1
|
Mostafa A, Barre RS, Allué-Guardia A, Escobedo RA, Shivanna V, Rothan H, Castro EM, Ma Y, Cupic A, Jackson N, Bayoumi M, Torrelles JB, Ye C, García-Sastre A, Martinez-Sobrido L. Replication kinetics, pathogenicity and virus-induced cellular responses of cattle-origin influenza A(H5N1) isolates from Texas, United States. Emerg Microbes Infect 2025; 14:2447614. [PMID: 39727152 PMCID: PMC11721806 DOI: 10.1080/22221751.2024.2447614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/06/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
The host range of HPAIV H5N1 was recently expanded to include ruminants, particularly dairy cattle in the United States (US). Shortly after, human H5N1 infection was reported in a dairy worker in Texas following exposure to infected cattle. Herein, we rescued the cattle-origin influenza A/bovine/Texas/24-029328-02/2024(H5N1, rHPbTX) and A/Texas/37/2024(H5N1, rHPhTX) viruses, identified in dairy cattle and human, respectively, and their low pathogenic forms, rLPbTX and rLPhTX, with monobasic HA cleavage sites. Intriguingly, rHPhTX replicated more efficiently than rHPbTX in mammalian and avian cells. Still, variations in the PA and NA proteins didn't affect their antiviral susceptibility to PA and NA inhibitors. Unlike rHPbTX and rLPbTX, both rHPhTX and rLPhTX exhibited higher pathogenicity and efficient replication in infected C57BL/6J mice. The lungs of rHPhTX-infected mice produced higher inflammatory cytokines/chemokines than rHPbTX-infected mice. Our results highlight the potential risk of HPAIV H5N1 virus adaptation in human and/or dairy cattle during the current multistate/multispecies outbreak in the US.
Collapse
Affiliation(s)
- Ahmed Mostafa
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
- International Center for the Advancement of Research and Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ramya S. Barre
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Anna Allué-Guardia
- Population Health Program, Tuberculosis Group, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ruby A. Escobedo
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Vinay Shivanna
- Southwest National Primate Research Center at the Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Hussin Rothan
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Esteban M. Castro
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Yao Ma
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathaniel Jackson
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Mahmoud Bayoumi
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
- Virology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Jordi B. Torrelles
- International Center for the Advancement of Research and Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, USA
- Population Health Program, Tuberculosis Group, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chengjin Ye
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luis Martinez-Sobrido
- Host-pathogen interactions (HPI) and Disease Intervention and Prevention (DIP) programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
2
|
Neumann G, Eisfeld AJ, Kawaoka Y. Viral factors underlying the pandemic potential of influenza viruses. Microbiol Mol Biol Rev 2025:e0006624. [PMID: 40340558 DOI: 10.1128/mmbr.00066-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025] Open
Abstract
SUMMARYOver the past 25 years, there has been an increasing number of mammalian (including human) infections caused by avian influenza A viruses that resulted in mild to severe illnesses. These viruses typically did not spread between mammals through aerosols in nature or in experimental settings. However, recently, this has changed, with several avian influenza A viruses exhibiting aerosol transmissibility among mammals, indicating that these viruses may pose a greater pandemic risk. In this review, we examine the current situation and discuss the mutations that may be necessary for avian influenza A viruses to efficiently replicate in mammals and transmit among them via aerosols.
Collapse
Affiliation(s)
- Gabriele Neumann
- Department of Pathobiological Sciences, Influenza Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amie J Eisfeld
- Department of Pathobiological Sciences, Influenza Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, Influenza Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced research center (UTOPIA), University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| |
Collapse
|
3
|
Nguyen TQ, Hutter CR, Markin A, Thomas M, Lantz K, Killian ML, Janzen GM, Vijendran S, Wagle S, Inderski B, Magstadt DR, Li G, Diel DG, Frye EA, Dimitrov KM, Swinford AK, Thompson AC, Snekvik KR, Suarez DL, Lakin SM, Schwabenlander S, Ahola SC, Johnson KR, Baker AL, Robbe-Austerman S, Torchetti MK, Anderson TK. Emergence and interstate spread of highly pathogenic avian influenza A(H5N1) in dairy cattle in the United States. Science 2025; 388:eadq0900. [PMID: 40273240 DOI: 10.1126/science.adq0900] [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: 05/01/2024] [Accepted: 02/14/2025] [Indexed: 04/26/2025]
Abstract
Highly pathogenic avian influenza (HPAI) viruses cross species barriers and have the potential to cause pandemics. In North America, HPAI A(H5N1) viruses related to the goose/Guangdong 2.3.4.4b hemagglutinin phylogenetic clade have infected wild birds, poultry, and mammals. Our genomic analysis and epidemiological investigation showed that a reassortment event in wild bird populations preceded a single wild bird-to-cattle transmission episode. The movement of asymptomatic or presymptomatic cattle has likely played a role in the spread of HPAI within the United States dairy herd. Some molecular markers that may lead to changes in transmission efficiency and phenotype were detected at low frequencies. Continued transmission of H5N1 HPAI within dairy cattle increases the risk for infection and subsequent spread of the virus to human populations.
Collapse
Affiliation(s)
- Thao-Quyen Nguyen
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Department of Computer Science, Iowa State University, Ames, IA, USA
| | - Carl R Hutter
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Alexey Markin
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Megan Thomas
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Kristina Lantz
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Services, United States Department of Agriculture, Ames, IA, USA
| | - Mary Lea Killian
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Services, United States Department of Agriculture, Ames, IA, USA
| | - Garrett M Janzen
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Sriram Vijendran
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Department of Computer Science, Iowa State University, Ames, IA, USA
| | - Sanket Wagle
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Department of Computer Science, Iowa State University, Ames, IA, USA
| | - Blake Inderski
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Drew R Magstadt
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Ganwu Li
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Elisha Anna Frye
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Kiril M Dimitrov
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, TX, USA
| | - Amy K Swinford
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, TX, USA
| | | | - Kevin R Snekvik
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
- The Washington Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - David L Suarez
- Southeast Poultry Research Laboratory, National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture, Athens, GA, USA
| | - Steven M Lakin
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Services, United States Department of Agriculture, Ames, IA, USA
| | - Stacey Schwabenlander
- Ruminant Health Center, Animal and Plant Health Inspection Services, United States Department of Agriculture, Riverdale, MD, USA
| | - Sara C Ahola
- Field Epidemiologic Investigation Services, Animal and Plant Health Inspection Services, United States Department of Agriculture, Ft. Collins, CO, USA
| | - Kammy R Johnson
- Field Epidemiologic Investigation Services, Animal and Plant Health Inspection Services, United States Department of Agriculture, Ft. Collins, CO, USA
| | - Amy L Baker
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Suelee Robbe-Austerman
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Services, United States Department of Agriculture, Ames, IA, USA
| | - Mia Kim Torchetti
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Services, United States Department of Agriculture, Ames, IA, USA
| | - Tavis K Anderson
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| |
Collapse
|
4
|
Zhang L, Mysore SP. The barn owl in systems and behavioral neuroscience: Progress and promise. Curr Opin Neurobiol 2025; 91:102983. [PMID: 39987690 DOI: 10.1016/j.conb.2025.102983] [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: 08/23/2024] [Revised: 12/24/2024] [Accepted: 01/31/2025] [Indexed: 02/25/2025]
Abstract
Though well-adapted to their evolutionary niches, animals exhibit a repertoire of behavioral functions that are common across species. Neuroscientific research that promotes the study of similar functions in multiple species, can illuminate shared versus specialized design features of the nervous system, revealing potentially profound insights into the neural basis of behavior and cognition. Here, we advance the idea that the barn owl is an excellent animal model in which to investigate such common functions. We do so by drawing attention to the range of exciting questions that can be asked in the owl beyond those deriving from its evolutionary specializations, by underscoring the variety of complex yet experimentally tractable behaviors it exhibits naturally, by emphasizing its complex network of brain systems, and by highlighting emerging opportunities for the application of modern neural technologies. Our goal is to motivate broader adoption of the powerful barn owl model for behavioral and systems neuroscience.
Collapse
Affiliation(s)
- Lilian Zhang
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, USA
| | - Shreesh P Mysore
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, 21218, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, USA.
| |
Collapse
|
5
|
Ryu HJ, Kim SE, Kim KH, Park JH, Jeong H, Shaleh I, Park TS. Signaling pathways of duck RIG-I in gene-edited DF1 chicken cells. Poult Sci 2025; 104:104739. [PMID: 39733732 PMCID: PMC11743317 DOI: 10.1016/j.psj.2024.104739] [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: 08/26/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024] Open
Abstract
Retinoic acid inducible gene I (RIG-I) is an innate immune RNA sensor which can detect viral infection such as influenza viruses. Duck but not chicken has an RIG-I gene. However, the immune responses could be induced in chicken cells by transferring the duck RIG-I transgene. However, effects of other pathogen-recognition receptor (PRR) genes such as Toll-like receptor 3 (TLR3) and melanoma differentiation-associated protein 5 (MDA5) could not be ruled out. In this study, we knocked out TLR3 and MDA5 genes using gene-editing protocol, and stably transferred the duck RIG-I transgene into TLR3/MDA5 double knockout (KO) chicken DF1 cells. We investigated the antiviral responses induced by duck RIG-I in chicken cells. Duck RIG-I induced the expression of interferon-stimulated genes (ISGs) and inflammatory cytokines such as interferon regulatory factor 7 (IRF7), interferon β (IFNβ), Mx1, and protein kinase R1 (PKR1) after treatment with polyinosinic: polycytidylic acid (poly I:C) in TLR3/MDA5 double KO DF1 cells. Additionally, to examine the duck RIG-I signaling cascade, we knocked out mitochondrial antiviral-signaling protein (MAVS), which encodes an antiviral signaling factor in innate immunity. Duck RIG-I in TLR3/MDA5/MAVS triple KO DF1 cells did not activate downstream expression of ISGs. Finally, to analyze the global signaling pathways of duck RIG-I in chicken cells, next-generation sequencing of total mRNAs with and without poly I:C treatment was conducted. In conclusion, duck RIG-I mediated antiviral signaling independently of TLR3 and MDA5, and MAVS induced and stimulated ISGs by duck RIG-I in chicken cells.
Collapse
Affiliation(s)
- Hyeong Ju Ryu
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea.
| | - Si Eun Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea.
| | - Ki Hyun Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea.
| | - Ji Hoon Park
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea.
| | - Heesu Jeong
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea.
| | - Ismail Shaleh
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea; Department of Biology, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, Indonesia.
| | - Tae Sub Park
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea; Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, South Korea.
| |
Collapse
|
6
|
Fujimoto Y, Ozaki K, Ono E. Chicken ANP32A-independent replication of highly pathogenic avian influenza viruses potentially leads to mammalian adaptation-related amino acid substitutions in viral PB2 and PA proteins. J Virol 2025; 99:e0184024. [PMID: 39570060 PMCID: PMC11784356 DOI: 10.1128/jvi.01840-24] [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: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
Acidic nuclear phosphoprotein 32 family member A (ANP32A) is an important host factor that supports the efficient replication of avian influenza viruses (AIVs). To develop an antiviral strategy against Gs/Gd-lineage H5 highly pathogenic avian influenza (HPAI) viruses in chickens, we established chicken ANP32-knockout (chANP32A-KO) DF-1 cells and evaluated their antiviral efficacy through in vitro validation. The replication of all HPAI viruses tested in chANP32A-KO cells was significantly lower compared to that of wild-type DF-1 cells. However, when HPAI strains A/mountain hawk-eagle/Kumamoto/1/2007 (H5N1; MHE) and A/chicken/Aichi/2/2011 (H5N1; H5Aichi) were passed in chANP32A-KO cells, mutant viruses were generated, which exhibited comparable replication levels in both chANP32A-KO and wild-type DF-1 cells. Sequence analysis revealed that mammalian-adaptive amino acid mutations PB2_D256G and PA_T97I were present in the MHE mutant virus, and the PB2_E627K mutation was identified in the H5Aichi mutant virus. These mutations have also been reported to enhance the polymerase activity of AIVs in mammalian cells; however, the minigenome assay in the present study showed that the polymerase activity of mutant viruses in chANP32A-KO cells was not restored to levels comparable to those in wild-type DF-1 cells. These findings suggest that ANP32A-independent viral replication may induce amino acid substitutions associated with mammalian adaptation in AIVs. They also imply that the high efficiency of viral replication mediated by these amino acid mutations may not result from enhanced polymerase activity but rather involve other undefined mechanisms.IMPORTANCEDuring the host-switching of avian influenza viruses (AIVs) to mammalian hosts, introducing adaptive mutations into viral proteins is essential to ensure optimal functionality through virus-host protein interactions in mammalian cells. However, the mechanisms leading to adaptive mutations in viral proteins remain unclear. Among several host proteins that promote viral growth, acidic nuclear phosphoprotein 32 family member A (ANP32A) is known to be an important factor for efficient viral replication. Here, we generated mutant highly pathogenic avian influenza viruses capable of ANP32A-independent replication in a chicken-derived cell line. We demonstrated that several amino acid mutations found in the mutant viruses correspond to those associated with the mammalian adaptation of AIVs. These results suggest that ANP32A-independent viral replication is one of the mechanisms for introducing amino acid mutations that are reportedly involved in the mammalian adaptation of AIVs.
Collapse
Affiliation(s)
- Yoshikazu Fujimoto
- Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
- Joint Graduate School of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Kinuyo Ozaki
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Etsuro Ono
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
7
|
Niu YJ, Wu J, Ren W, Liu G, Wu G, Peng Y, Zheng D, Jin K, Zuo Q, Li G, Han W, Cui XS, Chen G, Li B. Aflatoxin B1 impairs the growth and development of chicken PGCs through oxidative stress and mitochondrial dysfunction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117727. [PMID: 39818136 DOI: 10.1016/j.ecoenv.2025.117727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Aflatoxins harm the reproductive system and gamete development in animals. Primordial germ cells (PGCs) in chickens, as ancestral cells of gametes, are essential for genetic transmission, yet the impact and mechanisms of aflatoxins on them remain elusive. This study systematically investigated the effects of aflatoxin B1 (AFB1) on chicken PGCs and their potential mechanisms using an in vitro culture model. We observed a significant reduction in PGC numbers and an increase in apoptosis levels with AFB1 treatment. Further analysis revealed that AFB1 induced mitochondrial structural and functional abnormalities. Additionally, AFB1 treatment led to increased oxidative stress, lipid peroxidation, ferroptosis, and autophagy in chicken PGCs, ultimately affecting their biological characteristics. Interestingly, we found that the NRF2-mediated antioxidant pathway was activated in AFB1-treated PGCs. Inhibiting NRF2 exacerbated oxidative stress and cell death in PGCs, suggesting NRF2 upregulation plays a protective role under AFB1 regulation. This study illuminates AFB1's toxic effects on chicken PGCs and provides insights into potential mechanisms, establishing a basis for strategies to prevent and treat AFB1's adverse effects on poultry genetic transmission.
Collapse
Affiliation(s)
- Ying-Jie Niu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China.
| | - Jun Wu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Wenjie Ren
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Guangzheng Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Gaoyuan Wu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yixiu Peng
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Dan Zheng
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Kai Jin
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Qisheng Zuo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Guohui Li
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu 225125, China
| | - Wei Han
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu 225125, China
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, South Korea
| | - Guohong Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Bichun Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China; Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.
| |
Collapse
|
8
|
Gu Y, Wu K, Niu B, Wang Z, Jie Y, Fan Z, Li J, Sun C, Hou ZC, Shao LW. Hyperthermia suppresses the biological characteristics and migration of chicken primordial germ cells. Front Genome Ed 2025; 6:1512108. [PMID: 39845894 PMCID: PMC11751037 DOI: 10.3389/fgeed.2024.1512108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Primordial germ cells (PGCs) play a crucial role in transmitting genetic information to the next-generation. In chickens, genetically edited PGCs can be propagated in vitro and subsequently transplanted into recipient embryos to produce offspring with desired genetic traits. However, during early embryogenesis, the effects of external conditions on PGC migration through the vascular system to the gonads have yet to be explored, which may affect the efficiency of preparing gene-edited chickens. In this study, we investigated the effects of hyperthermia on the biological characteristics and migration of chicken PGCs. A gonad-derived PGC line of White Leghorn (WLH) chicken was established and verified through PAS staining and immunofluorescence of PGC-specific proteins. To visually observe PGC migration in vivo, GFP-positive PGCs were prepared and locations of chimeras were validated. Cell viability, glycogen granule contents, and mRNA expression levels of pluripotency markers (NANOG and POUV), germ cell-specific markers (DAZL and CVH), and telomerase reverse transcriptase (TERT) were reduced in PGCs cultured under high temperatures (43°C for 12, 24, and 48 h). After the heat treatment of donor PGCs (43°C) or recipient embryos (39.5°C), GFP-positive PGCs in gonads were rarely observed. Taken together, our results underscore the negative effects of hyperthermia on the biological characteristics and migration of chicken PGCs, which provides valuable insights for the implementation of PGC-based gene editing techniques in chickens.
Collapse
Affiliation(s)
- Yuzhou Gu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Kexin Wu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Bowen Niu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Zhiting Wang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Yuchen Jie
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Zixuan Fan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Junying Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Zhuo-Cheng Hou
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Li-Wa Shao
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| |
Collapse
|
9
|
Mostafa A, Naguib MM, Nogales A, Barre RS, Stewart JP, García-Sastre A, Martinez-Sobrido L. Avian influenza A (H5N1) virus in dairy cattle: origin, evolution, and cross-species transmission. mBio 2024; 15:e0254224. [PMID: 39535188 PMCID: PMC11633217 DOI: 10.1128/mbio.02542-24] [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] [Indexed: 11/16/2024] Open
Abstract
Since the emergence of highly pathogenic avian influenza virus (HPAIV) H5N1 of clade 2.3.4.4b as a novel reassortant virus from subtype H5N8, the virus has led to a massive number of outbreaks worldwide in wild and domestic birds. Compared to the parental HPAIV H5N8 clade 2.3.4.4b, the novel reassortant HPAIV H5N1 displayed an increased ability to escape species barriers and infect multiple mammalian species, including humans. The virus host range has been recently expanded to include ruminants, particularly dairy cattle in the United States, where cattle-to-cattle transmission was reported. As with the avian 2.3.4.4.b H5N1 viruses, the cattle-infecting virus was found to transmit from cattle to other contact animals including cats, raccoons, rodents, opossums, and poultry. Although replication of the virus in cows appears to be mainly confined to the mammary tissue, with high levels of viral loads detected in milk, infected cats and poultry showed severe respiratory disease, neurologic signs, and eventually died. Furthermore, several human infections with HPAIV H5N1 have also been reported in dairy farm workers and were attributed to exposures to infected dairy cattle. This is believed to represent the first mammalian-to-human transmission report of the HPAIV H5N1. Fortunately, infection in humans and cows, as opposed to other animals, appears to be mild in most cases. Nevertheless, the H5N1 bovine outbreak represents the largest outbreak of the H5N1 in a domestic mammal close to humans, increasing the risk that this already mammalian adapted H5N1 further adapts to human-to-human transmission and starts a pandemic. Herein, we discuss the epidemiology, evolution, pathogenesis, and potential impact of the recently identified HPAIV H5N1 clade 2.3.4.4b in dairy cattle in the United States. Eventually, interdisciplinary cooperation under a One Health framework is required to be able to control this ongoing HPAIV H5N1 outbreak to stop it before further expansion of its host range and geographical distribution.
Collapse
Affiliation(s)
- Ahmed Mostafa
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Mahmoud M. Naguib
- Department of Infection Biology and Microbiomes, University of Liverpool, Liverpool, United Kingdom
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, Madrid, Spain
| | - Ramya S. Barre
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - James P. Stewart
- Department of Infection Biology and Microbiomes, University of Liverpool, Liverpool, United Kingdom
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Luis Martinez-Sobrido
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
10
|
Kwon T, Artiaga BL, McDowell CD, Whitworth KM, Wells KD, Prather RS, Delhon G, Cigan M, White SN, Retallick J, Gaudreault NN, Morozov I, Richt JA. Gene editing of pigs to control influenza A virus infections. Emerg Microbes Infect 2024; 13:2387449. [PMID: 39083026 PMCID: PMC11346336 DOI: 10.1080/22221751.2024.2387449] [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: 04/28/2024] [Revised: 06/24/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
Proteolytic activation of the haemagglutinin (HA) glycoprotein by host cellular proteases is pivotal for influenza A virus (IAV) infectivity. Highly pathogenic avian influenza viruses possess the multibasic cleavage site of the HA which is cleaved by ubiquitous proteases, such as furin; in contrast, the monobasic HA motif is recognized and activated by trypsin-like proteases, such as the transmembrane serine protease 2 (TMPRSS2). Here, we aimed to determine the effects of TMPRSS2 on the replication of pandemic H1N1 and H3N2 subtype IAVs in the natural host, the pig. The use of the CRISPR/Cas 9 system led to the establishment of homozygous gene edited (GE) TMPRSS2 knockout (KO) pigs. Delayed IAV replication was demonstrated in primary respiratory cells of KO pigs in vitro. IAV infection in vivo resulted in a significant reduction of virus shedding in the upper respiratory tract, and lower virus titers and pathological lesions in the lower respiratory tract of TMPRSS2 KO pigs as compared to wild-type pigs. Our findings support the commercial use of GE pigs to mitigate influenza A virus infection in pigs, as an alternative approach to prevent zoonotic influenza A transmissions from pigs to humans.
Collapse
Affiliation(s)
- Taeyong Kwon
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Bianca L. Artiaga
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Chester D. McDowell
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Kristin M. Whitworth
- Division of Animal Science & National Swine Resource and Research Center, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Kevin D. Wells
- Division of Animal Science & National Swine Resource and Research Center, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Randall S. Prather
- Division of Animal Science & National Swine Resource and Research Center, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Gustavo Delhon
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | | | - Jamie Retallick
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Igor Morozov
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
11
|
Gu C, Maemura T, Guan L, Eisfeld AJ, Biswas A, Kiso M, Uraki R, Ito M, Trifkovic S, Wang T, Babujee L, Presler R, Dahn R, Suzuki Y, Halfmann PJ, Yamayoshi S, Neumann G, Kawaoka Y. A human isolate of bovine H5N1 is transmissible and lethal in animal models. Nature 2024; 636:711-718. [PMID: 39467571 DOI: 10.1038/s41586-024-08254-7] [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: 07/18/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
The outbreak of clade 2.3.4.4b highly pathogenic avian influenza viruses of the H5N1 subtype (HPAI H5N1) in dairy cattle in the USA has so far resulted in spillover infections of at least 14 farm workers1-3, who presented with mild respiratory symptoms or conjunctivitis, and one individual with no known animal exposure who was hospitalized but recovered3,4. Here we characterized A/Texas/37/2024 (huTX37-H5N1), a virus isolated from the eyes of an infected farm worker who developed conjunctivitis5. huTX37-H5N1 replicated efficiently in primary human alveolar epithelial cells, but less efficiently in corneal epithelial cells. Despite causing mild disease in the infected worker, huTX37-H5N1 proved lethal in mice and ferrets and spread systemically, with high titres in both respiratory and non-respiratory organs. Importantly, in four independent experiments in ferrets, huTX37-H5N1 transmitted by respiratory droplets in 17-33% of transmission pairs, and five of six exposed ferrets that became infected died. PB2-631L (encoded by bovine isolates) promoted influenza polymerase activity in human cells, suggesting a role in mammalian adaptation similar to that of PB2-627K (encoded by huTX37-H5N1). In addition, bovine HPAI H5N1 virus was found to be susceptible to polymerase inhibitors both in vitro and in mice. Thus, HPAI H5N1 virus derived from dairy cattle transmits by respiratory droplets in mammals without previous adaptation and causes lethal disease in animal models.
Collapse
Affiliation(s)
- Chunyang Gu
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Tadashi Maemura
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lizheng Guan
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Amie J Eisfeld
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Asim Biswas
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Maki Kiso
- Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ryuta Uraki
- Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Mutsumi Ito
- Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Sanja Trifkovic
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Tong Wang
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lavanya Babujee
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert Presler
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Randall Dahn
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yasuo Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Seiya Yamayoshi
- Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Gabriele Neumann
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), University of Tokyo, Tokyo, Japan.
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
| |
Collapse
|
12
|
Hu S, Gan M, Wei Z, Shang P, Song L, Feng J, Chen L, Niu L, Wang Y, Zhang S, Shen L, Zhu L, Zhao Y. Identification of host factors for livestock and poultry viruses: genome-wide screening technology based on the CRISPR system. Front Microbiol 2024; 15:1498641. [PMID: 39640855 PMCID: PMC11619636 DOI: 10.3389/fmicb.2024.1498641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Genome-wide CRISPR library screening technology is a gene function research tool developed based on the CRISPR/Cas9 gene-editing system. The clustered regularly interspaced short palindromic repeats/CRISPR-associated genes (CRISPR/Cas) system, considered the third generation of gene editing after zinc finger nucleases (ZFN) and transcription activator-like effector nucleases (TALEN), is widely used for screening various viral host factors. CRISPR libraries are classified into three main categories based on the different functions of Cas9 enzymes: CRISPR knockout (CRISPR KO) library screening, CRISPR transcriptional activation (CRISPRa) library screening, and CRISPR transcriptional interference (CRISPRi) library screening. Recently, genome-wide CRISPR library screening technology has been used to identify host factors that interact with viruses at various stages, including adsorption, endocytosis, and replication. By specifically modulating the expression of these host factors, it becomes possible to cultivate disease-resistant varieties, establish disease models, and design and develop vaccines, among other applications. This review provides an overview of the development and technical processes of genome-wide CRISPR library screening, as well as its applications in identifying viral host factors in livestock and poultry.
Collapse
Affiliation(s)
- Shijie Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Mailin Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Ziang Wei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Pan Shang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lei Song
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Jinkang Feng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lei Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Shunhua Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
| | - Linyuan Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| | - Ye Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Ya’an, China
| |
Collapse
|
13
|
Holmes EC, Krammer F, Goodrum FD. Virology-The next fifty years. Cell 2024; 187:5128-5145. [PMID: 39303682 PMCID: PMC11467463 DOI: 10.1016/j.cell.2024.07.025] [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: 02/29/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 09/22/2024]
Abstract
Virology has made enormous advances in the last 50 years but has never faced such scrutiny as it does today. Herein, we outline some of the major advances made in virology during this period, particularly in light of the COVID-19 pandemic, and suggest some areas that may be of research importance in the next 50 years. We focus on several linked themes: cataloging the genomic and phenotypic diversity of the virosphere; understanding disease emergence; future directions in viral disease therapies, vaccines, and interventions; host-virus interactions; the role of viruses in chronic diseases; and viruses as tools for cell biology. We highlight the challenges that virology will face moving forward-not just the scientific and technical but also the social and political. Although there are inherent limitations in trying to outline the virology of the future, we hope this article will help inspire the next generation of virologists.
Collapse
Affiliation(s)
- Edward C. Holmes
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Laboratory of Data Discovery for Health Limited, Hong Kong SAR, China
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Felicia D. Goodrum
- Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
14
|
Drobik-Czwarno W, Wolc A, Petal CR, Miedzinska K, Dekkers J, Fulton JE, Smith J. Candidate Genes Associated with Survival Following Highly Pathogenic Avian Influenza Infection in Chickens. Int J Mol Sci 2024; 25:10056. [PMID: 39337540 PMCID: PMC11432379 DOI: 10.3390/ijms251810056] [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: 08/19/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Highly pathogenic strains of avian influenza (HPAI) devastate poultry flocks and result in significant economic losses for farmers due to high mortality, reduced egg production, and mandated euthanization of infected flocks. Within recent years, HPAI outbreaks have affected egg production flocks across the world. The H5N2 outbreak in the US in 2015 resulted in over 99% mortality. Here, we analyze sequence data from chickens that survived (42 cases) along with uninfected controls (28 samples) to find genomic regions that differ between these two groups and that, therefore, may encompass prime candidates that are resistant to HPAI. Blood samples were obtained from survivors of the 2015 HPAI outbreak plus age and genetics-matched non-affected controls. A whole-genome sequence was obtained, and genetic variants were characterized and used in a genome-wide association study to identify regions showing significant association with survival. Regions associated with HPAI resistance were observed on chromosomes 1, 2, 5, 8, 10, 11, 15, 20, and 28, with a number of candidate genes identified. We did not detect a specific locus which could fully explain the difference between survivors and controls. Influenza virus replication depends on multiple components of the host cellular machinery, with many genes involved in the host response.
Collapse
Affiliation(s)
- Wioleta Drobik-Czwarno
- Department of Animal Genetics and Conservation, Institute of Animal Science, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Anna Wolc
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA; (A.W.); (J.D.)
- Hy-Line International, P.O. Box 310, 2583 240th St., Dallas Center, IA 50063, USA;
| | - Callie R. Petal
- The Roslin Institute and Royal (Dick) School of Veterinary Studies R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (C.R.P.); (K.M.)
| | - Katarzyna Miedzinska
- The Roslin Institute and Royal (Dick) School of Veterinary Studies R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (C.R.P.); (K.M.)
| | - Jack Dekkers
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA; (A.W.); (J.D.)
| | - Janet E. Fulton
- Hy-Line International, P.O. Box 310, 2583 240th St., Dallas Center, IA 50063, USA;
| | - Jacqueline Smith
- The Roslin Institute and Royal (Dick) School of Veterinary Studies R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (C.R.P.); (K.M.)
| |
Collapse
|
15
|
Eisfeld AJ, Biswas A, Guan L, Gu C, Maemura T, Trifkovic S, Wang T, Babujee L, Dahn R, Halfmann PJ, Barnhardt T, Neumann G, Suzuki Y, Thompson A, Swinford AK, Dimitrov KM, Poulsen K, Kawaoka Y. Pathogenicity and transmissibility of bovine H5N1 influenza virus. Nature 2024; 633:426-432. [PMID: 38977017 PMCID: PMC11390473 DOI: 10.1038/s41586-024-07766-6] [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: 06/01/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
Highly pathogenic H5N1 avian influenza (HPAI H5N1) viruses occasionally infect, but typically do not transmit, in mammals. In the spring of 2024, an unprecedented outbreak of HPAI H5N1 in bovine herds occurred in the USA, with virus spread within and between herds, infections in poultry and cats, and spillover into humans, collectively indicating an increased public health risk1-4. Here we characterize an HPAI H5N1 virus isolated from infected cow milk in mice and ferrets. Like other HPAI H5N1 viruses, the bovine H5N1 virus spread systemically, including to the mammary glands of both species, however, this tropism was also observed for an older HPAI H5N1 virus isolate. Bovine HPAI H5N1 virus bound to sialic acids expressed in human upper airways and inefficiently transmitted to exposed ferrets (one of four exposed ferrets seroconverted without virus detection). Bovine HPAI H5N1 virus thus possesses features that may facilitate infection and transmission in mammals.
Collapse
Affiliation(s)
- Amie J Eisfeld
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Asim Biswas
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lizheng Guan
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Chunyang Gu
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Tadashi Maemura
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Sanja Trifkovic
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Tong Wang
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lavanya Babujee
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Randall Dahn
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Gabriele Neumann
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yasuo Suzuki
- Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Alexis Thompson
- Texas A&M Veterinary Medical Diagnostic Laboratory, Canyon, TX, USA
| | - Amy K Swinford
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, TX, USA
| | - Kiril M Dimitrov
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, TX, USA
| | - Keith Poulsen
- Wisconsin Veterinary Diagnostic Laboratory, University of Wisconsin-Madison, Madison, WI, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), University of Tokyo, Tokyo, Japan.
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
| |
Collapse
|
16
|
Zapatero-Belinchón FJ, Kumar P, Ott M, Schwartz O, Sigal A. Understanding emerging and re-emerging viruses to facilitate pandemic preparedness. Nat Microbiol 2024; 9:2208-2211. [PMID: 39198691 DOI: 10.1038/s41564-024-01789-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
Affiliation(s)
| | - Priti Kumar
- Yale University School of Medicine, New Haven, CT, USA.
| | - Melanie Ott
- Gladstone Institute of Virology, University of California, San Francisco, San Francisco, CA, USA.
| | | | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa.
| |
Collapse
|
17
|
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
|
18
|
Matoušková M, Plachý J, Kučerová D, Pecnová Ľ, Reinišová M, Geryk J, Karafiát V, Hron T, Hejnar J. Rapid adaptive evolution of avian leukosis virus subgroup J in response to biotechnologically induced host resistance. PLoS Pathog 2024; 20:e1012468. [PMID: 39146367 PMCID: PMC11349186 DOI: 10.1371/journal.ppat.1012468] [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: 03/14/2024] [Revised: 08/27/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024] Open
Abstract
Genetic editing of the germline using CRISPR/Cas9 technology has made it possible to alter livestock traits, including the creation of resistance to viral diseases. However, virus adaptability could present a major obstacle in this effort. Recently, chickens resistant to avian leukosis virus subgroup J (ALV-J) were developed by deleting a single amino acid, W38, within the ALV-J receptor NHE1 using CRISPR/Cas9 genome editing. This resistance was confirmed both in vitro and in vivo. In vitro resistance of W38-/- chicken embryonic fibroblasts to all tested ALV-J strains was shown. To investigate the capacity of ALV-J for further adaptation, we used a retrovirus reporter-based assay to select adapted ALV-J variants. We assumed that adaptive mutations overcoming the cellular resistance would occur within the envelope protein. In accordance with this assumption, we isolated and sequenced numerous adapted virus variants and found within their envelope genes eight independent single nucleotide substitutions. To confirm the adaptive capacity of these substitutions, we introduced them into the original retrovirus reporter. All eight variants replicated effectively in W38-/- chicken embryonic fibroblasts in vitro while in vivo, W38-/- chickens were sensitive to tumor induction by two of the variants. Importantly, receptor alleles with more extensive modifications have remained resistant to the virus. These results demonstrate an important strategy in livestock genome engineering towards antivirus resistance and illustrate that cellular resistance induced by minor receptor modifications can be overcome by adapted virus variants. We conclude that more complex editing will be necessary to attain robust resistance.
Collapse
Affiliation(s)
- Magda Matoušková
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Plachý
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Dana Kučerová
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Ľubomíra Pecnová
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Markéta Reinišová
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Josef Geryk
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Vít Karafiát
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Hron
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Hejnar
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
19
|
Briggs K, Chrzastek K, Segovia K, Mo J, Kapczynski DR. Genetic insertion of mouse Myxovirus-resistance gene 1 increases innate resistance against both high and low pathogenic avian influenza virus by significantly decreasing replication in chicken DF1 cell line. Virology 2024; 595:110066. [PMID: 38574415 DOI: 10.1016/j.virol.2024.110066] [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: 01/11/2024] [Revised: 03/01/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024]
Abstract
Avian influenza virus (AIV) is a constant threat to animal health with recent global outbreaks resulting in the death of hundreds of millions of birds with spillover into mammals. Myxovirus-resistance (Mx) proteins are key mediators of the antiviral response that block virus replication. Mouse (Mu) Mx (Mx1) is a strong antiviral protein that interacts with the viral nucleoprotein to inhibit polymerase function. The ability of avian Mx1 to inhibit AIV is unclear. In these studies, Mu Mx1 was stably introduced into chicken DF1 cells to enhance the immune response against AIV. Following infection, titers of AIV were significantly decreased in cells expressing Mu Mx1. In addition, considerably less cytopathic effect (CPE) and matrix protein staining was observed in gene-edited cells expressing Mu Mx1, suggesting Mu Mx1 is broadly effective against multiple AIV subtypes. This work provides foundational studies for use of gene-editing to enhance innate disease resistance against AIV.
Collapse
Affiliation(s)
- Kelsey Briggs
- Exotic and Emerging Avian Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, Georgia
| | - Klaudia Chrzastek
- Exotic and Emerging Avian Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, Georgia
| | - Karen Segovia
- Exotic and Emerging Avian Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, Georgia
| | - Jongsuk Mo
- Exotic and Emerging Avian Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, Georgia
| | - Darrell R Kapczynski
- Exotic and Emerging Avian Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, Georgia.
| |
Collapse
|
20
|
Ledesma AV, Van Eenennaam AL. Global status of gene edited animals for agricultural applications. Vet J 2024; 305:106142. [PMID: 38788996 DOI: 10.1016/j.tvjl.2024.106142] [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: 01/09/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Gene editing (GnEd) involves using a site-directed nuclease to introduce a double-strand break (DSB) at a targeted location in the genome. A literature search was performed on the use of GnEd in animals for agricultural applications. Data was extracted from 212 peer-reviewed articles that described the production of at least one living animal employing GnEd technologies for agricultural purposes. The most common GnEd system reported was CRISPR/Cas9, and the most frequent type of edit was the unguided insertion or deletion resulting from the repair of the targeted DSB leading to a knock-out (KO) mutation. Animal groups included in the reviewed papers were ruminants (cattle, sheep, goats, n=63); monogastrics (pigs and rabbits, n=60); avian (chicken, duck, quail, n=17); aquatic (many species, n=65), and insects (honeybee, silkworm, n=7). Yield (32%), followed by reproduction (21%) and disease resistance (17%) were the most commonly targeted traits. Over half of the reviewed papers had Chinese first-authorship. Several countries, including Argentina, Australia, Brazil, Colombia and Japan, have adopted a regulatory policy that considers KO mutations introduced following GnEd DSB repair as akin to natural genetic variation, and therefore treat these GnEd animals analogously to those produced using conventional breeding. This approach has resulted in a non-GMO determination for a small number of GnEd food animal applications, including three species of GnEd KO fast-growing fish, (red sea bream, olive flounder and tiger pufferfish in Japan), KO fish and cattle in Argentina and Brazil, and porcine reproductive and respiratory syndrome (PRRS) virus disease-resistant KO pigs in Colombia.
Collapse
Affiliation(s)
- Alba V Ledesma
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | | |
Collapse
|
21
|
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
|
22
|
Xiao Y, Sheng ZM, Williams SL, Taubenberger JK. Two complete 1918 influenza A/H1N1 pandemic virus genomes characterized by next-generation sequencing using RNA isolated from formalin-fixed, paraffin-embedded autopsy lung tissue samples along with evidence of secondary bacterial co-infection. mBio 2024; 15:e0321823. [PMID: 38349163 PMCID: PMC10936189 DOI: 10.1128/mbio.03218-23] [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: 11/28/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024] Open
Abstract
The 1918 influenza pandemic was the most devastating respiratory pandemic in modern human history, with 50-100 million deaths worldwide. Here, we characterized the complete genomes of influenza A virus (IAV) from two fatal cases during the fall wave of 1918 influenza A (H1N1) pandemic in the United States, one from Walter Reed Army Hospital in Washington, DC, and the other from Camp Jackson, SC. The two complete IAV genomes were obtained by combining Illumina deep sequencing data from both total RNA and influenza viral genome-enriched libraries along with Sanger sequencing data from PCR across the sequencing gaps. This study confirms the previously reported 1918 IAV genomes and increases the total number of available complete or near-complete influenza viral genomes of the 1918 pandemic from four to six. Sequence comparisons among them confirm that the genomes of the 1918 pandemic virus were highly conserved during the main wave of the pandemic with geographic separation in North America and Europe. Metagenomic analyses revealed bacterial co-infections in both cases. Interestingly, in the Washington, DC, case, evidence is presented of the first reported Rhodococcus-influenza virus co-infection. IMPORTANCE This study applied modern molecular biotechnology and high-throughput sequencing to formalin-fixed, paraffin-embedded autopsy lung samples from two fatal cases during the fall wave of the 1918 influenza A (H1N1) pandemic in the United States. Complete influenza genomes were obtained from both cases, which increases the total number of available complete or near-complete influenza genomes of the 1918 pandemic virus from four to six. Sequence analysis confirms that the 1918 pandemic virus was highly conserved during the main wave of the pandemic with geographic separation in North America and Europe. Metagenomic analyses revealed bacterial co-infections in both cases, including the first reported evidence of Rhodococcus-influenza co-infection. Overall, this study offers a detailed view at the molecular level of the very limited samples from the most devastating influenza pandemic in modern human history.
Collapse
Affiliation(s)
- Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie L. Williams
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
23
|
Jibrilla M, Raji H, Okeke MI. Survey of attitude to human genome modification in Nigeria. J Community Genet 2024; 15:1-11. [PMID: 37995060 PMCID: PMC10857991 DOI: 10.1007/s12687-023-00689-1] [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: 08/03/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023] Open
Abstract
Gene editing and mitochondrial replacement therapy (MRT) are biotechnologies used to modify the host nuclear and mitochondrial DNA, respectively. Gene editing is the modification of a region of the host genome using site-specific nucleases, in particular the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system. Heritable and somatic genome editing (HGE and SGE) are used in gene therapy. MRT is a technique used to substitute the defective mitochondria in the recipient embryo with a female donor healthy mitochondrion in order to prevent the inheritance of mothers' defective mitochondria resulting in the change of mitochondria of the entire generation to come. To evaluate the perception of the Nigerian citizens on human genome modification, two survey forms were created and distributed in-person and majorly online. There was a total of 268 responses, 188 from the public and 80 from health workers and bio-scientists. The results showed poor knowledge about gene editing and MRT by the Nigerian public, but its use to prevent and cure inherited diseases was supported. Morality and religion have great influence on the attitude of Nigerians towards genome modification, but the influence of religion and morality is not unequivocal. Multiple regression analysis of Nigerian public responses shows that gender (females), age (19-30 years), monthly income (NGN 0 to 30,000), and level of education (tertiary) are significantly associated with approval of human genome editing, but the survey of health workers and bio-scientists shows no significant association except for females who approve and Muslims who disapprove of human genome editing.
Collapse
Affiliation(s)
- Maryam Jibrilla
- Department of Natural and Environmental Sciences, Biomedical Science Concentration, American University of Nigeria, 98 Lamido Zubairu Way, PMB, Yola, 2250, Adamawa State, Nigeria
| | - Hayatu Raji
- Department of Natural and Environmental Sciences, Biomedical Science Concentration, American University of Nigeria, 98 Lamido Zubairu Way, PMB, Yola, 2250, Adamawa State, Nigeria
| | - Malachy Ifeanyi Okeke
- Department of Natural and Environmental Sciences, Biomedical Science Concentration, American University of Nigeria, 98 Lamido Zubairu Way, PMB, Yola, 2250, Adamawa State, Nigeria.
| |
Collapse
|
24
|
Kwon T, Artiaga BL, McDowell CD, Whitworth KM, Wells KD, Prather RS, Delhon G, Cigan M, White SN, Retallick J, Gaudreault NN, Morozov I, Richt JA. Gene editing of pigs to control influenza A virus infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575771. [PMID: 38293027 PMCID: PMC10827075 DOI: 10.1101/2024.01.15.575771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Proteolytic activation of the hemagglutinin (HA) glycoprotein by host cellular proteases is pivotal for influenza A virus (IAV) infectivity. Highly pathogenic avian influenza viruses possess the multibasic cleavage site of the HA which is cleaved by ubiquitous proteases, such as furin; in contrast, the monobasic HA motif is recognized and activated by trypsin-like proteases, such as the transmembrane serine protease 2 (TMPRSS2). Here, we aimed to determine the effects of TMPRSS2 on the replication of pandemic H1N1 and H3N2 subtype IAVs in the natural host, the pig. The use of the CRISPR/Cas 9 system led to the establishment of homozygous gene edited (GE) TMPRSS2 knockout (KO) pigs. Delayed IAV replication was demonstrated in primary respiratory cells of KO pigs in vitro. IAV infection in vivo resulted in significant reduction of virus shedding in the upper respiratory tract, and lower virus titers and pathological lesions in the lower respiratory tract of TMPRSS2 KO pigs as compared to WT pigs. Our findings could support the commercial use of GE pigs to minimize (i) the economic losses caused by IAV infection in pigs, and (ii) the emergence of novel IAVs with pandemic potential through genetic reassortment in the "mixing vessel", the pig.
Collapse
Affiliation(s)
- Taeyong Kwon
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Bianca L. Artiaga
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Chester D. McDowell
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Kristin M. Whitworth
- Division of Animal Science & National Swine Resource and Research Center, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, Columbia, MO 65211, USA
| | - Kevin D. Wells
- Division of Animal Science & National Swine Resource and Research Center, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, Columbia, MO 65211, USA
| | - Randall S. Prather
- Division of Animal Science & National Swine Resource and Research Center, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, Columbia, MO 65211, USA
| | - Gustavo Delhon
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | | | | | - Jamie Retallick
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Igor Morozov
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
25
|
Brüssow H. Avian influenza virus cross-infections as test case for pandemic preparedness: From epidemiological hazard models to sequence-based early viral warning systems. Microb Biotechnol 2024; 17:e14389. [PMID: 38227348 PMCID: PMC10832514 DOI: 10.1111/1751-7915.14389] [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: 10/15/2023] [Revised: 11/17/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024] Open
Abstract
Pandemic preparedness starts with an early warning system of viruses with a pandemic potential. Based on information collected in a multitude of surveys, hazard models were developed identifying influenza viruses presenting a pandemic threat. Scores are attributed for 10 viral traits by expert panels which identified avian influenza viruses (AIV) belonging to subtypes H7N9 and H5N1 as representing the greatest pandemic risk. In 2013, more than 100 human cases infected with AIV H7N9 were observed in China. Case fatality rate (CFR) was high (27%), but the human-to-human transmission rate was low and by serological evidence H7N9 did not spread widely. Nevertheless, until 2019 more than 1500 H7N9 patients were identified characterized by a high CFR of 39%. Serology demonstrated that mild infections with H7N9 were widespread. In 2003, more than 400 people experienced AIV H7N7 cross-infection causing mainly conjunctivitis during a large poultry epidemic in The Netherlands. Between 1996 and 2019, a total of 881 human infections with avian H5N1 viruses were documented showing a CFR of 52%. Outbreaks were centred on South East Asia and showed close associations with epizootics in poultry. Mutations predisposing to human cross-infections were identified in the haemagglutinin (HA) and the RNA polymerase subunit PB2 of human H7N9 isolates. Human H5N1 isolates showed mutations in the receptor binding domain of HA and transmission in mammals could be obtained by as few as four additional aa changes introduced experimentally. Researchers have defined viral point mutations in HA, PB2 and the nucleoprotein NP that allowed AIV to cross the species barrier to mammals with respect to receptor recognition, RNA replication and escape from innate immunity respectively. Based on this insight a sequence-based early warning system for AIV preadapted to human transmission could be envisioned. Mink farms and live poultry markets are prime targets for such sequencing efforts.
Collapse
Affiliation(s)
- Harald Brüssow
- Division of Animal and Human Health Engineering, Department of BiosystemsKU LeuvenLeuvenBelgium
| |
Collapse
|