1
|
Fox CR, Yousef NN, Varudkar N, Shiffer EM, Aquino JR, Kedarinath K, Parks GD. Resistance to complement-mediated lysis of parainfluenza virus 5-infected cells is acquired after transition from acute to persistent infection. J Virol 2025; 99:e0189524. [PMID: 39791880 PMCID: PMC11852780 DOI: 10.1128/jvi.01895-24] [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: 11/13/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
Persistent viral infections can be an important medical problem, with persistently infected (PI) cells extending viral shedding, maintaining inflammation, and providing potential sources for new viral variants. Given that PI cells can acquire resistance to some innate immune pathways, we tested the hypothesis that complement (C')-mediated lysis of parainfluenza virus 5 (PIV5)-infected cells would differ between acute-infected and PI cells. Biochemical and real-time cell viability assays showed effective C'-mediated lysis of A549 lung cells acutely infected with PIV5, through pathways that depended on C3 and C5, but largely independent of C6. A PIV5 PI cell line established by long-term culturing of acutely infected A549 cells showed a high-level persistent expression of PIV5 proteins and infectious virus. Under conditions that led to effective lysis of acute PIV5-infected cells, the PI cells were nearly completely resistant to C'-mediated killing. This lack of C' killing was not due to failure to activate C', since C'-treated PIV5 PI cells had extensive C3 and membrane attack complex deposition, as well as production of C3a and C5a. Transcriptomics analysis revealed the C' cascade as the most significantly upregulated pathway in PIV5 PI cells versus acute infection. Biochemical analyses showed that resistance to C' killing correlated with increased expression in PI cells of two major C' inhibitors: complement factor H and Vitronectin. The finding of acquisition of C' resistance after the transition from acute PIV5 infection to PI cells raises the potential to inform therapeutics for PIs based on modulating C' pathways. IMPORTANCE A persistent infection (PI) with RNA viruses can extend virus shedding, prolong inflammation, and be a source of new viral variants. Since profound changes to innate immune pathways can occur in PI cells, it was important to test PI cells for changes in sensitivity to the complement (C') system, powerful innate immune pathways capable of lysing infected cells. Using parainfluenza virus 5 (PIV5) as a model system, we show that PI cells are nearly completely resistant to C'-mediated lysis, in stark contrast to high sensitivity of acute PIV5-infected cells to C' killing. A key finding was the upregulated expression in PI cells of two C' inhibitors: Vitronectin and complement factor H. These are important results with strong potential to inform therapeutics, given that polymorphisms in C' genes can correlate with severity of viral infections, and clinical trials are underway with new drugs that modulate C' responses.
Collapse
Affiliation(s)
- Candace R. Fox
- University of Central Florida, College of Medicine, Orlando, Florida, USA
| | - Nasser N. Yousef
- University of Central Florida, College of Medicine, Orlando, Florida, USA
| | - Namita Varudkar
- University of Central Florida, College of Medicine, Orlando, Florida, USA
| | | | - Jenna R. Aquino
- University of Central Florida, College of Medicine, Orlando, Florida, USA
| | - Kritika Kedarinath
- University of Central Florida, College of Medicine, Orlando, Florida, USA
| | - Griffith D. Parks
- University of Central Florida, College of Medicine, Orlando, Florida, USA
| |
Collapse
|
2
|
Savin IA, Sen’kova AV, Goncharova EP, Zenkova MA, Markov AV. Novel Core Gene Signature Associated with Inflammation-to-Metaplasia Transition in Influenza A Virus-Infected Lungs. Int J Mol Sci 2024; 25:11958. [PMID: 39596028 PMCID: PMC11594146 DOI: 10.3390/ijms252211958] [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: 10/09/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Respiratory infections caused by RNA viruses are a major contributor to respiratory disease due to their ability to cause annual epidemics with profound public health implications. Influenza A virus (IAV) infection can affect a variety of host signaling pathways that initiate tissue regeneration with hyperplastic and/or dysplastic changes in the lungs. Although these changes are involved in lung recovery after IAV infection, in some cases, they can lead to serious respiratory failure. Despite being ubiquitously observed, there are limited data on the regulation of long-term recovery from IAV infection leading to normal or dysplastic repair represented by inflammation-to-metaplasia transition in mice or humans. To address this knowledge gap, we used integrative bioinformatics analysis with further verification in vivo to elucidate the dynamic molecular changes in IAV-infected murine lung tissue and identified the core genes (Birc5, Cdca3, Plk1, Tpx2, Prc1. Rrm2, Nusap1, Spag5, Top2a, Mcm5) and transcription factors (E2F1, E2F4, NF-YA, NF-YB, NF-YC) involved in persistent lung injury and regeneration processes, which may serve as gene signatures reflecting the long-term effects of IAV proliferation on the lung. Further analysis of the identified core genes revealed their involvement not only in IAV infection but also in COVID-19 and lung neoplasm development, suggesting their potential role as biomarkers of severe lung disease and its complications represented by abnormal epithelial proliferation and oncotransformation.
Collapse
|
3
|
Guo YJ, Sun J, Li YL, Lai QR, Li L, Zhou HY, Li W. Epidemiology and genetic characterization of human parainfluenza virus-1 infection in pediatric patients from Hangzhou China, 2021-2022. Virol J 2024; 21:206. [PMID: 39223668 PMCID: PMC11367859 DOI: 10.1186/s12985-024-02479-8] [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: 04/01/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Human parainfluenza virus-1 (HPIV-1) is a notable pathogen instigating acute respiratory tract infections in children. The article is to elucidate the epidemiological and genetic characteristics of HPIV-1 circulating in Hangzhou during the period of 2021-2022. METHODS A cohort of 2360 nasopharyngeal swabs were amassed and subsequently examined via RT-PCR, with HPIV-1 positive samples undergoing P gene sequencing. RESULTS The highest HPIV-1 infection rates were found in children aged between 3 and 6 years. A pronounced positive rate persisted through the latter half of 2021, with a notable decline observed in the initial half of 2022. All HPIV-1 strains could be clustered into 2 groups: Cluster 1, with strains similar to those found in Japan (LC764865, LC764864), and Cluster 2, with strains similar to the Beijing strain (MW575643). CONCLUSION In conclusion, our study contributes to the comprehensive data on the epidemiological and genetic characteristics of HPIV-1 in pediatric patients from Hangzhou, post the COVID-19 peak.
Collapse
Affiliation(s)
- Ya-Jun Guo
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, PR China
| | - Jian Sun
- Department of Stomatology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Ya-Lin Li
- Zhejiang LAB, Hangzhou, 310003, PR China
| | - Qin-Rui Lai
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, PR China
| | - Lin Li
- Department of Infectious Diseases, Fujian Branch of Shanghai Children's Medical Center, Fujian Children's Hospital, Fuzhou, 350014, Fujian Province, China
| | - Hang-Yu Zhou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Wei Li
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, PR China.
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng road, Hangzhou, 310052, China.
| |
Collapse
|
4
|
Shiffer EM, Oyer JL, Copik AJ, Parks GD. Parainfluenza Virus 5 V Protein Blocks Interferon Gamma-Mediated Upregulation of NK Cell Inhibitory Ligands and Improves NK Cell Killing of Neuroblastoma Cells. Viruses 2024; 16:1270. [PMID: 39205244 PMCID: PMC11359056 DOI: 10.3390/v16081270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Natural killer (NK) cells can be effective immunotherapeutic anti-cancer agents due to their ability to selectively target and kill tumor cells. This activity is modulated by the interaction of NK cell receptors with inhibitory ligands on the surface of target cells. NK cell inhibitory ligands can be upregulated on tumor cell surfaces in response to interferon-gamma (IFN-γ), a cytokine which is produced by activated NK cells. We hypothesized that the resistance of tumor cells to NK cell killing could be overcome by expression of the parainfluenza virus 5 (PIV5) V protein, which has known roles in blocking IFN-γ signaling. This was tested with human PM21-NK cells produced through a previously developed particle-based method which yields superior NK cells for immunotherapeutic applications. Infection of human SK-N-SH neuroblastoma cells with PIV5 blocked IFN-γ-mediated upregulation of three NK cell inhibitory ligands and enhanced in vitro killing of these tumor cells by PM21-NK cells. SK-N-SH cells transduced to constitutively express the V protein alone were resistant to IFN-γ-mediated increases in cell surface expression of NK cell inhibitory ligands. Real-time in vitro cell viability assays demonstrated that V protein expression in SK-N-SH cells was sufficient to increase PM21-NK cell-mediated killing. Toward a potential therapeutic application, transient lentiviral delivery of the V gene also enhanced PM21-NK cell killing in vitro. Our results provide the foundation for novel therapeutic applications of V protein expression in combination with ex vivo NK cell therapy to effectively increase the killing of tumor cells.
Collapse
Affiliation(s)
| | | | | | - Griffith D. Parks
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA; (E.M.S.); (J.L.O.); (A.J.C.)
| |
Collapse
|
5
|
Saka N, Nishio M, Ohta K. Human parainfluenza virus type 2 V protein inhibits mitochondrial apoptosis pathway through two ways. Virology 2024; 594:110053. [PMID: 38492518 DOI: 10.1016/j.virol.2024.110053] [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: 11/20/2023] [Revised: 02/19/2024] [Accepted: 03/06/2024] [Indexed: 03/18/2024]
Abstract
Paramyxoviruses are reported to block apoptosis for their replication, but the mechanisms remain unclear. Furthermore, regulation of mitochondrial apoptosis by paramyxoviruses has been hardly reported. We investigated whether and how human parainfluenza virus type 2 (hPIV-2) counteracts apoptosis. Infection of recombinant hPIV-2 carrying mutated V protein showed higher caspase 3/7 activity and higher cytochrome c release from mitochondria than wild type hPIV-2 infection. This indicates that V protein controls mitochondrial apoptosis pathway. hPIV-2 V protein interacted with Bad, an apoptotic promoting protein, and this interaction inhibited the binding of Bad to Bcl-XL. V protein also bound to 14-3-3ε, which was essential for inhibition of 14-3-3ε cleavage. Our data collectively suggest that hPIV-2 V protein has two means of preventing mitochondrial apoptosis pathway: the inhibition of Bad-Bcl-XL interaction and the suppression of 14-3-3ε cleavage. This is the first report of the mechanisms behind how paramyxoviruses modulate mitochondrial apoptosis pathways.
Collapse
Affiliation(s)
- Naoki Saka
- Department of Microbiology, School of Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Machiko Nishio
- Department of Microbiology, School of Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Keisuke Ohta
- Department of Microbiology, School of Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
6
|
Zhang L, Meng Z, Calderone R, Liu W, She X, Li D. Mitochondria complex I deficiency in Candida albicans arrests the cell cycle at S phase through suppressive TOR and PKA pathways. FEMS Yeast Res 2024; 24:foae010. [PMID: 38592962 PMCID: PMC11008738 DOI: 10.1093/femsyr/foae010] [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: 08/22/2023] [Revised: 02/16/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024] Open
Abstract
How mutations in mitochondrial electron transport chain (ETC) proteins impact the cell cycle of Candida albicans was investigated in this study. Using genetic null mutants targeting ETC complexes I (CI), III (CIII), and IV (CIV), the cell cycle stages (G0/G1, S phase, and G2/M) were analyzed via fluorescence-activated cell sorting (FACS). Four CI null mutants exhibited distinct alterations, including extended S phase, shortened G2/M population, and a reduction in cells size exceeding 10 µM. Conversely, CIII mutants showed an increased population in G1/G0 phase. Among four CI mutants, ndh51Δ/Δ and goa1Δ/Δ displayed aberrant cell cycle patterns correlated with previously reported cAMP/PKA downregulation. Specifically, nuo1Δ/Δ and nuo2Δ/Δ mutants exhibited increased transcription of RIM15, a central hub linking cell cycle with nutrient-dependent TOR1 and cAMP/PKA pathways and Snf1 aging pathway. These findings suggest that suppression of TOR1 and cAMP/PKA pathways or enhanced Snf1 disrupts cell cycle progression, influencing cell longevity and growth among CI mutants. Overall, our study highlights the intricate interplay between mitochondrial ETC, cell cycle, and signaling pathways.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Dermatology, Jiangsu Province Hospital of Traditional Chinese Medicine, No.155 Hanzhong Road, Qinhuai District, Nanjing, 210029, China
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC, 20057, United States
| | - Zhou Meng
- Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), No. 12 Jiangwangmiao Street, Xuanwu District, Naning, 210042, China
| | - Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC, 20057, United States
| | - Weida Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), No. 12 Jiangwangmiao Street, Xuanwu District, Naning, 210042, China
| | - Xiaodong She
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC, 20057, United States
- Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), No. 12 Jiangwangmiao Street, Xuanwu District, Naning, 210042, China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC, 20057, United States
| |
Collapse
|
7
|
Cheng H, Zhang H, Cai H, Liu M, Wen S, Ren J. Molecular biology of canine parainfluenza virus V protein and its potential applications in tumor immunotherapy. Front Microbiol 2023; 14:1282112. [PMID: 38173672 PMCID: PMC10761501 DOI: 10.3389/fmicb.2023.1282112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
Canine parainfluenza virus (CPIV) is a zoonotic virus that is widely distributed and is the main pathogen causing canine infectious respiratory disease (CIRD), also known as "kennel cough," in dogs. The CPIV-V protein is the only nonstructural protein of the virus and plays an important role in multiple stages of the virus life cycle by inhibiting apoptosis, altering the host cell cycle and interfering with the interferon response. In addition, studies have shown that the V protein has potential applications in the field of immunotherapy in oncolytic virus therapy or self-amplifying RNA vaccines. In this review, the biosynthesis, structural characteristics and functions of the CPIV-V protein are reviewed with an emphasis on how it facilitates viral immune escape and its potential applications in the field of immunotherapy. Therefore, this review provides a scientific basis for research into the CPIV-V protein and its potential applications.
Collapse
Affiliation(s)
- Huai Cheng
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Hewei Zhang
- College of Food and Drugs, Luoyang Polytechnic, Luoyang, China
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang, China
| | - Huanchang Cai
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Min Liu
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Shubo Wen
- Preventive Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
| | - Jingqiang Ren
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang, China
| |
Collapse
|
8
|
Sajewicz-Krukowska J, Jastrzębski JP, Grzybek M, Domańska-Blicharz K, Tarasiuk K, Marzec-Kotarska B. Transcriptome Sequencing of the Spleen Reveals Antiviral Response Genes in Chickens Infected with CAstV. Viruses 2021; 13:2374. [PMID: 34960643 PMCID: PMC8708055 DOI: 10.3390/v13122374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Astrovirus infections pose a significant problem in the poultry industry, leading to multiple adverse effects such as a decreased egg production, breeding disorders, poor weight gain, and even increased mortality. The commonly observed chicken astrovirus (CAstV) was recently reported to be responsible for the "white chicks syndrome" associated with an increased embryo/chick mortality. CAstV-mediated pathogenesis in chickens occurs due to complex interactions between the infectious pathogen and the immune system. Many aspects of CAstV-chicken interactions remain unclear, and there is no information available regarding possible changes in gene expression in the chicken spleen in response to CAstV infection. We aim to investigate changes in gene expression triggered by CAstV infection. Ten 21-day-old SPF White Leghorn chickens were divided into two groups of five birds each. One group was inoculated with CAstV, and the other used as the negative control. At 4 days post infection, spleen samples were collected and immediately frozen at -70 °C for RNA isolation. We analyzed the isolated RNA, using RNA-seq to generate transcriptional profiles of the chickens' spleens and identify differentially expressed genes (DEGs). The RNA-seq findings were verified by quantitative reverse-transcription PCR (qRT-PCR). A total of 31,959 genes was identified in response to CAstV infection. Eventually, 45 DEGs (p-value < 0.05; log2 fold change > 1) were recognized in the spleen after CAstV infection (26 upregulated DEGs and 19 downregulated DEGs). qRT-PCR performed on four genes (IFIT5, OASL, RASD1, and DDX60) confirmed the RNA-seq results. The most differentially expressed genes encode putative IFN-induced CAstV restriction factors. Most DEGs were associated with the RIG-I-like signaling pathway or more generally with an innate antiviral response (upregulated: BLEC3, CMPK2, IFIT5, OASL, DDX60, and IFI6; downregulated: SPIK5, SELENOP, HSPA2, TMEM158, RASD1, and YWHAB). The study provides a global analysis of host transcriptional changes that occur during CAstV infection in vivo and proves that, in the spleen, CAstV infection in chickens predominantly affects the cell cycle and immune signaling.
Collapse
Affiliation(s)
- Joanna Sajewicz-Krukowska
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Jan Paweł Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Maciej Grzybek
- Department of Tropical Parasitology, Institute of Maritime and Tropical Medicine, Medical University of Gdansk, 81-519 Gdynia, Poland;
| | - Katarzyna Domańska-Blicharz
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Karolina Tarasiuk
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Barbara Marzec-Kotarska
- Department of Clinical Pathomorphology, The Medical University of Lublin, 20-090 Lublin, Poland;
| |
Collapse
|
9
|
Friedman N, Jacob-Hirsch J, Drori Y, Eran E, Kol N, Nayshool O, Mendelson E, Rechavi G, Mandelboim M. Transcriptomic profiling and genomic mutational analysis of Human coronavirus (HCoV)-229E -infected human cells. PLoS One 2021; 16:e0247128. [PMID: 33630927 PMCID: PMC7906355 DOI: 10.1371/journal.pone.0247128] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Human coronaviruses (HCoVs) cause mild to severe respiratory infection. Most of the common cold illnesses are caused by one of four HCoVs, namely HCoV-229E, HCoV-NL63, HCoV-HKU1 and HCoV-OC43. Several studies have applied global transcriptomic methods to understand host responses to HCoV infection, with most studies focusing on the pandemic severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome CoV (MERS-CoV) and the newly emerging SARS-CoV-2. In this study, Next Generation Sequencing was used to gain new insights into cellular transcriptomic changes elicited by alphacoronavirus HCoV-229E. HCoV-229E-infected MRC-5 cells showed marked downregulation of superpathway of cholesterol biosynthesis and eIF2 signaling pathways. Moreover, upregulation of cyclins, cell cycle control of chromosomal replication, and the role of BRCA1 in DNA damage response, alongside downregulation of the cell cycle G1/S checkpoint, suggest that HCoV-229E may favors S phase for viral infection. Intriguingly, a significant portion of key factors of cell innate immunity, interferon-stimulated genes (ISGs) and other transcripts of early antiviral response genes were downregulated early in HCoV-229E infection. On the other hand, early upregulation of the antiviral response factor Apolipoprotein B mRNA editing enzyme catalytic subunit 3B (APOBEC3B) was observed. APOBEC3B cytidine deaminase signature (C-to-T) was previously observed in genomic analysis of SARS-CoV-2 but not HCoV-229E. Higher levels of C-to-T mutations were found in countries with high mortality rates caused by SARS-CoV-2. APOBEC activity could be a marker for new emerging CoVs. This study will enhance our understanding of commonly circulating HCoVs and hopefully provide critical information about still-emerging coronaviruses.
Collapse
Affiliation(s)
- Nehemya Friedman
- Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jasmine Jacob-Hirsch
- Sheba Cancer Research Center (SCRC), Chaim Sheba Medical Center, Ramat Gan, Israel
- Wohl Centre for Translational Medicine, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Yaron Drori
- Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eyal Eran
- Sheba Cancer Research Center (SCRC), Chaim Sheba Medical Center, Ramat Gan, Israel
- Wohl Centre for Translational Medicine, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nitzan Kol
- Sheba Cancer Research Center (SCRC), Chaim Sheba Medical Center, Ramat Gan, Israel
- Wohl Centre for Translational Medicine, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Omri Nayshool
- Sheba Cancer Research Center (SCRC), Chaim Sheba Medical Center, Ramat Gan, Israel
- Wohl Centre for Translational Medicine, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Ella Mendelson
- Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gideon Rechavi
- Sheba Cancer Research Center (SCRC), Chaim Sheba Medical Center, Ramat Gan, Israel
- Wohl Centre for Translational Medicine, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Michal Mandelboim
- Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * E-mail:
| |
Collapse
|
10
|
Zaichuk TA, Nechipurenko YD, Adzhubey AA, Onikienko SB, Chereshnev VA, Zainutdinov SS, Kochneva GV, Netesov SV, Matveeva OV. The Challenges of Vaccine Development against Betacoronaviruses: Antibody Dependent Enhancement and Sendai Virus as a Possible Vaccine Vector. Mol Biol 2020; 54:812-826. [PMID: 32921819 PMCID: PMC7473411 DOI: 10.1134/s0026893320060151] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
To design an effective and safe vaccine against betacoronaviruses, it is necessary to use their evolutionarily conservative antigenic determinants that will elicit the combination of strong humoral and cell-mediated immune responses. Targeting such determinants minimizes the risk of antibody-dependent enhancement of viral infection. This phenomenon was observed in animal trials of experimental vaccines against SARS-CoV-1 and MERS-CoV that were developed based on inactivated coronavirus or vector constructs expressing the spike protein (S) of the virion. The substitution and glycosylation of certain amino acids in the antigenic determinants of the S-protein, as well as its conformational changes, can lead to the same effect in a new experimental vaccine against SARS-CoV-2. Using more conservative structural and accessory viral proteins for the vaccine antigenic determinants will help to avoid this problem. This review outlines approaches for developing vaccines against the new SARS-CoV-2 coronavirus that are based on non-pathogenic viral vectors. For efficient prevention of infections caused by respiratory pathogens the ability of the vaccine to stimulate mucosal immunity in the respiratory tract is important. Such a vaccine can be developed using non-pathogenic Sendai virus vector, since it can be administered intranasally and induce a mucosal immune response that strengthens the antiviral barrier in the respiratory tract and provides reliable protection against infection.
Collapse
Affiliation(s)
| | - Y D Nechipurenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A A Adzhubey
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia.,George Washington University, 20052 Washington, DC USA
| | - S B Onikienko
- Department of Military Field Therapy, Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - V A Chereshnev
- Institute of Immunology and Physiology, 620049 Yekaterinburg, Russia
| | - S S Zainutdinov
- State Research Center of Virology and Biotechnology "Vector,", 630559 Koltsovo, Russia
| | - G V Kochneva
- State Research Center of Virology and Biotechnology "Vector,", 630559 Koltsovo, Russia
| | - S V Netesov
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - O V Matveeva
- Sendai Viralytics, 117261 Acton, MA USA.,Biopolymer Design, 117281 Acton, MA USA
| |
Collapse
|
11
|
Feng C, Liang Y, Teodoro JG. The Role of Apoptin in Chicken Anemia Virus Replication. Pathogens 2020; 9:pathogens9040294. [PMID: 32316372 PMCID: PMC7238243 DOI: 10.3390/pathogens9040294] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 12/16/2022] Open
Abstract
Apoptin is the Vp3 protein of chicken anemia virus (CAV), which infects the thymocytes and erythroblasts in young chickens, causing chicken infectious anemia and immunosuppression. Apoptin is highly studied for its ability to selectively induce apoptosis in human tumor cells and, thus, is a protein of interest in anti-tumor therapy. CAV apoptin is known to localize to different subcellular compartments in transformed and non-transformed cells, depending on the DNA damage response, and the phosphorylation of several identified threonine residues. In addition, apoptin interacts with molecular machinery such as the anaphase promoting complex/cyclosome (APC/C) to inhibit the cell cycle and induce arrest in G2/M phase. While these functions of apoptin contribute to the tumor-selective effect of the protein, they also provide an important fundamental framework to apoptin’s role in viral infection, pathogenesis, and propagation. Here, we reviewed how the regulation, localization, and functions of apoptin contribute to the viral life cycle and postulated its importance in efficient replication of CAV. A model of the molecular biology of infection is critical to informing our understanding of CAV and other related animal viruses that threaten the agricultural industry.
Collapse
Affiliation(s)
- Cynthia Feng
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Yingke Liang
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Jose G. Teodoro
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
- Goodman Cancer Research Centre, Montreal, QC H3G 1A1, Canada
- Correspondence:
| |
Collapse
|
12
|
Wang Y, Wang R, Li Y, Sun Y, Song C, Zhan Y, Tan L, Liao Y, Meng C, Qiu X, Ding C. Newcastle disease virus induces G 0/G 1 cell cycle arrest in asynchronously growing cells. Virology 2018; 520:67-74. [PMID: 29793075 PMCID: PMC7112094 DOI: 10.1016/j.virol.2018.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/02/2022]
Abstract
The cell cycle, as a basic cellular process, is conservatively regulated. Consequently, subversion of the host cell replication cycle is a common strategy employed by many viruses to create a cellular environment favorable for viral replication. Newcastle disease virus (NDV) causes disease in poultry and is also an effective oncolytic agent. However, the effects of NDV infection on cell cycle progression are unknown. In this study, we showed that NDV replication in asynchronized cells resulted in the accumulation of infected cells in the G0/G1 phase of the cell cycle, which benefitted the proliferation of NDV. Examination of various cell cycle-regulatory proteins showed that expression of cyclin D1, was significantly reduced following NDV infection. Importantly, the decreased expression of cyclin D1 was reversed by inhibition of CHOP expression, indicating that induction of the PERK-eIF-2a-ATF4-CHOP signaling pathway was involved in the G0/G1 phase cell cycle arrest observed following NDV infection.
Collapse
Affiliation(s)
- Yan Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Rui Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Yangzhou University, Yangzhou 225000, PR China
| | - Yanrong Li
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Yuan Zhan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Lei Tan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Ying Liao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - ChunChun Meng
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Xusheng Qiu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China.
| |
Collapse
|
13
|
Su YC, Reshi L, Chen LJ, Li WH, Chiu HW, Hong JR. Nuclear targeting of the betanodavirus B1 protein via two arginine-rich domains induces G1/S cell cycle arrest mediated by upregulation of p53/p21. Sci Rep 2018; 8:3079. [PMID: 29449573 PMCID: PMC5814437 DOI: 10.1038/s41598-018-21340-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 02/02/2018] [Indexed: 12/20/2022] Open
Abstract
The molecular functions of betanodavirus non-structural protein B and its role in host cell survival remain unclear. In the present study, we examined the roles of specific nuclear targeting domains in B1 localization as well as the effect of B1 nuclear localization on the cell cycle and host cell survival. The B1 protein of the Red spotted grouper nervous necrosis virus (RGNNV) was detected in GF-1 grouper cells as early as 24 hours post-infection (hpi). Using an EYFP-B1 fusion construct, we observed nuclear localization of the B1 protein (up to 99%) in GF-1 cells at 48 hpi. The nuclear localization of B1 was mediated by two arginine-rich nuclear targeting domains (B domain: 46RRSRR51; C domain: 63RDKRPRR70) and domain C was more important than domain B in this process. B1 nuclear localization correlated with upregulation of p53 and p21(wef1/cip1); downregulation of Cyclin D1, CDK4 and Mdm2; and G1/S cell cycle arrest in GF-1 cells. In conclusion, nuclear targeting of the RGNNV B1 protein via two targeting domains causes cell cycle arrest by up-regulating p53/p21 and down-regulating Mdm2, thereby regulating host cell survival.
Collapse
Affiliation(s)
- Yu-Chin Su
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Latif Reshi
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan.,Department of Life Science, College of Bioscience & Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Lei-Jia Chen
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wei-Han Li
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Hsuan-Wen Chiu
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan
| | - Jiann-Ruey Hong
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan. .,Department of Biotechnology and Bioindustry, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
14
|
Graf1 Controls the Growth of Human Parainfluenza Virus Type 2 through Inactivation of RhoA Signaling. J Virol 2016; 90:9394-405. [PMID: 27512058 DOI: 10.1128/jvi.01471-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 08/01/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Rho GTPases are involved in a variety of cellular activities and are regulated by guanine nucleotide exchange factors and GTPase-activating proteins (GAPs). We found that the activation of Rho GTPases by lysophosphatidic acid promotes the growth of human parainfluenza virus type 2 (hPIV-2). Furthermore, hPIV-2 infection causes activation of RhoA, a Rho GTPase. We hypothesized that Graf1 (also known as ARHGAP26), a GAP, regulates hPIV-2 growth by controlling RhoA signaling. Immunofluorescence analysis showed that hPIV-2 infection altered Graf1 localization from a homogenous distribution within the cytoplasm to granules. Graf1 colocalized with hPIV-2 P, NP, and L proteins. Graf1 interacts with P and V proteins via their N-terminal common region, and the C-terminal Src homology 3 domain-containing region of Graf1 is important for these interactions. In HEK293 cells constitutively expressing Graf1, hPIV-2 growth was inhibited, and RhoA activation was not observed during hPIV-2 infection. In contrast, Graf1 knockdown restored hPIV-2 growth and RhoA activation. Overexpression of hPIV-2 P and V proteins enhanced hPIV-2-induced RhoA activation. These results collectively suggested that hPIV-2 P and V proteins enhanced hPIV-2 growth by binding to Graf1 and that Graf1 inhibits hPIV-2 growth through RhoA inactivation. IMPORTANCE Robust growth of hPIV-2 requires Rho activation. hPIV-2 infection causes RhoA activation, which is suppressed by Graf1. Graf1 colocalizes with viral RNP (vRNP) in hPIV-2-infected cells. We found that Graf1 interacts with hPIV-2 P and V proteins. We also identified regions in these proteins which are important for this interaction. hPIV-2 P and V proteins enhanced the hPIV-2 growth via binding to Graf1, while Graf1 inhibited hPIV-2 growth through RhoA inactivation.
Collapse
|
15
|
Xi D, Wang X, Teng D, Mao R. Mechanism of action of the tri-hybrid antimicrobial peptide LHP7 from lactoferricin, HP and plectasin on Staphylococcus aureus. Biometals 2015; 27:957-68. [PMID: 25015218 DOI: 10.1007/s10534-014-9768-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/24/2014] [Indexed: 11/28/2022]
Abstract
The tri-hybrid peptide-LHP7 has the potent activity against Gram-positive and Gram-negative as well as fungi, but its mechanism of action has remained elusive. The effluences of LHP7 on the Staphylococcus aureus cell membrane and targets of intracellular action were investigated. LHP7 exhibited an inhibitory effect on the S. aureus growth, similar to those achieved by plectasin, vancomycin and gramicidin. The membrane integrity studies confirmed that LHP7 disrupted the cell membrane, indicating a membrane permeabilizing killing action. A marginal decline in the intensity fluorescence indicated no significant depolarization of the membrane potential following LHP7 treatment. Furthermore, electron microscopy showed that cell shrinkage, cell wall thickening, cellular content leakage, and cell disruption were observed in the cells treated with LHP7. A gel retardation assay showed that LHP7 bound to the genomic DNA of S. aureus or plasmid DNA at a mass ratio of 2.5–10 (peptide/DNA). Circular dichroism indicated that LHP7 inserted into the groove of DNA. The cell cycle analysis showed that after the treatment with LHP7 for 30 and 60 min, the proportion of cells in I-phase increased from 8.71 to 12.09 % and from 8.71 to 15.68 %, indicating that LHP7 induced arrest of cells in the I-phase. These results would conduce to elucidate its underlying antibacterial mechanism.
Collapse
|
16
|
Zhou MT, Qin Y, Li M, Chen C, Chen X, Shu HB, Guo L. Quantitative Proteomics Reveals the Roles of Peroxisome-associated Proteins in Antiviral Innate Immune Responses. Mol Cell Proteomics 2015; 14:2535-49. [PMID: 26124285 DOI: 10.1074/mcp.m115.048413] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
Compared with whole-cell proteomic analysis, subcellular proteomic analysis is advantageous not only for the increased coverage of low abundance proteins but also for generating organelle-specific data containing information regarding dynamic protein movement. In the present study, peroxisome-enriched fractions from Sendai virus (SeV)-infected or uninfected HepG2 cells were obtained and subjected to quantitative proteomics analysis. We identified 311 proteins that were significantly changed by SeV infection. Among these altered proteins, 25 are immune response-related proteins. Further bioinformatic analysis indicated that SeV infection inhibits cell cycle-related proteins and membrane attack complex-related proteins, all of which are beneficial for the survival and replication of SeV within host cells. Using Luciferase reporter assays on several innate immune-related reporters, we performed functional analysis on 11 candidate proteins. We identified LGALS3BP and CALU as potential negative regulators of the virus-induced activation of the type I interferons.
Collapse
Affiliation(s)
- Mao-Tian Zhou
- From the ‡State Key Laboratory of Virology, College of Life Sciences
| | - Yue Qin
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University
| | - Mi Li
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University
| | - Chen Chen
- From the ‡State Key Laboratory of Virology, College of Life Sciences
| | - Xi Chen
- ¶Wuhan Institute of Biotechnology, Wuhan, China
| | - Hong-Bing Shu
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University;
| | - Lin Guo
- From the ‡State Key Laboratory of Virology, College of Life Sciences;
| |
Collapse
|
17
|
Cheng S, Zhang Y, Brooks CL. PCalign: a method to quantify physicochemical similarity of protein-protein interfaces. BMC Bioinformatics 2015; 16:33. [PMID: 25638036 PMCID: PMC4339745 DOI: 10.1186/s12859-015-0471-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 01/15/2015] [Indexed: 02/07/2023] Open
Abstract
Background Structural comparison of protein-protein interfaces provides valuable insights into the functional relationship between proteins, which may not solely arise from shared evolutionary origin. A few methods that exist for such comparative studies have focused on structural models determined at atomic resolution, and may miss out interesting patterns present in large macromolecular complexes that are typically solved by low-resolution techniques. Results We developed a coarse-grained method, PCalign, to quantitatively evaluate physicochemical similarities between a given pair of protein-protein interfaces. This method uses an order-independent algorithm, geometric hashing, to superimpose the backbone atoms of a given pair of interfaces, and provides a normalized scoring function, PC-score, to account for the extent of overlap in terms of both geometric and chemical characteristics. We demonstrate that PCalign outperforms existing methods, and additionally facilitates comparative studies across models of different resolutions, which are not accommodated by existing methods. Furthermore, we illustrate potential application of our method to recognize interesting biological relationships masked by apparent lack of structural similarity. Conclusions PCalign is a useful method in recognizing shared chemical and spatial patterns among protein-protein interfaces. It outperforms existing methods for high-quality data, and additionally facilitates comparison across structural models with different levels of details with proven robustness against noise. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0471-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shanshan Cheng
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA.
| | - Yang Zhang
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA. .,Department of Biological Chemistry, Medical School, University of Michigan, Ann Arbor, MI, USA.
| | - Charles L Brooks
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, USA. .,Department of Chemistry, University of Michigan, Ann Arbor, MI, USA. .,Biophysics Program, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Kim JY, Park SY, Lyoo HR, Koo ES, Kim MS, Jeong YS. Extended stability of cyclin D1 contributes to limited cell cycle arrest at G1-phase in BHK-21 cells with Japanese encephalitis virus persistent infection. J Microbiol 2015; 53:77-83. [PMID: 25557483 PMCID: PMC7090764 DOI: 10.1007/s12275-015-4661-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/01/2014] [Accepted: 12/01/2014] [Indexed: 11/16/2022]
Abstract
There is increasing evidence that many RNA viruses manipulate cell cycle control to achieve favorable cellular environments for their efficient replication during infection. Although virus-induced G0/G1 arrest often delays early apoptosis temporarily, a prolonged replication of the infected virus leads host cells to eventual death. In contrast, most mammalian cells with RNA virus persistent infection often escape cytolysis in the presence of productive viral replication. In this study, we demonstrated that the extended endurance of cyclin D1 was clearly associated with the suppression of glycogen synthase kinase-3ß (GSK-3ß) expression in BHK-21 cells that are persistently infected with Japanese encephalitis virus (JEV). The G0/G1 arrest of these cells turned much loose compared to the normal BHK-21 cells with JEV acute infection. After cycloheximide treatment, cyclin D1 in the persistently infected cells lasted several hours longer than those in acutely infected cells. Furthermore, both p21Cip1 and p27Kip1, positive regulators for cyclin D1 accumulation in the nucleus, were suppressed in their expression, which contrasts with those in JEV acute infection. Inhibition of the GSK-3ß by lithium chloride treatment rescued a significant number of cells from cytolysis in JEV acute infection, which coincided with the levels of cyclin D1 that escaped from proteolysis. Therefore, the limitation of G1/S arrest in the BHK-21 cells with JEV persistent infection is associated with the suppression of GSK-3ß expression, resulting in the extended duration of cyclin D1.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Biology and Research Institute of Basic Sciences, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
To replicate their genomes in cells and generate new progeny, viruses typically require factors provided by the cells that they have infected. Subversion of the cellular machinery that controls replication of the infected host cell is a common activity of many viruses. Viruses employ different strategies to deregulate cell cycle checkpoint controls and modulate cell proliferation pathways. A number of DNA and RNA viruses encode proteins that target critical cell cycle regulators to achieve cellular conditions that are beneficial for viral replication. Many DNA viruses induce quiescent cells to enter the cell cycle; this is thought to increase pools of deoxynucleotides and thus, facilitate viral replication. In contrast, some viruses can arrest cells in a particular phase of the cell cycle that is favorable for replication of the specific virus. Cell cycle arrest may inhibit early cell death of infected cells, allow the cells to evade immune defenses, or help promote virus assembly. Although beneficial for the viral life cycle, virus-mediated alterations in normal cell cycle control mechanisms could have detrimental effects on cellular physiology and may ultimately contribute to pathologies associated with the viral infection, including cell transformation and cancer progression and maintenance. In this chapter, we summarize various strategies employed by DNA and RNA viruses to modulate the replication cycle of the virus-infected cell. When known, we describe how these virus-associated effects influence replication of the virus and contribute to diseases associated with infection by that specific virus.
Collapse
Affiliation(s)
- Eishi Noguchi
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania USA
| | - Mariana C. Gadaleta
- Dept of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, USA
| |
Collapse
|
20
|
Li L, Gu B, Zhou F, Chi J, Feng D, Xie F, Wang F, Ma C, Li M, Wang J, Yao K. Cell cycle perturbations induced by human herpesvirus 6 infection and their effect on virus replication. Arch Virol 2013; 159:365-70. [PMID: 24013234 PMCID: PMC7086940 DOI: 10.1007/s00705-013-1826-0] [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: 02/20/2013] [Accepted: 07/08/2013] [Indexed: 11/28/2022]
Abstract
In this study, we demonstrate that infection of HSB-2 cells with human herpesvirus 6 (HHV-6) resulted in the accumulation of infected cells in the G2/M phase of the cell cycle. Analysis of various cell-cycle-regulatory proteins indicated that the levels of cyclins A2, B1, and E1 were increased in HHV-6-infected cells, but there was no difference in cyclin D1 levels between mock-infected and HHV-6-infected cells. Our data also showed that inducing G2/M phase arrest in cells infected by HHV-6 provided favorable conditions for viral replication.
Collapse
Affiliation(s)
- Lingyun Li
- Department of Developmental Genetics, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Briggs CM, Mayer AE, Parks GD. Mumps virus inhibits migration of primary human macrophages toward a chemokine gradient through a TNF-alpha dependent mechanism. Virology 2012; 433:245-52. [PMID: 22935226 PMCID: PMC3457059 DOI: 10.1016/j.virol.2012.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 06/22/2012] [Accepted: 08/10/2012] [Indexed: 01/04/2023]
Abstract
Macrophages are an important cell type for regulation of immunity, and can play key roles in virus pathogenesis. Here we address the effect of infection of primary human macrophages with the related paramyxoviruses Parainfluenza virus 5 (PIV5) and Mumps virus (MuV). Monocyte-derived macrophages infected with PIV5 or MuV showed very little cytopathic effect, but were found to be defective in migration toward a gradient of chemokines such as macrophage colony stimulating factor (MCSF) and vascular endothelial growth factor (VEGF). For MuV infection, the inhibition of migration required live virus infection, but was not caused by a loss of chemokine receptors on the surface of infected cells. MuV-mediated inhibition of macrophage chemotaxis was through a soluble factor released from infected cells. MuV infection enhanced secretion of TNF-α, but not macrophage inhibitory factor (MIF). Antibody inhibition and add-back experiments demonstrated that TNF-α was both necessary and sufficient for MuV-mediate chemotaxis inhibition.
Collapse
Affiliation(s)
- Caitlin M. Briggs
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157-1064
| | - Anne E. Mayer
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157-1064
| | - Griffith D. Parks
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157-1064
| |
Collapse
|
22
|
Bagchi P, Nandi S, Chattopadhyay S, Bhowmick R, Halder UC, Nayak MK, Kobayashi N, Chawla-Sarkar M. Identification of common human host genes involved in pathogenesis of different rotavirus strains: an attempt to recognize probable antiviral targets. Virus Res 2012; 169:144-53. [PMID: 22846921 DOI: 10.1016/j.virusres.2012.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 07/19/2012] [Accepted: 07/20/2012] [Indexed: 11/30/2022]
Abstract
Although two rotavirus vaccines have been licensed and approved by WHO and FDA; other parallel therapeutic strategies are needed to reduce the mortality and morbidity of rotavirus induced diarrhea worldwide. Since rotaviruses utilize the host cell machinery for their replication, study was initiated to identify host proteins which positively regulate rotavirus infection. To overcome the possible variations in host response due to existence of large variety of genotypes and human-animal reassortants, the total gene expression profile of HT29 cells infected with either simian (SA11) or bovine (A5-13) or human (Wa) rotavirus strains was analyzed using genome microarrays. Even though cells infected with human strain revealed some differences compared to the viruses of animal origin, 131 genes were similarly induced by all three strains. Genes involved in innate immune response, stress response, apoptosis and protein metabolism were induced by all viral strains. Results were validated by immunoblotting or RT-PCR. Role of some host genes in rotavirus infection was analyzed by using specific siRNAs.
Collapse
Affiliation(s)
- Parikshit Bagchi
- Division of Virology, National Institute of Cholera and Enteric Diseases, P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, India
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Ellis JA, Krakowka GS. A review of canine parainfluenza virus infection in dogs. J Am Vet Med Assoc 2012; 240:273-84. [DOI: 10.2460/javma.240.3.273] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
24
|
Dunn EF, Connor JH. HijAkt: The PI3K/Akt pathway in virus replication and pathogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 106:223-50. [PMID: 22340720 PMCID: PMC7149925 DOI: 10.1016/b978-0-12-396456-4.00002-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As obligate parasites of cellular processes, viruses must take over cellular macromolecular machinery. It is also becoming clear that viruses routinely control intracellular signaling pathways through the direct or indirect control of kinases and phosphatases. This control of cellular phosphoproteins is important to promote a variety of viral processes, from control of entry to nuclear function to the stimulation of viral protein synthesis. This review focuses on the takeover of the cellular phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway by a variety of retroviruses, DNA viruses, and RNA viruses, highlighting the functions ascribed to virus activation of PI3K and Akt activity. This review also describes the role that the PI3K/Akt pathway plays in the host response, noting that it that can trigger anti- as well as proviral functions.
Collapse
Affiliation(s)
- Ewan F Dunn
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
25
|
Proteomic profiling of the human cytomegalovirus UL35 gene products reveals a role for UL35 in the DNA repair response. J Virol 2011; 86:806-20. [PMID: 22072767 DOI: 10.1128/jvi.05442-11] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human cytomegalovirus infections involve the extensive modification of host cell pathways, including cell cycle control, the regulation of the DNA damage response, and averting promyelocytic leukemia (PML)-mediated antiviral responses. The UL35 gene from human cytomegalovirus is important for viral gene expression and efficient replication and encodes two proteins, UL35 and UL35a, whose mechanism of action is not well understood. Here, affinity purification coupled with mass spectrometry was used to identify previously unknown human cellular targets of UL35 and UL35a. We demonstrate that both viral proteins interact with the ubiquitin-specific protease USP7, and that UL35 expression can alter USP7 subcellular localization. In addition, UL35 (but not UL35a) was found to associate with three components of the Cul4(DCAF1) E3 ubiquitin ligase complex (DCAF1, DDB1, and DDA1) previously shown to be targeted by the HIV-1 Vpr protein. The coimmunoprecipitation and immunofluorescence microscopy of DCAF1 mutants revealed that the C-terminal region of DCAF1 is required for association with UL35 and mediates the dramatic relocalization of DCAF1 to UL35 nuclear bodies, which also contain conjugated ubiquitin. As previously reported for the Vpr-DCAF1 interaction, UL35 (but not UL35a) expression resulted in the accumulation of cells in the G(2) phase of the cell cycle, which is typical of a DNA damage response, and activated the G(2) checkpoint in a DCAF1-dependent manner. In addition, UL35 (but not UL35a) induced γ-H2AX and 53BP1 foci, indicating the activation of DNA damage and repair responses. Therefore, the identified interactions suggest that UL35 can contribute to viral replication through the manipulation of host responses.
Collapse
|
26
|
Characterization of the interaction between human respiratory syncytial virus and the cell cycle in continuous cell culture and primary human airway epithelial cells. J Virol 2011; 85:10300-9. [PMID: 21795354 DOI: 10.1128/jvi.05164-11] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Viruses can modify conditions inside cells to make them more favorable for replication and progeny virus production. One way of doing this is through manipulation of the cell cycle, a process that describes the ordered growth and division of cells. Analysis of model cell lines, such as A549 cells and primary airway epithelial cells, infected with human respiratory syncytial virus (HRSV) has shown alteration of the cell cycle during infection, although the signaling events were not clearly understood. In this study, targeted transcriptomic analysis of HRSV-infected primary airway epithelial cells revealed alterations in the abundances of many mRNAs encoding cell cycle-regulatory molecules, including decreases in the D-type cyclins and corresponding cyclin-dependent kinases (CDK4 and CDK6 [CDK4/6]). These alterations were reflected in changes in protein abundance and/or relocalization in HRSV-infected cells; taken together, they were predicted to result in G(0)/G(1) phase arrest. In contrast, there was no change in the abundances of D-type cyclins in A549 cells infected with HRSV. However, the abundance of the G(1)/S phase progression inhibitor p21(WAF1/CIP1) was increased over that in mock-treated cells, and this, again, was predicted to result in G(0)/G(1) phase arrest. The G(0)/G(1) phase arrest in both HRSV-infected primary cells and A549 cells was confirmed using dual-label flow cytometry that accurately measured the different stages of the cell cycle. Comparison of progeny virus production in primary and A549 cells enriched in G(0)/G(1) using a specific CDK4/6 kinase inhibitor with asynchronously replicating cells indicated that this phase of the cell cycle was more efficient for virus production.
Collapse
|
27
|
Identification of human parainfluenza virus type 2 (HPIV-2) V protein amino acid residues that reduce binding of V to MDA5 and attenuate HPIV-2 replication in nonhuman primates. J Virol 2011; 85:4007-19. [PMID: 21289116 DOI: 10.1128/jvi.02542-10] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human parainfluenza virus type 2 (HPIV-2), an important pediatric respiratory pathogen, encodes a V protein that inhibits type I interferon (IFN) induction and signaling. Using reverse genetics, we attempted the recovery of a panel of V mutant viruses that individually contained one of six cysteine-to-serine (residues 193, 197, 209, 211, 214, and 218) substitutions, one of two paired charge-to-alanine (R175A/R176A and R205A/K206A) substitutions, or a histidine-to-phenylalanine (H174F) substitution. This mutagenesis was performed using a cDNA-derived HPIV-2 virus that expressed the V and P coding sequences from separate mRNAs. Of the cysteine substitutions, only C193S, C214S, and C218S yielded viable virus, and only the C214S mutant replicated well enough for further analysis. The H174F, R175A/R176A, and R205A/K206A mutants were viable and replicated well. The H174F and R205A/K206A mutants did not differ from the wild-type (WT) V in their ability to physically interact with MDA5, a cytoplasmic sensor of nonself RNA that induces type I IFN. Like WT HPIV-2, these mutants inhibited IFN-β induction and replicated efficiently in African green monkeys (AGMs). In contrast, the C214S and R175A/R176A mutants did not bind MDA5 efficiently, did not inhibit interferon regulatory factor 3 (IRF3) dimerization or IFN-β induction, and were attenuated in AGMs. These findings indicate that V binding to MDA5 is important for HPIV-2 virulence in nonhuman primates and that some V protein residues involved in MDA5 binding are not essential for efficient HPIV-2 growth in vitro. Using a transient expression system, 20 additional mutant V proteins were screened for MDA5 binding, and the region spanning residues 175 to 180 was found to be essential for this activity.
Collapse
|
28
|
Schmitt PT, Ray G, Schmitt AP. The C-terminal end of parainfluenza virus 5 NP protein is important for virus-like particle production and M-NP protein interaction. J Virol 2010; 84:12810-23. [PMID: 20943976 PMCID: PMC3004301 DOI: 10.1128/jvi.01885-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 10/03/2010] [Indexed: 11/20/2022] Open
Abstract
Enveloped virus particles are formed by budding from infected-cell membranes. For paramyxoviruses, viral matrix (M) proteins are key drivers of virus assembly and budding. However, other paramyxovirus proteins, including glycoproteins, nucleocapsid (NP or N) proteins, and C proteins, are also important for particle formation in some cases. To investigate the role of NP protein in parainfluenza virus 5 (PIV5) particle formation, NP protein truncation and substitution mutants were analyzed. Alterations near the C-terminal end of NP protein completely disrupted its virus-like particle (VLP) production function and significantly impaired M-NP protein interaction. Recombinant viruses with altered NP proteins were generated, and these viruses acquired second-site mutations. Recombinant viruses propagated in Vero cells acquired mutations that mainly affected components of the viral polymerase, while recombinant viruses propagated in MDBK cells acquired mutations that mainly affected the viral M protein. Two of the Vero-propagated viruses acquired the same mutation, V/P(S157F), found previously to be responsible for elevated viral gene expression induced by a well-characterized variant of PIV5, P/V-CPI(-). Vero-propagated viruses caused elevated viral protein synthesis and spread rapidly through infected monolayers by direct cell-cell fusion, bypassing the need to bud infectious virions. Both Vero- and MDBK-propagated viruses exhibited infectivity defects and altered polypeptide composition, consistent with poor incorporation of viral ribonucleoprotein complexes (RNPs) into budding virions. Second-site mutations affecting M protein restored interaction with altered NP proteins in some cases and improved VLP production. These results suggest that multiple avenues are available to paramyxoviruses for overcoming defects in M-NP protein interaction.
Collapse
Affiliation(s)
- Phuong Tieu Schmitt
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, the Pennsylvania State University, University Park, Pennsylvania 16802
| | - Greeshma Ray
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, the Pennsylvania State University, University Park, Pennsylvania 16802
| | - Anthony P. Schmitt
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, the Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
29
|
Schaap-Nutt A, D'Angelo C, Amaro-Carambot E, Nolan SM, Davis S, Wise SM, Higgins C, Bradley K, Kim O, Mayor R, Skiadopoulos MH, Collins PL, Murphy BR, Schmidt AC. Recombinant human parainfluenza virus type 2 with mutations in V that permit cellular interferon signaling are not attenuated in non-human primates. Virology 2010; 406:65-79. [PMID: 20667570 PMCID: PMC2932766 DOI: 10.1016/j.virol.2010.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 06/15/2010] [Accepted: 07/06/2010] [Indexed: 02/06/2023]
Abstract
The HPIV2 V protein inhibits type I interferon (IFN) induction and signaling. To manipulate the V protein, whose coding sequence overlaps that of the polymerase-associated phosphoprotein (P), without altering the P protein, we generated an HPIV2 virus in which P and V are expressed from separate genes (rHPIV2-P+V). rHPIV2-P+V replicated like HPIV2-WT in vitro and in non-human primates. HPIV2-P+V was modified by introducing two separate mutations into the V protein to create rHPIV2-L101E/L102E and rHPIV2-Delta122-127. In contrast to HPIV2-WT, both mutant viruses were unable to degrade STAT2, leaving virus-infected cells susceptible to IFN. Neither mutant, nor HPIV2-WT, induced significant amounts of IFN-beta in infected cells. Surprisingly, neither rHPIV2-L101E/L102E nor rHPIV2-Delta122-127 was attenuated in two species of non-human primates. This indicates that loss of HPIV2's ability to inhibit IFN signaling is insufficient to attenuate virus replication in vivo as long as IFN induction is still inhibited.
Collapse
Affiliation(s)
- Anne Schaap-Nutt
- Laboratory of Infectious Diseases, RNA Viruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Many viruses interact with the host cell division cycle to favor their own growth. In this study, we examined the ability of influenza A virus to manipulate cell cycle progression. Our results show that influenza A virus A/WSN/33 (H1N1) replication results in G(0)/G(1)-phase accumulation of infected cells and that this accumulation is caused by the prevention of cell cycle entry from G(0)/G(1) phase into S phase. Consistent with the G(0)/G(1)-phase accumulation, the amount of hyperphosphorylated retinoblastoma protein, a necessary active form for cell cycle progression through late G(1) into S phase, decreased after infection with A/WSN/33 (H1N1) virus. In addition, other key molecules in the regulation of the cell cycle, such as p21, cyclin E, and cyclin D1, were also changed and showed a pattern of G(0)/G(1)-phase cell cycle arrest. It is interesting that increased viral protein expression and progeny virus production in cells synchronized in the G(0)/G(1) phase were observed compared to those in either unsynchronized cells or cells synchronized in the G(2)/M phase. G(0)/G(1)-phase cell cycle arrest is likely a common strategy, since the effect was also observed in other strains, such as H3N2, H9N2, PR8 H1N1, and pandemic swine H1N1 viruses. These findings, in all, suggest that influenza A virus may provide favorable conditions for viral protein accumulation and virus production by inducing a G(0)/G(1)-phase cell cycle arrest in infected cells.
Collapse
|
31
|
Ramachandran A, Horvath CM. Paramyxovirus disruption of interferon signal transduction: STATus report. J Interferon Cytokine Res 2010; 29:531-7. [PMID: 19694544 DOI: 10.1089/jir.2009.0070] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
RNA viruses in the paramyxovirus family have evolved a number of strategies to escape host cell surveillance and antiviral responses. One mechanism exploited by a number of viruses in this family is direct targeting of cytokine-inducible transcription regulators in the STAT family. Diverse members of this large virus family effectively suppress STAT signaling by the actions of their V proteins, or the related proteins derived from alternate viral mRNAs. These viral proteins have distinct means of targeting STATs, resulting in a variety of negative effects on STATs and their signal transduction. Recent developments in understanding STAT targeting will be reviewed.
Collapse
Affiliation(s)
- Aparna Ramachandran
- Department of Biochemistry, Molecular Biology, and Cell Biology, Northwestern University, Evanston, Illinois 60208, USA
| | | |
Collapse
|
32
|
Schaap-Nutt A, D'Angelo C, Scull MA, Amaro-Carambot E, Nishio M, Pickles RJ, Collins PL, Murphy BR, Schmidt AC. Human parainfluenza virus type 2 V protein inhibits interferon production and signaling and is required for replication in non-human primates. Virology 2009; 397:285-98. [PMID: 19969320 PMCID: PMC2822077 DOI: 10.1016/j.virol.2009.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 08/19/2009] [Accepted: 11/10/2009] [Indexed: 12/28/2022]
Abstract
In wild-type human parainfluenza virus type 2 (WT HPIV2), one gene (the P/V gene) encodes both the polymerase-associated phosphoprotein (P) and the accessory V protein. We generated a HPIV2 virus (rHPIV2-Vko) in which the P/V gene encodes only the P protein to examine the role of V in replication in vivo and as a potential live attenuated virus vaccine. Preventing expression of V protein severely impaired virus recovery from cDNA and growth in vitro, particularly in IFN-competent cells. rHPIV2-Vko, unlike WT HPIV2, strongly induced IFN-β and permitted IFN signaling, leading to establishment of a robust antiviral state. rHPIV2-Vko infection induced extensive syncytia and cytopathicity that was due to both apoptosis and necrosis. Replication of rHPIV2-Vko was highly restricted in the respiratory tract of African green monkeys and in differentiated primary human airway epithelial (HAE) cultures, suggesting that V protein is essential for efficient replication of HPIV2 in organized epithelial cells and that rHPIV2-Vko is over-attenuated for use as a live attenuated vaccine.
Collapse
Affiliation(s)
- Anne Schaap-Nutt
- Laboratory of Infectious Diseases, RNA Viruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Sun D, Luthra P, Li Z, He B. PLK1 down-regulates parainfluenza virus 5 gene expression. PLoS Pathog 2009; 5:e1000525. [PMID: 19629176 PMCID: PMC2709441 DOI: 10.1371/journal.ppat.1000525] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 06/30/2009] [Indexed: 12/28/2022] Open
Abstract
The paramyxoviruses are a family of negative-sense RNA viruses that includes many important human and animal pathogens. Paramyxovirus RNA synthesis requires the viral phosphoprotein (P) and the large (L) protein. Phosphorylation of P is thought to regulate viral gene expression, though direct proof remains elusive. Recently, we reported that phosphorylation of a specific residue (Ser157) of the P protein of parainfluenza virus 5 (PIV5), a prototypical paramyxovirus, correlates with decreased viral gene expression and cytokine expression in infected cells. Here, we show that: Polo-like kinase 1 (PLK1), a serine/theronine kinase that plays a critical role in regulating the cell cycle, interacts with PIV5 P through the S157 residue; PLK1 inhibition increases viral gene expression; PLK1 over-expression inhibits viral gene expression; and PLK1 directly phosphorylates P in vitro, indicating that PLK1 down-regulates viral gene expression by phosphorylating P. Furthermore, we have determined the PLK1 phosphorylation site on P and found that mutant recombinant PIV5 whose P proteins cannot either bind to or be phosphorylated by PLK1 have similar phenotypes. Increased viral gene expression in PIV5 with mutations in the PLK1 binding/phosphorylation sites correlates with increased induction of cell death and cytokine expression, suggesting that PIV5 limits its viral gene expression to avoid these host effects. It is possible that targeting PLK1 will enhance host innate immune responses, leading to a novel strategy of clearing paramyxovirus infections quickly.
Collapse
Affiliation(s)
- Dengyun Sun
- Intercollege Graduate Program in Cell and Developmental Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Priya Luthra
- Intercollege Graduate Program in Cell and Developmental Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Zhuo Li
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Biao He
- Intercollege Graduate Program in Cell and Developmental Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center of Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
34
|
Ramachandran A, Parisien JP, Horvath CM. STAT2 is a primary target for measles virus V protein-mediated alpha/beta interferon signaling inhibition. J Virol 2008; 82:8330-8. [PMID: 18579593 PMCID: PMC2519631 DOI: 10.1128/jvi.00831-08] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 06/17/2008] [Indexed: 12/11/2022] Open
Abstract
Measles virus, a member of the Morbillivirus family, infects millions of people each year despite the availability of effective vaccines. The V protein of measles virus is an important virulence factor that can interfere with host innate immunity by inactivating alpha/beta interferon (IFN-alpha/beta) and IFN-gamma signaling through protein interactions with signal transducer and activator of transcription proteins STAT1 and STAT2. Here we demonstrate that although STAT1 interference results from protein interactions within a V protein N-terminal region encompassed by amino acids 110 to 130, detection of STAT1 interaction and IFN-gamma signaling inhibition requires the presence of cellular STAT2. Cell-specific variability in STAT1 interference was observed to correlate with V protein expression level. A more direct target for measles virus V protein-mediated IFN-alpha/beta evasion is STAT2. Results indicate that the widely conserved C-terminal zinc finger domain of measles virus V protein is both necessary and sufficient to bind STAT2 and disrupt IFN-alpha/beta signal transduction. Mutagenesis and molecular modeling define a contact surface for STAT2 association that includes aspartic acid residue 248 as critical for STAT2 interference and IFN antiviral immune suppression. These findings clearly define the molecular determinants for measles virus IFN evasion and validate specific targets as candidates for therapeutic intervention.
Collapse
|
35
|
DeHart JL, Bosque A, Harris RS, Planelles V. Human immunodeficiency virus type 1 Vif induces cell cycle delay via recruitment of the same E3 ubiquitin ligase complex that targets APOBEC3 proteins for degradation. J Virol 2008; 82:9265-72. [PMID: 18596088 PMCID: PMC2546900 DOI: 10.1128/jvi.00377-08] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 06/27/2008] [Indexed: 12/18/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) Vif recruits a Cullin 5 ubiquitin ligase that targets APOBEC3 proteins for degradation. Recently, Vif has also been shown to induce cell cycle disturbance in G(2). We show that in contrast to the expression of Vpr, the expression of Vif does not preclude cell division, and therefore, Vif causes delay and not arrest in G(2). We also demonstrate that the interaction of Vif with the ubiquitin ligase is required for cell cycle disruption, as was previously shown for HIV-1 Vpr. The presence of APOBEC3 D/E, F, and G had no influence on Vif-induced alteration of the cell cycle. We conclude that cell cycle delay by Vif is a result of ubiquitination and degradation of a cellular protein that is different from the known APOBEC3 family members.
Collapse
Affiliation(s)
- Jason L DeHart
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East no. 2100, Room 2520, Salt Lake City, Utah 84112, USA
| | | | | | | |
Collapse
|
36
|
A single amino acid residue change in the P protein of parainfluenza virus 5 elevates viral gene expression. J Virol 2008; 82:9123-33. [PMID: 18614634 DOI: 10.1128/jvi.00289-08] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Parainfluenza virus 5 (PIV5) is a prototypical paramyxovirus. The V/P gene of PIV5 encodes two mRNA species through a process of pseudotemplated insertion of two G residues at a specific site during transcription, resulting in two viral proteins, V and P, whose N termini of 164 amino acid residues are identical. Previously it was reported that mutating six amino acid residues within this identical region results in a recombinant PIV5 (rPIV5-CPI-) that exhibits elevated viral protein expression and induces production of cytokines, such as beta interferon and interleukin 6. Because the six mutations correspond to the shared region of the V protein and the P protein, it is not clear whether the phenotypes associated with rPIV5-CPI- are due to mutations in the P protein and/or mutations in the V protein. To address this question, we used a minigenome system and recombinant viruses to study the effects of mutations on the functions of the P and V proteins. We found that the P protein with six amino acid residue changes (Pcpi-) was more efficient than wild-type P in facilitating replication of viral RNA, while the V protein with six amino acid residue changes (Vcpi-) still inhibits minigenome replication as does the wild-type V protein. These results indicate that elevated viral gene expression in rPIV5-CPI- virus-infected cells can be attributed to a P protein with an increased ability to facilitate viral RNA synthesis. Furthermore, we found that a single amino acid residue change at position 157 of the P protein from Ser (the residue in the wild-type P protein) to Phe (the residue in Pcpi-) is sufficient for elevated viral gene expression. Using mass spectrometry and (33)P labeling, we found that residue S157 of the P protein is phosphorylated. Based on these results, we propose that phosphorylation of the P protein at residue 157 plays an important role in regulating viral RNA replication.
Collapse
|
37
|
Lin Y, Sun M, Fuentes SM, Keim CD, Rothermel T, He B. Inhibition of interleukin-6 expression by the V protein of parainfluenza virus 5. Virology 2007; 368:262-72. [PMID: 17692882 PMCID: PMC2100396 DOI: 10.1016/j.virol.2007.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 06/29/2007] [Accepted: 07/06/2007] [Indexed: 01/22/2023]
Abstract
The V protein of parainfluenza virus 5 (PIV5) plays an important role in the evasion of host immune responses. The V protein blocks interferon (IFN) signaling in human cells by causing degradation of the STAT1 protein, a key component of IFN signaling, and blocks IFN-beta production by preventing nuclear translocation of IRF3, a key transcription factor for activating IFN-beta promoter. Interleukin-6 (IL-6), along with tumor necrosis factor (TNF)-alpha and IL-1beta, is a major proinflammatory cytokine that plays important roles in clearing virus infection through inflammatory responses. Many viruses have developed strategies to block IL-6 expression. Wild-type PIV5 infection induces little, if any, expression of cytokines such as IL-6 or TNF-alpha, whereas infection by a mutant PIV5 lacking the conserved C-terminal cysteine rich domain (rPIV5VDeltaC) induced high levels of IL-6 expression. Examination of mRNA levels of IL-6 indicated that the transcription activation of IL-6 played an important role in the increased IL-6 expression. Co-infection with wild-type PIV5 prevented the activation of IL-6 transcription by rPIV5VDeltaC, and a plasmid encoding the full-length PIV5 V protein prevented the activation of IL-6 promoter-driven reporter gene expression by rPIV5VDeltaC, indicating that the V protein played a role in inhibiting IL-6 transcription. The activation of IL-6 was independent of IFN-beta even though rPIV5VDeltaC-infected cells produced IFN-beta. Using reporter gene assays and chromatin immunoprecipitation (ChIP), it was found that NF-kappaB played an important role in activating expression of IL-6. We have proposed a model of activating and inhibiting IL-6 transcription by PIV5.
Collapse
Affiliation(s)
- Yuan Lin
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802
| | - Minghao Sun
- Graduate Program in Pathobiology, Pennsylvania State University, University Park, PA 16802
| | - Sandra M. Fuentes
- Graduate Program in Pathobiology, Pennsylvania State University, University Park, PA 16802
| | - Celia D. Keim
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802
| | - Terri Rothermel
- Graduate Program in Pathobiology, Pennsylvania State University, University Park, PA 16802
| | - Biao He
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802
- Graduate Program in Pathobiology, Pennsylvania State University, University Park, PA 16802
- The Huck Institutes of Life sciences, Pennsylvania State University, University Park, PA 16802
- Center of Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
38
|
Akt plays a critical role in replication of nonsegmented negative-stranded RNA viruses. J Virol 2007; 82:105-14. [PMID: 17959676 DOI: 10.1128/jvi.01520-07] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The order Mononegavirales (comprised of nonsegmented negative-stranded RNA viruses or NNSVs) contains many important pathogens. Parainfluenza virus 5 (PIV5), formerly known as simian virus 5, is a prototypical paramyxovirus and encodes a V protein, which has a cysteine-rich C terminus that is conserved among all paramyxoviruses. The V protein of PIV5, like that of many other paramyxoviruses, plays an important role in regulating viral RNA synthesis. In this work, we show that V interacts with Akt, a serine/threonine kinase, also known as protein kinase B. Both pharmacological inhibitors and small interfering RNA against Akt1 reduced PIV5 replication, indicating that Akt plays a critical role in PIV5 replication. Furthermore, treatment with Akt inhibitors also reduced the replication of several other paramyxoviruses, as well as vesicular stomatitis virus, the prototypical rhabdovirus, indicating that Akt may play a more universal role in NNSV replication. The phosphoproteins (P proteins) of NNSVs are essential cofactors for the viral RNA polymerase complex and require heavy phosphorylation for their activity. Inhibition of Akt activity reduced the level of P phosphorylation, suggesting that Akt is involved in regulating viral RNA synthesis. In addition, Akt1 phosphorylated a recombinant P protein of PIV5 purified from bacteria. The finding that Akt plays a critical role in replication of NNSV will lead to a better understanding of how these viruses replicate, as well as novel strategies to treat infectious diseases caused by NNSVs.
Collapse
|
39
|
Tan L, Ehrlich E, Yu XF. DDB1 and Cul4A are required for human immunodeficiency virus type 1 Vpr-induced G2 arrest. J Virol 2007; 81:10822-30. [PMID: 17626091 PMCID: PMC2045451 DOI: 10.1128/jvi.01380-07] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Accepted: 06/29/2007] [Indexed: 01/01/2023] Open
Abstract
Vpr-mediated induction of G2 cell cycle arrest has been postulated to be important for human immunodeficiency virus type 1 (HIV-1) replication, but the precise role of Vpr in this cell cycle arrest is unclear. In the present study, we have shown that HIV-1 Vpr interacts with damaged DNA binding protein 1 (DDB1) but not its partner DDB2. The interaction of Vpr with DDB1 was inhibited when DCAF1 (VprBP) expression was reduced by short interfering RNA (siRNA) treatment. The Vpr mutant (Q65R) that was defective for DCAF1 interaction also had a defect in DDB1 binding. However, Vpr binding to DDB1 was not sufficient to induce G2 arrest. A reduction in DDB1 or DDB2 expression in the absence of Vpr also did not induce G2 arrest. On the other hand, Vpr-induced G2 arrest was impaired when the intracellular level of DDB1 or Cullin 4A was reduced by siRNA treatment. Furthermore, Vpr-induced G2 arrest was largely abolished by a proteasome inhibitor. These data suggest that Vpr assembles with DDB1 through interaction with DCAF1 to form an E3 ubiquitin ligase that targets cellular substrates for proteasome-mediated degradation and G2 arrest.
Collapse
Affiliation(s)
- Lindi Tan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
40
|
Wen X, Duus KM, Friedrich TD, de Noronha CMC. The HIV1 protein Vpr acts to promote G2 cell cycle arrest by engaging a DDB1 and Cullin4A-containing ubiquitin ligase complex using VprBP/DCAF1 as an adaptor. J Biol Chem 2007; 282:27046-27057. [PMID: 17620334 DOI: 10.1074/jbc.m703955200] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The roles of the HIV1 protein Vpr in virus replication and pathogenesis remain unclear. Expression of Vpr in dividing cells causes cell cycle arrest in G(2). Vpr also facilitates low titer infection of terminally differentiated macrophages, enhances transcription, promotes apoptosis, and targets cellular uracil N-glycosylase for degradation. Using co-immunoprecipitation and tandem mass spectroscopy, we found that HIV1 Vpr engages a DDB1- and cullin4A-containing ubiquitin-ligase complex through VprBP/DCAF1. HIV2 Vpr has two Vpr-like proteins, Vpr and Vpx, which cause G(2) arrest and facilitate macrophage infection, respectively. HIV2 Vpr, but not Vpx, engages the same set of proteins. We further demonstrate that the interaction between Vpr and the ubiquitin-ligase components as well as further assembly of the ubiquitin-ligase are necessary for Vpr-mediated G(2) arrest. Our data support a model in which Vpr engages the ubiquitin ligase to deplete a cellular factor that is required for cell cycle progression into mitosis. Vpr, thus, functions like the HIV1 proteins Vif and Vpu to usurp cellular ubiquitin ligases for viral functions.
Collapse
Affiliation(s)
- Xiaoyun Wen
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| | - Karen M Duus
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| | - Thomas D Friedrich
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| | - Carlos M C de Noronha
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208.
| |
Collapse
|
41
|
Tompkins SM, Lin Y, Leser GP, Kramer KA, Haas DL, Howerth EW, Xu J, Kennett MJ, Durbin RK, Durbin JE, Tripp R, Lamb RA, He B. Recombinant parainfluenza virus 5 (PIV5) expressing the influenza A virus hemagglutinin provides immunity in mice to influenza A virus challenge. Virology 2007; 362:139-50. [PMID: 17254623 PMCID: PMC1995462 DOI: 10.1016/j.virol.2006.12.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2006] [Revised: 10/23/2006] [Accepted: 12/03/2006] [Indexed: 10/23/2022]
Abstract
Parainfluenza virus type 5 (PIV5), formerly known as simian virus 5 (SV5), is a non-segmented negative strand RNA virus that offers several advantages as a vaccine vector. PIV5 infects many cell types causing little cytopathic effect, it replicates in the cytoplasm of infected cells, and does not have a DNA phase in its life cycle thus avoiding the possibility of introducing foreign genes into the host DNA genome. Importantly, PIV5 can infect humans but it is not associated with any known human illness. PIV5 grows well in tissue culture cells, including Vero cells, which have been approved for vaccine production, and the virus can be obtained easily from the media. To test the feasibility of using PIV5 as a live vaccine vector, the hemagglutinin (HA) gene from influenza A virus strain A/Udorn/72 (H3N2) was inserted into the PIV5 genome as an extra gene between the hemagglutinin-neuraminidase (HN) gene and the large (L) polymerase gene. Recombinant PIV5 containing the HA gene of Udorn (rPIV5-H3) was recovered and it replicated similarly to wild type PIV5, both in vitro and in vivo. The HA protein expressed by rPIV5-H3-infected cells was incorporated into the virions and addition of the HA gene did not increase virus virulence in mice. The efficacy of rPIV5-H3 as a live vaccine was examined in 6-week-old BALB/c mice. The results show that a single dose inoculation provides broad and considerable immunity against influenza A virus infection.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Body Weight
- Cattle
- Cell Line
- Cells, Cultured
- Chlorocebus aethiops
- Disease Models, Animal
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immunoglobulin G/blood
- Immunoglobulin M/blood
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A virus/genetics
- Influenza A virus/immunology
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Lung/pathology
- Lung/virology
- Mice
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/pathology
- Orthomyxoviridae Infections/prevention & control
- Parainfluenza Virus 5/genetics
- Parainfluenza Virus 5/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- S. Mark Tompkins
- Department of Infectious Diseases, University of Georgia, Athens, GA
| | - Yuan Lin
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA
| | - George P. Leser
- Department of Biochemistry, Molecular Biology and Cellular Biology, Evanston, IL
| | - Kari A. Kramer
- Department of Infectious Diseases, University of Georgia, Athens, GA
| | - Debra L. Haas
- Department of Infectious Diseases, University of Georgia, Athens, GA
| | - Elizabeth W. Howerth
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Jie Xu
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA
| | - Mary J. Kennett
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA
| | | | - Joan E. Durbin
- Children's Hospital, Ohio State University, Columbus, OH
| | - Ralph Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA
| | - Robert A. Lamb
- Department of Biochemistry, Molecular Biology and Cellular Biology, Evanston, IL
- Howard Hughes Medical Institute, Northwestern University, Evanston, IL
| | - Biao He
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA
- Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, PA
| |
Collapse
|
42
|
Schröfelbauer B, Hakata Y, Landau NR. HIV-1 Vpr function is mediated by interaction with the damage-specific DNA-binding protein DDB1. Proc Natl Acad Sci U S A 2007; 104:4130-5. [PMID: 17360488 PMCID: PMC1820720 DOI: 10.1073/pnas.0610167104] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The Vpr accessory protein of HIV-1 induces a response similar to that of DNA damage. In cells expressing Vpr, the DNA damage sensing kinase, ATR, is activated, resulting in G(2) arrest and apoptosis. In addition, Vpr causes rapid degradation of the uracil-DNA glycosylases UNG2 and SMUG1. Although several cellular proteins have been reported to bind to Vpr, the mechanism by which Vpr mediates its biological effects is unknown. Using tandem affinity purification and mass spectrometry, we identified a predominant cellular protein that binds to Vpr as the damage-specific DNA-binding protein 1 (DDB1). In addition to its role in the repair of damaged DNA, DDB1 is a component of an E3 ubiquitin ligase that degrades numerous cellular substrates. Interestingly, DDB1 is targeted by specific regulatory proteins of other viruses, including simian virus 5 and hepatitis B. We show that the interaction with DDB1 mediates Vpr-induced apoptosis and UNG2/SMUG1 degradation and impairs the repair of UV-damaged DNA, which could account for G(2) arrest and apoptosis. The interaction with DDB1 may explain several of the diverse biological functions of Vpr and suggests potential roles for Vpr in HIV-1 replication.
Collapse
Affiliation(s)
- Bärbel Schröfelbauer
- *Infectious Disease Laboratory, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA 92037-1099; and
- Department of Biotechnology, Institute of Applied Microbiology, University of Natural Resources and Applied Life Sciences, A-1180 Vienna, Austria
| | - Yoshiyuki Hakata
- *Infectious Disease Laboratory, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA 92037-1099; and
| | - Nathaniel R. Landau
- *Infectious Disease Laboratory, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA 92037-1099; and
- To whom correspondence should be sent at the present address:
New York University School of Medicine, Department of Microbiology, 550 First Avenue, New York, NY 10016. E-mail:
| |
Collapse
|
43
|
Haralambieva I, Iankov I, Hasegawa K, Harvey M, Russell SJ, Peng KW. Engineering oncolytic measles virus to circumvent the intracellular innate immune response. Mol Ther 2007; 15:588-97. [PMID: 17245355 PMCID: PMC3833616 DOI: 10.1038/sj.mt.6300076] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The innate antiviral responses of tumor cells are often impaired but may still be sufficient to impede the intratumoral spread of an oncolytic virus. Here, we establish that the oncolytic measles virus (MV-eGFP) induces interferon (IFN) production in human myeloma and ovarian cancer cells. In addition, MV gene expression and virus progeny production were inhibited by IFN treatment of these tumor cells. The P gene of wild-type measles virus encodes P/V/C proteins known to antagonize IFN induction and/or response. We therefore engineered MV-eGFP for IFN evasion and more efficient intratumoral spread by arming it with the P gene from wild-type IC-B strain MV, thus generating MV-eGFP-Pwt. The chimeric virus exhibited reduced IFN sensitivity and diminished capacity to induce IFN in BJAB lymphoma, ARH-77 myeloma cells, and activated peripheral blood mononuclear cells. Interestingly, unlike the wild-type MV, MV-eGFP-Pwt was unable to shut down IFN induction completely. In immunocompromised mice bearing human myeloma xenografts, intravenously administered MV-eGFP-Pwt showed significantly enhanced oncolytic potency compared to MV-eGFP. These results indicate that oncolytic viruses are subject to control by the innate immune defenses of human tumor cells and may therefore be more effective if their natural ability to combat innate immunity is maintained.
Collapse
Affiliation(s)
- Iana Haralambieva
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Ianko Iankov
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Kosei Hasegawa
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Mary Harvey
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Stephen J Russell
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Kah-Whye Peng
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
44
|
Liang X, Pickering MT, Cho NH, Chang H, Volkert MR, Kowalik TF, Jung JU. Deregulation of DNA damage signal transduction by herpesvirus latency-associated M2. J Virol 2006; 80:5862-74. [PMID: 16731925 PMCID: PMC1472574 DOI: 10.1128/jvi.02732-05] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infected cells recognize viral replication as a DNA damage stress and elicit a DNA damage response that ultimately induces apoptosis as part of host immune surveillance. Here, we demonstrate a novel mechanism where the murine gamma herpesvirus 68 (gammaHV68) latency-associated, anti-interferon M2 protein inhibits DNA damage-induced apoptosis by interacting with the DDB1/COP9/cullin repair complex and the ATM DNA damage signal transducer. M2 expression constitutively induced DDB1 nuclear localization and ATM kinase activation in the absence of DNA damage. Activated ATM subsequently induced Chk activation and p53 phosphorylation and stabilization without eliciting H2AX phosphorylation and MRN recruitment to foci upon DNA damage. Consequently, M2 expression inhibited DNA repair, rendered cells resistant to DNA damage-induced apoptosis, and induced a G(1) cell cycle arrest. Our results suggest that gammaHV68 M2 blocks apoptosis-mediated intracellular innate immunity, which might ultimately contribute to its role in latent infection.
Collapse
Affiliation(s)
- Xiaozhen Liang
- Department of Microbiology and Molecular Genetics, New England Primate Research Center, Harvard Medical School, Southborough, MA 01772, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Cruz CD, Palosaari H, Parisien JP, Devaux P, Cattaneo R, Ouchi T, Horvath CM. Measles virus V protein inhibits p53 family member p73. J Virol 2006; 80:5644-50. [PMID: 16699046 PMCID: PMC1472123 DOI: 10.1128/jvi.02400-05] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Accepted: 03/11/2006] [Indexed: 01/23/2023] Open
Abstract
Paramyxovirus V proteins function as host interference factors that inactivate antiviral responses, including interferon. Characterization of cellular proteins that copurify with ectopically expressed measles virus V protein has revealed interactions with DNA binding domains of p53 family proteins, p53 and p73. Specific transcriptional assays reveal that expression of measles virus V cDNA inhibits p73, but not p53. Expression of measles virus V cDNA can delay cell death induced by genotoxic stress and also can decrease the abundance of the proapoptotic factor PUMA, a p73 target. Recombinant measles virus with an engineered deficiency in V protein is capable of inducing more severe cytopathic effects than the wild type, implicating measles virus V protein as an inhibitor of cell death. These findings also suggest that p73-PUMA signaling may be a previously unrecognized arm of cellular innate antiviral immunity.
Collapse
Affiliation(s)
- Cristian D Cruz
- Pancoe-ENH Research Pavilion, Northwestern University, 2200 Campus Drive, Evanston, IL 60208, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Zhang X, Zhang H, Ye L. Effects of hepatitis B virus X protein on the development of liver cancer. ACTA ACUST UNITED AC 2006; 147:58-66. [PMID: 16459163 DOI: 10.1016/j.lab.2005.10.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Revised: 10/08/2005] [Accepted: 10/20/2005] [Indexed: 02/08/2023]
Abstract
Hepatitis B virus (HBV) infections play an important role in the development of cirrhosis and hepatocellular carcinoma (HCC). The pathogenesis of HBV-related HCC, however, has not been fully described. Evidence suggests that the HBV X protein (HBx) plays a crucial role in the pathogenesis of HCC. The high occurrence of anti-HBx antibody in the serum of HCC patients indicates that it could be a prognostic marker of HBV infection and HCC. HBx stimulates and influences signal transduction pathways within cells. HBx also binds to such protein targets as p53, proteasome subunits, and UV-damaged DNA binding proteins. It also interacts with the cyclic AMP-responsive element binding protein, ATF-2, NFkappaB, and basal transcription factors. HBx is primarily localized to the cytoplasm, where it interacts with and stimulates protein kinases, including protein kinase C, Janus kinase/STAT, IKK, PI-3-K, stress-activated protein kinase/Jun N-terminal kinase, and protein kinase B/Akt. It is also found in the mitochondrion, where it influences the Bcl-2 family. This review examines the role of HBx in the life cycle of HBV as well as the various signal transduction pathways involved in the pathogenesis of HBV-induced hepatocarcinogenesis.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Cancer Research, Institute for Molecular Biology, Nankai University, Tianjin, P. R. China.
| | | | | |
Collapse
|
47
|
Dove B, Brooks G, Bicknell K, Wurm T, Hiscox JA. Cell cycle perturbations induced by infection with the coronavirus infectious bronchitis virus and their effect on virus replication. J Virol 2006; 80:4147-56. [PMID: 16571830 PMCID: PMC1440480 DOI: 10.1128/jvi.80.8.4147-4156.2006] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Accepted: 02/02/2006] [Indexed: 11/20/2022] Open
Abstract
In eukaryotic cells, cell growth and division occur in a stepwise, orderly fashion described by a process known as the cell cycle. The relationship between positive-strand RNA viruses and the cell cycle and the concomitant effects on virus replication are not clearly understood. We have shown that infection of asynchronously replicating and synchronized replicating cells with the avian coronavirus infectious bronchitis virus (IBV), a positive-strand RNA virus, resulted in the accumulation of infected cells in the G2/M phase of the cell cycle. Analysis of various cell cycle-regulatory proteins and cellular morphology indicated that there was a down-regulation of cyclins D1 and D2 (G1 regulatory cyclins) and that a proportion of virus-infected cells underwent aberrant cytokinesis, in which the cells underwent nuclear, but not cytoplasmic, division. We assessed the impact of the perturbations on the cell cycle for virus-infected cells and found that IBV-infected G2/M-phase-synchronized cells exhibited increased viral protein production when released from the block when compared to cells synchronized in the G0 phase or asynchronously replicating cells. Our data suggested that IBV induces a G2/M phase arrest in infected cells to promote favorable conditions for viral replication.
Collapse
Affiliation(s)
- Brian Dove
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | | | | | | |
Collapse
|
48
|
Nishio M, Tsurudome M, Ito M, Ito Y. Human parainfluenza virus type 4 is incapable of evading the interferon-induced antiviral effect. J Virol 2006; 79:14756-68. [PMID: 16282476 PMCID: PMC1287573 DOI: 10.1128/jvi.79.23.14756-14768.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The V proteins of some paramyxoviruses have developed the ability to efficiently inactivate STAT protein function as a countermeasure for evading interferon (IFN) responses. Human parainfluenza virus type 4 (hPIV4) is one of the rubulaviruses, which are members of the family Paramyxoviridae, and has a V protein with a highly conserved cysteine-rich domain that is the hallmark of paramyxovirus V proteins. In order to study the function of the hPIV4 V protein, we established HeLa cells expressing the hPIV4A V protein (HeLa/FlagPIV4V). The hPIV4 V protein had no ability to reduce the level of STAT1 or STAT2, although it associated with STAT1, STAT2, DDB1, and Cul4A. It interfered with neither STAT1 and STAT2 tyrosine phosphorylation nor IFN-induced STAT nuclear accumulation. In addition, HeLa/FlagPIV4V cells are fully sensitive to both beta interferon (IFN-beta) and IFN-gamma, indicating that the hPIV4 V protein has no ability to block IFN-induced signaling. We further established HeLa cells expressing various chimeric proteins between the hPIV2 and hPIV4A V proteins. The lack of IFN-antagonistic activity of the hPIV4 V protein is caused by both the P/V common and V-specific domains. At least two regions (amino acids [aa] 32 to 45 and aa 143 to 164) of hPIV4 V in the P/V common domain and one region (aa 200 to 212) of the C terminus are involved in the inability to evade the IFN-induced signaling. Moreover, we established HeLa cells persistently infected with hPIV4 to make sure of the inability to escape IFN and confirmed that hPIV4 is the only paramyxovirus analyzed to date that can't evade the IFN-induced antiviral responses.
Collapse
Affiliation(s)
- Machiko Nishio
- Department of Microbiology, Mie University School of Medicine, 2-174, Edobashi, Tsu-shi, Mie Prefecture, 514-8507 Japan.
| | | | | | | |
Collapse
|
49
|
Lin Y, Horvath F, Aligo JA, Wilson R, He B. The role of simian virus 5 V protein on viral RNA synthesis. Virology 2005; 338:270-80. [PMID: 15950997 DOI: 10.1016/j.virol.2005.05.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Revised: 03/10/2005] [Accepted: 05/06/2005] [Indexed: 12/01/2022]
Abstract
The paramyxovirus simian virus 5 (SV5) has seven genes but encodes eight known viral proteins. The V/P gene is transcribed into two mRNA species: V mRNA from a faithful transcription of the gene and P mRNA from transcription with addition of two G residues at a specific site of the gene. V, a 222-amino acid (AA) residue protein, and P, a 392 AA residue protein, share an identical N-terminus domain of 164 amino acid residues. P is essential for SV5 RNA replication and transcription. Whereas it is known that V plays important roles in virus pathogenesis, the role of V in SV5 replication and transcription is not clear. A mini-genome system, free of vaccinia virus gene expression system, consisting of plasmids expressing NP, P, and L, as well as a plasmid encoding a reporter gene, chloramphenicol acetyltransferase (CAT) flanked by SV5 trailer and leader sequences under control of a bacteriophage T7 RNA polymerase promoter, has been established to examine the role of V in SV5 RNA transcription and replication. Addition of V-expressing plasmid in the mini-genome system caused inhibition of the reporter gene expression, suggesting that V plays a role in regulating SV5 gene expression. By examining the amount of encapsidated viral RNA genome using reverse transcription with primer annealing to viral anti-genome RNA and PCR, it was found that expression of V reduced the amount of viral RNA genome in the mini-genome system, suggesting that V inhibits viral RNA replication. To examine whether the V protein inhibits viral RNA transcription as well, a mini-genome system with a defective anti-genome promoter (AGP) such that a reporter gene (luciferase, Luc) expression is only derived from transcription of newly produced mini-genome and not from de novo replicated viral genome due to the defect in replication element has been utilized. The V protein inhibited luciferase expression from the mini-genome with a defective AGP, suggesting V inhibits SV5 transcription. Thus, SV5 V inhibits both SV5 RNA replication and transcription.
Collapse
Affiliation(s)
- Yuan Lin
- Department of Veterinary Science, Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | |
Collapse
|
50
|
Nishio M, Tsurudome M, Ito M, Garcin D, Kolakofsky D, Ito Y. Identification of paramyxovirus V protein residues essential for STAT protein degradation and promotion of virus replication. J Virol 2005; 79:8591-601. [PMID: 15956600 PMCID: PMC1143765 DOI: 10.1128/jvi.79.13.8591-8601.2005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Some paramyxovirus V proteins induce STAT protein degradation, and the amino acids essential for this process in the human parainfluenza virus type 2 (hPIV2) V protein have been studied. Various recombinant hPIV2s and cell lines constitutively expressing various mutant V proteins were generated. We found that V proteins with replacement of Cys residues of the Cys cluster were still able to bind STATs but were unable to induce their degradation. The hPIV2 V protein binds STATs via a W-(X)3-W-(X)9-W Trp motif located just upstream of the Cys cluster. Replacements of two or more Trp residues in this motif resulted in a failure to form a V/STAT2 complex. We have also identified two Phe residues of the hPIV2 V protein that are essential for STAT degradation, namely, Phe207, lying within the Cys cluster, and Phe143, in the P/V common region of the protein. Interestingly, infection of BHK cells with hPIV2 led to the specific degradation of STAT1 and not STAT2. Other evidence for the cell species specificity of hPIV2-induced STAT degradation is presented. Finally, a V-minus hPIV2, which can express only the P protein from its P gene, was generated and partially characterized. In contrast to V-minus viruses of other paramyxovirus genera, this V-minus rubulavirus was highly debilitated, and its growth even in Vero cells was very limited. The structural rubulavirus V proteins, as expected, are thus clearly important in promoting virus growth, independent of their anti-interferon (IFN) activity. Interestingly, many of the residues that are essential for anti-IFN activity, e.g., the Cys of this cluster and Phe207 within this cluster, as well as the Trp of this motif, are also essential for promoting virus growth.
Collapse
Affiliation(s)
- Machiko Nishio
- Department of Microbiology, Mie University School of Medicine, Tsu-shi, Mie Prefecture, Japan.
| | | | | | | | | | | |
Collapse
|