Genome-wide variations of SARS-CoV-2 infer evolution relationship and transmission route ======================================================================================== * Lehai Zhang * Shifu Wang * Qian Ren * Junjie Yang * Yanqin Lu * Lei Zhang * Zhongtao Gai ## Abstract In the epidemic evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the issues of mutation, origin, typing and the effect of mutation on molecular detection remain to be unrevealed. In order to identify the evolutionary relationship of SARS-CoV-2 and evaluate the detection efficiency of primers that are currently used in different countries, we retrieved genomic sequences of 373 SARS-CoV-2 strains from multiple databases and performed genome-wide variation analysis. According to the nucleotide C28144T variation, the SARS-CoV-2 can be divided into group A (117 strains) and group B (256 strains). The spike protein gene (S gene) coding region 1841 (total 23403) A1841G, formed a B1 subgroup (40 strains) in group B, of which 30 strains were from European and American countries in March (especially Washington, USA). These mutations are likely to be influenced by the environment or the immunization selection pressure of different populations. Although the mutation is not in the receptor binding region (RBD) and alkaline cleavage region, it may also affect the ability of transmission and pathogenicity; however, the significance is not yet clear. As the ratio of A / B strains in the epidemic months showed an increasing trend (0.35: 1 in January, 0.62: 1 in February and 0.76: 1 in March), it seems that the transmissibility of group A strains becomes stronger with time. Based on the variation of 11 nucleotide sites during the epidemic process, it is speculated that the Washington strain is more like an ancestor type, and the Wuhan strain is the offspring of the group A virus strain. By comparing the detection capabilities of primers in different countries, the SARS-CoV-2 nucleotide variation may only affect molecular detection of very few strains. The differences in the transmissibility, pathogenicity and clinical manifestations of different types of strains require further investigations. Keywords * SARS-CoV-2 * Molecular typing * Source of infection * Mutation transmission route ## Introduction The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) started to emerge in Wuhan in December 2019. As of April 20, 2020, more than 2.4 million cases have been reported in more than 211 countries and regions. While the outbreak and pandemic of SARS-CoV-2 poses a great threat to public health, in-depth studies have been conducted on key issues such as transmission ability, epidemiology, clinical characteristics, pathogenic mechanism, clinical treatment, and laboratory testing. Regarding the origin of SARS-CoV-2, bat coronavirus RaTG13 was found to be highly similar to SARS-CoV-2, so it was speculated that bats might be the original host of the virus [1]. It was further found that the virus isolated from pangolin was similar to SARS-CoV-2, suggesting that pangolin might be an intermediate host [2, 3]. By comparing the genomes of existing coronavirus such as SARS-CoV-2, MERS-CoV and SARS-CoV, it was found that RNA editing in the human body is related to certain mutations obtained by virus evolution [4]. In order to reveal more clues of the origin of SARS-CoV-2, which is still an urgent problem to be solved, we retrieved the publicly available virus sequences from global database sources such as GISAID and NCBI to analyze the emerged mutation, evolution and typing of SARS-CoV-2, and to evaluate the influence of its mutations on molecular detection in current clinical settings. ### Materials and methods By February 25, 2020, our first retrieval downloaded a total of 188 complete genomes from GISAID, NCBI, the Global Coronavirus Data Sharing and Analysis System of the National Microbiology Data Center of China ([http://nmdc.cn/coronavirus](http://nmdc.cn/coronavirus)) and the 2019 Novel Coronavirus Resource (2019nCoVR) of the China National Center for Bioinformation ([https://bigd.big.ac.cn/ncov/release_genome?lang=en](https://bigd.big.ac.cn/ncov/release_genome?lang=en)). BioEdit was used to compare the genome sequence and remove those with ambiguous loci. After continued retrieval, 373 full genomes were finally obtained by March 31, 2020, and used to construct the phylogenetic tree by MEGA-X and FigTreev1.4.3. The variation percentage and pairwise distance were calculated using MEGA-X32. BioEdit was used to calculate the number of mutation sites, and DNAsp was used to calculate the nonsynonymous mutation rate and synonymous mutation rate. ## Results ### 1. Mutation and evolution of SARS-CoV-2 The total number of nucleotides was 29,838bp after alignment of 373 full genome sequences and the deleted sequences carried no mutations. Representative strains, such as USA/WA1-F6, Chongqing/YC01, Fujian/8, MN908947.3, MN938384.1, WA-UW82, were selected from each cluster of the phylogenetic tree to analyze the mutation rate of nucleic acids, the nonsynonymous mutation rate and synonymous mutation rate, the amino acids variations of nonsynonymous mutations, and their evolutionary relationship (Table 1, 2 and 3). View this table: [Table 1](http://medrxiv.org/content/early/2020/05/03/2020.04.27.20081349/T1) Table 1 Number of nucleotide variation in the genome of the representative strains of SARS-CoV-2 View this table: [Table 2](http://medrxiv.org/content/early/2020/05/03/2020.04.27.20081349/T2) Table 2 The nucleotide variation loci in the genome of the representative strains of SARS-CoV-2 View this table: [Table 3](http://medrxiv.org/content/early/2020/05/03/2020.04.27.20081349/T3) Table 3 Genome evolution analysis of the representative strains of SARS-CoV-2 Based on the position and nucleotide variation of the phylogenetic tree, we compared the evolutionary variation between SARS-CoV-2 representative strains. WA1-F6 (sample collected on January 25, 2020) strain has one nucleotide variation compared with the closed bat strain (bat/Yunnan/RaTG13). Compared with WA1-F6 strain, we find that Chongqing/YC01, Fujian/8 and HKU-SZ-002a strain (in the same evolutionary branch with WA1-F6 strain) showed 2, 1 and 2 nucleotide variation respectively; While Wuhan epidemic strain Wuhan-Hu-1 (sample collected in November 2019), which is far from the outer group, has 3 nucleotide variation; and WA-UW82, which is far from the outer group, has 8 nucleotide variation (sample collected in March 2020). Interestingly, the WA-UW82 strain appeared one-and-a-half month after the WA1-F6 strain became popular in the Washington area of the United States. The WA-UW82 strain mutated and evolved from WA1-F6 in the 5 ‘UTR, ORF1ab, S, ORF3a and ORF8 coding regions, whereas no positive selection was found (Table 3). ### 2. Non-synonymous mutation and phylogenetic clustering Compared with bat/Yunnan/RaTG13, Wuhan-Hu-1 has a non-synonymous mutation (S84L) at position 251 (28,144 in total) in the coding region of ORF8, which is speculated to be closely related to its evolutionary position. The nucleotides and amino acids in the ORF8 region of the sequence were analyzed, and the positions of different mutated strains in the evolutionary tree were compared in Figure 1. It was found that nucleotides T251C (28144 in total) or 197 strains of amino acid L84S formed two clusters in the phylogenetic tree. To test this assumption, all 373 SARS-CoV-2 genome sequences were integrated into the phylogenetic tree in batches, and the results remained consistent. According to the nucleotide C28144T variation, SARS-CoV-2 can be clustered into two large groups. The group A virus has a C at position 28,144, whereas group B virus has a T. Among the 39 strains of the group B virus, the A1841G non-synonymous mutation (D614G) occurred in the 1841 position (23403 positions) of the S gene coding region during the SARS-CoV-2 pandemic, forming a B1 subgroup (Supplementary Figure 1 and Table 1). ![Figure 1](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/05/03/2020.04.27.20081349/F1.medium.gif) [Figure 1](http://medrxiv.org/content/early/2020/05/03/2020.04.27.20081349/F1) Figure 1 The geographical distribution of mutant strain in the S gene coding region D614G Compared with the other 5 SARS-CoV-2 representative strains, the non-synonymous mutation of A1841G (D614G) occurred in the S gene coding region 1841 (total 23403), but no mutation occurred in the receptor binding region (RBD, S protein amino acid 451–509) and alkaline cleavage region (667–694) in WA-UW82. Further analysis of the S-coding regions of 373 strains of SARS-CoV-2 genome collected from December 2019 to March 31, 2020 revealed that 39 strains had mutations of D614G, and 334 strains had no mutations. Of the 39 strains, only BetaCoV/Germany/BavPat1/2020 was mutated at the end of January, 10 strains were mutated in February, and the remaining 28 strains were mutated in March. Except for the Wuhan/HBCDC-HB-06/2020 strain from Asia, the vast majority of mutations were from European and American countries (Figure1). ### 3. The influence of the variation to the nucleic acid detection In order to evaluate the impact of nucleotide variation on clinical test, we compared the 373 SARS-CoV-2 genome sequences with currently used primers that were reported by the Chinese Center for Disease Control and Prevention (CDC) [5], Europe and Hong Kong [6] (Table 4). View this table: [Table 4](http://medrxiv.org/content/early/2020/05/03/2020.04.27.20081349/T4) Table 4 Primers for nucleic acid detection of SARS-CoV-2 in China CDC and Europe The primer sequences reported by China CDC and Europe did not find nucleotide variation in the ORF1ab and RdRp amplification regions, respectively, while several nucleotides in the N amplification region mutated, including 10 strains from Mexico, Netherlands, Sweden, Finland, Portugal and Peru. The nucleotides corresponding to the 5’ initiation segment of the forward primer of N region have all been mutated from GGG to AAC (Table 4), but this variation may not lead to false-negative results in PCR detection. Whereas the nucleotide mutations have occurred in the N or E amplification region near the 3 ‘end of the European primers, that may affect the amplification results. Although European primers use degenerate primers at the RdRp amplification region, fortunately there is no mutation observed yet. The difference between German and European primers is that the former amplifies RdRP, E, and N region, and the latter amplifies only RdRP and E region. The RdRP reverse primer “S” is designed according to the base C of SARS-CoV-2 and the base G of the bat SARS-related coronavirus, while no mutation of base T has been found at this location in SARS-CoV-2. Therefore, the use of Chinese CDC and European primer sequence in PCR detection will not be affected by this mutation, which can ensure the accuracy and quality of clinical PCR screening of COVID - 19. ## Discussion Since the outbreak of COVID-19 in Wuhan in December 2019, it has infected more than 1.7 million people in more than 200 countries and regions worldwide, resulting in 160,000 deaths (as of 2020–4–20 9:30).The origin of SARS-CoV-2 has been described by international authorities [7, 8], and four distinct characteristics of the virus have been found. There have also been in-depth studies on the evolution of variation [9, 10], which however only studied the variation and evolution of SARS-CoV-2 strain in the early stage of the pandemic. In this study, 373 SARS-CoV-2 genome sequences were downloaded from GISAID, NCBI and the Global Coronavirus Data Sharing and Analysis System of the National Microbiology Data Center of China from the end of 2019 to March 31, 2020. More than 188 virus sequences from different epidemic times and countries and regions were selected to construct the phylogenetic tree. Several distinct clusters were shown in the phylogenetic tree, and the evolutionary relationships of each cluster were analyzed. It was found that the strains of bat/Yunnan/RaTG13 clustered at the lower part of the phylogenetic tree had non-synonymous mutations (T251C, S84L) at 251 (28,144 in total), and those without mutations were in other clusters. More importantly, there were such mutations in the further analysis of 373 strains [9]. Accordingly, popular strain can be divided into group A (117 strains) and group B (256 strains) clusters. The A/B ratio showed an interesting trend of increase of strains (January 0.35:1, February 0.62:1 and March 0.76:1), indicating that the transmissibility of class A strain got stronger over time. However, the differences in the transmission ability, pathogenic ability and clinical performance of the two types of strains have not been reported, and it is unknown whether there is a difference in the mortality caused by them. It is worthy of further follow-up studies by molecular epidemiology experts and clinical experts. Singapore [11] reported that the regionally popular strains spanned ORF7b and ORF8 mutations, including ORF8 transcriptional regulatory sequence (TRS), which eliminated ORF8 transcription, thereby enhancing the transcription efficiency of downstream N-terminal genes. The ORF8 region is considered to be an evolutionary hotspot of SARS related coronaviruses (SARSr-CoVs) and an important region for the evolution of SARSr-CoVs. The ORF8 region has been shown to play an important role in the virus replication efficiency of SARS-CoV, interspecies transmission, and adaptation to human hosts. It is speculated that it may be the result of the adaptation of SARS-CoV-2 to the human host, and it has undergone reorganization selection during the transmission from animal to human [12]. In the SARS-CoV-2 pandemic, 39 strains of the same group B (group B1) had A1841G non-synonymous mutation (D614G) in the 1,841 position (23,403 positions in total) in the S gene coding region. Of these, 30 strains were isolated from European and American countries in March (especially Washington, USA), whereas in comparison, only 1 strain was isolated in January and 10 strains were isolated in February. At the end of the writing this article, we downloaded 62 new sequences that were deposited in April into the GISAID database. After deleting the low-quality sequences, 45 were retained for analysis (data not shown). According to the above typing method (C28144T, A23403G), except for hCoV-19/England/20144018704 and hCoV-19/ Belgium/ULG-10095/2020 (Group B), the other 43 strains are all divided into B1 subgroup. With the exception of 3 strains from South Africa and 1 from Georgia, the remaining 39 strains were all from Europe and the United States. This may be mutated by the selection pressure of the environment or the immunity of different population. Although the mutation is not in the RBD and alkaline lysis zone, it may also affect the transmission and pathogenic ability, but its significance is not yet clear. These results suggest that the virus may have undergone new mutations under the pressure of environmental selection, forming a new epidemic strain. The difference in the ability of the mutation to spread, pathogenicity, clinical manifestations and outcome of the strain needs to be determined by molecular epidemiologists and clinicians in further follow-up studies. Of the first set of 41 infected patients in Wuhan, 27 were associated with the South China Seafood Market of Wuhan city, and the infected subjects spread them to others outside the market, however, this conclusion has been controversial [13]. In this study, the strains that have a close phylogenetic relationship with the outer group bat/Yunnan/RaTG13 are all group A strains. There is no obvious difference in the time of initial epidemic between group A and B strains. The original virus strains belonging to group A, such as Chongqing (Chongqing/YC01), Fujian (Fujian/8), Shenzhen (HKU-SZ-002a), and Washington (USA/WA1-F6), etc., are all phylogenetically far away from the group B strain that were popular in Wuhan. Four family members from Shenzhen (Wuhan-Hu-1) visited Wuhan from December 29, 2019 to January 4, 2020 [14], and the first confirmed patient in the United States (USA / WA1-F6) visited Wuhan on January 15, 2020 and returned to Washington State [15], and the Chongqing (Chongqing/YC01) patients returned to their hometowns after working in Wuhan. Thus, it is speculated that there are at least two or more original infection sources, or that two types of strains are spreading simultaneously in a certain infection source. By analyzing the 11 nucleotide sites that can be mutated in the epidemic process, we found that the first patient strain (WA1) in Washington, USA, was mutated by 1 nucleotide based on the bat strain bat/Yunnan/RaTG13, the Chongqing strain Chongqing/YC01 and Shenzhen strain HKU-SZ-002a mutated 2 nucleotides at different positions respectively, while the American WA-UW82 mutated 9 nucleotides. It can be considered that the Washington strain of the United States is more like an ancestor type, although its onset time is later than Wuhan patients. The epidemic strain in Wuhan should be a variant of group A strains. We thus speculated that the group A strains were already prevalent before the outbreak in Wuhan at the end of December 2019. Through analysis of 10 strains from Mexico, the Netherlands, Sweden, Finland, Portugal, and Peru, it was found that the nucleotides corresponding to the 5 ‘initiation segment of the N region forward primer were all mutated from GGG to AAC. This mutation may cause false negative results in PCR amplification detection, leading to the missed diagnosis and the spread of the virus. However, the primer sequences provided by the Chinese CDC and Europe did not find nucleotide variations in the amplified regions of ORF1ab and RdRp. Therefore, the quality of screening COVID-19 by clinical PCR could be guaranteed by using the detection kits that include Chinese CDC and European primer. In conclusion, the global spread of SARS-CoV-2 has not been effectively controlled at present,. Group B should be the mutated strain of group A, and the group A strain is easier to spread. We should pay close attention to the variation and significance in the epidemic process, especially the pathogenicity, transmissibility, clinical process and prognosis of different groups of strains are not clear. Its origin is still a major question to be explored. ## Data Availability We downloaded a total of 188 complete genomes from GISAID, NCBI, the Global Coronavirus Data Sharing and Analysis System of the National Microbiology Data Center of China ([http://nmdc.cn/coronavirus](http://nmdc.cn/coronavirus)) and the 2019 Novel Coronavirus Resource (2019nCoVR) of the China National Center for Bioinformation ([https://bigd.big.ac.cn/ncov/release_genome?lang=en](https://bigd.big.ac.cn/ncov/release_genome?lang=en)) ## Declaration of interest statement All authors declare that there is no conflict of interest. This research plan was approved by the Medical Ethics Committee of the Qilu Children’s Hospital of Shandong University. ## Acknowledgments This study was supported by Shandong Province Science Foundation for Talent Cultivation [ZR2015PH059], the outstanding young technical backbone training project of Jinan [Shi-fu Wang], the State Major Infectious Disease Research Program [2012ZX10004206–007 and 2017zx10103004–007]. Lei Zhang is supported by the TaiShan Scholars Program of Shandong Province (No. tshw20120206) and TaiShan Industrial Experts Program (No. tscy20190612).ZT.G. and L.Z. design the research; LH.Z. and SF.W. performed research; LH.Z. and J.J.Y. analyzed data; LH.Z. Q.R. and Y.Q.L.performed statistical analyses; LH.Z., SF.W.and L.Z. wrote the manuscript. ## Appendices **Table1**. The number of nucleotide variation in the genome of SARS-CoV-2 represents strain **Table2**. The nucleotide variation sites in the genome of SARS-CoV-2 represent strains **Table3**. The genome evolution analysis of the SARS-CoV-2 represents strain **Table4**. The primers for nucleic acid detection of SARS-CoV-2 in China CDC and Europe **Supplementary Table1**. Group A evolved into group B, subgroup B1, and amino acid changes of protein S **Figure1**. The geographical distribution of mutant strain in the S gene coding region D614G **Supplementary Figure 1**. the comparison and analysis of subgroups of SARS-CoV-2 A/B group * Received April 27, 2020. * Revision received April 27, 2020. * Accepted May 3, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## References 1. [1].Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin[J/OL].Nature,[2020-03-23].[https://www.nature.com/articles/s41586-020-2012-7](https://www.nature.com/articles/s41586-020-2012-7). D0I:doi:10.1038/s41586-020-2012-7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-020-2012-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32015507&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F03%2F2020.04.27.20081349.atom) 2. [2]. Tommy Tsan-Yuk Lam, Marcus Ho-Hin Shum, Hua-Chen Zhu, Identification of 2019-nCoV related coronaviruses in Malayan pangolins in southern China. [J/OL]. BioRxiv, [2020-03-23]. [https://www.biorxiv.org/content/10.1101/2020.02.13.945485v1](https://www.biorxiv.org/content/10.1101/2020.02.13.945485v1). DOI: 10.1101/2020.02.13.945485. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4wMi4xMy45NDU0ODV2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA1LzAzLzIwMjAuMDQuMjcuMjAwODEzNDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 3. [3]. Kangpeng Xiao, Junqiong Zhai, Yaoyu Feng, et al. Isolation and Characterization of 2019-nCoV-like Coronavirus from Malayan Pangolins. [J/OL]..BioRxiv, [2020-03-23]. [https://www.biorxiv.org/content/10.1101/2020.02.17.951335v1](https://www.biorxiv.org/content/10.1101/2020.02.17.951335v1). DOI: 10.1101/2020.02.17.951335 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4wMi4xNy45NTEzMzV2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA1LzAzLzIwMjAuMDQuMjcuMjAwODEzNDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 4. [4]. Salvatore Di Giorgio, Filippo Martignano, Maria Gabriella Torcia, et al. Evidence for RNA editing in the transcriptome of 2019 Novel Coronavirus. [J/OL]. BioRxiv, [2020-03-23]. [https://www.biorxiv.org/content/10.1101/2020.03.02.973255v1](https://www.biorxiv.org/content/10.1101/2020.03.02.973255v1). DOI: 10.1101/2020.03.02.973255 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4wMy4wMi45NzMyNTV2MiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA1LzAzLzIwMjAuMDQuMjcuMjAwODEzNDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 5. [5].[http://ivdc.chinacdc.cn/kyjz/202001/t20200121\_211337.html](http://ivdc.chinacdc.cn/kyjz/202001/t20200121_211337.html) 6. [6].Corman V M, Landt O, Kaiser M, et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. [J/OL].eurosurveillance,[2020-03-26].[https://doi.org/10.2807/1560-7917.ES.2020.25.3.2000045](https://doi.org/10.2807/1560-7917.ES.2020.25.3.2000045).DOI: 10.2807/1560-7917.ES.2020.25.3.2000045. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2807/1560-7917.ES.2020.25.3.2000045&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31992387&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F03%2F2020.04.27.20081349.atom) 7. [7].Andersenl K G, Rambaut A, Lipkin W I, et al. The proximal origin of SARS-CoV-2. [J/OL]. Nature,[2020-04-8]. [https://doi.org/10.1038/s41591-020-0820-9](https://doi.org/10.1038/s41591-020-0820-9).DOI: 10.1038/s41591-020-0820-9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-0820-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32284615&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F05%2F03%2F2020.04.27.20081349.atom) 8. [8].Bonil MF, Lemey P, Xiaowei Jiang XW et al.Evolutionary origins of the SARS CoV 2 sarbecovirus lineage responsible for the COVID-19 pandemic. [J/OL]. bioRxiv,[2020-04-8]. [https://www.biorxiv.org/content/10.1101/2020.03.30.015008v1](https://www.biorxiv.org/content/10.1101/2020.03.30.015008v1). doi: 10.1101/2020.03.30.015008 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4wMy4zMC4wMTUwMDh2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA1LzAzLzIwMjAuMDQuMjcuMjAwODEzNDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 9. [9].Tang XL, Wu CC, Li X. et al.On the origin and continuing evolution of SARS-CoV-2. [J/OL].National Science Review. [2020-04-8]. [https://doi.org/10.1093/nsr/nwaa036](https://doi.org/10.1093/nsr/nwaa036). doi: 10.1093/nsr/nwaa036 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nsr/nwaa036&link_type=DOI) 10. [10].Yu WB, Tang GD, Zhang L, et al. Decoding the evolution and transmissions of the novel pneumonia coronavirus(SARS-CoV-2) using whole genomic data.[J/OL].chinaXiv.[2020-04-8]. [http://eprint.las.ac.cn/abs/202002.00033](http://eprint.las.ac.cn/abs/202002.00033). doi: 10.12074/202002.00033. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.12074/202002.00033&link_type=DOI) 11. [11].Su Y C F, Anderson D E, Young B E, et al., Discovery of a 382-nt deletion during the early evolution of SARS-CoV-2. [J/OL]. bioRxiv, [2020-04-8].[https://www.biorxiv.org/content/10.1101/2020.03.11.987222v1](https://www.biorxiv.org/content/10.1101/2020.03.11.987222v1). doi: 10.1101/2020.03.11.987222 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4wMy4xMS45ODcyMjJ2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA1LzAzLzIwMjAuMDQuMjcuMjAwODEzNDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 12. [12].Lau, S.K., et al., Severe Acute Respiratory Syndrome (SARS) Coronavirus ORF8 Protein Is Acquired from SARS-Related Coronavirus from Greater Horseshoe Bats through Recombination. [J/OL]. J Virol, 2015. 89(20): p. 10532–47 13. [13].Cohen J. Wuhan seafood market may not be source of novel virus spreading globally. Science 2020: 10.1126/science.abb0 611. 14. [14].Chan JF-W, Yuan S, Kok K-H, et al. A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: a study of a family cluster. Lancet 2020: 10.1016/S0140–6736(20)30154–9. 15. [15].Holshue ML, DeBolt C, Lindquist S, et al. First Case of 2019 Novel Coronavirus in the United States. N Engl J Med 2020: 10.1056/NEJMoa2001191.