Skip to main content
Erschienen in: Virology Journal 1/2018

Open Access 01.12.2018 | Research

Full-length genome and molecular characterization of dengue virus serotype 2 isolated from an imported patient from Myanmar

verfasst von: Zhaoping Zeng, Jiandong Shi, Xiaofang Guo, Ling Mo, Ningzhu Hu, Jing Sun, Meini Wu, Hongning Zhou, Yunzhang Hu

Erschienen in: Virology Journal | Ausgabe 1/2018

Abstract

Background

Dengue is the most common mosquito-borne infection worldwide and a serious threat to global public health. Sporadic dengue virus serotype 2 (DENV-2) imported cases from Myanmar have been documented almost every year in Yunnan Province of China since 2005. However, the complete genome sequences of DENV-2 isolates imported from Myanmar are not available.

Methods

The full-length genome of the DENV-2 strain (YNPE2), isolated from an imported case from Myanmar in 2013, was identified by the next-generation sequencing. The extreme ends of the viral genome were validated by 5′/3′ RACE and Sanger sequencing. Furthermore, phylogenetic, recombination and selection pressure analyses were conducted for the molecular characterization of YNPE2 strain.

Results

Whole-genome sequencing revealed that the full-length sequence of YNPE2 strain was 10,724 bases, with an open reading frame encoding for 3391 amino acids. The YNPE2 strain had 99.0% nucleotide identity and 99.8% amino acid identity with two closely related strains, ThD2_0078_01 strain (DQ181797) and DENV-2/TH/BID-V2157/200 strain (FJ639832). The phylogenetic analysis suggested that the YNPE2 strain belonged to Asian I genotype and was likely derived from Thailand strain (DQ181797). Moreover, selection pressure analysis revealed two amino acid sites of the NS4B and NS5 proteins, with important evidence of positive selection.

Conclusion

This study revealed the first complete genome sequence and molecular characterization of a DENV-2 strain (YNPE2) isolated from an imported case from Myanmar, thus providing a valuable reference genome source for future surveillance, epidemiology and vaccine development of DENV-2 virus in Yunnan, China.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12985-018-1043-2) contains supplementary material, which is available to authorized users.
Zhaoping Zeng and Jiandong Shi contributed equally to this work.
Abkürzungen
DENV-2
Dengue virus serotype 2
DF
Dengue fever
DHF
Dengue hemorrhagic fever
DSS
Dengue shock syndrome
FBS
Fetal bovine serum
FEL
Fixed effects likelihood
IFEL
Internal fixed effect likelihoods
MEME
Mixed effects model of evolution
ML
Maximum likelihood
NGS
Next-generation sequencing
NS
Non-structural
nts
Nucleotides
ORF
Open reading frame
PCR
Polymerase chain reaction
RDP
Recombination Detection Program
SLAC
Single-likelihood ancestor counting
UTR
Untranslated regions

Background

Dengue, a re-emerging mosquito-borne human viral disease, is caused by the dengue virus (DENV). The global prevalence of the disease has markedly increased in recent decades, with about half of the world′s population under threat [1]. More than 390 million people worldwide are infected by DENV every year, of which 96 million have clinical symptoms, ranging from mild dengue fever (DF) to dengue hemorrhagic fever (DHF) or Dengue shock syndrome (DSS) [2].
DENV is a single stranded and positive polarity RNA virus with a genome of about 11,000 bases in length [3], and belongs to the genus Flavivirus, family Flaviviridae [4]. The viral genome only has a long open reading frame (ORF) that encodes a polyprotein, and is flanked by 5′ untranslated region (UTR) and 3′ UTR. The polyprotein can be processed into three structural proteins (capsid, membrane and envelope) and seven non-structural (NS) proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5) [3]. The DENV is mainly transmitted by Aedes aegypti mosquitoes in tropical and sub-tropical countries [5]. Serologically, DENV is classified into four serotypes (DENV-1 to DENV-4), which share 65–70% homology [6]. Due to its high mutation rate, DENV has generated different genotypes and lineages within each serotype [7]. Revealing the genotype of the circulating DENV in epidemic areas is critical to disease surveillance, epidemiology and vaccine development.
With the increase in population mobility, the DENV and mosquitoes can quickly travel by modern transportation across the globe. Thus, the endemic areas of dengue have expanded from 9 countries before 1970 to over 100 countries worldwide [1]. Among them, South-East Asia, America and the Western Pacific regions are seriously affected by dengue, due to their tropical and sub-tropical climate. In South-East Asia, all countries have reported outbreaks of dengue since 1950 [814]. In China, the first dengue epidemic was documented in Guangdong Province in 1978 [15]. In recent decades, the number of dengue patients have gradually increased in some provinces of China, including Guangdong, Yunnan, Fujian and Zhejiang [1619]. Generally, numerous dengue outbreaks in China are closely related to imported cases from countries in the South-East Asia. Thus, dengue remains an imported disease in China [20].
Yunnan Province is located in the Southwest border of China, and shares a 4060-km border with three dengue-endemic countries: Myanmar, Laos and Vietnam. With the construction of the China-ASEAN Free Trade Area and the transport infrastructure in the Greater Mekong Subregion, Yunnan and South Asia and South-East Asia countries are experiencing an increase in trade and tourism. Hence, Yunnan Province faces an increasing need for dengue prevention and control due to numerous imported cases from the neighboring countries. The national dengue surveillance program was implemented in Yunnan province in 2005, and an increasing number of imported cases have been reported each year [21, 22]. In 2008, an outbreak of dengue was caused by imported cases from Myanmar [21]. Moreover, a large-scale dengue outbreak was reported in 2013 in Dehong and Xishuangbanna Prefectures of Yunnan Province. The outbreak was predominantly related to imported cases from Myanmar and Laos [23]. Therefore, imported cases from Myanmar and Laos were likely the main cause of the dengue outbreak in Yunnan Province. However, majority of the previous studies on the imported DENV-2 strain were based on partial sequencing data of the envelope (E) gene. The complete genome sequence of any DENV-2 strain imported from Myanmar is not available. In this study, we reported full-length genome sequence and molecular characterization of the YNPE2 strain isolated from an imported case from Myanmar. This study will likely facilitate future surveillance, epidemiology studies and vaccine development of DENV-2 virus in Yunnan, China.

Methods

Case presentation

In September 2013, a 32-year-old female patient, who came from Myanmar, was clinically diagnosed with dengue fever in Ruli, Yunnan, China. She had dengue-like symptoms including fever, joint pain, myalgia and headache. The patient′s acute serum was positive for NS1 antigen by One Step Dengue nonstructural protein (NS) 1 RapiDipTM InstaTest (Cortez). The patient’s acute serum (2–5 ml) was collected with informed consent and stored at -80 °C until testing.

Virus isolation, identification and serotyping

Acute-phase serum samples collected from the dengue patient were used for virus isolation. Briefly, the serum was diluted at a ratio of 1:10 using RPMI 1640 medium (Biological Industries, USA), followed by incubation for 1 h with Aedes albopictus C6/36 cells for virus adsorption in a 75 cm2 flask (Corning, USA). Then, RPMI 1640 medium supplemented with 2% fetal bovine serum (FBS; Gibco) was added to maintain cell survival. Cells were incubated at 28 °C in 5% CO2 and examined daily for cytopathic effects (CPE). When CPE was observed, the cell culture supernatant was collected, which served as virus stock. The virus stock was used for infecting C6/36 cells. Once CPE was observed, the cell culture supernatant was collected and analyzed by reverse transcription PCR (RT-PCR) to determine the serotype of the virus. The viral RNA was extracted from infected cell culture supernatant using TRIzol reagent (Tiangen, Beijing, China) according to the manufacturer′s protocol. Total RNA was reverse transcribed using GoScript™ Reverse Transcriptase (Promega, WI, USA) according to the manufacturer′s instructions. The reaction was conducted for 5 min at 25 °C, followed by 1 h at 42 °C and 15 min at 70 °C in a BioRad C1000 Cycler system. Then, PCR was performed using Taq polymerase (TaKaRa, Dalian, China) using a reference protocol with DENV universal primers (D1: 5′ -TGAATATGCTGAAACGCGCGAGAAACCG-3′ and D2: 5′ -TTGCACCAACAGTCAATGTCTTCAGGTTC-3′) and DENV-2 typing primer (D1 and TS2: 5′ -CGCCAGAAGGGCCATGAACAG-3′) [24]. Agarose gel electrophoresis was conducted to check the specificity of amplicons [24].

Whole-genome sequencing analysis

The viral RNA was extracted from infected cell culture supernatant using the TRIzol reagent (Invitrogen, CA, USA) according to the manufacturer′s protocol. The total RNA was depleted of rRNA with an epicentre Ribo-ZeroTM kit (Illumina, CA, USA). The rRNA-depleted RNA was purified and fragmented using a RNA fragmentation kit (Ambion, USA). The sequencing library was constructed as previously described [25]. The sequencing was performed on an Illumina Hiseq 4000 platform (LC Sciences, Hangzhou, China) according to the manufacturer′s standard protocol, and 2 × 150 bp paired-end reads (PE150) were generated. The joint and low quality reads were removed from the raw sequencing reads to obtain the clean reads. The viral genome was assembled from the clean reads by the SPAdes (v3.10.1) program [26].

5′ / 3′ rapid amplification of cDNA ends (RACE)

The viral RNA was extracted from infected cell culture supernatant using the TRIzol reagent (Invitrogen, CA, USA). Since the total RNA was extracted from a non-eukaryotic organism and lacked a polyadenylated tail, the 5′-first-strand cDNA was synthesized with random primers and the 3′-first-strand cDNA was synthesized after adding a poly(A) tail using Poly(A) Polymerase (TaKaRa, Dalian, China) according to the instruction of SMARTer 5′/3′ RACE kit (Clontech, CA, USA). The 5′/3′ RACE PCR was performed by using SeqAmp DNA Polymerase (TaKaRa, Dalian, China) with Universal Primer Mix and 5′ Gene-Specific Primer (5′-CGCCATTATGGTGAAGCCTGGATGTCTC-3′) / 3′ Gene-Specific Primer (5′-ATCTGGGAGGCCACAAACCATGGAAGC-3′) according to the instruction of SMARTer 5′/ 3′ RACE kit. The PCR reaction involved 40 cycles of 94 °C for 30 s, 60 °C for 30 s, and 72 °C for 2 min in a BioRad C1000 Cycler system. The PCR products of 5′ RACE and 3′ RACE were diluted at a ratio of 1:10 with nuclease-free water. The second round PCRs were performed in a reaction mixture of 50 μL containing 5 μL of 10 × La Taq Buffer (Mg2+ Plus), 8 μL of dNTPs (2.5 mM), 5 μL of 10 × Universal Primer Mix, 1 μL of 5′/3′ Gene-Specific Primer, 2.5 μL of diluted 5′/3′ RACE PCR products and 1 unit of high fidelity La Taq DAN polymerase (TaKaRa, Dalian, China). The second round PCRs were conducted under conditions of one cycle of 94 °C for 5 min followed by 40 cycles of 94 °C for 30 s, 60 °C for 30 s, and 72 °C for 2 min. The amplified products were purified from 1.5% agarose gel using the Wizard SV Gel and PCR Clean-Up System (Promega, WI, USA). The purified products were ligated to the T4 vector (Promega, WI, USA), transformed into E. coli, and positive clones were selected for Sanger sequencing (BGI, Shenzhen).

Accession number

The full-length genome sequence of the DENV-2 strain YNPE2 was deposited in GenBank (accession number MF459663.3).

Genomic characterization analysis

The viral genomic characterization and amino acid sequence was analyzed using the DNASTAR Lasergene 11.1 software package (DNASTAR Inc., USA). Multiple sequence alignments were performed using the ClustalX2.1 program [27]. The Mfold software package (http://​unafold.​rna.​albany.​edu/​?​q=​mfold) was utilized to predict the secondary structure of the UTRs of the viral genome with the default folding parameters [28].

Phylogenetic analysis

Phylogenetic analysis based on the DENV-2 coding sequence and the envelope gene were performed using the MEGA program (version 6.06) [29, 30]. The phylogenetic trees were deduced by employing the Maximum Likelihood (ML) method based on the General Time Reversible model [31]. The bootstrap replications was set at 1000 for estimating the Branch topology of the phylogenetic tree.

Recombination analysis

Recombination detection of the DENV-2 was conducted by seven methods, including RDP [32], Chimaera [33], BootScan [34], 3Seq [35], GENCONV [36], MaxChi [37] and SiScan [38] available in the Recombination Detection Program (RDP version 4.95), with p < 0.01. The recombinant event was identified if a sequence was detected by at least three methods under the multiple comparison correction setting option.

Selection pressure analysis

The selection pressure analysis for codons of the open reading frame (ORF) of the viruses (excluding potential recombinants) was performed by single-likelihood ancestor counting (SLAC) [39], fixed effects likelihood (FEL) [40], internal fixed effect likelihoods (IFEL) [41] and mixed effects model of evolution (MEME) [42] available in HyPhy (Hypothesis testing using phylogenies) open-source software package (http://​www.​hyphy.​org/). The ratio of non-synonymous to synonymous substitutions (dN/dS ratio, ω) were calculated in four methods. The sites were considered significant if they were detected by three or more methods.

Results

Identification and serotyping of virus

To observe the proliferation properties of the virus, C6/36 cells were repeatedly infected with virus stock. At 8 days post-infection, typical CPE was observed in the C6/36 cells. Furthermore, the RT-PCR analysis showed two specific bands, 511 bp and 119 bp in length, which were specifically amplified by DENV universal primers and DENV-2 typing primers (Fig. 1), indicating that the virus was dengue virus serotype 2. The virus was named as YNPE2 strain.

Full-length sequence and genomic characterization of YNPE2

The results of RNA-seq yielded 7,811,508 clean reads from 8,328,038 raw reads after removing unqualified reads. The full-length sequence of the YNPE2 strain was assembled by SPAdes software based on the RNA-seq data and the referenced genome of DENV-2 (DQ181797). The assembled full-length sequence of YNPE2 was 10,741 nucleotides (nts) with an ORF encoding a polyprotein of 3391 amino acids, flanked by 160 nts and 405 nts at the 5′ UTR and 3′ UTR, respectively. The RACE was considered a reliable method for confirming the extreme ends of RNA genome. Thus, the UTR sequences of viral genome were verified by 5′/3′ RACE and Sanger sequencing. Interestingly, the Sanger sequencing results of 5′ UTR and 3′ UTR were different from the Illumina′s next-generation sequencing (NGS). Sanger sequencing showed that the 5′ UTR and 3′ UTR were 96 nts and 452 nts, respectively (Fig. 2). Thus, based on 5′/3′ RACE and Sanger sequencing, the complete genome of YNPE2 was corrected to 10, 724 nts, flanked by 96 nts and 452 nts at the 5′ UTR and 3′ UTR, respectively. To determine the sequence identity between the YNPE2 strain and other DENV-2 strains, the complete genome sequence of YNPE2 was aligned with the prototype strain of DENV-2 (New Guinea C, AF038403) and 38 other globally representative DENV-2 strains (Additional file 1: Table S1). As shown in Table 1, the YNPE2 strain showed 89.5–99.0% sequence identity with the other strains. No insertion or deletion of nucleotides were observed in the protein-coding region when aligned with these representative strains. Moreover, the alignment results revealed that base substitutions were distributed over the whole stretch of the alignment. A single base substitution at position 54 in the 5′ UTR was observed when aligned with the prototype. As compared to the 3′ UTR of the prototype, the YNPE2 had 14 substitutions (position 10,275 [G → A], 10,279 [A → G], 10,289 [G → A], 10,321 [T → C], 10,387 [T → C], 10,389 [C → T], 10,390 [A → T], 10,413 [G → A], 10,418 [A → T], 10,553 [G → A], 10,555 [T → C], 10,570 [G → A], 10,575 [A → G] and 10,609 [A → G]), a single base (G) deletion at position 10,415 and a single nucleotide (A) insertion at position 10,455. The RNA secondary structure prediction results showed that these nucleotides substitutions in the 3′ UTR of YNPE2 led to minor change in the secondary stem loop structure as compared to the prototype (Fig. 3). The predicted RNA secondary structure of 3′ UTR of YNPE2 had fewer loops than the prototype.
Table 1
Nucleotide (NT) and amino acid (AA) identity of YNPE2 (MF459663) complete genome compared to representative globally diverse full-length DENV-2 sequences
Serial number
Accession number
NT identity (%)
AA identity (%)
1
EU056811
89.5
96.7
2
GQ868588
89.5
96.7
3
AY702040
89.7
96.7
4
HM582105
89.8
96.8
5
HM582108
89.8
96.9
6
HM582099
89.9
96.9
7
JN851123
91.1
97.2
8
HQ012538
92.1
97.1
9
JX470186
91.2
97.1
10
FJ898454
91.3
97.4
11
FJ850088
91.4
96.7
12
GQ252676
91.4
97.4
13
EU179858
91.5
97.0
14
EU482636
91.7
97.0
15
FJ639703
91.8
97.4
16
EU482788
91.9
97.4
17
AY702039
92.0
97.0
18
EU529695
92.2
97.1
19
GQ398271
92.1
97.1
20
GQ868540
92.2
97.1
21
FJ898450
92.3
97.3
22
GQ398264
92.3
97.3
23
DQ181801
92.4
97.4
24
AF119661
92.4
97.4
25
GQ398269
92.4
97.3
26
JF730055
94.4
98.0
27
HQ891024
94.4
97.9
28
AF204178
94.4
97.7
29
EU854293
94.6
98.1
30
AF038403
94.5
98.1
31
GQ398268
94.6
98.3
32
FJ196851
96.0
98.2
33
FJ639705
98.2
99.5
34
FJ906957
98.3
99.5
35
FJ410215
98.4
99.7
36
EU482445
98.5
99.7
37
GQ868543
98.7
99.5
38
FJ639832
99.0
99.8
39
DQ181797
99.0
99.8

Amino acid sequence diversity

In order to characterize viral amino acid sequences, the deduced amino acid sequence of the YNPE2 strain was aligned with 39 complete genome sequences of DENV-2 representing the global diversity of DENV-2 (Additional file 1: Table S1). The sequence alignment revealed 96.7–99.8% identity among the sequences (Table 1) and amino acid substitutions throughout the complete coding region. The result indicated 66 amino acid replacements between YNPE2 and the prototype strain of DENV-2 (New Guinea C, AF038403) (Table 2). In addition, based on nucleotide and amino acid homology, two DENV-2 strains, ThD2_0078_01 strain (DQ181797) and DENV-2/TH/BID-V2157/200 strain (FJ639832), were the most closely related to YNPE2.
Table 2
Description of amino acid substitutions in YNPE2 compared to the prototype (New Guinea C)
Serial number
AAa position (ORF)
AA position (Protein)
MF459663 (YNPE2)
AF038403 (New Guinea)
Capsid
 1
9
9
K
R
 2
101
101
S
T
prM
 3
129
15
G
S
4
130
16
I
R
 5
169
55
L
F
 6
266
152
V
A
E
 7
351
71
E
D
 8
363
83
K
N
9
406
126
E
K
 10
409
129
I
V
 11
421
141
V
I
 12
444
164
V
I
13
456
176
A
T
 14
506
226
K
T
 15
508
228
E
G
16
626
346
Y
H
 17
682
402
F
I
 18
764
484
I
V
NS1
 19
825
50
Q
H
20
855
80
A
S
 21
880
105
R
Q
22
904
129
Y
H
 23
947
172
K
R
 24
952
177
A
V
 25
997
222
N
S
 26
1022
247
L
F
27
1036
261
Y
H
28
1039
264
I
T
 29
1056
281
D
E
 30
1061
286
I
V
NS2A
31
1132
5
V
T
 32
1178
51
K
R
 33
1184
57
V
M
 34
1192
65
A
T
 35
1247
120
V
I
 36
1263
136
I
M
 37
1266
139
N
K
 38
1285
158
T
A
 39
1308
181
L
F
 40
1316
189
T
A
NS2B
 41
1402
57
T
A
 42
1457
112
I
V
 43
NS3
   
 44
1488
13
M
V
 45
1595
120
T
A
 46
1662
187
R
K
 47
1808
333
I
M
 48
1820
345
N
S
 49
2024
549
K
R
 50
2081
606
A
T
NS4A
 51
2114
21
T
A
NS4B
 52
2262
19
A
T
 53
2355
112
L
F
 54
2418
175
I
V
NS5
 55
2626
135
I
T
 56
2792
301
R
K
 57
2826
335
L
V
 58
2828
337
T
M
 59
2920
429
G
S
 60
3122
631
N
S
 61
3132
641
V
I
 62
3167
676
H
S
 63
3178
687
I
V
 64
3189
698
K
R
 65
3310
819
R
Q
 66
3356
865
A
T
Note: The amino acid site including replacement of hydrophilic to hydrophobic or vice versa are marked in bold font. The amino acid substitution of negatively charged to positively charged amino acid is written in bold and italic font
a AA amino acid

Phylogenetic analysis

To determine the genetic relationship between YNPE2 and other DENV-2 strains isolated from geographically diverse areas, phylogenetic analysis of the whole coding sequence (Additional file 2: Table S2) and the envelope gene (Additional file 3: Table S3) were performed. The phylogenetic tree based on the complete coding sequences revealed that the DENV-2 strain belonged to the Asian I genotype along with multiple DENV-2 isolates from Vietnam, Thailand, and Cambodia. The YNPE2 strain was clustered into one group, together with Cambodia strain (FJ639718) and Thailand strain (DQ181797) (Fig. 4). The phylogenetic analysis suggested that the YNPE2 strain was likely derived from the Cambodia strain (FJ639718) or the Thailand strain (DQ181797). Moreover, the phylogenetic tree based on 73 envelope gene sequences isolated from geographically diverse areas showed that YNPE2 was clustered into a group with three Myanmar isolates (KJ470758, KJ470752 and KJ470762) and three Chinese isolates (KX262954, KX262957 and KY038916) circulating in 2013, which suggested their common origin (Fig. 5). In this study, the phylogenetic trees of the whole coding sequence and envelope gene were similar, with little diversity.

Recombination analysis

The potential recombination events of the YNPE2 strain and other representative DENV-2 isolates (Additional file 1: Table S1) were analyzed by RDP4 software. The results of preliminary analysis revealed that four recombination events might have occurred among these DENV-2 strains (p < 0.01) (Additional file 4: Table S4). These recombination events seemed to be limited to some strains, accounting for 10% (4 out of 40 strains). However, no potential recombination event was detected for the YNPE2 strain within the tested isolates.

Selection pressure analysis

To determine the influence of the selection pressure acting on complete coding region of DENV-2 isolates (Additional file 1: Table S1), an analysis based on the selection coefficient ω (dN/dS ratio) at every codon was conducted with a cutoff value (ω > 1) of positive selection. The results showed that most of the codons in DENV-2 were under negative selection. However, a total of 13 sites were verified to be under positive selection pressure through at least three out of the four approaches (Table 3). Moreover, two of these sites, positioned on NS4B and NS5 respectively, indicated important evidence of episodic positive selection through at least two different methods with a low p-value (p < 0.1). Limited evidence of positive selection was confirmed by SLAC, FEL and IFEL .
Table 3
Selection pressure analysis of the ORF (3391 codons) of DENV-2 using MEME, SLAC, FEL and IFEL
AAa position (ORF)
MEME
SLAC
FEL
IFEL
ω
p-Value
ω
p-Value
ω
p-Value
ω
p-Value
prM
 248
55.584
0.062
4.778
0.445
1.525
0.277
1.123
0.278
 253
> 100
0.089
2.368
0.672
0.789
0.446
< 0
1
Envelope
 408
> 100
0.093
2.309
0.712
0.824
0.406
2.171
0.333
 735
> 100
0.096
1.602
0.995
0.486
9.56
< 0
1
NS1
 986
> 100
0.054
2.417
0.68
0.826
0.322
< 0
1
 1010
> 100
0.017
2.431
0.664
0.713
0.416
2.098
0.305
NS2A
 1142
> 100
< 0.001
3.231
0.988
0.997
0.929
< 0
1
 1195
> 100
0.093
1.685
0.946
0.576
0.819
< 0
1
NS2B
 1439
> 100
0.019
4.765
0.447
1.485
0.246
< 0
1
 1459
> 100
0.014
5146
0.8
1.81
0.649
1.044
0.718
NS3
 1489
> 100
0.067
9.674
0.198
3.111
0.979
< 0
1
NS4B
2262
5.444
0.0838
13.16
0.108
3.4
0.088
6.196
0.082
NS5
3310
2.249
0.107
8.065
0.296
4.012
0.085
8.109
0.058
Note: Criteria to determine codons with important evidence of positive selection: p-value < 0.1 in MEME, SLAC, FEL and IFEL. Codons that are detected positive by at least three approaches are listed in the table. Codons with important evidence of positive selection by at least two approaches are marked in bold font
aAA amino acid

Discussion

For human viral diseases, knowledge of the full-length genome sequence, genotype and genomic characterization of a pathogen are essential for disease surveillance, epidemiology, and vaccine development. In this study, we used complete genome sequencing and comparative sequence analysis to determine the complete genome sequence and the molecular characterization of a DENV-2 strain (YNPE2) isolated from an imported case from Myanmar. It provides critical information on the whole genome sequence and genomic characterization of DENV-2 isolates imported from Myanmar to China.
Geographically, Yunnan Province in China borders with three dengue hyper-endemic countries, including Myanmar, Laos and Vietnam. With the increase in border trade between China and Myanmar, the risk of dengue import is also increasing. In 2008, a dengue outbreak caused by imported case from Myanmar to Ruili City was monitored [21]. Forty-nine dengue patients were clinically diagnosed based on the IgM antibody and viral nucleic acid detection, of which 48 were imported from Myanmar [21]. Moreover, a recent study on dengue epidemic characterization in Yunnan also demonstrated that the imported cases from Myanmar and Laos were mainly responsible for the dengue outbreak in Yunnan [43]. Thus, the investigation of complete genome and molecular characterization of different imported DENV isolates is essential for control and prevention of DENV circulating in Yunnan. In the present study, next-generation sequencing (NGS) Illumina Hiseq 4000 was employed for whole genome sequencing of the YNPE2 strain based on its advantages of high throughput, ultra-high speed, no mark and low cost [44, 45]. The UTR sequences were confirmed by 5′/3′ RACE, which is a credible method for determining the precise UTRs of RNA genome. The 5′/3′ RACE results were inconsistent with NGS, perhaps due to some artefacts of deep sequencing or assembly protocols. Therefore, 5′/3′ RACE and Sanger sequencing are essential for determining the precise viral genome. A highly reliable full-length genome sequence of YNPE2 was obtained using NGS, 5′/3′ RACE and Sanger sequencing. The conserved UTRs at both ends of the viral genome can form stable secondary structures, which play important roles in viral translation, replication and assembly [46, 47]. Interestingly, genomic characterization analysis from this study revealed some base substitutions in the 3′ UTR. The potential biological implications of the nucleotide substitution remains unknown, but their molecular configuration was slightly changed. Further studies are needed to determine the influence of the substitution on DENV virulence and replication in host cells.
Numerous amino acid substitutions were observed in the structural and non-structural proteins of the YNPE2 strain as compared to the prototype of DENV-2 (Table 2). These amino acid substitutions may potentially influence viral fitness and virulence in host cells. Interestingly, some amino acid substitutions, such as E71D, E126K and F402I observed in the envelope protein in this study, have been confirmed in previous studies [48, 49]. These amino acid substitutions enhanced the neurovirulence of the virus. Moreover, multiple amino acid substitutions in NS1, NS2A, NS2B and NS5 proteins of YNPE2 were observed. The potential influence of these substitutions needs further investigation.
Gene recombination plays a key role in the evolution and adaptation of DENV [50]. For molecular characterization of the YNPE2 strain, we selected 39 other representative globally diverse DENV-2 complete sequences to identify potential recombination events. No potential recombination event was found in the YNPE2 strain. Perhaps, more DENV-2 complete sequences are needed to uncover any potential recombination events of YNPE2. The analysis of site-specific selection pressures for DENV-2 indicated that strong negative selection was present, which was ascribed to the evolutionary constraints of arboviruses [7]. The positive selection from host-specific immunity contributes to viral protein mutations that allow the virus to escape host immune recognition [51, 52]. In this study, two sites with important evidence of positive selection were determined. One site in NS4B was reported in an earlier study of sylvatic DENV-2 and had an inhibitory effect on the interferon signaling pathway [53, 54]. Another site in NS5 was reported for the first time in our study, and its functional importance needs further investigation.
The phylogenetic analysis, based on the complete coding sequence, showed a close relationship between the YNPE2 strain and DQ181797 (Thailand, 2001), which suggested that the YNPE2 strain very likely originated in Thailand. This conclusion was further supported by the results of the sequence and amino acid identity. A phylogenetic tree based on the envelope gene revealed different subgenotype circulating in Myanmar. Moreover, the YNPE2 strain and seven Yunnan strains are closely related to isolates from Myanmar, suggesting that the dengue outbreaks in Yunnan Province are mainly caused by imported cases from Myanmar.

Conclusion

This study presented the first complete sequence of DENV-2 imported from Myanmar, and its molecular characterization. These results will stimulate further investigations of DENV infection, proliferation, pathogenicity, epidemiology and vaccine development.

Acknowledgements

We would like to thank Dr. Nigel W. Fraser of Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA, for proof-reading and reviewing our manuscript.

Funding

This work was supported by grants from the CAMS Innovation Fund for Medical Sciences (CIFMS) (grant no. 2017-I2M-3-022), the National Key Research and Development Program of the Ministry of Science and Technology of China (2016YFC1202300), the State Project for Essential Drug Researchand Development (SQ2018ZX091510), the National Natural Science Foundation of China (31500724), the Yunnan Applied Basic Research Projects (2017FB115, 2017ZF007), and the Peking Union Medical College Youth Fund (3332016113), and the Fundamental Research Funds for the Central Universities, China.

Availability of data and materials

The genome sequence of the DENV-2 (YNPE2 strain) has been deposited into GenBank database under the accession number MF459663.3.
Written consent was obtained from the patient. The study protocol was approved by the Institutional Ethics Committee of Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College.
All the authors consent to publish.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, Drake JM, Brownstein JS, Hoen AG, Sankoh O, et al. The global distribution and burden of dengue. Nature. 2013;496:504–7.CrossRefPubMedPubMedCentral Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, Drake JM, Brownstein JS, Hoen AG, Sankoh O, et al. The global distribution and burden of dengue. Nature. 2013;496:504–7.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Chambers TJ, Hahn CS, Galler R, Rice CM. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol. 1990;44:649–88.CrossRefPubMed Chambers TJ, Hahn CS, Galler R, Rice CM. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol. 1990;44:649–88.CrossRefPubMed
5.
Zurück zum Zitat Salazar MI, Richardson JH, Sánchez-Vargas I, Olson KE, Beaty BJ. Dengue virus type 2: replication and tropisms in orally infected Aedes aegypti mosquitoes. BMC Microbiol. 2007;7:9.CrossRefPubMedPubMedCentral Salazar MI, Richardson JH, Sánchez-Vargas I, Olson KE, Beaty BJ. Dengue virus type 2: replication and tropisms in orally infected Aedes aegypti mosquitoes. BMC Microbiol. 2007;7:9.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Anoop M, Mathew AJ, Jayakumar B, Issac A, Nair S, Abraham R, Anupriya MG, Sreekumar E. Complete genome sequencing and evolutionary analysis of dengue virus serotype 1 isolates from an outbreak in Kerala, South India. Virus Genes. 2012;45:1–13.CrossRefPubMed Anoop M, Mathew AJ, Jayakumar B, Issac A, Nair S, Abraham R, Anupriya MG, Sreekumar E. Complete genome sequencing and evolutionary analysis of dengue virus serotype 1 isolates from an outbreak in Kerala, South India. Virus Genes. 2012;45:1–13.CrossRefPubMed
7.
Zurück zum Zitat Holmes E, Twiddy S. The origin, emergence and evolutionary genetics of dengue virus. Infect Genet Evol. 2003;3:19–28.CrossRefPubMed Holmes E, Twiddy S. The origin, emergence and evolutionary genetics of dengue virus. Infect Genet Evol. 2003;3:19–28.CrossRefPubMed
8.
Zurück zum Zitat Khampapongpane B, Lewis HC, Ketmayoon P, Phonekeo D, Somoulay V, Khamsing A, Phengxay M, Sisouk T, Vongphrachanha P, Bryantcd JE. National dengue surveillance in the Lao People’s Democratic Republic,2006–2012: epidemiological and laboratory findings. Surveillance Report. 2014;5:7–13. Khampapongpane B, Lewis HC, Ketmayoon P, Phonekeo D, Somoulay V, Khamsing A, Phengxay M, Sisouk T, Vongphrachanha P, Bryantcd JE. National dengue surveillance in the Lao People’s Democratic Republic,2006–2012: epidemiological and laboratory findings. Surveillance Report. 2014;5:7–13.
9.
Zurück zum Zitat Cuong HQ, Vu NT, Cazelles B, Boni MF, Thai KTD, Rabaa MA, Quang LC, Simmons CP, Huu TN, Anders KL. Spatiotemporal dynamics of dengue epidemics, Southern Vietnam. Emerg Infect Dis. 2013;19:945–53.CrossRefPubMedPubMedCentral Cuong HQ, Vu NT, Cazelles B, Boni MF, Thai KTD, Rabaa MA, Quang LC, Simmons CP, Huu TN, Anders KL. Spatiotemporal dynamics of dengue epidemics, Southern Vietnam. Emerg Infect Dis. 2013;19:945–53.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Chen SP. Molecular evolution and epidemiology of four serotypes of dengue virus in Thailand from 1973 to 2007. Epidemiol Infect. 2013;141:419–24.CrossRefPubMed Chen SP. Molecular evolution and epidemiology of four serotypes of dengue virus in Thailand from 1973 to 2007. Epidemiol Infect. 2013;141:419–24.CrossRefPubMed
11.
Zurück zum Zitat Kyaw AK, Ngwe Tun MM, Moi ML, Nabeshima T, Soe KT, Thwe SM, Myint AA, Maung KTT, Aung W, Hayasaka D, et al. Clinical, virological and epidemiological characterization of dengue outbreak in Myanmar, 2015. Epidemiol Infect. 2017;145:1886–97.CrossRefPubMed Kyaw AK, Ngwe Tun MM, Moi ML, Nabeshima T, Soe KT, Thwe SM, Myint AA, Maung KTT, Aung W, Hayasaka D, et al. Clinical, virological and epidemiological characterization of dengue outbreak in Myanmar, 2015. Epidemiol Infect. 2017;145:1886–97.CrossRefPubMed
12.
Zurück zum Zitat Massad E, Coutinho FA, Ma S, Burattini MN. A hypothesis for the 2007 dengue outbreak in Singapore. Epidemiol Infect. 2010;138:951–7.CrossRefPubMed Massad E, Coutinho FA, Ma S, Burattini MN. A hypothesis for the 2007 dengue outbreak in Singapore. Epidemiol Infect. 2010;138:951–7.CrossRefPubMed
13.
Zurück zum Zitat Mohd-Zaki AH, Brett J, Ismail E, L’Azou M. Epidemiology of dengue disease in Malaysia (2000-2012): a systematic literature review. PLoS Negl Trop Dis. 2014;8:e3159.CrossRefPubMedPubMedCentral Mohd-Zaki AH, Brett J, Ismail E, L’Azou M. Epidemiology of dengue disease in Malaysia (2000-2012): a systematic literature review. PLoS Negl Trop Dis. 2014;8:e3159.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Vong S, Khieu V, Glass O, Ly S, Duong V, Huy R, Ngan C, Wichmann O, Letson GW, Margolis HS, Buchy P. Dengue incidence in urban and rural Cambodia: results from population-based active fever surveillance, 2006–2008. PLoS Negl Trop Dis. 2010;4:e903.CrossRefPubMedPubMedCentral Vong S, Khieu V, Glass O, Ly S, Duong V, Huy R, Ngan C, Wichmann O, Letson GW, Margolis HS, Buchy P. Dengue incidence in urban and rural Cambodia: results from population-based active fever surveillance, 2006–2008. PLoS Negl Trop Dis. 2010;4:e903.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Fan W, Yu S, Cosgriff TM. The reemergence of dengue in China. Rev Infect Dis. 1989;11:S847–53.CrossRefPubMed Fan W, Yu S, Cosgriff TM. The reemergence of dengue in China. Rev Infect Dis. 1989;11:S847–53.CrossRefPubMed
16.
Zurück zum Zitat Guo R, Lin J, Li L, Ke C, He J, Zhong H, Zhou H, Peng Z, Yang F, Liang W. The prevalence and endemic nature of dengue infections in Guangdong, South China: an epidemiological, serological, and etiological study from 2005–2011. PLoS One. 2014;9:e85596.CrossRefPubMedPubMedCentral Guo R, Lin J, Li L, Ke C, He J, Zhong H, Zhou H, Peng Z, Yang F, Liang W. The prevalence and endemic nature of dengue infections in Guangdong, South China: an epidemiological, serological, and etiological study from 2005–2011. PLoS One. 2014;9:e85596.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Wang B, Li Y, Feng Y, Zhou H, Liang Y, Dai J, Qin W, Hu Y, Wang Y, Zhang L, et al. Phylogenetic analysis of dengue virus reveals the high relatedness between imported and local strains during the 2013 dengue outbreak in Yunnan, China: a retrospective analysis. BMC Infect Dis. 2015;15:142.CrossRefPubMedPubMedCentral Wang B, Li Y, Feng Y, Zhou H, Liang Y, Dai J, Qin W, Hu Y, Wang Y, Zhang L, et al. Phylogenetic analysis of dengue virus reveals the high relatedness between imported and local strains during the 2013 dengue outbreak in Yunnan, China: a retrospective analysis. BMC Infect Dis. 2015;15:142.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Wang J, Chen H, Huang M, Zhang Y, Xie J, Yan Y, Zheng K, Weng Y. Epidemiological and etiological investigation of dengue fever in the Fujian province of China during 2004–2014. Sci China Life Sci. 2017;60:72–80.CrossRefPubMed Wang J, Chen H, Huang M, Zhang Y, Xie J, Yan Y, Zheng K, Weng Y. Epidemiological and etiological investigation of dengue fever in the Fujian province of China during 2004–2014. Sci China Life Sci. 2017;60:72–80.CrossRefPubMed
19.
Zurück zum Zitat Sun J, Lin J, Yan J, Fan W, Lu L, Lv H, Hou J, Ling F, Fu T, Chen Z, et al. Dengue virus serotype 3 subtype III, Zhejiang Province, China. Emerg Infect Dis. 2011;17:321–3.CrossRefPubMedPubMedCentral Sun J, Lin J, Yan J, Fan W, Lu L, Lv H, Hou J, Ling F, Fu T, Chen Z, et al. Dengue virus serotype 3 subtype III, Zhejiang Province, China. Emerg Infect Dis. 2011;17:321–3.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Sang S, Chen B, Wu H, Yang Z, Di B, Wang L, Tao X, Liu X, Liu Q. Dengue is still an imported disease in China: a case study in Guangzhou. Infect Genet Evol. 2015;32:178–90.CrossRefPubMed Sang S, Chen B, Wu H, Yang Z, Di B, Wang L, Tao X, Liu X, Liu Q. Dengue is still an imported disease in China: a case study in Guangzhou. Infect Genet Evol. 2015;32:178–90.CrossRefPubMed
21.
Zurück zum Zitat Zhang H, Fu S, Deng Z, Yuan J, Jiang H, Li M, Gao X, Wang J, Liu Y. An outbreak of imported dengue fever from Myanmar to the border of China, with its viral molecular epidemiological features. Zhonghua Liu Xing Bing Xue Za Zhi. 2013;34:428–32.PubMed Zhang H, Fu S, Deng Z, Yuan J, Jiang H, Li M, Gao X, Wang J, Liu Y. An outbreak of imported dengue fever from Myanmar to the border of China, with its viral molecular epidemiological features. Zhonghua Liu Xing Bing Xue Za Zhi. 2013;34:428–32.PubMed
22.
Zurück zum Zitat Jinyong J, Xiaofang G, Yerong T, Mingdon Y, Chun W, Hongning Z. Surveillance and control of imported dengue cases in Yunnan from 2004 to 2014. Chin J Vec Biol Control. 2016;27:5–8. Jinyong J, Xiaofang G, Yerong T, Mingdon Y, Chun W, Hongning Z. Surveillance and control of imported dengue cases in Yunnan from 2004 to 2014. Chin J Vec Biol Control. 2016;27:5–8.
23.
Zurück zum Zitat Wang B, Yang H, Feng Y, Zhou H, Dai J, Hu Y, Zhang L, Wang Y, Baloch Z, Xia X. The distinct distribution and phylogenetic characteristics of dengue virus serotypes/genotypes during the 2013 outbreak in Yunnan, China phylogenetic characteristics of 2013 dengue outbreak in Yunnan, China. Infect Genet Evol. 2016;37:1–7.CrossRefPubMed Wang B, Yang H, Feng Y, Zhou H, Dai J, Hu Y, Zhang L, Wang Y, Baloch Z, Xia X. The distinct distribution and phylogenetic characteristics of dengue virus serotypes/genotypes during the 2013 outbreak in Yunnan, China phylogenetic characteristics of 2013 dengue outbreak in Yunnan, China. Infect Genet Evol. 2016;37:1–7.CrossRefPubMed
24.
Zurück zum Zitat Ministry of Health PsRoC. WS 216-2008 diagnostic criteria and principle of management of dengue fever. Beijing: China Standard Publishing House; 2008. Ministry of Health PsRoC. WS 216-2008 diagnostic criteria and principle of management of dengue fever. Beijing: China Standard Publishing House; 2008.
25.
Zurück zum Zitat Riesgo A, Farrar N, Windsor PJ, Giribet G, Leys SP. The analysis of eight transcriptomes from all poriferan classes reveals surprising genetic complexity in sponges. Mol Biol Evol. 2014;31:1102–20.CrossRefPubMed Riesgo A, Farrar N, Windsor PJ, Giribet G, Leys SP. The analysis of eight transcriptomes from all poriferan classes reveals surprising genetic complexity in sponges. Mol Biol Evol. 2014;31:1102–20.CrossRefPubMed
26.
Zurück zum Zitat Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, Lesin VM, Nikolenko SI, Pham S, Prjibelski AD, et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19:455–77.CrossRefPubMedPubMedCentral Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, Lesin VM, Nikolenko SI, Pham S, Prjibelski AD, et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19:455–77.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Larkin MA, Blackshields G, Brown NP, Chenna R, McGettigan PA, McWilliam H, Valentin F, Wallace IM, Wilm A, Lopez R, et al. Clustal W and Clustal X version 2.0. Bioinformatics. 2007;23:2947–8.CrossRefPubMed Larkin MA, Blackshields G, Brown NP, Chenna R, McGettigan PA, McWilliam H, Valentin F, Wallace IM, Wilm A, Lopez R, et al. Clustal W and Clustal X version 2.0. Bioinformatics. 2007;23:2947–8.CrossRefPubMed
29.
Zurück zum Zitat Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S. MEGA5: molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods. Mol Biol Evol. 2011;28:2731–9.CrossRefPubMedPubMedCentral Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S. MEGA5: molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods. Mol Biol Evol. 2011;28:2731–9.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Tamura K, Stecher G, Peterson D, Filipski A, Kumar S. MEGA6: molecular evolutionary genetics analysis version 6.0. Mol Biol Evol. 2013;30:2725–9.CrossRefPubMedPubMedCentral Tamura K, Stecher G, Peterson D, Filipski A, Kumar S. MEGA6: molecular evolutionary genetics analysis version 6.0. Mol Biol Evol. 2013;30:2725–9.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat NMaK S. Molecular evolution and Phylogenetics: Oxford University Press; 2000. NMaK S. Molecular evolution and Phylogenetics: Oxford University Press; 2000.
32.
Zurück zum Zitat Martin DP, Murrell B, Golden M, Khoosal A, Muhire B. RDP4: detection and analysis of recombination patterns in virus genomes. Virus Evol. 2015;1:vev003.CrossRefPubMedPubMedCentral Martin DP, Murrell B, Golden M, Khoosal A, Muhire B. RDP4: detection and analysis of recombination patterns in virus genomes. Virus Evol. 2015;1:vev003.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Posada D, Crandall KA. Evaluation of methods for detecting recombination from DNA sequences: computer simulations. Proc Natl Acad Sci U S A. 2001;98:13757–62.CrossRefPubMedPubMedCentral Posada D, Crandall KA. Evaluation of methods for detecting recombination from DNA sequences: computer simulations. Proc Natl Acad Sci U S A. 2001;98:13757–62.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Martin DP, Posada D, Crandall KA, Williamson C. A modified bootscan algorithm for automated identification of recombinant sequences and recombination breakpoints. AIDS Res Hum Retrovir. 2005;21:98–102.CrossRefPubMed Martin DP, Posada D, Crandall KA, Williamson C. A modified bootscan algorithm for automated identification of recombinant sequences and recombination breakpoints. AIDS Res Hum Retrovir. 2005;21:98–102.CrossRefPubMed
35.
36.
Zurück zum Zitat Padidam M, Sawyer S, Fauquet CM. Possible emergence of new Geminiviruses by frequent recombination. Virology. 1999;265:218–25.CrossRefPubMed Padidam M, Sawyer S, Fauquet CM. Possible emergence of new Geminiviruses by frequent recombination. Virology. 1999;265:218–25.CrossRefPubMed
37.
Zurück zum Zitat Smith JM. Analyzing the mosaic structure of genes. J Mol Evol. 1992;34:126–9.PubMed Smith JM. Analyzing the mosaic structure of genes. J Mol Evol. 1992;34:126–9.PubMed
38.
Zurück zum Zitat Gibbs MJ, Gibbs AJ. Sister-scanning: a Monte Carlo procedure for assessing signals in recombinant sequences. Bioinformatics. 2000;16:573–82.CrossRefPubMed Gibbs MJ, Gibbs AJ. Sister-scanning: a Monte Carlo procedure for assessing signals in recombinant sequences. Bioinformatics. 2000;16:573–82.CrossRefPubMed
39.
Zurück zum Zitat Pond SL, Frost SD. Datamonkey: rapid detection of selective pressure on individual sites of codon alignments. Bioinformatics. 2005;21:2531–3.CrossRefPubMed Pond SL, Frost SD. Datamonkey: rapid detection of selective pressure on individual sites of codon alignments. Bioinformatics. 2005;21:2531–3.CrossRefPubMed
40.
Zurück zum Zitat Kosakovsky Pond SL, Frost SD. Not so different after all: a comparison of methods for detecting amino acid sites under selection. Mol Biol Evol. 2005;22:1208–22.CrossRefPubMed Kosakovsky Pond SL, Frost SD. Not so different after all: a comparison of methods for detecting amino acid sites under selection. Mol Biol Evol. 2005;22:1208–22.CrossRefPubMed
41.
Zurück zum Zitat Pond SL, Frost SD, Grossman Z, Gravenor MB, Richman DD, Brown AJ. Adaptation to different human populations by HIV-1 revealed by codon-based analyses. PLoS Comput Biol. 2006;2:e62.CrossRefPubMed Pond SL, Frost SD, Grossman Z, Gravenor MB, Richman DD, Brown AJ. Adaptation to different human populations by HIV-1 revealed by codon-based analyses. PLoS Comput Biol. 2006;2:e62.CrossRefPubMed
42.
Zurück zum Zitat Murrell B, Wertheim JO, Moola S, Weighill T, Scheffler K, Kosakovsky Pond SL. Detecting individual sites subject to episodic diversifying selection. PLoS Genet. 2012;8:e1002764.CrossRefPubMedPubMedCentral Murrell B, Wertheim JO, Moola S, Weighill T, Scheffler K, Kosakovsky Pond SL. Detecting individual sites subject to episodic diversifying selection. PLoS Genet. 2012;8:e1002764.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Hu T, Zhang H, Feng Y, Fan J, Tang T, Liu Y, Zhang L, Yin X, Chen G, Li H, et al. Epidemiological and molecular characteristics of emergent dengue virus in Yunnan Province near the China-MyanmarLaos border, 2013–2015. BMC Infect Dis. 2017;17:331.CrossRefPubMedPubMedCentral Hu T, Zhang H, Feng Y, Fan J, Tang T, Liu Y, Zhang L, Yin X, Chen G, Li H, et al. Epidemiological and molecular characteristics of emergent dengue virus in Yunnan Province near the China-MyanmarLaos border, 2013–2015. BMC Infect Dis. 2017;17:331.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Liu L, Li Y, Li S, Hu N, He Y, Pong R, Lin D, Lu L, Law M. Comparison of next-generation sequencing systems. J Biomed Biotechnol. 2012;2012:251364.PubMedPubMedCentral Liu L, Li Y, Li S, Hu N, He Y, Pong R, Lin D, Lu L, Law M. Comparison of next-generation sequencing systems. J Biomed Biotechnol. 2012;2012:251364.PubMedPubMedCentral
45.
Zurück zum Zitat van Nimwegen KJM, van Soest RA, Veltman JA, Nelen MR, van der Wilt GJ, Vissers LELM, Grutters JPC. Is the $1000 genome as near as we think? A cost analysis of next-generation sequencing. Clin Chem. 2016;62:1458–64.CrossRefPubMed van Nimwegen KJM, van Soest RA, Veltman JA, Nelen MR, van der Wilt GJ, Vissers LELM, Grutters JPC. Is the $1000 genome as near as we think? A cost analysis of next-generation sequencing. Clin Chem. 2016;62:1458–64.CrossRefPubMed
46.
Zurück zum Zitat Chiu WW, Kinney RM, Dreher TW. Control of translation by the 5′- and 3′-terminal regions of the dengue virus genome. J Virol. 2005;79:8303–15.CrossRefPubMedPubMedCentral Chiu WW, Kinney RM, Dreher TW. Control of translation by the 5′- and 3′-terminal regions of the dengue virus genome. J Virol. 2005;79:8303–15.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Leardkamolkarn V, Sirigulpanit W, Kinney RM. Characterization of recombinant dengue-2 virus derived from a single nucleotide substitution in the 5′ noncoding region. J Biomed Biotechnol. 2010;2010:934694.CrossRefPubMedPubMedCentral Leardkamolkarn V, Sirigulpanit W, Kinney RM. Characterization of recombinant dengue-2 virus derived from a single nucleotide substitution in the 5′ noncoding region. J Biomed Biotechnol. 2010;2010:934694.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Bray M, Men R, Tokimatsu I, Lai C. Genetic determinants responsible for acquisition of dengue type 2 virus mouse neurovirulence. J Virol. 1998;72:1647–51.PubMedPubMedCentral Bray M, Men R, Tokimatsu I, Lai C. Genetic determinants responsible for acquisition of dengue type 2 virus mouse neurovirulence. J Virol. 1998;72:1647–51.PubMedPubMedCentral
49.
Zurück zum Zitat Gualano R, Pryor M, Cauchi M, Wright P, Davidson A. Identification of a major determinant of mouse neurovirulence of dengue virus type 2 using stably cloned genomic-length cDNA. J Gen Virol. 1998;79:437–46.CrossRefPubMed Gualano R, Pryor M, Cauchi M, Wright P, Davidson A. Identification of a major determinant of mouse neurovirulence of dengue virus type 2 using stably cloned genomic-length cDNA. J Gen Virol. 1998;79:437–46.CrossRefPubMed
50.
Zurück zum Zitat Worobey M, Rambaut A, Holmes EC. Widespread intra-serotype recombination in natural populations of dengue virus. Proc Natl Acad Sci U S A. 1999;96:7352–7.CrossRefPubMedPubMedCentral Worobey M, Rambaut A, Holmes EC. Widespread intra-serotype recombination in natural populations of dengue virus. Proc Natl Acad Sci U S A. 1999;96:7352–7.CrossRefPubMedPubMedCentral
51.
52.
Zurück zum Zitat Seoighe C, Ketwaroo F, Pillay V, Scheffler K, Wood N, Duffet R, Zvelebil M, Martinson N, McIntyre J, Morris L, Hide W. A model of directional selection applied to the evolution of drug resistance in HIV-1. Mol Biol Evol. 2007;24:1025–31.CrossRefPubMed Seoighe C, Ketwaroo F, Pillay V, Scheffler K, Wood N, Duffet R, Zvelebil M, Martinson N, McIntyre J, Morris L, Hide W. A model of directional selection applied to the evolution of drug resistance in HIV-1. Mol Biol Evol. 2007;24:1025–31.CrossRefPubMed
53.
Zurück zum Zitat Vasilakis N, Holmes EC, Fokam EB, Faye O, Diallo M, Sall AA, Weaver SC. Evolutionary processes among sylvatic dengue type 2 viruses. J Virol. 2007;81:9591–5.CrossRefPubMedPubMedCentral Vasilakis N, Holmes EC, Fokam EB, Faye O, Diallo M, Sall AA, Weaver SC. Evolutionary processes among sylvatic dengue type 2 viruses. J Virol. 2007;81:9591–5.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Dash PK, Sharma S, Soni M, Agarwal A, Parida M, Rao PVL. Complete genome sequencing and evolutionary analysis of Indian isolates of dengue virus type 2. Biochem Biophys Res Commun. 2013;436:478–85.CrossRefPubMed Dash PK, Sharma S, Soni M, Agarwal A, Parida M, Rao PVL. Complete genome sequencing and evolutionary analysis of Indian isolates of dengue virus type 2. Biochem Biophys Res Commun. 2013;436:478–85.CrossRefPubMed
Metadaten
Titel
Full-length genome and molecular characterization of dengue virus serotype 2 isolated from an imported patient from Myanmar
verfasst von
Zhaoping Zeng
Jiandong Shi
Xiaofang Guo
Ling Mo
Ningzhu Hu
Jing Sun
Meini Wu
Hongning Zhou
Yunzhang Hu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2018
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-018-1043-2

Weitere Artikel der Ausgabe 1/2018

Virology Journal 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.