Skip to main content
Erschienen in: Virology Journal 1/2019

Open Access 01.12.2019 | Research

Heparin sulfate is the attachment factor of duck Tembus virus on both BHK21 and DEF cells

verfasst von: Shaoxiong Wu, Zhen Wu, Yuanyuan Wu, Tao Wang, Mingshu Wang, Renyong Jia, Dekang Zhu, Mafeng Liu, Xinxin Zhao, Qiao Yang, Ying Wu, Shaqiu Zhang, Yunya Liu, Ling Zhang, Yanling Yu, Leichang Pan, Shun Chen, Anchun Cheng

Erschienen in: Virology Journal | Ausgabe 1/2019

Abstract

Background

Duck tembusu virus (DTMUV, genus Flaviviruses, family Flaviviridae) is an emerging flavivirus that can infect a wide range of cells and cell lines in vitro, though the initial step of virus invasion remains obscure.

Methods

In this study, drug treatments that including heparin, chondroitin sulfate, heparinase I, chondroitinase ABC and trypsin were applied to detect the influence of DTMUV absorption, subsequently, the copy number of viral genome RNA was analyzed by quantitative real-time PCR. The inhibition process of viral absorption or entry by heparin was determined by western blotting, and the cytotoxicity of drug treated cells was detected by cell counting kit-8.

Results

We found that the desulfation of glycosaminoglycans (GAGs) with sodium chlorate had a significant effect on the adsorption of DTMUV in both BHK21 and DEF cells. Based on this result, we incubated cells with a mixture of DTMUV and GAGs competition inhibitors or pre-treated cells with inhibitors, after incubation with the virus, the NS5 expression of DTMUV and viral titers were detected. The data suggested that heparin can significantly inhibit the absorption of DTMUV in a dose dependent manner but not at the step of viral entry in BHK21 and DEF cells. Meanwhile, heparinase I can significantly inhibit DTMUV attachment step.

Conclusions

Our results clearly proved that heparin sulfate plays an important role in the first step of DTMUV entry, viral attachment, in both BHK21 and DEF cells, which sheds light on the entry mechanism of DTMUV.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
DC-SIGN
Dendritic Cell -specific ICAM-3 grabbing non-integrin
DENV
Dengue Virus
DTMUV
Duck tembusu virus
GAGs
Glycosaminoglycans
GRP78
Glucose-regulated protein 78
HRP
Fluoridehorseradish peroxidase
HSPGs
Heparin sulfate proteoglycans
JEV
Japanese encephalitis virus
L-SIGN
Liver/Lymph Node-Specific ICAM-3 Grabbing Non-Integrin
PtdSer
Phosphatidylserine
PVDF
Polyvinylidene
qRT-PCR
Real-time PCR
TAM
TYRO3, AXL and MER
TCID50
Fifty percent tissue culture infectious dose
TIM
T-cell immunoglobulin and mucin domain
VOPBA
Virus overlay protein binding assay
YFV
Yellow fever virus

Background

DTMUV is an arthropod-borne flavivirus responsible for the severe decline in egg production in ducks. Since 2010, a newly emerging disease characterized by egg-drop syndrome has occurred in ducks in China. Egg production in sick ducks was seriously decreased within 2 weeks after disease onset. The infected ducks mainly showed clinical symptoms such as depression, loss of appetite, growth retardation, even paralysis or death. The disease has caused serious economic losses to the duck industry. Additionally, DTMUV can exhibit pathogenicity to Kunming mice by intracerebral inoculation [15]. DTMUV is similar to other flaviviruses, consisting of an 11-kb positive-sense single-stranded RNA genome composed of one long open-reading frame, encoding three structural proteins (capsid [C]; pre-membrane [prM], which is post-translationally cleaved to produce the pr and M proteins; and envelope [E]) and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, 2KNS4B and NS5) [68]. Flavivirus invasion into cells is a complex process involving many cellular receptors and viral components [9]. Binding to the cellular surface is the first step for flavivirus entry, which mainly depends on E protein. Moreover, prM and phosphatidylserine (PtdSer) can also recognize some cellular receptors [10]. The crystal structure revealed that flavivirus E protein structures have the following three distinct domains: the N-terminal central β-barrel-shaped domain I; the elongated finger-like domain II, which mediates the low-pH-driven membrane fusion of the viral membrane with the host endosomal membrane, and the C-terminal immunoglobulin-like domain III, which is required for binding to the cellular receptor [1114].
Flaviviruses are arboviruses that can be transmitted by mosquitoes, and most mammalian cells are susceptible to flavivirus infection. Flavivirus require two different sets of receptors to enter the cell as previously reported [15]. The first step in the flavivirus infectious cell entry pathway is concentration of the virus on the cell surface by attachment factors. Then, the virion bind to the greater affinitive entry receptor and begin to enter the cells [9]. Heparin sulfate proteoglycans (HSPGs) are involved in the adsorption of flaviviruses and comprise a core protein structure and GAGs chains. HSPGs are divided into the following three families according to their core protein structure: glypicans, syndecans and perlecans [16]. Meanwhile, sulfated GAGs, which include heparin sulfate, chondroitin sulfate and dextran sulfate, are expressed on a number of cell types and are utilized as attachment factor by some flaviviruses [1719]. Flavivirus binding to GAGs is due to the electrostatic interaction of positively charged residues on the surface of the E glycoprotein with negatively charged sulfate groups. Significantly, heparin sulfate, but not other GAGs components, is confirmed as attachment factors for some flaviviruses, such as Dengue Virus (DENV) [20], Yellow fever virus (YFV) [21] and Japanese encephalitis virus (JEV) [22].
DTMUV is a relatively recent virus and has attracted much attention. DTMUV is spread mainly through birds such as ducks and geese, but recent studies have also found that DTMUV can also infect mice [23]. To further understand the molecular mechanisms of DTMUV invasion, DEF and BHK21 cells were used for subsequent experiments. We first proved that GAGs were involved in the adsorption of DTMUV by sodium chlorate desulfated GAGs. We then indicated that heparin but not chondroitin sulfate A inhibited the absorption of DTMUV. Furthermore, we verified the results with corresponding GAG-lyases including heparinase I and chondroitinase ABC. Finally, we found that heparin only affected virus adsorption but did not affect its entry. Additionally, we briefly discussed which DTMUV structures are involved in adsorption and we found that E protein was important for the adsorption by treating DTMUV with trypsin.

Methods

Virus, cells and chemical

The DTMUV CQW1 strain was propagated in DEF cells as previously described [2] and the virus titer was 105 TCID50/0.1 ml. Baby Hamster Kidney (BHK21) cells were provided by our laboratory and maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Sigma, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco, Gaithersburg, MD, USA), penicillin 100 IU/ml and streptomycin 100 μg/ml at 37 °C in a 5% CO2 incubator. Primary duck embryo fibroblast (DEF) cells were obtained from duck embryo and cultured at 37 °C and 5% CO2 in DMEM with 10% FBS. Heparin and chondroitin sulfate A were purchase from Solarbio (Beijing, China). Heparinase I, chondroitinase ABC and sodium chlorate were purchased from Sigma (St. Louis, MO, USA). The anti-flavivirus group antigen antibody (MAB10216) was purchased from Merck (New Jersey, USA).

DTMUV binding assay

BHK21 or DEF cells were grown into monolayers in 12-well cell culture plates and then incubated with 1000TCID50 DTMUV in the absence or presence of heparin and chondroitin sulfate A at 4 °C for 1 h. The unbound DTMUV was removed by washing cells three times with PBS. Total DTMUV RNA levels were determined by qRT-PCR.

Sodium chlorate treatment

The addition of sodium chlorate can block cellular ATP-sulfurylase and sulfate adenyl transferase activities and reduce the sulfation levels of GAGs as previous described [20, 21]. BHK21 cells were cultured for 1 week in low-sulfated culture medium RPMI with 10% FBS containing the different concentration of sodium chlorate or 4 mM sodium sulfate as a supplement. For virus binding, the cells were grown into monolayers in 12-well plates that were incubated with 1000TCID50 DTMUV at 4 °C for 1 h. The unbound DTMUV was removed by washing the cells three times with PBS. Total cellular RNA was extracted from infected cells using Trizol Reagent, and DTMUV viral RNA levels were determined by quantitative real-time PCR (qRT-PCR) with mouse β-actin as the endogenous control.

GAG-lyases treatment

Heparinase I can cleave glycosidic linkages in heparin sulfate on the cellular surface and chondroitinase ABC specifically cleaves glycosidic linkages in chondroitin sulfate A, B and C [24]. BHK21 or DEF cells were grown into monolayers in 12-well cell culture plates and then incubated for 1 h at 37 °C with 300 μl buffer (20 mM Tris-HCl, pH 7.5, 50 mM NaCl, 4 mM CaCl2, and 0.01% BSA) containing heparinase I or 300 μl buffer (50 mM Trizma HCl, pH 8.0, with 60 mM sodium acetate and 0.02% BSA) containing chondroitinase ABC. The cells were washed with PBS three times and then incubated with 1000TCID50 DTMUV at 4 °C for 1 h. The unbound DTMUV was removed by washing the cells three times with PBS. Total DTMUV RNA levels were determined by qRT-PCR.

DTMUV RNA levels were determined by qRT-PCR

The cellular total RNA was extracted with the Trizol Reagent according to the manufacturer’s instructions. Then 1 μg of RNA per sample was synthesized into cDNA using a One Step TB Green™ PrimeScript™ RT-PCR Kit II (Takara, Dalian, China). Subsequently, relative mRNA levels were quantified using a SYBR Green qPCR kit (Abm, Richmond, BC, Canada) and a real-time cycler (CFX96 Bio-Rad, Hercules, CA, USA). The primers used for qRT-PCR were as described previously [25]. The reaction conditions were as follow: 94 °C for 3 min, followed by 40 cycles of 94 °C for 10 s and 63 °C for 1 min.

Fifty percent tissue culture infectious dose (TCID50) assay

Viral titers were determined using an endpoint dilution assay. Briefly, BHK21 cells in 96-well tissue culture plates were infected with serial 10-fold dilutions of DTMUV in eight replicates. The plates were incubated for 144 h at 37 °C. The viral titers were calculated using the Reed–Muench method.

Western blotting

Total cellular proteins were boiled in 6x protein loading buffer before separation by 10% SDS-PAGE electrophoresis. Then, the proteins were transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore, Bedford, MA, USA). The membranes were blocked with 5% skim milk in TBST at 37 °C for 1 h. Subsequently, The membranes were incubated with mouse anti-NS5 polyclonal antibodies provided our lab or anti-β-actin antibody (Ruiying Biological, Suzhou, China) overnight at 4 °C. Then, horseradish peroxidase (HRP)-conjugated goat anti-mouse IgG antibody was used as a secondary antibody. Finally, the proteins were visualized by chemiluminescence using an ECL kit (Bio-Rad).

Cell cytotoxicity assay by cell counting kit-8

DEF and BHK21 cells were seeded into 96-well microtiter plates. After 1 day, 100 μl of spent medium was replaced with an equal volume of fresh medium containing 10% CCK8 (MedChemExpress, New Jersey, USA). Then, the cells were incubated at 37 °C for 2 h. The optical density (OD) at 450 nm was measured with a microplate spectrophotometer and was proportional to the number of viable cells in the wells.

Statistical analysis

The unpaired Student’s t-test (GraphPad Prism software) was used to determine the statistical significance of the differences between the experimental groups. Error bars represented the standard error of the mean. The p value < 0.05 was considered statistically significant, and the degree of significance was indicated as follows: * p < 0.05, ** p < 0.01, and *** p < 0.001.

Result

Desulfation treatment on DTMUV attachment

To examine whether GAGs affect DTMUV absorption, BHK21 cells were treated with different concentrations of sodium chlorate or 4 mM sodium sulfate as a supplement. We found that the virus adsorption capacity significantly decreased, even at low concentrations such as 5 mM sodium chlorate (Fig. 1a). When BHK21 cells were treated with 4 mM sodium sulfate as a supplement, the virus adsorption capacity did not significantly decrease (Fig. 1b). GAGs desulfation had a significant effect on DTMUV adsorption, indicating that they were involved in the DTMUV adsorption process. Finally, the cells treated with sodium chlorate or sodium sulfate showed no significant cytotoxicity (Fig. 1c).

Effects of heparin and chondroitin sulfate on DTMUV attachment

To determine the DTMUV attachment factors, we tested two well-known GAGs with cell-surface related receptors for virus binding. The results suggested that incubating cells with a mixture of virus and heparin (Fig. 2a) or pre-treating cells with heparin at 4 °C for 1 h (Fig. 2b), reduced the amount of attached DTMUV in a dose-dependent manner. The decrease in the adsorbed DTMUV reached a maximum in the 500 μg/ml heparin-treated group. Compared with DEF cells, the effect of heparin on virus adsorption was more obvious on BHK21 cells. However, we found that DTMUV absorption decreased significantly when both DEF and BHK21 cells were incubated with a mixture of virus and 500 μg/ml chondroitin sulfate A (Fig. 2c). Additionally, chondroitin sulfate did not inhibited the absorption of DTMUV even after pre-treating the cells at the 500 μg/ml concentration (Fig. 2d). The results indicated that heparin, but not chondroitin sulfate A, can significantly inhibit the absorption of DTMUV. These results indirectly proved that heparin sulfate participate in the DTMUV adsorption process.

Effects of GAG-lyases on DTMUV attachment

To further confirm that heparin sulfate plays a crucial role in DTMUV attachment, monolayer BHK21 and DEF cells were pre-treated with different concentrations of heparinase I (specific for heparin sulfate) and chondroitinase ABC (specific for chondroitin sulfate ABC) at 37 °C for 1 h, and then incubated with DTMUV. The results (Fig. 3a) showed that heparinase I can significantly inhibit DTMUV binding to BHK21 cells at a concentration of 0.5 U/ml. Interestingly, the downward trend is more obvious in BHK21 cells than DEF cells. Heparinase I showed a 40% inhibitory effect on viral attachment to DEF at 2 U/ml, whereas the amount of DTMUV attachment decreased by 49% to BHK21 cells compared with the control group. In contrast, chondroitinase ABC had no significant effect on DTMUV attachment to either BHK21 or DEF cells (Fig. 3b). The results indicated that heparinase I, but not chondroitinase ABC, can significantly inhibit the absorption of DTMUV both on BHK21 and DEF cells. These results proved that heparin sulfate was involved in DTMUV attachment.

Heparin inhibits DTMUV attachment but not entry

To confirm whether heparin sulfate affects the adsorption or entry of DTMUV, DEF cells were incubated with DTMUV at 4 °C for 1 h in the presence of various concentrations of heparin. Unbound virus was removed by washing the cells with PBS, and then the cells were incubated in medium at 37 °C. At 24 h post-infection, the DTMUV in lysed cell was detected by Western-blotting using an anti-NS5 antibody (Fig. 4a) and DTMUV in the supernatants was titrated by TCID50 (Fig. 4b). The results showed that both DTMUV NS5 expression and the viral titers were decreased.
To determine the effects of heparin on DTMUV entry, DEF cells were incubated with DTMUV at 4 °C for 1 h and the unbound virus was removed by washing the cells with PBS. The cells were then incubated in culture medium with different concentrations of heparin at 37 °C for an additional 5 h. Then, the medium was replaced with fresh culture medium and the cells were incubated at 37 °C. As expected, there was no significant decrease in the NS5 protein levels or DTMUV titer compared with the control group. Collectively, these results showed that heparin only affects DTMUV adsorption and does not affect to its cellular entry.

Trypsin treatment on DTMUV attachment

Previous investigations had suggested that the DENV envelope protein was sensitive to proteases, indicating that trypsin treatment can suppress virus attachment [20]. To determine the effects of trypsin on DTMUV attachment, DTMUV was pre-treated with different concentrations of trypsin and then added to monolayer BHK21 cells. The results (Fig. 4c) showed that trypsin had no effect on DTMUV adsorption at 5 μg/ml. However, both BHK21 and DEF cells showed significant inhibition of attachment when using trypsin at 50 μg/ml concentrations. Additionally, the cells treated with trypsin had no significantly cytotoxicity (Fig. 4d). Then we directly treated the virion with trypsin, these results showed the DTMUV envelope protein can be degraded by trypsin. These indicated that the envelope protein mainly is involved in virus adsorption.

Discussion

Invading the host cell is the first step in the flavivirus infection cycle, and they can utilize multiple receptors to initiate infection [17]. Virion is first concentrated on the cell surface by binding to attachment factors, and then they are introduced into the endocytosis pathway by a large number of different types of entry receptors, which are internalized into the cell by clathrin-mediated endocytosis [10, 26]. Finally, the low-pH environment within endosomes interacts within the E glycoprotein leading to membrane fusion of the viral membrane with the endosomal membrane, subsequently promoting nucleocapsid release into the cell cytosol [17]. Flaviviruses can infect different animals and spread widely. The identification of the flavivirus receptor will allow us to further explore the mechanism of its cross-host transmission. DTMUV, as a member of flaviviruses, can be transmitted by duck and geese and infect mice by intracranial injection [4, 23]. However, the mechanism of cross-host transmission has remained obscure.
Several studies have indicated that flaviviruses build initial contacts with the host cell by binding to GAGs, such as heparin sulfate and chondroitin sulfate [10]. GAGs are long, unbranched, sulfated polysaccharides that are exposed on the cell surfaces of all tissues and are primarily involved in electrostatic interactions with concentrated virion at the cell surface [27, 28]. To determine the DTMUV attachment factors on BHK21 and DEF cells, we compared the effects of GAGs desulfation (Fig. 1), competitive inhibitors (Fig. 2), and GAG-lyses (Fig. 3) on DTMUV adsorption. These results clearly indicated that heparin sulfate but not chondroitin sulfate was involved in DTMUV adsorption on both BHK21 and DEF cells. Heparin sulfate may be required to concentrate virion at the cell surface. Additionally, heparin sulfate not only initiates the infection by some flaviviruses [16, 1921] but can also interact with other viruses, such as Sindbis virus [29], Foot-and-mouth disease virus (FMDV) [30], Herpes simplex virus (HSV) [31], Vaccinia virus [32], Venezuelan equine encephalitis virus (VEEV) [33], Papillomavirus [34] and Echovirus [35]. Additionally, Watterson found that interaction between DENV and heparin sulfate occurred through two lysine residues located in domain III of E protein by site-directed mutagenesis studies [36]. Meanwhile, studies have indicated that N- and 6-O-sulfation but not 2-O-sulfation are important for HCV infection by silencing of the enzymes involved in the heparin sulfate biosynthesis pathway [9].
Entry receptors can bind specifically to flaviviruses and direct them to the endocytic pathway after flaviviruses attach to the cell surface [10]. Recently, it has been shown that dendritic cell-specific ICAM-3 grabbing non-integrin (DC-SIGN) and liver/lymph node-specific ICAM-3 grabbing non-Integrin (L-SIGN) that belong to C-type lectins can bind to several flaviviruses, such as Dengue virus [3739], West Nile virus [40] and Japanese encephalitis virus [41]. Additionally, some investigations have also suggested that the T-cell immunoglobulin and mucin domain (TIM) and TYRO3, AXL and MER (TAM) can enhance flavivirus infection by binding to PtdSer on the virion surface rather than E glycoprotein [42, 43]. To verify the role of heparin sulfate in DTUMV invasion, we compared the effects of heparin on DTMUV adsorption and entry. The results clearly indicated that heparin only affects DTMUV adsorption and does not affect the process of virus internalization. Meanwhile, Philip Hilgard found that DENV E protein is extremely sensitive to trypsin [20]; so we treated DTMUV with trypsin and conducted DTMUV adsorption experiments. We found that trypsin could degrade E protein showing that E protein was mainly involved in virus adsorption. Despite intensive research, little is known about the identity of the cell receptors that mediate flavivirus entry and infection. Many molecules have been described as candidate receptors for flavivirus on different cell types, but their exact role in viral entry remains unclear. So far, there have been several investigations focused on determining the DTMUV entry receptor by virus overlay protein binding assay (VOPBA). Glucose-regulated protein 78 (GRP78), a receptor on BHK21 cells, and heat shock protein A9, a receptor on DF-1 cell, bind to DTMUV to allow cellular entry [44, 45]. However, this is far from enough to thoroughly understand DTMUV invasion mechanism in cells and even in animals, which requires further research.

Conclusion

Heparin sulfate, a component of GAGs exposed on the cell surface, is involved in DTMUV attachment but not entry. Moreover, heparinase I but not chondroitinase ABC can inhibit DTMUV attachment. Thus, our results may provide a molecular explanation for the pathogenesis of DTMUV.

Acknowledgments

We thank AJE Group America (www.​aje.​cn/​ac), for editing the English text of a draft of this manuscript.
This study was approved by the Animal Ethics Committee of SichuanAgricultural University (2018–19).
Not applicable.

Competing interests

The authors declare that they have no competing interests.
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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Yan P, Zhao Y, Zhang X, Xu D, Dai X, Teng Q, Yan L, Zhou J, Ji X, Zhang S, Liu G, Zhou Y, Kawaoka Y, Tong G, Li Z. An infectious disease of ducks caused by a newly emerged Tembusu virus strain in mainland China. Virology. 2011;417:1–8.CrossRefPubMed Yan P, Zhao Y, Zhang X, Xu D, Dai X, Teng Q, Yan L, Zhou J, Ji X, Zhang S, Liu G, Zhou Y, Kawaoka Y, Tong G, Li Z. An infectious disease of ducks caused by a newly emerged Tembusu virus strain in mainland China. Virology. 2011;417:1–8.CrossRefPubMed
2.
Zurück zum Zitat Zhu K, Huang J, Jia R, Zhang B, Wang M, Zhu D, Chen S, Liu M, Yin Z, Cheng A. Identification and molecular characterization of a novel duck Tembusu virus isolate from Southwest China. Arch Virol. 2015;160:2781–90.CrossRefPubMed Zhu K, Huang J, Jia R, Zhang B, Wang M, Zhu D, Chen S, Liu M, Yin Z, Cheng A. Identification and molecular characterization of a novel duck Tembusu virus isolate from Southwest China. Arch Virol. 2015;160:2781–90.CrossRefPubMed
3.
Zurück zum Zitat Yan Z, Shen H, Wang Z, Lin W, Xie Q, Bi Y, Chen F. Isolation and characterization of a novel Tembusu virus circulating in Muscovy ducks in South China. Transbound Emerg Dis. 2017;64:e15–7.CrossRefPubMed Yan Z, Shen H, Wang Z, Lin W, Xie Q, Bi Y, Chen F. Isolation and characterization of a novel Tembusu virus circulating in Muscovy ducks in South China. Transbound Emerg Dis. 2017;64:e15–7.CrossRefPubMed
4.
Zurück zum Zitat Zhang W, Chen S, Mahalingam S, Wang M, Cheng A. An updated review of avian-origin Tembusu virus: a newly emerging avian Flavivirus. J Gen Virology. 2017;98:2413–20.CrossRef Zhang W, Chen S, Mahalingam S, Wang M, Cheng A. An updated review of avian-origin Tembusu virus: a newly emerging avian Flavivirus. J Gen Virology. 2017;98:2413–20.CrossRef
5.
Zurück zum Zitat Ti J, Zhang M, Li Z, Li X, Diao Y. Duck Tembusu virus exhibits pathogenicity to Kunming mice by Intracerebral inoculation. Front Microbiol. 2016;7:190.CrossRefPubMedPubMedCentral Ti J, Zhang M, Li Z, Li X, Diao Y. Duck Tembusu virus exhibits pathogenicity to Kunming mice by Intracerebral inoculation. Front Microbiol. 2016;7:190.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Yu G, Lin Y, Tang Y, Diao Y. Evolution of Tembusu virus in ducks, chickens, geese, sparrows, and mosquitoes in northern China. Viruses. 2018;10:485–94. Yu G, Lin Y, Tang Y, Diao Y. Evolution of Tembusu virus in ducks, chickens, geese, sparrows, and mosquitoes in northern China. Viruses. 2018;10:485–94.
8.
Zurück zum Zitat Chen P, Liu J, Jiang Y, Zhao Y, Li Q, Wu L, He X, Chen H. The vaccine efficacy of recombinant duck enteritis virus expressing secreted E with or without PrM proteins of duck tembusu virus. Vaccine. 2014;32:5271–7.CrossRefPubMed Chen P, Liu J, Jiang Y, Zhao Y, Li Q, Wu L, He X, Chen H. The vaccine efficacy of recombinant duck enteritis virus expressing secreted E with or without PrM proteins of duck tembusu virus. Vaccine. 2014;32:5271–7.CrossRefPubMed
9.
Zurück zum Zitat Xu Y, Martinez P, Seron K, Luo G, Allain F, Dubuisson J, Belouzard S. Characterization of hepatitis C virus interaction with heparan sulfate proteoglycans. J Virol. 2015;89:3846–58.CrossRefPubMedPubMedCentral Xu Y, Martinez P, Seron K, Luo G, Allain F, Dubuisson J, Belouzard S. Characterization of hepatitis C virus interaction with heparan sulfate proteoglycans. J Virol. 2015;89:3846–58.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Dai L, Song J, Lu X, Deng YQ, Musyoki AM, Cheng H, Zhang Y, Yuan Y, Song H, Haywood J, Xiao H, Yan J, Shi Y, Qin CF, Qi J, Gao GF. Structures of the Zika virus envelope protein and its complex with a Flavivirus broadly protective antibody. Cell Host Microbe. 2016;19:696–704.CrossRefPubMed Dai L, Song J, Lu X, Deng YQ, Musyoki AM, Cheng H, Zhang Y, Yuan Y, Song H, Haywood J, Xiao H, Yan J, Shi Y, Qin CF, Qi J, Gao GF. Structures of the Zika virus envelope protein and its complex with a Flavivirus broadly protective antibody. Cell Host Microbe. 2016;19:696–704.CrossRefPubMed
12.
Zurück zum Zitat Pokidysheva E, Zhang Y, Battisti AJ, Bator-Kelly CM, Chipman PR, Xiao C, Gregorio GG, Hendrickson WA, Kuhn RJ, Rossmann MG. Cryo-EM reconstruction of dengue virus in complex with the carbohydrate recognition domain of DC-SIGN. Cell. 2006;124:485–93.CrossRefPubMed Pokidysheva E, Zhang Y, Battisti AJ, Bator-Kelly CM, Chipman PR, Xiao C, Gregorio GG, Hendrickson WA, Kuhn RJ, Rossmann MG. Cryo-EM reconstruction of dengue virus in complex with the carbohydrate recognition domain of DC-SIGN. Cell. 2006;124:485–93.CrossRefPubMed
13.
Zurück zum Zitat Das S, Laxminarayana SV, Chandra N, Ravi V, Desai A. Heat shock protein 70 on Neuro2a cells is a putative receptor for Japanese encephalitis virus. Virology. 2009;385:47–57.CrossRefPubMed Das S, Laxminarayana SV, Chandra N, Ravi V, Desai A. Heat shock protein 70 on Neuro2a cells is a putative receptor for Japanese encephalitis virus. Virology. 2009;385:47–57.CrossRefPubMed
14.
Zurück zum Zitat Rodenhuis-Zybert IA, Moesker B, da Silva Voorham JM, van der Ende-Metselaar H, Diamond MS, Wilschut J, Smit JM. A fusion-loop antibody enhances the infectious properties of immature flavivirus particles. J Virol. 2011;85:11800–8.CrossRefPubMedPubMedCentral Rodenhuis-Zybert IA, Moesker B, da Silva Voorham JM, van der Ende-Metselaar H, Diamond MS, Wilschut J, Smit JM. A fusion-loop antibody enhances the infectious properties of immature flavivirus particles. J Virol. 2011;85:11800–8.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Rey FA, Stiasny K, Heinz FX. Flavivirus structural heterogeneity: implications for cell entry. Current Opinion Virology. 2017;24:132–9.CrossRef Rey FA, Stiasny K, Heinz FX. Flavivirus structural heterogeneity: implications for cell entry. Current Opinion Virology. 2017;24:132–9.CrossRef
16.
Zurück zum Zitat Lin X. Functions of heparan sulfate proteoglycans in cell signaling during development. Development. 2004;131:6009–21.CrossRefPubMed Lin X. Functions of heparan sulfate proteoglycans in cell signaling during development. Development. 2004;131:6009–21.CrossRefPubMed
18.
Zurück zum Zitat Nakamura M, Uehara Y, Asada M, Suzuki M, Imamura T. Sulfated glycosaminoglycan-assisted receptor specificity of human fibroblast growth factor (FGF) 19 signaling in a mouse system is different from that in a human system. J Biomol Screen. 2013;18:321–30.CrossRefPubMed Nakamura M, Uehara Y, Asada M, Suzuki M, Imamura T. Sulfated glycosaminoglycan-assisted receptor specificity of human fibroblast growth factor (FGF) 19 signaling in a mouse system is different from that in a human system. J Biomol Screen. 2013;18:321–30.CrossRefPubMed
19.
Zurück zum Zitat Su CM, Liao CL, Lee YL, Lin YL. Highly sulfated forms of heparin sulfate are involved in japanese encephalitis virus infection. Virology. 2001;286:206–15.CrossRefPubMed Su CM, Liao CL, Lee YL, Lin YL. Highly sulfated forms of heparin sulfate are involved in japanese encephalitis virus infection. Virology. 2001;286:206–15.CrossRefPubMed
20.
Zurück zum Zitat Hilgard P, Stockert R. Heparan sulfate proteoglycans initiate dengue virus infection of hepatocytes. Hepatology. 2000;32:1069–77.CrossRefPubMed Hilgard P, Stockert R. Heparan sulfate proteoglycans initiate dengue virus infection of hepatocytes. Hepatology. 2000;32:1069–77.CrossRefPubMed
21.
Zurück zum Zitat Germi R, Crance JM, Garin D, Guimet J, Lortat-Jacob H, Ruigrok RW, Zarski JP, Drouet E. Heparan sulfate-mediated binding of infectious dengue virus type 2 and yellow fever virus. Virology. 2002;292:162–8.CrossRefPubMed Germi R, Crance JM, Garin D, Guimet J, Lortat-Jacob H, Ruigrok RW, Zarski JP, Drouet E. Heparan sulfate-mediated binding of infectious dengue virus type 2 and yellow fever virus. Virology. 2002;292:162–8.CrossRefPubMed
22.
Zurück zum Zitat Liu H, Chiou SS, Chen WJ. Differential binding efficiency between the envelope protein of Japanese encephalitis virus variants and heparan sulfate on the cell surface. J Med Virol. 2004;72:618–24.CrossRefPubMed Liu H, Chiou SS, Chen WJ. Differential binding efficiency between the envelope protein of Japanese encephalitis virus variants and heparan sulfate on the cell surface. J Med Virol. 2004;72:618–24.CrossRefPubMed
23.
24.
Zurück zum Zitat Misinzo G, Delputte PL, Meerts P, Lefebvre DJ, Nauwynck HJ. Porcine circovirus 2 uses heparan sulfate and chondroitin sulfate B glycosaminoglycans as receptors for its attachment to host cells. J Virol. 2006;80:3487–94.CrossRefPubMedPubMedCentral Misinzo G, Delputte PL, Meerts P, Lefebvre DJ, Nauwynck HJ. Porcine circovirus 2 uses heparan sulfate and chondroitin sulfate B glycosaminoglycans as receptors for its attachment to host cells. J Virol. 2006;80:3487–94.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Chen S, Wang T, Liu P, Yang C, Wang M, Jia R, Zhu D, Liu M, Yang Q, Wu Y. Duck interferon regulatory factor 7 (IRF7) can control duck Tembusu virus (DTMUV) infection by triggering type I interferon production and its signal transduction pathway. Cytokine. 2019;113:31–8.CrossRefPubMed Chen S, Wang T, Liu P, Yang C, Wang M, Jia R, Zhu D, Liu M, Yang Q, Wu Y. Duck interferon regulatory factor 7 (IRF7) can control duck Tembusu virus (DTMUV) infection by triggering type I interferon production and its signal transduction pathway. Cytokine. 2019;113:31–8.CrossRefPubMed
26.
Zurück zum Zitat Hackett BA, Cherry S. Flavivirus internalization is regulated by a size-dependent endocytic pathway. Proc Natl Acad Sci U S A. 2018;115:4246–51.CrossRefPubMedPubMedCentral Hackett BA, Cherry S. Flavivirus internalization is regulated by a size-dependent endocytic pathway. Proc Natl Acad Sci U S A. 2018;115:4246–51.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Zhang L. Glycosaminoglycan (GAG) biosynthesis and GAG-binding proteins. Progress Mol Biol Translational Sci. 2010;93:1.CrossRef Zhang L. Glycosaminoglycan (GAG) biosynthesis and GAG-binding proteins. Progress Mol Biol Translational Sci. 2010;93:1.CrossRef
28.
Zurück zum Zitat Chen Y, Maguire T, Hileman RE, Fromm JR, Esko JD, Linhardt RJ, Marks RM. Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nat Med. 1997;3:866–71.CrossRefPubMed Chen Y, Maguire T, Hileman RE, Fromm JR, Esko JD, Linhardt RJ, Marks RM. Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nat Med. 1997;3:866–71.CrossRefPubMed
29.
30.
Zurück zum Zitat Sa-Carvalho D, Rieder E, Baxt B, Rodarte R, Tanuri A, Mason PW. Tissue culture adaptation of foot-and-mouth disease virus selects viruses that bind to heparin and are attenuated in cattle. J Virol. 1997;71:5115–23.PubMedPubMedCentral Sa-Carvalho D, Rieder E, Baxt B, Rodarte R, Tanuri A, Mason PW. Tissue culture adaptation of foot-and-mouth disease virus selects viruses that bind to heparin and are attenuated in cattle. J Virol. 1997;71:5115–23.PubMedPubMedCentral
31.
Zurück zum Zitat Shukla D, Liu J, Blaiklock P, Shworak NW, Bai X, Esko JD, Cohen GH, Eisenberg RJ, Rosenberg RD, Spear PG. A novel role for 3-O-sulfated heparan sulfate in herpes simplex virus 1 entry. Cell. 1999;99:13.CrossRefPubMed Shukla D, Liu J, Blaiklock P, Shworak NW, Bai X, Esko JD, Cohen GH, Eisenberg RJ, Rosenberg RD, Spear PG. A novel role for 3-O-sulfated heparan sulfate in herpes simplex virus 1 entry. Cell. 1999;99:13.CrossRefPubMed
32.
Zurück zum Zitat Lin CL, Chung CS, Heine HG, Chang W. Vaccinia virus envelope H3L protein binds to cell surface heparan sulfate and is important for intracellular mature virion morphogenesis and virus infection in vitro and in vivo. J Virol. 2000;74:3353–65.CrossRefPubMedPubMedCentral Lin CL, Chung CS, Heine HG, Chang W. Vaccinia virus envelope H3L protein binds to cell surface heparan sulfate and is important for intracellular mature virion morphogenesis and virus infection in vitro and in vivo. J Virol. 2000;74:3353–65.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Bernard KA, Klimstra WB, Johnston RE. Mutations in the E2 glycoprotein of Venezuelan equine encephalitis virus confer heparan sulfate interaction, low morbidity, and rapid clearance from blood of mice. Virology. 2000;276:93–103.CrossRefPubMed Bernard KA, Klimstra WB, Johnston RE. Mutations in the E2 glycoprotein of Venezuelan equine encephalitis virus confer heparan sulfate interaction, low morbidity, and rapid clearance from blood of mice. Virology. 2000;276:93–103.CrossRefPubMed
34.
Zurück zum Zitat Joyce JG, Tung JS, Przysiecki CT, Cook JC, Lehman ED, Sands JA, Jansen KU, Keller PM. The L1 major capsid protein of human papillomavirus type 11 recombinant virus-like particles interacts with heparin and cell-surface glycosaminoglycans on human keratinocytes. J Biol Chem. 1999;274:5810–22.CrossRefPubMed Joyce JG, Tung JS, Przysiecki CT, Cook JC, Lehman ED, Sands JA, Jansen KU, Keller PM. The L1 major capsid protein of human papillomavirus type 11 recombinant virus-like particles interacts with heparin and cell-surface glycosaminoglycans on human keratinocytes. J Biol Chem. 1999;274:5810–22.CrossRefPubMed
36.
Zurück zum Zitat Watterson D, Kobe B, Young PR. Residues in domain III of the dengue virus envelope glycoprotein involved in cell-surface glycosaminoglycan binding. J General Virology. 2012;93:72–82.CrossRef Watterson D, Kobe B, Young PR. Residues in domain III of the dengue virus envelope glycoprotein involved in cell-surface glycosaminoglycan binding. J General Virology. 2012;93:72–82.CrossRef
37.
Zurück zum Zitat Alen MMF, Kaptein SJF, Burghgraeve TD, Balzarini J, Neyts J, Schols D. Antiviral activity of carbohydrate-binding agents and the role of DC-SIGN in dengue virus infection. Virology. 2009;387:67–75.CrossRefPubMed Alen MMF, Kaptein SJF, Burghgraeve TD, Balzarini J, Neyts J, Schols D. Antiviral activity of carbohydrate-binding agents and the role of DC-SIGN in dengue virus infection. Virology. 2009;387:67–75.CrossRefPubMed
38.
Zurück zum Zitat Boonrat T, Timothy HB, Angela G-P, Christine T, Jennifer F, Wellington S, Michael AE, Kovit P, Suttipant S, Deborah LB. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med. 2003;197:823–9.CrossRef Boonrat T, Timothy HB, Angela G-P, Christine T, Jennifer F, Wellington S, Michael AE, Kovit P, Suttipant S, Deborah LB. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med. 2003;197:823–9.CrossRef
39.
Zurück zum Zitat Chen ST, Lin YL, Huang MT, Wu MF, Cheng SC, Lei HY, Lee CK, Chiou TW, Wong CH, Hsieh SL. CLEC5A is critical for dengue-virus-induced lethal disease. Nature. 2008;453:672–6.CrossRefPubMed Chen ST, Lin YL, Huang MT, Wu MF, Cheng SC, Lei HY, Lee CK, Chiou TW, Wong CH, Hsieh SL. CLEC5A is critical for dengue-virus-induced lethal disease. Nature. 2008;453:672–6.CrossRefPubMed
40.
Zurück zum Zitat Davis CW, Hai-Yen N, Hanna SL, Sánchez MD, Doms RW, Pierson TC. West Nile virus discriminates between DC-SIGN and DC-SIGNR for cellular attachment and infection. J Virol. 2006;80:1290.CrossRefPubMedPubMedCentral Davis CW, Hai-Yen N, Hanna SL, Sánchez MD, Doms RW, Pierson TC. West Nile virus discriminates between DC-SIGN and DC-SIGNR for cellular attachment and infection. J Virol. 2006;80:1290.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Wang P, Hu K, Luo S, Zhang M, Deng X, Li C, Jin W, Hu B, He S, Li M. DC-SIGN as an attachment factor mediates Japanese encephalitis virus infection of human dendritic cells via interaction with a single high-mannose residue of viral E glycoprotein. Virology. 2016;488:108–19.CrossRefPubMed Wang P, Hu K, Luo S, Zhang M, Deng X, Li C, Jin W, Hu B, He S, Li M. DC-SIGN as an attachment factor mediates Japanese encephalitis virus infection of human dendritic cells via interaction with a single high-mannose residue of viral E glycoprotein. Virology. 2016;488:108–19.CrossRefPubMed
42.
Zurück zum Zitat Meertens L, Carnec X, Lecoin MP, Ramdasi R, Guivel-Benhassine F, Lew E, Lemke G, Schwartz O, Amara A. The TIM and TAM families of Phosphatidylserine receptors mediate dengue virus entry. Cell Host Microbe. 2012;12:544–57.CrossRefPubMedPubMedCentral Meertens L, Carnec X, Lecoin MP, Ramdasi R, Guivel-Benhassine F, Lew E, Lemke G, Schwartz O, Amara A. The TIM and TAM families of Phosphatidylserine receptors mediate dengue virus entry. Cell Host Microbe. 2012;12:544–57.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Kuadkitkan A, Wikan N, Fongsaran C, Smith DR. Identification and characterization of prohibitin as a receptor protein mediating DENV-2 entry into insect cells. Virology. 2010;406:149–61.CrossRefPubMed Kuadkitkan A, Wikan N, Fongsaran C, Smith DR. Identification and characterization of prohibitin as a receptor protein mediating DENV-2 entry into insect cells. Virology. 2010;406:149–61.CrossRefPubMed
44.
Zurück zum Zitat Zhao D, Liu Q, Han K, Wang H, Yang J, Bi K, Liu Y, Liu N, Tian Y, Li Y. Identification of glucose-regulated protein 78 (GRP78) as a receptor in BHK-21 cells for duck Tembusu virus infection. Front Microbiol. 2018;9:694.CrossRefPubMedPubMedCentral Zhao D, Liu Q, Han K, Wang H, Yang J, Bi K, Liu Y, Liu N, Tian Y, Li Y. Identification of glucose-regulated protein 78 (GRP78) as a receptor in BHK-21 cells for duck Tembusu virus infection. Front Microbiol. 2018;9:694.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Liu Q, Huang X, Zhao D, Han K, Liu Y, Yang J, Bi K, Li Y. Identification of heat shock protein A9 as a Tembusu virus binding protein on DF-1 cells. Virus Res. 2017;227:110–4.CrossRefPubMed Liu Q, Huang X, Zhao D, Han K, Liu Y, Yang J, Bi K, Li Y. Identification of heat shock protein A9 as a Tembusu virus binding protein on DF-1 cells. Virus Res. 2017;227:110–4.CrossRefPubMed
Metadaten
Titel
Heparin sulfate is the attachment factor of duck Tembus virus on both BHK21 and DEF cells
verfasst von
Shaoxiong Wu
Zhen Wu
Yuanyuan Wu
Tao Wang
Mingshu Wang
Renyong Jia
Dekang Zhu
Mafeng Liu
Xinxin Zhao
Qiao Yang
Ying Wu
Shaqiu Zhang
Yunya Liu
Ling Zhang
Yanling Yu
Leichang Pan
Shun Chen
Anchun Cheng
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2019
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-019-1246-1

Weitere Artikel der Ausgabe 1/2019

Virology Journal 1/2019 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.