1
|
Parducci NS, Garnique ADMB, de Almeida BO, Machado-Neto JA. Exploring the dual role of SIVA1 in cancer biology. Gene 2025; 950:149365. [PMID: 40024298 DOI: 10.1016/j.gene.2025.149365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/08/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
The intricate molecular mechanisms associated with cancer development continue to engage researchers due to the significant impact of the disease on global mortality. This review delves into the role of the apoptosis regulatory protein SIVA1, which has emerged as a significant player in cellular homeostasis. SIVA1, initially characterized as a pro-apoptotic protein interacting with the TNF receptor CD27, has since been implicated in various cellular contexts, revealing its complex functional dynamics. The SIVA1 gene, located on chromosome 14, encodes a protein containing distinctive structural features, including an amphipathic helix and a death domain homology region. Localization studies show that SIVA1 is present in both the cytoplasm and nucleus, with its expression linked to tumor differentiation. Investigations into SIVA1's interactions have uncovered its pro-apoptotic mechanisms, such as binding to anti-apoptotic proteins from the BCL2 family, thus promoting apoptosis under stress conditions. Interestingly, SIVA1 also exhibits tumor-promoting properties in specific cancer types, suggesting a dual role in apoptosis induction and tumor progression. As research progresses, understanding the regulatory mechanisms governing SIVA1's multifaceted functions could pave the way for novel therapeutic strategies aimed at manipulating its activity for improved cancer treatment outcomes. Future studies are warranted to clarify SIVA1's contextual roles and explore its potential clinical implications.
Collapse
Affiliation(s)
- Natália Sudan Parducci
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil.
| | | | - Bruna Oliveira de Almeida
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil.
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil.
| |
Collapse
|
2
|
Liu Q, Shang Y, Tao Z, Li X, Shen L, Zhang H, Liu Z, Rao Z, Yu X, Cao Y, Zeng L, Huang X. Coxsackievirus group B3 regulates ASS1-mediated metabolic reprogramming and promotes macrophage inflammatory polarization in viral myocarditis. J Virol 2024; 98:e0080524. [PMID: 39194244 PMCID: PMC11406948 DOI: 10.1128/jvi.00805-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Coxsackievirus group B3 (CVB3) belongs to the genus Enteroviruses of the family Picornaviridae and is the main pathogen underlying viral myocarditis (VMC). No specific therapeutic is available for this condition. Argininosuccinate synthase 1 (ASS1) is a key enzyme in the urea cycle that converts citrulline and aspartic acid to argininosuccinate. Here, we found that CVB3 and its capsid protein VP2 inhibit the autophagic degradation of ASS1 and that CVB3 consumes citrulline to upregulate ASS1, triggers urea cycle metabolic reprogramming, and then activates macrophages to develop pro-inflammatory polarization, thereby promoting the occurrence and development of VMC. Conversely, citrulline supplementation to prevent depletion can downregulate ASS1, rescue macrophage polarization, and alleviate the pathogenicity of VMC. These findings provide a new perspective on the occurrence and development of VMC, revealing ASS1 as a potential new target for treating this disease. IMPORTANCE Viral myocarditis (VMC) is a common and potentially life-threatening myocardial inflammatory disease, most commonly caused by CVB3 infection. So far, the pathogenesis of VMC caused by CVB3 is mainly focused on two aspects: one is the direct myocardial injury caused by a large number of viral replication in the early stage of infection, and the other is the local immune cell infiltration and inflammatory damage of the myocardium in the adaptive immune response stage. There are few studies on the early innate immunity of CVB3 infection in myocardial tissue, but the appearance of macrophages in the early stage of CVB3 infection suggests that they can play a regulatory role as early innate immune response cells in myocardial tissue. Here, we discovered a possible new mechanism of VMC caused by CVB3, revealed new drug targets for anti-CVB3, and discovered the therapeutic potential of citrulline for VMC.
Collapse
Affiliation(s)
- Qiong Liu
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yinpan Shang
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ziwei Tao
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xuan Li
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lu Shen
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hanchi Zhang
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Zhili Liu
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- HuanKui Academy, Nanchang University, Nanchang, China
| | - Zhirong Rao
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- HuanKui Academy, Nanchang University, Nanchang, China
| | - Xiaomin Yu
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanli Cao
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lingbing Zeng
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The First Affiliated Hospital of Nanchang University, School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- The First Affiliated Hospital of Nanchang University and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Mitochondria Dysfunction at the Heart of Viral Myocarditis: Mechanistic Insights and Therapeutic Implications. Viruses 2023; 15:v15020351. [PMID: 36851568 PMCID: PMC9963085 DOI: 10.3390/v15020351] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
The myocardium/heart is the most mitochondria-rich tissue in the human body with mitochondria comprising approximately 30% of total cardiomyocyte volume. As the resident "powerhouse" of cells, mitochondria help to fuel the high energy demands of a continuously beating myocardium. It is no surprise that mitochondrial dysfunction underscores the pathogenesis of many cardiovascular ailments, including those of viral origin such as virus-induced myocarditis. Enteroviruses have been especially linked to injuries of the myocardium and its sequelae dilated cardiomyopathy for which no effective therapies currently exist. Intriguingly, recent mechanistic insights have demonstrated viral infections to directly damage mitochondria, impair the mitochondrial quality control processes of the cell, such as disrupting mitochondrial antiviral innate immune signaling, and promoting mitochondrial-dependent pathological inflammation of the infected myocardium. In this review, we briefly highlight recent insights on the virus-mitochondria crosstalk and discuss the therapeutic implications of targeting mitochondria to preserve heart function and ultimately combat viral myocarditis.
Collapse
|
4
|
Yang Y, Li W, You B, Zhou C. Advances in cell death mechanisms involved in viral myocarditis. Front Cardiovasc Med 2022; 9:968752. [PMID: 36017100 PMCID: PMC9395613 DOI: 10.3389/fcvm.2022.968752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
Viral myocarditis is an acute inflammatory disease of the myocardium. Although many etiopathogenic factors exist, coxsackievirus B3 is a the leading cause of viral myocarditis. Abnormal cardiomyocyte death is the underlying problem for most cardiovascular diseases and fatalities. Various types of cell death occur and are regulated to varying degrees. In this review, we discuss the different cell death mechanisms in viral myocarditis and the potential interactions between them. We also explore the role and mechanism of cardiomyocyte death with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Exploring the mechanisms may help in the early identification and the development of effective treatments, thus improving the quality of life of patients with viral myocarditis. We believe that the inhibition of cardiomyocyte death has immense therapeutic potential in increasing the longevity and health of the heart.
Collapse
Affiliation(s)
- Yang Yang
- Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
- Clinical Laboratory Center, Jiangsu Taizhou People’s Hospital, Taizhou, China
- *Correspondence: Yang Yang,
| | - Wang Li
- Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
- Clinical Laboratory Center, Jiangsu Taizhou People’s Hospital, Taizhou, China
| | - Benshuai You
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chenglin Zhou
- Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
- Clinical Laboratory Center, Jiangsu Taizhou People’s Hospital, Taizhou, China
- Chenglin Zhou,
| |
Collapse
|
5
|
Kim SR, Song JH, Ahn JH, Jeong MS, Yang YM, Cho J, Jeong JH, Cha Y, Kim KN, Kim HP, Chang SY, Ko HJ. Obesity Exacerbates Coxsackievirus Infection via Lipid-Induced Mitochondrial Reactive Oxygen Species Generation. Immune Netw 2022; 22:e19. [PMID: 35573153 PMCID: PMC9066006 DOI: 10.4110/in.2022.22.e19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/16/2022] [Accepted: 03/27/2022] [Indexed: 12/03/2022] Open
Abstract
Coxsackievirus B3 (CVB3) infection causes acute pancreatitis and myocarditis. However, its pathophysiological mechanism is unclear. Here, we investigated how lipid metabolism is associated with exacerbation of CVB3 pathology using high-fat diet (HFD)-induced obese mice. Mice were intraperitoneally inoculated with 1×106 pfu/mouse of CVB3 after being fed a control or HFD to induce obesity. Mice were treated with mitoquinone (MitoQ) to reduce the level of mitochondrial ROS (mtROS). In obese mice, lipotoxicity of white adipose tissue-induced inflammation caused increased replication of CVB3 and mortality. The coxsackievirus adenovirus receptor increased under obese conditions, facilitating CVB3 replication in vitro. However, lipid-treated cells with receptor-specific inhibitors did not reduce CVB3 replication. In addition, lipid treatment increased mitochondria-derived vesicle formation and the number of multivesicular bodies. Alternatively, we found that inhibition of lipid-induced mtROS decreased viral replication. Notably, HFD-fed mice were more susceptible to CVB3-induced mortality in association with increased levels of CVB3 replication in adipose tissue, which was ameliorated by administration of the mtROS inhibitor, MitoQ. These results suggest that mtROS inhibitors can be used as potential treatments for CVB3 infection.
Collapse
Affiliation(s)
- Seong-Ryeol Kim
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hyoung Song
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Myeong Seon Jeong
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon 24341, Korea
| | - Yoon Mee Yang
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Jaewon Cho
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hyeon Jeong
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Younggil Cha
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Kil-Nam Kim
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon 24341, Korea
| | - Hong Pyo Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea
| | | | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
6
|
Abstract
Although a broad range of viruses cause myocarditis, the mechanisms that underlie viral myocarditis are poorly understood. Here, we report that the M2 gene is a determinant of reovirus myocarditis. The M2 gene encodes outer capsid protein μ1, which mediates host membrane penetration during reovirus entry. We infected newborn C57BL/6 mice with reovirus strain type 1 Lang (T1L) or a reassortant reovirus in which the M2 gene from strain type 3 Dearing (T3D) was substituted into the T1L genetic background (T1L/T3DM2). T1L was non-lethal in wild-type mice, whereas greater than 90% of mice succumbed to T1L/T3DM2 infection. T1L/T3DM2 produced higher viral loads than T1L at the site of inoculation. In secondary organs, T1L/T3DM2 was detected with more rapid kinetics and reached higher peak titers than T1L. We found that hearts from T1L/T3DM2-infected mice were grossly abnormal, with large lesions indicative of substantial inflammatory infiltrate. Lesions in T1L/T3DM2-infected mice contained necrotic cardiomyocytes with pyknotic debris, and extensive lymphocyte and histiocyte infiltration. In contrast, T1L induced the formation of small purulent lesions in a small subset of animals, consistent with T1L being mildly myocarditic. Finally, more activated caspase-3-positive cells were observed in hearts from animals infected with T1L/T3DM2 compared to T1L. Together, our findings indicate that substitution of the T3D M2 allele into an otherwise T1L genetic background is sufficient to change a non-lethal infection into a lethal infection. Our results further indicate that T3D M2 enhances T1L replication and dissemination in vivo, which potentiates the capacity of reovirus to cause myocarditis. IMPORTANCE Reovirus is a non-enveloped virus with a segmented double-stranded RNA genome that serves as a model for studying viral myocarditis. The mechanisms by which reovirus drives myocarditis development are not fully elucidated. We found that substituting the M2 gene from strain type 3 Dearing (T3D) into an otherwise type 1 Lang (T1L) genetic background (T1L/T3DM2) was sufficient to convert the non-lethal T1L strain into a lethal infection in neonatal C57BL/6 mice. T1L/T3DM2 disseminated more efficiently and reached higher maximum titers than T1L in all organs tested, including the heart. T1L is mildly myocarditic and induced small areas of cardiac inflammation in a subset of mice. In contrast, hearts from mice infected with T1L/T3DM2 contained extensive cardiac inflammatory infiltration and more activated caspase-3-positive cells, which is indicative of apoptosis. Together, our findings identify the reovirus M2 gene as a new determinant of reovirus-induced myocarditis.
Collapse
|
7
|
Wang Q, Zhu Q, Ye Q, Wang J, Dong Q, Chen Y, Wang M, Fu Y, Wu R, Wu T. STAT3 Suppresses Cardiomyocytes Apoptosis in CVB3-Induced Myocarditis Via Survivin. Front Pharmacol 2021; 11:613883. [PMID: 33658937 PMCID: PMC7919905 DOI: 10.3389/fphar.2020.613883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Viral myocarditis (VMC) is a common inflammatory cardiovascular disease with unclear mechanisms, which mainly affects children and adolescents. Apoptosis is the key to CVB3-induced myocarditis, and blocking this process may be beneficial to the therapy of VMC. Hence, this study aimed to explore the protective function of STAT3 on cardiomyocyte apoptosis of VMC and its underlying mechanisms. Methods and Results: In this research, we confirmed that STAT3 was significantly activated in both animal and cell models of VMC. To further clarify what role did STAT3 play in VMC, AG490, an inhibitor of STAT3, was used to suppress p-STAT3. Our results demonstrated that decreased expression of p-STAT3 caused by AG490 significantly aggravated severity of VMC with elevated myocardial inflammation, deteriorative ventricular systolic function and increased mortality. It suggested that STAT3 plays a protective role in VMC. To further identify the anti-apoptosis impact that activated STAT3 made, we constructed lentivirus to regulate the expression of STAT3 in NMCs. We found that up-regulated activated STAT3 attenuated cardiomyocyte apoptosis, but down-regulated one aggravated that, which verified activated STAT3 played an anti-apoptosis role in VMC. Following that, we explored what elements are involved in the anti-apoptotic mechanism of activated STAT3 by using survivin inhibitor YM155. The result showed the anti-apoptotic effect of activated STAT3 does not work in the case of survivin inhibition. Conclusion: Our findings demonstrated STAT3 by targeting survivin alleviated cardiomyocyte apoptosis in CVB3-induced myocarditis.
Collapse
Affiliation(s)
- Qiaoyu Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiongjun Zhu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiaofang Ye
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiajun Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qianqian Dong
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Youran Chen
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Minna Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Fu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Rongzhou Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Tingting Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Politano G, Benso A. IL6-mediated HCoV-host interactome regulatory network and GO/Pathway enrichment analysis. PLoS Comput Biol 2020; 16:e1008238. [PMID: 32997660 PMCID: PMC7561109 DOI: 10.1371/journal.pcbi.1008238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/15/2020] [Accepted: 08/11/2020] [Indexed: 11/19/2022] Open
Abstract
During these days of global emergency for the COVID-19 disease outbreak, there is an urgency to share reliable information able to help worldwide life scientists to get better insights and make sense of the large amount of data currently available. In this study we used the results presented in [1] to perform two different Systems Biology analyses on the HCoV-host interactome. In the first one, we reconstructed the interactome of the HCoV-host proteins, integrating it with highly reliable miRNA and drug interactions information. We then added the IL-6 gene, identified in recent publications [2] as heavily involved in the COVID-19 progression and, interestingly, we identified several interactions with the reconstructed interactome. In the second analysis, we performed a Gene Ontology and a Pathways enrichment analysis on the full set of the HCoV-host interactome proteins and on the ones belonging to a significantly dense cluster of interacting proteins identified in the first analysis. Results of the two analyses provide a compact but comprehensive glance on some of the current state-of-the-art regulations, GO, and pathways involved in the HCoV-host interactome, and that could support all scientists currently focusing on SARS-CoV-2 research.
Collapse
Affiliation(s)
| | - Alfredo Benso
- Computer Science and Automation Department, Politecnico di Torino, Italy
| |
Collapse
|
9
|
Wan Q, Song D, Li H, He ML. Stress proteins: the biological functions in virus infection, present and challenges for target-based antiviral drug development. Signal Transduct Target Ther 2020; 5:125. [PMID: 32661235 PMCID: PMC7356129 DOI: 10.1038/s41392-020-00233-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/26/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
Stress proteins (SPs) including heat-shock proteins (HSPs), RNA chaperones, and ER associated stress proteins are molecular chaperones essential for cellular homeostasis. The major functions of HSPs include chaperoning misfolded or unfolded polypeptides, protecting cells from toxic stress, and presenting immune and inflammatory cytokines. Regarded as a double-edged sword, HSPs also cooperate with numerous viruses and cancer cells to promote their survival. RNA chaperones are a group of heterogeneous nuclear ribonucleoproteins (hnRNPs), which are essential factors for manipulating both the functions and metabolisms of pre-mRNAs/hnRNAs transcribed by RNA polymerase II. hnRNPs involve in a large number of cellular processes, including chromatin remodelling, transcription regulation, RNP assembly and stabilization, RNA export, virus replication, histone-like nucleoid structuring, and even intracellular immunity. Dysregulation of stress proteins is associated with many human diseases including human cancer, cardiovascular diseases, neurodegenerative diseases (e.g., Parkinson’s diseases, Alzheimer disease), stroke and infectious diseases. In this review, we summarized the biologic function of stress proteins, and current progress on their mechanisms related to virus reproduction and diseases caused by virus infections. As SPs also attract a great interest as potential antiviral targets (e.g., COVID-19), we also discuss the present progress and challenges in this area of HSP-based drug development, as well as with compounds already under clinical evaluation.
Collapse
Affiliation(s)
- Qianya Wan
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Dan Song
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Huangcan Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China. .,CityU Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
10
|
Lai Y, Wang M, Cheng A, Mao S, Ou X, Yang Q, Wu Y, Jia R, Liu M, Zhu D, Chen S, Zhang S, Zhao XX, Huang J, Gao Q, Wang Y, Xu Z, Chen Z, Zhu L, Luo Q, Liu Y, Yu Y, Zhang L, Tian B, Pan L, Rehman MU, Chen X. Regulation of Apoptosis by Enteroviruses. Front Microbiol 2020; 11:1145. [PMID: 32582091 PMCID: PMC7283464 DOI: 10.3389/fmicb.2020.01145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/05/2020] [Indexed: 01/14/2023] Open
Abstract
Enterovirus infection can cause a variety of diseases and severely impair the health of humans, animals, poultry, and other organisms. To resist viral infection, host organisms clear infected cells and viruses via apoptosis. However, throughout their long-term competition with host cells, enteroviruses have evolved a series of mechanisms to regulate the balance of apoptosis in order to replicate and proliferate. In the early stage of infection, enteroviruses mainly inhibit apoptosis by regulating the PI3K/Akt pathway and the autophagy pathway and by impairing cell sensors, thereby delaying viral replication. In the late stage of infection, enteroviruses mainly regulate apoptotic pathways and the host translation process via various viral proteins, ultimately inducing apoptosis. This paper discusses the means by which these two phenomena are balanced in enteroviruses to produce virus-favoring conditions – in a temporal sequence or through competition with each other. This information is important for further elucidation of the relevant mechanisms of acute infection by enteroviruses and other members of the picornavirus family.
Collapse
Affiliation(s)
- Yalan Lai
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sai Mao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xumin Ou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dekang Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shaqiu Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xin-Xin Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qun Gao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yin Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Zhiwen Xu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Zhengli Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Qihui Luo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yunya Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yanling Yu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bin Tian
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Leichang Pan
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mujeeb Ur Rehman
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoyue Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
11
|
Zhao G, Zhang HM, Qiu Y, Ye X, Yang D. Cleavage of Desmosomal Cadherins Promotes γ-Catenin Degradation and Benefits Wnt Signaling in Coxsackievirus B3-Induced Destruction of Cardiomyocytes. Front Microbiol 2020; 11:767. [PMID: 32457708 PMCID: PMC7225294 DOI: 10.3389/fmicb.2020.00767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/31/2020] [Indexed: 01/01/2023] Open
Abstract
Coxsackievirus B3 (CVB3) is the primary etiologic agent of viral myocarditis, a major heart disease that occurs predominantly in children and young adolescents. In the heart, intercalated disks (ICD) are important structural formations that connect adjacent cardiomyocytes to maintain cardiac architecture and mediate signal communication. Deficiency in ICD components, such as desmosome proteins, leads to heart dysfunction. γ-catenin, a component protein of desmosomes, normally binds directly to desmocollin-2 and desmoglein-2. In this study, we found that CVB3 infection downregulated γ-catenin at the protein level but not the mRNA level in mouse HL-1 cardiomyocytes. We further found that this reduction of γ-catenin protein is a result of ubiquitin proteasome-mediated degradation, since the addition of proteasome inhibitor MG132 inhibited γ-catenin downregulation. In addition, we found that desmocollin-2 and desmoglein-2 were cleaved by both viral protease 3C and virus-activated cellular caspase, respectively. These cleavages led to the release of bound γ-catenin from the desmosome into the cytosol, resulting in rapid degradation of γ-catenin. Since γ-catenin shares high sequence homology with β-catenin in binding the TCF/LEF transcription factor, we further studied the effect of γ-catenin degradation on Wnt/β-catenin signaling. Luciferase assay showed that γ-catenin expression inhibited Wnt/β-catenin signaling. This finding was substantiated by qPCR to show that overexpression of γ-catenin downregulated transcription of Wnt signal target genes, c-myc and MMP9, while silencing γ-catenin upregulated these target genes. Finally, we demonstrated that γ-catenin expression inhibited CVB3 replication. In search for the underlying mechanism, we found that silencing γ-catenin caused down-regulation of interferon-β and its stimulated antiviral genes MDA5, MAVS, and ISG15. Taken together, our results indicate, for the first time, that CVB3 infection causes cardiomyocyte death through, at least in part, direct damage to the desmosome structure and reduction of γ-catenin protein, which in return promotes Wnt/β-catenin signaling and downregulates interferon-β stimulated immune responses.
Collapse
Affiliation(s)
- Guangze Zhao
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Huifang M Zhang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Ye Qiu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,College of Biology, Hunan University, Changsha, China
| | - Xin Ye
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Decheng Yang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
12
|
Palrasu M, Zaika E, El-Rifai W, Garcia-Buitrago M, Piazuelo MB, Wilson KT, Peek RM, Zaika AI. Bacterial CagA protein compromises tumor suppressor mechanisms in gastric epithelial cells. J Clin Invest 2020; 130:2422-2434. [PMID: 32250340 PMCID: PMC7190987 DOI: 10.1172/jci130015] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 01/22/2020] [Indexed: 01/01/2023] Open
Abstract
Approximately half of the world's population is infected with the stomach pathogen Helicobacter pylori. Infection with H. pylori is the main risk factor for distal gastric cancer. Bacterial virulence factors, such as the oncoprotein CagA, augment cancer risk. Yet despite high infection rates, only a fraction of H. pylori-infected individuals develop gastric cancer. This raises the question of defining the specific host and bacterial factors responsible for gastric tumorigenesis. To investigate the tumorigenic determinants, we analyzed gastric tissues from human subjects and animals infected with H. pylori bacteria harboring different CagA status. For laboratory studies, well-defined H. pylori strain B128 and its cancerogenic derivative strain 7.13, as well as various bacterial isogenic mutants were employed. We found that H. pylori compromises key tumor suppressor mechanisms: the host stress and apoptotic responses. Our studies showed that CagA induces phosphorylation of XIAP E3 ubiquitin ligase, which enhances ubiquitination and proteasomal degradation of the host proapoptotic factor Siva1. This process is mediated by the PI3K/Akt pathway. Inhibition of Siva1 by H. pylori increases survival of human cells with damaged DNA. It occurs in a strain-specific manner and is associated with the ability to induce gastric tumor.
Collapse
Affiliation(s)
- Manikandan Palrasu
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Elena Zaika
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Wael El-Rifai
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, Florida, USA
| | - Monica Garcia-Buitrago
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Maria Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, VA Tennessee Valley Health Care System, Nashville, Tennessee, USA
| | - Richard M. Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alexander I. Zaika
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, Florida, USA
| |
Collapse
|
13
|
Wu R, Wu T, Li P, Wang Q, Shi Y, Zhan Y, Zhang S, Xia T, Wang Z, Lv H. The protection effects of survivin in the cell model of CVB3-induced viral myocarditis. Heart Vessels 2020; 35:1171-1179. [PMID: 32328712 DOI: 10.1007/s00380-020-01607-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/10/2020] [Indexed: 01/24/2023]
Abstract
Viral myocarditis (VMC) is a widely studied but poorly understood inflammatory cardiomyopathy which mainly affects children and young adults and results in adverse outcomes. Cardiomyocyte apoptosis was reported important in the progress of coxsackievirus B3 (CVB3)-induced VMC and the blocking of this process may contribute to the therapeutic effect towards VMC. Therefore, this study was designed to explore whether survivin, one of the strongest antiapoptotic proteins, can protect cardiomyocytes from apoptosis in VMC and to discover its related mechanisms. Here, the cultured neonatal mouse cardiomyocytes (NMCs) were exposed to CVB3 to establish the cell model of VMC and the results of Western Blot showed that the protein expression of survivin in CVB3-infected NMCs varied at different post-infection time. Lentivirus was next used to examine the function of survivin in CVB3-infected NMCs. TUNEL assay demonstrated that the overexpression of survivin interrupted CVB3-induced apoptosis. It was next examined whether caspase-3 and -9 were involved in the antiapoptotic pathway initiated by survivin via Western Blot. The results showed a reverse relationship between the protein expression of survivin and that of cleaved caspase-3 and cleaved caspase-9, suggesting that survivin may attenuate apoptosis through restraining the activity of caspase-3 and -9. Moreover, the supernatant fluid of cultured NMCs was extracted to detect the quantitation of released lactate dehydrogenase (LDH) and a sharp decrease was discovered in the survivin-overexpressed group compared to the CVB3-infected group, indicating a protective role of survivin in the cell model of CVB3-induced myocarditis. This study demonstrated that survivin was triggered by CVB3 infection in NMCs and survivin executed its antiapoptotic effects via caspase-3- and caspase-9-dependent signaling pathway.
Collapse
Affiliation(s)
- Rongzhou Wu
- Department of Pediatric Cardiology, Children's Hospital of Soochow University, Suzhou, 215025, China
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Tingting Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Ping Li
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Qiaoyu Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Youyang Shi
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yi Zhan
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Songyue Zhang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Tianhe Xia
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zhenquan Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Haitao Lv
- Department of Pediatric Cardiology, Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
14
|
Coccia E, Planells-Ferrer L, Badillos-Rodríguez R, Pascual M, Segura MF, Fernández-Hernández R, López-Soriano J, Garí E, Soriano E, Barneda-Zahonero B, Moubarak RS, Pérez-García MJ, Comella JX. SIVA-1 regulates apoptosis and synaptic function by modulating XIAP interaction with the death receptor antagonist FAIM-L. Cell Death Dis 2020; 11:82. [PMID: 32015347 PMCID: PMC6997380 DOI: 10.1038/s41419-020-2282-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
The long isoform of Fas apoptosis inhibitory molecule (FAIM-L) is a neuron-specific death receptor antagonist that modulates apoptotic cell death and mechanisms of neuronal plasticity. FAIM-L exerts its antiapoptotic action by binding to X-linked inhibitor of apoptosis protein (XIAP), an inhibitor of caspases, which are the main effectors of apoptosis. XIAP levels are regulated by the ubiquitin-proteasome pathway. FAIM-L interaction with XIAP prevents the ubiquitination and degradation of the latter, thereby allowing it to inhibit caspase activation. This interaction also modulates non-apoptotic functions of caspases, such as the endocytosis of AMPA receptor (AMPAR) in hippocampal long-term depression (LTD). The molecular mechanism of action exerted by FAIM-L is unclear since the consensus binding motifs are still unknown. Here, we performed a two-hybrid screening to discover novel FAIM-L-interacting proteins. We found a functional interaction of SIVA-1 with FAIM-L. SIVA-1 is a proapoptotic protein that has the capacity to interact with XIAP. We describe how SIVA-1 regulates FAIM-L function by disrupting the interaction of FAIM-L with XIAP, thereby promoting XIAP ubiquitination, caspase-3 activation and neuronal death. Furthermore, we report that SIVA-1 plays a role in receptor internalization in synapses. SIVA-1 is upregulated upon chemical LTD induction, and it modulates AMPAR internalization via non-apoptotic activation of caspases. In summary, our findings uncover SIVA-1 as new functional partner of FAIM-L and demonstrate its role as a regulator of caspase activity in synaptic function.
Collapse
Affiliation(s)
- Elena Coccia
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Laura Planells-Ferrer
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Raquel Badillos-Rodríguez
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Marta Pascual
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Bellaterra, Spain.,Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, 08031, Barcelona, Spain
| | - Miguel F Segura
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR)-UAB, 08035, Barcelona, Spain
| | - Rita Fernández-Hernández
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRBLleida), and Departament de Ciències Mèdiques Bàsiques; Facultat de Medicina, Universitat de Lleida, 25198, Lleida, Catalonia, Spain
| | - Joaquin López-Soriano
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Eloi Garí
- Cell Cycle Laboratory, Institut de Recerca Biomèdica de Lleida (IRBLleida), and Departament de Ciències Mèdiques Bàsiques; Facultat de Medicina, Universitat de Lleida, 25198, Lleida, Catalonia, Spain
| | - Eduardo Soriano
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Bellaterra, Spain.,Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, 08031, Barcelona, Spain.,ICREA Academia, Barcelona, Spain
| | - Bruna Barneda-Zahonero
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain
| | - Rana S Moubarak
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain.,Department of Pathology, NYU Langone Health, New York, 10016, NY, USA
| | - M Jose Pérez-García
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain. .,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain.
| | - Joan X Comella
- Cell Signaling and Apoptosis Group, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031, Madrid, Spain. .,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08031, Bellaterra, Spain.
| |
Collapse
|
15
|
Liu T, Ma Y, Wang Z, Zhang W, Yang X. Siva 1 Inhibits Cervical Cancer Progression and Its Clinical Prognosis Significance. Cancer Manag Res 2020; 12:303-311. [PMID: 32021444 PMCID: PMC6970243 DOI: 10.2147/cmar.s232994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/05/2019] [Indexed: 01/20/2023] Open
Abstract
Background Cervical cancer is the second most common female malignancies. But the exact etiology of cervical cancer is still under investigation. Recent observations revealed that the loss expression of Siva 1 was related to several different types of tumors. It could play an indispensable role in both exogenous and endogenous apoptotic signaling pathways. Nevertheless, the relationship between Siva 1 expression and cervical cancer progression has not yet been fully clarified. This study aimed to explore the functional role of Siva1 in cervical cancer. Materials and Methods In this present experiment, expression of Siva 1 was detected in 87 cervical cancer, 34 CIN and 20 normal samples by immunohistochemistry. The correlation of Siva 1 expression and overall survival times (OS) was analyzed by Kaplan–Meier analysis. We up-regulated the expression of Siva 1 by plasmid pCMV3-Siva 1 in C33A cells. CCK8, flow cytometry, wound-healing, and transwell assays were performed to examine the influences of Siva 1 expression on cell proliferation, apoptosis, migration and invasion. Results The expression of Siva 1 was decreased in cervical cancer tissues compared with CIN and normal tissues. In addition, the Siva 1 immunoreactivity was significantly associated with tumor differentiation. Patients with Siva 1 negative staining exhibited a significantly decreased overall survival. Then, we established stable Siva 1 ectopic expression cells, and we found that elevated expression of Siva 1 promoted apoptosis, inhibited proliferation, and suppressed migration and invasion of cervical cancer cells. Conclusion The present study revealed a crucial role of Siva 1 in tumor progression and it may be a valuable prognostic indicator of cervical cancer.
Collapse
Affiliation(s)
- Ting Liu
- Department of Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Yifei Ma
- Department of Obstetrics and Gynecology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Zhiling Wang
- Department of Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Wenjing Zhang
- Department of Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Xingsheng Yang
- Department of Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, People's Republic of China
| |
Collapse
|
16
|
The VP3 protein of duck hepatitis A virus mediates host cell adsorption and apoptosis. Sci Rep 2019; 9:16783. [PMID: 31727985 PMCID: PMC6856352 DOI: 10.1038/s41598-019-53285-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
Duck hepatitis A virus (DHAV) causes an infectious disease that mainly affects 1- to 4-week-old ducklings, resulting in considerable loss to the duck industry. Although there have been many studies on DHAV in recent years, the effects on host infection and pathogenesis of DHAV-1 remain largely unknown. This study investigated the effects of the DHAV-1 structural protein VP3 on DHAV-1 virus adsorption and apoptosis to explore the role of VP3 in the viral life cycle. The effects of DHAV-1 VP3 and an antibody against the protein on virion adsorption was analyzed by qRT-PCR. The results showed that the virus copy number for the rabbit anti-VP3 IgG-treated group was significantly lower than that for the negative control group but higher than that for the rabbit anti-DHAV-1 IgG-treated group. This result indicates that VP3 mediates DHAV-1 virus adsorption but that it is not the only protein that involved in this process. In addition, a eukaryotic recombinant plasmid, pCAGGS/VP3, was transfected into duck embryo fibroblasts (DEFs), and the apoptotic rate was determined by DAPI staining, the TUNEL assay and flow cytometry. DAPI staining showed nucleus fragmentation and nuclear edge shifting. TUNEL assay results revealed yellow nuclei, and flow cytometry indicated a significant increase in the apoptotic rate. In addition, qRT-PCR revealed increased in the transcriptional levels of the apoptotic caspase-3, −8 and −9, with the largest increase for caspase-3, followed by caspase-9 and caspase-8. Enzyme activity analysis confirmed these results. Furthermore, the VP3 protein decreased the mitochondrial membrane potential, and the transcriptional levels of the proapoptotic factors Bak, Cyt c and Apaf-1 in the mitochondrial apoptotic pathway were significantly upregulated. These data suggest that expression of VP3 in DEFs induces apoptosis and may primarily activate caspase-3-induced apoptosis through mitochondrion-mediated intrinsic pathways. The findings provide scientific data to clarify DHAV-1 infection and pathogenesis.
Collapse
|
17
|
Nicotinic Agonist Inhibits Cardiomyocyte Apoptosis in CVB3-Induced Myocarditis via α3 β4-nAChR/PI3K/Akt-Dependent Survivin Upregulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9496419. [PMID: 30984342 PMCID: PMC6431489 DOI: 10.1155/2019/9496419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/05/2018] [Indexed: 12/27/2022]
Abstract
Background Cardiomyocyte apoptosis is critical for the development of coxsackievirus B3- (CVB3-) induced myocarditis, which is a common cardiac disease that may result in heart failure or even sudden death. Previous studies have associated CVB3-induced apoptosis with the downregulation of antiapoptotic proteins. Here, attempts were made to examine whether nicotinic acetylcholine receptors (nAChRs), especially α3β4-nAChRs, were a novel therapeutic antiapoptotic target via the activation of survivin, a strong antiapoptotic protein, in viral myocarditis (VMC). Methods and Results In the present study, we demonstrated that nAChRs, α3β4-nAChR subunits in particular, were present and upregulated in CVB3-infected neonatal rat cardiomyocytes (NRC) and H9c2 cells by RT-qPCR. The function of α3β4-nAChRs was next examined using its specific blocker α-CTX AuIB in vitro. The results of the TUNEL assay and western blot experiments showed that the block of α3β4-nAChRs abrogated nicotine-mediated protection of NRC from CVB3-induced apoptosis, and this effect displayed a substantial correlation with the protein expressions of pAkt, survivin, and Cleaved Caspase-3. Hence, the involvement of the PI3K/Akt pathway was further verified by LY294002, a selective inhibitor of PI3K. As a result, nicotine-mediated induction of pAkt and survivin was abolished by LY294002; meanwhile, apoptotic NRC were increased accompanied by an increase of Cleaved Caspase-3 expression. Regarding CVB3-infected BALB/c mice, the α-CTX AuIB- and LY294002-treated groups had a lower survival rate, deteriorative ventricular systolic function, and more severe inflammation than the nicotine-treated group and the modulation of pAkt, survivin, and Cleaved Caspase-3 protein expressions was similar to that in CVB3-infected NRC. In addition, we found that a nicotinic agonist reduced CVB3 replication in a dose-dependent manner in vitro, which indicates that nAChR activation may serve as a possible protection mechanism of CVB3-induced myocarditis. Conclusions Our study demonstrated that α3β4-nAChR subunits are essential in the nicotine-mediated antiapoptotic effect of protecting cardiomyocytes from CVB3-induced apoptosis in vivo and in vitro. This protection correlated with the PI3K/Akt pathway and the inducement of the antiapoptotic protein survivin. A combination of these mechanisms serves as a novel protective response to treat viral myocarditis.
Collapse
|
18
|
Rivadeneyra L, Charó N, Kviatcovsky D, de la Barrera S, Gómez RM, Schattner M. Role of neutrophils in CVB3 infection and viral myocarditis. J Mol Cell Cardiol 2018; 125:149-161. [PMID: 30393107 DOI: 10.1016/j.yjmcc.2018.08.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/05/2018] [Indexed: 12/11/2022]
Abstract
Coxsackievirus B3 (CVB3) is a globally prevalent enterovirus of the Picornaviridae family that is frequently associated with viral myocarditis (VM). Neutrophils, as first responders, may be key cells in determining viral disease outcomes; however, neutrophils have been poorly studied with respect to viral infection. Although neutrophils have been ascribed a relevant role in early cardiac inflammation, their precise role in CVB3 infection has not yet been evaluated. In this study, we aimed to determine if the interaction between human neutrophils and CVB3 could lead to viral replication and/or modulation of neutrophil survival and biological functions, and whether neutrophil depletion in a murine model has a beneficial or harmful effect on CVB3 infection. Our results show that CVB3 interacted with but did not replicate in human neutrophils. Neutrophils recognized CVB3 mainly through endosomal TLR-8, and infection triggered NFκB activation. Virus internalization resulted in increased cell survival, up-regulation of CD11b, enhanced adhesion to fibrinogen and fibronectin, and the secretion of IL-6, IL-1β, TNF-α, and IL-8. Supernatants from infected neutrophils exerted chemotactic activity partly mediated by IL-8. The infected neutrophils released myeloperoxidase and triggered neutrophil extracellular trap formation in the presence of TNF-α. In mice infected with CVB3, viral RNA was detected in neutrophils as well as in mononuclear cells. After neutrophil depletion, mice showed reduced VM reflected by a reduction in viral titers, cell exudates, and CCL-2 mRNA levels, as well as the abrogation of reactive cardiomyocyte hypertrophy. Our results indicate that neutrophils have relevant direct and indirect roles in the pathogenesis of CVB3-induced VM.
Collapse
Affiliation(s)
- Leonardo Rivadeneyra
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina.
| | - Nancy Charó
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina
| | - Denise Kviatcovsky
- Laboratory of Immunology of Respiratory Diseases, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina
| | - Silvia de la Barrera
- Laboratory of Immunology of Respiratory Diseases, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina.
| | - Ricardo Martín Gómez
- Biotechnology and Molecular Biology Institute, CONICET-UNLP, La Plata, Argentina.
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-ANM, Buenos Aires, Argentina.
| |
Collapse
|
19
|
Tian L, Yang Y, Li C, Chen J, Li Z, Li X, Li S, Wu F, Hu Z, Yang Z. The cytotoxicity of coxsackievirus B3 is associated with a blockage of autophagic flux mediated by reduced syntaxin 17 expression. Cell Death Dis 2018; 9:242. [PMID: 29445155 PMCID: PMC5833838 DOI: 10.1038/s41419-018-0271-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/08/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022]
Abstract
Coxsackievirus B3 (CVB3) is an important human pathogen linked to cardiac arrhythmias and acute heart failure. CVB3 infection has been reported to induce the formation of autophagosomes that support the viral replication in host cells. Interestingly, our study shows that the accumulation of autophagosomes during CVB3 infection is caused by a blockage of autophagosome–lysosome fusion rather than the induction of autophagosome biogenesis. Moreover, CVB3 decreases the transcription and translation of syntaxin 17 (STX17), a SNARE (soluble N-ethylmaleimide-sensitive factor activating protein receptor) protein involved in autophagosome–lysosome fusion. Overexpression of STX17 restored the autophagic flux, alleviated the virus-induced lysosomal dysfunction, and decreased the apoptosis induced by CVB3 infection in HeLa cells. Taken together, our results suggest that CVB3 infection impairs the autophagic flux by blocking autophagosome–lysosome fusion. These findings thus point to potential new therapeutic strategies targeting STX17 or autophagosome–lysosome fusion for treating CVB3-associated diseases.
Collapse
Affiliation(s)
- Lang Tian
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 410013, Changsha, China
| | - Yeyi Yang
- Department of Medicine, The Third Xiangya Hospital, Central South University, 410013, Changsha, China
| | - Chunyun Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 410013, Changsha, China
| | - Jia Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 410013, Changsha, China
| | - Zhuoying Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 410013, Changsha, China
| | - Xin Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 410013, Changsha, China
| | - Shentang Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 410013, Changsha, China
| | - Fang Wu
- Department of Pediatrics, Daping Hospital and Field Surgery Institute, Third Military Medical University, 400042, Chongqing, China
| | - Zhangxue Hu
- Department of Pediatrics, Daping Hospital and Field Surgery Institute, Third Military Medical University, 400042, Chongqing, China.
| | - Zuocheng Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 410013, Changsha, China.
| |
Collapse
|
20
|
Niu L, Li C, Wang Z, Xu H, An X. Effects of the MAPK pathway and the expression of CAR in a murine model of viral myocarditis. Exp Ther Med 2017; 13:230-234. [PMID: 28123495 PMCID: PMC5244858 DOI: 10.3892/etm.2016.3909] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 11/08/2016] [Indexed: 11/06/2022] Open
Abstract
The pathogenesis of viral myocarditis (VMC) is not fully understood. This study aimed to examine the relationship between coxsackie-adenovirus receptor (CAR) and the p38 mitogen activated protein kinase (MAPK) pathway mechanisms in a mouse model. Three groups of mice were established: 5 mice in a control group injected with saline, 15 in the model group injected with coxsackie virus B3 (CVB) and 15 in the intervention group injected with CVB3 but treated with the p38 MAPK inhibitor SB203580. Mice were sacrificed at days 1, 5, 10, 15 and 30 and cardiac tissues were isolated to perform the tests. Quantitative PCR and western blot analysis showed CAR mRNA and protein expression levels were highest in the model group at all time-points (P<0.05). The expression levels of p38 MAPK protein by western blot analysis at days 1, 5 and 10 were obviously higher in the model group (P>0.05). H&E staining used to observe myocardial pathological changes showed the inflammatory infiltration was also higher in the model group at all the time-points (P<0.05). Our results show a direct relationship between CAR and p38 MAPK levels, and since the p38 MAPK inhibitor treatment resulted in reduced levels of CAR as well as lower inflammatory infiltration, it is possible that the signaling pathway may mediate CAR expression during the pathogenesis of VMC.
Collapse
Affiliation(s)
- Ling Niu
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Chunli Li
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Zhenzhou Wang
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Hui Xu
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Xinjiang An
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
21
|
The Transcriptome of Rhabdomyosarcoma Cells Infected with Cytolytic and Non-Cytolytic Variants of Coxsackievirus B2 Ohio-1. PLoS One 2016; 11:e0164548. [PMID: 27760161 PMCID: PMC5070843 DOI: 10.1371/journal.pone.0164548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/07/2016] [Indexed: 11/19/2022] Open
Abstract
The transcriptomes of cells infected with lytic and non-lytic variants of coxsackievirus B2 Ohio-1 (CVB2O) were analyzed using next generation sequencing. This approach was selected with the purpose of elucidating the effects of lytic and non-lytic viruses on host cell transcription. Total RNA was extracted from infected cells and sequenced. The resulting reads were subsequently mapped against the human and CVB2O genomes. The amount of intracellular RNA was measured, indicating lower proportions of human RNA in the cells infected with the lytic virus compared to the non-lytic virus after 48 hours. This may be explained by reduced activity of the cellular transcription/translation machinery in lytic enteroviral replication due to activities of the enteroviral proteases 2A and/or 3C. Furthermore, differential expression in the cells infected with the two virus variants was identified and a number of transcripts were singled out as possible answers to the question of how the viruses interact with the host cells, resulting in lytic or non-lytic infections.
Collapse
|
22
|
Harris KG, Coyne CB. Unc93b Induces Apoptotic Cell Death and Is Cleaved by Host and Enteroviral Proteases. PLoS One 2015; 10:e0141383. [PMID: 26509685 PMCID: PMC4624986 DOI: 10.1371/journal.pone.0141383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/06/2015] [Indexed: 02/02/2023] Open
Abstract
Unc93b is an endoplasmic reticulum (ER)-resident transmembrane protein that serves to bind and traffic toll-like receptors (TLRs) from the ER to their appropriate subcellular locations for ligand sensing. Because of its role in TLR trafficking, Unc93b is necessary for an effective innate immune response to coxsackievirus B3 (CVB), a positive-sense single stranded RNA virus belonging to the enterovirus family. Here, we show that Unc93b is cleaved by a CVB-encoded cysteine protease (3Cpro) during viral replication. Further, we define a role for Unc93b in the induction of apoptotic cell death and show that expression of wild-type Unc93b, but not a mutant incapable of binding TLRs or exiting the ER (H412R), induces apoptosis. Furthermore, we show that cellular caspases activated during apoptosis directly cleave Unc93b. Interestingly, we show that the 3Cpro- and caspase-mediated cleavage of Unc93b both occur within ten amino acids in the distal N-terminus of Unc93b. Mechanistically, neither caspase-mediated nor 3Cpro-mediated cleavage of Unc93b altered its trafficking function, inhibited its role in facilitating TLR3 or TLR8 signaling, or altered its apoptosis-inducing effects. Taken together, our studies show that Unc93b is targeted by both viral- and host cell-specific proteases and identify a function of Unc93b in the induction of apoptotic cell death.
Collapse
Affiliation(s)
- Katharine G. Harris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Carolyn B. Coyne
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
23
|
Abstract
Type 1 diabetes (T1D) results from genetic predisposition and environmental factors leading to the autoimmune destruction of pancreatic beta cells. Recently, a rapid increase in the incidence of childhood T1D has been observed worldwide; this is too fast to be explained by genetic factors alone, pointing to the spreading of environmental factors linked to the disease. Enteroviruses (EVs) are perhaps the most investigated environmental agents in relationship to the pathogenesis of T1D. While several studies point to the likelihood of such correlation, epidemiological evidence in its support is inconclusive or in some instances even against it. Hence, it is still unknown if and how EVs are involved in the development of T1D. Here we review recent findings concerning the biology of EV in beta cells and the potential implications of this knowledge for the understanding of beta cell dysfunction and autoimmune destruction in T1D.
Collapse
Affiliation(s)
- Antje Petzold
- />Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscherstr.74, 01307 Dresden, Germany
- />German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Michele Solimena
- />Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscherstr.74, 01307 Dresden, Germany
- />German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- />Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Klaus-Peter Knoch
- />Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscherstr.74, 01307 Dresden, Germany
- />German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
24
|
Wen X, Cheng A, Wang M, Jia R, Zhu D, Chen S, Liu M, Sun K, Yang Q, Wu Y, Chen X. Recent advances from studies on the role of structural proteins in enterovirus infection. Future Microbiol 2015; 10:1529-42. [DOI: 10.2217/fmb.15.62] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Enteroviruses are a large group of small nonenveloped viruses that cause common and debilitating illnesses affecting humans and animals worldwide. The capsid composed by viral structural proteins packs the RNA genome. It is becoming apparent that structural proteins of enteroviruses play versatile roles in the virus–host interaction in the viral life cycle, more than just a shell. Furthermore, structural proteins to some extent may be associated with viral virulence and pathogenesis. Better understanding the roles of structural proteins in enterovirus infection may lead to the development of potential antiviral strategies. Here, we discuss recent advances from studies on the role of structural proteins in enterovirus infection and antiviral therapeutics targeted structural proteins.
Collapse
Affiliation(s)
- Xingjian Wen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Engineering & Technology Center for Laboratory Animals of Sichuan Agricultural University, Ya'an, Sichuan 625014, China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Dekang Zhu
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Kunfeng Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| | - Xiaoyue Chen
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
- Key Laboratory of Animal Disease & Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan 611130, PR China
| |
Collapse
|
25
|
Li X, Xia Y, Huang S, Liu F, Ying Y, Xu Q, Liu X, Jin G, Papasian CJ, Chen J, Fu M, Huang X. Identification of the interaction of VP1 with GM130 which may implicate in the pathogenesis of CVB3-induced acute pancreatitis. Sci Rep 2015; 5:13324. [PMID: 26314804 PMCID: PMC4551966 DOI: 10.1038/srep13324] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 07/21/2015] [Indexed: 11/09/2022] Open
Abstract
Coxsackievirus B3 (CVB3) is a causative agent of viral myocarditis, pancreatitis, and meningitis in humans. Although the susceptibility of CVB3-induced acute pancreatitis is age-dependent, the underlying mechanisms remain unclear. Here we identified the host factor Golgi matrix protein 130 (GM130) as a novel target of CVB3 during CVB3-induced acute pancreatitis. The viral protein VP1 interacted with GM130, disrupted GM130-GRASP65 complexes, and caused GM130 degradation, which may lead to disruption of the Golgi ribbon and development of acute pancreatitis in mice. Interestingly, the expression level of GM130 in mouse pancreas was age-dependent, which was nicely correlated with the age-associated susceptibility of CVB3-induced acute pancreatitis. Furthermore, interference RNA-mediated knockdown of GM130 significantly reduced CVB3 replication in HeLa cells. Taken together, the study identified GM130 as a novel target of CVB3, which may implicate in the pathogenesis of CVB3-induced acute pancreatitis.
Collapse
Affiliation(s)
- Xiuzhen Li
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Yanhua Xia
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Shengping Huang
- Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, MO, USA
| | - Fadi Liu
- Children’s Hospital of Jiangxi Province, Nanchang, Jiangxi, China
| | - Ying Ying
- Department of Pathophysiology, School of Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Qiufang Xu
- Shanghai Qingpu Center for Disease Control and Prevention, Shanghai, China
| | - Xin Liu
- Children’s Hospital of Jiangxi Province, Nanchang, Jiangxi, China
| | - Guili Jin
- The affiliated hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Christopher J. Papasian
- Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, MO, USA
| | - Jack Chen
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - Mingui Fu
- Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, MO, USA
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
26
|
Harris KG, Coyne CB. Death waits for no man--does it wait for a virus? How enteroviruses induce and control cell death. Cytokine Growth Factor Rev 2014; 25:587-96. [PMID: 25172372 DOI: 10.1016/j.cytogfr.2014.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/05/2014] [Indexed: 12/29/2022]
Abstract
Enteroviruses (EVs) are the most common human viral pathogens. They cause a variety of pathologies, including myocarditis and meningoencephalopathies, and have been linked to the onset of type I diabetes. These pathologies result from the death of cells in the myocardium, central nervous system, and pancreas, respectively. Understanding the role of EVs in inducing cell death is crucial to understanding the etiologies of these diverse pathologies. EVs both induce and delay host cell death, and their exquisite control of this balance is crucial for their success as human viral pathogens. Thus, EVs are tightly involved with cell death signaling pathways and interact with host cell signaling at multiple points. Here, we review the literature detailing the mechanisms of EV-induced cell death. We discuss the mechanisms by which EVs induce cell death, the signaling pathways involved in these pathways, and the strategies by which EVs antagonize cell death pathways. We also discuss the role of cell death in both the resulting pathology in the host and in the facilitation of viral spread.
Collapse
Affiliation(s)
- Katharine G Harris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Carolyn B Coyne
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219, United States.
| |
Collapse
|
27
|
Sheng XD, Zhang WP, Zhang QR, Gu CQ, Hu XY, Cheng GF. Apoptosis induction in duck tissues during duck hepatitis A virus type 1 infection. Poult Sci 2014; 93:527-34. [PMID: 24604844 DOI: 10.3382/ps.2013-03510] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
To investigate the role of apoptosis in duck viral hepatitis pathogenesis, 4- and 21-d-old ducks were inoculated with duck hepatitis A virus serotype 1 and killed at 2, 6, 12, 24, and 48 h postinfection. TdT-mediated dUTP nick-end labeling was used to detect apoptosis cells. Expression profiles of apoptosis-related genes including caspase-3, -8, -9, and Bcl-2 in spleen, bursa of Fabricius, liver, and the quantity of virus in blood were examined using real-time PCR. The TdT-mediated dUTP nick-end labeling analysis indicated there was a significant difference of apoptotic cells between treatments and controls. The same difference also appeared in virus amount variation in blood during infection. Gene expression analysis revealed that the apoptosis-related gene expression profile was different in the 2 groups, and also different between various organs. This study suggested that apoptosis may play an important role in duck hepatitis A virus serotype 1 infection, and apoptosis suppression might facilitate virus multiplication, resulting in the highest virus concentration in the host.
Collapse
Affiliation(s)
- X D Sheng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | | | | | | | | | | |
Collapse
|
28
|
Characterization of coxsackievirus B3 replication in human umbilical vein endothelial cells. Med Microbiol Immunol 2014; 203:217-29. [DOI: 10.1007/s00430-014-0333-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
|
29
|
Zins K, Pomyje J, Hofer E, Abraham D, Lucas T, Aharinejad S. Egr-1 upregulates Siva-1 expression and induces cardiac fibroblast apoptosis. Int J Mol Sci 2014; 15:1538-53. [PMID: 24451137 PMCID: PMC3907885 DOI: 10.3390/ijms15011538] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 12/21/2013] [Accepted: 01/13/2014] [Indexed: 12/24/2022] Open
Abstract
The early growth response transcription factor Egr-1 controls cell specific responses to proliferation, differentiation and apoptosis. Expression of Egr-1 and downstream transcription is closely controlled and cell specific upregulation induced by processes such as hypoxia and ischemia has been previously linked to multiple aspects of cardiovascular injury. In this study, we showed constitutive expression of Egr-1 in cultured human ventricular cardiac fibroblasts, used adenoviral mediated gene transfer to study the effects of continuous Egr-1 overexpression and studied downstream transcription by Western blotting, immunohistochemistry and siRNA transfection. Apoptosis was assessed by fluorescence microscopy and flow cytometry in the presence of caspase inhibitors. Overexpression of Egr-1 directly induced apoptosis associated with caspase activation in human cardiac fibroblast cultures in vitro assessed by fluorescence microscopy and flow cytometry. Apoptotic induction was associated with a caspase activation associated loss of mitochondrial membrane potential and transient downstream transcriptional up-regulation of the pro-apoptotic gene product Siva-1. Suppression of Siva-1 induction by siRNA partially reversed Egr-1 mediated loss of cell viability. These findings suggest a previously unknown role for Egr-1 and transcriptional regulation of Siva-1 in the control of cardiac accessory cell death.
Collapse
Affiliation(s)
- Karin Zins
- Laboratory for Molecular Cellular Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna A-1090, Austria.
| | - Jiri Pomyje
- Molecular Vascular Biology, Department of Vascular Biology and Thrombosis Research, Vienna Competence Center, Vienna Medical University, Vienna A-1090, Austria.
| | - Erhard Hofer
- Molecular Vascular Biology, Department of Vascular Biology and Thrombosis Research, Vienna Competence Center, Vienna Medical University, Vienna A-1090, Austria.
| | - Dietmar Abraham
- Laboratory for Molecular Cellular Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna A-1090, Austria.
| | - Trevor Lucas
- Laboratory for Molecular Cellular Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna A-1090, Austria.
| | - Seyedhossein Aharinejad
- Laboratory for Molecular Cellular Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna A-1090, Austria.
| |
Collapse
|
30
|
Gladue DP, Baker-Bransetter R, Holinka LG, Fernandez-Sainz IJ, O’Donnell V, Fletcher P, Lu Z, Borca MV. Interaction of CSFV E2 protein with swine host factors as detected by yeast two-hybrid system. PLoS One 2014; 9:e85324. [PMID: 24416391 PMCID: PMC3885694 DOI: 10.1371/journal.pone.0085324] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/26/2013] [Indexed: 01/31/2023] Open
Abstract
E2 is one of the envelope glycoproteins of pestiviruses, including classical swine fever virus (CSFV) and bovine viral diarrhea virus (BVDV). E2 is involved in several critical functions, including virus entry into target cells, induction of a protective immune response and virulence in swine. However, there is no information regarding any host binding partners for the E2 proteins. Here, we utilized the yeast two-hybrid system and identified fifty-seven host proteins as positive binding partners which bound E2 from both CSFV and BVDV with the exception of two proteins that were found to be positive for binding only to CSFV E2. Alanine scanning of CSFV E2 demonstrated that the binding sites for these cellular proteins on E2 are likely non-linear binding sites. The possible roles of the identified host proteins are discussed as the results presented here will be important for future studies to elucidate mechanisms of host protein-virus interactions during pestivirus infection. However, due to the limitations of the yeast two hybrid system, the proteins identified is not exhaustive and each interaction identified needs to be confirmed by independent experimental approaches in the context of virus-infected cells before any definitive conclusion can be drawn on relevance for the virus life cycle.
Collapse
Affiliation(s)
- Douglas P. Gladue
- Plum Island Animal Disease Center, Agriculture Research Service, United States Department of Agriculture, Greenport, New York, United States of America
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, Connecticut, United States of America
| | - Ryan Baker-Bransetter
- Plum Island Animal Disease Center, Agriculture Research Service, United States Department of Agriculture, Greenport, New York, United States of America
| | - Lauren G. Holinka
- Plum Island Animal Disease Center, Agriculture Research Service, United States Department of Agriculture, Greenport, New York, United States of America
| | - Ignacio J. Fernandez-Sainz
- Plum Island Animal Disease Center, Agriculture Research Service, United States Department of Agriculture, Greenport, New York, United States of America
| | - Vivian O’Donnell
- Plum Island Animal Disease Center, Agriculture Research Service, United States Department of Agriculture, Greenport, New York, United States of America
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, Connecticut, United States of America
| | - Paige Fletcher
- Plum Island Animal Disease Center, Agriculture Research Service, United States Department of Agriculture, Greenport, New York, United States of America
| | - Zhiqiang Lu
- Plum Island Animal Disease Center, Department of Homeland Security, Greenport, New York, United States of America
| | - Manuel V. Borca
- Plum Island Animal Disease Center, Agriculture Research Service, United States Department of Agriculture, Greenport, New York, United States of America
- * E-mail:
| |
Collapse
|
31
|
Chen JY, Yang LX, Huang ZF. The N-terminal 33 amino acid domain of Siva-1 is sufficient for nuclear localization. Braz J Med Biol Res 2013; 46:1021-1027. [PMID: 24345910 PMCID: PMC3935273 DOI: 10.1590/1414-431x20132833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 07/29/2013] [Indexed: 01/21/2023] Open
Abstract
Siva-1 induces apoptosis in multiple pathological processes and plays an important role in the suppression of tumor metastasis, protein degradation, and other functions. Although many studies have demonstrated that Siva-1 functions in the cytoplasm, a few have found that Siva-1 can relocate to the nucleus. In this study, we found that the first 33 amino acid residues of Siva-1 are required for its nuclear localization. Further study demonstrated that the green fluorescent protein can be imported into the nucleus after fusion with these 33 amino acid residues. Other Siva-1 regions and domains showed less effect on Siva-1 nuclear localization. By site-mutagenesis of all of these 33 amino acid residues, we found that mutants of the first 1-18 amino acids affected Siva-1 nuclear compartmentalization but could not complete this localization independently. In summary, we demonstrated that the N-terminal 33 amino acid residues were sufficient for Siva-1 nuclear localization, but the mechanism of this translocation needs additional investigation.
Collapse
Affiliation(s)
- J Y Chen
- Sun Yat-sen University, Zhongshan School of Medicine, Institute of Human Virology, Guangzhou, China
| | - L X Yang
- Sun Yat-sen University, Zhongshan School of Medicine, Institute of Human Virology, Guangzhou, China
| | - Z F Huang
- Sun Yat-sen University, Zhongshan School of Medicine, Institute of Human Virology, Guangzhou, China
| |
Collapse
|
32
|
Chen Z, Yang L, Liu Y, Tang A, Li X, Zhang J, Yang Z. LY294002 and Rapamycin promote coxsackievirus-induced cytopathic effect and apoptosis via inhibition of PI3K/AKT/mTOR signaling pathway. Mol Cell Biochem 2013; 385:169-77. [PMID: 24072614 DOI: 10.1007/s11010-013-1825-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/13/2013] [Indexed: 01/23/2023]
Abstract
Coxsackievirus B3 (CVB3) is a common human pathogen for acute myocarditis, pancreatitis, non-septic meningitis, and encephalitis; it induces a direct cytopathic effect (CPE) and apoptosis on infected cells. The Phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT/PKB)/mammalian target of Rapamycin (mTOR) signaling pathway regulates several cellular processes and it is one of the most important pathways in human networks. However, the effect and mechanism of PI3K/AKT/mTOR signaling pathway in CVB3 infected cells are poorly understood. In this study, we demonstrate that inhibition of PI3K/AKT/mTOR signaling pathway increased CVB3-induced CPE and apoptosis in HeLa cells. The activity of downstream targets of PI3K and mTOR is attenuated after CVB3 infection and inhibitors of PI3K and mTOR made their activity to decrease more significantly. We further show that LY294002 and Rapamycin, the inhibitor of PI3K and mTOR respectively, promote CVB3-induced CPE and apoptosis. Taken together, these data illustrate a new and imperative role for PI3K/AKT/mTOR signaling in CVB3 infection in HeLa cells and suggest an useful approach for the therapy of CVB3 infection.
Collapse
Affiliation(s)
- Zhiheng Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China,
| | | | | | | | | | | | | |
Collapse
|
33
|
Liu JY, Wang SM, Chen IC, Yu CK, Liu CC. Hepatic damage caused by coxsackievirus B3 is dependent on age-related tissue tropisms associated with the coxsackievirus-adenovirus receptor. Pathog Dis 2013; 68:52-60. [DOI: 10.1111/2049-632x.12044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 03/25/2013] [Accepted: 04/16/2013] [Indexed: 11/28/2022] Open
Affiliation(s)
- Jung-Yen Liu
- Department of Microbiology & Immunology; National Cheng Kung University Medical College; Tainan; Taiwan
| | | | - I-Chun Chen
- Institute of Basic Medical Sciences; National Cheng Kung University; Tainan; Taiwan
| | | | | |
Collapse
|
34
|
Abstract
Capsid proteins are obligatory components of infectious virions. Their primary structural function is to protect viral genomes during entry and exit from host cells. Evidence suggests that these proteins can also modulate the activity and specificity of viral replication complexes. More recently, it has become apparent that they play critical roles at the virus–host interface. Here, we discuss how capsid proteins of RNA viruses interact with key host cell proteins and pathways to modulate cell physiology in order to benefit virus replication. Capsid–host cell interactions may also have implications for viral disease. Understanding how capsids regulate virus–host interactions may lead to the development of novel antiviral therapies based on targeting the activities of cellular proteins.
Collapse
Affiliation(s)
- Steven Willows
- Department of Cell Biology, University of Alberta, 5–14 Medical Sciences Building, Edmonton, T6G 2H7, Canada
| | - Shangmei Hou
- Department of Cell Biology, University of Alberta, 5–14 Medical Sciences Building, Edmonton, T6G 2H7, Canada
| | - Tom C Hobman
- Department of Li Ka Shing Institute of Virology, University of Alberta, 5–14 Medical Sciences Building, Edmonton, T6G 2H7, Canada
| |
Collapse
|
35
|
Rassmann A, Martin U, Saluz HP, Peter S, Munder T, Henke A. Identification of gene expression profiles in HeLa cells and HepG2 cells infected with Coxsackievirus B3. J Virol Methods 2013; 187:190-4. [DOI: 10.1016/j.jviromet.2012.08.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 08/20/2012] [Accepted: 08/22/2012] [Indexed: 10/27/2022]
|
36
|
Mishra P, Samanta L. Oxidative stress and heart failure in altered thyroid States. ScientificWorldJournal 2012; 2012:741861. [PMID: 22649319 PMCID: PMC3354657 DOI: 10.1100/2012/741861] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/25/2011] [Indexed: 02/07/2023] Open
Abstract
Increased or reduced action of thyroid hormone on certain molecular pathways in the heart and vasculature causes relevant cardiovascular derangements. It is well established that hyperthyroidism induces a hyperdynamic cardiovascular state, which is associated with a faster heart rate, enhanced left ventricular systolic and diastolic function whereas hypothyroidism is characterized by the opposite changes. Hyperthyroidism and hypothyroidism represent opposite clinical conditions, albeit not mirror images. Recent experimental and clinical studies have suggested the involvement of ROS tissue damage under altered thyroid status. Altered-thyroid state-linked changes in heart modify their susceptibility to oxidants and the extent of the oxidative damage they suffer following oxidative challenge. Chronic increase in the cellular levels of ROS can lead to a catastrophic cycle of DNA damage, mitochondrial dysfunction, further ROS generation and cellular injury. Thus, these cellular events might play an important role in the development and progression of myocardial remodeling and heart failure in altered thyroid states (hypo- and hyper-thyroidism). The present review aims at elucidating the various signaling pathways mediated via ROS and their modulation under altered thyroid state and the possibility of antioxidant therapy.
Collapse
Affiliation(s)
- Pallavi Mishra
- Department of Zoology, Utkal University, Odisha, Bhubaneswar 751004, India
| | | |
Collapse
|
37
|
Wang YC, Yi TY, Lin KH. In VitroActivity ofParis polyphyllaSmith Against Enterovirus 71 and Coxsackievirus B3 and Its Immune Modulation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 39:1219-34. [DOI: 10.1142/s0192415x11009512] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Enterovirus 71 (EV71) and coxsackievirus B3 (CVB3) have resulted in severe pathogenesis caused by the host's immune response, including the cytokine cascade. Paris polyphylla Smith is a folk medicinal plant in Asia traditionally prescribed for the reduction of pain and elimination of poisoning. In this study, we investigated the anti-EV71 and CVB3 activity of P. polyphylla Smith as well as its immune modulation. The IC50for the P. polyphylla Smith 95% ethanol extract against EV71 and CVB3 were 12.5–23% and 99–156% of that of ribavirin, a positive control. Prevention of viral infection, viral inactivation, and anti-viral replication effects against both EV71 and CVB3 were demonstrated by the extract, the anti-viral replication effect being dominant. The extract significantly increased IL-6 production in both EV71- and CVB3-infected cells. A high correlation was possibly demonstrated between the high amounts of IL-6 induction in the EV71 and CVB3-infected cells and the anti-viral replication activity of the extract. In conclusion, good anti-EV71 and CVB3 activity was observed in the P. polyphylla Smith 95% ethanol extract. The high amounts of IL-6 induction in the virus-infected cells played a key role in the anti-viral activity of the extract.
Collapse
Affiliation(s)
- Yuan-Chuen Wang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Tsu-Yi Yi
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Kuei-Hsiang Lin
- Department of Clinical Laboratory, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
38
|
Multifaceted functions of Siva-1: more than an Indian God of Destruction. Protein Cell 2012; 3:117-22. [PMID: 22426980 DOI: 10.1007/s13238-012-2018-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/04/2012] [Indexed: 10/28/2022] Open
Abstract
Siva-1, as a p53-inducible gene, has been shown to induce extensive apoptosis in a number of different cell lines. Recent evidence suggests that Siva-1 functions as a part of the auto-regulatory feedback loop that restrains p53 through facilitating Mdm2-mediated p53 degradation. Also, Siva-1 plays an important role in suppressing tumor metastasis. Here we review the current understanding of Siva-1-mediated apoptotic signaling pathway. We also add comments on the p53-Siva-1 feedback loop, the novel function of Siva-1 in suppressing tumor metastasis, and their potential implications.
Collapse
|
39
|
Wiesener N, Schütze T, Lapp S, Lehmann M, Jarasch-Althof N, Wutzler P, Henke A. Analysis of different DNA vaccines for protection of experimental influenza A virus infection. Viral Immunol 2011; 24:321-30. [PMID: 21830903 DOI: 10.1089/vim.2011.0001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Influenza viruses cause acute respiratory infections in humans that result in significant excessive morbidity and mortality rates every year. Current vaccines are limited in several aspects, including laborious manufacturing technology, non-sufficient efficacy, and time-consuming adjustments to new emerging virus variants. An alternative vaccine approach utilizes plasmid DNA encoding influenza virus antigens. Previous experiments have evaluated the protective efficacy of DNA vaccines expressing variable as well as conserved antigens. In this present study, several different combinations of influenza A virus (IAV) HA, NA, M1, M2, NS1, NS2, and NP sequences were cloned into the plasmid pVIVO, which allows the independent expression of two genes separately. These DNA vaccines were administered to induce protection against a lethal IAV infection, and to reduce immunopathology in lung tissue of surviving animals. The highest efficacy was provided by vaccines expressing HA and NA, as well as a mixture of plasmids encoding HA, NA, M1, M2, NS1, NS2, and NP (Mix). Three days post-infection, more than a 99.99% reduction of viral load and no inflammation was achieved in lung tissue of pVIVO/HA-NA-vaccinated mice. Animals vaccinated with pVIVO/HA-NA, pVIVO/HA-M2, or vaccine Mix, survived a lethal challenge with minor or no obvious pathologic abnormities in the lungs. All other surviving mice revealed extensive changes in the lung tissue, indicating possibly an ongoing bronchiolitis obliterans. In addition, pVIVO/HA-NA and the vaccine Mix were also protective against a heterologous IAV infection. Taken together, next to all combinations of different DNA vaccines, the intramuscular application of pVIVO/HA-NA was the most efficient procedure to decrease virus replication and to prevent immunopathology in lung tissue of IAV-infected mice.
Collapse
Affiliation(s)
- Nadine Wiesener
- Department of Virology and Antiviral Therapy, University Hospital Jena, Friedrich Schiller University, Hans-Knöll-Strasse 2, Jena, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Barkinge JL, Gudi R, Sarah H, Chu F, Borthakur A, Prabhakar BS, Prasad KVS. The p53-induced Siva-1 plays a significant role in cisplatin-mediated apoptosis. J Carcinog 2011; 8:2. [PMID: 19240372 PMCID: PMC2678867 DOI: 10.4103/1477-3163.45389] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: The pro-apoptotic protein Siva-1 functions in both extrinsic and intrinsic cell death signaling; however, the exact contribution of the endogenous Siva-1 to DNA damage-induced apoptosis is unclear. Using cisplatin, a chemotherapeutic drug, to induce DNA damage and cell death, we determined the role of Siva-1. Methods: Cisplatin treated HCT116 colorectal carcinoma cells (p53+/+ and -/-) were used in the study. With the help of recombinant lentivirus that can express siSiva (siRNA that specifically targets Siva-1), we also generated Siva-1 knockdown HCT116 cells. Apoptosis was determined by tetramethyl rhodamine methyl ester (TMRM) staining and propidium iodide (PI) staining. Results: Treatment with cisplatin induced Siva-1 expression in a p53 dependent manner. In Siva-1 knockdown p53+/+ HCT116 colorectal carcinoma cells, loss of Siva-1 expression conferred significant resistance to cisplatin-induced apoptosis. Although Siva-1 levels were positively regulated by p53, Siva-1-induced apoptosis did not require p53. Despite the fact that Siva-1 lacks even a minimal BH3 domain, similar to other proapoptotic Bcl2 family members induced by p53, we showed that Siva-1 mediated apoptosis is characterized by Bax oligomerization and cytochrome c leakage from mitochondria. The putative amphipathic helical region in Siva-1 (SAH) appeared to function analogously to a BH3 domain. Conclusion: The p53 induced Siva-1 is one of the effector molecules, which plays a significant role in DNA damage-induced cell death.
Collapse
Affiliation(s)
- John L Barkinge
- Department of Microbiology and Immunology, University of Illinois at Chicago, 835 S. Wolcott Ave., MC 790, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Yi L, Lu J, Kung HF, He ML. The virology and developments toward control of human enterovirus 71. Crit Rev Microbiol 2011; 37:313-27. [PMID: 21651436 DOI: 10.3109/1040841x.2011.580723] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Enterovirus 71 (EV71), a member of the Enterovirus genus in the Picornaviridae family, was first recognized as a dermotrophic virus that usually cause mild, self-limiting hand-foot-and-mouth disease (HFMD). However, EV71 infection can sometimes induce a variety of severe neurological complications and even death. Current large outbreaks of EV71 make this virus being a major public health issue. Intense effort has been made to address its underlying pathogenesis and to develop effective means for combating EV71 infections. Here, we aimed to provide an overview of cellular mechanisms underlying EV71 infection and to assess potential agents for prevention and treatment of EV71 infections.
Collapse
Affiliation(s)
- Lina Yi
- Stanley Ho Center for Emerging Infectious Diseases, School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
42
|
Mukherjee A, Morosky SA, Delorme-Axford E, Dybdahl-Sissoko N, Oberste MS, Wang T, Coyne CB. The coxsackievirus B 3C protease cleaves MAVS and TRIF to attenuate host type I interferon and apoptotic signaling. PLoS Pathog 2011; 7:e1001311. [PMID: 21436888 PMCID: PMC3059221 DOI: 10.1371/journal.ppat.1001311] [Citation(s) in RCA: 243] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 02/02/2011] [Indexed: 02/06/2023] Open
Abstract
The host innate immune response to viral infections often involves the activation of parallel pattern recognition receptor (PRR) pathways that converge on the induction of type I interferons (IFNs). Several viruses have evolved sophisticated mechanisms to attenuate antiviral host signaling by directly interfering with the activation and/or downstream signaling events associated with PRR signal propagation. Here we show that the 3C(pro) cysteine protease of coxsackievirus B3 (CVB3) cleaves the innate immune adaptor molecules mitochondrial antiviral signaling protein (MAVS) and Toll/IL-1 receptor domain-containing adaptor inducing interferon-beta (TRIF) as a mechanism to escape host immunity. We found that MAVS and TRIF were cleaved in CVB3-infected cells in culture. CVB3-induced cleavage of MAVS and TRIF required the cysteine protease activity of 3C(pro), occurred at specific sites and within specialized domains of each molecule, and inhibited both the type I IFN and apoptotic signaling downstream of these adaptors. 3C(pro)-mediated MAVS cleavage occurred within its proline-rich region, led to its relocalization from the mitochondrial membrane, and ablated its downstream signaling. We further show that 3C(pro) cleaves both the N- and C-terminal domains of TRIF and localizes with TRIF to signalosome complexes within the cytoplasm. Taken together, these data show that CVB3 has evolved a mechanism to suppress host antiviral signal propagation by directly cleaving two key adaptor molecules associated with innate immune recognition.
Collapse
Affiliation(s)
- Amitava Mukherjee
- Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stefanie A. Morosky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth Delorme-Axford
- Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Naomi Dybdahl-Sissoko
- Picornavirus Laboratory, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - M. Steven Oberste
- Picornavirus Laboratory, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Tianyi Wang
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Carolyn B. Coyne
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
43
|
Jarasch-Althof N, Wiesener N, Schmidtke M, Wutzler P, Henke A. Antibody-dependent enhancement of coxsackievirus B3 infection of primary CD19+ B lymphocytes. Viral Immunol 2011; 23:369-76. [PMID: 20712481 DOI: 10.1089/vim.2010.0018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Coxsackievirus B3 (CVB3) is associated with several different acute and chronic forms of human disease, including myocarditis, aseptic meningitis, and pancreatitis. Moreover, CVB3 also infects immune cells like CD19+ B lymphocytes, but the viral uptake mechanism into these cells is not well understood. Therefore, primary murine and human CD19+ B cells were isolated by magnetic-activated cell separation technology and analyzed for virus receptor expression, antibody-dependent enhancement of viral infection, and different cellular surface proteins, that might be involved in mechanisms of viral uptake. Western blot analysis of these cells revealed no significant expression of the coxsackievirus-adenovirus receptor CAR. But incubation of CVB3 with serum dilutions, which exhibited binding but not neutralizing characteristics, increased viral uptake and replication significantly in a dose-dependent manner. Viral entry was reduced when Fc portions of immunoglobulins were blocked by protein A treatment. Moreover, the classical complement system rather than Fc-gamma-receptor-mediated mechanisms could be involved in viral uptake. Taken together, these data suggest an antibody-dependent enhancement of CVB3 infection of primary murine and human CD19+ B cells.
Collapse
Affiliation(s)
- Nadine Jarasch-Althof
- Department of Virology and Antiviral Therapy, University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | | | | | | |
Collapse
|
44
|
Wiesener N, Zimmer C, Jarasch-Althof N, Wutzler P, Henke A. Therapy of experimental influenza virus infection with pyrrolidine dithiocarbamate. Med Microbiol Immunol 2010; 200:115-26. [PMID: 21174118 DOI: 10.1007/s00430-010-0182-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Indexed: 12/27/2022]
Abstract
The search for new antiviral strategies to treat influenza A virus (IAV) infections is one major international health care activity. Hereby, the IAV-caused misuse of cellular nuclear factor kappa B (NF-κB) signaling pathways in infected cells represents one target for antiviral therapy. In the present study, pyrrolidine dithiocarbamate (PDTC), which is known as an antioxidant and as an inhibitor of IAV-induced NF-κB activation, was studied in vivo. After the antiviral activity of PDTC was confirmed in MDCK cells, mice-infected with the mouse-adapted strain of IAV A/PR/8/34 (H1N1)-were treated intraperitoneally simultaneously with PDTC (75, 150, 200 mg/kg body weight). The influence of PDTC administrations was evaluated on viral replication and inflammatory reactions in lung tissue up to 14 days postinfection (p. i.). This therapy increased survival up to 80% and reduced IAV-caused weight loss and viral replication in lung tissue in a dose-dependent manner. Protective effects were less pronounced, if the therapy started later on during an ongoing IAV infection. In addition, simultaneous PDTC treatment also limited IAV-caused infiltration of immune cells as well as local interferon-γ expression in lung tissue. These results imply that PDTC decreases IAV-caused disease in mice significantly. Therefore, the development of drugs like PDTC that interfere with NF-κB signaling may represent a modern focus of anti-IAV therapy.
Collapse
Affiliation(s)
- Nadine Wiesener
- Department of Virology and Antiviral Therapy, Jena University Hospital, Friedrich Schiller University, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | | | | | | | | |
Collapse
|
45
|
Shimoda HK, Shide K, Kameda T, Matsunaga T, Shimoda K. Tyrosine kinase 2 interacts with the proapoptotic protein Siva-1 and augments its apoptotic functions. Biochem Biophys Res Commun 2010; 400:252-7. [DOI: 10.1016/j.bbrc.2010.08.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 08/15/2010] [Indexed: 01/16/2023]
|
46
|
A single coxsackievirus B2 capsid residue controls cytolysis and apoptosis in rhabdomyosarcoma cells. J Virol 2010; 84:5868-79. [PMID: 20375176 DOI: 10.1128/jvi.02383-09] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Coxsackievirus B2 (CVB2), one of six human pathogens of the group B coxsackieviruses within the enterovirus genus of Picornaviridae, causes a wide spectrum of human diseases ranging from mild upper respiratory illnesses to myocarditis and meningitis. The CVB2 prototype strain Ohio-1 (CVB2O) was originally isolated from a patient with summer grippe in the 1950s. Later on, CVB2O was adapted to cytolytic replication in rhabdomyosarcoma (RD) cells. Here, we present analyses of the correlation between the adaptive mutations of this RD variant and the cytolytic infection in RD cells. Using reverse genetics, we identified a single amino acid change within the exposed region of the VP1 protein (glutamine to lysine at position 164) as the determinant for the acquired cytolytic trait. Moreover, this cytolytic virus induced apoptosis, including caspase activation and DNA degradation, in RD cells. These findings contribute to our understanding of the host cell adaptation process of CVB2O and provide a valuable tool for further studies of virus-host interactions.
Collapse
|
47
|
Gomes R, Guerra-Sá R, Arruda E. Coxsackievirus B5 induced apoptosis of HeLa cells: effects on p53 and SUMO. Virology 2009; 396:256-63. [PMID: 19906394 DOI: 10.1016/j.virol.2009.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 09/27/2009] [Accepted: 10/03/2009] [Indexed: 12/21/2022]
Abstract
Coxsackievirus B5 (CVB5), a human enterovirus of the family Picornaviridae, is a frequent cause of acute and chronic human diseases. The pathogenesis of enteroviral infections is not completely understood, and the fate of the CVB5-infected cell has a pivotal role in this process. We have investigated the CVB5-induced apoptosis of HeLa cells and found that it happens by the intrinsic pathway by a mechanism dependent on the ubiquitin-proteasome system, associated with nuclear aggregation of p53. Striking redistribution of both SUMO and UBC9 was noted at 4 h post-infection, simultaneously with a reduction in the levels of the ubiquitin-ligase HDM2. Taken together, these results suggest that CVB5 infection of HeLa cells elicit the intrinsic pathway of apoptosis by MDM2 degradation and p53 activation, destabilizing protein sumoylation, by a mechanism that is dependent on a functional ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Rogério Gomes
- Department of Cell Biology, University of São Paulo School of Medicine at Ribeirão Preto, Av. dos Bandeirantes, 3900, 14049-900, SP, Brazil.
| | | | | |
Collapse
|
48
|
Cottle DL, McGrath MJ, Wilding BR, Cowling BS, Kane JM, D'Arcy CE, Holdsworth M, Hatzinisiriou I, Prescott M, Brown S, Mitchell CA. SLIMMER (FHL1B/KyoT3) interacts with the proapoptotic protein Siva-1 (CD27BP) and delays skeletal myoblast apoptosis. J Biol Chem 2009; 284:26964-77. [PMID: 19643733 DOI: 10.1074/jbc.m109.036293] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The fhl1 gene encoding four-and-a-half LIM protein-1 (FHL1) and its spliced isoform, SLIMMER, is mutated in reducing body myopathy, X-linked myopathy with postural muscle atrophy, scapuloperoneal myopathy, and rigid spine syndrome. In this study we have identified a novel function for SLIMMER in delaying skeletal muscle apoptosis via an interaction with the proapoptotic protein Siva-1. Siva-1 was identified as a SLIMMER-specific-interacting protein using yeast two-hybrid screening, direct-binding studies, and glutathione S-transferase pulldown analysis of murine skeletal muscle lysates. In C2C12 skeletal myoblasts, SLIMMER and Siva co-localized in the nucleus; however, both proteins exhibited redistribution to the cytoplasm following the differentiation of mononucleated myoblasts to multinucleated myotubes. In sections of mature skeletal muscle from wild type mice, SLIMMER and Siva-1 co-localized at the Z-line. SLIMMER and Siva-1 were also enriched in Pax-7-positive satellite cells, muscle stem cells that facilitate repair and regeneration. Significantly, SLIMMER delayed Siva-1-dependent apoptosis in C2C12 myoblasts. In skeletal muscle sections from the mdx mouse model of Duchenne muscular dystrophy, SLIMMER and Siva-1 co-localized in the nucleus of apoptotic myofibers. Therefore, SLIMMER may protect skeletal muscle from apoptosis.
Collapse
Affiliation(s)
- Denny L Cottle
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800 Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Interactions between viral and prokaryotic pathogens in a mixed infection with cardiovirus and mycoplasma. J Virol 2009; 83:9940-51. [PMID: 19605479 DOI: 10.1128/jvi.01167-09] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In the natural environment, animal and plant viruses often share ecological niches with microorganisms, but the interactions between these pathogens, although potentially having important implications, are poorly investigated. The present report demonstrates, in a model system, profound mutual effects of mycoplasma and cardioviruses in animal cell cultures. In contrast to mycoplasma-free cells, cultures contaminated with Mycoplasma hyorhinis responded to infection with encephalomyocarditis virus (EMCV), a picornavirus, but not with poliovirus (also a picornavirus), with a strong activation of a DNase(s), as evidenced by the TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling) immunofluorescence assay and electrophoretic analysis of host DNA. This degradation was reminiscent of that observed upon apoptosis but was caspase independent, judging by the failure of the specific pan-caspase inhibitor Q-VD-OPh to prevent it. The electrophoretic mobility of the enzyme responsible for DNA degradation and dependence of its activity on ionic conditions strongly suggested that it was represented by a DNase(s) of mycoplasma origin. In cells not infected with EMCV, the relevant DNase was dormant. The possibility is discussed that activation of the mycoplasma DNase might be linked to a relatively early increase in permeability of plasma membrane of the infected cells caused by EMCV. This type of unanticipated virus-mycoplasma "cooperation" may exemplify the complexity of pathogen-host interactions under conditions when viruses and microorganisms are infecting the same host. In the course of the present study, it was also demonstrated that pan-caspase inhibitor zVAD(OMe).fmk strongly suppressed cardiovirus polyprotein processing, illustrating an additional pitfall in investigations of viral effects on the apoptotic system of host cells.
Collapse
|
50
|
Resch U, Schichl YM, Winsauer G, Gudi R, Prasad K, de Martin R. Siva1 is a XIAP-interacting protein that balances NFkappaB and JNK signalling to promote apoptosis. J Cell Sci 2009; 122:2651-61. [PMID: 19584092 DOI: 10.1242/jcs.049940] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
XIAP is known as a potent inhibitor of apoptosis, but in addition is involved in cellular signalling, including the NFkappaB, JNK and TGFbeta pathways. Our search for XIAP-interacting partners led us to Siva1, a proapoptotic protein that is known to play a role in T-cell apoptosis through a caspase-dependent mitochondrial pathway. The interaction sites between XIAP and Siva1 were mapped to the RING domain of XIAP and the N-terminal, SAH-containing and death-homology-region-containing domains of Siva1. Co-immunoprecipitation experiments showed that XIAP, Siva1 and TAK1 form a ternary complex in Jurkat T cells. Reporter-gene analysis revealed that Siva1 inhibits XIAP- and TAK1-TAB1-mediated NFkappaB activation. By contrast, Siva1 increased XIAP- and TNFalpha-mediated AP1 activity and prolonged TNFalpha-induced JNK activation, whereas knock down of Siva1 resulted in reduced JNK activation. This suggests that Siva1 differentially modulates signalling by JNK and NFkappaB and shifts the balance between these pathways towards enhanced JNK activation, a situation that promotes apoptosis. Ectopically expressed Siva1 increased caspase-3 activity, which was inhibited by XIAP in a ubiquitin-ligase-dependent manner. In line with this, Siva1 was lysine-48-linked polyubiquitylated by XIAP. Our findings suggest that, via physical interaction with XIAP and TAK1, Siva1 diminishes NFkappaB and enhances JNK activity to favour apoptosis.
Collapse
Affiliation(s)
- Ulrike Resch
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Lazarettg. 19, A-1090, Vienna, Austria.
| | | | | | | | | | | |
Collapse
|