| | - Wolfgang A. Linke
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Sophie Van Linthout
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Klinikum, Berlin, Germany
| | - Carsten Tschöpe
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Klinikum, Berlin, Germany
- * E-mail:
| |
Collapse
|
31
|
Li XQ, Liu XX, Wang XY, Xie YH, Yang Q, Liu XX, Ding YY, Cao W, Wang SW. Cinnamaldehyde Derivatives Inhibit Coxsackievirus B3-Induced Viral Myocarditis. Biomol Ther (Seoul) 2017; 25:279-287. [PMID: 27737525 PMCID: PMC5424638 DOI: 10.4062/biomolther.2016.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/15/2016] [Accepted: 06/13/2016] [Indexed: 12/30/2022] Open
Abstract
The chemical property of cinnamaldehyde is unstable in vivo, although early experiments have shown its obvious therapeutic effects on viral myocarditis (VMC). To overcome this problem, we used cinnamaldehyde as a leading compound to synthesize derivatives. Five derivatives of cinnamaldehyde were synthesized: 4-methylcinnamaldehyde (1), 4-chlorocinnamaldehyde (2), 4-methoxycinnamaldehyde (3), α-bromo-4-methylcinnamaldehyde (4), and α-bromo-4-chlorocinnamaldehyde (5). Neonatal rat cardiomyocytes and HeLa cells infected by coxsackievirus B3 (CVB3) were used to evaluate their antiviral and cytotoxic effects. In vivo BALB/c mice were infected with CVB3 for establishing VMC models. Among the derivatives, compound 4 and 5 inhibited the CVB3 in HeLa cells with the half-maximal inhibitory concentrations values of 11.38 ± 2.22 μM and 2.12 ± 0.37 μM, respectively. The 50% toxic concentrations of compound 4 and 5-treated cells were 39-fold and 87-fold higher than in the cinnamaldehyde group. Compound 4 and 5 effectively reduced the viral titers and cardiac pathological changes in a dose-dependent manner. In addition, compound 4 and 5 significantly inhibited the secretion, mRNA and protein expressions of inflammatory cytokines TNF-α, IL-1β and IL-6 in CVB3-infected cardiomyocytes, indicating that brominated cinnamaldehyde not only improved the anti-vital activities for VMC, but also had potent anti-inflammatory effects in cardiomyocytes induced by CVB3.
Collapse
Affiliation(s)
- Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiao-Xiao Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xue-Ying Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yan-Hua Xie
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Qian Yang
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xin-Xin Liu
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan-Yuan Ding
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Cao
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Si-Wang Wang
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
32
|
Wang C, Dong C, Xiong S. IL-33 enhances macrophage M2 polarization and protects mice from CVB3-induced viral myocarditis. J Mol Cell Cardiol 2016; 103:22-30. [PMID: 28041873 DOI: 10.1016/j.yjmcc.2016.12.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 01/13/2023]
Abstract
Viral myocarditis is the inflammation caused by myocardial virus infection, and the coxsackievirus group B3 virus (CVB3) is the most common pathogen. An efficient therapeutic agent against viral myocarditis is currently unavailable. IL-33, a new member of the IL-1 cytokine superfamily, exhibits potential immunotherapeutic effect against inflammatory and autoimmune diseases. However, the functional role of IL-33 in viral myocarditis has not been investigated. To examine the therapeutic role of IL-33 in viral myocarditis, an IL-33 overexpression plasmid (pDisplay-IL-33) and IL-33 knockdown plasmid (pLL3.7-IL-33) were packaged with polyethylenimine and delivered intravenously at the orbital area of BALB/c male mice after CVB3 infection. Then, myocarditis severity was assessed 7days after infection. Results showed that IL-33 up-regulation significantly alleviated the severity of viral myocarditis with an increased cardiac contractive function and survival rate. Mechanistic studies demonstrated that IL-33 can stimulate ST2L+F4/80+ macrophages and ST2L+CD4+T cells in cardiac tissue to express IL-4, which is a potent inducer for macrophage M2 polarization. Mice with adoptive transfer of M2 macrophages exhibited less cardiac inflammation and attenuated myocarditis, suggesting the protective role of M2 macrophage in viral myocarditis. Additionally, IL-4 neutralization abolished the IL-33-mediated cardiac functional improvement in myocarditis mice. Collectively, our findings provide a novel therapeutic role for IL-33 in CVB3-induced myocarditis.
Collapse
Affiliation(s)
- Chao Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China
| | - Chunsheng Dong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China.
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
33
|
Tang Q, Lu M, Zhou H, Chen D, Liu L. Gambogic acid inhibits the growth of ovarian cancer tumors by regulating p65 activity. Oncol Lett 2016; 13:384-388. [PMID: 28123571 DOI: 10.3892/ol.2016.5433] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 10/19/2016] [Indexed: 12/26/2022] Open
Abstract
Ovarian cancer patients often have poor prognosis, therefore, it is important to search for more effective therapeutic strategies to treat them. Gambogic acid (GA) exhibits an anti-tumor effect through various mechanisms, and has multiple targets in tumor cells. The present study aimed to elucidate the efficacy of GA in the treatment of ovarian cancer both in vivo and in vitro by analyzing its impact on cell survival and tumor growth through cell cycle and apoptosis analysis. GA inhibited the growth of ovarian cancer cells in a dose and time dependent manner, and arrested the cell cycle in ovarian cancer cells. Furthermore, GA increased caspase-3 and caspase-9 activity and inhibited RELA/NF-κB p65 (p65) DNA binding activity. Finally, GA suppressed tumor growth in vivo. Therefore, the current study suggests that GA inhibits the growth of ovarian cancer by regulating p65 activity, and may be developed as a novel therapeutic strategy to treat ovarian cancer.
Collapse
Affiliation(s)
- Qiusha Tang
- Department of Pathology and Pathophysiology, Medical College, Southeast University, Nanjing, Jiangsu 210096, P.R. China
| | - Mudan Lu
- Genetic Laboratory, Wuxi Hospital for Maternal and Child Health Care, The Affiliated Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| | - Huan Zhou
- Department of Pathology and Pathophysiology, Medical College, Southeast University, Nanjing, Jiangsu 210096, P.R. China
| | - Daozhen Chen
- Genetic Laboratory, Wuxi Hospital for Maternal and Child Health Care, The Affiliated Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| | - Lu Liu
- Department of Pathology and Pathophysiology, Medical College, Southeast University, Nanjing, Jiangsu 210096, P.R. China
| |
Collapse
|
34
|
Janus effects of ADAR1 on CVB3-induced viral myocarditis at different infection stages. Int J Cardiol 2016; 223:898-905. [DOI: 10.1016/j.ijcard.2016.08.315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 01/05/2023]
|
35
|
Lu M, Xu W, Gao B, Xiong S. Blunting Autoantigen-induced FOXO3a Protein Phosphorylation and Degradation Is a Novel Pathway of Glucocorticoids for the Treatment of Systemic Lupus Erythematosus. J Biol Chem 2016; 291:19900-12. [PMID: 27481940 DOI: 10.1074/jbc.m116.728840] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Indexed: 11/06/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease affecting multiple organs. Glucocorticoids (GCs), the potent anti-inflammatory drugs, remain as a cornerstone in the treatment for SLE; nevertheless, their clinical efficacy is compromised by the side effects of long term treatment and resistance. To improve the therapeutic efficacy of GCs in SLE, it is important to further decipher the molecular mechanisms of how GCs exert their anti-inflammatory effects. In this investigation, FOXO3a was identified as a molecule that was down-regulated in the course of SLE. Of interest, GC treatment was found to rescue FOXO3a expression both in SLE mice and in SLE patients. Gain- and loss-of-function studies demonstrated that FOXO3a played a crucial role in GC treatment of SLE via inhibiting inflammatory responses. Further studies showed that the up-regulation of FOXO3a by GCs relied on the suppression of pI3K/AKT-mediated FOXO3a phosphorylation and the arrest of FOXO3a in the nucleus. Finally, our data revealed that FOXO3a was critical for GC-mediated inhibition of NF-κB activity, which might involve its interaction with NF-κB p65 protein. Collectively, these data indicated that FOXO3a played an important role in GC treatment of SLE by suppressing pro-inflammatory response, and targeting FOXO3a might provide a novel therapeutic strategy against SLE.
Collapse
Affiliation(s)
- Mudan Lu
- From the Institute for Immunobiology, Department of Immunology, Shanghai Medical College of Fudan University, Shanghai 200032 and
| | - Wei Xu
- the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215006, China
| | - Bo Gao
- From the Institute for Immunobiology, Department of Immunology, Shanghai Medical College of Fudan University, Shanghai 200032 and
| | - Sidong Xiong
- From the Institute for Immunobiology, Department of Immunology, Shanghai Medical College of Fudan University, Shanghai 200032 and the Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215006, China
| |
Collapse
|
36
|
Hong JY, Bae WJ, Yi JK, Kim GT, Kim EC. Anti-inflammatory and anti-osteoclastogenic effects of zinc finger protein A20 overexpression in human periodontal ligament cells. J Periodontal Res 2015; 51:529-39. [PMID: 26548452 DOI: 10.1111/jre.12332] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND OBJECTIVE Although overexpression of the nuclear factor κB inhibitory and ubiquitin-editing enzyme A20 is thought to be involved in the pathogenesis of inflammatory diseases, its function in periodontal disease remains unknown. The aims of the present study were to evaluate A20 expression in patients with periodontitis and to study the effects of A20 overexpression, using a recombinant adenovirus encoding A20 (Ad-A20), on the inflammatory response and on osteoclastic differentiation in lipopolysaccharide (LPS)- and nicotine-stimulated human periodontal ligament cells (hPDLCs). MATERIAL AND METHODS The concentration of prostaglandin E2 was measured by radioimmunoassay. Reverse transcription-polymerase chain reactions and western blot analyses were used to measure mRNA and protein levels, respectively. Osteoclastic differentiation was assessed in mouse bone marrow-derived macrophages using conditioned medium from LPS- and nicotine-treated hPDLCs. RESULTS A20 was upregulated in the gingival tissues and neutrophils from patients with periodontitis and in LPS- and nicotine-exposed hPDLCs. Pretreatment with A20 overexpression by Ad-A20 markedly attenuated LPS- and nicotine-induced production of prostaglandin E2 , as well as expression of cyclooxygenase-2 and proinflammatory cytokines. Moreover, A20 overexpression inhibited the number and size of tartrate-resistant acid phosphatase-stained osteoclasts, and downregulated osteoclast-specific gene expression. LPS- and nicotine-induced p38 phosphorylation and nuclear factor κB activation were blocked by Ad-A20. Ad-A20 inhibited the effects of nicotine and LPS on the activation of pan-protein kinase C, Akt, GSK-3β and protein kinase Cα. CONCLUSIONS This study is the first to demonstrate that A20 overexpression has anti-inflammatory effects and blocks osteoclastic differentiation in a nicotine- and LPS-stimulated hPDLC model. Thus, A20 overexpression may be a potential therapeutic target in inflammatory bone loss diseases, such as periodontal disease.
Collapse
Affiliation(s)
- J-Y Hong
- Department of Periodontology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - W-J Bae
- Department of Oral and Maxillofacial Pathology & Research Center for Tooth and Periodontal Tissue Regeneration (MRC), School of Dentistry, Kyung Hee University, Seoul, Korea
| | - J-K Yi
- Department of Conservative Dentistry, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - G-T Kim
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - E-C Kim
- Department of Oral and Maxillofacial Pathology & Research Center for Tooth and Periodontal Tissue Regeneration (MRC), School of Dentistry, Kyung Hee University, Seoul, Korea
| |
Collapse
|
37
|
Sun S, Ma J, Zhang Q, Wang Q, Zhou L, Bai F, Hu H, Chang P, Yu J, Gao B. Argonaute proteins in cardiac tissue contribute to the heart injury during viral myocarditis. Cardiovasc Pathol 2015; 25:120-6. [PMID: 26764146 DOI: 10.1016/j.carpath.2015.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/10/2015] [Accepted: 10/13/2015] [Indexed: 10/22/2022] Open
Abstract
MicroRNAs (miRNAs) are a group of short, noncoding, regulatory RNA molecules the dysregulation of which contributes to the pathogenesis of myocarditis. Argonaute proteins are essential components of miRNA-induced silencing complex and play important roles during miRNA biogenesis and function. However, the expression pattern of four AGO family members has not yet been detected in the coxsackievirus B3 (CVB3)-induced myocarditis tissue samples. In this study, we detected the expression of four AGOs in the CVB3-infected mouse heart tissues and found that AGO1 and AGO3 up-regulated significantly at 4 and 8h after CVB3 infection. Further in vitro research indicated that up-regulated AGO1 and AGO3 are related to the down-regulated TNFAIP3, which is a negative regulator of NF-κB pathway. Subsequently, we confirmed that TNFAIP3 is a direct target of miR-19a/b, and during CVB3 infection, the expression of miR-19a/b and miR-125a/b is not significantly changed. TNFAIP3 level is mainly reduced by up-regulated AGO1 and AGO3. This research sheds light on the relationship between overexpressed AGO proteins and CVB3-induced myocarditis, and this provides potential therapeutic target for viral myocarditis.
Collapse
Affiliation(s)
- Shougang Sun
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Jialiang Ma
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Quan Zhang
- Department of Cardiology, Pingliang First People's Hospital, Pingliang, 744000, China
| | - Qiongying Wang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Lei Zhou
- Department of Nephrology, the 306th Hospital of the Chinese PLA, Beijing 100101, China
| | - Feng Bai
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Hao Hu
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Peng Chang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Jing Yu
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Bingren Gao
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
38
|
Li M, Shi X, Qian T, Li J, Tian Z, Ni B, Hao F. A20 overexpression alleviates pristine-induced lupus nephritis by inhibiting the NF-κB and NLRP3 inflammasome activation in macrophages of mice. Int J Clin Exp Med 2015; 8:17430-17440. [PMID: 26770333 PMCID: PMC4694233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/13/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Lupus nephritis is an autoimmune inflammatory disease and urgently needs effective anti-inflammation therapies. A20, tumor necrosis factor alpha induced protein 3 (TNFAIP3), is a key negative regulator of inflammation, however whether A20 can regulate lupus nephritis has not been clarified. This study aimed at investigating the potential therapeutic effect of A20 on renal inflammation in mouse pristine model oflupus. METHODOLOGY/PRINCIPAL FINDINGS Female BALB/c mice were intraperitoneally injected with pristine to establish lupus renal injury. The levels of serum IL-1β, IL-6 and autoantibodies and the degrees of renal injury and CCL2 and F4/80 levels were measured. The levels of the NF-κB and NLRP3 inflammasome activation in peritoneal macrophages were determined. We found that injection with pristine increased the levels of serum IL-1β, IL-6, autoantibodies and CCL20 and F4/80 expression in the kidney and induced renal injury, accompanied by enhancing the NF-κB and NLRP3 inflammasome activation in macrophages of mice. In contrast, treatment with Ad-A20, but not with Ad-control, significantly mitigated pristine-induced inflammatory responses and renal injury,and reduced the NF-κB and NLRP3 inflammasome activation in macrophages in mice. CONCLUSION/SIGNIFICANCE Our data indicated that induction of A20 overexpression inhibited pristane induced lupus inflammation and renal injury in mice and may be a new therapeutic strategy for treatment of lupus nephritis.
Collapse
Affiliation(s)
- Min Li
- Department of Dermatology, Southwest Hospital, Third Military Medical UniversityChongqing, 400038, China
| | - Xiaowei Shi
- Department of Dermatology, Southwest Hospital, Third Military Medical UniversityChongqing, 400038, China
| | - Tian Qian
- Department of Dermatology, Southwest Hospital, Third Military Medical UniversityChongqing, 400038, China
| | - Jian Li
- Department of Dermatology, Southwest Hospital, Third Military Medical UniversityChongqing, 400038, China
| | - Zhiqiang Tian
- Institute of Immunology PLA, Third Military Medical UniversityChongqing 400038, China
| | - Bing Ni
- Institute of Immunology PLA, Third Military Medical UniversityChongqing 400038, China
| | - Fei Hao
- Department of Dermatology, Southwest Hospital, Third Military Medical UniversityChongqing, 400038, China
| |
Collapse
|
39
|
In vitro interaction between coxsackievirus B3 VP1 protein and human pleckstrin homology domain retinal protein (PHR1). Virus Genes 2015; 51:182-9. [PMID: 26318175 DOI: 10.1007/s11262-015-1241-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/20/2015] [Indexed: 12/11/2022]
Abstract
Coxsackievirus B3 (CVB3) infection causes central nervous system diseases including aseptic meningitis and encephalitis. To understand the mechanism of this virus, a yeast two-hybrid system was used to screen cellular proteins from a human heart cDNA library. The results revealed that the human Pleckstrin Homology Domain Retinal protein (PHR1), a PH domain-containing protein with low expression in the heart and high expression in the brain, interacts with CVB3 VP1, a major structural protein of CVB3. Yeast mating assays and in vitro coimmunoprecipitation verified the interaction between CVB3 VP1 and PHR1. An α-galactosidase assay indicated that of α-galactosidase activity was higher in positive clones than in controls suggesting a strong interaction. Furthermore, assay of deletion mutants defined the minimal region of PHR1 required for its interaction with VP1 as amino acids 95-172 and two regions of VP1 required for its interaction with PHR1 as amino acids 729-767 and 811-859. The results revealed multiple binding sites between PHR1 and CVB3 VP1 and suggested that the strong interaction between these two proteins might play an important role in central nervous system disease in the human brain.
Collapse
|
40
|
Inhibition of Histone Deacetylase Activity Aggravates Coxsackievirus B3-Induced Myocarditis by Promoting Viral Replication and Myocardial Apoptosis. J Virol 2015; 89:10512-23. [PMID: 26269170 PMCID: PMC4580191 DOI: 10.1128/jvi.01028-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/28/2015] [Indexed: 01/09/2023] Open
Abstract
Viral myocarditis, which is most prevalently caused by coxsackievirus B3 (CVB3), is a serious clinical condition characterized by excessive myocardial inflammation. Recent studies suggest that regulation of protein acetylation levels by inhibiting histone deacetylase (HDAC) activity modulates inflammatory response and shows promise as a therapy for several inflammatory diseases. However, the role of HDAC activity in viral myocarditis is still not fully understood. Here, we aim to investigate the role of HDAC activity in viral myocarditis and its underlying mechanism. CVB3-infected BALB/c mice were treated with the HDAC inhibitor (HDACI) suberoylanilide hydroxamic acid (SAHA) or trichostatin A (TSA). We found inhibition of HDAC activity aggravated rather than ameliorated the severity of CVB3-induced myocarditis, which was contrary to our expectations. The aggravated myocarditis by HDACI treatment seemed not to be caused by an elevated inflammatory response but by the increased CVB3 replication. Further, it was revealed that the increased CVB3 replication was closely associated with the HDACI-enhanced autophagosome formation. Inhibition of autophagosome formation by wortmannin or ATG5 short hairpin RNA dramatically suppressed the HDACI-increased CVB3 replication. The increased viral replication subsequently elevated CVB3-induced myocardial apoptosis. Conversely, inhibition of CVB3 replication and ensuing myocardial apoptosis by the antiviral drug ribavirin significantly reversed the HDACI-aggravated viral myocarditis. In conclusion, we elucidate that the inhibition of HDAC activity increases CVB3 replication and ensuing myocardial apoptosis, resulting in aggravated viral myocarditis. Possible adverse consequences of administering HDACI should be considered in patients infected (or coinfected) with CVB3. IMPORTANCE Viral myocarditis, which is most prevalently caused by CVB3, is characterized by excessive myocardial inflammation. Inhibition of HDAC activity was originally identified as a powerful anti-cancer therapeutic strategy and was recently found to be implicated in the regulation of inflammatory response. HDACI has been demonstrated to be efficacious in animal models of several inflammatory diseases. Thus, we hypothesize that inhibition of HDAC activity also protects against CVB3-induced viral myocarditis. Surprisingly, we found inhibition of HDAC activity enhanced myocardial autophagosome formation, which led to the elevated CVB3 viral replication and ensuing increased myocardial apoptosis. Viral myocarditis was eventually aggravated rather than ameliorated by HDAC inhibition. In conclusion, we elucidate the role of HDAC activity in viral myocarditis. Moreover, given the importance of HDACI in preclinical and clinical treatments, the possible unfavorable effect of HDACI should be carefully evaluated in patients infected with viruses, including CVB3.
Collapse
|
41
|
Papageorgiou AP, Heggermont W, Rienks M, Carai P, Langouche L, Verhesen W, De Boer RA, Heymans S. Liver X receptor activation enhances CVB3 viral replication during myocarditis by stimulating lipogenesis. Cardiovasc Res 2015; 107:78-88. [PMID: 25998987 DOI: 10.1093/cvr/cvv157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/13/2015] [Indexed: 12/16/2022] Open
Abstract
AIMS Viral myocarditis (VM) is severe cardiac inflammation that can result in sudden death or congestive heart failure in previously healthy adults, with no effective therapy. Liver X receptor (LXR) agonists have both anti-inflammatory and lipid-lowering properties. This study investigates whether LXR agonist T0901317 may modulate viral replication and cardiac inflammation during VM. METHODS AND RESULTS (i) Adult mice were administered T0901317 or vehicle with the onset of inflammation during CVB3 virus myocarditis or (ii) treated 2 days prior to CVB3 infection. Against what we expected, T0901317 treatment did not alter leucocyte infiltration after CVB3 infection; yet pre-administration with T0901317 resulted in increased mortality upon CVB3 infection, higher cardiac viral presence, and increased cardiomyocyte damage when compared with the vehicle. Furthermore, we show a correlation of fatty acid synthase (FAS) and sterol regulatory element-binding protein 1c (SREBP-1c) with CVB3 viral load in the heart and that T0901317 is able to enhance the cardiac expression of FAS and SREBP-1c. Finally, we show in vitro that T0901317 is able to exaggerate CVB3-mediated damage of Vero cells, whereas inhibitors of FAS and the SREBP-1c reduce the viral presence of CVB3 in neonatal cardiomyocytes. CONCLUSION LXR agonism does not modulate cardiac inflammation, but exacerbates virus-mediated myocardial damage during VM by stimulating lipid biosynthesis and enhancing CVB3 replication.
Collapse
Affiliation(s)
- Anna-Pia Papageorgiou
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Ward Heggermont
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Marieke Rienks
- CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Paolo Carai
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Lies Langouche
- Laboratory of Intensive Care Medicine, KU Leuven, Leuven, Belgium
| | - Wouter Verhesen
- CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Rudolf A De Boer
- University Medical Center, Groningen University, Groningen, The Netherlands
| | - Stephane Heymans
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands ICIN - Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
42
|
Yu JY, Zhang B, Peng L, Wu CH, Cao H, Zhong JF, Hoffman J, Huang SH. Repositioning of Memantine as a Potential Novel Therapeutic Agent against Meningitic E. coli-Induced Pathogenicities through Disease-Associated Alpha7 Cholinergic Pathway and RNA Sequencing-Based Transcriptome Analysis of Host Inflammatory Responses. PLoS One 2015; 10:e0121911. [PMID: 25993608 PMCID: PMC4437645 DOI: 10.1371/journal.pone.0121911] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/07/2015] [Indexed: 01/09/2023] Open
Abstract
Neonatal sepsis and meningitis (NSM) remains a leading cause worldwide of mortality and morbidity in newborn infants despite the availability of antibiotics over the last several decades. E. coli is the most common gram-negative pathogen causing NSM. Our previous studies show that α7 nicotinic receptor (α7 nAChR), an essential regulator of inflammation, plays a detrimental role in the host defense against NSM. Despite notable successes, there still exists an unmet need for new effective therapeutic approaches to treat this disease. Using the in vitro/in vivo models of the blood-brain barrier (BBB) and RNA-seq, we undertook a drug repositioning study to identify unknown antimicrobial activities for known drugs. We have demonstrated for the first time that memantine (MEM), a FDA-approved drug for treatment of Alzheimer’s disease, could very efficiently block E. coli-caused bacteremia and meningitis in a mouse model of NSM in a manner dependent on α7 nAChR. MEM was able to synergistically enhance the antibacterial activity of ampicillin in HBMEC infected with E. coli K1 (E44) and in neonatal mice with E44-caused bacteremia and meningitis. Differential gene expression analysis of RNA-Seq data from mouse BMEC infected with E. coli K1 showed that several E44-increased inflammatory factors, including IL33, IL18rap, MMP10 and Irs1, were significantly reduced by MEM compared to the infected cells without drug treatment. MEM could also significantly up-regulate anti-inflammatory factors, including Tnfaip3, CISH, Ptgds and Zfp36. Most interestingly, these factors may positively and negatively contribute to regulation of NF-κB, which is a hallmark feature of bacterial meningitis. Furthermore, we have demonstrated that circulating BMEC (cBMEC) are the potential novel biomarkers for NSM. MEM could significantly reduce E44-increased blood level of cBMEC in mice. Taken together, our data suggest that memantine can efficiently block host inflammatory responses to bacterial infection through modulation of both inflammatory and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Jing-Yi Yu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Bao Zhang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Liang Peng
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America; Department of Clinic Laboratory, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Chun-Hua Wu
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China
| | - John F Zhong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States of America; Department of Perio, Diagnostic Sciences & Biomedical Sciences, School of Dentistry, University of Southern California, Los Angeles, CA, 93003, United States of America; Department of Pediatrics, School of Medicine, University of Southern California, Los Angeles, CA, 93003, United States of America
| | - Jill Hoffman
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Sheng-He Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| |
Collapse
|
43
|
Li X, Yue Y, Zhu Y, Xiong S. Extracellular, but not intracellular HMGB1, facilitates self-DNA induced macrophage activation via promoting DNA accumulation in endosomes and contributes to the pathogenesis of lupus nephritis. Mol Immunol 2015; 65:177-88. [DOI: 10.1016/j.molimm.2015.01.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 01/18/2015] [Accepted: 01/19/2015] [Indexed: 01/01/2023]
|
44
|
Zha X, Yue Y, Dong N, Xiong S. Endoplasmic Reticulum Stress Aggravates Viral Myocarditis by Raising Inflammation Through the IRE1-Associated NF-κB Pathway. Can J Cardiol 2015; 31:1032-40. [PMID: 26111668 DOI: 10.1016/j.cjca.2015.03.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 02/20/2015] [Accepted: 03/03/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Viral myocarditis, which is mostly caused by coxsackievirus infection, is characterized by myocardial inflammation. Abnormal endoplasmic reticulum (ER) stress participates in many heart diseases, but its role in viral myocarditis remains unsolved. METHODS We investigated the influence of ER stress in coxsackievirus B3 (CVB3)-induced viral myocarditis by dynamically detecting its activation in CVB3-infected hearts, analyzing its association with myocarditis severity, and exploring its impact on disease development by modulating the strength of ER stress with the chemical activator tunicamycin (Tm) or the inhibitor tauroursodeoxycholic acid (TUDCA). The underlying signal pathway of ER stress in CVB3-induced myocarditis was also deciphered. RESULTS We found that myocardial expression of Grp78 and Grp94, 2 ER stress markers, was significantly increased after CVB3 infection and positively correlated with myocarditis severity. Consistently, Tm-augmented ER stress obviously aggravated myocarditis, as shown by more severe myocardial inflammation, reduced cardiac function, and a lower survival rate, whereas TUDCA decreased ER stress and obviously alleviated myocarditis. This pathologic effect of ER stress could be attributed to increased levels of proinflammatory cytokine (interleukin [IL]-6, IL-12, tumor necrosis factor-alpha, and monocyte chemoattractant protein-1) production through the IRE1-associated nuclear factor-κB (NF-kB) pathway. CONCLUSIONS ER stress accentuated CVB3-induced myocardial inflammation through the IRE1-associated NF-κB pathway. This study may help us understand the role of ER stress in viral myocarditis and promote the development of corresponding therapeutic strategies based on manipulating ER stress.
Collapse
Affiliation(s)
- Xi Zha
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, Suzhou, People's Republic of China
| | - Yan Yue
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, Suzhou, People's Republic of China
| | - Ning Dong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, Suzhou, People's Republic of China
| | - Sidong Xiong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, Suzhou, People's Republic of China.
| |
Collapse
|
45
|
Gui J, Chen R, Xu W, Xiong S. Remission of CVB3-induced myocarditis with Astragaloside IV treatment requires A20 (TNFAIP3) up-regulation. J Cell Mol Med 2015; 19:850-64. [PMID: 25728713 PMCID: PMC4395199 DOI: 10.1111/jcmm.12459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 09/22/2014] [Indexed: 12/17/2022] Open
Abstract
Viral myocarditis (VMC) most prevalently caused by coxsackievirus B3 (CVB3) infection is characterized by severe cardiac inflammation. Therapeutic options for the disease are still limited. Astragaloside IV (AST-IV), a purified small molecular saponin (C41H68O14, MW 784), is the main active component of Chinese medical herb Astragalus which has been empirically prescribed for the treatment of heart dysfunction for centuries. In this study, we investigated the effect of AST-IV on CVB3-induced myocarditis and explored its possible mechanism involved. The results showed that AST-IV administration alleviated the severity of myocarditis and attenuated cardiac inflammation, which was mediated by inhibition of nuclear factor-kappaB (NF-κB) signalling. Importantly, we further identified that the inhibitory effect of AST-IV on NF-κB signalling was through increasing A20 (TNFAIP3) expression. Moreover, we validated that A20 was critical for the therapeutic efficacy of AST-IV on CVB3-induced myocarditis. Finally, we revealed that AST-IV enhanced A20 expression at post-transcriptional level by stabilization of mRNA. Our findings uncover a previously unknown mechanism for AST-IV in the treatment of VMC because of modulating inflammatory response via increasing A20 expression, which provide a potential target for screening new drugs and are helpful for optimization of the therapeutic strategies for VMC.
Collapse
Affiliation(s)
- Jun Gui
- Institute for Immunobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
46
|
Wu Z, Peng H, Du Q, Lin W, Liu Y. GYY4137, a hydrogen sulfide‑releasing molecule, inhibits the inflammatory response by suppressing the activation of nuclear factor‑kappa B and mitogen‑activated protein kinases in Coxsackie virus B3‑infected rat cardiomyocytes. Mol Med Rep 2014; 11:1837-44. [PMID: 25377925 DOI: 10.3892/mmr.2014.2901] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 07/21/2014] [Indexed: 11/06/2022] Open
Abstract
GYY4137 is a water‑soluble, small molecule hydrogen sulfide (H2S)‑release agent that possesses potent cardioprotective and anti‑inflammatory properties in experimental models. Coxsackie virus B3 (CVB3) infection commonly causes viral myocarditis, which mainly involves immune cell infiltration, eventually resulting in heart failure. In the present study, the effects and underlying mechanisms of GYY4137 treatment of CVB3‑induced myocarditis were investigated. The effects of GYY4137 on CVB3‑induced nuclear factor‑kappa B (NF‑κB) activity were examined by western blotting, immunofluorescence and electrophoretic mobility shift assay. Mitogen‑activated protein kinase (MAPK) signaling protein expression levels were detected by western blotting. Cardiomyocyte damage‑related enzyme activities, such as lactate dehydrogenase (LDH) and creatine kinase MB (CK‑MB), were measured by ELISA, as well as the production of proinflammatory cytokines. The results revealed that GYY4137 suppressed CVB3‑induced secretion of LDH, CK‑MB and pro‑inflammatory cytokines, such as tumor necrosis factor‑α, interleukin (IL)‑1β and IL‑6. Furthermore, the activation of NF‑κB and the IκBα degradation induced by CVB3 were also inhibited by GYY4137. Notably, the phosphorylation of p38, ERK1/2 and JNK1/2 induced by CVB3 was also inhibited by GYY4137. In conclusion, the data demonstrate that GYY4137 exerts anti‑inflammatory effects in CVB3‑infected cardiomyocytes. This anti‑inflammatory mechanism may be associated with suppression of NF‑κB and MAPK signaling pathway activation.
Collapse
Affiliation(s)
- Zubo Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hua Peng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qing Du
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wen Lin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yali Liu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
47
|
Massilamany C, Gangaplara A, Reddy J. Intricacies of cardiac damage in coxsackievirus B3 infection: implications for therapy. Int J Cardiol 2014; 177:330-339. [PMID: 25449464 DOI: 10.1016/j.ijcard.2014.09.136] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/27/2014] [Accepted: 09/15/2014] [Indexed: 02/06/2023]
Abstract
Heart disease is the leading cause of death in humans, and myocarditis is one predominant cause of heart failure in young adults. Patients affected with myocarditis can develop dilated cardiomyopathy (DCM), a common reason for heart transplantation, which to date is the only viable option for combatting DCM. Myocarditis/DCM patients show antibodies to coxsackievirus B (CVB)3 and cardiac antigens, suggesting a role for CVB-mediated autoimmunity in the disease pathogenesis; however, a direct causal link remains to be determined clinically. Experimentally, myocarditis can be induced in susceptible strains of mice using the human isolates of CVB3, and the disease pathogenesis of postinfectious myocarditis resembles that of human disease, making the observations made in animals relevant to humans. In this review, we discuss the complex nature of CVB3-induced myocarditis as it relates to the damage caused by both the virus and the host's response to infection. Based on recent data we obtained in the mouse model of CVB3 infection, we provide evidence to suggest that CVB3 infection accompanies the generation of cardiac myosin-specific CD4 T cells that can transfer the disease to naïve recipients. The therapeutic implications of these observations are also discussed.
Collapse
Affiliation(s)
| | - Arunakumar Gangaplara
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of health, Bethesda, MD
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583
| |
Collapse
|
48
|
Zhang X, Dong C, Sun X, Li Z, Zhang M, Guan Z, Duan M. Induction of the cellular miR-29c by influenza virus inhibits the innate immune response through protection of A20 mRNA. Biochem Biophys Res Commun 2014; 450:755-61. [PMID: 24953694 DOI: 10.1016/j.bbrc.2014.06.059] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 06/11/2014] [Indexed: 11/18/2022]
Abstract
Influenza A viruses (IAVs) are negative-sense, single-stranded, segmented RNA viruses, which primarily targets respiratory epithelial cells and produces clinical outcomes ranging from mild upper respiratory infection to severe pneumonia. MicroRNAs (miRNAs) represent a family of small noncoding RNAs controlling translation and transcription of many genes. The human miR-29 family of miRNAs has three mature members, miR-29a, miR-29b, and miR-29c. Recent studies have revealed that miR-29 is involved in regulation of the innate and adaptive immune responses. However, the function of miR-29 in the immune response to IAV infection remains to be further explored. Our previous study has shown that miR-29 family members are up-regulated during IAV infection, especially miR-29c. Here we report that miR-29c is involved in inhibition of IAV-induced innate immune responses. We found that posttranscriptional regulation was involved in IAV-induced A20 expression in A549 cells. Consistent with a previous report, miR-29c functionally protected A20 transcripts in A549 cells. Overexpression of miR-29c with miR-29c mimic enhanced IAV-induced A20 protein expression and conversely that miR-29c inhibitor significantly blocked IAV-induced A20 protein expression in A549 cells. Furthermore, functional results showed that IAV-induced miR-29c expression correlated with decreased NF-κB activity and expression of several antiviral and proinflammatory cytokines via up-regulation of A20. Together, the findings indicate a new role of miR-29c in IAV infection and suggest its induction may contribute to counteract the innate immune response.
Collapse
Affiliation(s)
- Xiaoyang Zhang
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, 130062 Changchun, PR China
| | - Chunyan Dong
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, 130062 Changchun, PR China
| | - Xiaoning Sun
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, 130062 Changchun, PR China
| | - Zhongyi Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, 130062 Changchun, PR China
| | - Maolin Zhang
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, 130062 Changchun, PR China
| | - Zhenhong Guan
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, 130062 Changchun, PR China
| | - Ming Duan
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, 130062 Changchun, PR China.
| |
Collapse
|
49
|
Zou XL, Pei DA, Yan JZ, Xu G, Wu P. A20 overexpression inhibits lipopolysaccharide-induced NF-κB activation, TRAF6 and CD40 expression in rat peritoneal mesothelial cells. Int J Mol Sci 2014; 15:6592-608. [PMID: 24747594 PMCID: PMC4013649 DOI: 10.3390/ijms15046592] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/04/2014] [Accepted: 03/24/2014] [Indexed: 12/31/2022] Open
Abstract
Zinc finger protein A20 is a key negative regulator of inflammation. However, whether A20 may affect inflammation during peritoneal dialysis (PD)-associated peritonitis is still unclear. This study was aimed to investigate the effect of A20 overexpression on lipopolysaccharide (LPS)-induced inflammatory response in rat peritoneal mesothelial cells (RPMCs). Isolated and cultured RPMCs in vitro. Plasmid pGEM-T easy-A20 was transfected into RPMCs by Lipofectamine™2000. The protein expression of A20, phospho-IκBα, IκBα, TNF receptor-associated factor (TRAF) 6 and CD40 were analyzed by Western blot. The mRNA expression of TRAF6, CD40, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were determined by real time-PCR. NF-κB p65 DNA binding activity, IL-6 and TNF-α levels in cells culture supernatant were determined by ELISA. Our results revealed that RPMCs overexpression of A20 lead to significant decrease of LPS-induced IκBα phosphorylation and NF-κB DNA binding activity (all p<0.01). In addition, A20 also attenuated the expression of TRAF6, CD40, IL-6 and TNF-α as well as levels of IL-6 and TNF-α in cells culture supernatant (all p<0.05). However, A20 only partly inhibited CD40 expression. Our study indicated that A20 overexpression may depress the inflammatory response induced by LPS in cultured RPMCs through negatively regulated the relevant function of adaptors in LPS signaling pathway.
Collapse
Affiliation(s)
- Xun-Liang Zou
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| | - De-An Pei
- Division of Cardiology, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, China.
| | - Ju-Zhen Yan
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| | - Gang Xu
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| | - Ping Wu
- Department of Nephrology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China.
| |
Collapse
|
50
|
Ye X, Zhang HM, Qiu Y, Hanson PJ, Hemida MG, Wei W, Hoodless PA, Chu F, Yang D. Coxsackievirus-induced miR-21 disrupts cardiomyocyte interactions via the downregulation of intercalated disk components. PLoS Pathog 2014; 10:e1004070. [PMID: 24722419 PMCID: PMC3983067 DOI: 10.1371/journal.ppat.1004070] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/25/2014] [Indexed: 01/28/2023] Open
Abstract
Intercalated disks (ICDs) are substantial connections maintaining cardiac structures and mediating signal communications among cardiomyocytes. Deficiency in ICD components such as desmosomes, fascia adherens and gap junctions leads to heart dysfunction. Coxsackievirus B3 (CVB3) infection induces cardiac failure but its pathogenic effect on ICDs is unclear. Here we show that CVB3-induced miR-21 expression affects ICD structure, i.e., upregulated miR-21 targets YOD1, a deubiquitinating enzyme, to enhance the K48-linked ubiquitination and degradation of desmin, resulting in disruption of desmosomes. Inhibition of miR-21 preserves desmin during CVB3 infection. Treatment with proteasome inhibitors blocks miR-21-mediated desmin degradation. Transfection of miR-21 or knockdown of YOD1 triggers co-localization of desmin with proteasomes. We also identified K108 and K406 as important sites for desmin ubiquintination and degradation. In addition, miR-21 directly targets vinculin, leading to disturbed fascia adherens evidenced by the suppression and disorientation of pan-cadherin and α-E-catenin proteins, two fascia adherens-components. Our findings suggest a new mechanism of miR-21 in modulating cell-cell interactions of cardiomyocytes during CVB3 infection. Coxsackievirus B3 (CVB3) is one of most common causes of heart inflammation and failure. However, the mechanism by which CVB3 induces cardiac damage has not been fully elucidated. Particularly, the involvement of microRNAs (miRNAs), a family of small RNAs controlling the progression of a wide range of diseases, in CVB3 infection is still unclear. These small RNAs are essential to understand the CVB3-caused heart muscle cell injury and have great potential to serve therapeutic purposes. Here, we systematically analyzed the miRNA changes during CVB3 infection and found that miR-21 is increased by viral infection. We further demonstrated that the CVB3-induced miR-21 triggers heart muscle cell damage by interfering with the cell-cell interactions. miR-21 suppresses the levels of components in cell-cell interactions by either promoting the degradation of those proteins or directly inhibiting the protein production. Inhibition of miR-21 can reduce the host injury caused by CVB3 infection. Our findings will shed new lights on the pathogenesis of CVB3-induced heart failure.
Collapse
Affiliation(s)
- Xin Ye
- Department of Pathology and Laboratory Medicine, University of British Columbia, The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Huifang Mary Zhang
- Department of Pathology and Laboratory Medicine, University of British Columbia, The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Ye Qiu
- Department of Pathology and Laboratory Medicine, University of British Columbia, The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Paul J. Hanson
- Department of Pathology and Laboratory Medicine, University of British Columbia, The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Maged Gomaa Hemida
- Department of Pathology and Laboratory Medicine, University of British Columbia, The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Wei Wei
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Pamela A. Hoodless
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Fanny Chu
- Department of Pathology and Laboratory Medicine, University of British Columbia, The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Decheng Yang
- Department of Pathology and Laboratory Medicine, University of British Columbia, The Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|