1
|
Blondin-Brosseau M, Zhang W, Gravel C, Harlow J, Li X, Nasheri N. Comparison of Methods for Extraction of Infectious Influenza Virus from Raw Milk Cheeses. J Food Prot 2025; 88:100529. [PMID: 40345493 DOI: 10.1016/j.jfp.2025.100529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
In recent years, the highly pathogenic avian influenza (HPAI) H5N1 viruses have spread widely among birds and multiple mammal species. The HPAI spillover to dairy cattle, and its excretion in milk in high-titers has created a new interface for human exposure and has raised food safety concerns. Multiple lines of evidence show that pasteurization is effective in inactivation of influenza viruses. In Canada, dairy products must be pasteurized with the exception of cheese. Since influenza viruses were not considered as foodborne, there is no data available regarding their survival in cheeses and no standard method exists for their extraction from food commodities, including dairy products. Herein, we examined the efficacy of multiple methods for the extraction of infectious H1N1 virus (as a representative for type A influenza viruses) from cream cheese made from unpasteurized milk. We used murine norovirus (MNV) as a surrogate for human norovirus and also as a process control virus and examined the efficacy of the employed methods by plaque assay. The limit of detection for the two best-performing methods was determined using a variety of soft and firm raw-milk cheeses. The described methods assist health authorities for the surveillance of foodborne viruses in dairy products.
Collapse
Affiliation(s)
| | - Wanyue Zhang
- Centre for Oncology, Radiopharmaceuticals and Research, Health Canada, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Caroline Gravel
- Centre for Oncology, Radiopharmaceuticals and Research, Health Canada, Ottawa, Ontario, Canada
| | - Jennifer Harlow
- Bureau Microbial Hazards, Health Canada, Ottawa, Ontario, Canada
| | - Xuguang Li
- Centre for Oncology, Radiopharmaceuticals and Research, Health Canada, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Neda Nasheri
- Bureau Microbial Hazards, Health Canada, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
2
|
Jain R, Sharma H, Pena L, Jit S, Rathi B, De Oliveira RN, Verma M. Influenza virus: Genomic insights, evolution, and its clinical presentation. Microb Pathog 2025; 205:107671. [PMID: 40345348 DOI: 10.1016/j.micpath.2025.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Influenza viruses belong to the family Orthomyxoviridae and have been a concern for public health due to frequent epidemics and sporadic waves of pandemics. According to WHO, seasonal strains of the influenza A virus (IAV) infect 3 to 5 million people every year and result in 0.29-0.65 million deaths globally. Among all proteins of IAV, the hemagglutinin (HA) and neuraminidase (NA) have high mutation rates and have been reported to undergo antigenic drift and shift leading to novel strains. Hence continuous revision in drugs and vaccines regime is an economic burden as these reassortments can cause high morbidity among the immunologically naive population. Infection rates are higher among older age groups, infants and patients reported with other respiratory distress. Routine diagnostic tests include reverse transcription polymerase chain reaction (RT-PCR), viral culture, and immunofluorescence assays; however, the diagnosis may be hindered due to the resemblance of the clinical presentation of other respiratory viruses with influenza viruses. Here we present a comprehensive review which sheds light on the genomic structure, evolution, pathogenicity and clinical presentation of influenza virus that can help to distinguish it from other respiratory viruses. A detailed analysis will ultimately pave the way for the development of more effective antiviral drugs and a universal vaccine.
Collapse
Affiliation(s)
- Rishika Jain
- Department of Zoology, Hansraj College, University of Delhi, Mahatma Hansraj Marg, Malkaganj, Delhi, 110007, India
| | - Himanshu Sharma
- Department of Zoology, Hansraj College, University of Delhi, Mahatma Hansraj Marg, Malkaganj, Delhi, 110007, India
| | - Lindomar Pena
- Department of Virology and Experimental Therapy Aggeu Magalhães Institute - Fiocruz-PE, Recife, Pernambuco, Brazil
| | - Simran Jit
- Department of Zoology, Miranda House, University of Delhi, Delhi, 110007, India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi, 110007, India
| | - Ronaldo Nascimento De Oliveira
- Department of Chemistry, Laboratory of Synthesis of Bioactive Compounds, Federal Rural University of Pernambuco, Recife, Pernambuco, Brazil
| | - Mansi Verma
- Department of Zoology, Hansraj College, University of Delhi, Mahatma Hansraj Marg, Malkaganj, Delhi, 110007, India.
| |
Collapse
|
3
|
Jiang S, Ding Y, Zhao G, Ye S, Liu S, Yin Y, Li Z, Zou X, Xie D, You C, Guo X. Species-specific RNA barcoding technology for rapid and accurate identification of four types of influenza virus. BMC Genomics 2025; 26:409. [PMID: 40295995 PMCID: PMC12036255 DOI: 10.1186/s12864-025-11602-0] [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: 09/24/2024] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The influenza virus (IV) is responsible for seasonal flu epidemics. Constant mutation of the virus results in new strains and widespread reinfections across the globe, bringing great challenges to disease prevention and control. Research has demonstrated that barcoding technology efficiently and cost-effectively differentiates closely related species on a large scale. We screened and validated species-specific RNA barcode segments based on the genetic relationships of four types of IVs, facilitating their precise identification in high-throughput sequencing viral samples. RESULTS Through the analysis of single nucleotide polymorphism, population genetic characteristics, and phylogenetic relationships in the training set, 7 IVA type, 29 IVB type, 40 IVC type, and 5 IVD type barcode segments were selected. In the testing set, the nucleotide-level recall rate for all barcode segments reached 96.86%, the average nucleotide-level specificity was approximately 55.27%, the precision rate was 100%, and the false omission rate was 0%, demonstrating high accuracy, specificity, and generalization capabilities for species identification. Ultimately, all four types of IVs were visualized in a combination of one-dimensional and two-dimensional codes and stored in an online database named Influenza Virus Barcode Database (FluBarDB, http://virusbarcodedatabase.top/database/index.html ). CONCLUSION This study validates the effective application of RNA barcoding technology in the detection of IVs and establishes criteria and procedures for selecting species-specific molecular markers. These advancements enhance the understanding of the genetic and epidemiological characteristics of IVs and enable rapid responses to viral genetic mutations.
Collapse
Affiliation(s)
- Shuai Jiang
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Yunyun Ding
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Gaili Zhao
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Shunxing Ye
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Shucan Liu
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Yan Yin
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Zeqi Li
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Xiaoxiao Zou
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Daolong Xie
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Changqiao You
- College of Biology, Hunan University, Changsha, Hunan, 410082, China.
| | - Xinhong Guo
- College of Biology, Hunan University, Changsha, Hunan, 410082, China.
| |
Collapse
|
4
|
Domínguez-Odio A, Rodríguez-Martínez E, Paneque Zayas M, Cala-Delgado DL. Research publications and global manufacture of veterinary vaccines against avian influenza A (2019-2023). Front Vet Sci 2025; 12:1394675. [PMID: 40144520 PMCID: PMC11936895 DOI: 10.3389/fvets.2025.1394675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
The characteristics of the avian influenza virus and its worldwide spread have led to intense and unprecedented scientific activity and industrial production for preventive veterinary vaccines. However, knowledge gaps remain regarding the best strategies to prevent epidemiological events in the future. In this context, the present study aimed to provide a global analysis on the scientific and industrial production of avian influenza type A vaccines for farm animals and pets during the period 2019 2023. The Scopus database was used as the primary source of information (12,162 keywords, 2,437 scientific articles, 659 academic journals, and 46 countries) for the academic analysis, while technical information posted on official institutional websites (136 commercial formulations, 24 vaccines manufacturers, and 17 countries) was collected to conduct the industrial analysis. 3,045, 25.0%) exhibited the highest levels of co-occurrence in the sciences; the journal Vaccine was the most productive in terms of articles (11.8%, 288/2,437), and the countries with the most publications were the USA (25.5%, 622/2,437) and China (23.1%, 564/2,437). The most internationally marketed vaccines were inactivated (86.0%, 117/136), avian (47.1%, 64/136), and combined (52.2%, 71/136) vaccines as well as those containing Newcastle antigens (38.0%, 27/71). In conclusion, the study demonstrated the fundamental role of classical production methods (based on the use of the whole pathogen) in avian influenza A research and the production of veterinary vaccines.
Collapse
|
5
|
Srivastava R, Jaiswal N, Kharkwal H, Dubey NK, Srivastava R. Phytomedical Properties of Carica papaya for Boosting Human Immunity Against Viral Infections. Viruses 2025; 17:271. [PMID: 40007026 PMCID: PMC11861161 DOI: 10.3390/v17020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Carica papaya, a tropical fruit-bearing plant, has attracted significant attention for its diverse phytomedical properties and its ability to regulate both innate and adaptive immunity, making it a promising natural therapeutic agent. C. papaya is rich in bioactive compounds that play a multifaceted role in immunomodulation. These bioactive constituents have demonstrated efficacy not only against the dengue virus but also against other viral infections, including COVID-19 (Corona Virus Disease 2019), Human Immunodeficiency Virus (HIV), Zika virus, and others. The antiviral effects of C. papaya are achieved through its ability to enhance host immunity, mitigate inflammation, reduce oxidative stress, inhibit viral replication, and modulate immune responses. These mechanisms highlight its potential as a candidate for antiviral therapies, paving the way for further exploration of its pharmacological applications and promoting eco-friendly, accessible healthcare solutions for combating viral diseases. This review highlights the antiviral potential of C. papaya extracts in inhibiting viral replication and modulating immune responses, emphasizing the need for further studies and clinical trials to validate their efficacy against other medically significant viruses causing human diseases.
Collapse
Affiliation(s)
- Rashmi Srivastava
- School of Life Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, Uttar Pradesh, India
| | - Neeshma Jaiswal
- School of Life Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, Uttar Pradesh, India
| | - Harsha Kharkwal
- Amity Institute of Phytochemistry and Phytomedicine, Amity University, Noida 201313, Uttar Pradesh, India
| | - Neeraj Kumar Dubey
- Botany Department, Rashtriya PG College, Jaunpur 222003, Uttar Pradesh, India
| | - Rakesh Srivastava
- Research and Development, Helix Biosciences, New Delhi 110028, Delhi, India
| |
Collapse
|
6
|
Xiang H, Hong C, Tian Y, Gao Y, Cun Y, Zhao H, Li G, Chen Y, Zhou J. Design, development, and validation of a strand-specific RT-qPCR assay to differentiate replicating versus nonreplicating Rabies virus. J Virol Methods 2025; 332:115054. [PMID: 39522826 DOI: 10.1016/j.jviromet.2024.115054] [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: 07/22/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
The Rabies virus, a single-strand RNA virus with a negative-sense polarity, is responsible for causing encephalitis and is a zoonotic disease. If not promptly treated after infection, it has a close to 100 % fatality rate. Similar to other negative-sense polarity single-strand RNA viruses, the Rabies virus requires the creation of a positive-strand RNA intermediate for replication. One approach to identify this replication activity is to detect the complementary strand of the viral RNA genome in suspected infected cells or tissues. The reported Rabies virus RT-qPCR detection methods are designed to detect total viral load in samples without distinguishing between positive- and negative-strand for RNA viruses. As such, in this study, a sensitive Taqman-based strand-specific RT-qPCR assay has been developed to quantitatively detect both the positive- and negative-strand of the Rabies virus. This method demonstrates good reproducibility across a wide dynamic range and exhibits linearity of 8 logs with a lower limit of detection of 103 copies/μL for the positive-strand and 9 logs with a lower limit of detection of 102 copies/μL for the negative-strand. Notably, it can accurately detect a specific viral RNA strand even in the presence of high levels of the opposite strand, confirming the method's specificity. In summary, a reliable strand-specific RT-qPCR assay has been developed and validated to differentiate replicating from non-replicating Rabies virus.
Collapse
Affiliation(s)
- Hong Xiang
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Chao Hong
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Yuan Tian
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Yu Gao
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Yina Cun
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Hongling Zhao
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Guilan Li
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Yu Chen
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China
| | - Jian Zhou
- Institute of Medical Biology Chinese Academy of Medical Science & Peking Union Medical College, Kunming, PR China.
| |
Collapse
|
7
|
Lv X, Zheng T, Lei X, Ren L, Zhao S, Wang J, Zhuo Z, Wang J. RTP4 restricts influenza A virus infection by targeting the viral NS1 protein. Virology 2025; 603:110397. [PMID: 39798334 DOI: 10.1016/j.virol.2025.110397] [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: 11/14/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The influenza A virus evades the host innate immune response to establish infection by inhibiting RIG-I activation through its nonstructural protein 1 (NS1). Here, we reported that receptor-transporting protein 4 (RTP4), an interferon-stimulated gene (ISG), targets NS1 to inhibit influenza A virus infection. Depletion of RTP4 significantly increased influenza A virus multiplication, while NS1-deficient viruses were unaffected. Mechanistically, RTP4 interacts with NS1 in an RNA-dependent manner and sequesters it from the TRIM25-RIG-I complex, thereby restoring TRIM25-mediated RIG-I K63-linked ubiquitination and subsequent activation of IRF3. Antiviral activity of RTP4 requires the evolutionarily conserved CXXC motifs and an H149 residue in the zinc finger domain, mutations of which disrupted RTP4-NS1 interaction and abrogated the ability of RTP4 to rescue RIG-I-mediated signaling. Collectively, our findings provided insights into the mechanism by which an ISG restricts influenza A virus replication by reactivating host antiviral signaling.
Collapse
Affiliation(s)
- Xueying Lv
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, China
| | - Tian Zheng
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Lei
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China
| | - Lili Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China
| | - Shiyi Zhao
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Jingfeng Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Zhou Zhuo
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China; National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
8
|
Teo QW, Wang Y, Lv H, Oade MS, Mao KJ, Tan TJC, Huan YW, Rivera-Cardona J, Shao EK, Choi D, Wang C, Tavakoli Dargani Z, Brooke CB, Te Velthuis AJW, Wu NC. Probing the functional constraints of influenza A virus NEP by deep mutational scanning. Cell Rep 2025; 44:115196. [PMID: 39817904 PMCID: PMC11830162 DOI: 10.1016/j.celrep.2024.115196] [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: 06/10/2024] [Revised: 11/24/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025] Open
Abstract
The influenza A virus nuclear export protein (NEP) is a multifunctional protein that is essential for the viral life cycle and has very high sequence conservation. However, since the open reading frame of NEP largely overlaps with that of another influenza viral protein, non-structural protein 1, it is difficult to infer the functional constraints of NEP based on sequence conservation analysis. In addition, the N-terminal of NEP is structurally disordered, which further complicates the understanding of its function. Here, we systematically measure the replication fitness effects of >1,800 mutations of NEP. Our results show that the N-terminal domain has high mutational tolerance. Additional experiments show that N-terminal domain mutations affect viral transcription and replication dynamics, host cellular responses, and mammalian adaptation of avian influenza virus. Overall, our study not only advances the functional understanding of NEP but also provides insights into its evolutionary constraints.
Collapse
Affiliation(s)
- Qi Wen Teo
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Yiquan Wang
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Huibin Lv
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Michael S Oade
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Kevin J Mao
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Timothy J C Tan
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Yang Wei Huan
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Joel Rivera-Cardona
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Evan K Shao
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Danbi Choi
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Chaoyang Wang
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Zahra Tavakoli Dargani
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Christopher B Brooke
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Aartjan J W Te Velthuis
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Nicholas C Wu
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
9
|
Chenavier F, Zarkadas E, Freslon LL, Stelfox A, Schoehn G, Ruigrok RH, Ballandras-Colas A, Crépin T. Influenza a virus antiparallel helical nucleocapsid-like pseudo-atomic structure. Nucleic Acids Res 2025; 53:gkae1211. [PMID: 39673795 PMCID: PMC11797009 DOI: 10.1093/nar/gkae1211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/29/2024] [Accepted: 11/21/2024] [Indexed: 12/16/2024] Open
Abstract
Influenza A viruses are responsible for human seasonal epidemics and severe animal pandemics with a risk of zoonotic transmission to humans. The viral segmented RNA genome is encapsidated by nucleoproteins (NP) and attached to the heterotrimeric polymerase, forming the viral ribonucleoproteins (vRNPs). Flexible helical vRNPs are central for viral transcription and replication. In this study, we present an advanced biological tool, the antiparallel helical RNP-like complex, assembled from recombinant N-terminally truncated NP and short synthetic RNA. The 3.0 Å cryo-electron microscopy structure details for the first time the whole RNA pathway across NP as well as NP-NP interactions that drive the antiparallel helical assembly accommodating major and minor grooves. Our findings show that the surface of the protein can harbour several conformations of the RNA, confirming that the number of nucleobases that binds to NP is not fixed, but ranges probably between 20 and 24. Taking all together, our data provide details to further understand the genome encapsidation and explain the inherent flexibility of influenza A virus vRNPs.
Collapse
Affiliation(s)
- Florian Chenavier
- Univ. Grenoble Alpes, CNRS, CEA, IBS, 71 avenue des Martyrs, F-38000 Grenoble, France
| | - Eleftherios Zarkadas
- Univ. Grenoble Alpes, CNRS, CEA, EMBL, ISBG, 71 avenue des Martyrs, F-38000 Grenoble, France
| | - Lily-Lorette Freslon
- Univ. Grenoble Alpes, CNRS, CEA, IBS, 71 avenue des Martyrs, F-38000 Grenoble, France
| | - Alice J Stelfox
- Univ. Grenoble Alpes, CNRS, CEA, IBS, 71 avenue des Martyrs, F-38000 Grenoble, France
| | - Guy Schoehn
- Univ. Grenoble Alpes, CNRS, CEA, IBS, 71 avenue des Martyrs, F-38000 Grenoble, France
| | - Rob W H Ruigrok
- Univ. Grenoble Alpes, CNRS, CEA, IBS, 71 avenue des Martyrs, F-38000 Grenoble, France
| | | | - Thibaut Crépin
- Univ. Grenoble Alpes, CNRS, CEA, IBS, 71 avenue des Martyrs, F-38000 Grenoble, France
| |
Collapse
|
10
|
Hama S, Watanabe-Takahashi M, Nishimura H, Omi J, Tamada M, Saitoh T, Maenaka K, Okuda Y, Ikegami A, Kitagawa A, Furuta K, Izumi K, Shimizu E, Nishizono T, Fujiwara M, Miyasaka T, Takamori S, Takayanagi H, Nishikawa K, Kobayashi T, Toyama-Sorimachi N, Yamashita M, Senda T, Hirokawa T, Bito H, Nishikawa K. CaMKII-dependent non-canonical RIG-I pathway promotes influenza virus propagation in the acute-phase of infection. mBio 2025; 16:e0008724. [PMID: 39601535 PMCID: PMC11708044 DOI: 10.1128/mbio.00087-24] [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: 01/26/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is one of hundreds of host-cell factors involved in the propagation of type A influenza virus (IAV), although its mechanism of action is unknown. Here, we identified CaMKII inhibitory peptide M3 by targeting its kinase domain using affinity-based screening of a tailored random peptide library. M3 inhibited IAV cytopathicity and propagation in cells by specifically inhibiting the acute-phase activation of retinoic acid-inducible gene I (RIG-I), which is uniquely regulated by CaMKII. Downstream of the RIG-I pathway activated TBK1 and then IRF3, which induced small but sufficient amounts of transcripts of the genes for IFN α/β to provide the capped 5'-ends that were used preferentially as primers to synthesize viral mRNAs by the cap-snatching mechanism. Importantly, knockout of RIG-I in cells almost completely inhibited the expression of IFN mRNAs and subsequent viral NP mRNA early in infection (up to 6 h after infection), which then protected cells from cytopathicity 24 h after infection. Thus, CaMKII-dependent acute-phase activation of RIG-I promoted IAV propagation, whereas the canonical RIG-I pathway stimulated antiviral activity by inducing large amounts of mRNA for IFNs and then for antiviral proteins later in infection. Co-administration of M3 with IAV infection rescued mice from the lethality and greatly reduced proinflammatory cytokine mRNA expression in the lung, indicating that M3 is highly effective against IAV in vivo. Thus, regulation of the CaMKII-dependent non-canonical RIG-I pathway may provide a novel host-factor-directed antiviral therapy.IMPORTANCEThe recent emergence of IAV strains resistant to commonly used therapeutic agents that target viral proteins has exacerbated the need for innovative strategies. Here, we originally identified CaMKII-inhibitory peptide M3, which efficiently inhibits IAV-lethality in vitro and in vivo. M3 specifically inhibited the acute-phase activation of RIG-I, which is a novel pathway to promote IAV propagation. Thus, this pathway acts in an opposite manner compared with the canonical RIG-I pathway, which plays essential roles in antiviral innate immune response later in infection. The CaMKII-dependent non-canonical RIG-I pathway can be a promising and novel drug target for the treatment of infections.
Collapse
Affiliation(s)
- Shinichiro Hama
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Miho Watanabe-Takahashi
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Hiroki Nishimura
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Jumpei Omi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masakazu Tamada
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Takashi Saitoh
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Hokkaido, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Hokkaido, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development, HU-IVReD, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuta Okuda
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Aoi Ikegami
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Asami Kitagawa
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Koudai Furuta
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Kana Izumi
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Eiko Shimizu
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Takashi Nishizono
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Makoto Fujiwara
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Tomohiro Miyasaka
- Department of Physiology and Anatomy, Faculty of Pharmacy, Nihon University, Funabashi, Japan
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Keizo Nishikawa
- Department of Cell Biology and Metabolic Biochemistry, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Toshihiko Kobayashi
- Division of Human Immunology, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Noriko Toyama-Sorimachi
- Division of Human Immunology, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Makoto Yamashita
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, Japan
| | - Toshiya Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba, Ibaraki, Japan
| | - Takatsugu Hirokawa
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Division of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kiyotaka Nishikawa
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| |
Collapse
|
11
|
Dutta S, Zhu Y, Almuntashiri S, Peh HY, Zuñiga J, Zhang D, Somanath PR, Ramírez G, Irineo-Moreno V, Jiménez-Juárez F, López-Salinas K, Regino N, Campero P, Crocker SJ, Owen CA, Wang X. PDGFRα-positive cell-derived TIMP-1 modulates adaptive immune responses to influenza A viral infection. Am J Physiol Lung Cell Mol Physiol 2025; 328:L60-L74. [PMID: 39585242 PMCID: PMC11905806 DOI: 10.1152/ajplung.00104.2024] [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: 03/20/2024] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1) is a physiologic inhibitor of the matrix metalloproteinases (MMPs), but little is known about the role of TIMP-1 in regulating the pathogenesis of influenza A virus (IAV) infection. Here, we performed both in vivo and in vitro experiments to investigate the regulation and function of TIMP-1 during IAV infection. Specifically, plasma levels of TIMP-1 are significantly increased in human subjects and wild-type (WT) mice infected with 2009 H1N1 IAV compared with levels in uninfected controls. Also, TIMP-1 is strikingly upregulated in PDGFRα positive (PDGFRα+) cells in IAV-infected murine lungs as demonstrated using conditional KO (cKO) mice with a specific deletion of Timp-1 in PDGFRα+ cells. Our in vitro data indicated that TIMP-1 is induced by transforming growth factor-β (TGF-β) during lipofibroblasts (lipoFBs)-to-myofibroblast (myoFB) transdifferentiation. Timp-1 deficiency protects mice from H1N1 IAV-induced weight loss, mortality, and lung injury. IAV-infected Timp-1-deficient mice showed increased macrophages, and B and T cell counts in bronchoalveolar lavage (BAL) on day 7 postinfection (p.i.), but reduced BAL neutrophil counts. Increased Cxcl12 levels were detected in both BAL cells and lungs from Timp-1-deficient mice on day 3 p.i. Taken together, our data strongly link TIMP-1 to IAV pathogenesis. We identified that PDGFRα-lineage cells are the main cellular source of elevated TIMP-1 during IAV infection. Loss of Timp-1 attenuates IAV-induced mortality and promotes T and B cell recruitment. Thus, TIMP-1 may be a novel therapeutic target for IAV infection.NEW & NOTEWORTHY Our data strongly link tissue inhibitor of metalloproteinases-1 (TIMP-1) to influenza A virus (IAV) pathogenesis. TIMP-1 is highly increased in PDGFRα-lineage cells during IAV infection. Transforming growth factor-β (TGF-β) induces TIMP-1 during lipofibroblast (lipoFB)-to- myofibroblast (myoFB) transdifferentiation. Timp-1 deficiency protects mice from H1N1 IAV-induced weight loss, mortality, and lung injury. TIMP-1 may be a novel therapeutic target for IAV infection.
Collapse
Affiliation(s)
- Saugata Dutta
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Hong Yong Peh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| | - Joaquin Zuñiga
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Gustavo Ramírez
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Valeria Irineo-Moreno
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Karen López-Salinas
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Nora Regino
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Paloma Campero
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, United States
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
12
|
Hutchinson EC, Amorim MJ, Yamauchi Y. Understanding Influenza. Methods Mol Biol 2025; 2890:1-26. [PMID: 39890719 DOI: 10.1007/978-1-0716-4326-6_1] [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] [Indexed: 02/03/2025]
Abstract
Influenza, a serious illness of humans and domesticated animals, has been studied intensively for many years. It therefore provides an example of how much we can learn from detailed studies of an infectious disease, and of how even the most intensive scientific research leaves further questions to answer. This introduction is written for researchers who have become interested in one of these unanswered questions, but who may not have previously worked on influenza. To investigate these questions, researchers must not only have a firm grasp of relevant methods and protocols; they must also be familiar with the basic details of our current understanding of influenza. This chapter briefly covers the burden of disease that has driven influenza research, summarizes how our thinking about influenza has evolved over time, and sets out key features of influenza viruses by discussing how we classify them and what we currently understand of their replication. It does not aim to be comprehensive, as any researcher will read deeply into the specific areas that have grasped their interest. Instead, it aims to provide a general summary of how we came to think about influenza in the way we do now, in the hope that the reader's own research will help us to understand it better.
Collapse
Affiliation(s)
| | - Maria João Amorim
- Católica Biomedical Research Centre (CBR), Católica Medical School, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Yohei Yamauchi
- Institute of Pharmaceutical Sciences, ETH Zurich, Zürich, Switzerland
| |
Collapse
|
13
|
Nakano M, Noda T. Purification and Ultramicroscopic Observation of the Influenza A Virus Ribonucleoprotein Complex. Methods Mol Biol 2025; 2890:141-149. [PMID: 39890725 DOI: 10.1007/978-1-0716-4326-6_7] [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] [Indexed: 02/03/2025]
Abstract
Influenza A virus (IAV) has an eight-segmented, single-stranded, negative-sense viral genomic RNA (vRNA). Each vRNA strand associates with nucleoproteins and an RNA-dependent RNA polymerase complex to form a viral ribonucleoprotein (vRNP) complex. IAV vRNPs adopt a flexible double-helical configuration that varies in length. Although the transcription and replication of vRNA take place in the context of vRNPs, the precise structural conformation of vRNPs during RNA synthesis remains partially elucidated. To unravel the intricate ultrastructure of the vRNP, it is necessary to purify it while preserving its native functionality. Herein, we introduce a comprehensive protocol for the purification of IAV vRNPs using glycerol gradient ultracentrifugation. Furthermore, we provide a method for the high-speed atomic force microscopy observation of vRNPs during viral RNA synthesis.
Collapse
Affiliation(s)
- Masahiro Nakano
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan.
| | - Takeshi Noda
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan.
| |
Collapse
|
14
|
Xue R, Bao D, Ma T, Niu S, Wu Z, Lv X, Zhang Y, Xu G, Yan D, Zhang Z, Pan X, Yan M, Teng Q, Yuan C, Li Z, Liu Q. hnRNPH1 Inhibits Influenza Virus Replication by Binding Viral RNA. Microorganisms 2024; 13:24. [PMID: 39858792 PMCID: PMC11767954 DOI: 10.3390/microorganisms13010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/27/2025] Open
Abstract
During the life cycle of the influenza virus, viral RNPs (vRNPs) are transported to the nucleus for replication. Given that a large number of progeny viral RNA occupies the nucleus, whether there is any host protein located in the nucleus that recognizes the viral RNA and inhibits the viral replication remains largely unknown. In this study, to explore the role of hnRNPH1 in influenza virus infection, we knocked down and over-expressed the hnRNPH1 proteins in 293T cells, then infected the cells with the influenza virus. The results showed that the host hnRNPH1 inhibits the replication of H1N1 and H9N2 influenza viruses by restraining the polymerase activity of viruses. hnRNPH1 contains two RNA recognition motifs (RRM1) and RRM2. Further studies indicated that hnRNPH1 specifically binds to the viral RNA of the PB1, PA, and NP genes. Mutation of the key residues tryptophan and tyrosine in RRM1 and RRM2 abolished the binding affinity to viral RNA and the suppression of polymerase activity of the influenza virus. All the results suggested that hnRNPH1 suppresses polymerase activity and replication of the influenza virus by binding viral RNA.
Collapse
Affiliation(s)
- Ruixue Xue
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
- Shandong Provincial Center for Animal Disease Control and Prevention (Shandong Provincial Center for Zoonoses Epidemiology Investigation and Surveillance), 4566 Tangye West Road, Licheng District, Jinan 250100, China
| | - Danqi Bao
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Tianxin Ma
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Shiqi Niu
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Zihua Wu
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Xuehua Lv
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Yunxiang Zhang
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Guanlong Xu
- China Institute of Veterinary Drug Control, Beijing 100082, China;
| | - Dawei Yan
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Zhifei Zhang
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Xue Pan
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Minghao Yan
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Qiaoyang Teng
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Chunxiu Yuan
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Zejun Li
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| | - Qinfang Liu
- Shanghai Veterinary Research Institute, 518 Ziyue Road, Minhang District, Shanghai 200241, China; (R.X.); (D.B.); (T.M.); (S.N.); (Z.W.); (X.L.); (Y.Z.); (D.Y.); (Z.Z.); (X.P.); (M.Y.); (Q.T.); (C.Y.)
| |
Collapse
|
15
|
Uchikubo-Kamo T, Ishimoto N, Umezawa H, Hirohama M, Ono M, Kawabata H, Kamata K, Ohki M, Yoshida H, Park JH, Tame JRH, Kawaguchi A, Park SY. Structural insights into Influenza A virus RNA polymerase PB1 binding to nuclear import host factor RanBP5. Biochem Biophys Res Commun 2024; 739:150952. [PMID: 39536408 DOI: 10.1016/j.bbrc.2024.150952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 10/27/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
The genome of influenza A viruses consists of eight RNA segments that form a heterotrimer, and the viral genome undergoes transcription and replication in the nucleus. Thus, during infection, newly synthesized RNA polymerase subunits must be imported into the nucleus. Although several models have been proposed for this process, the consensus is that the RNA polymerase subunits PB1 and PA form a dimer in the cytoplasm and are transported into the nucleus by Ran binding protein 5 (RanBP5). The PB2 subunit undergoes separate transport to complete the nuclear import. However, the molecular mechanism of nuclear import by host factors and their interactions with proteins are largely unknown. Here we present the structural analysis of the RanBP5 and PB1 NLS domain complex by cryo-EM at 3.2 Å resolution. The pattern shows that the NLS domain of PB1 does not exist in a secondary structure and interacts with RanBP5 in a wrapped state. In addition, biochemical analyses of the mutant have identified critical amino acid sites involved in complex binding. The results suggest a stepwise assembly of influenza virus structural components regulated by nuclear import mechanisms and host factor binding, with important implications for drug discovery research.
Collapse
Affiliation(s)
- Tomomi Uchikubo-Kamo
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Naito Ishimoto
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Haruka Umezawa
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Mikako Hirohama
- Department of Infection Biology, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Maasa Ono
- Department of Infection Biology, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Haruka Kawabata
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Kenichi Kamata
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Mio Ohki
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Hisashi Yoshida
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Jae-Hyun Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan; Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jeremy R H Tame
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan
| | - Atsushi Kawaguchi
- Department of Infection Biology, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, 230-0045, Japan.
| |
Collapse
|
16
|
Williams SL, Qi L, Sheng ZM, Xiao Y, Freeman A, Matthews L, Legaspi SF, Fodor E, Taubenberger JK. Effect of pandemic influenza A virus PB1 genes of avian origin on viral RNA polymerase activity and pathogenicity. SCIENCE ADVANCES 2024; 10:eads5735. [PMID: 39671482 PMCID: PMC11641000 DOI: 10.1126/sciadv.ads5735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Zoonotic influenza A virus (IAV) infections pose a substantial threat to global health. The influenza RNA-dependent RNA polymerase (RdRp) comprises the PB2, PB1, and PA proteins. Of the last four pandemic IAVs, three featured avian-origin PB1 genes. Prior research linked these avian PB1 genes to increased viral fitness when reassorted with human IAV genes. This study evaluated chimeric RdRps with PB1 genes from the 1918, 1957, and 1968 pandemic IAVs in a low pathogenic avian influenza (LPAI) virus background to assess polymerase activity and pathogenicity. Substituting in the pandemic PB1 genes reduced polymerase activity, virulence, and altered lung pathology, while the native LPAI PB1 showed the highest pathogenicity and polymerase activity. The native LPAI PB1 virus caused severe pneumonia and high early viral RNA levels, correlating with elevated host cytokine signaling. Increased genetic distance from the LPAI PB1 sequence correlated with reduced polymerase activity, IFN-β expression, viral replication, and pathogenicity.
Collapse
Affiliation(s)
- Stephanie L. Williams
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Li Qi
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Ashley Freeman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Lex Matthews
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Sharon Fong Legaspi
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Ervin Fodor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Disease, National Institutes of Health, National Institute for Allergy and Infectious Diseases, Bethesda, MD, USA
| |
Collapse
|
17
|
Cheng Z, Ma J, Zhao C. Advantages of Broad-Spectrum Influenza mRNA Vaccines and Their Impact on Pulmonary Influenza. Vaccines (Basel) 2024; 12:1382. [PMID: 39772044 PMCID: PMC11680418 DOI: 10.3390/vaccines12121382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Influenza poses a significant global health challenge due to its rapid mutation and antigenic variability, which often leads to seasonal epidemics and frequent outbreaks. Traditional vaccines struggle to offer comprehensive protection because of mismatches with circulating viral strains. The development of a broad-spectrum vaccine is therefore crucial. This paper explores the potential of mRNA vaccine technology to address these challenges by providing a swift, adaptable, and broad protective response against evolving influenza strains. We detail the mechanisms of antigenic variation in influenza viruses and discuss the rapid design and production, enhanced immunogenicity, encoding of multiple antigens, and safety and stability of mRNA vaccines compared to traditional methods. By leveraging these advantages, mRNA vaccines represent a revolutionary approach in influenza prevention, potentially offering broad-spectrum protection and significantly improving global influenza management and response strategies.
Collapse
Affiliation(s)
- Ziqi Cheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China;
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Junfeng Ma
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Chenyan Zhao
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| |
Collapse
|
18
|
Panthi S, Hong JY, Satange R, Yu CC, Li LY, Hou MH. Antiviral drug development by targeting RNA binding site, oligomerization and nuclear export of influenza nucleoprotein. Int J Biol Macromol 2024; 282:136996. [PMID: 39486729 DOI: 10.1016/j.ijbiomac.2024.136996] [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: 08/12/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
The quasispecies of the influenza virus poses a significant challenge for developing effective therapies. Current antiviral drugs such as oseltamivir, zanamivir, peramivir and baloxavir marboxil along with seasonal vaccines have limitations due to viral variability caused by antigenic drift and shift as well as the development of drug resistance. Therefore, there is a clear need for novel antiviral agents targeting alternative mechanisms, either independently or in combination with existing modalities, to reduce the impact of influenza virus-related infections. The influenza nucleoprotein (NP) is a key component of the viral ribonucleoprotein complex. The multifaceted nature of the NP makes it an attractive target for antiviral intervention. Recent reports have identified inhibitors that specifically target this protein. Recognizing the importance of developing influenza treatments for potential pandemics, this review explores the structural and functional aspects of NP and highlights its potential as an emerging target for anti-influenza drugs. We discuss various strategies for targeting NP, including RNA binding, oligomerization, and nuclear export, and also consider the potential of NP-based vaccines. Overall, this review provides insights into recent developments and future perspectives on targeting influenza NP for antiviral therapies.
Collapse
Affiliation(s)
- Sankar Panthi
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Jhen-Yi Hong
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Roshan Satange
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Ching-Ching Yu
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Long-Yuan Li
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.
| | - Ming-Hon Hou
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan; Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan; Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
19
|
Keown JR, Carrique L, Nilsson-Payant BE, Fodor E, Grimes JM. Structural characterization of the full-length Hantaan virus polymerase. PLoS Pathog 2024; 20:e1012781. [PMID: 39652621 DOI: 10.1371/journal.ppat.1012781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 12/19/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
Hantaviridae are a family of segmented negative-sense RNA viruses that contain important human and animal pathogens. Hantaviridae contain a viral RNA-dependent RNA polymerase that replicates and transcribes the viral genome. Here we establish the expression and purification of the polymerase from the Old World Hantaan virus and characterise the structure using Cryo-EM. We determine a series of structures at resolutions between 2.7 and 3.3 Å of RNA free polymerase comprising the core, core and endonuclease, and a full-length polymerase. The full-length polymerase structure depicts the location of the cap binding and C-terminal domains which are arranged in a conformation that is incompatible with transcription and in a novel conformation not observed in previous conformations of cap-snatching viral polymerases. We further describe structures with 5' vRNA promoter in the presence and absence of a nucleotide triphosphate. The nucleotide bound structure mimics a replication pre-initiation complex and the nucleotide stabilises the motif E in a conformation distinct from those previously observed. We observe motif E in four distinct conformations including β-sheet, two helical arrangements, and nucleotide primed arrangement. The insights gained here guide future mechanistic studies of both the transcription and replication activities of the hantavirus polymerase and for the development of therapeutic targets.
Collapse
Affiliation(s)
- Jeremy R Keown
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Loïc Carrique
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Benjamin E Nilsson-Payant
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Cluster of Excellence RESIST (EXC2155), Hannover Medical School, Hannover, Germany
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ervin Fodor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Jonathan M Grimes
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Piesche R, Breithaupt A, Pohlmann A, Ahrens AK, Beer M, Harder T, Grund C. Dominant HPAIV H5N1 genotypes of Germany 2021/2022 are linked to high virulence in Pekin ducklings. NPJ VIRUSES 2024; 2:53. [PMID: 40295819 PMCID: PMC11721377 DOI: 10.1038/s44298-024-00062-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/25/2024] [Indexed: 04/30/2025]
Abstract
Highly pathogenic avian influenza viruses (HPAIV) of H5 clade 2.3.4.4b pose an ongoing threat worldwide. It remains unclear whether this panzootic situation would favor low virulent phenotypes expected by the 'avirulence hypothesis' of viral evolution. Assessing virulence in Pekin ducklings in an intramuscular infection model revealed that the two genotypes that dominated the epidemiological situation in Germany during the period 2021 and 2022 (EU-RL:CH and EU-RL:AB) were of high virulence. In contrast, rare genotypes were of intermediate virulence. The genetic constellation of these reassortants pointed to an important role of the viral polymerase complex (RdRP), particularly the PB1 genome segment, in shaping virulence in ducklings. Occulo-nasal infection of ducklings confirmed the phenotypes for two representative viruses and indicated a more efficient replication for the high virulence strain. These observations would be in line with the 'virulence-transmission trade-off' model for describing HPAIV epidemiology in wild birds in Germany.
Collapse
Affiliation(s)
- Ronja Piesche
- Friedrich- Loeffler- Institute, Institute of Diagnostic Virology, Greifswald, Germany
| | - Angele Breithaupt
- Friedrich- Loeffler- Institute, Department of Experimental Animal Facilities and Biorisk Management (ATB), Greifswald, Germany
| | - Anne Pohlmann
- Friedrich- Loeffler- Institute, Institute of Diagnostic Virology, Greifswald, Germany
| | - Ann Kathrin Ahrens
- Friedrich- Loeffler- Institute, Institute of Diagnostic Virology, Greifswald, Germany
| | - Martin Beer
- Friedrich- Loeffler- Institute, Institute of Diagnostic Virology, Greifswald, Germany.
| | - Timm Harder
- Friedrich- Loeffler- Institute, Institute of Diagnostic Virology, Greifswald, Germany
| | - Christian Grund
- Friedrich- Loeffler- Institute, Institute of Diagnostic Virology, Greifswald, Germany.
| |
Collapse
|
21
|
Bonomini A, Felicetti T, Pacetti M, Bertagnin C, Coletti A, Giammarino F, De Angelis M, Poggialini F, Macchiarulo A, Sabatini S, Mercorelli B, Nencioni L, Vicenti I, Dreassi E, Cecchetti V, Tabarrini O, Loregian A, Massari S. Optimization of potent, broad-spectrum, and specific anti-influenza compounds targeting RNA polymerase PA-PB1 heterodimerization. Eur J Med Chem 2024; 277:116737. [PMID: 39153334 DOI: 10.1016/j.ejmech.2024.116737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
Influenza viruses (IV) are single-stranded RNA viruses with a negative-sense genome and have the potential to cause pandemics. While vaccines exist for influenza, their protection is only partial. Additionally, there is only a limited number of approved anti-IV drugs, which are associated to emergence of drug resistance. To address these issues, for years we have focused on the development of small-molecules that can interfere with the heterodimerization of PA and PB1 subunits of the IV RNA-dependent RNA polymerase (RdRP). In this study, starting from a cycloheptathiophene-3-carboxamide compound that we recently identified, we performed iterative cycles of medicinal chemistry optimization that led to the identification of compounds 43 and 45 with activity in the nanomolar range against circulating A and B strains of IV. Mechanistic studies demonstrated the ability of 43 and 45 to interfere with viral RdRP activity by disrupting PA-PB1 subunits heterodimerization and to bind to the PA C-terminal domain through biophysical assays. Most important, ADME studies of 45 also showed an improvement in the pharmacokinetic profile with respect to the starting hit.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Martina Pacetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Alice Coletti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Federica Giammarino
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185, Rome, Italy
| | - Federica Poggialini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | | | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185, Rome, Italy
| | - Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy.
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy.
| |
Collapse
|
22
|
Sun J, Kuai L, Zhang L, Xie Y, Zhang Y, Li Y, Peng Q, Shao Y, Yang Q, Tian WX, Zhu J, Qi J, Shi Y, Deng T, Gao GF. NS2 induces an influenza A RNA polymerase hexamer and acts as a transcription to replication switch. EMBO Rep 2024; 25:4708-4727. [PMID: 39026012 PMCID: PMC11549089 DOI: 10.1038/s44319-024-00208-4] [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: 06/14/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
Genome transcription and replication of influenza A virus (FluA), catalyzed by viral RNA polymerase (FluAPol), are delicately controlled across the virus life cycle. A switch from transcription to replication occurring at later stage of an infection is critical for progeny virion production and viral non-structural protein NS2 has been implicated in regulating the switch. However, the underlying regulatory mechanisms and the structure of NS2 remained elusive for years. Here, we determine the cryo-EM structure of the FluAPol-NS2 complex at ~3.0 Å resolution. Surprisingly, three domain-swapped NS2 dimers arrange three symmetrical FluPol dimers into a highly ordered barrel-like hexamer. Further structural and functional analyses demonstrate that NS2 binding not only hampers the interaction between FluAPol and the Pol II CTD because of steric conflicts, but also impairs FluAPol transcriptase activity by stalling it in the replicase conformation. Moreover, this is the first visualization of the full-length NS2 structure. Our findings uncover key molecular mechanisms of the FluA transcription-replication switch and have implications for the development of antivirals.
Collapse
Affiliation(s)
- Junqing Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, 030032, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lu Kuai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lei Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, 518026, China
| | - Yufeng Xie
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yanfang Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yan Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Peng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuekun Shao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiuxian Yang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wen-Xia Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, China.
| | - Junhao Zhu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yi Shi
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, 030032, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- International Institute of Vaccine Research and Innovation (iVac), Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Tao Deng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - George F Gao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, China.
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, 030032, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- International Institute of Vaccine Research and Innovation (iVac), Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
23
|
Martinez-Sobrido L, Nogales A. Recombinant Influenza A Viruses Expressing Reporter Genes from the Viral NS Segment. Int J Mol Sci 2024; 25:10584. [PMID: 39408912 PMCID: PMC11476892 DOI: 10.3390/ijms251910584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Studying influenza A viruses (IAVs) requires secondary experimental procedures to detect the presence of the virus in infected cells or animals. The ability to generate recombinant (r)IAV using reverse genetics techniques has allowed investigators to generate viruses expressing foreign genes, including fluorescent and luciferase proteins. These rIAVs expressing reporter genes have allowed for easily tracking viral infections in cultured cells and animal models of infection without the need for secondary approaches, representing an excellent option to study different aspects in the biology of IAV where expression of reporter genes can be used as a readout of viral replication and spread. Likewise, these reporter-expressing rIAVs provide an excellent opportunity for the rapid identification and characterization of prophylactic and/or therapeutic approaches. To date, rIAV expressing reporter genes from different viral segments have been described in the literature. Among those, rIAV expressing reporter genes from the viral NS segment have been shown to represent an excellent option to track IAV infection in vitro and in vivo, eliminating the need for secondary approaches to identify the presence of the virus. Here, we summarize the status on rIAV expressing traceable reporter genes from the viral NS segment and their applications for in vitro and in vivo influenza research.
Collapse
Affiliation(s)
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, 28130 Madrid, Spain
| |
Collapse
|
24
|
Zhang Z, Uribe I, Davis KA, McPherson RL, Larson GP, Badiee M, Tran V, Ledwith MP, Feltman E, Yú S, Caì Y, Chang CY, Yang X, Ma Z, Chang P, Kuhn JH, Leung AKL, Mehle A. Global remodeling of ADP-ribosylation by PARP1 suppresses influenza A virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613696. [PMID: 39345583 PMCID: PMC11430048 DOI: 10.1101/2024.09.19.613696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
ADP-ribosylation is a highly dynamic and fully reversible post-translational modification performed by poly(ADP-ribose) polymerases (PARPs) that modulates protein function, abundance, localization and turnover. Here we show that influenza A virus infection causes a rapid and dramatic upregulation of global ADP-ribosylation that inhibits viral replication. Mass spectrometry defined for the first time the global ADP-ribosylome during infection, creating an infection-specific profile with almost 4,300 modification sites on ~1,080 host proteins, as well as over 100 modification sites on viral proteins. Our data indicate that the global increase likely reflects a change in the form of ADP-ribosylation rather than modification of new targets. Functional assays demonstrated that modification of the viral replication machinery antagonizes its activity and further revealed that the anti-viral activity of PARPs and ADP-ribosylation is counteracted by the influenza A virus protein NS1, assigning a new activity to the primary viral antagonist of innate immunity. We identified PARP1 as the enzyme producing the majority of poly(ADP-ribose) present during infection. Influenza A virus replicated faster in cells lacking PARP1, linking PARP1 and ADP-ribosylation to the anti-viral phenotype. Together, these data establish ADP-ribosylation as an anti-viral innate immune-like response to viral infection antagonized by a previously unknown activity of NS1.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, WI
| | - Isabel Uribe
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kaitlin A. Davis
- Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, WI
| | - Robert Lyle McPherson
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Gloria P Larson
- Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, WI
| | - Mohsen Badiee
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Vy Tran
- Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, WI
| | - Mitchell P. Ledwith
- Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, WI
| | - Elizabeth Feltman
- Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, WI
| | - Shuǐqìng Yú
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Yíngyún Caì
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Che-Yuan Chang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xingyi Yang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Zhuo Ma
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Paul Chang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Anthony K. L. Leung
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Molecular Biology and Genetics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Mehle
- Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, WI
- Lead Contact
| |
Collapse
|
25
|
Arragain B, Krischuns T, Pelosse M, Drncova P, Blackledge M, Naffakh N, Cusack S. Structures of influenza A and B replication complexes give insight into avian to human host adaptation and reveal a role of ANP32 as an electrostatic chaperone for the apo-polymerase. Nat Commun 2024; 15:6910. [PMID: 39160148 PMCID: PMC11333492 DOI: 10.1038/s41467-024-51007-3] [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: 02/08/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024] Open
Abstract
Replication of influenza viral RNA depends on at least two viral polymerases, a parental replicase and an encapsidase, and cellular factor ANP32. ANP32 comprises an LRR domain and a long C-terminal low complexity acidic region (LCAR). Here we present evidence suggesting that ANP32 is recruited to the replication complex as an electrostatic chaperone that stabilises the encapsidase moiety within apo-polymerase symmetric dimers that are distinct for influenza A and B polymerases. The ANP32 bound encapsidase, then forms the asymmetric replication complex with the replicase, which is embedded in a parental ribonucleoprotein particle (RNP). Cryo-EM structures reveal the architecture of the influenza A and B replication complexes and the likely trajectory of the nascent RNA product into the encapsidase. The cryo-EM map of the FluB replication complex shows extra density attributable to the ANP32 LCAR wrapping around and stabilising the apo-encapsidase conformation. These structures give new insight into the various mutations that adapt avian strain polymerases to use the distinct ANP32 in mammalian cells.
Collapse
Affiliation(s)
- Benoît Arragain
- European Molecular Biology Laboratory, Grenoble, Cedex 9, France
| | - Tim Krischuns
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, RNA Biology of Influenza Virus, Paris, France
- Heidelberg University, Department of Infectious Diseases, Virology, Schaller Research Group, Heidelberg, Germany
| | - Martin Pelosse
- European Molecular Biology Laboratory, Grenoble, Cedex 9, France
| | - Petra Drncova
- European Molecular Biology Laboratory, Grenoble, Cedex 9, France
| | - Martin Blackledge
- Institut de Biologie Structurale, Université Grenoble-Alpes-CEA-CNRS UMR5075, Grenoble, France
| | - Nadia Naffakh
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, RNA Biology of Influenza Virus, Paris, France
| | - Stephen Cusack
- European Molecular Biology Laboratory, Grenoble, Cedex 9, France.
| |
Collapse
|
26
|
Hover S, Barr JN, Fontana J. Insights into the structure of RNPs from segmented negative-sense RNA viruses. Structure 2024; 32:1027-1028. [PMID: 39121836 DOI: 10.1016/j.str.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 08/12/2024]
Abstract
The genome of segmented negative-sense single-stranded RNA viruses, such as influenza virus and bunyaviruses, is coated by viral nucleoproteins (NPs), forming a ribonucleoprotein (RNP). In this issue of Structure, Dick et al.1 expand our knowledge on the RNPs of these viruses by solving the structures of Thogoto virus NP and RNP.
Collapse
Affiliation(s)
- Samantha Hover
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - John N Barr
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Juan Fontana
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
27
|
Xiao R, Jin H, Huang F, Huang B, Wang H, Wang YG. Oncolytic virotherapy for hepatocellular carcinoma: A potent immunotherapeutic landscape. World J Gastrointest Oncol 2024; 16:2867-2876. [PMID: 39072175 PMCID: PMC11271782 DOI: 10.4251/wjgo.v16.i7.2867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/24/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a systemic disease with augmented malignant degree, high mortality and poor prognosis. Since the establishment of the immune mechanism of tumor therapy, people have realized that immunotherapy is an effective means for improvement of HCC patient prognosis. Oncolytic virus is a novel immunotherapy drug, which kills tumor cells and exempts normal cells by directly lysing tumor and inducing anti-tumor immune response, and it has been extensively examined as an HCC therapy. This editorial discusses oncolytic viruses for the treatment of HCC, emphasizing viral immunotherapy strategies and clinical applications related to HCC.
Collapse
Affiliation(s)
- Rong Xiao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Hao Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Fang Huang
- Department of Pathology, Laboratory Medicine Center, Zhejiang Provincial Peoples’ Hospital, Peoples’ Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Hui Wang
- Department of Oncology, Zhejiang Xiaoshan Hospital, Hangzhou 310018, Zhejiang Province, China
| | - Yi-Gang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
28
|
Peng B, Wang Y, Xie Y, Dong X, Liu W, Li D, Chen H. An overview of influenza A virus detection methods: from state-of-the-art of laboratories to point-of-care strategies. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:4496-4515. [PMID: 38946516 DOI: 10.1039/d4ay00508b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Influenza A virus (IAV), a common respiratory infectious pathogen, poses a significant risk to personal health and public health safety due to rapid mutation and wide host range. To better prevent and treat IAV, comprehensive measures are needed for early and rapid screening and detection of IAV. Although traditional laboratory-based techniques are accurate, they are often time-consuming and not always feasible in emergency or resource-limited areas. In contrast, emerging point-of-care strategies provide faster results but may compromise sensitivity and specificity. Here, this review critically evaluates various detection methods for IAV from established laboratory-based procedures to innovative rapid diagnosis. By analyzing the recent research progress, we aim to address significant gaps in understanding the effectiveness, practicality, and applicability of these methods in different scenarios, which could provide information for healthcare strategies, guide public health response measures, and ultimately strengthen patient care in the face of the ongoing threat of IAV. Through a detailed comparison of diagnostic models, this review can provide a reliable reference for rapid, accurate and efficient detection of IAV, and to contribute to the diagnosis, treatment, prevention, and control of IAV.
Collapse
Affiliation(s)
- Bin Peng
- Guangzhou Huashang Vocational College, Guangzhou, 510000, China
| | - Yaqi Wang
- Guangzhou Institute for Food Inspection, Guangzhou, 510000, China
| | - Yueliang Xie
- Guangdong Agriculture Industry Business Polytechnic College, Guangzhou, 510000, China
| | - Xiangyan Dong
- State Key Laboratory of Chemical Oncogenomics, Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
| | - Wen Liu
- Guangdong Agriculture Industry Business Polytechnic College, Guangzhou, 510000, China
| | - Dan Li
- College of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, China
| | - Hui Chen
- State Key Laboratory of Chemical Oncogenomics, Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
| |
Collapse
|
29
|
Wu W, Lan W, Jiao X, Shao A, Wu P, Wang K, Zhan S. Mechanisms underlying the therapeutic effects of Gang Huo Qing wen granules in the treatment of influenza based on network pharmacology, molecular docking and molecular dynamics. Sci Rep 2024; 14:15853. [PMID: 38982082 PMCID: PMC11233559 DOI: 10.1038/s41598-024-62469-2] [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: 09/22/2023] [Accepted: 05/17/2024] [Indexed: 07/11/2024] Open
Abstract
Influenza (Flu) is a severe health, medical, and economic problem, but no medication that has excellent outcomes and lowers the occurrence of these problems is now available. GanghuoQingwenGranules (GHQWG) is a common Chinese herbal formula for the treatment of influenza (flu). However, its methods of action remain unknown. We used network pharmacology, molecular docking, and molecular dynamics simulation techniques to investigate the pharmacological mechanism of GHQWG in flu. TCMSP and various types of literature were used to obtain active molecules and targets of GHQWG. Flu-related targets were found in the Online Mendelian Inheritance in Man (OMIM) database, the DisFeNET database, the Therapeutic Target Database (TTD), and the DrugBank database. To screen the key targets, a protein-protein interaction (PPI) network was constructed. DAVID was used to analyze GO and KEGG pathway enrichment. Target tissue and organ distribution was assessed. Molecular docking was used to evaluate interactions between possible targets and active molecules. For the ideal core protein-compound complexes obtained using molecular docking, a molecular dynamics simulation was performed. In total, 90 active molecules and 312 GHQWG targets were discovered. The PPI network's topology highlighted six key targets. GHQWG's effects are mediated via genes involved in inflammation, apoptosis, and oxidative stress, as well as the TNF and IL-17 signaling pathways, according to GO and KEGG pathway enrichment analysis. Molecular docking and molecular dynamics simulations demonstrated that the active compounds and tested targets had strong binding capabilities. This analysis accurately predicts the effective components, possible targets, and pathways involved in GHQWG flu treatment. We proposed a novel study strategy for future studies on the molecular processes of GHQWG in flu treatment. Furthermore, the possible active components provide a dependable source for flu drug screening.
Collapse
Affiliation(s)
- Wenyu Wu
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wanning Lan
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Jiao
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Axue Shao
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Wu
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Kai Wang
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Shaofeng Zhan
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
30
|
Alshiban NM, Aleyiydi MS, Nassar MS, Alhumaid NK, Almangour TA, Tawfik YM, Damiati LA, Almutairi AS, Tawfik EA. Epidemiologic and clinical updates on viral infections in Saudi Arabia. Saudi Pharm J 2024; 32:102126. [PMID: 38966679 PMCID: PMC11223122 DOI: 10.1016/j.jsps.2024.102126] [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] [Indexed: 07/06/2024] Open
Abstract
In the past two decades, the world has witnessed devastating pandemics affecting the global healthcare infrastructure and disrupting society and the economy worldwide. Among all pathogens, viruses play a critical role that is associated with outbreaks due to their wide range of species, involvement of animal hosts, easily transmitted to humans, and increased rates of infectivity. Viral disease outbreaks threaten public health globally due to the challenges associated with controlling and eradicating them. Implementing effective viral disease control programs starts with ongoing surveillance data collection and analyses to detect infectious disease trends and patterns, which is critical for maintaining public health. Viral disease control strategies include improved hygiene and sanitation facilities, eliminating arthropod vectors, vaccinations, and quarantine. The Saudi Ministry of Health (MOH) and the Public Health Authority (also known as Weqayah) in Saudi Arabia are responsible for public health surveillance to control and prevent infectious diseases. The notifiable viral diseases based on the Saudi MOH include hepatitis diseases, viral hemorrhagic fevers, respiratory viral diseases, exanthematous viral diseases, neurological viral diseases, and conjunctivitis. Monitoring trends and detecting changes in these viral diseases is essential to provide proper interventions, evaluate the established prevention programs, and develop better prevention strategies. Therefore, this review aims to highlight the epidemiological updates of the recently reported viral infections in Saudi Arabia and to provide insights into the recent clinical treatment and prevention strategies.
Collapse
Affiliation(s)
- Noura M. Alshiban
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - Munirah S. Aleyiydi
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - Majed S. Nassar
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - Nada K. Alhumaid
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - Thamer A. Almangour
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yahya M.K. Tawfik
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Laila A. Damiati
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah 23218, Saudi Arabia
| | | | - Essam A. Tawfik
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| |
Collapse
|
31
|
Urmi UL, Vijay AK, Willcox MDP, Attard S, Enninful G, Kumar N, Islam S, Kuppusamy R. Exploring the Efficacy of Peptides and Mimics against Influenza A Virus, Adenovirus, and Murine Norovirus. Int J Mol Sci 2024; 25:7030. [PMID: 39000138 PMCID: PMC11240954 DOI: 10.3390/ijms25137030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The ongoing battle against viral pandemics continues, with the possibility of future outbreaks. The search for effective antiviral compounds that can combat a diverse range of viruses continues to be a focal point of research. This study investigated the efficacy of two natural antimicrobial peptides (AMPs) (lactoferricin and LL-37), two synthetic AMPs (melimine and Mel4), and nine AMP mimics (758, 1091, 1096, 1083, 610, NAPL, 3-BIPL, 4-BIPL, and Sau-22) against influenza A virus strains H1N1 and H3N2, human adenovirus 5 (HAdV-5), and murine norovirus 1 (MNV-1). These compounds were tested using virus pre-treatment, cell pre-treatment, or post-cell entry treatment assays, electron microscopy, and circular dichroism (CD), alongside evaluations of cytotoxicity against the host cells. After virus pre-treatment, the AMP mimics 610 and Sau-22 had relatively low IC50 values for influenza strains H1N1 (2.35 and 6.93 µM, respectively) and H3N2 (3.7 and 5.34 µM, respectively). Conversely, natural and synthetic AMPs were not active against these strains. For the non-enveloped viruses, the AMP Mel4 and mimic 1083 had moderate activity against HAdV-5 (Mel4 IC50 = 47.4 µM; 1083 IC50 = 47.2 µM), whereas all AMPs, but none of the mimics, were active against norovirus (LL-37 IC50 = 4.2 µM; lactoferricin IC50 = 23.18 µM; melimine IC50 = 4.8 µM; Mel4 IC50 = 8.6 µM). Transmission electron microscopy demonstrated that the mimics targeted the outer envelope of influenza viruses, while the AMPs targeted the capsid of non-enveloped viruses. CD showed that Mel4 adopted an α-helical structure in a membrane mimetic environment, but mimic 758 remained unstructured. The diverse activity against different virus groups is probably influenced by charge, hydrophobicity, size, and, in the case of natural and synthetic AMPs, their secondary structure. These findings underscore the potential of peptides and mimics as promising candidates for antiviral therapeutics against both enveloped and non-enveloped viruses.
Collapse
Affiliation(s)
- Umme Laila Urmi
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ajay Kumar Vijay
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mark D P Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Samuel Attard
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - George Enninful
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Salequl Islam
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh
| | - Rajesh Kuppusamy
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
- The Drug Discovery Initiative, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
32
|
Hagan MF, Zandi R, Uetrecht C. Overview of the 2023 Physical Virology Gordon Research Conference-Viruses at Multiple Levels of Complexity. Viruses 2024; 16:895. [PMID: 38932189 PMCID: PMC11209351 DOI: 10.3390/v16060895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
This review accompanies the Special Issue on the subject of physical virology, which features work presented at the recent Gordon Research Conference (GRC) on this topic [...].
Collapse
Affiliation(s)
- Michael F. Hagan
- Martin Fisher School of Physics, Brandeis University, Waltham, MA 02453, USA
| | - Roya Zandi
- Department of Physics and Astronomy, University of California Riverside, Riverside, CA 92521, USA
| | - Charlotte Uetrecht
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen Synchrotron DESY, Leibniz Institute of Virology (LIV), 22607 Hamburg, Germany
- Institute of Chemistry and Metabolomics, University Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
33
|
Teo QW, Wang Y, Lv H, Mao KJ, Tan TJC, Huan YW, Rivera-Cardona J, Shao EK, Choi D, Dargani ZT, Brooke CB, Wu NC. Deep mutational scanning of influenza A virus NEP reveals pleiotropic mutations in its N-terminal domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594574. [PMID: 38798526 PMCID: PMC11118461 DOI: 10.1101/2024.05.16.594574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The influenza A virus nuclear export protein (NEP) is a multifunctional protein that is essential for the viral life cycle and has very high sequence conservation. However, since the open reading frame of NEP largely overlaps with that of another influenza viral protein, non-structural protein 1, it is difficult to infer the functional constraints of NEP based on sequence conservation analysis. Besides, the N-terminal of NEP is structurally disordered, which further complicates the understanding of its function. Here, we systematically measured the replication fitness effects of >1,800 mutations of NEP. Our results show that the N-terminal domain has high mutational tolerance. Additional experiments demonstrate that N-terminal domain mutations pleiotropically affect viral transcription and replication dynamics, host cellular responses, and mammalian adaptation of avian influenza virus. Overall, our study not only advances the functional understanding of NEP, but also provides insights into its evolutionary constraints.
Collapse
|
34
|
Staller E, Carrique L, Swann OC, Fan H, Keown JR, Sheppard CM, Barclay WS, Grimes JM, Fodor E. Structures of H5N1 influenza polymerase with ANP32B reveal mechanisms of genome replication and host adaptation. Nat Commun 2024; 15:4123. [PMID: 38750014 PMCID: PMC11096171 DOI: 10.1038/s41467-024-48470-3] [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: 01/05/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Avian influenza A viruses (IAVs) pose a public health threat, as they are capable of triggering pandemics by crossing species barriers. Replication of avian IAVs in mammalian cells is hindered by species-specific variation in acidic nuclear phosphoprotein 32 (ANP32) proteins, which are essential for viral RNA genome replication. Adaptive mutations enable the IAV RNA polymerase (FluPolA) to surmount this barrier. Here, we present cryo-electron microscopy structures of monomeric and dimeric avian H5N1 FluPolA with human ANP32B. ANP32B interacts with the PA subunit of FluPolA in the monomeric form, at the site used for its docking onto the C-terminal domain of host RNA polymerase II during viral transcription. ANP32B acts as a chaperone, guiding FluPolA towards a ribonucleoprotein-associated FluPolA to form an asymmetric dimer-the replication platform for the viral genome. These findings offer insights into the molecular mechanisms governing IAV genome replication, while enhancing our understanding of the molecular processes underpinning mammalian adaptations in avian-origin FluPolA.
Collapse
Affiliation(s)
- Ecco Staller
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Loïc Carrique
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Olivia C Swann
- Section of Molecular Virology, Imperial College London, London, UK
| | - Haitian Fan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- School of Basic Medical Sciences, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jeremy R Keown
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, UK
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Carol M Sheppard
- Section of Molecular Virology, Imperial College London, London, UK
| | - Wendy S Barclay
- Section of Molecular Virology, Imperial College London, London, UK
| | - Jonathan M Grimes
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Ervin Fodor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
Keown J, Baazaoui A, Šebesta M, Štefl R, Carrique L, Fodor E, Grimes JM. Structural and functional characterization of the interaction between the influenza A virus RNA polymerase and the CTD of host RNA polymerase II. J Virol 2024; 98:e0013824. [PMID: 38563748 PMCID: PMC11092357 DOI: 10.1128/jvi.00138-24] [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: 01/19/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
Influenza A viruses, causing seasonal epidemics and occasional pandemics, rely on interactions with host proteins for their RNA genome transcription and replication. The viral RNA polymerase utilizes host RNA polymerase II (Pol II) and interacts with the serine 5 phosphorylated (pS5) C-terminal domain (CTD) of Pol II to initiate transcription. Our study, using single-particle electron cryomicroscopy (cryo-EM), reveals the structure of the 1918 pandemic influenza A virus polymerase bound to a synthetic pS5 CTD peptide composed of four heptad repeats mimicking the 52 heptad repeat mammalian Pol II CTD. The structure shows that the CTD peptide binds at the C-terminal domain of the PA viral polymerase subunit (PA-C) and reveals a previously unobserved position of the 627 domain of the PB2 subunit near the CTD. We identify crucial residues of the CTD peptide that mediate interactions with positively charged cavities on PA-C, explaining the preference of the viral polymerase for pS5 CTD. Functional analysis of mutants targeting the CTD-binding site within PA-C reveals reduced transcriptional function or defects in replication, highlighting the multifunctional role of PA-C in viral RNA synthesis. Our study provides insights into the structural and functional aspects of the influenza virus polymerase-host Pol II interaction and identifies a target for antiviral development.IMPORTANCEUnderstanding the intricate interactions between influenza A viruses and host proteins is crucial for developing targeted antiviral strategies. This study employs advanced imaging techniques to uncover the structural nuances of the 1918 pandemic influenza A virus polymerase bound to a specific host protein, shedding light on the vital process of viral RNA synthesis. The study identifies key amino acid residues in the influenza polymerase involved in binding host polymerase II (Pol II) and highlights their role in both viral transcription and genome replication. These findings not only deepen our understanding of the influenza virus life cycle but also pinpoint a potential target for antiviral development. By elucidating the structural and functional aspects of the influenza virus polymerase-host Pol II interaction, this research provides a foundation for designing interventions to disrupt viral replication and transcription, offering promising avenues for future antiviral therapies.
Collapse
Affiliation(s)
- Jeremy Keown
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Alaa Baazaoui
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Marek Šebesta
- CEITEC–Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Richard Štefl
- CEITEC–Central European Institute of Technology, Masaryk University, Brno, Czechia
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czechia
| | - Loïc Carrique
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ervin Fodor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Jonathan M. Grimes
- Division of Structural Biology, Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
36
|
Sheen K, Myung S, Lee DM, Yu S, Choi Y, Kim T, Kim J, Ji SG, Kim MS, Kim W, Lee Y, Kim MS, Park YC. RNA-Seq of an LPS-Induced Inflammation Model Reveals Transcriptional Profile Patterns of Inflammatory Processes. Life (Basel) 2024; 14:558. [PMID: 38792580 PMCID: PMC11121855 DOI: 10.3390/life14050558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/10/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
The LPS-induced inflammation model is widely used for studying inflammatory processes due to its cost-effectiveness, reproducibility, and faithful representation of key hallmarks. While researchers often validate this model using clinical cytokine markers, a comprehensive understanding of gene regulatory mechanisms requires extending investigation beyond these hallmarks. Our study leveraged multiple whole-blood bulk RNA-seq datasets to rigorously compare the transcriptional profiles of the well-established LPS-induced inflammation model with those of several human diseases characterized by systemic inflammation. Beyond conventional inflammation-associated systems, we explored additional systems indirectly associated with inflammatory responses (i.e., ISR, RAAS, and UPR) using a customized core inflammatory gene list. Our cross-condition-validation approach spanned four distinct conditions: systemic lupus erythematosus (SLE) patients, dengue infection, candidemia infection, and staphylococcus aureus exposure. This analysis approach, utilizing the core gene list aimed to assess the model's suitability for understanding the gene regulatory mechanisms underlying inflammatory processes triggered by diverse factors. Our analysis resulted in elevated expressions of innate immune-associated genes, coinciding with suppressed expressions of adaptive immune-associated genes. Also, upregulation of genes associated with cellular stresses and mitochondrial innate immune responses underscored oxidative stress as a central driver of the corresponding inflammatory processes in both the LPS-induced and other inflammatory contexts.
Collapse
Affiliation(s)
- Kisung Sheen
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Seokho Myung
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Medicine, Kyung Hee University College of Medicine, Seoul 02453, Republic of Korea
| | - Dong-Min Lee
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Acupuncture & Moxibustion, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Sanghyeon Yu
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yueun Choi
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Taeyoon Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Jihan Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Medicine, Kyung Hee University College of Medicine, Seoul 02453, Republic of Korea
| | - Sang-Gu Ji
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Myung-Seo Kim
- Department of Orthopaedic Surgery, Shoulder & Elbow Clinic, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| | - Wonnam Kim
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Yoonsung Lee
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
| | - Man S. Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea; (K.S.); (S.M.); (D.-M.L.); (S.Y.); (Y.C.); (T.K.); (J.K.); (S.-G.J.); (Y.L.)
| | - Yeon-Cheol Park
- Department of Acupuncture & Moxibustion, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| |
Collapse
|
37
|
Quignon E, Ferhadian D, Hache A, Vivet-Boudou V, Isel C, Printz-Schweigert A, Donchet A, Crépin T, Marquet R. Structural Impact of the Interaction of the Influenza A Virus Nucleoprotein with Genomic RNA Segments. Viruses 2024; 16:421. [PMID: 38543786 PMCID: PMC10974462 DOI: 10.3390/v16030421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
Influenza A viruses (IAVs) possess a segmented genome consisting of eight viral RNAs (vRNAs) associated with multiple copies of viral nucleoprotein (NP) and a viral polymerase complex. Despite the crucial role of RNA structure in IAV replication, the impact of NP binding on vRNA structure is not well understood. In this study, we employed SHAPE chemical probing to compare the structure of NS and M vRNAs of WSN IAV in various states: before the addition of NP, in complex with NP, and after the removal of NP. Comparison of the RNA structures before the addition of NP and after its removal reveals that NP, while introducing limited changes, remodels local structures in both vRNAs and long-range interactions in the NS vRNA, suggesting a potentially biologically relevant RNA chaperone activity. In contrast, NP significantly alters the structure of vRNAs in vRNA/NP complexes, though incorporating experimental data into RNA secondary structure prediction proved challenging. Finally, our results suggest that NP not only binds single-stranded RNA but also helices with interruptions, such as bulges or small internal loops, with a preference for G-poor and C/U-rich regions.
Collapse
Affiliation(s)
- Erwan Quignon
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, 67000 Strasbourg, France; (E.Q.); (A.H.); (V.V.-B.); (C.I.)
| | - Damien Ferhadian
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, 67000 Strasbourg, France; (E.Q.); (A.H.); (V.V.-B.); (C.I.)
| | - Antoine Hache
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, 67000 Strasbourg, France; (E.Q.); (A.H.); (V.V.-B.); (C.I.)
| | - Valérie Vivet-Boudou
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, 67000 Strasbourg, France; (E.Q.); (A.H.); (V.V.-B.); (C.I.)
| | - Catherine Isel
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, 67000 Strasbourg, France; (E.Q.); (A.H.); (V.V.-B.); (C.I.)
| | - Anne Printz-Schweigert
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, 67000 Strasbourg, France; (E.Q.); (A.H.); (V.V.-B.); (C.I.)
| | - Amélie Donchet
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France (T.C.)
| | - Thibaut Crépin
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France (T.C.)
| | - Roland Marquet
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, 67000 Strasbourg, France; (E.Q.); (A.H.); (V.V.-B.); (C.I.)
| |
Collapse
|
38
|
Carter T, Iqbal M. The Influenza A Virus Replication Cycle: A Comprehensive Review. Viruses 2024; 16:316. [PMID: 38400091 PMCID: PMC10892522 DOI: 10.3390/v16020316] [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: 01/18/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Influenza A virus (IAV) is the primary causative agent of influenza, colloquially called the flu. Each year, it infects up to a billion people, resulting in hundreds of thousands of human deaths, and causes devastating avian outbreaks with worldwide losses worth billions of dollars. Always present is the possibility that a highly pathogenic novel subtype capable of direct human-to-human transmission will spill over into humans, causing a pandemic as devastating if not more so than the 1918 influenza pandemic. While antiviral drugs for influenza do exist, they target very few aspects of IAV replication and risk becoming obsolete due to antiviral resistance. Antivirals targeting other areas of IAV replication are needed to overcome this resistance and combat the yearly epidemics, which exact a serious toll worldwide. This review aims to summarise the key steps in the IAV replication cycle, along with highlighting areas of research that need more focus.
Collapse
Affiliation(s)
- Toby Carter
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK;
| | | |
Collapse
|
39
|
Krischuns T, Arragain B, Isel C, Paisant S, Budt M, Wolff T, Cusack S, Naffakh N. The host RNA polymerase II C-terminal domain is the anchor for replication of the influenza virus genome. Nat Commun 2024; 15:1064. [PMID: 38316757 PMCID: PMC10844641 DOI: 10.1038/s41467-024-45205-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/16/2024] [Indexed: 02/07/2024] Open
Abstract
The current model is that the influenza virus polymerase (FluPol) binds either to host RNA polymerase II (RNAP II) or to the acidic nuclear phosphoprotein 32 (ANP32), which drives its conformation and activity towards transcription or replication of the viral genome, respectively. Here, we provide evidence that the FluPol-RNAP II binding interface, beyond its well-acknowledged function in cap-snatching during transcription initiation, has also a pivotal role in replication of the viral genome. Using a combination of cell-based and in vitro approaches, we show that the RNAP II C-terminal-domain, jointly with ANP32, enhances FluPol replication activity. We observe successive conformational changes to switch from a transcriptase to a replicase conformation in the presence of the bound RNPAII C-terminal domain and propose a model in which the host RNAP II is the anchor for transcription and replication of the viral genome. Our data open new perspectives on the spatial coupling of viral transcription and replication and the coordinated balance between these two activities.
Collapse
Affiliation(s)
- Tim Krischuns
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, RNA Biology of Influenza Virus, Paris, France.
| | | | - Catherine Isel
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, RNA Biology of Influenza Virus, Paris, France
| | - Sylvain Paisant
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, RNA Biology of Influenza Virus, Paris, France
| | - Matthias Budt
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
| | - Thorsten Wolff
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
| | - Stephen Cusack
- European Molecular Biology Laboratory, Grenoble, France.
| | - Nadia Naffakh
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, RNA Biology of Influenza Virus, Paris, France.
| |
Collapse
|
40
|
Zhang L, Shao Y, Wang Y, Yang Q, Guo J, Gao GF, Deng T. Twenty natural amino acid substitution screening at the last residue 121 of influenza A virus NS2 protein reveals the critical role of NS2 in promoting virus genome replication by coordinating with viral polymerase. J Virol 2024; 98:e0116623. [PMID: 38054704 PMCID: PMC10804943 DOI: 10.1128/jvi.01166-23] [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: 08/02/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023] Open
Abstract
Both influenza A virus genome transcription (vRNA→mRNA) and replication (vRNA→cRNA→vRNA), catalyzed by the influenza RNA polymerase (FluPol), are dynamically regulated across the virus life cycle. It has been reported that the last amino acid I121 of the viral NS2 protein plays a critical role in promoting viral genome replication in influenza mini-replicon systems. Here, we performed a 20 natural amino acid substitution screening at residue NS2-I121 in the context of virus infection. We found that the hydrophobicity of the residue 121 is essential for virus survival. Interestingly, through serial passage of the rescued mutant viruses, we further identified adaptive mutations PA-K19E and PB1-S713N on FluPol which could effectively compensate for the replication-promoting defect caused by NS2-I121 mutation in the both mini-replicon and virus infection systems. Structural analysis of different functional states of FluPol indicates that PA-K19E and PB1-S713N could stabilize the replicase conformation of FluPol. By using a cell-based NanoBiT complementary reporter assay, we further demonstrate that both wild-type NS2 and PA-K19E/PB1-S713N could enhance FluPol dimerization, which is necessary for genome replication. These results reveal the critical role NS2 plays in promoting viral genome replication by coordinating with FluPol.IMPORTANCEThe intrinsic mechanisms of influenza RNA polymerase (FluPol) in catalyzing viral genome transcription and replication have been largely resolved. However, the mechanisms of how transcription and replication are dynamically regulated remain elusive. We recently reported that the last amino acid of the viral NS2 protein plays a critical role in promoting viral genome replication in an influenza mini-replicon system. Here, we conducted a 20 amino acid substitution screening at the last residue 121 in virus rescue and serial passage. Our results demonstrate that the replication-promoting function of NS2 is important for virus survival and efficient multiplication. We further show evidence that NS2 and NS2-I121 adaptive mutations PA-K19E/PB1-S713N regulate virus genome replication by promoting FluPol dimerization. This work highlights the coordination between NS2 and FluPol in fulfilling efficient genome replication. It further advances our understanding of the regulation of viral RNA synthesis for influenza A virus.
Collapse
Affiliation(s)
- Lei Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- />Institute of Pediatrics, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yuekun Shao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yingying Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Qiuxian Yang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiamei Guo
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - George F. Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tao Deng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
41
|
Li X, Ye Z, Plant EP. 5' copyback defective viral genomes are major component in clinical and non-clinical influenza samples. Virus Res 2024; 339:199274. [PMID: 37981214 PMCID: PMC10701078 DOI: 10.1016/j.virusres.2023.199274] [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: 09/11/2023] [Revised: 11/02/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
Clinical samples from people with influenza disease have been analyzed to assess the presence and abundance of Defective Viral Genomes (DVGs), but these have not been assessed using the same bioinformatic pipeline. The type of DVG most described for influenza infections (deletion DVGs) differs from the most commonly described DVGs from non-segmented negative stranded viruses (5' copyback). This could be attributed to either differences between viruses or the tools used to detect and characterize DVGs. Here we analyze several NGS datasets from people infected with different types of influenza virus using the same bioinformatic pipeline. We observe that 5' copyback DVGs are prevalent in all human clinical samples but not in the cultured samples. To address this discrepancy between clinical and laboratory cultures, we infected cell culture and ferrets with an H5N8 influenza A virus (FLUAV) and analyzed the DVG composition. The results demonstrate that the DVG population is skewed toward 5' copyback DVGs in the in vivo infections and deletion DVGs in the in vitro infections. This demonstrates that there are differences in vivo genome production and in vitro genome production, and this has implications for how the role of DVGs in clinical disease is studied. We also investigate the role the host cofactor ANP32B has in DVG production.
Collapse
Affiliation(s)
- Xing Li
- Laboratory of Pediatric and Respiratory Viral Disease, Office of Vaccine Research and Review, CBER, FDA, Silver Spring, MD, USA
| | - Zhiping Ye
- Laboratory of Pediatric and Respiratory Viral Disease, Office of Vaccine Research and Review, CBER, FDA, Silver Spring, MD, USA
| | - Ewan P Plant
- Laboratory of Pediatric and Respiratory Viral Disease, Office of Vaccine Research and Review, CBER, FDA, Silver Spring, MD, USA.
| |
Collapse
|
42
|
Sabsay KR, te Velthuis AJW. Negative and ambisense RNA virus ribonucleocapsids: more than protective armor. Microbiol Mol Biol Rev 2023; 87:e0008223. [PMID: 37750733 PMCID: PMC10732063 DOI: 10.1128/mmbr.00082-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
SUMMARYNegative and ambisense RNA viruses are the causative agents of important human diseases such as influenza, measles, Lassa fever, and Ebola hemorrhagic fever. The viral genome of these RNA viruses consists of one or more single-stranded RNA molecules that are encapsidated by viral nucleocapsid proteins to form a ribonucleoprotein complex (RNP). This RNP acts as protection, as a scaffold for RNA folding, and as the context for viral replication and transcription by a viral RNA polymerase. However, the roles of the viral nucleoproteins extend beyond these functions during the viral infection cycle. Recent advances in structural biology techniques and analysis methods have provided new insights into the formation, function, dynamics, and evolution of negative sense virus nucleocapsid proteins, as well as the role that they play in host innate immune responses against viral infection. In this review, we discuss the various roles of nucleocapsid proteins, both in the context of RNPs and in RNA-free states, as well as the open questions that remain.
Collapse
Affiliation(s)
- Kimberly R. Sabsay
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Aartjan J. W. te Velthuis
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
43
|
Liu C, Hu L, Dong G, Zhang Y, Ferreira da Silva-Júnior E, Liu X, Menéndez-Arias L, Zhan P. Emerging drug design strategies in anti-influenza drug discovery. Acta Pharm Sin B 2023; 13:4715-4732. [PMID: 38045039 PMCID: PMC10692392 DOI: 10.1016/j.apsb.2023.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/12/2023] [Accepted: 08/03/2023] [Indexed: 12/05/2023] Open
Abstract
Influenza is an acute respiratory infection caused by influenza viruses (IFV), According to the World Health Organization (WHO), seasonal IFV epidemics result in approximately 3-5 million cases of severe illness, leading to about half a million deaths worldwide, along with severe economic losses and social burdens. Unfortunately, frequent mutations in IFV lead to a certain lag in vaccine development as well as resistance to existing antiviral drugs. Therefore, it is of great importance to develop anti-IFV drugs with high efficiency against wild-type and resistant strains, needed in the fight against current and future outbreaks caused by different IFV strains. In this review, we summarize general strategies used for the discovery and development of antiviral agents targeting multiple IFV strains (including those resistant to available drugs). Structure-based drug design, mechanism-based drug design, multivalent interaction-based drug design and drug repurposing are amongst the most relevant strategies that provide a framework for the development of antiviral drugs targeting IFV.
Collapse
Affiliation(s)
- Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Lide Hu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Guanyu Dong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Ying Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Edeildo Ferreira da Silva-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió 57072-970, Alagoas, Brazil
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Luis Menéndez-Arias
- Centro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid), Madrid 28049, Spain
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
44
|
Zhu Z, Fan H, Fodor E. Defining the minimal components of the influenza A virus replication machinery via an in vitro reconstitution system. PLoS Biol 2023; 21:e3002370. [PMID: 37943954 PMCID: PMC10662765 DOI: 10.1371/journal.pbio.3002370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/21/2023] [Accepted: 10/09/2023] [Indexed: 11/12/2023] Open
Abstract
During influenza A virus infection, the viral RNA polymerase transcribes the viral negative-sense segmented RNA genome and replicates it in a two-step process via complementary RNA within viral ribonucleoprotein (vRNP) complexes. While numerous viral and host factors involved in vRNP functions have been identified, dissecting the roles of individual factors remains challenging due to the complex cellular environment in which vRNP activity has been studied. To overcome this challenge, we reconstituted viral transcription and a full cycle of replication in a test tube using vRNPs isolated from virions and recombinant factors essential for these processes. This novel system uncovers the minimal components required for influenza virus replication and also reveals new roles of regulatory factors in viral replication. Moreover, it sheds light on the molecular interplay underlying the temporal regulation of viral transcription and replication. Our highly robust in vitro system enables systematic functional analysis of factors modulating influenza virus vRNP activity and paves the way for imaging key steps of viral transcription and replication.
Collapse
Affiliation(s)
- Zihan Zhu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Haitian Fan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Ervin Fodor
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Abstract
The nonsegmented, negative-strand RNA viruses (nsNSVs), also known as the order Mononegavirales, have a genome consisting of a single strand of negative-sense RNA. Integral to the nsNSV replication cycle is the viral polymerase, which is responsible for transcribing the viral genome, to produce an array of capped and polyadenylated messenger RNAs, and replicating it to produce new genomes. To perform the different steps that are necessary for these processes, the nsNSV polymerases undergo a series of coordinated conformational transitions. While much is still to be learned regarding the intersection of nsNSV polymerase dynamics, structure, and function, recently published polymerase structures, combined with a history of biochemical and molecular biology studies, have provided new insights into how nsNSV polymerases function as dynamic machines. In this review, we consider each of the steps involved in nsNSV transcription and replication and suggest how these relate to solved polymerase structures.
Collapse
Affiliation(s)
- Mohamed Ouizougun-Oubari
- Department of Virology, Immunology & Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA;
| | - Rachel Fearns
- Department of Virology, Immunology & Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA;
| |
Collapse
|
46
|
Camacho-Zarco AR, Yu L, Krischuns T, Dedeoglu S, Maurin D, Bouvignies G, Crépin T, Ruigrok RWH, Cusack S, Naffakh N, Blackledge M. Multivalent Dynamic Colocalization of Avian Influenza Polymerase and Nucleoprotein by Intrinsically Disordered ANP32A Reveals the Molecular Basis of Human Adaptation. J Am Chem Soc 2023; 145:20985-21001. [PMID: 37707433 PMCID: PMC10540212 DOI: 10.1021/jacs.3c06965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Indexed: 09/15/2023]
Abstract
Adaptation of avian influenza RNA polymerase (FluPol) to human cells requires mutations on the 627-NLS domains of the PB2 subunit. The E627K adaptive mutation compensates a 33-amino-acid deletion in the acidic intrinsically disordered domain of the host transcription regulator ANP32A, a deletion that restricts FluPol activity in mammalian cells. The function of ANP32A in the replication transcription complex and in particular its role in host restriction remains poorly understood. Here we characterize ternary complexes formed between ANP32A, FluPol, and the viral nucleoprotein, NP, supporting the putative role of ANP32A in shuttling NP to the replicase complex. We demonstrate that while FluPol and NP can simultaneously bind distinct linear motifs on avian ANP32A, the deletion in the shorter human ANP32A blocks this mode of colocalization. NMR reveals that NP and human-adapted FluPol, containing the E627 K mutation, simultaneously bind the identical extended linear motif on human ANP32A in an electrostatically driven, highly dynamic and multivalent ternary complex. This study reveals a probable molecular mechanism underlying host adaptation, whereby E627K, which enhances the basic surface of the 627 domain, is selected to confer the necessary multivalent properties to allow ANP32A to colocalize NP and FluPol in human cells.
Collapse
Affiliation(s)
- Aldo R. Camacho-Zarco
- Institut
de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS
UMR5075, 71 Avenue des
Martyrs, 38000 Grenoble, France
| | - Lefan Yu
- Institut
de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS
UMR5075, 71 Avenue des
Martyrs, 38000 Grenoble, France
| | - Tim Krischuns
- Institut
Pasteur, Université Paris Cité,
CNRS UMR3569, Unité Biologie des ARN et Virus Influenza, 75015 Paris, France
| | - Selin Dedeoglu
- Institut
de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS
UMR5075, 71 Avenue des
Martyrs, 38000 Grenoble, France
| | - Damien Maurin
- Institut
de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS
UMR5075, 71 Avenue des
Martyrs, 38000 Grenoble, France
| | - Guillaume Bouvignies
- Laboratoire
des Biomolécules, Département de Chimie, École
Normale Supérieur, UPMC Université Paris 06, CNRS, PSL Research University, 24 rue Lhomond, 75005 Paris, France
| | - Thibaut Crépin
- Institut
de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS
UMR5075, 71 Avenue des
Martyrs, 38000 Grenoble, France
| | - Rob W. H. Ruigrok
- Institut
de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS
UMR5075, 71 Avenue des
Martyrs, 38000 Grenoble, France
| | - Stephan Cusack
- European
Molecular Biology Laboratory, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Nadia Naffakh
- Institut
Pasteur, Université Paris Cité,
CNRS UMR3569, Unité Biologie des ARN et Virus Influenza, 75015 Paris, France
| | - Martin Blackledge
- Institut
de Biologie Structurale, Université Grenoble Alpes-CEA-CNRS
UMR5075, 71 Avenue des
Martyrs, 38000 Grenoble, France
| |
Collapse
|
47
|
Petrone-García VM, Castellanos-Huerta I, Tellez-Isaias G. Editorial: High-impact respiratory RNA virus diseases. Front Vet Sci 2023; 10:1273650. [PMID: 37675076 PMCID: PMC10478262 DOI: 10.3389/fvets.2023.1273650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023] Open
|
48
|
Villa R, Renzi S, Dotti S, Lucchini F. siRNAs pools generated in Escherichia coli exhibit strong RNA-interference activity against influenza virus genomic sequences. Virology 2023; 579:38-45. [PMID: 36599198 DOI: 10.1016/j.virol.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023]
Abstract
Due to the recurrent pandemic outbreaks that occurred during the last century, Influenza A viruses are considered a serious potential danger to human health. Among the innate immune pathways in eukaryotes, RNA interference plays a significant role in the interaction between viruses and host cells. RNA interference is addressed by small dsRNA molecules produced by the host itself (miRNAs, i.e. "micro-RNAs") but can be triggered also by the administration of exogenous short RNAs (siRNAs, "short interfering RNAs"). In this work, artificial siRNA pools targeting NP and PB genomic regions of the Influenza virus were produced in engineered Escherichia coli, adapting a published protocol. In a MDCK cell in vitro model, these preparations were challenged against reporter vectors bearing viral genomic sequences. A strong and specific RNA interference activity was observed, and the details of this action were indagated.
Collapse
Affiliation(s)
- Riccardo Villa
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Laboratorio di Controllo di Prodotti Biologici, Farmaceutici e Convalida di Processi Produttivi, via A. Bianchi 9, Brescia, Italy
| | - Sabrina Renzi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Laboratorio di Controllo di Prodotti Biologici, Farmaceutici e Convalida di Processi Produttivi, via A. Bianchi 9, Brescia, Italy
| | - Silvia Dotti
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna (IZSLER), Laboratorio di Controllo di Prodotti Biologici, Farmaceutici e Convalida di Processi Produttivi, via A. Bianchi 9, Brescia, Italy
| | - Franco Lucchini
- Università Cattolica del Sacro Cuore, Facoltà di Scienze Agrarie, Alimentari ed Ambientali, DiSTAS - Dipartimento di Scienze e Tecnologie Alimentari per una filiera agro-alimentare sostenibile, via L. Bissolati 74, Cremona, Italy.
| |
Collapse
|