分享

Inter- and intra-lineage genetic diversity of wild-type Zika viruses reveals both common and distinc

 医学abeycd 2019-08-26

ABSTRACT

Zika virus (ZIKV) strains belong to the East African, West African, and Asian/American phylogenetic lineages. RNA viruses, like ZIKV, exist as populations of genetically-related sequences whose heterogeneity may impact viral fitness, evolution, and virulence. Genetic diversity of representative ZIKVs from each lineage was examined using next generation sequencing (NGS) paired with downstream entropy and single nucleotide variant (SNV) analysis. Comparisons showed that inter-lineage diversity was statistically supported, while intra-lineage diversity varied. Intra-lineage diversity was significant for East but not West Africa strains. Furthermore, intra-lineage diversity for the Asian/American lineage was not supported for human serum isolates; however, a placenta isolate differed significantly. Relative entropy values were higher in the pre-membrane/membrane (prM/M) gene of several ZIKV strains. Additionally, the East African lineage contained a greater number of synonymous SNVs, while a greater number of non-synonymous SNVs were identified for American strains. Further, inter-lineage SNVs were dispersed throughout the genome, whereas intra-lineage non-synonymous SNVs for Asian/American strains clustered within prM/M and NS1 gene. This comprehensive analysis of ZIKV genetic diversity provides a repository of SNV positions across lineages. We posit that increased non-synonymous SNV populations and increased relative genetic diversity of the prM/M and NS1 proteins provides more evidence for their role in ZIKV virulence and fitness.

KEYWORDS: Next generation sequencingZika virusgenetic diversityZika lineageShannon entropy

Introduction

Zika virus (ZIKV) is a mosquito-borne flavivirus that was originally discovered in Uganda in 1947 after a sentinel Rhesus monkey developed fever and viremia [1Dick GWKitchen SHaddow A. Zika virus (I). Isolations and serological specificity. Trans R Soc Trop Med Hyg. 1952;46:509520. doi: 10.1016/0035-9203(52)90042-4[Crossref][Web of Science ®] [Google Scholar]]. Historically, human ZIKV infections occurred infrequently in Africa and Asia, and symptoms were limited to mild febrile disease [2Musso DGubler DJ. Zika virus. Clin Microbiol Rev. 2016;29:487524. doi: 10.1128/CMR.00072-15[Crossref][Web of Science ®] [Google Scholar]]. In 2007, ZIKV infections on the island of Yap led to clinical signs including rash, fever, conjunctivitis, and arthralgia [3Duffy MChen THancock W, et al. Zika virus outbreak on Yap island, Federated States of Micronesia. N Engl J Med. 2009;360:25362543. doi: 10.1056/NEJMoa0805715[Crossref][Web of Science ®] [Google Scholar]]. After initial spread out of Asia, ZIKV continued to cause clinical disease, including a 2013 outbreak in French Polynesia associated with Guillain-Barré syndrome [4Weaver SCCosta FGarcia-Blanco MA, et al. Zika virus: history, emergence, biology, and prospects for control. Antiviral Res. 2016;130:6980. doi: 10.1016/j.antiviral.2016.03.010[Crossref][Web of Science ®] [Google Scholar],5Cao-Lormeau V-MBlake AMons S, et al. Guillain-Barré syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study. Lancet. 2016;387:15311539. doi: 10.1016/S0140-6736(16)00562-6[Crossref][Web of Science ®] [Google Scholar]]. Beginning in 2015, a ZIKV epidemic spread throughout the Americas, leading to foetal neurodevelopmental deficiencies, including microcephaly, termed Congenital Zika Syndrome [6Costello ADua TDuran P, et al. Defining the syndrome associated with congenital Zika virus infection. Bull World Health Organ. 2016;94:406406A. doi: 10.2471/BLT.16.176990[Crossref][Web of Science ®] [Google Scholar]].

ZIKV strains are grouped into three major phylogenetic lineages: East African, West African, and Asian/American [4Weaver SCCosta FGarcia-Blanco MA, et al. Zika virus: history, emergence, biology, and prospects for control. Antiviral Res. 2016;130:6980. doi: 10.1016/j.antiviral.2016.03.010[Crossref][Web of Science ®] [Google Scholar],7–11Faye OFreire CCMIamarino A, et al. Molecular evolution of Zika virus during its emergence in the 20th Century. PLoS Negl Trop Dis. 2014;8:e2636. doi: 10.1371/journal.pntd.0002636
Lanciotti RSLambert AJHolodniy M, et al. Phylogeny of Zika virus in Western Hemisphere, 2015. Emerg Infect Dis J. 2016;22:933935. doi: 10.3201/eid2205.160065
Ye QLiu Z-YHan J-F, et al. Genomic characterization and phylogenetic analysis of Zika virus circulating in the Americas. Infect Genet Evol. 2016;43:4349. doi: 10.1016/j.meegid.2016.05.004
Kochakarn TKotanan NKümpornsin K, et al. Comparative genome analysis between Southeast Asian and South American Zika viruses. Asian Pac J Trop Med. 2016;9:10481054. doi: 10.1016/j.apjtm.2016.10.002
Pettersson JHEldholm VSeligman SJ, et al. How did Zika virus emerge in the Pacific islands and Latin America. MBio. 2016;7:e012396. doi: 10.1128/mBio.01239-16 
]. Strains from the recent epidemic in the Americas form a clade within the Asian lineage.

ZIKV has a typical flavivirus genome organization of a positive-sense, single-stranded RNA of approximately 10,800 nucleotides in length. The genome encodes for a single polyprotein that is co- and post-translationally processed to generate ten viral proteins [12Kuno GChang GJTsuchiya KR, et al. Phylogeny of the genus flavivirus. J Virol. 1998;72:7383.[Web of Science ®] [Google Scholar]]. The three structural proteins: capsid (C), pre-membrane/membrane (prM/M), and envelope (E) have roles in viral maturation, entry and fusion with the host cell, and humoral immunity [13Heinz FXAllison SL. Flavivirus structure and membrane fusion. Adv Virus Res. 2003;59:6397. doi: 10.1016/S0065-3527(03)59003-0[Crossref][Web of Science ®] [Google Scholar],14Lindenbach BDRice C. Molecular biology of flaviviruses. Adv Virus Res. 2003;59:2361. doi: 10.1016/S0065-3527(03)59002-9[Crossref][Web of Science ®] [Google Scholar]]. The seven nonstructural proteins: NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5 encode for proteins involved in innate immunity, the replication complex, or viral enzymes, such as a helicase, methyl-transferase, or RNA-dependent RNA-polymerase (RdRp) [15Chambers TJHahn CSGaller R, et al. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol. 1990;44:649688. doi: 10.1146/annurev.mi.44.100190.003245[Crossref][Web of Science ®] [Google Scholar]]. Studies with flaviviruses of medical importance, such as dengue, West Nile, and Japanese encephalitis, have shown that molecular determinants of virulence are present throughout the genome [16–19Bray MMen RTokimatsu I, et al. Genetic determinants responsible for acquisition of dengue type 2 virus mouse neurovirulence. J Virol. 1998;72:16471651.
de Borba LStrottmann DMde Noronha L, et al. Synergistic interactions between the NS3 hel and E proteins contribute to the virulence of dengue virus type 1. PLoS Negl Trop Dis. 2012;6:e1624.
Davis CTGalbraith SEZhang S, et al. A combination of Naturally Occurring mutations in North American West Nile virus nonstructural protein genes and in the 3’ Untranslated region Alters virus phenotype. J Virol. 2007;81:61116116. doi: 10.1128/JVI.02387-06
Yu Y. Phenotypic and genotypic characteristics of Japanese encephalitis attenuated live vaccine virus SA14-14-2 and their stabilities. Vaccine. 2010;28:36353641. doi: 10.1016/j.vaccine.2010.02.105 
].

RNA virus genomes exist as viral populations composed of genetically-related sequences. This is due, in part, to the error rate of the RdRp, which leads to nucleotide misincorporation during viral replication. As mutations accumulate, viral genomes can become more heterogeneous. Next generation sequencing (NGS) provides a platform to gain deeper understanding of viral diversity by increasing the depth of sequencing coverage, thereby allowing detection of single nucleotide variants (SNV) in the population. Evaluating genetic diversity has expanded the understanding of virulence, evolution, and host-specific adaptations. Flavivirus genetic diversity has been examined for the prototypical member, yellow fever virus (YFV). Comparisons between wild-type (WT) and live-attenuated YFV strains show that higher entropy is associated with a virulent, WT phenotype [20Beck ATesh RBWood TG, et al. Comparison of the live attenuated yellow fever vaccine 17D-204 strain to its virulent parental strain Asibi by deep sequencing. J Infect Dis. 2014;209:334344. doi: 10.1093/infdis/jit546[Crossref][Web of Science ®] [Google Scholar],21Collins NDBeck ASWiden SG, et al. Structural and nonstructural genes contribute to the genetic diversity of RNA Viruses. MBio. 2018;9:113. doi: 10.1128/mBio.01871-18[Crossref][Web of Science ®] [Google Scholar]].

In the present study, NGS has been used to gain insight into the genetic diversity of representative ZIKV strains from different lineages. Intra-lineage diversity for Asian/American strains was not supported for human serum isolates; however, the diversity of a placenta isolate was significantly different. Furthermore, relative genomic diversity was highest in the prM/M and NS1 gene regions of Asian/American strains isolated from human serum. This study provides insight into the intra-host diversity of contemporary ZIKV that may contribute to mammalian infection or virulence.

Materials and methods

Viruses

ZIKV strains acquired from the World Reference Center for Emerging Viruses and Arboviruses (WRCEVA) (www./wrceva) were chosen as representatives of the different genetic lineages from East Africa: MR766 and MR766ΔE153-156, from West Africa: DakAr41667 and DakAr41524, from Asia: FSS13025 and from the Americas PA259249 and R103451. Stocks were generated by passaging in monkey kidney Vero cells in minimum essential media (MEM) supplemented with L-glutamine, non-essential amino acids, penicillin–streptomycin, and 2% fetal bovine serum. Isolation and passage histories are described in Table 1 and selection details are provided in Supplementary Materials and Methods.

Table 1. Description of ZIKV strains used in study.


Phylogenetic analysis

Complete coding sequences of ZIKVs available from the WRCEVA were analysed using maximum-likelihood phylogeny. Nucleotide sequences were aligned with MUSCLE [22Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 2004;32:17921797. doi: 10.1093/nar/gkh340[Crossref][Web of Science ®] [Google Scholar]], and phylogeny was constructed using the RaXML-HPC Backbox tool [23Stamatakis A. RAxML version 8: A tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics. 2014;30:13121313. doi: 10.1093/bioinformatics/btu033[Crossref][Web of Science ®] [Google Scholar]] in the CIPRES Scientific Gateway using default parameters [24Miller MAPfeiffer WSchwartz T. Creating the CIPRES Science Gateway for inference of large phylogenetic trees. 2010 Gateway Computing Environments Workshop; 2010 Nov 14; New Orleans, LA; 2010. [Google Scholar]].

Next generation sequence analysis

Viral RNA was sequenced at the UTMB Next Generation Sequencing (NGS) Core Facility using the Illumina platform. The analysis pipeline used in this study is described in Supplementary Materials and Methods and in Figure S1 according to the guidelines for computational reproducibility [25Piccolo SRFrampton MB. Tools and techniques for computational reproducibility. Gigascience. 2016;5:113. doi: 10.1186/s13742-016-0135-4[Crossref][Web of Science ®] [Google Scholar]]. De novo consensus sequences were compared to reference sequences deposited in Genbank, with the exception of DakAr41667, for which there was no available sequence until this study (accession MF510857). Sequencing data are available in the ArrayExpress repository (accession E-MTAB-7771).

Determination of diversity indices

Diversity indices were determined from sequence alignment data following removal of PCR duplicates. V-phaser2 v2.0, a software package for inferring low frequency variants, was used to determine variant population. All SNVs controlled for false discovery and strand-bias were assessed. The results in the form of a list of SNV positions, frequencies, and identities for each ZIKV strain were exported to Excel to facilitate data sorting and table organization. The amino acid identity encoded by each SNV was determined using the gene lengths in Table S3 and compared among the strains. Shannon entropy index was calculated to assess ZIKV genomic diversity. The variability (or uncertainty) at each nucleotide position from 107 to 10,700 was determined as previously described [26Fishman SLBranch AD. The quasispecies nature and biological implications of the hepatitis C virus. Infect Genet Evol. 2009;9:11581167. doi: 10.1016/j.meegid.2009.07.011[Crossref][Web of Science ®] [Google Scholar],27Nasu AMarusawa HUeda Y, et al. Genetic heterogeneity of hepatitis C virus in association with antiviral therapy determined by ultra-deep sequencing. PLoS One. 2011;6:e24907. doi: 10.1371/journal.pone.0024907[Crossref][Web of Science ®] [Google Scholar]]. Briefly, deepSNV v1.16.0 bam2R with a quality score of 30 was used to determine nucleotide counts for genomic positions and then converted to relative nucleotide frequencies using RF. Shannon entropy was calculated utilizing local R script v3.2.4 (Figure S1) and relative nucleotide frequencies, according to previously reported analyses [28Nishijima NMarusawa HUeda Y, et al. Dynamics of hepatitis B virus quasispecies in association with nucleos(t)ide analogue treatment determined by ultra-deep sequencing. PLoS One. 2012;7:e35052. doi: 10.1371/journal.pone.0035052[Crossref][Web of Science ®] [Google Scholar]], with a maximum positional value of 1.61 indicating equal representation of all nucleotides (A, U, G, C, and gap). Data were evaluated for each strain (n = 2 each) or for each lineage (East Africa: n = 4, West Africa: n = 4, Asia/ America: n = 6). The inter- and intra-lineage entropy statistics were determined by combining the replicates for each strain within a lineage and non-parametric analysis.

Structural renderings

Protein structures were rendered using Pymol v1.8.4.0: Capsid: PDB 5YGH, Membrane-Envelope: PDB 6CO8, NS1: PDB 5K6 K, NS3: PDB 5GJB, NS5: PDB 5UOB.

Statistical analysis

Statistical tests (Mann–Whitney, Kruskal–Wallis and post-tests), correlations, (Spearman and Pearson) and calculations (mean, median, error) were determined using Prism v7.0 (GraphPad). Plots were generated using Prism v7.0 or 8.0.

Results

NGS generated a rich dataset to study genetic diversity

Maximum likelihood phylogeny was constructed using ZIKV strains available from the WRCEVA (Figure 1). Tree topology was consistent with three lineages consisting of strains from East Africa, West Africa, and Asia and the Americas. Representative strains from each lineage were selected for genetic analysis (Table 1). The two East African strains originated from monkey serum in 1947 in Uganda [1Dick GWKitchen SHaddow A. Zika virus (I). Isolations and serological specificity. Trans R Soc Trop Med Hyg. 1952;46:509520. doi: 10.1016/0035-9203(52)90042-4[Crossref][Web of Science ®] [Google Scholar]]. Strain MR766 and strain MR766 stock 30306 (subsequently referred to as MR766ΔE153-156) differ in consensus sequences by four amino acids in the E protein (153-154 deletion), due to lengthy passage histories. Two West African strains, DakAr41524 and DakAr41667, were isolated in Senegal in 1984 from Aedes africanus and Ae. taylori mosquitoes, respectively. Next, three Asian/American lineage strains were selected for the study. Strain FSS13025 was isolated in Cambodia in 2010 from the acute phase serum of a child [29Heang VYasuda CYSovann L, et al. Zika virus infection, Cambodia, 2010. Emerg Infect Dis. 2012;18:349351. doi: 10.3201/eid1802.111224[Crossref][Web of Science ®] [Google Scholar]]. Strain PA259249 was isolated from human serum in Panama in December 2015. Lastly, a human placenta isolate, strain R103451, was isolated in Honduras in January 2016. Strains PA259249 and R103451, have an identical low number of passages in Vero cells.

Figure 1. Phylogenetic analysis. Maximum-likelihood phylogeny of ZIKV strains from the WRCEVA supported the groupings into the East Africa, West Africa, and Asian lineages, the latter containing strains from the recent epidemic. Arrows point to the ZIKVs chosen for genetic diversity analysis.

Display full size

Two biological replicates of the seven ZIKVs were subjected to NGS and downstream analysis. Mapped reads ranged from 415,219–2,526,545. After processing the mean map quality of the strains was approximately 44 (Table S1), and the mean depth of coverage across the genome ranged from 1,906–10,762, with consistent positional coverage for both replicates of each strain (Figure 2). Mean genome coverage and mapped reads were highly correlated (r = 0.9961) (Figure S2). De novo consensus sequences for strains MR766, MR766ΔE153-156, DakAr41524, FSS13025, PA259249, and R103451 were aligned to the corresponding publicly available sequences using MUSCLE, which showed that some de novo sequences were missing up to 10 nucleotides from the 5’ and/or 3’ ends, so the sequences were adjusted with N placeholders accordingly. Additionally, FSS13025 and R103451 each contained the amino acid substitutions K1059E and K733R, respectively, compared to published sequences (Table S2). Overall, the NGS results provided a rich dataset to perform downstream genetic analysis.

Figure 2. ZIKV NGS sequencing dataset description. Duplicate samples (N = 14) of each strain were analysed. The positional read coverage depth of each replicate (after filtering out low-quality reads and PCR replicates) was plotted along the genome and was similar for both replicates of each strain. Coverage range: 1,906-10,762.

Inter- and intra-lineage comparisons reveal differences among Asian/American lineage ZIKVs

To determine the nucleotide diversity of ZIKVs, Shannon entropy was computed. This calculates the nucleotides at a given position, and higher values indicate that all nucleotides are equally represented at that position. To account for bias introduced from lower depths of coverage at the 5’ and 3’ ends of the genome, Shannon entropy was calculated for every position starting at nucleotide 107 (the A of the first codon in African lineage strains) through nucleotide 10,700, approximately 100 nucleotides from the 3’ terminus of the ZIKV genome and the mean of the two replicates was determined (Figure S3).

Inter-lineage nucleotide diversity was assessed by grouping the strains phylogenetically prior to performing statistical analysis (Figure 3a). Results show that East African, West African, and Asian/American lineages differ in genomic nucleotide diversity (Kruskal–Wallis, Dunn’s post-test, p < 0.0001), with mean entropy values of 2.88 × 10−3, 3.24 × 10−3, and 3.52 × 10−3, respectively, indicating increased nucleotide diversity for the Asian/American lineage.

Figure 3. Shannon entropy analysis shows inter- and intra-lineage genetic diversity relationships. The mean Shannon entropy calculated from positions 107-10,700 from two biological replicates of each strain for the East Africa (n = 4), West Africa (n = 4), and Asia/America (n = 6) lineages (A) or by strain (each, n = 2) (B) is shown as violin plots; solid lines: median, dashed lines: interquartile range, statistics determined using Kruskal-Wallis with Dunn’s post-test. C) Mean entropy values for each strain (n = 2) with standard error of the mean (SEM) and standard deviation (SD). The number of positions with entropy values greater than two times the SD are shown; arrows denote that strains PA259249 and R103451 contain more high entropy positions. D) Statistical analysis (Kruskal-Wallis with Dunn’s post-test) of mean genomic Shannon entropy comparing (n = 2) of each strain. E). Relative diversity heat map shows the mean Shannon entropy of each gene region relative to each gene length.

To assess within-lineage diversity, entropy was calculated for each strain. The intra-lineage diversity is statistically significant (Kruskal–Wallis, Dunn’s post-test) for strains of the East African lineage (p < 0.0001) and for strains of the Asian/American lineage (p < 0.0001) but not for strains of the West African lineage (Figure 3b-d). Interestingly the diversity of human placenta strain R103451 was significantly different (p < 0.0001) to that of both strains FSS13025 and PA259249, but diversity of the latter two was not significantly different to each other.

The mean nucleotide diversity for the ZIKVs ranged from 2.56 × 10−3–3.60 × 10−3 (Figure 3c). MR766 has higher mean genomic entropy than MR766ΔE153-156, indicating that passaging of the latter led to both a sequence deletion and lower genomic diversity. The contemporary strains (Asian/American) had increased genomic diversity compared to strains from Africa. Specifically, strain R103451 had the highest mean entropy in the dataset. Next, high diversity values for nucleotide positions in a given strain were defined as those with entropy values greater than 2σ (two times the standard deviation) of the Shannon entropy of the population for each strain. The number of nucleotide positions containing high nucleotide diversity was greater for strains PA259249 and R103451 (148 and 151, respectively), accounting for 1.4% of the positions examined, but for other strains, high entropy positions (range of 55-97) comprised less than 1% of the positions (Figure 3c and S3). Overall, strains PA259249 and R103451 contained 1.7-, 2.3-, and 2.5-fold greater numbers of high diversity positions than East African, West African, or Asian (FSS13025) (p = 0.066), respectively.

Shannon entropy of each gene region (Table S3) was analysed to determine the distribution of genetic diversity. Cumulatively, genetic diversity corresponded to the gene length (Figure S4), with the NS5 gene containing highest entropy values, followed by NS3, E, and NS1. However, relative diversity, expressed relative to gene length, had a different distribution (Figure 3e). The two human serum strains exhibited highest relative diversity in many gene regions compared to other strains. Specifically, the prM/M and NS2B gene region were highest for strain FSS13025, and prM/M and NS1 gene regions were highest for strain PA259249 (Figure 3e). These results indicate that high relative genetic diversity may be important for the role of the prM/M during infection.

Single nucleotide variant sites are distributed throughout the ZIKV genome

The contribution of SNVs to the genetic population of ZIKVs was determined using Vphaser2 v2.0. Results from duplicate datasets were combined to yield a total number of SNV positions ranging from 44 to 170 depending on the strain. SNVs were categorized as encoding synonymous or non-synonymous substitutions (Tables S4 and S5, respectively), and comprehensive lists are provided in the supplement. The frequency of each SNV was plotted against the corresponding positional entropy value to reveal a strong correlation between entropy value and SNV frequency (Figure 4a). Subsequent analysis was performed only with SNVs that were present in both biological replicates of each ZIKV. Strains MR766, MR766ΔE153-156, and DakAr41667, and FSS13025 have greater number of synonymous SNVs, whereas strains DakAr41524, PA259249, and R103451 have greater numbers of non-synonymous SNVs (Figure 4b).

SNV frequency was plotted along the genome, and all strains had SNVs exceeding 1% frequency of the population (Figure 4c). Of these, a majority were non-synonymous for American strains: PA259249 (12 of 14) and R103451 (4 of 6), but not for the others: MR766 (0 of 11), MR766ΔE153-156 (2 of 11), DakAr41667 (1of 3), and DakAr41524 (1of 2), and FSS13025 (7 of 15).

Several non-synonymous SNVs with greater than 1% frequency were located in the prM/M gene encoding for: DakAr41524 A136; FSS13025 K104, S109, L145; PA259249 T107, T119; and R103451 I163. Furthermore, Asian/American strains had several non-synonymous SNVs with greater than 1% frequency in the NS1 protein: FSS13025 M220; PA259249 R103, K128, M220 (2 nucleotides), K265, and E342; and R103451 K94 and Y175. Surprisingly, strain PA259249 contained 21 SNVs in the NS1 gene; 20 were non-synonymous, and 6 were present at a frequency of 1% or greater. Furthermore, strains MR766, FSS13025, and PA25924 contained multiple SNVs at a given amino acid position (Table 2). Most were located in the NS genes, specifically in NS1. These data showed a trend for flexible codon usage in the NS1 gene, particularly for strain PA259249, suggesting it possesses a high degree of variant diversity.

Table 2. ZIKV amino acid positions containing multiple variants in the same codon.


Common inter- and intra-lineage SNVs

In order to discover advantageous SNVs that were maintained in the population, variants shared by one or more ZIKV strains were identified (Table 3 and Figure 5). Fourteen of the seventeen SNVs shared by at least two ZIKVs are located in ZIKV proteins with solved structures: C, (prM/)M, E, NS1, NS3, and NS5 proteins (Figure S5). No SNVs were shared between East and West African lineages. However, the Asian/American lineage shared 11 SNV amino acid positions with either the East or West African lineages (Table 3). Seven SNV positions were shared between the East African and Asian/American lineages: C G73, prM/M F130, E A270, NS1 L145, NS1 K265, NS4B F86, and NS5 H855. Two SNVs, NS1 L145 and NS1 K265, were less than 1% and synonymous in the East African lineage but greater than 1% and non-synonymous in Asian/American lineage. Four SNV positions were shared between the West African and Asian/American lineages: prM/M S109, NS1 M220, NS2A A117 and NS5 C269. Three non-synonymous SNVs (prM/M S109, NS1 M220 and NS2A A117). were present at greater than 1% frequency in Asian/American strains yet were less than 1% frequency in the West African Strains.

Table 3. Common inter- and intra-lineage SNVs per protein amino acid position.


Intra-lineage comparisons revealed two SNV positions common to only East African strains (NS1 S228 and NS4B L206) and four SNV positions common to only Asian/American strains (prM/M T107, E H401, E A501, and NS3 R583) (Table 3). The high frequency SNV prM/M T107 was only shared between strains FSS13025 and PA2592495. It was a synonymous, codon position 3, for strain FSS13025 but non-synonymous, codon position 2, for strain PA259249 (Tables S4 and S5). Likewise, strains R103451 and FSS13025 both had a SNV in NS3 583: synonymous, codon position 3, in strain FSS13025 but non-synonymous, codon position 2, in strain R103451. All of the SNVs shared between PA259249 and R103451 were low frequency SNVs. Several shared SNVs were located in the carboxy-terminus of the E protein and the transmembrane portion of the cleaved prM/M protein located in the mature virion (Figure S5b). SNVs shared in the NS1 protein localized to the beta-ladder (220, 228, 265) and to the wing domain (145). Together, these data demonstrate that specific SNVs are maintained in the genomic populations of ZIKVs among lineages.

Comparison of strains from the Americas

The genomes of the two American strains were examined in detail in order to compare the diversity of two strains with identical passage history. The mean genomic entropy was higher for strain R103451 (Figure 3c). However, strain PA259249 had approximately twice the number of SNVs. Approximately 25% of SNVs from both strains exceeded 1% of the population (Figure 4c). Strains PA259249 and R103451 differ in consensus genomic sequence by 38 nucleotides and 9 amino acid substitutions throughout the genome (Figure 5), 4 of which were located in the NS1 protein (F58S, G100A, Y175S, and W324R). SNVs were detected for two of these changes in the dataset, F58S (7.72%) in strain PA259249 and S175Y (2.94%) in strain R103451. Additionally, the consensus change NS2A G224R was also a SNV in R103451 (2.91%). Lastly, the consensus change NS3 M572L was a SNV detected in DakAr41667, albeit at a low frequency (0.06%). These data take into account the similarity in cell culture passaging of the two strains from the Americas to reveal differences in genomic diversity.

Figure 5. SNVs were detected in four of the nine consensus sequence changes between strains PA259249 and R103451. A) Genome schematic depicting the amino acid differences between strains PA259249 and R103451, red: denotes positions of SNVs in the dataset across lineages. B) Nucleotide, polyprotein, and protein number of coding differences between the two strains, as well as the variants detected (% denotes frequency of SNV) are listed. Structural depiction of each consensus coding change, in black, or consensus coding change with SNV detected, in red, for the NS1 dimer (C), NS3 helicase (D), and NS5 polymerase (E). No structure is available for amino acids C 107 and NS2A 224.

Display full size

Analysis of NGS data from the public repository

A limited set of NGS data deposited in Sequence Read Archive (SRA) was analysed in order to corroborate the genetic diversity identified in this study with existing sequence data. Public data for strains MR766ΔE153-156, DakAr41524, PA259249, and R103451 were analysed using the pipeline described in this study to determine the SNVs in each sample (Figure S6). Because the deposited sample data contained 4–10 runs for each sample entry, the data were pooled during analysis. However, the mean coverage across the genome was still lower in the downloaded dataset, with a range of 332-1,611 after processing. Strain PA259249 (sample #1) was the only sample with coverage >1500, which is the cut-off typically used for downstream entropy and SNV analysis. Nevertheless, a SNV analysis was performed. For MR766ΔE153-156 (#4) the overwhelming majority of SNVs were synonymous (Figure S6), as previously characterized (Figure 4), whereas the majority of SNVs for strains PA259249 (#1) and R103451 (#5) were non-synonymous (Figure S6), as previously characterized (Figure 4). However, analysis of two different entries of strain DakAr41524 (#2 and #6) resulted in different numbers of SNVs (64 and 3, respectively). The number of SNVs that were shared among the stocks sequenced in this study and the downloaded data are listed in Table S4 as follows: MR766ΔE153-156 (#4)−4; DakAr41524 (#2)−3; DakAr41524 (#6)−1; PA259249 (#1)−18; and R103451 (#5)−5. Notably NS1 145, NS1 220, and NS2A 117 were shared among several strains, as previously characterized (Table 3 and S6). Overall, analyses of the sequence data downloaded from the SRA and the data generated in this study had similaries. However, some strains differed in the SNVs, possibly due to differences in sample passages, sequencing platforms, genome coverage depth, and number of runs. These factors can influence genetic diversity studies.

Figure 4. SNVs detected in all ZIKV strains. A) For all positions with SNVs, the frequency percentage and entropy values are correlated (Spearman correlation, r = 0.9793). B) The total number of SNVs identified and common to both replicates of each ZIKV are shown: synonymous change (blue bar), non-synonymous change (red bar). C) Mean frequency values of all SNVs common to both replicates were plotted by nucleotide position, synonymous (blue), non-synonymous (red). Genomic regions (UTRs and proteins) are denoted by dotted lines and alternating background shading under a genome map. The number of SNVs present at greater than 1% frequency out of the total number of SNVs is provided on the right.

Display full size

Discussion

The present study evaluated genetic diversity across lineages of ZIKV analysing NGS of representative strains from various sources (mosquito and mammalian), including a human placenta isolate. The dataset was robust with sufficient coverage and map quality scores (Figure 2Table 2) to analyse intra-host diversity of ZIKV. The mean genomic diversity of the Asian/American lineage was higher than those of the African lineages, and American isolates had a greater number of high entropy positions (Figure 3a-c and S3). Intra-lineage diversity differed between the East African strains MR766 and MR766ΔE153-156, likely due to the number of passages since isolation in 1947. However, there was no statistical support for intra-lineage diversity difference between the two West African strains, whose passage histories differed by one passage (Table 1). The Asian/American lineage strains FSS13025 and PA259249 were not statistically different in diversity, yet the diversity of strain R103451 differed significantly from both. Because both strains PA259249 and R103451 were passaged similarly, the intra-host diversity differences may be due to other characteristics, such as the geographic location of isolation (Panama versus Honduras) or to tissue-specific (serum versus placenta) effects on the genomic diversity of ZIKV. Other studies have shown that the genetic diversity of serum samples collected during the outbreak in the Americas does not differ from that of Asian strains [30Metsky HCMatranga CBWohl S, et al. Zika virus evolution and spread in the Americas. Nature. 2017;546:411415. doi: 10.1038/nature22402[Crossref][Web of Science ®] [Google Scholar],31Faria NRQuick JClaro IM, et al. Establishment and cryptic transmission of Zika virus in Brazil and the Americas. Nature. 2017;546:406410. doi: 10.1038/nature22401[Crossref][Web of Science ®] [Google Scholar]], which supports the data in this study. One limitation of this study is that, due to widespread isolation times and locations, the ZIKV strains selected were passaged a number of times in different cells or tissues which may not reflect the natural diversity of these ZIKV strains. For each ZIKV, the lowest passage possible was used. It is worth noting that strains FSS13025 and PA259249 did not differ in diversity even though the former was passaged twice in C6/36 mosquito cells, while the latter was not passaged in C6/36 cells at all.

Amino acids in prM/M and NS1 proteins have been associated with virulence of ZIKV strains [32–34Xia HLuo HShan C, et al. An evolutionary NS1 mutation enhances Zika virus evasion of host interferon induction. Nat Commun. 2018;9:414. doi: 10.1038/s41467-017-02816-2
Yuan LHuang X-YLiu Z-Y, et al. Single mutation in the prM protein of Zika virus contributes to microcephaly. Science. 2017;358:933936. doi: 10.1126/science.aam7120
Liu YLiu JDu S, et al. Evolutionary enhancement of Zika virus infectivity in Aedes aegypti mosquitoes. Nature. 2017;545:482486. doi: 10.1038/nature22365 
]. The present analysis showed that Asian/American strains (and strain MR766) had higher relative entropy values in the prM/M gene region, and additional regions of high relative diversity for the Asian/American lineage include NS1 (FSS13025, PA259249, and R103451), E (R103451), NS2B (FSS13025 and R103451), NS4B (FSS13025), and 3’UTR (R103451). Therefore, the clusters of highest genomic diversity for the Asian/American lineage are located in the prM/M and NS1 gene regions.

Sub-consensus-level diversity was also measured by SNV analysis. Strain MR766 contained a greater number of SNVs than all other strains, especially compared to MR766ΔE153-156, which suggests that passaging of the latter led to not only a deletion in the E protein, but also decreased the SNV population. Interestingly, the mosquito isolates from West Africa had the fewest number of high frequency SNVs. The possibility that this is due to a bottleneck caused by virus propagation in arthropod cells followed by mammalian cells cannot be excluded; however, molecular epidemiology showed that contemporary (American) isolates of human serum, urine, or plasma clustered together with those from mosquito pools from the Americas [30Metsky HCMatranga CBWohl S, et al. Zika virus evolution and spread in the Americas. Nature. 2017;546:411415. doi: 10.1038/nature22402[Crossref][Web of Science ®] [Google Scholar]], which may or may not be extrapolated to other ZIKV lineages. The Asian/American lineage strains had four non-synonymous SNVs in the prM/M gene region and at least two in the NS1 gene region. Most notably, strain PA259249 had 20 non-synonymous SNVs in NS1, which was the greatest number of non-synonymous SNVs per gene region of the dataset (Figure 4c). Lastly, inter-lineage SNVs were dispersed throughout the genome.

Several SNVs detected in this study correspond to positions of evolutionary importance identified in phylogenetic studies. Specifically, 17 of the amino acid changes reported by Pettersson and colleagues [11Pettersson JHEldholm VSeligman SJ, et al. How did Zika virus emerge in the Pacific islands and Latin America. MBio. 2016;7:e012396. doi: 10.1128/mBio.01239-16[Crossref][Web of Science ®] [Google Scholar]] contained SNVs in at least one strain replicate in the present study (Tables S4 and S5), and several were located in both replicates of at least one strain: C 110 (R103451), NS3 583 (FSS13025 and R103451) and NS5 784 (R103451). These three amino acids and nine more were also reported by Wang and colleagues [35Wang LValderramos SGWu A, et al. From Mosquitos to humans: genetic evolution of Zika virus. Cell Host Microbe. 2016;19:561565. doi: 10.1016/j.chom.2016.04.006[Crossref][Web of Science ®] [Google Scholar]]. Ultimately, sequence analyses can work in concert to provide insight into the composition and genetic diversity of ZIKVs.

Previous variant analyses of ZIKVs from different sources are in accordance with the present study. NGS analysis of ZIKV isolated from macaques experimentally infected with the French Polynesia strain led to identification of eight variants with frequencies greater than 5%, most of which were located between the carboxy-terminus of the E protein through NS2B [36Dudley DMAliota MTMohr EL, et al. A rhesus macaque model of Asian-lineage Zika virus infection. Nat Commun. 2016;7:12204. doi: 10.1038/ncomms12204[Crossref][Web of Science ®] [Google Scholar]]. Likewise, NGS analysis of WT strain PRVABC59 amplicons showed that the majority of variants are located in the E and NS1 protein regions [37Weger-Lucarelli JAl E. Development and Characterization of recombinant virus generated from a New World Zika virus Infectious Clone. J Virol. 2017;91:110.[Web of Science ®] [Google Scholar]]. Several NGS analyses of American ZIKV strains have examined the molecular epidemiology of ZIKV during the recent epidemic [30Metsky HCMatranga CBWohl S, et al. Zika virus evolution and spread in the Americas. Nature. 2017;546:411415. doi: 10.1038/nature22402[Crossref][Web of Science ®] [Google Scholar],31Faria NRQuick JClaro IM, et al. Establishment and cryptic transmission of Zika virus in Brazil and the Americas. Nature. 2017;546:406410. doi: 10.1038/nature22401[Crossref][Web of Science ®] [Google Scholar],38Grubaugh NDLadner JTKraemer MUG, et al. Genomic epidemiology reveals multiple introductions of Zika virus into the United States. Nature. 2017;546:401405. doi: 10.1038/nature22400[Crossref][Web of Science ®] [Google Scholar]]. The sequences of 110 patient and mosquito samples resulted in identification of 21 SNVs, the majority of which (18) occurred in nucleotides 118-3,987 (spanning from C to NS2A protein) [30Metsky HCMatranga CBWohl S, et al. Zika virus evolution and spread in the Americas. Nature. 2017;546:411415. doi: 10.1038/nature22402[Crossref][Web of Science ®] [Google Scholar]]. Five of those 21 SNVs were detected in the present study, and three additional SNVs were detected in the same codon (but not nucleotide position) [30Metsky HCMatranga CBWohl S, et al. Zika virus evolution and spread in the Americas. Nature. 2017;546:411415. doi: 10.1038/nature22402[Crossref][Web of Science ®] [Google Scholar]], which provides additional evidence for clusters of diversity.

A previous study evaluating similar ZIKV strains identified consensus differences within and between lineages and conducted an extensive phylogenetic analysis, while the variant analysis was restricted to high frequency SNVs. Interestingly, five SNVs identified by Shrivastava and colleagues were also identified in this study (prM/M 46, E 161, E 401, NS2A 117, and NS2A 157), albeit in lower frequencies and not necessarily in the same strain. The differences in number, strain, and frequencies are likely due to the combination of different variant analysis software used and the restriction to high frequency SNVs. Vphaser2, used in the present study, was validated with a Flavivirus dataset and designed to identify low frequency SNVs, thus allowing analysis of all identified SNVs [39Shrivastava SPuri VDilley KA, et al. Whole genome sequencing, variant analysis, phylogenetics, and deep sequencing of Zika virus strains. Sci Rep. 2018;8:111. doi: 10.1038/s41598-017-17765-5[Crossref][Web of Science ®] [Google Scholar]].

Although, the present study focused on genetic analysis, examination of these isolates in vitro and in vivoin the literature has shown that ZIKVs belong to a single serotype and share a highly virulent phenotype in immunocompromised mice, while NHPs recover from infection. Strains MR766 and FSS13025 have been characterized in immunocompromised mouse [40–42Lazear HMGovero JSmith AM, et al. A mouse model of Zika virus Pathogenesis. Cell Host Microbe. 2016;19:111. doi: 10.1016/j.chom.2016.03.010
Rossi SLTesh RBAzar SR, et al. Characterization of a Novel Murine model to study Zika virus. Am J Trop Med Hyg. 2016;94:13621369. doi: 10.4269/ajtmh.16-0111
Zmurko JMarques RESchols D, et al. The viral polymerase inhibitor 7-deaza-2’-C-methyladenosine is a potent inhibitor of in vitro Zika virus replication and delays disease progression in a robust mouse infection model. PLoS Negl Trop Dis. 2016;10:115. doi: 10.1371/journal.pntd.0004695 
] and non-human primate (NHP) models [43–46Aliota MTDudley DMNewman CM, et al. Heterologous protection against Asian Zika virus challenge in Rhesus macaques. PLoS Negl Trop Dis. 2016;10:122.
Adams Waldorf KMStencel-Baerenwald JEKapur RP, et al. Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate. Nat Med. 2016;22:12561259. doi: 10.1038/nm.4193
Azar SRRossi SLHaller SH, et al. ZIKV demonstrates minimal pathologic effects and mosquito infectivity in viremic cynomolgus macaques. Viruses. 2018;10:120. doi: 10.3390/v10110661
Koide FGoebel SSnyder B, et al. Development of a zika virus infection model in cynomolgus macaques. Front Microbiol. 2016;7:18. doi: 10.3389/fmicb.2016.02028 
]. An early study in NHPs found that the E protein non-glycosylated mutant (equivalent to MR766ΔE153-156) regained the glycosylation site following in vivo infection [43Aliota MTDudley DMNewman CM, et al. Heterologous protection against Asian Zika virus challenge in Rhesus macaques. PLoS Negl Trop Dis. 2016;10:122.[Crossref][Web of Science ®] [Google Scholar]]. Strain DakAr41667 is also virulent in immunocompromised mice [40Lazear HMGovero JSmith AM, et al. A mouse model of Zika virus Pathogenesis. Cell Host Microbe. 2016;19:111. doi: 10.1016/j.chom.2016.03.010[Crossref][Web of Science ®] [Google Scholar]]. Strains PA259249 and R103451 have been used for baby mouse challenge [47Tai WHe LWang Y, et al. Critical neutralizing fragment of Zika virus EDIII elicits cross-neutralization and protection against divergent Zika viruses article. Emerg Microbes Infect. 2018;7:18. doi: 10.1038/s41426-017-0007-8[Taylor & Francis Online][Web of Science ®] [Google Scholar]] and in vitro analyses [48Moser LABoylan BTMoreira FR, et al. Growth and adaptation of Zika virus in mammalian and mosquito cells. PLoS Negl Trop Dis. 2018;12:119.[Crossref][Web of Science ®] [Google Scholar],49Ojha CRRodriguez MKaruppan MKM, et al. Toll-like receptor 3 regulates Zika virus infection and associated host inflammatory response in primary human astrocytes. PLoS One. 2019;14:126. doi: 10.1371/journal.pone.0208543[Crossref][Web of Science ®] [Google Scholar]]. Lastly, the virulence of strain DakAr41524 has been shown to be comparable to that of MR766 in dendritic cells [50Bowen JRQuicke KMMaddur MS, et al. Zika virus Antagonizes type I Interferon Responses during infection of human dendritic cells. PLoS Pathog. 2017;13:130. doi: 10.1371/journal.ppat.1006164[Crossref][Web of Science ®] [Google Scholar]].

This study provides a broad view of ZIKV genetic diversity and posits that recent American isolates harbors increased non-synonymous SNV populations that may contribute to the virulence of contemporary ZIKVs in humans. The present analyses did not include phenotypic studies; however, studies have shown that both prM/M and NS1 may contribute to fitness or virulence. The prM/M S17N substitution increased infection of neural cells and neonatal mice, with the latter exhibiting increased mortality. Also, embryonic infection of mouse brains with virus encoding prM/M S17N led to microcephaly, including smaller brain size and increased apoptosis in brain cortices [33Yuan LHuang X-YLiu Z-Y, et al. Single mutation in the prM protein of Zika virus contributes to microcephaly. Science. 2017;358:933936. doi: 10.1126/science.aam7120[Crossref][Web of Science ®] [Google Scholar]]. Also, a recent study found that the prM/M protein has a role in neural cell cytotoxicity and apoptosis in vitro [51Li GBos STsetsarkin KA, et al. The roles of prM-E proteins in historical and epidemic zika virus-mediated infection and neurocytotoxicity. Viruses. 2019;11:E157. doi: 10.3390/v11020157[Crossref][Web of Science ®] [Google Scholar]]. Vaccine studies showed that inclusion of the prM/M sequence in an adenovirus-vectored vaccine containing the E protein increased the ZIKV-specific neutralizing antibody response and provided sterilizing immunity in a mouse challenge model [52Guo QChan JFWPoon VKM, et al. Immunization with a novel human type 5 adenovirus-vectored vaccine expressing the premembrane and envelope proteins of zika virus provides consistent and sterilizing protection in multiple immunocompetent and immunocompromised animal models. J Infect Dis. 2018;218:365377. doi: 10.1093/infdis/jiy187[Crossref][Web of Science ®] [Google Scholar]]. These results suggested that interactions between prM/M and E are important for epitope display in ZIKV. Furthermore, subsequent studies found that presence of both prM/M and NS1 in the adenovirus-vectored vaccine was important for effective protection in mouse models because an Ad2-prM-E-NS1 vaccine provided better protection to neonatal mice than Ad2-prM-E [53Liu XQu LYe X, et al. Incorporation of NS1 and prM/M are important to confer effective protection of adenovirus-vectored Zika virus vaccine carrying E protein. npj Vaccines. 2018;3:18. doi: 10.1038/s41541-017-0041-5[Crossref][Web of Science ®] [Google Scholar]]. Moreover, an evaluation of a prM-E-NS1 recombinant VSV-vectored vaccine showed that NS1 likely plays a role in modulating the cellular immune response during vaccination [54Li AYu JLu M, et al. A Zika virus vaccine expressing premembrane-envelope-NS1 polyprotein. Nat Commun. 2018;9:3067. doi: 10.1038/s41467-018-05276-4[Crossref][Web of Science ®] [Google Scholar]]. Furthermore, NS1 V188A was implicated in antagonism of IFN-beta induction by decreasing the phosphorylation of the TBK1 intermediate [32Xia HLuo HShan C, et al. An evolutionary NS1 mutation enhances Zika virus evasion of host interferon induction. Nat Commun. 2018;9:414. doi: 10.1038/s41467-017-02816-2[Crossref][Web of Science ®] [Google Scholar]]. The NS1 V188A mutation also increased infectivity in Aedes aegypti mosquitoes [34Liu YLiu JDu S, et al. Evolutionary enhancement of Zika virus infectivity in Aedes aegypti mosquitoes. Nature. 2017;545:482486. doi: 10.1038/nature22365[Crossref][Web of Science ®] [Google Scholar]].

The American ZIKV strain PRVABC59 was originally isolated from human serum. A recent study showed that Vero cell passaging led to increased emergence of the variants NS1 W98G and E V330L, presumably to increase fitness in Vero cell culture. The presence of both SNVs attenuated strain PRVABC59 in mice [55Duggal NKMcDonald EMWeger-Lucarelli J, et al. Mutations present in a low-passage Zika virus isolate result in attenuated pathogenesis in mice. Virology. 2019;530:1926. doi: 10.1016/j.virol.2019.02.004[Crossref][Web of Science ®] [Google Scholar]]. In the present study, SNVs were also detected at NS1 W98 in all three codon positions of strain PA259249; however, the resulting substitutions differed W98R/S/C and were low frequency (0.2-0.5%). Another NS1 mutation, K265E, increased ZIKV replication in Vero cells by increasing virion assembly; addition of a second mutation at prM/M H83R further increased viral titers in Vero cells [56Yang YShan CZou J, et al. A cDNA Clone-Launched platform for high-yield Production of Inactivated Zika vaccine. EBioMedicine. 2017;17:145156. doi: 10.1016/j.ebiom.2017.02.003[Crossref][Web of Science ®] [Google Scholar]]. Both of these amino acids were detected as SNVs in the present analysis. The SNV at NS1 K265E was detected in both DakAr41524 (0.2%) and in PA259249 (3.7%), while a synonymous SNV (0.8%) at this position was detected in MR766ΔE153-156. Lastly, prM/M H83R was identified as a SNV (0.3%) in R103451.

The present study shows a clustering of non-synonymous SNVs in the prM/M and NS1 proteins, especially in the contemporary ZIKVs. These results are in accordance with previous ZIKV characterizations in the literature. Subsequent analyses will be required in order to determine the contribution of SNVs to virulence and fitness.

Acknowledgements

This work was supported by a John Sealy Memorial Research Endowment pilot grant to VVS. We thank Jill K. Thompson for assistance with NGS, Thomas G. Wood and Alan D.T. Barrett for helpful discussions, and Hilda Guzman from the World Reference Center for Emerging Viruses and Arboviruses.

Disclosure statement

No potential conflict of interest was reported by the authors.

ORCID

Vanessa V. Sarathy http:///0000-0002-1262-2101

Correction Statement

This article has been republished with minor changes. These changes do not impact the academic content of the article.

Supplemental material

Inter- and intra-lineage genetic diversity of wild-type Zika viruses reveals both common and distinctive nucleotide variants and clusters of genomic diversity

    References

  • Dick GWKitchen SHaddow A. Zika virus (I). Isolations and serological specificity. Trans R Soc Trop Med Hyg.1952;46:509520. doi: 10.1016/0035-9203(52)90042-4 



  • Musso DGubler DJ. Zika virus. Clin Microbiol Rev. 2016;29:487524

  • Duffy MChen THancock W, et al. Zika virus outbreak on Yap island, Federated States of Micronesia. N Engl J Med. 2009;360:25362543

  • Weaver SCCosta FGarcia-Blanco MA, et al. Zika virus: history, emergence, biology, and prospects for control. Antiviral Res. 2016;130:6980

  • Cao-Lormeau V-MBlake AMons S, et al. Guillain-Barré syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study. Lancet. 2016;387:15311539. doi: 10.1016/S0140-6736(16)00562-6 



  • Costello ADua TDuran P, et al. Defining the syndrome associated with congenital Zika virus infection. Bull World Health Organ. 2016;94:406406A

  • Faye OFreire CCMIamarino A, et al. Molecular evolution of Zika virus during its emergence in the 20th Century. PLoS Negl Trop Dis. 2014;8:e2636

  • Lanciotti RSLambert AJHolodniy M, et al. Phylogeny of Zika virus in Western Hemisphere, 2015. Emerg Infect Dis J. 2016;22:933935

  • Ye QLiu Z-YHan J-F, et al. Genomic characterization and phylogenetic analysis of Zika virus circulating in the Americas. Infect Genet Evol. 2016;43:4349



  • Kochakarn TKotanan NKümpornsin K, et al. Comparative genome analysis between Southeast Asian and South American Zika viruses. Asian Pac J Trop Med. 2016;9:10481054. doi: 10.1016/j.apjtm.2016.10.002 



  • Pettersson JHEldholm VSeligman SJ, et al. How did Zika virus emerge in the Pacific islands and Latin America. MBio. 2016;7:e012396. doi: 10.1128/mBio.01239-16 



  • Kuno GChang GJTsuchiya KR, et al. Phylogeny of the genus flavivirus. J Virol. 1998;72:7383



  • Heinz FXAllison SL. Flavivirus structure and membrane fusion. Adv Virus Res. 2003;59:6397. doi:10.1016/S0065-3527(03)59003-0 



  • Lindenbach BDRice C. Molecular biology of flaviviruses. Adv Virus Res. 2003;59:2361. doi: 10.1016/S0065-3527(03)59002-9 



  • Chambers TJHahn CSGaller R, et al. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol. 1990;44:649688. doi: 10.1146/annurev.mi.44.100190.003245 



  • Bray MMen RTokimatsu I, et al. Genetic determinants responsible for acquisition of dengue type 2 virus mouse neurovirulence. J Virol. 1998;72:16471651



  • de Borba LStrottmann DMde Noronha L, et al. Synergistic interactions between the NS3 hel and E proteins contribute to the virulence of dengue virus type 1. PLoS Negl Trop Dis. 2012;6:e1624. 



  • Davis CTGalbraith SEZhang S, et al. A combination of Naturally Occurring mutations in North American West Nile virus nonstructural protein genes and in the 3’ Untranslated region Alters virus phenotype. J Virol.2007;81:61116116



  • Yu Y. Phenotypic and genotypic characteristics of Japanese encephalitis attenuated live vaccine virus SA14-14-2 and their stabilities. Vaccine. 2010;28:36353641



  • Beck ATesh RBWood TG, et al. Comparison of the live attenuated yellow fever vaccine 17D-204 strain to its virulent parental strain Asibi by deep sequencing. J Infect Dis. 2014;209:334344. doi: 10.1093/infdis/jit546 


    21.Collins NDBeck ASWiden SG, et al. Structural and nonstructural genes contribute to the genetic diversity of RNA Viruses. MBio. 2018;9:113



  • Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res.2004;32:17921797. doi: 10.1093/nar/gkh340 



  • Stamatakis A. RAxML version 8: A tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics. 2014;30:13121313. doi: 10.1093/bioinformatics/btu033 



  • Miller MAPfeiffer WSchwartz T. Creating the CIPRES Science Gateway for inference of large phylogenetic trees. 2010 Gateway Computing Environments Workshop; 2010 Nov 14; New Orleans, LA; 2010


  • Piccolo SRFrampton MB. Tools and techniques for computational reproducibility. Gigascience. 2016;5:113. doi:10.1186/s13742-016-0135-4 



  • Fishman SLBranch AD. The quasispecies nature and biological implications of the hepatitis C virus. Infect Genet Evol. 2009;9:11581167. doi: 10.1016/j.meegid.2009.07.011 



  • Nasu AMarusawa HUeda Y, et al. Genetic heterogeneity of hepatitis C virus in association with antiviral therapy determined by ultra-deep sequencing. PLoS One. 2011;6:e24907. doi: 10.1371/journal.pone.0024907 



  • Nishijima NMarusawa HUeda Y, et al. Dynamics of hepatitis B virus quasispecies in association with nucleos(t)ide analogue treatment determined by ultra-deep sequencing. PLoS One. 2012;7:e35052. doi:10.1371/journal.pone.0035052 



  • Heang VYasuda CYSovann L, et al. Zika virus infection, Cambodia, 2010. Emerg Infect Dis. 2012;18:349351. doi: 10.3201/eid1802.111224 



  • Metsky HCMatranga CBWohl S, et al. Zika virus evolution and spread in the Americas. Nature. 2017;546:411415. doi: 10.1038/nature22402 



  • Faria NRQuick JClaro IM, et al. Establishment and cryptic transmission of Zika virus in Brazil and the Americas. Nature. 2017;546:406410



  • Xia HLuo HShan C, et al. An evolutionary NS1 mutation enhances Zika virus evasion of host interferon induction. Nat Commun. 2018;9:414. doi: 10.1038/s41467-017-02816-2 



  • Yuan LHuang X-YLiu Z-Y, et al. Single mutation in the prM protein of Zika virus contributes to microcephaly. Science. 2017;358:933936. doi: 10.1126/science.aam7120 



  • Liu YLiu JDu S, et al. Evolutionary enhancement of Zika virus infectivity in Aedes aegypti mosquitoes. Nature.2017;545:482486. doi: 10.1038/nature22365 



  • Wang LValderramos SGWu A, et al. From Mosquitos to humans: genetic evolution of Zika virus. Cell Host Microbe. 2016;19:561565. doi: 10.1016/j.chom.2016.04.006 



  • Dudley DMAliota MTMohr EL, et al. A rhesus macaque model of Asian-lineage Zika virus infection. Nat Commun. 2016;7:12204. doi: 10.1038/ncomms12204 



  • Weger-Lucarelli JAl E. Development and Characterization of recombinant virus generated from a New World Zika virus Infectious Clone. J Virol. 2017;91:110



  • Grubaugh NDLadner JTKraemer MUG, et al. Genomic epidemiology reveals multiple introductions of Zika virus into the United States. Nature. 2017;546:401405. doi: 10.1038/nature22400 



  • Shrivastava SPuri VDilley KA, et al. Whole genome sequencing, variant analysis, phylogenetics, and deep sequencing of Zika virus strains. Sci Rep. 2018;8:111. doi: 10.1038/s41598-017-17765-5 



  • Lazear HMGovero JSmith AM, et al. A mouse model of Zika virus Pathogenesis. Cell Host Microbe. 2016;19:111. doi: 10.1016/j.chom.2016.03.010 



  • Rossi SLTesh RBAzar SR, et al. Characterization of a Novel Murine model to study Zika virus. Am J Trop Med Hyg. 2016;94:13621369. doi: 10.4269/ajtmh.16-0111 



  • Zmurko JMarques RESchols D, et al. The viral polymerase inhibitor 7-deaza-2’-C-methyladenosine is a potent inhibitor of in vitro Zika virus replication and delays disease progression in a robust mouse infection model. PLoS Negl Trop Dis. 2016;10:115. doi: 10.1371/journal.pntd.0004695 



  • Aliota MTDudley DMNewman CM, et al. Heterologous protection against Asian Zika virus challenge in Rhesus macaques. PLoS Negl Trop Dis. 2016;10:122



  • Adams Waldorf KMStencel-Baerenwald JEKapur RP, et al. Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate. Nat Med. 2016;22:12561259. doi: 10.1038/nm.4193 



  • Azar SRRossi SLHaller SH, et al. ZIKV demonstrates minimal pathologic effects and mosquito infectivity in viremic cynomolgus macaques. Viruses. 2018;10:120. doi: 10.3390/v10110661 



  • Koide FGoebel SSnyder B, et al. Development of a zika virus infection model in cynomolgus macaques. Front Microbiol. 2016;7:18. doi: 10.3389/fmicb.2016.02028 



  • Tai WHe LWang Y, et al. Critical neutralizing fragment of Zika virus EDIII elicits cross-neutralization and protection against divergent Zika viruses article. Emerg Microbes Infect. 2018;7:18. doi: 10.1038/s41426-017-0007-8 



  • Moser LABoylan BTMoreira FR, et al. Growth and adaptation of Zika virus in mammalian and mosquito cells. PLoS Negl Trop Dis. 2018;12:119



  • Ojha CRRodriguez MKaruppan MKM, et al. Toll-like receptor 3 regulates Zika virus infection and associated host inflammatory response in primary human astrocytes. PLoS One. 2019;14:126. doi:10.1371/journal.pone.0208543 



  • Bowen JRQuicke KMMaddur MS, et al. Zika virus Antagonizes type I Interferon Responses during infection of human dendritic cells. PLoS Pathog. 2017;13:130. doi: 10.1371/journal.ppat.1006164 



  • Li GBos STsetsarkin KA, et al. The roles of prM-E proteins in historical and epidemic zika virus-mediated infection and neurocytotoxicity. Viruses. 2019;11:E157. doi: 10.3390/v11020157 



  • Guo QChan JFWPoon VKM, et al. Immunization with a novel human type 5 adenovirus-vectored vaccine expressing the premembrane and envelope proteins of zika virus provides consistent and sterilizing protection in multiple immunocompetent and immunocompromised animal models. J Infect Dis. 2018;218:365377. doi:10.1093/infdis/jiy187 



  • Liu XQu LYe X, et al. Incorporation of NS1 and prM/M are important to confer effective protection of adenovirus-vectored Zika virus vaccine carrying E protein. npj Vaccines. 2018;3:18. doi: 10.1038/s41541-017-0041-5 



  • Li AYu JLu M, et al. A Zika virus vaccine expressing premembrane-envelope-NS1 polyprotein. Nat Commun.2018;9:3067. doi: 10.1038/s41467-018-05276-4 



  • Duggal NKMcDonald EMWeger-Lucarelli J, et al. Mutations present in a low-passage Zika virus isolate result in attenuated pathogenesis in mice. Virology. 2019;530:1926. doi: 10.1016/j.virol.2019.02.004 


    56.Yang YShan CZou J, et al. A cDNA Clone-Launched platform for high-yield Production of Inactivated Zika vaccine. EBioMedicine. 2017;17:145156. doi: 10.1016/j.ebiom.2017.02.003 

    本站是提供个人知识管理的网络存储空间,所有内容均由用户发布,不代表本站观点。请注意甄别内容中的联系方式、诱导购买等信息,谨防诈骗。如发现有害或侵权内容,请点击一键举报。
    转藏 分享 献花(0

    0条评论

    发表

    请遵守用户 评论公约

    类似文章 更多