1
|
Zhang X, Ji X, Wang L, Chi L, Li C, Wen S, Chen H. STRsensor: a computationally efficient method for STR allele-typing from massively parallel sequencing data. Brief Bioinform 2024; 26:bbae637. [PMID: 39665493 PMCID: PMC11635639 DOI: 10.1093/bib/bbae637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/16/2024] [Accepted: 11/22/2024] [Indexed: 12/13/2024] Open
Abstract
Short tandem repeats (STRs) represent one of the most polymorphic variations in the human genome, finding extensive applications in forensics, population genetics and medical genetics. In contrast to the traditional capillary electrophoresis (CE) method, genotyping STRs using massive parallel sequencing technology offers enhanced sensitivity and accuracy. However, current methods are mainly designed for target sequencing with higher coverage for a specific STR locus, thereby constraining the utility of STRs in low- and medium-coverage whole genome sequencing (WGS) data. Here, we introduce STRsensor, a method designed to type STR alleles in low-coverage WGS data and target sequencing data, achieving a significant high detection ratio and accuracy. STRsensor employs two methods for STR allele-typing: the Kmers-based method and the CIGAR-based method. Furthermore, by incorporating a model for PCR stutters, STRsensor greatly enhances the accuracy of STR allele typing. With simulation data, we demonstrate that STRsensor achieves a detection ratio of 100$\%$ and an accuracy of 99.37$\%$ for a 30$\times $ WGS data, outperforming the existing methods, such as STRait Razor, STRinNGS, and HipSTR. When applied to real target sequencing data from 687 individuals, STRsensor achieves a detection ratio of 99.64$\%$ and an accuracy of 99.99$\%$. Moreover, STRsensor is a computationally efficient method that runs 79 times faster than HipSTR and 10 000 times faster than STRinNGS. STRsensor is freely available on GitHub: https://github.com/ChenHuaLab/STRsensor.
Collapse
Affiliation(s)
- Xiaolong Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianchao Ji
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingxiang Wang
- Institute of Archaeological Science, Fudan University, Shanghai 200032, China
| | - Lianjiang Chi
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Chengtao Li
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shaoqing Wen
- Institute of Archaeological Science, Fudan University, Shanghai 200032, China
| | - Hua Chen
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650023, China
| |
Collapse
|
2
|
Yilmaz G, Chan M, Lau CHF, Capitani S, Kang M, Charron P, Hoover E, Topp E, Guan J. How Gut Microbiome Perturbation Caused by Antibiotic Pre-Treatments Affected the Conjugative Transfer of Antimicrobial Resistance Genes. Microorganisms 2024; 12:2148. [PMID: 39597538 PMCID: PMC11596856 DOI: 10.3390/microorganisms12112148] [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: 09/21/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
The global spread of antimicrobial resistance genes (ARGs) poses a significant threat to public health. While antibiotics effectively treat bacterial infections, they can also induce gut dysbiosis, the severity of which varies depending on the specific antibiotic treatment used. However, it remains unclear how gut dysbiosis affects the mobility and dynamics of ARGs. To address this, mice were pre-treated with streptomycin, ampicillin, or sulfamethazine, and then orally inoculated with Salmonella enterica serovar Typhimurium and S. Heidelberg carrying a multi-drug resistance IncA/C plasmid. The streptomycin pre-treatment caused severe microbiome perturbation, promoting the high-density colonization of S. Heidelberg and S. Typhimurium, and enabling an IncA/C transfer from S. Heidelberg to S. Typhimurium and a commensal Escherichia coli. The ampicillin pre-treatment induced moderate microbiome perturbation, supporting only S. Heidelberg colonization and the IncA/C transfer to commensal E. coli. The sulfamethazine pre-treatment led to mild microbiome perturbation, favoring neither Salmonella spp. colonization nor a conjugative plasmid transfer. The degree of gut dysbiosis also influenced the enrichment or depletion of the ARGs associated with mobile plasmids or core commensal bacteria, respectively. These findings underscore the significance of pre-existing gut dysbiosis induced by various antibiotic treatments on ARG dissemination and may inform prudent antibiotic use practices.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Maria Chan
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Calvin Ho-Fung Lau
- Ottawa Laboratory-Carling, Canadian Food Inspection Agency, Ottawa, ON K1A 0Z, Canada; (C.H.-F.L.)
| | - Sabrina Capitani
- Ottawa Laboratory-Carling, Canadian Food Inspection Agency, Ottawa, ON K1A 0Z, Canada; (C.H.-F.L.)
| | - Mingsong Kang
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Philippe Charron
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Emily Hoover
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Edward Topp
- Agroecology Research Unit, INRAE, University of Burgundy, 21065 Dijon, France;
| | - Jiewen Guan
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| |
Collapse
|
3
|
Wang Y, Wang S, Zang Z, Li B, Liu G, Huang H, Zhao X. Molecular and transcriptomic analysis of the ovary during laying and brooding stages in Zhedong white geese ( Anser cygnoides domesticus). Br Poult Sci 2024; 65:631-644. [PMID: 38916443 DOI: 10.1080/00071668.2024.2364351] [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: 12/06/2023] [Accepted: 05/13/2024] [Indexed: 06/26/2024]
Abstract
1. This study investigates the molecular mechanisms affecting brooding in Zhedong white geese by examining differences in reproductive endocrine levels, ovarian histology and transcriptomics.2. Twenty 18-month-old Zhedong white geese were selected to examine their ovaries using histological, biochemical, molecular biological, and high-throughput sequencing techniques during the laying and brooding periods.3. The results showed that the number of atretic follicles and apoptotic cells in the ovaries increased significantly (p < 0.05), the levels of follicle-stimulating hormone, luteinising hormone, gonadotropin-releasing hormone and oestradiol decreased significantly (p < 0.05), and the level of prolactin increased significantly (p < 0.01) during the brooding stage.4. In broody geese, the expression of CASP3, CASP9, P53, BAX, and Cyt-c were considerably higher (p < 0.05), but BCL2 expression was significantly lower (p < 0.05).5. In ovarian tissues, 260 differentially expressed lncRNAs, 13 differentially expressed miRNA and 60 differentially expressed mRNA were all discovered using transcriptome sequencing analysis. Functional enrichment analysis revealed that the differentially expressed mRNA and non-coding RNA target genes were primarily involved in ECM-receptor interaction, cell adhesion, cardiac muscle contraction, mTOR signalling, and the calcium signalling pathway.6. In conclusion, follicular atrophy and apoptosis occurred in the ovaries and serum reproductive hormone levels were significantly changed during the brooding period of Zhedong white geese. COL3A1, COL1A2, GRIA1, RNF152, miR-192, and miR-194 may be important candidates for the regulation of brooding behaviour, with the mTOR signalling pathway playing a key role.
Collapse
Affiliation(s)
- Y Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - S Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Z Zang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - B Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - G Liu
- Animal Husbandry Institute, Heilongjiang Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - H Huang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - X Zhao
- Animal Husbandry Institute, Heilongjiang Academy of Agricultural Sciences, Harbin, People's Republic of China
| |
Collapse
|
4
|
Piñana M, González-Sánchez A, Andrés C, Abanto M, Vila J, Esperalba J, Moral N, Espartosa E, Saubi N, Creus A, Codina MG, Folgueira D, Martinez-Urtaza J, Pumarola T, Antón A. The emergence, impact, and evolution of human metapneumovirus variants from 2014 to 2021 in Spain. J Infect 2023:S0163-4453(23)00262-1. [PMID: 37178807 DOI: 10.1016/j.jinf.2023.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Human metapneumovirus (HMPV) is an important aetiologic agent of respiratory tract infection (RTI). This study aimed to describe the prevalence, genetic diversity, and evolutionary dynamics of HMPV. METHODS Laboratory-confirmed HMPV were characterised based on partial-coding G gene sequences with MEGA.v6.0. WGS was performed with Illumina, and evolutionary analyses with Datamonkey and Nextstrain. RESULTS HMPV prevalence was 2.5%, peaking in February-April and with an alternation in the predominance of HMPV-A and -B until the emergence of SARS-CoV-2, not circulating until summer and autumn-winter 2021, with a higher prevalence and with the almost only circulation of A2c111dup. G and SH proteins were the most variable, and 70% of F protein was under negative selection. Mutation rate of HMPV genome was 6.95 ×10-4 substitutions/site/year. CONCLUSION HMPV showed a significant morbidity until the emergence of SARS-CoV-2 pandemic in 2020, not circulating again until summer and autumn 2021, with a higher prevalence and with almost the only circulation of A2c111dup, probably due to a more efficient immune evasion mechanism. The F protein showed a very conserved nature, supporting the need for steric shielding. The tMRCA showed a recent emergence of the A2c variants carrying duplications, supporting the importance of virological surveillance.
Collapse
Affiliation(s)
- Maria Piñana
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alejandra González-Sánchez
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Andrés
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Michel Abanto
- Genomics and Bioinformatics Unit, Scientific and Technological Bioresource Nucleus (BIOREN). Universidad de La Frontera, Temuco, Chile
| | - Jorgina Vila
- Paediatric Hospitalization Unit, Paediatrics Department, Hospital Universitari Maternoinfantil Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juliana Esperalba
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Noelia Moral
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Elena Espartosa
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Narcís Saubi
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Creus
- Paediatric Hospitalization Unit, Paediatrics Department, Hospital Universitari Maternoinfantil Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Gema Codina
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Dolores Folgueira
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Jaime Martinez-Urtaza
- Department of Genetics and Microbiology, School of Biosciences, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | - Tomàs Pumarola
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Andrés Antón
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Local monitoring of SARS-CoV-2 variants in two large California counties in 2021. Sci Rep 2022; 12:17046. [PMID: 36221029 PMCID: PMC9553084 DOI: 10.1038/s41598-022-21481-0] [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: 11/17/2021] [Accepted: 09/27/2022] [Indexed: 12/30/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to persist due to mutations resulting in newer, more infectious variants of concern. We aimed to leverage an ongoing private SARS-CoV-2 testing laboratory's infrastructure to monitor SARS-CoV-2 variants in two large California counties. Study enrollment was offered to adults aged 18 years or older in Los Angeles County and Riverside County who recently tested positive for SARS-CoV-2 with a polymerase chain reaction (PCR) assay. A cycle threshold value less than or equal to 30 cycles was considered a positive test for sequencing purposes. Within 5 days of study enrollment, clinician-monitored, self-collected oral fluid and anterior nares swab specimens were obtained from participants. Specimens were transported and stored at 8 °C or cooler. Samples underwent ribonucleic acid extraction, library preparation, and sequencing. SARS-CoV-2 lineages were identified using sequencing data. Participant and genomic data were analyzed using statistical tools and visualized with toolkits. The study was approved by Advarra Institutional Review Board (Pro00053729). From May 27, 2021 to September 9, 2021, 503 individuals were enrolled and underwent specimen collection. Of the 503 participants, 238 (47.3%) participants were women, 329 (63.6%) participants were vaccinated, and 221 (43.9%) participants were of Hispanic or Spanish origin. Of the cohort, 496 (98.6%) participants had symptoms at the time of collection. Among the 503 samples, 443 (88.1%) nasal specimens and 353 (70.2%) oral specimens yielded positive sequencing results. Over our study period, the prevalence of the Alpha variant of SARS-CoV-2 decreased (initially 23.1% [95% confidence interval (95% CI): 0-0.49%] to 0% [95% CI 0.0-0.0%]) as the prevalence of the Delta variant of SARS-CoV-2 increased (initially 33.3% [95% CI 0.0-100.0%] to 100.0% [95% CI 100.0-100.0%]). A strain that carried mutations of both Delta and Mu was identified. We found that outpatient SARS-CoV-2 variant surveillance could be conducted in a timely and accurate manner. The prevalence of different variants changed over time. A higher proportion of nasal specimens yielded results versus oral specimens. Timely and regional outpatient SARS-CoV-2 variant surveillance could be used for public health efforts to identify changes in SARS-CoV-2 strain epidemiology.
Collapse
|
6
|
COVID-19 Whole-Genome Resequencing with Redundant Tiling PCR and Subtract-Based Amplicon Normalization Successfully Characterized SARS-CoV-2 Variants in Clinical Specimens. Interdiscip Perspect Infect Dis 2022; 2022:2109641. [PMID: 36212105 PMCID: PMC9534710 DOI: 10.1155/2022/2109641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
With an increasing number of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) sequences gathered worldwide, we recognize that deletion mutants and nucleotide substitutions that may affect whole-genome sequencing are accumulating. Here, we propose an additional strategy for tiling PCR for whole-genome resequencing, which can make the pipeline robust for mutations at the primer annealing site by a redundant amplicon scheme. We further demonstrated that subtracting overrepresented amplicons from the multiplex PCR products reduced the bias of the next-generation sequencing (NGS) library, resulting in decreasing required sequencing reads per sample. We applied this sequencing strategy to clinical specimens collected in Bangladesh. More than 80% out of the 304 samples were successfully sequenced. Less than 5% were ambiguous nucleotides, and several known variants were detected. With the additional strategies presented here, we believe that whole-genome resequencing of SARS-CoV-2 from clinical samples can be optimized.
Collapse
|
7
|
Harari S, Tahor M, Rutsinsky N, Meijer S, Miller D, Henig O, Halutz O, Levytskyi K, Ben-Ami R, Adler A, Paran Y, Stern A. Drivers of adaptive evolution during chronic SARS-CoV-2 infections. Nat Med 2022; 28:1501-1508. [PMID: 35725921 PMCID: PMC9307477 DOI: 10.1038/s41591-022-01882-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022]
Abstract
In some immunocompromised patients with chronic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, considerable adaptive evolution occurs. Some substitutions found in chronic infections are lineage-defining mutations in variants of concern (VOCs), which has led to the hypothesis that VOCs emerged from chronic infections. In this study, we searched for drivers of VOC-like emergence by consolidating sequencing results from a set of 27 chronic infections. Most substitutions in this set reflected lineage-defining VOC mutations; however, a subset of mutations associated with successful global transmission was absent from chronic infections. We further tested the ability to associate antibody evasion mutations with patient-specific and virus-specific features and found that viral rebound is strongly correlated with the emergence of antibody evasion. We found evidence for dynamic polymorphic viral populations in most patients, suggesting that a compromised immune system selects for antibody evasion in particular niches in a patient’s body. We suggest that a tradeoff exists between antibody evasion and transmissibility and that extensive monitoring of chronic infections is necessary to further understanding of VOC emergence. Analysis of mutations that arise in chronic SARS-CoV-2 infections shows both overlap and differences with mutations present in pandemic viral variants of concern, highlighting their distinct drivers of evolution.
Collapse
Affiliation(s)
- Sheri Harari
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel.,Edmond J. Safra Center for Bioinformatics at Tel Aviv University, Tel Aviv, Israel
| | - Maayan Tahor
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Natalie Rutsinsky
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Suzy Meijer
- Department of Infectious Diseases and Epidemiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Danielle Miller
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel.,Edmond J. Safra Center for Bioinformatics at Tel Aviv University, Tel Aviv, Israel
| | - Oryan Henig
- Department of Infectious Diseases and Epidemiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ora Halutz
- Clinical Microbiology Laboratory, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Katia Levytskyi
- Department of Infectious Diseases and Epidemiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronen Ben-Ami
- Department of Infectious Diseases and Epidemiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amos Adler
- Department of Infectious Diseases and Epidemiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Paran
- Department of Infectious Diseases and Epidemiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Stern
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel. .,Edmond J. Safra Center for Bioinformatics at Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
8
|
Kasugai Y, Kohmoto T, Taniyama Y, Koyanagi YN, Usui Y, Iwase M, Oze I, Yamaguchi R, Ito H, Imoto I, Matsuo K. Association between germline pathogenic variants and breast cancer risk in Japanese women: The HERPACC study. Cancer Sci 2022; 113:1451-1462. [PMID: 35218119 PMCID: PMC8990868 DOI: 10.1111/cas.15312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 12/24/2022] Open
Abstract
Approximately 5%-10% of breast cancers are hereditary, caused by germline pathogenic variants (GPVs) in breast cancer predisposition genes. To date, most studies of the prevalence of GPVs and risk of breast cancer for each gene based on cases and noncancer controls have been conducted in Europe and the United States, and little information from Japanese populations is available. Furthermore, no studies considered confounding by established environmental factors and single-nucleotide polymorphisms (SNPs) identified in genome-wide association studies (GWAS) together in GPV evaluation. To evaluate the association between GPVs in nine established breast cancer predisposition genes including BRCA1/2 and breast cancer risk in Japanese women comprehensively, we conducted a case-control study within the Hospital-based Epidemiologic Research Program at Aichi Cancer Center (629 cases and 1153 controls). The associations between GPVs and the risk of breast cancer were assessed by odds ratios (OR) and 95% confidence intervals (CI) using logistic regression models adjusted for potential confounders. A total of 25 GPVs were detected among all cases (4.0%: 95% CI: 2.6-5.9), whereas four individuals carried GPVs in all controls (0.4%). The OR for breast cancer by all GPVs and by GPVs in BRCA1/2 was 12.2 (4.4-34.0, p = 1.74E-06) and 16.0 (4.2-60.9, p = 5.03E-0.5), respectively. A potential confounding with GPVs was observed for the GWAS-identified SNPs, whereas not for established environmental risk factors. In conclusion, GPVs increase the risk of breast cancer in Japanese women regardless of environmental factors and GWAS-identified SNPs. Future studies investigating interactions with environment and SNPs are warranted.
Collapse
Grants
- Aichi Cancer Center Joint Research Project on Priority Areas
- Grant-in-Aid for the Third Term Comprehensive 10-year Strategy for Cancer Control from the Ministry of Health, Labour and Welfare of Japan
- JP15ck0106177 AMED
- JP21ck0106553 AMED
- Cancer BioBank Aichi
- 17015018 Grants-in-Aid for Scientific Research from the Ministry of Education, Science, Sports, Culture and Technology of Japan
- 221S0001 Grants-in-Aid for Scientific Research from the Ministry of Education, Science, Sports, Culture and Technology of Japan
- JP16H06277(CoBiA) Grants-in-Aid for Scientific Research from the Ministry of Education, Science, Sports, Culture and Technology of Japan
- JP18H03045 Grants-in-Aid for Scientific Research from the Ministry of Education, Science, Sports, Culture and Technology of Japan
- AMED
- Grants‐in‐Aid for Scientific Research from the Ministry of Education, Science, Sports, Culture and Technology of Japan
Collapse
Affiliation(s)
- Yumiko Kasugai
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
- Department of Cancer EpidemiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Tomohiro Kohmoto
- Division of Cancer Systems BiologyAichi Cancer Center Research InstituteNagoyaJapan
- Department of Human GeneticsGraduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
| | - Yukari Taniyama
- Division of Cancer Information and ControlAichi Cancer Center Research InstituteNagoyaJapan
| | - Yuriko N. Koyanagi
- Division of Cancer Information and ControlAichi Cancer Center Research InstituteNagoyaJapan
| | - Yoshiaki Usui
- Division of Cancer Information and ControlAichi Cancer Center Research InstituteNagoyaJapan
- Laboratory for Genotyping DevelopmentRIKEN Center for Integrative Medical SciencesYokohamaJapan
| | - Madoka Iwase
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
| | - Isao Oze
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
| | - Rui Yamaguchi
- Division of Cancer Systems BiologyAichi Cancer Center Research InstituteNagoyaJapan
| | - Hidemi Ito
- Division of Cancer Information and ControlAichi Cancer Center Research InstituteNagoyaJapan
| | - Issei Imoto
- Aichi Cancer Center Research InstituteNagoyaJapan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
- Department of Cancer EpidemiologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
9
|
The ViReflow pipeline enables user friendly large scale viral consensus genome reconstruction. Sci Rep 2022; 12:5077. [PMID: 35332213 PMCID: PMC8943356 DOI: 10.1038/s41598-022-09035-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/15/2022] [Indexed: 11/18/2022] Open
Abstract
Throughout the COVID-19 pandemic, massive sequencing and data sharing efforts enabled the real-time surveillance of novel SARS-CoV-2 strains throughout the world, the results of which provided public health officials with actionable information to prevent the spread of the virus. However, with great sequencing comes great computation, and while cloud computing platforms bring high-performance computing directly into the hands of all who seek it, optimal design and configuration of a cloud compute cluster requires significant system administration expertise. We developed ViReflow, a user-friendly viral consensus sequence reconstruction pipeline enabling rapid analysis of viral sequence datasets leveraging Amazon Web Services (AWS) cloud compute resources and the Reflow system. ViReflow was developed specifically in response to the COVID-19 pandemic, but it is general to any viral pathogen. Importantly, when utilized with sufficient compute resources, ViReflow can trim, map, call variants, and call consensus sequences from amplicon sequence data from 1000 SARS-CoV-2 samples at 1000X depth in < 10 min, with no user intervention. ViReflow’s simplicity, flexibility, and scalability make it an ideal tool for viral molecular epidemiological efforts.
Collapse
|
10
|
SARS-CoV-2 Genome Sequences, Including One with an ORF7a Deletion, Obtained from Patients in Bangladesh. Microbiol Resour Announc 2021; 10:e0076421. [PMID: 34881982 PMCID: PMC8656380 DOI: 10.1128/mra.00764-21] [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] [Indexed: 11/20/2022] Open
Abstract
Genomic sequences from a complete SARS-CoV-2 open reading frame (ORF) were obtained from 24 patients diagnosed in May 2020 in Dhaka, Bangladesh. All sequences belonged to clade 20A or 20B, and none were variants of concern. Interestingly, one sequence showed a 161-nucleotide deletion in ORF7a.
Collapse
|
11
|
Kayani MUR, Huang W, Feng R, Chen L. Genome-resolved metagenomics using environmental and clinical samples. Brief Bioinform 2021; 22:bbab030. [PMID: 33758906 PMCID: PMC8425419 DOI: 10.1093/bib/bbab030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/29/2020] [Accepted: 01/20/2021] [Indexed: 12/25/2022] Open
Abstract
Recent advances in high-throughput sequencing technologies and computational methods have added a new dimension to metagenomic data analysis i.e. genome-resolved metagenomics. In general terms, it refers to the recovery of draft or high-quality microbial genomes and their taxonomic classification and functional annotation. In recent years, several studies have utilized the genome-resolved metagenome analysis approach and identified previously unknown microbial species from human and environmental metagenomes. In this review, we describe genome-resolved metagenome analysis as a series of four necessary steps: (i) preprocessing of the sequencing reads, (ii) de novo metagenome assembly, (iii) genome binning and (iv) taxonomic and functional analysis of the recovered genomes. For each of these four steps, we discuss the most commonly used tools and the currently available pipelines to guide the scientific community in the recovery and subsequent analyses of genomes from any metagenome sample. Furthermore, we also discuss the tools required for validation of assembly quality as well as for improving quality of the recovered genomes. We also highlight the currently available pipelines that can be used to automate the whole analysis without having advanced bioinformatics knowledge. Finally, we will highlight the most widely adapted and actively maintained tools and pipelines that can be helpful to the scientific community in decision making before they commence the analysis.
Collapse
Affiliation(s)
- Masood ur Rehman Kayani
- Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Shanghai Jiao Tong University, School of Medicine, Shanghai 2,000,025, China
| | - Wanqiu Huang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, School of Medicine, Shanghai 200,000, China
| | - Ru Feng
- Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Shanghai Jiao Tong University, School of Medicine, Shanghai 2,000,025, China
| | - Lei Chen
- Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Shanghai Jiao Tong University, School of Medicine, Shanghai 2,000,025, China
| |
Collapse
|
12
|
Kustin T, Harel N, Finkel U, Perchik S, Harari S, Tahor M, Caspi I, Levy R, Leshchinsky M, Ken Dror S, Bergerzon G, Gadban H, Gadban F, Eliassian E, Shimron O, Saleh L, Ben-Zvi H, Keren Taraday E, Amichay D, Ben-Dor A, Sagas D, Strauss M, Shemer Avni Y, Huppert A, Kepten E, Balicer RD, Netzer D, Ben-Shachar S, Stern A. Evidence for increased breakthrough rates of SARS-CoV-2 variants of concern in BNT162b2-mRNA-vaccinated individuals. Nat Med 2021; 27:1379-1384. [PMID: 34127854 PMCID: PMC8363499 DOI: 10.1038/s41591-021-01413-7] [Citation(s) in RCA: 248] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/26/2021] [Indexed: 12/28/2022]
Abstract
The BNT162b2 mRNA vaccine is highly effective against SARS-CoV-2. However, apprehension exists that variants of concern (VOCs) may evade vaccine protection, due to evidence of reduced neutralization of the VOCs B.1.1.7 and B.1.351 by vaccine sera in laboratory assays. We performed a matched cohort study to examine the distribution of VOCs in infections of BNT162b2 mRNA vaccinees from Clalit Health Services (Israel) using viral genomic sequencing, and hypothesized that if vaccine effectiveness against a VOC is reduced, its proportion among breakthrough cases would be higher than in unvaccinated controls. Analyzing 813 viral genome sequences from nasopharyngeal swabs, we showed that vaccinees who tested positive at least 7 days after the second dose were disproportionally infected with B.1.351, compared with controls. Those who tested positive between 2 weeks after the first dose and 6 days after the second dose were disproportionally infected by B.1.1.7. These findings suggest reduced vaccine effectiveness against both VOCs within particular time windows. Our results emphasize the importance of rigorously tracking viral variants, and of increasing vaccination to prevent the spread of VOCs.
Collapse
Affiliation(s)
- Talia Kustin
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Noam Harel
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Uriah Finkel
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat Gan, Israel
| | - Shay Perchik
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat Gan, Israel
| | - Sheri Harari
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Maayan Tahor
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Itamar Caspi
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rachel Levy
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michael Leshchinsky
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat Gan, Israel
| | - Shifra Ken Dror
- Clalit Health Services, Central Laboratories, Haifa and Western Galilee, Nesher, Israel
| | - Galit Bergerzon
- Clalit Health Services, Central Laboratories, Haifa and Western Galilee, Nesher, Israel
| | - Hala Gadban
- Clalit Health Services, Central Laboratories, Haifa and Western Galilee, Nesher, Israel
| | - Faten Gadban
- Clalit Health Services, Central Laboratories, Haifa and Western Galilee, Nesher, Israel
| | - Eti Eliassian
- Progenin Laboratories, Jerusalem District, Clalit Health Services, Tel Aviv, Israel
| | - Orit Shimron
- Progenin Laboratories, Jerusalem District, Clalit Health Services, Tel Aviv, Israel
| | - Loulou Saleh
- Microbiology lab, Rabin Medical Center, Beilinson Hospital, Petah Tiqva, Israel
| | - Haim Ben-Zvi
- Microbiology lab, Rabin Medical Center, Beilinson Hospital, Petah Tiqva, Israel
| | | | - Doron Amichay
- Central Laboratory, Clalit Health Services, Tel Aviv, Israel
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheba, Israel
| | - Anat Ben-Dor
- Central Laboratory, Clalit Health Services, Tel Aviv, Israel
| | - Dana Sagas
- Microbiology Laboratory, Emek Medical Center, Afula, Israel
| | - Merav Strauss
- Microbiology Laboratory, Emek Medical Center, Afula, Israel
| | - Yonat Shemer Avni
- Laboratory of Clinical Virology, Soroka University Medical Center, Beersheba, Israel
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheba, Israel
| | - Amit Huppert
- The Bio-statistical and Bio-mathematical Unit, The Gertner Institute for Epidemiology and Health Policy Research, Chaim Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel
- The Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Eldad Kepten
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat Gan, Israel
| | - Ran D Balicer
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat Gan, Israel
| | | | - Shay Ben-Shachar
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat Gan, Israel.
- The Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| | - Adi Stern
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
13
|
Lind A, Barlinn R, Landaas ET, Andresen LL, Jakobsen K, Fladeby C, Nilsen M, Bjørnstad PM, Sundaram AYM, Ribarska T, Müller F, Gilfillan GD, Holberg-Petersen M. Rapid SARS-CoV-2 variant monitoring using PCR confirmed by whole genome sequencing in a high-volume diagnostic laboratory. J Clin Virol 2021; 141:104906. [PMID: 34273860 PMCID: PMC8262397 DOI: 10.1016/j.jcv.2021.104906] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The emerging SARS-CoV-2 variants of concern (VoC), B.1.1.7, B.1.351 and P.1, with increased transmission and/or immune evasion, emphasise the need for broad and rapid variant monitoring. Our high-volume laboratory introduced a PCR variant assay (Variant PCR) in January 2021 based on the protocol by Vogels et al. STUDY DESIGN: To assess whether Variant PCR could be used for rapid B.1.1.7, B.1.351 and P.1 screening, all positive SARS-CoV-2 airway samples were prospectively tested in parallel using both the Variant PCR and whole genome sequencing (WGS). RESULTS In total 1,642 SARS-CoV-2 positive samples from individual patients were tested within a time span of 4 weeks. For all samples with valid results from both Variant PCR and WGS, no VoC was missed by Variant PCR (totalling 399 VoC detected). Conversely, all of the samples identified as "other lineages" (i.e., "non-VoC lineages") by the Variant PCR, were confirmed by WGS. CONCLUSIONS The Variant PCR based on the protocol by Vogels et al., is an effective method for rapid screening for VoC, applicable for most diagnostic laboratories within a pandemic setting. WGS is still required to confirm the identity of certain variants and for continuous surveillance of emerging VoC.
Collapse
Affiliation(s)
- Andreas Lind
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Regine Barlinn
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Toverud Landaas
- Department of Microbiology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Kirsti Jakobsen
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Cathrine Fladeby
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Mariann Nilsen
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Pål Marius Bjørnstad
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Arvind Y M Sundaram
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Teodora Ribarska
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Fredrik Müller
- Department of Microbiology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gregor D Gilfillan
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | | |
Collapse
|
14
|
Mrozek D, Stępień K, Grzesik P, Małysiak-Mrozek B. A Large-Scale and Serverless Computational Approach for Improving Quality of NGS Data Supporting Big Multi-Omics Data Analyses. Front Genet 2021; 12:699280. [PMID: 34326863 PMCID: PMC8314304 DOI: 10.3389/fgene.2021.699280] [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: 04/23/2021] [Accepted: 05/28/2021] [Indexed: 11/13/2022] Open
Abstract
Various types of analyses performed over multi-omics data are driven today by next-generation sequencing (NGS) techniques that produce large volumes of DNA/RNA sequences. Although many tools allow for parallel processing of NGS data in a Big Data distributed environment, they do not facilitate the improvement of the quality of NGS data for a large scale in a simple declarative manner. Meanwhile, large sequencing projects and routine DNA/RNA sequencing associated with molecular profiling of diseases for personalized treatment require both good quality data and appropriate infrastructure for efficient storing and processing of the data. To solve the problems, we adapt the concept of Data Lake for storing and processing big NGS data. We also propose a dedicated library that allows cleaning the DNA/RNA sequences obtained with single-read and paired-end sequencing techniques. To accommodate the growth of NGS data, our solution is largely scalable on the Cloud and may rapidly and flexibly adjust to the amount of data that should be processed. Moreover, to simplify the utilization of the data cleaning methods and implementation of other phases of data analysis workflows, our library extends the declarative U-SQL query language providing a set of capabilities for data extraction, processing, and storing. The results of our experiments prove that the whole solution supports requirements for ample storage and highly parallel, scalable processing that accompanies NGS-based multi-omics data analyses.
Collapse
Affiliation(s)
- Dariusz Mrozek
- Department of Applied Informatics, Silesian University of Technology, Gliwice, Poland
| | - Krzysztof Stępień
- Department of Applied Informatics, Silesian University of Technology, Gliwice, Poland
| | - Piotr Grzesik
- Department of Applied Informatics, Silesian University of Technology, Gliwice, Poland
| | - Bożena Małysiak-Mrozek
- Department of Graphics, Computer Vision and Digital Systems, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
15
|
Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Lorenzi JCC, Park S, Schmidt F, Wang Z, Huang Y, Luo Y, Nair MS, Wang P, Schulz JE, Tessarollo L, Bylund T, Chuang GY, Olia AS, Stephens T, Teng IT, Tsybovsky Y, Zhou T, Munster V, Ho DD, Hatziioannou T, Bieniasz PD, Nussenzweig MC, Kwong PD, Casellas R. Nanobodies from camelid mice and llamas neutralize SARS-CoV-2 variants. Nature 2021; 595:278-282. [PMID: 34098567 PMCID: PMC8260353 DOI: 10.1038/s41586-021-03676-z] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/27/2021] [Indexed: 11/08/2022]
Abstract
Since the start of the COVID-19 pandemic, SARS-CoV-2 has caused millions of deaths worldwide. Although a number of vaccines have been deployed, the continual evolution of the receptor-binding domain (RBD) of the virus has challenged their efficacy. In particular, the emerging variants B.1.1.7, B.1.351 and P.1 (first detected in the UK, South Africa and Brazil, respectively) have compromised the efficacy of sera from patients who have recovered from COVID-19 and immunotherapies that have received emergency use authorization1-3. One potential alternative to avert viral escape is the use of camelid VHHs (variable heavy chain domains of heavy chain antibody (also known as nanobodies)), which can recognize epitopes that are often inaccessible to conventional antibodies4. Here, we isolate anti-RBD nanobodies from llamas and from mice that we engineered to produce VHHs cloned from alpacas, dromedaries and Bactrian camels. We identified two groups of highly neutralizing nanobodies. Group 1 circumvents antigenic drift by recognizing an RBD region that is highly conserved in coronaviruses but rarely targeted by human antibodies. Group 2 is almost exclusively focused to the RBD-ACE2 interface and does not neutralize SARS-CoV-2 variants that carry E484K or N501Y substitutions. However, nanobodies in group 2 retain full neutralization activity against these variants when expressed as homotrimers, and-to our knowledge-rival the most potent antibodies against SARS-CoV-2 that have been produced to date. These findings suggest that multivalent nanobodies overcome SARS-CoV-2 mutations through two separate mechanisms: enhanced avidity for the ACE2-binding domain and recognition of conserved epitopes that are largely inaccessible to human antibodies. Therefore, although new SARS-CoV-2 mutants will continue to emerge, nanobodies represent promising tools to prevent COVID-19 mortality when vaccines are compromised.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/isolation & purification
- CRISPR-Cas Systems
- Camelids, New World/genetics
- Camelids, New World/immunology
- Female
- Gene Editing
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Models, Molecular
- Mutation
- Neutralization Tests
- SARS-CoV-2/chemistry
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Single-Domain Antibodies/chemistry
- Single-Domain Antibodies/genetics
- Single-Domain Antibodies/immunology
- Single-Domain Antibodies/isolation & purification
- Somatic Hypermutation, Immunoglobulin/genetics
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/immunology
Collapse
Affiliation(s)
- Jianliang Xu
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA.
| | - Kai Xu
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Andrea Conte
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA
| | | | - Frauke Muecksch
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | | | - Solji Park
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA
| | - Fabian Schmidt
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
| | - Zijun Wang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Yang Luo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Pengfei Wang
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Jonathan E Schulz
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, CCR, NCI, NIH, Frederick, MD, USA
| | | | - Gwo-Yu Chuang
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Adam S Olia
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Tyler Stephens
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA
| | - I-Ting Teng
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Tongqing Zhou
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Vincent Munster
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | | | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA.
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| | - Peter D Kwong
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA.
| | - Rafael Casellas
- Lymphocyte Nuclear Biology, NIAMS, NIH, Bethesda, MD, USA.
- The NIH Regulome Project, NIH, Bethesda, MD, USA.
- Center for Cancer Research, NCI, NIH, Bethesda, MD, USA.
| |
Collapse
|
16
|
Miller D, Martin MA, Harel N, Tirosh O, Kustin T, Meir M, Sorek N, Gefen-Halevi S, Amit S, Vorontsov O, Shaag A, Wolf D, Peretz A, Shemer-Avni Y, Roif-Kaminsky D, Kopelman NM, Huppert A, Koelle K, Stern A. Full genome viral sequences inform patterns of SARS-CoV-2 spread into and within Israel. Nat Commun 2020; 11:5518. [PMID: 33139704 DOI: 10.1101/2020.05.21.20104521] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/02/2020] [Indexed: 05/22/2023] Open
Abstract
Full genome sequences are increasingly used to track the geographic spread and transmission dynamics of viral pathogens. Here, with a focus on Israel, we sequence 212 SARS-CoV-2 sequences and use them to perform a comprehensive analysis to trace the origins and spread of the virus. We find that travelers returning from the United States of America significantly contributed to viral spread in Israel, more than their proportion in incoming infected travelers. Using phylodynamic analysis, we estimate that the basic reproduction number of the virus was initially around 2.5, dropping by more than two-thirds following the implementation of social distancing measures. We further report high levels of transmission heterogeneity in SARS-CoV-2 spread, with between 2-10% of infected individuals resulting in 80% of secondary infections. Overall, our findings demonstrate the effectiveness of social distancing measures for reducing viral spread.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Base Sequence
- Basic Reproduction Number/statistics & numerical data
- Betacoronavirus/genetics
- COVID-19
- Child
- Child, Preschool
- Communicable Diseases, Imported/epidemiology
- Communicable Diseases, Imported/virology
- Coronavirus Infections/epidemiology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/transmission
- Female
- Genome, Viral/genetics
- Humans
- Infant
- Infant, Newborn
- Israel/epidemiology
- Male
- Middle Aged
- Pandemics/prevention & control
- Phylogeny
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/transmission
- Psychological Distance
- RNA, Viral/genetics
- SARS-CoV-2
- Sequence Analysis, RNA
- United States
- Young Adult
Collapse
Affiliation(s)
- Danielle Miller
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michael A Martin
- Department of Biology, Emory University, Atlanta, GA, USA
- Population Biology, Ecology, and Evolution Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Noam Harel
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Omer Tirosh
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Talia Kustin
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Moran Meir
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nadav Sorek
- Microbiology Laboratory, Assuta Ashdod University-Affiliated Hospital, Ashdod, Israel
| | | | - Sharon Amit
- Clinical Microbiology Laboratory, Sheba Medical Center, Ramat-Gan, Israel
| | - Olesya Vorontsov
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Avraham Shaag
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Dana Wolf
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Avi Peretz
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Clinical Microbiology Laboratory, The Baruch Padeh Medical Center, Poriya, Tiberias, Israel
| | - Yonat Shemer-Avni
- Clinical Virology Laboratory, Soroka Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Naama M Kopelman
- Department of Computer Science, Holon Institute of Technology, Holon, Israel
| | - Amit Huppert
- Bio-statistical and Bio-mathematical Unit, The Gertner Institute for Epidemiology and Health Policy Research, Chaim Sheba Medical Center, 52621, Tel Hashomer, Israel
- School of Public Health, The Sackler Faculty of Medicine, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Katia Koelle
- Department of Biology, Emory University, Atlanta, GA, USA
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta, GA, USA
| | - Adi Stern
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
17
|
Miller D, Martin MA, Harel N, Tirosh O, Kustin T, Meir M, Sorek N, Gefen-Halevi S, Amit S, Vorontsov O, Shaag A, Wolf D, Peretz A, Shemer-Avni Y, Roif-Kaminsky D, Kopelman NM, Huppert A, Koelle K, Stern A. Full genome viral sequences inform patterns of SARS-CoV-2 spread into and within Israel. Nat Commun 2020; 11:5518. [PMID: 33139704 PMCID: PMC7606475 DOI: 10.1038/s41467-020-19248-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
Full genome sequences are increasingly used to track the geographic spread and transmission dynamics of viral pathogens. Here, with a focus on Israel, we sequence 212 SARS-CoV-2 sequences and use them to perform a comprehensive analysis to trace the origins and spread of the virus. We find that travelers returning from the United States of America significantly contributed to viral spread in Israel, more than their proportion in incoming infected travelers. Using phylodynamic analysis, we estimate that the basic reproduction number of the virus was initially around 2.5, dropping by more than two-thirds following the implementation of social distancing measures. We further report high levels of transmission heterogeneity in SARS-CoV-2 spread, with between 2-10% of infected individuals resulting in 80% of secondary infections. Overall, our findings demonstrate the effectiveness of social distancing measures for reducing viral spread.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Base Sequence
- Basic Reproduction Number/statistics & numerical data
- Betacoronavirus/genetics
- COVID-19
- Child
- Child, Preschool
- Communicable Diseases, Imported/epidemiology
- Communicable Diseases, Imported/virology
- Coronavirus Infections/epidemiology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/transmission
- Female
- Genome, Viral/genetics
- Humans
- Infant
- Infant, Newborn
- Israel/epidemiology
- Male
- Middle Aged
- Pandemics/prevention & control
- Phylogeny
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/transmission
- Psychological Distance
- RNA, Viral/genetics
- SARS-CoV-2
- Sequence Analysis, RNA
- United States
- Young Adult
Collapse
Affiliation(s)
- Danielle Miller
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michael A Martin
- Department of Biology, Emory University, Atlanta, GA, USA
- Population Biology, Ecology, and Evolution Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Noam Harel
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Omer Tirosh
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Talia Kustin
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Moran Meir
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nadav Sorek
- Microbiology Laboratory, Assuta Ashdod University-Affiliated Hospital, Ashdod, Israel
| | | | - Sharon Amit
- Clinical Microbiology Laboratory, Sheba Medical Center, Ramat-Gan, Israel
| | - Olesya Vorontsov
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Avraham Shaag
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Dana Wolf
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Avi Peretz
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Clinical Microbiology Laboratory, The Baruch Padeh Medical Center, Poriya, Tiberias, Israel
| | - Yonat Shemer-Avni
- Clinical Virology Laboratory, Soroka Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Naama M Kopelman
- Department of Computer Science, Holon Institute of Technology, Holon, Israel
| | - Amit Huppert
- Bio-statistical and Bio-mathematical Unit, The Gertner Institute for Epidemiology and Health Policy Research, Chaim Sheba Medical Center, 52621, Tel Hashomer, Israel
- School of Public Health, The Sackler Faculty of Medicine, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Katia Koelle
- Department of Biology, Emory University, Atlanta, GA, USA
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta, GA, USA
| | - Adi Stern
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|