1
|
Li C, Culhane MR, Schroeder DC, Cheeran MCJ, Galina Pantoja L, Jansen ML, Torremorell M. Quantifying the impact of vaccination on transmission and diversity of influenza A variants in pigs. J Virol 2024; 98:e0124524. [PMID: 39530665 DOI: 10.1128/jvi.01245-24] [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: 07/22/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Global evolutionary dynamics of influenza A virus (IAV) are fundamentally driven by the extent of virus diversity generated, transmitted, and shaped in individual hosts. How vaccination affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs is unknown. To evaluate the effect of vaccination on the transmission of genetically distinct IAV variants and their diversity after transmission in pigs, we examined the whole genome of IAV recovered from the nasal cavities of pigs vaccinated with different influenza immunization regimens after being infected simultaneously by H1N1 and H3N2 IAVs using a seeder pig model. We found that the seeder pigs harbored more diversified virus populations than the contact pigs. Among contact pigs, H3N2 and H1N1 viruses recovered from pigs vaccinated with a single dose of an unmatched modified live vaccine generally accumulated more extensive genetic mutations than non-vaccinated pigs. Furthermore, the non-sterilizing immunity elicited by the single-dose-modified live vaccine may have exerted positive selection on H1 antigenic regions as we detected significantly higher nonsynonymous but lower synonymous evolutionary rates in H1 antigenic regions than non-antigenic regions. In addition, we observed that the vaccinated pigs shared significantly less proportion of H3N2 variants with seeder pigs than unvaccinated pigs. These results indicated that vaccination might reduce the impact of transmitted influenza variants on the overall diversity of IAV populations harbored in recipient pigs and that within-host genetic selection of IAV is more likely to occur in pigs vaccinated with improperly matched vaccines.IMPORTANCEUnderstanding how vaccination shapes the diversity of influenza variants that transmit and propagate among pigs is essential for designing effective IAV surveillance and control programs. Current knowledge about the transmission of IAV variants has primarily been explored in humans during natural infection. However, how immunity elicited by improperly matched vaccines affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs at the whole-genome level is unknown. We analyzed IAV sequences from samples collected daily from experimentally infected pigs vaccinated with various protocols in a field-represented IAV co-infection model. We found that vaccine-induced non-sterilizing immunity might promote genetic variation on the IAV genome and drive positive selection at antigenic sites during infection. In addition, a smaller proportion of H3N2 viral variants were shared between seeder pigs and vaccinated pigs, suggesting the influence of vaccination on shaping the virus genomic diversity in recipient pigs during the transmission events.
Collapse
Affiliation(s)
- Chong Li
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Marie R Culhane
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Declan C Schroeder
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Maxim C-J Cheeran
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | | | | | | |
Collapse
|
2
|
M Hassen B, Rashedy SH, Mostafa A, Mahrous N, Nafie MS, Elebeedy D, Abdel Azeiz AZ. Identification of potential antiviral compounds from Egyptian marine algae against influenza A virus. Nat Prod Res 2024; 38:4411-4418. [PMID: 37990847 DOI: 10.1080/14786419.2023.2284865] [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: 06/06/2023] [Revised: 11/01/2023] [Accepted: 11/12/2023] [Indexed: 11/23/2023]
Abstract
Influenza is a contagious viral infection of the respiratory tract, affecting nearly 10% of the world's population, each year. The aim of this study was to extract and identify antiviral compounds against the influenza-A virus (H1N1) from different species of Egyptian marine algae. Three samples of marine macroalgae species were extracted and the antiviral activity of the extracts were tested on Madin Darby Canine Kidney cells. The bioactive compounds present in the most active fractions were identified using gas chromatography-mass spectrometry (GC-MS), then the binding potentials of the identified compounds were examined towards neuraminidase (NA) of the influenza-A virus using molecular docking. The methanolic extract of Sargassum aquifolium showed promising in-vitro antiviral activity with a selectivity index (SI) value of 101. The GC-MS analysis showed twelve compounds and the molecular docking analysis found that tetradecanoic acid showed the strongest binding affinities towards the NA enzyme.
Collapse
Affiliation(s)
- Bassel M Hassen
- College of Biotechnology, Misr University for Science and Technology (MUST), 6th of October, Egypt
| | - Sarah H Rashedy
- National Institute of Oceanography and Fisheries (NIOF), Cairo, Egypt
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Noura Mahrous
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Mohamed S Nafie
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Dalia Elebeedy
- College of Biotechnology, Misr University for Science and Technology (MUST), 6th of October, Egypt
| | - A Z Abdel Azeiz
- College of Biotechnology, Misr University for Science and Technology (MUST), 6th of October, Egypt
| |
Collapse
|
3
|
Pauciullo S, Zulian V, La Frazia S, Paci P, Garbuglia AR. Spillover: Mechanisms, Genetic Barriers, and the Role of Reservoirs in Emerging Pathogens. Microorganisms 2024; 12:2191. [PMID: 39597581 PMCID: PMC11596118 DOI: 10.3390/microorganisms12112191] [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: 09/20/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Viral spillover represents the transmission of pathogen viruses from one species to another that can give rise to an outbreak. It is a critical concept that has gained increasing attention, particularly after the SARS-CoV-2 pandemic. However, the term is often used inaccurately to describe events that do not meet the true definition of spillover. This review aims to clarify the proper use of the term and provides a detailed analysis of the mechanisms driving zoonotic spillover, with a focus on the genetic and environmental factors that enable viruses to adapt to new hosts. Key topics include viral genetic variability in reservoir species, biological barriers to cross-species transmission, and the factors that influence viral adaptation and spread in novel hosts. The review also examines the role of evolutionary processes such as mutation and epistasis, alongside ecological conditions that facilitate the emergence of new pathogens. Ultimately, it underscores the need for more accurate predictive models and improved surveillance to better anticipate and mitigate future spillover events.
Collapse
Affiliation(s)
- Silvia Pauciullo
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.)
| | - Verdiana Zulian
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.)
| | - Simone La Frazia
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy;
| | - Paola Paci
- Department of Computer, Control, and Management Engineering “A. Ruberti” (DIAG), Sapienza University of Rome, 00185 Rome, Italy;
| | - Anna Rosa Garbuglia
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.)
| |
Collapse
|
4
|
Shi YT, Harris JD, Martin MA, Koelle K. Transmission Bottleneck Size Estimation from De Novo Viral Genetic Variation. Mol Biol Evol 2024; 41:msad286. [PMID: 38158742 PMCID: PMC10798134 DOI: 10.1093/molbev/msad286] [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: 08/14/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
Sequencing of viral infections has become increasingly common over the last decade. Deep sequencing data in particular have proven useful in characterizing the roles that genetic drift and natural selection play in shaping within-host viral populations. They have also been used to estimate transmission bottleneck sizes from identified donor-recipient pairs. These bottleneck sizes quantify the number of viral particles that establish genetic lineages in the recipient host and are important to estimate due to their impact on viral evolution. Current approaches for estimating bottleneck sizes exclusively consider the subset of viral sites that are observed as polymorphic in the donor individual. However, these approaches have the potential to substantially underestimate true transmission bottleneck sizes. Here, we present a new statistical approach for instead estimating bottleneck sizes using patterns of viral genetic variation that arise de novo within a recipient individual. Specifically, our approach makes use of the number of clonal viral variants observed in a transmission pair, defined as the number of viral sites that are monomorphic in both the donor and the recipient but carry different alleles. We first test our approach on a simulated dataset and then apply it to both influenza A virus sequence data and SARS-CoV-2 sequence data from identified transmission pairs. Our results confirm the existence of extremely tight transmission bottlenecks for these 2 respiratory viruses.
Collapse
Affiliation(s)
| | | | - Michael A Martin
- Department of Biology, Emory University, Atlanta, GA, USA
- Graduate Program in Population Biology, Ecology, and Evolution, Emory University, Atlanta, GA, USA
| | - Katia Koelle
- Department of Biology, Emory University, Atlanta, GA, USA
- Emory Center of Excellence for Influenza Research and Response (CEIRR), Atlanta, GA, USA
| |
Collapse
|
5
|
Zhong H, Wu M, Sonne C, Lam SS, Kwong RW, Jiang Y, Zhao X, Sun X, Zhang X, Li C, Li Y, Qu G, Jiang F, Shi H, Ji R, Ren H. The hidden risk of microplastic-associated pathogens in aquatic environments. ECO-ENVIRONMENT & HEALTH 2023; 2:142-151. [PMID: 38074987 PMCID: PMC10702891 DOI: 10.1016/j.eehl.2023.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/01/2023] [Accepted: 07/10/2023] [Indexed: 06/16/2024]
Abstract
Increasing studies of plastisphere have raised public concern about microplastics (MPs) as vectors for pathogens, especially in aquatic environments. However, the extent to which pathogens affect human health through MPs remains unclear, as controversies persist regarding the distinct pathogen colonization on MPs as well as the transmission routes and infection probability of MP-associated pathogens from water to humans. In this review, we critically discuss whether and how pathogens approach humans via MPs, shedding light on the potential health risks involved. Drawing on cutting-edge multidisciplinary research, we show that some MPs may facilitate the growth and long-range transmission of specific pathogens in aquatic environments, ultimately increasing the risk of infection in humans. We identify MP- and pathogen-rich settings, such as wastewater treatment plants, aquaculture farms, and swimming pools, as possible sites for human exposure to MP-associated pathogens. This review emphasizes the need for further research and targeted interventions to better understand and mitigate the potential health risks associated with MP-mediated pathogen transmission.
Collapse
Affiliation(s)
- Huan Zhong
- School of Environment, Nanjing University, State Key Laboratory of Pollution Control and Resource Reuse, Nanjing 210023, China
| | - Mengjie Wu
- School of Environment, Nanjing University, State Key Laboratory of Pollution Control and Resource Reuse, Nanjing 210023, China
| | - Christian Sonne
- Department of Bioscience, Arctic Research Centre, Aarhus University, Roskilde, Denmark
| | - Su Shiung Lam
- Higher Institution Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries (AKUATROP), Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
- University Centre for Research and Development, Department of Chemistry, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Raymond W.M. Kwong
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Yuelu Jiang
- Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| | - Xiaoli Zhao
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Xuemei Sun
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Xuxiang Zhang
- School of Environment, Nanjing University, State Key Laboratory of Pollution Control and Resource Reuse, Nanjing 210023, China
| | - Chengjun Li
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health (HUST), Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Feng Jiang
- School of Environmental Science and Engineering, Guangdong Provincial Key Lab of Environmental Pollution Control and Remediation Technology, Sun Yat-sen University, Guangzhou 510275, China
| | - Huahong Shi
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200241, China
| | - Rong Ji
- School of Environment, Nanjing University, State Key Laboratory of Pollution Control and Resource Reuse, Nanjing 210023, China
| | - Hongqiang Ren
- School of Environment, Nanjing University, State Key Laboratory of Pollution Control and Resource Reuse, Nanjing 210023, China
| |
Collapse
|
6
|
Shi T, Harris JD, Martin MA, Koelle K. Transmission bottleneck size estimation from de novo viral genetic variation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553219. [PMID: 37645981 PMCID: PMC10462048 DOI: 10.1101/2023.08.14.553219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Sequencing of viral infections has become increasingly common over the last decade. Deep sequencing data in particular have proven useful in characterizing the roles that genetic drift and natural selection play in shaping within-host viral populations. They have also been used to estimate transmission bottleneck sizes from identified donor-recipient pairs. These bottleneck sizes quantify the number of viral particles that establish genetic lineages in the recipient host and are important to estimate due to their impact on viral evolution. Current approaches for estimating bottleneck sizes exclusively consider the subset of viral sites that are observed as polymorphic in the donor individual. However, allele frequencies can change dramatically over the course of an individual's infection, such that sites that are polymorphic in the donor at the time of transmission may not be polymorphic in the donor at the time of sampling and allele frequencies at donor-polymorphic sites may change dramatically over the course of a recipient's infection. Because of this, transmission bottleneck sizes estimated using allele frequencies observed at a donor's polymorphic sites may be considerable underestimates of true bottleneck sizes. Here, we present a new statistical approach for instead estimating bottleneck sizes using patterns of viral genetic variation that arose de novo within a recipient individual. Specifically, our approach makes use of the number of clonal viral variants observed in a transmission pair, defined as the number of viral sites that are monomorphic in both the donor and the recipient but carry different alleles. We first test our approach on a simulated dataset and then apply it to both influenza A virus sequence data and SARS-CoV-2 sequence data from identified transmission pairs. Our results confirm the existence of extremely tight transmission bottlenecks for these two respiratory viruses, using an approach that does not tend to underestimate transmission bottleneck sizes.
Collapse
Affiliation(s)
- Teresa Shi
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Jeremy D. Harris
- Department of Biology, Emory University, Atlanta, GA, USA
- Department of Mathematics, Rose-Hulman Institute of Technology, Terre Haute, IN, USA
| | - Michael A. Martin
- Department of Biology, Emory University, Atlanta, GA, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Graduate Program in Population Biology, Ecology, and Evolution, Emory University, Atlanta, GA, USA
| | - Katia Koelle
- Department of Biology, Emory University, Atlanta, GA, USA
- Emory Center of Excellence for Influenza Research and Response (CEIRR), Atlanta GA, USA
| |
Collapse
|
7
|
Pedrazzoli P, Lasagna A, Cassaniti I, Piralla A, Squeri A, Bruno R, Sacchi P, Baldanti F, Di Maio M, Beretta GD, Cinieri S, Silvestris N. Vaccination for seasonal flu, pneumococcal infection, and SARS-CoV-2 in patients with solid tumors: recommendations of the Associazione Italiana di Oncologia Medica (AIOM). ESMO Open 2023; 8:101215. [PMID: 37104930 PMCID: PMC10067463 DOI: 10.1016/j.esmoop.2023.101215] [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: 03/03/2023] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Patients with cancer have a well-known and higher risk of vaccine-preventable diseases (VPDs). VPDs may cause severe complications in this setting due to the immune system impairment, malnutrition and oncological treatments. Despite this evidence, vaccination rates are inadequate. The Italian Association of Medical Oncology (AIOM) has been involved in vaccination awareness since 2014. Based on a careful review of the available data about the immunogenicity, effectiveness and safety of flu, pneumococcal and anti-SARS-CoV-2 vaccines, we report the recommendations of the Associazione Italiana di Oncologia Medica about these vaccinations in adult patients with solid tumors. AIOM recommends comprehensive education on the issue of VPDs. We believe that a multidisciplinary care model may improve the vaccination coverage in immunocompromised patients. Continued surveillance, implementation of preventive practices and future well-designed immunological prospective studies are essential for a better management of our patients with cancer.
Collapse
Affiliation(s)
- P Pedrazzoli
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy; Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A Lasagna
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - I Cassaniti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A Piralla
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - A Squeri
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy; School of Specialization in Medical Oncology, University of Messina, Messina, Italy
| | - R Bruno
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Division of Infectious Diseases I, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - P Sacchi
- Division of Infectious Diseases I, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - F Baldanti
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - M Di Maio
- Department of Oncology, University of Turin, Division of Medical Oncology, Ordine Mauriziano Hospital, Turin, Italy
| | - G D Beretta
- Medical Oncology Unit, Santo Spirito Hospital, Pescara, Italy
| | - S Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| | - N Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| |
Collapse
|
8
|
High-throughput droplet-based analysis of influenza A virus genetic reassortment by single-virus RNA sequencing. Proc Natl Acad Sci U S A 2023; 120:e2211098120. [PMID: 36730204 PMCID: PMC9963642 DOI: 10.1073/pnas.2211098120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The segmented RNA genome of influenza A viruses (IAVs) enables viral evolution through genetic reassortment after multiple IAVs coinfect the same cell, leading to viruses harboring combinations of eight genomic segments from distinct parental viruses. Existing data indicate that reassortant genotypes are not equiprobable; however, the low throughput of available virology techniques does not allow quantitative analysis. Here, we have developed a high-throughput single-cell droplet microfluidic system allowing encapsulation of IAV-infected cells, each cell being infected by a single progeny virion resulting from a coinfection process. Customized barcoded primers for targeted viral RNA sequencing enabled the analysis of 18,422 viral genotypes resulting from coinfection with two circulating human H1N1pdm09 and H3N2 IAVs. Results were highly reproducible, confirmed that genetic reassortment is far from random, and allowed accurate quantification of reassortants including rare events. In total, 159 out of the 254 possible reassortant genotypes were observed but with widely varied prevalence (from 0.038 to 8.45%). In cells where eight segments were detected, all 112 possible pairwise combinations of segments were observed. The inclusion of data from single cells where less than eight segments were detected allowed analysis of pairwise cosegregation between segments with very high confidence. Direct coupling analysis accurately predicted the fraction of pairwise segments and full genotypes. Overall, our results indicate that a large proportion of reassortant genotypes can emerge upon coinfection and be detected over a wide range of frequencies, highlighting the power of our tool for systematic and exhaustive monitoring of the reassortment potential of IAVs.
Collapse
|
9
|
Wang M, Li H, Liu S, Ge L, Muhmood A, Liu D, Gan F, Liu Y, Chen X, Huang K. Lipopolysaccharide aggravates canine influenza a (H3N2) virus infection and lung damage via mTOR/autophagy in vivo and in vitro. Food Chem Toxicol 2023; 172:113597. [PMID: 36596444 DOI: 10.1016/j.fct.2022.113597] [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: 07/26/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
Influenza A (H3N2) accounts for the majority of influenza worldwide and continues to challenge human health. Disturbance in the gut microbiota caused by many diseases leads to increased production of lipopolysaccharide (LPS), and LPS induces sepsis and conditions associated with local or systemic inflammation. However, to date, little attention has been paid to the potential impact of LPS on influenza A (H3N2) infection and the potential mechanism. Hence, in this study we used canine influenza A (H3N2) virus (CIV) as a model of influenza A virus to investigate the effect of low-dose of LPS on CIV replication and lung damage and explore the underlying mechanism in mice and A549 and HPAEpiC cells. The results showed that LPS (25 μg/kg) increased CIV infection and lung damage in mice, as indicated by pulmonary virus titer, viral NP levels, lung index, and pulmonary histopathology. LPS (1 μg/ml) also increased CIV replication in A549 cells as indicated by the above same parameters. Furthermore, low doses of LPS reduced CIV-induced p-mTOR protein expression and enhanced CIV-induced autophagy-related mRNA/protein expressions in vivo and in vitro. In addition, the use of the mTOR activator, MHY1485, reversed CIV-induced autophagy and CIV replication in A549 and HPAEpiC cells, respectively. siATG5 alleviated CIV replication exacerbated by LPS in the two lines. In conclusion, LPS aggravates CIV infection and lung damage via mTOR/autophagy.
Collapse
Affiliation(s)
- Mengmeng Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Haolei Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Lei Ge
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Azhar Muhmood
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Fang Gan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Yunhuan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
10
|
Bendall EE, Callear AP, Getz A, Goforth K, Edwards D, Monto AS, Martin ET, Lauring AS. Rapid transmission and tight bottlenecks constrain the evolution of highly transmissible SARS-CoV-2 variants. Nat Commun 2023; 14:272. [PMID: 36650162 PMCID: PMC9844183 DOI: 10.1038/s41467-023-36001-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Transmission bottlenecks limit the spread of novel mutations and reduce the efficiency of selection along a transmission chain. While increased force of infection, receptor binding, or immune evasion may influence bottleneck size, the relationship between transmissibility and the transmission bottleneck is unclear. Here we compare the transmission bottleneck of non-VOC SARS-CoV-2 lineages to those of Alpha, Delta, and Omicron. We sequenced viruses from 168 individuals in 65 households. Most virus populations had 0-1 single nucleotide variants (iSNV). From 64 transmission pairs with detectable iSNV, we identify a per clade bottleneck of 1 (95% CI 1-1) for Alpha, Delta, and Omicron and 2 (95% CI 2-2) for non-VOC. These tight bottlenecks reflect the low diversity at the time of transmission, which may be more pronounced in rapidly transmissible variants. Tight bottlenecks will limit the development of highly mutated VOC in transmission chains, adding to the evidence that selection over prolonged infections may drive their evolution.
Collapse
Affiliation(s)
- Emily E Bendall
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Amy P Callear
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Getz
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Kendra Goforth
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Drew Edwards
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Arnold S Monto
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Emily T Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Adam S Lauring
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA.
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
11
|
Bendall EE, Callear A, Getz A, Goforth K, Edwards D, Monto AS, Martin ET, Lauring AS. Rapid transmission and tight bottlenecks constrain the evolution of highly transmissible SARS-CoV-2 variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.10.12.511991. [PMID: 36263068 PMCID: PMC9580385 DOI: 10.1101/2022.10.12.511991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Transmission bottlenecks limit the spread of novel mutations and reduce the efficiency of natural selection along a transmission chain. Many viruses exhibit tight bottlenecks, and studies of early SARS-CoV-2 lineages identified a bottleneck of 1-3 infectious virions. While increased force of infection, host receptor binding, or immune evasion may influence bottleneck size, the relationship between transmissibility and the transmission bottleneck is unclear. Here, we compare the transmission bottleneck of non-variant-of-concern (non-VOC) SARS-CoV-2 lineages to those of the Alpha, Delta, and Omicron variants. We sequenced viruses from 168 individuals in 65 multiply infected households in duplicate to high depth of coverage. In 110 specimens collected close to the time of transmission, within-host diversity was extremely low. At a 2% frequency threshold, 51% had no intrahost single nucleotide variants (iSNV), and 42% had 1-2 iSNV. In 64 possible transmission pairs with detectable iSNV, we identified a bottleneck of 1 infectious virion (95% CI 1-1) for Alpha, Delta, and Omicron lineages and 2 (95% CI 2-2) in non-VOC lineages. The latter was driven by a single iSNV shared in one non-VOC household. The tight transmission bottleneck in SARS-CoV-2 is due to low genetic diversity at the time of transmission, a relationship that may be more pronounced in rapidly transmissible variants. The tight bottlenecks identified here will limit the development of highly mutated VOC in typical transmission chains, adding to the evidence that selection over prolonged infections in immunocompromised patients may drive their evolution.
Collapse
Affiliation(s)
- Emily E. Bendall
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Callear
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Getz
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Kendra Goforth
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Drew Edwards
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Arnold S. Monto
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Adam S. Lauring
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
12
|
Neopane P, Nypaver J, Shrestha R, Beqaj S. Performance Evaluation of TaqMan SARS-CoV-2, Flu A/B, RSV RT-PCR Multiplex Assay for the Detection of Respiratory Viruses. Infect Drug Resist 2022; 15:5411-5423. [PMID: 36119638 PMCID: PMC9480588 DOI: 10.2147/idr.s373748] [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: 05/11/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To detect and differentiate co-infection with influenza and respiratory syncytial virus during the COVID pandemic, a rapid method that can detect multiple pathogens in a single test is a significant diagnostic advance to analyze the outcomes and clinical implications of co-infection. Therefore, we validated and evaluated the performance characteristics of TaqMan SARS-CoV-2, Flu A/B, RSV RT-PCR multiplex assay for the detection of SARS-CoV-2, Flu A/B, and RSV using nasopharyngeal and saliva samples. Materials and Methods The method validation was performed by using culture fluids of Influenza A virus (H3N2) (A/Wisconsin/67/2005), Influenza B virus (B/Virginia/ATCC4/2009), RSV A2 cpts-248, SARS-CoV-2 (USA-WA1/2020) and quantitative RNA controls of Influenza A virus (H1N1) strain A/PR/8/34 (VR-95DQ), RSV A2 (VR-1540DQ) and SARS-CoV-2 (MN908947.3 Wuhan-Hu-1) from ATCC and Zeptometrix, NY, USA. A total of 110 nasopharyngeal specimens and 70 saliva samples were used for the SARS-CoV-2 detection, and a total of 70 nasopharyngeal specimens were used for Influenza and RSV detection. Total RNA was extracted from all the samples and multiplex PCR was performed using TaqMan SARS-CoV-2, Flu A/B, RSV RT-PCR multiplex assay. The assay was used for SARS-CoV-2 variant (B.1.1.7_601443, B.1.617.1_1662307, P.1_792683, B.1.351_678597, B.1.1.529/BA.1). Results Validation controls showed accurate and precise results. The correlation study found the accuracy of 96.38 to 100% (95% CI) in nasopharyngeal and 94.87 to 100% (95% CI) in saliva for SARS-CoV-2 and 91.1 to 100% (95% CI) for both Influenza A/B and RSV. The diagnostic efficiency of this assay was not affected by SARS-CoV-2 variant, including Omicron. Conclusion The TaqMan SARS-CoV-2, Flu A/B, RSV RT-PCR multiplex assay is a rapid method to detect and differentiate SAR-CoV-2, Flu A and B, and RSV in nasopharyngeal and saliva samples. It has a significant role in the diagnosis and management of respiratory illnesses and the clinical implications of co-infection.
Collapse
Affiliation(s)
- Puja Neopane
- Patients Choice Laboratories, Indianapolis, IN, 46278, USA
| | - Jerome Nypaver
- Patients Choice Laboratories, Indianapolis, IN, 46278, USA
| | | | - Safedin Beqaj
- Patients Choice Laboratories, Indianapolis, IN, 46278, USA
| |
Collapse
|
13
|
Chan ER, Jones LD, Linger M, Kovach JD, Torres-Teran MM, Wertz A, Donskey CJ, Zimmerman PA. COVID-19 infection and transmission includes complex sequence diversity. PLoS Genet 2022; 18:e1010200. [PMID: 36074769 PMCID: PMC9455841 DOI: 10.1371/journal.pgen.1010200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/27/2022] [Indexed: 12/16/2022] Open
Abstract
SARS-CoV-2 whole genome sequencing has played an important role in documenting the emergence of polymorphisms in the viral genome and its continuing evolution during the COVID-19 pandemic. Here we present data from over 360 patients to characterize the complex sequence diversity of individual infections identified during multiple variant surges (e.g., Alpha and Delta). Across our survey, we observed significantly increasing SARS-CoV-2 sequence diversity during the pandemic and frequent occurrence of multiple biallelic sequence polymorphisms in all infections. This sequence polymorphism shows that SARS-CoV-2 infections are heterogeneous mixtures. Convention for reporting microbial pathogens guides investigators to report a majority consensus sequence. In our study, we found that this approach would under-report sequence variation in all samples tested. As we find that this sequence heterogeneity is efficiently transmitted from donors to recipients, our findings illustrate that infection complexity must be monitored and reported more completely to understand SARS-CoV-2 infection and transmission dynamics. Many of the nucleotide changes that would not be reported in a majority consensus sequence have now been observed as lineage defining SNPs in Omicron BA.1 and/or BA.2 variants. This suggests that minority alleles in earlier SARS-CoV-2 infections may play an important role in the continuing evolution of new variants of concern.
Collapse
Affiliation(s)
- Ernest R. Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Lucas D. Jones
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Marlin Linger
- The Center for Global Health & Diseases, Pathology Department, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jeffrey D. Kovach
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- The Center for Global Health & Diseases, Pathology Department, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Maria M. Torres-Teran
- Pathology Department, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Audric Wertz
- Biology Department, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Curtis J. Donskey
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Geriatric Research, Education, and Clinical Center, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States of America
| | - Peter A. Zimmerman
- The Center for Global Health & Diseases, Pathology Department, Case Western Reserve University, Cleveland, Ohio, United States of America
- Master of Public Health Program, Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
14
|
Bull MB, Gu H, Ma FNL, Perera LP, Poon LLM, Valkenburg SA. Next-generation T cell-activating vaccination increases influenza virus mutation prevalence. SCIENCE ADVANCES 2022; 8:eabl5209. [PMID: 35385318 PMCID: PMC8986104 DOI: 10.1126/sciadv.abl5209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/11/2022] [Indexed: 06/14/2023]
Abstract
To determine the potential for viral adaptation to T cell responses, we probed the full influenza virus genome by next-generation sequencing directly ex vivo from infected mice, in the context of an experimental T cell-based vaccine, an H5N1-based viral vectored vaccinia vaccine Wyeth/IL-15/5Flu, versus the current standard-of-care, seasonal inactivated influenza vaccine (IIV) and unvaccinated conditions. Wyeth/IL-15/5Flu vaccination was coincident with increased mutation incidence and frequency across the influenza genome; however, mutations were not enriched within T cell epitope regions, but high allele frequency mutations within conserved hemagglutinin stem regions and PB2 mammalian adaptive mutations arose. Depletion of CD4+ and CD8+ T cell subsets led to reduced frequency of mutants in vaccinated mice; therefore, vaccine-mediated T cell responses were important drivers of virus diversification. Our findings suggest that Wyeth/IL-15/5Flu does not generate T cell escape mutants but increases stochastic events for virus adaptation by stringent bottlenecks.
Collapse
Affiliation(s)
- Maireid B. Bull
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Haogao Gu
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Fionn N. L. Ma
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Liyanage P. Perera
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1374, USA
| | - Leo L. M. Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie A. Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Microbiology and Immunology, at The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Swine H1N1 Influenza Virus Variants with Enhanced Polymerase Activity and HA Stability Promote Airborne Transmission in Ferrets. J Virol 2022; 96:e0010022. [DOI: 10.1128/jvi.00100-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Diverse IAVs circulate in animals, yet few acquire the viral traits needed to start a human pandemic. A stabilized HA and mammalian-adapted polymerase have been shown to promote the adaptation of IAVs to humans and ferrets (the gold-standard model for IAV replication, pathogenicity, and transmissibility).
Collapse
|
16
|
Daniels RS, Galiano M, Ermetal B, Kwong J, Lau CS, Xiang Z, McCauley JW, Lo J. Temporal and Gene Reassortment Analysis of Influenza C Virus Outbreaks in Hong Kong, SAR, China. J Virol 2022; 96:e0192821. [PMID: 34787455 PMCID: PMC8826914 DOI: 10.1128/jvi.01928-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 11/29/2022] Open
Abstract
From 2014 to week 07/2020 the Centre for Health Protection in Hong Kong conducted screening for influenza C virus (ICV). A retrospective analysis of ICV detections to week 26/2019 revealed persistent low-level circulation with outbreaks occurring biennially in the winters of 2015 to 2016 and 2017 to 2018 (R. S. Daniels et al., J Virol 94:e01051-20, 2020, https://doi.org/10.1128/JVI.01051-20). Here, we report on an outbreak occurring in 2019 to 2020, reinforcing the observation of biennial seasonality in Hong Kong. All three outbreaks occurred in similar time frames, were subsequently dwarfed by seasonal epidemics of influenza types A and B, and were caused by similar proportions of C/Kanagawa/1/76 (K)-lineage and C/São Paulo/378/82 S1- and S2-sublineage viruses. Ongoing genetic drift was observed in all genes, with some evidence of amino acid substitution in the hemagglutinin-esterase-fusion (HEF) glycoprotein possibly associated with antigenic drift. A total of 61 ICV genomes covering the three outbreaks were analyzed for reassortment, and 9 different reassortant constellations were identified, 1 K-lineage, 4 S1-sublineage, and 4 S2-sublineage, with 6 of these being identified first in the 2019-1920 outbreak (2 S2-lineage and 4 S1-lineage). The roles that virus interference/enhancement, ICV persistent infection, genome evolution, and reassortment might play in the observed seasonality of ICV in Hong Kong are discussed. IMPORTANCE Influenza C virus (ICV) infection of humans is common, with the great majority of people being infected during childhood, though reinfection can occur throughout life. While infection normally results in "cold-like" symptoms, severe disease cases have been reported in recent years. However, knowledge of ICV is limited due to poor systematic surveillance and an inability to propagate the virus in large amounts in the laboratory. Following recent systematic surveillance in Hong Kong SAR, China, and direct ICV gene sequencing from clinical specimens, a 2-year cycle of disease outbreaks (epidemics) has been identified, with gene mixing playing a significant role in ICV evolution. Studies like those reported here are key to developing an understanding of the impact of influenza C virus infection in humans, notably where comorbidities exist and severe respiratory disease can develop.
Collapse
Affiliation(s)
- Rodney S. Daniels
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Monica Galiano
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Burcu Ermetal
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Jasmine Kwong
- Centre for Health Protection, Department of Health, Hong Kong SAR, China
| | - Chi S. Lau
- Centre for Health Protection, Department of Health, Hong Kong SAR, China
| | - Zheng Xiang
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - John W. McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Janice Lo
- Centre for Health Protection, Department of Health, Hong Kong SAR, China
| |
Collapse
|
17
|
Morales-Arce AY, Johri P, Jensen JD. Inferring the distribution of fitness effects in patient-sampled and experimental virus populations: two case studies. Heredity (Edinb) 2022; 128:79-87. [PMID: 34987185 PMCID: PMC8728706 DOI: 10.1038/s41437-021-00493-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/19/2022] Open
Abstract
We here propose an analysis pipeline for inferring the distribution of fitness effects (DFE) from either patient-sampled or experimentally-evolved viral populations, that explicitly accounts for non-Wright-Fisher and non-equilibrium population dynamics inherent to pathogens. We examine the performance of this approach via extensive power and performance analyses, and highlight two illustrative applications - one from an experimentally-passaged RNA virus, and the other from a clinically-sampled DNA virus. Finally, we discuss how such DFE inference may shed light on major research questions in virus evolution, ranging from a quantification of the population genetic processes governing genome size, to the role of Hill-Robertson interference in dictating adaptive outcomes, to the potential design of novel therapeutic approaches to eradicate within-patient viral populations via induced mutational meltdown.
Collapse
Affiliation(s)
- Ana Y Morales-Arce
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Parul Johri
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Jeffrey D Jensen
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
18
|
Li B, Deng A, Li K, Hu Y, Li Z, Shi Y, Xiong Q, Liu Z, Guo Q, Zou L, Zhang H, Zhang M, Ouyang F, Su J, Su W, Xu J, Lin H, Sun J, Peng J, Jiang H, Zhou P, Hu T, Luo M, Zhang Y, Zheng H, Xiao J, Liu T, Tan M, Che R, Zeng H, Zheng Z, Huang Y, Yu J, Yi L, Wu J, Chen J, Zhong H, Deng X, Kang M, Pybus OG, Hall M, Lythgoe KA, Li Y, Yuan J, He J, Lu J. Viral infection and transmission in a large, well-traced outbreak caused by the SARS-CoV-2 Delta variant. Nat Commun 2022; 13:460. [PMID: 35075154 PMCID: PMC8786931 DOI: 10.1038/s41467-022-28089-y] [Citation(s) in RCA: 217] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/04/2022] [Indexed: 12/30/2022] Open
Abstract
The SARS-CoV-2 Delta variant has spread rapidly worldwide. To provide data on its virological profile, we here report the first local transmission of Delta in mainland China. All 167 infections could be traced back to the first index case. Daily sequential PCR testing of quarantined individuals indicated that the viral loads of Delta infections, when they first become PCR-positive, were on average ~1000 times greater compared to lineage A/B infections during the first epidemic wave in China in early 2020, suggesting potentially faster viral replication and greater infectiousness of Delta during early infection. The estimated transmission bottleneck size of the Delta variant was generally narrow, with 1-3 virions in 29 donor-recipient transmission pairs. However, the transmission of minor iSNVs resulted in at least 3 of the 34 substitutions that were identified in the outbreak, highlighting the contribution of intra-host variants to population-level viral diversity during rapid spread.
Collapse
Affiliation(s)
- Baisheng Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Aiping Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Kuibiao Li
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Yao Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhencui Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yaling Shi
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Qianling Xiong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Zhe Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Qianfang Guo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lirong Zou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huan Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Meng Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Fangzhu Ouyang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Juan Su
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Wenzhe Su
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jing Xu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huifang Lin
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jing Sun
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jinju Peng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Huiming Jiang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Pingping Zhou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Ting Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Luo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yingtao Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huanying Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianpeng Xiao
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Tao Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Mingkai Tan
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Rongfei Che
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hanri Zeng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhonghua Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yushi Huang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianxiang Yu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lina Yi
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jie Wu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jingdiao Chen
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Haojie Zhong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiaoling Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Kang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | - Matthew Hall
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Katrina A Lythgoe
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Yan Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China. .,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jun Yuan
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
| | - Jianfeng He
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China. .,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jing Lu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China. .,Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China. .,Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Li B, Deng A, Li K, Hu Y, Li Z, Shi Y, Xiong Q, Liu Z, Guo Q, Zou L, Zhang H, Zhang M, Ouyang F, Su J, Su W, Xu J, Lin H, Sun J, Peng J, Jiang H, Zhou P, Hu T, Luo M, Zhang Y, Zheng H, Xiao J, Liu T, Tan M, Che R, Zeng H, Zheng Z, Huang Y, Yu J, Yi L, Wu J, Chen J, Zhong H, Deng X, Kang M, Pybus OG, Hall M, Lythgoe KA, Li Y, Yuan J, He J, Lu J. Viral infection and transmission in a large, well-traced outbreak caused by the SARS-CoV-2 Delta variant. Nat Commun 2022. [PMID: 35075154 DOI: 10.1101/2021.07.07.21260122] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
The SARS-CoV-2 Delta variant has spread rapidly worldwide. To provide data on its virological profile, we here report the first local transmission of Delta in mainland China. All 167 infections could be traced back to the first index case. Daily sequential PCR testing of quarantined individuals indicated that the viral loads of Delta infections, when they first become PCR-positive, were on average ~1000 times greater compared to lineage A/B infections during the first epidemic wave in China in early 2020, suggesting potentially faster viral replication and greater infectiousness of Delta during early infection. The estimated transmission bottleneck size of the Delta variant was generally narrow, with 1-3 virions in 29 donor-recipient transmission pairs. However, the transmission of minor iSNVs resulted in at least 3 of the 34 substitutions that were identified in the outbreak, highlighting the contribution of intra-host variants to population-level viral diversity during rapid spread.
Collapse
Affiliation(s)
- Baisheng Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Aiping Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Kuibiao Li
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Yao Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhencui Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yaling Shi
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Qianling Xiong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Zhe Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Qianfang Guo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lirong Zou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huan Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Meng Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Fangzhu Ouyang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Juan Su
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Wenzhe Su
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jing Xu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huifang Lin
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jing Sun
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jinju Peng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Huiming Jiang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Pingping Zhou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Ting Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Luo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yingtao Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huanying Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianpeng Xiao
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Tao Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Mingkai Tan
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Rongfei Che
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hanri Zeng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhonghua Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yushi Huang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianxiang Yu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lina Yi
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jie Wu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jingdiao Chen
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Haojie Zhong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiaoling Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Kang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | - Matthew Hall
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Katrina A Lythgoe
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Yan Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jun Yuan
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
| | - Jianfeng He
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jing Lu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Li B, Deng A, Li K, Hu Y, Li Z, Shi Y, Xiong Q, Liu Z, Guo Q, Zou L, Zhang H, Zhang M, Ouyang F, Su J, Su W, Xu J, Lin H, Sun J, Peng J, Jiang H, Zhou P, Hu T, Luo M, Zhang Y, Zheng H, Xiao J, Liu T, Tan M, Che R, Zeng H, Zheng Z, Huang Y, Yu J, Yi L, Wu J, Chen J, Zhong H, Deng X, Kang M, Pybus OG, Hall M, Lythgoe KA, Li Y, Yuan J, He J, Lu J. Viral infection and transmission in a large, well-traced outbreak caused by the SARS-CoV-2 Delta variant. Nat Commun 2022; 13:460. [PMID: 35075154 DOI: 10.21203/rs.3.rs-738164/v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/04/2022] [Indexed: 05/18/2023] Open
Abstract
The SARS-CoV-2 Delta variant has spread rapidly worldwide. To provide data on its virological profile, we here report the first local transmission of Delta in mainland China. All 167 infections could be traced back to the first index case. Daily sequential PCR testing of quarantined individuals indicated that the viral loads of Delta infections, when they first become PCR-positive, were on average ~1000 times greater compared to lineage A/B infections during the first epidemic wave in China in early 2020, suggesting potentially faster viral replication and greater infectiousness of Delta during early infection. The estimated transmission bottleneck size of the Delta variant was generally narrow, with 1-3 virions in 29 donor-recipient transmission pairs. However, the transmission of minor iSNVs resulted in at least 3 of the 34 substitutions that were identified in the outbreak, highlighting the contribution of intra-host variants to population-level viral diversity during rapid spread.
Collapse
Affiliation(s)
- Baisheng Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Aiping Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Kuibiao Li
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Yao Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhencui Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yaling Shi
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Qianling Xiong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Zhe Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Qianfang Guo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lirong Zou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huan Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Meng Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Fangzhu Ouyang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Juan Su
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Wenzhe Su
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jing Xu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huifang Lin
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jing Sun
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jinju Peng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Huiming Jiang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Pingping Zhou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Ting Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Luo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yingtao Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huanying Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianpeng Xiao
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Tao Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Mingkai Tan
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Rongfei Che
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hanri Zeng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhonghua Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yushi Huang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianxiang Yu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lina Yi
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jie Wu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jingdiao Chen
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Haojie Zhong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiaoling Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Kang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | - Matthew Hall
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Katrina A Lythgoe
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Yan Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jun Yuan
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
| | - Jianfeng He
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jing Lu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China.
| |
Collapse
|
21
|
Li B, Deng A, Li K, Hu Y, Li Z, Shi Y, Xiong Q, Liu Z, Guo Q, Zou L, Zhang H, Zhang M, Ouyang F, Su J, Su W, Xu J, Lin H, Sun J, Peng J, Jiang H, Zhou P, Hu T, Luo M, Zhang Y, Zheng H, Xiao J, Liu T, Tan M, Che R, Zeng H, Zheng Z, Huang Y, Yu J, Yi L, Wu J, Chen J, Zhong H, Deng X, Kang M, Pybus OG, Hall M, Lythgoe KA, Li Y, Yuan J, He J, Lu J. Viral infection and transmission in a large, well-traced outbreak caused by the SARS-CoV-2 Delta variant. Nat Commun 2022; 13:460. [PMID: 35075154 DOI: 10.1101/2021.1107.1107.21260122v21260122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/04/2022] [Indexed: 05/23/2023] Open
Abstract
The SARS-CoV-2 Delta variant has spread rapidly worldwide. To provide data on its virological profile, we here report the first local transmission of Delta in mainland China. All 167 infections could be traced back to the first index case. Daily sequential PCR testing of quarantined individuals indicated that the viral loads of Delta infections, when they first become PCR-positive, were on average ~1000 times greater compared to lineage A/B infections during the first epidemic wave in China in early 2020, suggesting potentially faster viral replication and greater infectiousness of Delta during early infection. The estimated transmission bottleneck size of the Delta variant was generally narrow, with 1-3 virions in 29 donor-recipient transmission pairs. However, the transmission of minor iSNVs resulted in at least 3 of the 34 substitutions that were identified in the outbreak, highlighting the contribution of intra-host variants to population-level viral diversity during rapid spread.
Collapse
Affiliation(s)
- Baisheng Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Aiping Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Kuibiao Li
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Yao Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhencui Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yaling Shi
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Qianling Xiong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Zhe Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Qianfang Guo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lirong Zou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huan Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Meng Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Fangzhu Ouyang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Juan Su
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Wenzhe Su
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jing Xu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huifang Lin
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jing Sun
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jinju Peng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Huiming Jiang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Pingping Zhou
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Ting Hu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Luo
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yingtao Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huanying Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianpeng Xiao
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Tao Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Mingkai Tan
- Guangzhou 8th People's Hospital, Guangzhou, Guangdong, China
| | - Rongfei Che
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hanri Zeng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhonghua Zheng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yushi Huang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jianxiang Yu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Lina Yi
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China
| | - Jie Wu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jingdiao Chen
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Haojie Zhong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiaoling Deng
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Min Kang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | - Matthew Hall
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Katrina A Lythgoe
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Yan Li
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jun Yuan
- Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
| | - Jianfeng He
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Jing Lu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China.
- Guangdong Workstation for Emerging Infectious Disease Control and Prevention, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Guangdong Provincial Institution of Public Health, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Zan A, Xie ZR, Hsu YC, Chen YH, Lin TH, Chang YS, Chang KY. DeepFlu: a deep learning approach for forecasting symptomatic influenza A infection based on pre-exposure gene expression. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 213:106495. [PMID: 34798406 DOI: 10.1016/j.cmpb.2021.106495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/19/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Not everyone gets sick after an exposure to influenza A viruses (IAV). Although KLRD1 has been identified as a potential biomarker for influenza susceptibility, it remains unclear whether forecasting symptomatic flu infection based on pre-exposure host gene expression might be possible. METHOD To examine this hypothesis, we developed DeepFlu using the state-of-the-art deep learning approach on the human gene expression data infected with IAV subtype H1N1 or H3N2 viruses to forecast who would catch the flu prior to an exposure to IAV. RESULTS The results indicated that such forecast is possible and, in other words, gene expression could reflect the strength of host immunity. In the leave-one-person-out cross-validation, DeepFlu based on deep neural network outperformed the models using convolutional neural network, random forest, or support vector machine, achieving 70.0% accuracy, 0.787 AUROC, and 0.758 AUPR for H1N1 and 73.8% accuracy, 0.847 AUROC, and 0.901 AUPR for H3N2. In the external validation, DeepFlu also reached 71.4% accuracy, 0.700 AUROC, and 0.723 AUPR for H1N1 and 73.5% accuracy, 0.732 AUROC, and 0.749 AUPR for H3N2, surpassing the KLRD1 biomarker. In addition, DeepFlu which was trained only by pre-exposure data worked the best than by other time spans and mixed training data of H1N1 and H3N2 did not necessarily enhance prediction. DeepFlu is available at https://github.com/ntou-compbio/DeepFlu. CONCLUSIONS DeepFlu is a prognostic tool that can moderately recognize individuals susceptible to the flu and may help prevent the spread of IAV.
Collapse
Affiliation(s)
- Anna Zan
- Computational Biology Laboratory, Department of Computer Science & Engineering, National Taiwan Ocean University, Keelung Taiwan, ROC
| | - Zhong-Ru Xie
- Computational Drug Discovery Laboratory, School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens GA, USA
| | - Yi-Chen Hsu
- Computational Biology Laboratory, Department of Computer Science & Engineering, National Taiwan Ocean University, Keelung Taiwan, ROC
| | - Yu-Hao Chen
- Computational Biology Laboratory, Department of Computer Science & Engineering, National Taiwan Ocean University, Keelung Taiwan, ROC
| | - Tsung-Hsien Lin
- Computational Biology Laboratory, Department of Computer Science & Engineering, National Taiwan Ocean University, Keelung Taiwan, ROC
| | - Yong-Shan Chang
- Computational Biology Laboratory, Department of Computer Science & Engineering, National Taiwan Ocean University, Keelung Taiwan, ROC
| | - Kuan Y Chang
- Computational Biology Laboratory, Department of Computer Science & Engineering, National Taiwan Ocean University, Keelung Taiwan, ROC.
| |
Collapse
|
23
|
Quéromès G, Destras G, Bal A, Regue H, Burfin G, Brun S, Fanget R, Morfin F, Valette M, Trouillet-Assant S, Lina B, Frobert E, Josset L. Characterization of SARS-CoV-2 ORF6 deletion variants detected in a nosocomial cluster during routine genomic surveillance, Lyon, France. Emerg Microbes Infect 2021; 10:167-177. [PMID: 33399033 PMCID: PMC7850418 DOI: 10.1080/22221751.2021.1872351] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/10/2020] [Accepted: 12/30/2020] [Indexed: 12/21/2022]
Abstract
During routine molecular surveillance of SARS-CoV-2 performed at the National Reference Center of Respiratory Viruses (Lyon, France) (n = 229 sequences collected February-April 2020), two frameshifting deletions were detected in the open reading frame 6, at the same position (27267). While a 26-nucleotide deletion variant (D26) was only found in one nasopharyngeal sample in March 2020, the 34-nucleotide deletion (D34) was found within a single geriatric hospital unit in 5/9 patients and one health care worker in April 2020. Phylogeny analysis strongly suggested a nosocomial transmission of D34, with potential fecal transmission, as also identified in a stool sample. No difference in disease severity was observed between patients hospitalized in the geriatric unit infected with WT or D34. In vitro D26 and D34 characterization revealed comparable replication kinetics with the wild-type (WT), but differential host immune responses. While interferon-stimulated genes were similarly upregulated after infection with WT and ORF6 variants, the latter specifically induced overexpression of 9 genes coding for inflammatory cytokines in the NF-kB pathway, including CCL2/MCP1, PTX3, and TNFα, for which high plasma levels have been associated with severe COVID-19. Our findings emphasize the need to monitor the occurrence of ORF6 deletions and assess their impact on the host immune response.
Collapse
Affiliation(s)
- Grégory Quéromès
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
| | - Grégory Destras
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
| | - Antonin Bal
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
| | - Hadrien Regue
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
| | - Gwendolyne Burfin
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des virus des infections respiratoires, Lyon, Fracne
| | - Solenne Brun
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des virus des infections respiratoires, Lyon, Fracne
| | - Rémi Fanget
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des virus des infections respiratoires, Lyon, Fracne
| | - Florence Morfin
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
| | - Martine Valette
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des virus des infections respiratoires, Lyon, Fracne
| | | | - Bruno Lina
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
- Centre National de Référence des virus des infections respiratoires, Lyon, Fracne
| | - Emilie Frobert
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
| | - Laurence Josset
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Lyon, Fracne
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
24
|
Ftouh M, Kalboussi N, Abid N, Sfar S, Mignet N, Bahloul B. Contribution of Nanotechnologies to Vaccine Development and Drug Delivery against Respiratory Viruses. PPAR Res 2021; 2021:6741290. [PMID: 34721558 PMCID: PMC8550859 DOI: 10.1155/2021/6741290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
According to the Center for Disease Control and Prevention (CDC), the coronavirus disease 2019, a respiratory viral illness linked to significant morbidity, mortality, production loss, and severe economic depression, was the third-largest cause of death in 2020. Respiratory viruses such as influenza, respiratory syncytial virus, SARS-CoV-2, and adenovirus, are among the most common causes of respiratory illness in humans, spreading as pandemics or epidemics throughout all continents. Nanotechnologies are particles in the nanometer range made from various compositions. They can be lipid-based, polymer-based, protein-based, or inorganic in nature, but they are all bioinspired and virus-like. In this review, we aimed to present a short review of the different nanoparticles currently studied, in particular those which led to publications in the field of respiratory viruses. We evaluated those which could be beneficial for respiratory disease-based viruses; those which already have contributed, such as lipid nanoparticles in the context of COVID-19; and those which will contribute in the future either as vaccines or antiviral drug delivery systems. We present a short assessment based on a critical selection of evidence indicating nanotechnology's promise in the prevention and treatment of respiratory infections.
Collapse
Affiliation(s)
- Mahdi Ftouh
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Nesrine Kalboussi
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
- Sahloul University Hospital, Pharmacy Department, Sousse, Tunisia
| | - Nabil Abid
- Department of Biotechnology, High Institute of Biotechnology of Sidi Thabet, University of Manouba, BP-66, 2020 Ariana, Tunis, Tunisia
- Laboratory of Transmissible Diseases and Biological Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Rue Ibn Sina, 5000 Monastir, Tunisia
| | - Souad Sfar
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Nathalie Mignet
- University of Paris, INSERM, CNRS, UTCBS, Faculté de Pharmacie, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Badr Bahloul
- Drug Development Laboratory LR12ES09, Faculty of Pharmacy, University of Monastir, Tunisia
| |
Collapse
|
25
|
Review of Influenza Virus Vaccines: The Qualitative Nature of Immune Responses to Infection and Vaccination Is a Critical Consideration. Vaccines (Basel) 2021; 9:vaccines9090979. [PMID: 34579216 PMCID: PMC8471734 DOI: 10.3390/vaccines9090979] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/28/2021] [Accepted: 08/30/2021] [Indexed: 01/06/2023] Open
Abstract
Influenza viruses have affected the world for over a century, causing multiple pandemics. Throughout the years, many prophylactic vaccines have been developed for influenza; however, these viruses are still a global issue and take many lives. In this paper, we review influenza viruses, associated immunological mechanisms, current influenza vaccine platforms, and influenza infection, in the context of immunocompromised populations. This review focuses on the qualitative nature of immune responses against influenza viruses, with an emphasis on trained immunity and an assessment of the characteristics of the host–pathogen that compromise the effectiveness of immunization. We also highlight innovative immunological concepts that are important considerations for the development of the next generation of vaccines against influenza viruses.
Collapse
|
26
|
Lin GL, Drysdale SB, Snape MD, O’Connor D, Brown A, MacIntyre-Cockett G, Mellado-Gomez E, de Cesare M, Bonsall D, Ansari MA, Öner D, Aerssens J, Butler C, Bont L, Openshaw P, Martinón-Torres F, Nair H, Bowden R, RESCEU Investigators CampbellHarry13CunninghamSteve13BogaertDebby814BeutelsPhilippe15WildenbeestJoanne8ClutterbuckElizabeth1McGinleyJoseph1ThwaitesRyan10WisemanDexter10Gómez-CarballaAlberto12Rodriguez-TenreiroCarmen12Rivero-CalleIrene12Dacosta-UrbietaAna12HeikkinenTerho16MeijerAdam17FischerThea Kølsen18van den BergeMaarten19GiaquintoCarlo20AbramMichael21DormitzerPhilip22StoszekSonia23GallichanScott24RosenBrian25MoleroEva26MachinNuria26SpadettoMartina26, Golubchik T, Pollard AJ. Distinct patterns of within-host virus populations between two subgroups of human respiratory syncytial virus. Nat Commun 2021; 12:5125. [PMID: 34446722 PMCID: PMC8390747 DOI: 10.1038/s41467-021-25265-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infection in young children globally, but little is known about within-host RSV diversity. Here, we characterised within-host RSV populations using deep-sequencing data from 319 nasopharyngeal swabs collected during 2017-2020. RSV-B had lower consensus diversity than RSV-A at the population level, while exhibiting greater within-host diversity. Two RSV-B consensus sequences had an amino acid alteration (K68N) in the fusion (F) protein, which has been associated with reduced susceptibility to nirsevimab (MEDI8897), a novel RSV monoclonal antibody under development. In addition, several minor variants were identified in the antigenic sites of the F protein, one of which may confer resistance to palivizumab, the only licensed RSV monoclonal antibody. The differences in within-host virus populations emphasise the importance of monitoring for vaccine efficacy and may help to explain the different prevalences of monoclonal antibody-escape mutants between the two subgroups.
Collapse
Affiliation(s)
- Gu-Lung Lin
- grid.4991.50000 0004 1936 8948Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK ,grid.454382.cNIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Simon B. Drysdale
- grid.4991.50000 0004 1936 8948Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK ,grid.454382.cNIHR Oxford Biomedical Research Centre, Oxford, UK ,grid.4464.20000 0001 2161 2573Present Address: Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George’s, University of London, London, UK
| | - Matthew D. Snape
- grid.4991.50000 0004 1936 8948Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK ,grid.454382.cNIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Daniel O’Connor
- grid.4991.50000 0004 1936 8948Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK ,grid.454382.cNIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Anthony Brown
- grid.4991.50000 0004 1936 8948Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - George MacIntyre-Cockett
- grid.4991.50000 0004 1936 8948Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Esther Mellado-Gomez
- grid.4991.50000 0004 1936 8948Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Mariateresa de Cesare
- grid.4991.50000 0004 1936 8948Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - David Bonsall
- grid.4991.50000 0004 1936 8948Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - M. Azim Ansari
- grid.4991.50000 0004 1936 8948Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Deniz Öner
- grid.419619.20000 0004 0623 0341Translational Biomarkers, Infectious Diseases Therapeutic Area, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jeroen Aerssens
- grid.419619.20000 0004 0623 0341Translational Biomarkers, Infectious Diseases Therapeutic Area, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Christopher Butler
- grid.4991.50000 0004 1936 8948Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Louis Bont
- grid.7692.a0000000090126352Department of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands ,ReSViNET Foundation, Zeist, Netherlands
| | - Peter Openshaw
- grid.7445.20000 0001 2113 8111National Heart and Lung Institute, Imperial College London, London, UK
| | - Federico Martinón-Torres
- grid.411048.80000 0000 8816 6945Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain ,grid.488911.d0000 0004 0408 4897Genetics, Vaccines, Infectious Diseases, and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Harish Nair
- grid.4305.20000 0004 1936 7988Centre for Global Health, Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - Rory Bowden
- grid.4991.50000 0004 1936 8948Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK ,grid.1042.7Present Address: Division of Advanced Technology and Biology, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia
| | | | - Tanya Golubchik
- grid.4991.50000 0004 1936 8948Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J. Pollard
- grid.4991.50000 0004 1936 8948Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK ,grid.454382.cNIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
27
|
Faleye TOC, Adams D, Adhikari S, Sandrolini H, Halden RU, Varsani A, Scotch M. Use of hemagglutinin and neuraminidase amplicon-based high-throughput sequencing with variant analysis to detect co-infection and resolve identical consensus sequences of seasonal influenza in a university setting. BMC Infect Dis 2021; 21:810. [PMID: 34388979 PMCID: PMC8360813 DOI: 10.1186/s12879-021-06526-5] [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: 03/01/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
Background Local transmission of seasonal influenza viruses (IVs) can be difficult to resolve. Here, we study if coupling high-throughput sequencing (HTS) of hemagglutinin (HA) and neuraminidase (NA) genes with variant analysis can resolve strains from local transmission that have identical consensus genome. We analyzed 24 samples collected over four days in January 2020 at a large university in the US. We amplified complete hemagglutinin (HA) and neuraminidase (NA) genomic segments followed by Illumina sequencing. We identified consensus complete HA and NA segments using BLASTn and performed variant analysis on strains whose HA and NA segments were 100% similar. Results Twelve of the 24 samples were PCR positive, and we detected complete HA and/or NA segments by de novo assembly in 83.33% (10/12) of them. Similarity and phylogenetic analysis showed that 70% (7/10) of the strains were distinct while the remaining 30% had identical consensus sequences. These three samples also had IAV and IBV co-infection. However, subsequent variant analysis showed that they had distinct variant profiles. While the IAV HA of one sample had no variant, another had a T663C mutation and another had both C1379T and C1589A. Conclusion In this study, we showed that HTS coupled with variant analysis of only HA and NA genes can help resolve variants that are closely related. We also provide evidence that during a short time period in the 2019–2020 season, co-infection of IAV and IBV occurred on the university campus and both 2020/2021 and 2021/2022 WHO recommended H1N1 vaccine strains were co-circulating. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06526-5.
Collapse
Affiliation(s)
- Temitope O C Faleye
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Deborah Adams
- Biodesign Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Sangeet Adhikari
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA.,School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, 85287, USA
| | - Helen Sandrolini
- Arizona State University Health Services, Arizona State University, Tempe, AZ, 85287, USA
| | - Rolf U Halden
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA.,School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, 85287, USA
| | - Arvind Varsani
- Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Matthew Scotch
- Biodesign Center for Environmental Health Engineering, Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA. .,College of Health Solutions, Arizona State University, Phoenix, AZ, 85004, USA.
| |
Collapse
|
28
|
Han AX, Felix Garza ZC, Welkers MRA, Vigeveno RM, Tran ND, Le TQM, Pham Quang T, Dang DT, Tran TNA, Ha MT, Nguyen TH, Le QT, Le TH, Hoang TBN, Chokephaibulkit K, Puthavathana P, Nguyen VVC, Nghiem MN, Nguyen VK, Dao TT, Tran TH, Wertheim HFL, Horby PW, Fox A, van Doorn HR, Eggink D, de Jong MD, Russell CA. Within-host evolutionary dynamics of seasonal and pandemic human influenza A viruses in young children. eLife 2021; 10:e68917. [PMID: 34342576 PMCID: PMC8382297 DOI: 10.7554/elife.68917] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/02/2021] [Indexed: 01/14/2023] Open
Abstract
The evolution of influenza viruses is fundamentally shaped by within-host processes. However, the within-host evolutionary dynamics of influenza viruses remain incompletely understood, in part because most studies have focused on infections in healthy adults based on single timepoint data. Here, we analyzed the within-host evolution of 82 longitudinally sampled individuals, mostly young children, infected with A/H1N1pdm09 or A/H3N2 viruses between 2007 and 2009. For A/H1N1pdm09 infections during the 2009 pandemic, nonsynonymous minority variants were more prevalent than synonymous ones. For A/H3N2 viruses in young children, early infection was dominated by purifying selection. As these infections progressed, nonsynonymous variants typically increased in frequency even when within-host virus titers decreased. Unlike the short-lived infections of adults where de novo within-host variants are rare, longer infections in young children allow for the maintenance of virus diversity via mutation-selection balance creating potentially important opportunities for within-host virus evolution.
Collapse
Affiliation(s)
- Alvin X Han
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Zandra C Felix Garza
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Matthijs RA Welkers
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - René M Vigeveno
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Nhu Duong Tran
- National Institute of Hygiene and EpidemiologyHanoiViet Nam
| | | | | | | | | | | | | | | | - Thanh Hai Le
- Vietnam National Children's HospitalHanoiViet Nam
| | | | | | | | | | | | | | | | - Tinh Hien Tran
- Siriraj Hospital, Mahidol UniversityBangkokThailand
- Oxford University Clinical Research UnitHo Chi Minh cityViet Nam
| | - Heiman FL Wertheim
- Oxford University Clinical Research UnitHo Chi Minh cityViet Nam
- Radboud Medical Centre, Radboud UniversityNijmegenNetherlands
- Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Peter W Horby
- Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Oxford University Clinical Research UnitHanoiViet Nam
| | - Annette Fox
- Oxford University Clinical Research UnitHanoiViet Nam
- Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
- WHO Collaborating Centre for Reference and Research on InfluenzaMelbourneAustralia
| | - H Rogier van Doorn
- Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Oxford University Clinical Research UnitHanoiViet Nam
| | - Dirk Eggink
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the EnvironmentBilthovenNetherlands
| | - Menno D de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| | - Colin A Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical CenterAmsterdamNetherlands
| |
Collapse
|
29
|
Abstract
Human respiratory virus infections lead to a spectrum of respiratory symptoms and disease severity, contributing to substantial morbidity, mortality and economic losses worldwide, as seen in the COVID-19 pandemic. Belonging to diverse families, respiratory viruses differ in how easy they spread (transmissibility) and the mechanism (modes) of transmission. Transmissibility as estimated by the basic reproduction number (R0) or secondary attack rate is heterogeneous for the same virus. Respiratory viruses can be transmitted via four major modes of transmission: direct (physical) contact, indirect contact (fomite), (large) droplets and (fine) aerosols. We know little about the relative contribution of each mode to the transmission of a particular virus in different settings, and how its variation affects transmissibility and transmission dynamics. Discussion on the particle size threshold between droplets and aerosols and the importance of aerosol transmission for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus is ongoing. Mechanistic evidence supports the efficacies of non-pharmaceutical interventions with regard to virus reduction; however, more data are needed on their effectiveness in reducing transmission. Understanding the relative contribution of different modes to transmission is crucial to inform the effectiveness of non-pharmaceutical interventions in the population. Intervening against multiple modes of transmission should be more effective than acting on a single mode.
Collapse
Affiliation(s)
- Nancy H L Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
30
|
Liu R, Wu P, Ogrodzki P, Mahmoud S, Liang K, Liu P, Francis SS, Khalak H, Liu D, Li J, Ma T, Chen F, Liu W, Huang X, He W, Yuan Z, Qiao N, Meng X, Alqarni B, Quilez J, Kusuma V, Lin L, Jin X, Yang C, Anton X, Koshy A, Yang H, Xu X, Wang J, Xiao P, Al Kaabi N, Fasihuddin MS, Selvaraj FA, Weber S, Al Hosani FI, Liu S, Zaher WA. Genomic epidemiology of SARS-CoV-2 in the UAE reveals novel virus mutation, patterns of co-infection and tissue specific host immune response. Sci Rep 2021; 11:13971. [PMID: 34234167 PMCID: PMC8263779 DOI: 10.1038/s41598-021-92851-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/14/2021] [Indexed: 01/08/2023] Open
Abstract
To unravel the source of SARS-CoV-2 introduction and the pattern of its spreading and evolution in the United Arab Emirates, we conducted meta-transcriptome sequencing of 1067 nasopharyngeal swab samples collected between May 9th and Jun 29th, 2020 during the first peak of the local COVID-19 epidemic. We identified global clade distribution and eleven novel genetic variants that were almost absent in the rest of the world and that defined five subclades specific to the UAE viral population. Cross-settlement human-to-human transmission was related to the local business activity. Perhaps surprisingly, at least 5% of the population were co-infected by SARS-CoV-2 of multiple clades within the same host. We also discovered an enrichment of cytosine-to-uracil mutation among the viral population collected from the nasopharynx, that is different from the adenosine-to-inosine change previously reported in the bronchoalveolar lavage fluid samples and a previously unidentified upregulation of APOBEC4 expression in nasopharynx among infected patients, indicating the innate immune host response mediated by ADAR and APOBEC gene families could be tissue-specific. The genomic epidemiological and molecular biological knowledge reported here provides new insights for the SARS-CoV-2 evolution and transmission and points out future direction on host-pathogen interaction investigation.
Collapse
Affiliation(s)
- Rong Liu
- Group42 Healthcare, Abu Dhabi, United Arab Emirates
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Pei Wu
- Group42 Healthcare, Abu Dhabi, United Arab Emirates
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | | | | | - Ke Liang
- MGI, BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Pengjuan Liu
- MGI, BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Stephen S Francis
- Department of Neurological Surgery, University of California, San Francisco, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - Hanif Khalak
- Group42 Healthcare, Abu Dhabi, United Arab Emirates
| | - Denghui Liu
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, 518100, China
| | - Junhua Li
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, China
| | - Tao Ma
- MGI, BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Fang Chen
- MGI, BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Weibin Liu
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Xinyu Huang
- MGI, BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Wenjun He
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, 518100, China
| | - Zhaorong Yuan
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, 518100, China
| | - Nan Qiao
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, 518100, China
| | - Xin Meng
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd., Shenzhen, 518100, China
| | | | | | - Vinay Kusuma
- Group42 Healthcare, Abu Dhabi, United Arab Emirates
| | - Long Lin
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Chongguang Yang
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, USA
| | - Xavier Anton
- Group42 Healthcare, Abu Dhabi, United Arab Emirates
| | - Ashish Koshy
- Group42 Healthcare, Abu Dhabi, United Arab Emirates
| | | | - Xun Xu
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China
| | - Peng Xiao
- Group42 Healthcare, Abu Dhabi, United Arab Emirates
| | - Nawal Al Kaabi
- SEHA, Abu Dhabi Health Services Co., Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | | | | | - Stefan Weber
- SEHA, Abu Dhabi Health Services Co., Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | | | - Siyang Liu
- BGI-Shenzhen, Shenzhen, 518083, Guangdong, China.
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 510006, Guangdong, China.
| | | |
Collapse
|
31
|
Fuhrmann L, Jablonski KP, Beerenwinkel N. Quantitative measures of within-host viral genetic diversity. Curr Opin Virol 2021; 49:157-163. [PMID: 34153841 DOI: 10.1016/j.coviro.2021.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/22/2022]
Abstract
The genetic diversity of virus populations within their hosts is known to influence disease progression, treatment outcome, drug resistance, cell tropism, and transmission risk, and the study of dynamic changes of genetic heterogeneity can provide insights into the evolution of viruses. Several measures to quantify within-host genetic diversity capturing different aspects of diversity patterns in a sample or population are used, based on incidence, relative frequencies, pairwise distances, or phylogenetic trees. Here, we review and compare several of these measures.
Collapse
Affiliation(s)
- Lara Fuhrmann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| | - Kim Philipp Jablonski
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland.
| |
Collapse
|
32
|
Xie C, Su W, Sia SF, Choy KT, Morrell S, Zhou J, Peiris M, Bloom J, Yen HL. A(H1N1)pdm09 influenza viruses replicating in ferret upper or lower respiratory tract differed in onward transmission potential by air. J Infect Dis 2021; 225:65-74. [PMID: 34036370 DOI: 10.1093/infdis/jiab286] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/21/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND A(H1N1)pdm09 influenza viruses replicate efficiently in respiratory epithelia and are transmitted via respiratory droplets and aerosols expelled by infected hosts. The relative onward transmission potential of influenza viruses replicating in the upper and lower respiratory epithelial cells has not been fully defined. METHODS Wild-type and barcoded A(H1N1)pdm09 viruses that differed by 2 synonymous mutations per gene segment were inoculated into ferrets via intra-nasal and intra-tracheal routes. Naïve recipients were exposed to the exhaled breath of inoculated donors for 8 hours on day 2 post-inoculation. Onward transmission potential of wild-type and barcoded genotypes were monitored by next generation sequencing. RESULTS Transmissible airborne particles were respired from the upper but not the lower respiratory epithelial cells of donor ferrets. There was limited mixing of viral populations replicating in the upper and lower respiratory tissues. CONCLUSIONS The ferret upper respiratory epithelium was mapped as the anatomic site that generated influenza virus-laden particles mediating onward transmission by air. Our results suggest that vaccines and antivirals should aim to reduce viral loads in the upper respiratory tract for prevention of influenza transmission.
Collapse
Affiliation(s)
- Chenyi Xie
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Wen Su
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Sin Fun Sia
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Ka-Tim Choy
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Steven Morrell
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Jie Zhou
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Jesse Bloom
- Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Howard Hughes Medical Institutes, Seattle, WA, USA
| | - Hui-Ling Yen
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
33
|
Wagner J, Yuen L, Littlejohn M, Sozzi V, Jackson K, Suri V, Tan S, Feierbach B, Gaggar A, Marcellin P, Buti Ferret M, Janssen HLA, Gane E, Chan HLY, Colledge D, Rosenberg G, Bayliss J, Howden BP, Locarnini SA, Wong D, Thompson AT, Revill PA. Analysis of Hepatitis B Virus Haplotype Diversity Detects Striking Sequence Conservation Across Genotypes and Chronic Disease Phase. Hepatology 2021; 73:1652-1670. [PMID: 32780526 DOI: 10.1002/hep.31516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/01/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS We conducted haplotype analysis of complete hepatitis B virus (HBV) genomes following deep sequencing from 368 patients across multiple phases of chronic hepatitis B (CHB) infection from four major genotypes (A-D), analyzing 4,110 haplotypes to identify viral variants associated with treatment outcome and disease progression. APPROACH AND RESULTS Between 18.2% and 41.8% of nucleotides and between 5.9% and 34.3% of amino acids were 100% conserved in all genotypes and phases examined, depending on the region analyzed. Hepatitis B e antigen (HBeAg) loss by week 192 was associated with different haplotype populations at baseline. Haplotype populations differed across the HBV genome and CHB history, this being most pronounced in the precore/core gene. Mean number of haplotypes (frequency) per patient was higher in immune-active, HBeAg-positive chronic hepatitis phase 2 (11.8) and HBeAg-negative chronic hepatitis phase 4 (16.2) compared to subjects in the "immune-tolerant," HBeAg-positive chronic infection phase 1 (4.3, P< 0.0001). Haplotype frequency was lowest in genotype B (6.2, P< 0.0001) compared to the other genotypes (A = 11.8, C = 11.8, D = 13.6). Haplotype genetic diversity increased over the course of CHB history, being lowest in phase 1, increasing in phase 2, and highest in phase 4 in all genotypes except genotype C. HBeAg loss by week 192 of tenofovir therapy was associated with different haplotype populations at baseline. CONCLUSIONS Despite a degree of HBV haplotype diversity and heterogeneity across the phases of CHB natural history, highly conserved sequences in key genes and regulatory regions were identified in multiple HBV genotypes that should be further investigated as targets for antiviral therapies and predictors of treatment response.
Collapse
Affiliation(s)
- Josef Wagner
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Lilly Yuen
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Margaret Littlejohn
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Vitina Sozzi
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Kathy Jackson
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | | | | | | | | | | | - Maria Buti Ferret
- Liver Unit, Valle d'Hebron University Hospital, Ciberehd del Insituto Carlos III Barcelona, Barcelona, Spain
| | - Harry L A Janssen
- Toronto Center for Liver Diseases, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Ed Gane
- New Zealand Liver Transplant Unit, Auckland City Hospital, Auckland, New Zealand
| | - Henry L Y Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Danni Colledge
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Gillian Rosenberg
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Julianne Bayliss
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin P Howden
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Stephen A Locarnini
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| | - Darren Wong
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia.,Department of Gastroenterology, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Alexander T Thompson
- Department of Gastroenterology, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Peter A Revill
- Division of Molecular Research and Development, Victorian Infectious Diseases, Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne Healthy, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
34
|
Sapoval N, Mahmoud M, Jochum MD, Liu Y, Elworth RAL, Wang Q, Albin D, Ogilvie HA, Lee MD, Villapol S, Hernandez KM, Maljkovic Berry I, Foox J, Beheshti A, Ternus K, Aagaard KM, Posada D, Mason CE, Sedlazeck FJ, Treangen TJ. SARS-CoV-2 genomic diversity and the implications for qRT-PCR diagnostics and transmission. Genome Res 2021; 31:635-644. [PMID: 33602693 PMCID: PMC8015855 DOI: 10.1101/gr.268961.120] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
The COVID-19 pandemic has sparked an urgent need to uncover the underlying biology of this devastating disease. Though RNA viruses mutate more rapidly than DNA viruses, there are a relatively small number of single nucleotide polymorphisms (SNPs) that differentiate the main SARS-CoV-2 lineages that have spread throughout the world. In this study, we investigated 129 RNA-seq data sets and 6928 consensus genomes to contrast the intra-host and inter-host diversity of SARS-CoV-2. Our analyses yielded three major observations. First, the mutational profile of SARS-CoV-2 highlights intra-host single nucleotide variant (iSNV) and SNP similarity, albeit with differences in C > U changes. Second, iSNV and SNP patterns in SARS-CoV-2 are more similar to MERS-CoV than SARS-CoV-1. Third, a significant fraction of insertions and deletions contribute to the genetic diversity of SARS-CoV-2. Altogether, our findings provide insight into SARS-CoV-2 genomic diversity, inform the design of detection tests, and highlight the potential of iSNVs for tracking the transmission of SARS-CoV-2.
Collapse
Affiliation(s)
- Nicolae Sapoval
- Department of Computer Science, Rice University, Houston, Texas 77005, USA
| | - Medhat Mahmoud
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Michael D Jochum
- Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030, USA
| | - Yunxi Liu
- Department of Computer Science, Rice University, Houston, Texas 77005, USA
| | - R A Leo Elworth
- Department of Computer Science, Rice University, Houston, Texas 77005, USA
| | - Qi Wang
- Systems, Synthetic, and Physical Biology (SSPB) Graduate Program, Rice University, Houston, Texas 77005, USA
| | - Dreycey Albin
- Systems, Synthetic, and Physical Biology (SSPB) Graduate Program, Rice University, Houston, Texas 77005, USA
| | - Huw A Ogilvie
- Department of Computer Science, Rice University, Houston, Texas 77005, USA
| | - Michael D Lee
- Exobiology Branch, NASA Ames Research Center, Mountain View, California 94043, USA
- Blue Marble Space Institute of Science, Seattle, Washington 98104, USA
| | - Sonia Villapol
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Kyle M Hernandez
- Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA
- Center for Translational Data Science, University of Chicago, Chicago, Illinois 60637, USA
| | | | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, California 94035, USA
| | | | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030, USA
| | - David Posada
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Department of Biochemistry, Genetics, and Immunology, Universidade de Vigo, 36310 Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36213 Vigo, Spain
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Todd J Treangen
- Department of Computer Science, Rice University, Houston, Texas 77005, USA
| |
Collapse
|
35
|
Yamamoto T, Sawai K, Nishi T, Fukai K, Kato T, Hayama Y, Murato Y, Shimizu Y, Yamaguchi E. Subgrouping and analysis of relationships between classical swine fever virus identified during the 2018-2020 epidemic in Japan by a novel approach using shared genomic variants. Transbound Emerg Dis 2021; 69:1166-1177. [PMID: 33730417 DOI: 10.1111/tbed.14076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/01/2021] [Accepted: 03/15/2021] [Indexed: 11/29/2022]
Abstract
Classical swine fever (CSF) is a worldwide devastating disease of the pig industry caused by classical swine fever virus (CSFV). In September 2018, an outbreak of CSF occurred in Japan where the disease had been eradicated and was officially designated a CSF-free country since 2015. Following the detection of the first 2018 case on a farm in Gifu Prefecture, the disease spread among both farm pigs and wild boars and still continues. Epigenome analysis using whole-genome information is helpful in identifying the infection route, but the current approaches provide an insufficient resolution. In this study, a novel method of using single-nucleotide variants (SNVs) was employed to identify the associations among 158 isolates (65 from farms and 93 from wild boars). The identified groups of CSFV strains were plotted in different colours on a map, identifying the location where each strain was collected. The lack of an SNV set shared between the index case and the other strains suggested the first infection in Japan during the outbreak occurred in wild boars, not at the index farm. For the Atsumi Peninsula outbreaks, where nine farms were found infected within a 10-km radius area, the farm strains were assembled into three groups, suggesting these outbreaks resulted from at least three different infection events in this area. For the infections in the area around Saitama Prefecture, an area remote from the epicentre, strains from both the farms and wild boars were identified as being in the same group, suggesting they resulted from one viral introduction. Likewise, seven infected farms in Okinawa Prefecture, almost 1,500 km from Gifu Prefecture, were identified as being in a common, but separate group. By demonstrating the variety of transmission routes and possibility of long-distance infection, these results will help improve disease control measures.
Collapse
Affiliation(s)
- Takehisa Yamamoto
- Epidemiology Unit, Viral Disease and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Kotaro Sawai
- Epidemiology Unit, Viral Disease and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Tatsuya Nishi
- Foot and Mouth Disease Unit, Division of Transboundary Animal Diseases, National Institute of Animal Health, National Agriculture and Food Research Organization, Kodaira, Japan
| | - Katsuhiko Fukai
- Foot and Mouth Disease Unit, Division of Transboundary Animal Diseases, National Institute of Animal Health, National Agriculture and Food Research Organization, Kodaira, Japan
| | - Tomoko Kato
- Foot and Mouth Disease Unit, Division of Transboundary Animal Diseases, National Institute of Animal Health, National Agriculture and Food Research Organization, Kodaira, Japan
| | - Yoko Hayama
- Epidemiology Unit, Viral Disease and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Yoshinori Murato
- Epidemiology Unit, Viral Disease and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Yumiko Shimizu
- Epidemiology Unit, Viral Disease and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Emi Yamaguchi
- Epidemiology Unit, Viral Disease and Epidemiology Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Ibaraki, Japan
| |
Collapse
|
36
|
Ramazzotti D, Angaroni F, Maspero D, Gambacorti-Passerini C, Antoniotti M, Graudenzi A, Piazza R. VERSO: A comprehensive framework for the inference of robust phylogenies and the quantification of intra-host genomic diversity of viral samples. PATTERNS (NEW YORK, N.Y.) 2021; 2:100212. [PMID: 33728416 PMCID: PMC7953447 DOI: 10.1016/j.patter.2021.100212] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/30/2020] [Accepted: 01/22/2021] [Indexed: 12/22/2022]
Abstract
We introduce VERSO, a two-step framework for the characterization of viral evolution from sequencing data of viral genomes, which is an improvement on phylogenomic approaches for consensus sequences. VERSO exploits an efficient algorithmic strategy to return robust phylogenies from clonal variant profiles, also in conditions of sampling limitations. It then leverages variant frequency patterns to characterize the intra-host genomic diversity of samples, revealing undetected infection chains and pinpointing variants likely involved in homoplasies. On simulations, VERSO outperforms state-of-the-art tools for phylogenetic inference. Notably, the application to 6,726 amplicon and RNA sequencing samples refines the estimation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolution, while co-occurrence patterns of minor variants unveil undetected infection paths, which are validated with contact tracing data. Finally, the analysis of SARS-CoV-2 mutational landscape uncovers a temporal increase of overall genomic diversity and highlights variants transiting from minor to clonal state and homoplastic variants, some of which fall on the spike gene. Available at: https://github.com/BIMIB-DISCo/VERSO.
Collapse
Affiliation(s)
- Daniele Ramazzotti
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - Fabrizio Angaroni
- Department of Informatics, Systems and Communication, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Davide Maspero
- Department of Informatics, Systems and Communication, Università degli Studi di Milano-Bicocca, Milan, Italy
- Inst. of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
| | | | - Marco Antoniotti
- Department of Informatics, Systems and Communication, Università degli Studi di Milano-Bicocca, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre – B4, Milan, Italy
| | - Alex Graudenzi
- Inst. of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre – B4, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| |
Collapse
|
37
|
Graudenzi A, Maspero D, Angaroni F, Piazza R, Ramazzotti D. Mutational signatures and heterogeneous host response revealed via large-scale characterization of SARS-CoV-2 genomic diversity. iScience 2021; 24:102116. [PMID: 33532709 PMCID: PMC7842190 DOI: 10.1016/j.isci.2021.102116] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/09/2020] [Accepted: 01/22/2021] [Indexed: 01/03/2023] Open
Abstract
To dissect the mechanisms underlying the inflation of variants in the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) genome, we present a large-scale analysis of intra-host genomic diversity, which reveals that most samples exhibit heterogeneous genomic architectures, due to the interplay between host-related mutational processes and transmission dynamics. The decomposition of minor variants profiles unveils three non-overlapping mutational signatures related to nucleotide substitutions and likely ruled by APOlipoprotein B Editing Complex (APOBEC), Reactive Oxygen Species (ROS), and Adenosine Deaminase Acting on RNA (ADAR), highlighting heterogeneous host responses to SARS-CoV-2 infections. A corrected-for-signatures dN/dS analysis demonstrates that such mutational processes are affected by purifying selection, with important exceptions. In fact, several mutations appear to transit toward clonality, defining new clonal genotypes that increase the overall genomic diversity. Furthermore, the phylogenomic analysis shows the presence of homoplasies and supports the hypothesis of transmission of minor variants. This study paves the way for the integrated analysis of intra-host genomic diversity and clinical outcomes of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Alex Graudenzi
- Inst. of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre – B4, Milan, Italy
| | - Davide Maspero
- Inst. of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
- Department of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy
| | - Fabrizio Angaroni
- Department of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, Univ. of Milan-Bicocca, Monza, Italy
| | - Daniele Ramazzotti
- Department of Medicine and Surgery, Univ. of Milan-Bicocca, Monza, Italy
| |
Collapse
|
38
|
Wang D, Wang Y, Sun W, Zhang L, Ji J, Zhang Z, Cheng X, Li Y, Xiao F, Zhu A, Zhong B, Ruan S, Li J, Ren P, Ou Z, Xiao M, Li M, Deng Z, Zhong H, Li F, Wang WJ, Zhang Y, Chen W, Zhu S, Xu X, Jin X, Zhao J, Zhong N, Zhang W, Zhao J, Li J, Xu Y. Population Bottlenecks and Intra-host Evolution During Human-to-Human Transmission of SARS-CoV-2. Front Med (Lausanne) 2021; 8:585358. [PMID: 33659260 PMCID: PMC7917136 DOI: 10.3389/fmed.2021.585358] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
The emergence of the novel human coronavirus, SARS-CoV-2, causes a global COVID-19 (coronavirus disease 2019) pandemic. Here, we have characterized and compared viral populations of SARS-CoV-2 among COVID-19 patients within and across households. Our work showed an active viral replication activity in the human respiratory tract and the co-existence of genetically distinct viruses within the same host. The inter-host comparison among viral populations further revealed a narrow transmission bottleneck between patients from the same households, suggesting a dominated role of stochastic dynamics in both inter-host and intra-host evolutions.
Collapse
Affiliation(s)
- Daxi Wang
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wanying Sun
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Lu Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Infectious Disease, Guangzhou Eighth People's Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingkai Ji
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinyi Cheng
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yimin Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fei Xiao
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Department of Infectious Diseases, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bei Zhong
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | | | - Jiandong Li
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Peidi Ren
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Zhihua Ou
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Minfeng Xiao
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Min Li
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Ziqing Deng
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Huanzi Zhong
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Fuqiang Li
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI-Shenzhen, Shenzhen, China
| | - Wen-jing Wang
- BGI-Shenzhen, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI-Shenzhen, Shenzhen, China
| | | | - Weijun Chen
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
- BGI PathoGenesis Pharmaceutical Technology, BGI-Shenzhen, Shenzhen, China
| | - Shida Zhu
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Engineering Laboratory for Innovative Molecular Diagnostics, BGI-Shenzhen, Shenzhen, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Infectious Disease, Guangzhou Eighth People's Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junhua Li
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yonghao Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
39
|
Posada-Céspedes S, Seifert D, Topolsky I, Jablonski KP, Metzner KJ, Beerenwinkel N. V-pipe: a computational pipeline for assessing viral genetic diversity from high-throughput data. Bioinformatics 2021; 37:1673-1680. [PMID: 33471068 PMCID: PMC8289377 DOI: 10.1093/bioinformatics/btab015] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/09/2020] [Accepted: 01/08/2021] [Indexed: 12/30/2022] Open
Abstract
Motivation High-throughput sequencing technologies are used increasingly not only in viral genomics research but also in clinical surveillance and diagnostics. These technologies facilitate the assessment of the genetic diversity in intra-host virus populations, which affects transmission, virulence and pathogenesis of viral infections. However, there are two major challenges in analysing viral diversity. First, amplification and sequencing errors confound the identification of true biological variants, and second, the large data volumes represent computational limitations. Results To support viral high-throughput sequencing studies, we developed V-pipe, a bioinformatics pipeline combining various state-of-the-art statistical models and computational tools for automated end-to-end analyses of raw sequencing reads. V-pipe supports quality control, read mapping and alignment, low-frequency mutation calling, and inference of viral haplotypes. For generating high-quality read alignments, we developed a novel method, called ngshmmalign, based on profile hidden Markov models and tailored to small and highly diverse viral genomes. V-pipe also includes benchmarking functionality providing a standardized environment for comparative evaluations of different pipeline configurations. We demonstrate this capability by assessing the impact of three different read aligners (Bowtie 2, BWA MEM, ngshmmalign) and two different variant callers (LoFreq, ShoRAH) on the performance of calling single-nucleotide variants in intra-host virus populations. V-pipe supports various pipeline configurations and is implemented in a modular fashion to facilitate adaptations to the continuously changing technology landscape. Availabilityand implementation V-pipe is freely available at https://github.com/cbg-ethz/V-pipe. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Susana Posada-Céspedes
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| | - David Seifert
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| | - Ivan Topolsky
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| | - Kim Philipp Jablonski
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| | - Karin J Metzner
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, 8091, Switzerland.,4 Institute of Medical Virology, University of Zurich, Zurich, 8091, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, 4058, Switzerland
| |
Collapse
|
40
|
Takayama I, Nguyen BG, Dao CX, Pham TT, Dang TQ, Truong PT, Do TV, Pham TTP, Fujisaki S, Odagiri T, Hasegawa H, Nakajima N. Next-Generation Sequencing Analysis of the Within-Host Genetic Diversity of Influenza A(H1N1)pdm09 Viruses in the Upper and Lower Respiratory Tracts of Patients with Severe Influenza. mSphere 2021; 6:e01043-20. [PMID: 33408229 PMCID: PMC7845592 DOI: 10.1128/msphere.01043-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/16/2020] [Indexed: 01/22/2023] Open
Abstract
The influenza A(H1N1)pdm09 virus emerged in April 2009 with an unusual incidence of severe disease and mortality, and currently circulates as a seasonal influenza virus. Previous studies using consensus viral genome sequencing data have overlooked the viral genomic and phenotypic diversity. Next-generation sequencing (NGS) may instead be used to characterize viral populations in an unbiased manner and to measure within-host genetic diversity. In this study, we used NGS analysis to investigate the within-host genetic diversity of influenza A(H1N1)pdm09 virus in the upper and lower respiratory samples from nine patients who were admitted to the intensive care unit (ICU). A total of 47 amino acid substitution positions were found to differ between the upper and lower respiratory tract samples from all patients. However, the D222G/N substitution in hemagglutinin (HA) protein was the only amino acid substitution common to multiple patients. Furthermore, the substitution was detected only in the six samples from the lower respiratory tract. Therefore, it is important to investigate influenza A(H1N1)pdm09 virus populations using multiple paired samples from the upper and lower respiratory tract to avoid overlooking potentially important substitutions, especially in patients with severe disease.IMPORTANCE The D222G/N substitution in the hemagglutinin (HA) protein of influenza A(H1N1)pdm09 virus has been reported to be associated with disease severity and mortality in numerous previous studies. In the present study, 75% of lower respiratory samples contained heterogeneous influenza populations that carried different amino acids at position 222 of the HA protein, whereas all upper respiratory samples only contained the wild-type 222D. These results suggest the influenza A(H1N1)pdm09 virus has diversified inside the host owing to differences in tissue specificity. In this study, the within-host genetic diversity of influenza A(H1N1)pdm09 virus was investigated for the first time using next-generation sequencing analysis of the viral whole-genome in samples extracted from the upper and lower respiratory tracts of patients with severe disease.
Collapse
Affiliation(s)
- Ikuyo Takayama
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | | | | | | | | | - Seiichiro Fujisaki
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takato Odagiri
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hideki Hasegawa
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Noriko Nakajima
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
41
|
Mikhaylova YV, Shelenkov AA, Yanushevich YG, Shagin DA. Increasing the Uniformity of Genome Fragment Coverage for High-Throughput Sequencing of Influenza A Virus. Mol Biol 2021. [DOI: 10.1134/s0026893320060084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Schreiber SJ, Ke R, Loverdo C, Park M, Ahsan P, Lloyd-Smith JO. Cross-scale dynamics and the evolutionary emergence of infectious diseases. Virus Evol 2021; 7:veaa105. [PMID: 35186322 PMCID: PMC8087961 DOI: 10.1093/ve/veaa105] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
When emerging pathogens encounter new host species for which they are poorly adapted, they must evolve to escape extinction. Pathogens experience selection on traits at multiple scales, including replication rates within host individuals and transmissibility between hosts. We analyze a stochastic model linking pathogen growth and competition within individuals to transmission between individuals. Our analysis reveals a new factor, the cross-scale reproductive number of a mutant virion, that quantifies how quickly mutant strains increase in frequency when they initially appear in the infected host population. This cross-scale reproductive number combines with viral mutation rates, single-strain reproductive numbers, and transmission bottleneck width to determine the likelihood of evolutionary emergence, and whether evolution occurs swiftly or gradually within chains of transmission. We find that wider transmission bottlenecks facilitate emergence of pathogens with short-term infections, but hinder emergence of pathogens exhibiting cross-scale selective conflict and long-term infections. Our results provide a framework to advance the integration of laboratory, clinical, and field data in the context of evolutionary theory, laying the foundation for a new generation of evidence-based risk assessment of emergence threats.
Collapse
Affiliation(s)
| | - Ruian Ke
- T-6: Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Claude Loverdo
- Laboratoire Jean Perrin, Sorbonne Université, CNRS, Paris 75005, France
| | - Miran Park
- Department of Ecology & Evolution, University of California, Los Angeles, CA 90095, USA
| | - Prianna Ahsan
- Department of Ecology & Evolution, University of California, Los Angeles, CA 90095, USA
| | - James O Lloyd-Smith
- Department of Ecology & Evolution, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
43
|
Popa A, Genger JW, Nicholson MD, Penz T, Schmid D, Aberle SW, Agerer B, Lercher A, Endler L, Colaço H, Smyth M, Schuster M, Grau ML, Martínez-Jiménez F, Pich O, Borena W, Pawelka E, Keszei Z, Senekowitsch M, Laine J, Aberle JH, Redlberger-Fritz M, Karolyi M, Zoufaly A, Maritschnik S, Borkovec M, Hufnagl P, Nairz M, Weiss G, Wolfinger MT, von Laer D, Superti-Furga G, Lopez-Bigas N, Puchhammer-Stöckl E, Allerberger F, Michor F, Bock C, Bergthaler A. Genomic epidemiology of superspreading events in Austria reveals mutational dynamics and transmission properties of SARS-CoV-2. Sci Transl Med 2020; 12:eabe2555. [PMID: 33229462 PMCID: PMC7857414 DOI: 10.1126/scitranslmed.abe2555] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022]
Abstract
Superspreading events shaped the coronavirus disease 2019 (COVID-19) pandemic, and their rapid identification and containment are essential for disease control. Here, we provide a national-scale analysis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) superspreading during the first wave of infections in Austria, a country that played a major role in initial virus transmissions in Europe. Capitalizing on Austria's well-developed epidemiological surveillance system, we identified major SARS-CoV-2 clusters during the first wave of infections and performed deep whole-genome sequencing of more than 500 virus samples. Phylogenetic-epidemiological analysis enabled the reconstruction of superspreading events and charts a map of tourism-related viral spread originating from Austria in spring 2020. Moreover, we exploited epidemiologically well-defined clusters to quantify SARS-CoV-2 mutational dynamics, including the observation of low-frequency mutations that progressed to fixation within the infection chain. Time-resolved virus sequencing unveiled viral mutation dynamics within individuals with COVID-19, and epidemiologically validated infector-infectee pairs enabled us to determine an average transmission bottleneck size of 103 SARS-CoV-2 particles. In conclusion, this study illustrates the power of combining epidemiological analysis with deep viral genome sequencing to unravel the spread of SARS-CoV-2 and to gain fundamental insights into mutational dynamics and transmission properties.
Collapse
Affiliation(s)
- Alexandra Popa
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Jakob-Wendelin Genger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Michael D Nicholson
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Thomas Penz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Daniela Schmid
- Austrian Agency for Health and Food Safety (AGES), 1220 Vienna, Austria
| | - Stephan W Aberle
- Center for Virology, Medical University of Vienna, 1090 Vienna, Austria
| | - Benedikt Agerer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Alexander Lercher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Lukas Endler
- Bioinformatics and Biostatistics Platform, Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Henrique Colaço
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Mark Smyth
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Michael Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Miguel L Grau
- Institute for Research in Biomedicine (IRB), 08028 Barcelona, Spain
| | | | - Oriol Pich
- Institute for Research in Biomedicine (IRB), 08028 Barcelona, Spain
| | - Wegene Borena
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Erich Pawelka
- Department of Medicine IV, Kaiser Franz Josef Hospital, 1100 Vienna, Austria
| | - Zsofia Keszei
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Martin Senekowitsch
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Jan Laine
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Judith H Aberle
- Center for Virology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Mario Karolyi
- Department of Medicine IV, Kaiser Franz Josef Hospital, 1100 Vienna, Austria
| | - Alexander Zoufaly
- Department of Medicine IV, Kaiser Franz Josef Hospital, 1100 Vienna, Austria
| | | | - Martin Borkovec
- Austrian Agency for Health and Food Safety (AGES), 1220 Vienna, Austria
| | - Peter Hufnagl
- Austrian Agency for Health and Food Safety (AGES), 1220 Vienna, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michael T Wolfinger
- Department of Theoretical Chemistry, University of Vienna, 1090 Vienna, Austria
- Research Group Bioinformatics and Computational Biology, Faculty of Computer Science, University of Vienna, 1090 Vienna, Austria
| | - Dorothee von Laer
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Nuria Lopez-Bigas
- Institute for Research in Biomedicine (IRB), 08028 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | | | - Franz Allerberger
- Austrian Agency for Health and Food Safety (AGES), 1220 Vienna, Austria
| | - Franziska Michor
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria.
| |
Collapse
|
44
|
Primary Swine Respiratory Epithelial Cell Lines for the Efficient Isolation and Propagation of Influenza A Viruses. J Virol 2020; 94:JVI.01091-20. [PMID: 32967961 DOI: 10.1128/jvi.01091-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Influenza virus isolation from clinical samples is critical for the identification and characterization of circulating and emerging viruses. Yet efficient isolation can be difficult. In these studies, we isolated primary swine nasal and tracheal respiratory epithelial cells and immortalized swine nasal epithelial cells (siNEC) and tracheal epithelial cells (siTEC) that retained the abilities to form tight junctions and cilia and to differentiate at the air-liquid interface like primary cells. Critically, both human and swine influenza viruses replicated in the immortalized cells, which generally yielded higher-titer viral isolates from human and swine nasal swabs, supported the replication of isolates that failed to grow in Madin-Darby canine kidney (MDCK) cells, and resulted in fewer dominating mutations during viral passaging than MDCK cells.IMPORTANCE Robust in vitro culture systems for influenza virus are critically needed. MDCK cells, the most widely used cell line for influenza isolation and propagation, do not adequately model the respiratory tract. Therefore, many clinical isolates, both animal and human, are unable to be isolated and characterized, limiting our understanding of currently circulating influenza viruses. We have developed immortalized swine respiratory epithelial cells that retain the ability to differentiate and can support influenza replication and isolation. These cell lines can be used as additional tools to enhance influenza research and vaccine development.
Collapse
|
45
|
Dimas Martins A, Gjini E. Modeling Competitive Mixtures With the Lotka-Volterra Framework for More Complex Fitness Assessment Between Strains. Front Microbiol 2020; 11:572487. [PMID: 33072034 PMCID: PMC7536265 DOI: 10.3389/fmicb.2020.572487] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/12/2020] [Indexed: 11/13/2022] Open
Abstract
With increasing resolution of microbial diversity at the genomic level, experimental and modeling frameworks that translate such diversity into phenotypes are highly needed. This is particularly important when comparing drug-resistant with drug-sensitive pathogen strains, when anticipating epidemiological implications of microbial diversity, and when designing control measures. Classical approaches quantify differences between microbial strains using the exponential growth model, and typically report a selection coefficient for the relative fitness differential between two strains. The apparent simplicity of such approaches comes with the costs of limiting the range of biological scenarios that can be captured, and biases strain fitness estimates to polarized extremes of competitive exclusion. Here, we propose a mathematical and statistical framework based on the Lotka-Volterra model, that can capture frequency-dependent competition between microbial strains within-host and upon transmission. As a proof-of-concept, the model is applied to a previously-published dataset from in-vivo competitive mixture experiments with influenza strains in ferrets (McCaw et al., 2011). We show that for the same data, our model predicts a scenario of coexistence between strains, and supports a higher bottleneck size in the range of 35–145 virions transmitted from donor to recipient host. Thanks to its simplicity and generality, such framework could be applied to other ecological scenarios of microbial competition, enabling a more complex and nuanced view of possible outcomes between two strains, beyond competitive exclusion.
Collapse
Affiliation(s)
- Afonso Dimas Martins
- Mathematical Modeling of Biological Processes Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Departamento de Estatística e Investigacão Operacional, Faculdade de Ciências, Universidade de Lisbon, Lisbon, Portugal
| | - Erida Gjini
- Mathematical Modeling of Biological Processes Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
46
|
Lumby CK, Zhao L, Breuer J, Illingworth CJR. A large effective population size for established within-host influenza virus infection. eLife 2020; 9:e56915. [PMID: 32773034 PMCID: PMC7431133 DOI: 10.7554/elife.56915] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
Strains of the influenza virus form coherent global populations, yet exist at the level of single infections in individual hosts. The relationship between these scales is a critical topic for understanding viral evolution. Here we investigate the within-host relationship between selection and the stochastic effects of genetic drift, estimating an effective population size of infection Ne for influenza infection. Examining whole-genome sequence data describing a chronic case of influenza B in a severely immunocompromised child we infer an Ne of 2.5 × 107 (95% confidence range 1.0 × 107 to 9.0 × 107) suggesting that genetic drift is of minimal importance during an established influenza infection. Our result, supported by data from influenza A infection, suggests that positive selection during within-host infection is primarily limited by the typically short period of infection. Atypically long infections may have a disproportionate influence upon global patterns of viral evolution.
Collapse
Affiliation(s)
- Casper K Lumby
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
| | - Lei Zhao
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
| | - Judith Breuer
- Great Ormond Street HospitalLondonUnited Kingdom
- Division of Infection and Immunity, University College LondonLondonUnited Kingdom
| | - Christopher JR Illingworth
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
- Department of Applied Mathematics and Theoretical Physics, University of CambridgeCambridgeUnited Kingdom
- Department of Computer Science, Institute of Biotechnology, University of HelsinkiHelsinkiFinland
| |
Collapse
|
47
|
Abstract
Infectious disease research spans scales from the molecular to the global—from specific mechanisms of pathogen drug resistance, virulence, and replication to the movement of people, animals, and pathogens around the world. All of these research areas have been impacted by the recent growth of large-scale data sources and data analytics. Some of these advances rely on data or analytic methods that are common to most biomedical data science, while others leverage the unique nature of infectious disease, namely its communicability. This review outlines major research progress in the past few years and highlights some remaining opportunities, focusing on data or methodological approaches particular to infectious disease.
Collapse
Affiliation(s)
- Peter M. Kasson
- Department of Biomedical Engineering and Department of Molecular Physiology, University of Virginia, Charlottesville, Virginia 22908, USA
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 752 37 Uppsala, Sweden
| |
Collapse
|
48
|
Cheng C, Li J, Liu W, Xu L, Zhang Z. Modeling analysis revealed the distinct global transmission patterns of influenza A viruses and their influencing factors. Integr Zool 2020; 16:788-797. [PMID: 32649020 PMCID: PMC9292709 DOI: 10.1111/1749-4877.12469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Influenza A virus has caused huge damage to human health and poultry production worldwide, but its global transmission patterns and influencing factors remain unclear. Here, by using the Nearest Genetic Distance Approach with genetic sequences data, we reconstructed the global transmission patterns of 4 most common subtypes of influenza A virus (H1N1, H3N2, H5N1, and H7N9) and analyzed associations of transmission velocity of these influenza viruses with environmental factors. We found that the transmission patterns of influenza viruses and their associations with environmental factors were closely related to their host properties. H1N1 and H3N2, which are mainly held by humans, are transmitted between regions at high velocity and over long distances, which may be due to human transportation via airplane; while H5N1 and H7N9, which are mainly carried by animals, are transmitted locally at short distances and at low velocity, which may be facilitated by poultry transportation via railways or high ways. H1N1 and H3N2 spread faster in cold seasons, while H5N1 spread faster in both cold and warm seasons, and H7N9 spread faster in wet seasons. H1N1, H3N2, and H5N1 spread faster in places with both high and low human densities. Our study provided novel insights into the global transmission patterns, processes, and management strategies for influenza under accelerated global change.
Collapse
Affiliation(s)
- Chaoyuan Cheng
- State Key Laboratory of Integrated Management on Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jing Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lei Xu
- Ministry of Education Key Laboratory for Earth System Modeling, Department of Earth System Science, Tsinghua University, Beijing, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management on Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
49
|
Sapoval N, Mahmoud M, Jochum MD, Liu Y, Elworth RAL, Wang Q, Albin D, Ogilvie H, Lee MD, Villapol S, Hernandez KM, Berry IM, Foox J, Beheshti A, Ternus K, Aagaard KM, Posada D, Mason CE, Sedlazeck F, Treangen TJ. Hidden genomic diversity of SARS-CoV-2: implications for qRT-PCR diagnostics and transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.02.184481. [PMID: 32637955 PMCID: PMC7337385 DOI: 10.1101/2020.07.02.184481] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The COVID-19 pandemic has sparked an urgent need to uncover the underlying biology of this devastating disease. Though RNA viruses mutate more rapidly than DNA viruses, there are a relatively small number of single nucleotide polymorphisms (SNPs) that differentiate the main SARS-CoV-2 clades that have spread throughout the world. In this study, we investigated over 7,000 SARS-CoV-2 datasets to unveil both intrahost and interhost diversity. Our intrahost and interhost diversity analyses yielded three major observations. First, the mutational profile of SARS-CoV-2 highlights iSNV and SNP similarity, albeit with high variability in C>T changes. Second, iSNV and SNP patterns in SARS-CoV-2 are more similar to MERS-CoV than SARS-CoV-1. Third, a significant fraction of small indels fuel the genetic diversity of SARS-CoV-2. Altogether, our findings provide insight into SARS-CoV-2 genomic diversity, inform the design of detection tests, and highlight the potential of iSNVs for tracking the transmission of SARS-CoV-2.
Collapse
Affiliation(s)
- Nicolae Sapoval
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Medhat Mahmoud
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Michael D. Jochum
- Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
| | - Yunxi Liu
- Department of Computer Science, Rice University, Houston, TX, USA
| | | | - Qi Wang
- Systems, Synthetic, and Physical Biology (SSPB) Graduate Program, Houston, TX
| | - Dreycey Albin
- Systems, Synthetic, and Physical Biology (SSPB) Graduate Program, Houston, TX
| | - Huw Ogilvie
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Michael D. Lee
- Exobiology Branch, NASA Ames Research Center, Mountain View, CA
- Blue Marble Space Institute of Science, Seattle, WA
| | | | - Kyle M. Hernandez
- Department of Medicine, University of Chicago, Chicago, IL
- Center for Translational Data Science, University of Chicago, Chicago, IL
| | | | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA
| | - Krista Ternus
- Signature Science, LLC, 8329 North Mopac Expressway, Austin TX 78759
| | | | - David Posada
- Biomedical Research Center (CINBIO), University of Vigo, 36310 Vigo, Spain
- Department of Biochemistry, Genetics and Immunology School of Biology, University of Vigo, Vigo, Spain
- Galicia Sur Health Research Institute, 36310 Vigo, Spain
| | - Christopher E. Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Fritz Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Todd J. Treangen
- Department of Computer Science, Rice University, Houston, TX, USA
| |
Collapse
|
50
|
Inferring Transmission Bottleneck Size from Viral Sequence Data Using a Novel Haplotype Reconstruction Method. J Virol 2020; 94:JVI.00014-20. [PMID: 32295920 PMCID: PMC7307158 DOI: 10.1128/jvi.00014-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
Viral populations undergo a repeated cycle of within-host growth followed by transmission. Viral evolution is affected by each stage of this cycle. The number of viral particles transmitted from one host to another, known as the transmission bottleneck, is an important factor in determining how the evolutionary dynamics of the population play out, restricting the extent to which the evolved diversity of the population can be passed from one host to another. Previous study of viral sequence data has suggested that the transmission bottleneck size for influenza A transmission between human hosts is small. Reevaluating these data using a novel and improved method, we largely confirm this result, albeit that we infer a slightly higher bottleneck size in some cases, of between 1 and 13 virions. While a tight bottleneck operates in human influenza transmission, it is not extreme in nature; some diversity can be meaningfully retained between hosts. The transmission bottleneck is defined as the number of viral particles that transmit from one host to establish an infection in another. Genome sequence data have been used to evaluate the size of the transmission bottleneck between humans infected with the influenza virus; however, the methods used to make these estimates have some limitations. Specifically, viral allele frequencies, which form the basis of many calculations, may not fully capture a process which involves the transmission of entire viral genomes. Here, we set out a novel approach for inferring viral transmission bottlenecks; our method combines an algorithm for haplotype reconstruction with maximum likelihood methods for bottleneck inference. This approach allows for rapid calculation and performs well when applied to data from simulated transmission events; errors in the haplotype reconstruction step did not adversely affect inferences of the population bottleneck. Applied to data from a previous household transmission study of influenza A infection, we confirm the result that the majority of transmission events involve a small number of viruses, albeit with slightly looser bottlenecks being inferred, with between 1 and 13 particles transmitted in the majority of cases. While influenza A transmission involves a tight population bottleneck, the bottleneck is not so tight as to universally prevent the transmission of within-host viral diversity. IMPORTANCE Viral populations undergo a repeated cycle of within-host growth followed by transmission. Viral evolution is affected by each stage of this cycle. The number of viral particles transmitted from one host to another, known as the transmission bottleneck, is an important factor in determining how the evolutionary dynamics of the population play out, restricting the extent to which the evolved diversity of the population can be passed from one host to another. Previous study of viral sequence data has suggested that the transmission bottleneck size for influenza A transmission between human hosts is small. Reevaluating these data using a novel and improved method, we largely confirm this result, albeit that we infer a slightly higher bottleneck size in some cases, of between 1 and 13 virions. While a tight bottleneck operates in human influenza transmission, it is not extreme in nature; some diversity can be meaningfully retained between hosts.
Collapse
|