Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Apr 18.
Published in final edited form as: Cell Host Microbe. 2012 Oct 18;12(4):544–557. doi: 10.1016/j.chom.2012.08.009

The TIM and TAM Families of Phosphatidylserine Receptors Mediate Dengue Virus Entry

Laurent Meertens 1,2,3,6, Xavier Carnec 1,2,3,6, Manuel Perera Lecoin 1,2,3,6, Rasika Ramdasi 1,2,3, Florence Guivel-Benhassine 4, Erin Lew 5, Greg Lemke 5, Olivier Schwartz 4, Ali Amara 1,2,3,*
PMCID: PMC3572209  NIHMSID: NIHMS442207  PMID: 23084921

SUMMARY

Dengue viruses (DVs) are responsible for the most medically relevant arboviral diseases. However, the molecular interactions mediating DV entry are poorly understood. We determined that TIM and TAM proteins, two receptor families that mediate the phosphatidylserine (PtdSer)-dependent phagocytic removal of apoptotic cells, serve as DV entry factors. Cells poorly susceptible to DV are robustly infected after ectopic expression of TIM or TAM receptors. Conversely, DV infection of susceptible cells is inhibited by anti-TIM or anti-TAM antibodies or knockdown of TIM and TAM expression. TIM receptors facilitate DV entry by directly interacting with virion-associated PtdSer. TAM-mediated infection relies on indirect DV recognition, in which the TAM ligand Gas6 acts as a bridging molecule by binding to PtdSer within the virion. This dual mode of virus recognition by TIM and TAM receptors reveals how DVs usurp the apoptotic cell clearance pathway for infectious entry.

INTRODUCTION

Dengue disease, which is caused by four dengue virus (DV) serotypes (DV1 to DV4), has emerged as a major global health problem during the last decades (Halstead, 2007; Kyle and Harris, 2008). It is estimated that 50–100 million dengue cases occur annually, with more than 2.5 billion people at risk of infection. Infection by any of the four serotypes causes disease, ranging from mild fever to life-threatening dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) (Halstead, 2007; Kyle and Harris, 2008). Despite the importance and increasing incidence of DV as a human pathogen, there is currently no licensed vaccine against DV, and the lack of antiviral drugs severely restricts therapeutic options.

DV entry into target cells is a promising target for preventive as well as therapeutic antiviral strategies since it is a major determinant of host-range, cellular tropism, and viral pathogenesis. During primary infection, DV enters host cells by clathrin-mediated endocytosis (Krishnan et al., 2007; van der Schaar et al., 2008), a process driven by the interaction between viral particles and cellular receptors. Virus-receptor complexes are then targeted to early endosomes, where the acidic environment triggers an irreversible trimerization of the viral E glycoprotein that results in fusion of the viral and cell membranes (Bressanelli et al., 2004; Modis et al., 2004) and the release of genomic RNA into the cytosol. To date, the molecular bases of DV-host interactions leading to virus entry are poorly understood and little is known about the identity of DV cellular receptor(s). DVs infect a wide range of cell types (Anderson, 2003; Balsitis et al., 2009; Couvelard et al., 1999; Jessie et al., 2004; Wu et al., 2000) and may therefore exploit multiple different receptors or use widely expressed entry molecules. Earlier studies indicated that DV virions make initial contact with the host by binding to membrane-associated heparan-sulfate proteoglycans (Chen et al., 1997). These molecules recognize positively charged residues on the surface of E protein and are thought to concentrate the virus at the target cell surface prior to its interaction with entry factors. Numerous cellular proteins such as heat shock protein 70 (HSP70), HSP90, GRP78/Bip, the lipopolysaccharide receptor CD14, or the 37/67 kDa high-affinity laminin have been proposed as putative DV entry receptors (Ansarah-Sobrinho et al., 2007; Cabrera-Hernandez et al., 2007; Jindadamrongwech et al., 2004; Thepparit and Smith, 2004). However, their function in viral entry remains obscure. To date, the known cellular receptors that promote DV infection are DC-SIGN in dendritic cells (Fernandez-Garcia et al., 2009; Lozach et al., 2005; Tassaneetrithep et al., 2003), L-SIGN in liver endothelial cells (Tassaneetrithep et al., 2003), and the mannose receptor in macrophages (Miller et al., 2008a), all of which are C-type lectins (Robinson et al., 2006) that bind glycans on the viral envelope glycoprotein E (Dejnirattisai et al., 2011; Lozach et al., 2005; Miller et al., 2008a). However, DVs also infect cells that lack these lectins, indicating that other unidentified receptor(s) should exist.

In this study, we have conducted a gain-of-function cDNA screen to identify plasma membrane proteins that enhance DV infection. We discovered that TIM and TAM proteins, two distinct families of transmembrane receptors that participate in the phosphatidylserine (PtdSer)-dependent phagocytic engulfment and removal of apoptotic cells, are DV entry factors. We found that TIM proteins bind directly to PtdSer on the surface of DV particles, while TAM proteins bind indirectly to viral PtdSer via their natural ligands Gas6 and ProS, which act as bridging molecules. These results indicate that DVs have evolved to exploit TIM and TAM receptors and the apoptotic cell clearance pathway for entry into target cells and suggest that inhibitors of these receptors may represent a promising class of antiviral compounds.

RESULTS

A cDNA Screen Identifies Cell Surface Receptors that Enhance DV2 Infection

To identify DV entry factors, we carried out a gain-of-function cDNA screen for human genes that render the poorly susceptible cell line 293T (Lozach et al., 2005) infectable by the DV2 strain JAM (DV2-JAM). This screen identified L-SIGN, confirming the validity of our approach, but also T cell immunoglobulin domain and mucin domain (TIM)-3, TYRO3, and AXL as potential DV receptors (data not shown). These belong to two distinct families of transmembrane receptors that bind directly (TIMs) or indirectly (TAMs) to phosphatidylserine (PtdSer), an “eat me” signal that promotes the engulfment of apoptotic cells (Freeman et al., 2010; Lemke and Rothlin, 2008; Ravichandran, 2011). We further characterized the role of these receptors and of PtdSer during DV infection.

TIM Receptors Enhance DV Infection

TIM-3, along with TIM-1 and TIM-4, is a cell surface glycoprotein that binds to PtdSer on the surface of apoptotic cells (Freeman et al., 2010) and are thought to be key regulators of immune tolerance (Freeman et al., 2010; Umetsu et al., 2008). To examine whether expression of TIM receptors enhances DV infection, 293T cells stably expressing TIM-1 and TIM-4 (which were not initially present in our cDNA library) or TIM-3 (Figure 1A) were generated and challenged with mosquito-derived DV2-JAM at different multiplicities of infection (moi) (Figure 1B). Parental cells were minimally infected by the virus even at high moi. TIM-3 expression resulted in a modest increase in the percentage of infected cells. Strikingly, TIM-1 or TIM-4 expression potentiated infection up to 500-fold. Of note, infection was assessed by measuring newly synthesized NS1 proteins, indicating that TIMs mediate productive DV infection. We obtained similar results using DV2 particles grown in Vero cells (Figure S1A). Titration of cell-free supernatants collected from cells challenged with DV2-JAM showed that TIM-1- and TIM-4-infected cells released large amounts of infectious particles (Figure 1C). Interestingly TIM-mediated enhancement of DV infection was also observed when HeLa cells were used as recipients (Figure S1B) and did not occur in cells expressing BAl1 (Figure S1C), a recently identified PtdSer receptor (Park et al., 2007). TIM-1 or TIM-4 also enhances infection by the three other DV serotypes (Figure 1D). The laboratory-adapted DV2 New Guinea C (NGC) and 16681 strains infected parental 293T cells, suggesting that some DV isolates may use other receptor(s) (Figure S1D). However, DV2 NGC or 16681 infection was also strongly enhanced by TIM-1 or TIM-4 (Figure S1D). To determine whether TIM receptors mediated infection by other viral species, we challenged TIM-1- and TIM-4-expressing cells with West Nile virus (WNV), yellow fever virus vaccine strain (YFV-17D), and herpes simplex virus 1 (HSV-1). Viral infection was quantified by flow cytometry using specific Ab (Figure 1E). TIM-1 and TIM-4 massively enhanced WNV infection, slightly upregulated sensitivity to YFV-17D, but had no effect on HSV-1. Together, these data indicate the PtdSer receptors TIM-1 and TIM-4, and to a lesser extent TIM-3, are cellular factors promoting flavivirus infection.

Figure 1. TIM Receptors Enhance DV Infection.

Figure 1

(A) Surface levels of TIM molecules on 293T parental (red line) and transduced cells (blue line) were evaluated by flow cytometry using polyclonal anti-TIM antibodies. Gray shading represents cell staining with a control IgG.

(B–E) Parental and 293T cells expressing TIM receptors (B) were challenged with DV2-JAM. Infection levels were assessed by flow cytometry using the anti-NS1 mAb. Parental, TIM-expressing 293T (C) were infected with DV2-JAM. Supernatants were collected 48 hr later. Virus titers were determined by plaque assay and expressed as plaque forming unit per ml. TIM receptors are used by the four DV serotypes (D). TIM receptors are shared by other flaviviruses (E). Data are represented as mean ± SEM of at least three independent experiments. See also Figure S1.

TIM Receptors Directly Interact with DV Particles and Enhance Virus Internalization

To determine whether DV virions directly bind TIM receptors, we conducted a pull-down assay with soluble TIM-Fc. We incubated DV2 particles with TIM-1-Fc or TIM-4-Fc, or as a positive control with DC-SIGN-Fc and protein G-sepharose beads. Precipitated virus was analyzed by western blotting using the anti-DV E protein mAb 4G2. DV bound to TIM-1, TIM-4, and DC-SIGN constructs and not to NKG2D-Fc or IgG1-Fc negative control constructs (Figure 2A). DV-TIM-1 interaction was also confirmed by ELISA using TIM-1-Fc coating on well plates (Figure 2B). The binding of DV to TIM-1-Fc was inhibited by anti-TIM-1 polyclonal Ab but not by control Ig (Figure S2). DV/TIM-1-Fc interaction was inhibited by EDTA (Figure S2), suggesting that it is Ca2+ dependent. Moreover, DV efficiently attached to 293T-TIM-1 and 293T-TIM-4 but not to control cells (Figure 2C). Together, these results show that TIM-1 and TIM-4 bind DV and mediate virus attachment to target cells.

Figure 2. TIM-1 and TIM-4 Molecules Bind to DV and Enhance Virus Endocytosis.

Figure 2

(A) Western blot analysis of DV2-JAM preincubated with control Fc, NKG2D-Fc, TIM-1-Fc, or TIM-4-Fc bound to protein A-agarose beads. Pulled-down virus was detected using the 4G2 anti-DV E protein mAb.

(B) Control Fc, NKG2D-Fc, or TIM-1-Fc were coated on 96-well plates and incubated with DV2-JAM for 1 hr at 4°C. Bound virus was detected using the biotinylated 4G2 mAb and HRP-conjugated streptavidin See also Figure S2.

(C) Cells transfected with an empty vector or plasmids encoding TIM-1 or TIM-4 were incubated 1 hr at 4°C with DV2-JAM. Bound virus was detected by flow cytometry using the 4G2 mAb.

(D) Cells were incubated with DV2-JAM for 2 hr at 37°C and treated with proteinase K. Total RNA was extracted from infected cells, and DV2 viral RNA level was determined by real-time quantitative PCR with human GAPDH as endogenous control. Results are expressed as the fold difference using expression in 293T infected cells as calibrator value.

(E) Parental and CHO-745 cells expressing TIM-1, TIM-4, or DC-SIGN were incubated with DV2-JAM for 1 hr at 4°C and shifted at 37°C for 45 min. Cells were labeled with DAPI (blue) and the anti-DV E protein 4G2 (green) to detect virus uptake in unpermeabilized and permeabilized cells.

(F) 293 T cells were transfected with plasmid encoding for TIM-1 WT, TIM-1 lacking its entire cytoplasmic tail (TIM-1 Δcyt), or an empty vector (pcDNA3). Cells were stained for TIM-1 expression (left panel) and challenged with DV2-JAM (right panel). Data are represented as mean ± SEM of at least three independent experiments.

We then investigated whether TIM expression enhances DV internalization. We challenged parental, TIM-1, or TIM-4-expressing 293T cells with DV2-JAM for 2 hr at 37°C. Cells were treated with proteinase K to eliminate bound virus, as previously described (Fernandez-Garcia et al., 2011). Total RNA was extracted and viral RNA levels were quantified by qPCR. TIM receptors strongly increased DV RNA uptake into 293T cells (18-fold and 10-fold enhancement with TIM-1 and TIM-4, respectively) (Figure 2D). In a second approach, we expressed TIM-1, TIM-4, and DC-SIGN into CHO-745 cells, which are a mutant CHO cell line that does not express cell surface heparan sulfate proteoglycans (Esko et al., 1985). Cells were incubated with purified DV2-JAM particles for 1 hr at 4°C and shifted to 37°C for 45 min to allow endocytosis, as previously described (Fernandez-Garcia et al., 2011). Virus uptake was monitored by fluorescence microscopy using anti-DV E mAb 4G2 in cells that were permeabilized, or not, with saponin (Figure 2E). We observed a massive intracellular accumulation of DV E protein in permeabilized cells expressing TIM-1, TIM-4, or DC-SIGN, but not in parental CHO (Figure 2E). Few virus particles were detected in cells that were not permeabilized. To study the role of TIM-1 cytoplasmic tail in enhancement of infection, we transfected 293T cells with TIM-1 WT or a TIM-1 mutant lacking its entire cytoplasmic tail (TIM-1 Δcyt). Cell surface expression levels of TIM-1Δcyt were found to be similar to WT TIM-1 (Figure 2F), however our infection studies show that TIM-1Δcyt is still able to enhance DV2-JAM infection. Thus, TIM-1 enhances DV entry via a mechanism that is independent of its cytoplasmic tail.

TIM Receptors Recognize PdtSer Associated with DV Particles

TIMs specifically recognize PtdSer exposed on the surface of apoptotic cells (Freeman et al., 2010; Kobayashi et al., 2007; Santiago et al., 2007a, 2007b). We therefore investigated whether PtdSer is associated with DV virions. For this aim, we purified DV2-JAM particles using sucrose gradient as previously described (Navarro-Sanchez et al., 2003). Purified viruses retain their ability to use TIM receptors (Figure S3). Sucrose gradient purified DV2-JAM were coated on ELISA wells and incubated with the anti-PtdSer mAb 1H6. We found that the 1H6 mAb, but not its isotype control, bound to DV-coated ELISA plates (Figure 3A). To examine if TIM-mediated DV infection is dependent on viral PtdSer, we preincubated purified DV2 with annexin V (ANX5), a PtdSer-binding protein. ANX5 inhibited infection of 293T-TIM-1 and 293T-TIM-4 but not of 293T-DC-SIGN cells (Figure 3B). Structural studies of the TIM immunoglobulin (Ig) domain previously demonstrated that PtdSer binds in a cavity built up by the CC′ and FG loops, termed the metal ion-dependent ligand-binding site (MILIBS) (Kobayashi et al., 2007; Santiago et al., 2007a, 2007b). We designed mutants of highly conserved amino acids (TIM-1 N114A or D115A, TIM-4 N121A) that line the binding cavity and are essential to PtdSer recognition (Kobayashi et al., 2007; Santiago et al., 2007a, 2007b). We found that the mutated TIM receptors were unable to mediate DV2 infection even though they were correctly expressed at the cell surface (Figure 3C). Together, these data suggest that PtdSer is associated with the surface of DV virions and is required for TIM-mediated DV infection.

Figure 3. TIM-1 and TIM-4 Molecules Recognize PtdSer Associated with DV Particles.

Figure 3

(A) Sucrose gradient purified DV2 particles were coated on well plates and incubated with the anti-PtdSer 1H6 mAb. See also Figure S3.

(B) DV2-JAM preincubated with Annexin V (ANX5; 25 μg/ml) was used to infect the indicated cells. Infection was normalized to infection without ANX5.

(C) TIM molecules mutated in the PtdSer binding domain do not mediate DV infection. Left panel: Cell surface expression of WT and TIM-1 or TIM-4 mutants. Right panel: Transfected cells were infected with DV2-JAM. Data are represented as mean ± SEM of at least three independent experiments.

TAM Ligands Bind to Viral PtdSer and Bridge DV Particles to TAM Receptors

TYRO3 and AXL belong to the TAM family, a group of three receptor protein tyrosine kinases that mediate the clearance of apoptotic cells (Lemke and Rothlin, 2008). The TAM ligands Gas6 and ProS are essential to this process (Fernández-Fernández et al., 2008; Lemke and Rothlin, 2008). Via their N-terminal Gla domain, they recognize the PtdSer expressed on apoptotic cells and bridge these cells to a TAM receptor on the surface of phagocytes (Lemke and Rothlin, 2008).

We found that 293T cells expressing high levels of TYRO3 or AXL (Figure 4A) were efficiently infected by DV2-JAM as assessed by immunofluorescence (Figure 4B) and flow cytometry using anti-NS1 Ab (Figure 4C) and released many infectious particles (Figure S4A). Ectopic expression of AXL and TYRO3 in 293T cells strongly potentiated infection by the four different DV serotypes and by WNV, but not by HSV-1 (Figures S4B and S4C). In addition, TYRO3 and AXL act at a very early stage during DV infection by enhancing virus uptake (Figure S4D). We expressed WT, kinase-dead forms of AXL (K567A mutation of the ATP-binding site) and AXL ΔCyt (complete cytoplasmic domain deletion) in 293T cells and compared their ability to mediate endocytosis and infection. Interestingly, our data indicated that tailless and kinase-dead AXL variants are unable to enhance DV infection (Figure S4E). However, these mutated receptors mediate DV internalization as efficiently as AXL WT (Figure S4F). This suggests that the AXL cytoplasmic tail and tyrosine kinase activity are essential for enhancement of DV infectivity but dispensable for virus endocytosis.

Figure 4. TYRO3 and AXL Enhance DV Infection.

Figure 4

(A) Surface levels of TAM molecules on 293T parental (red) and cells stably expressing human TYRO3 and AXL (blue). Gray: isotype control Ab.

(B and C) Cells were challenged by DV2-JAM and infection was assessed 48 hr later. Immunofluorescence (B) with an anti DV prM glycoprotein (2H2) (red) and DAPI (blue). Flow cytometry analysis (C) with an anti-NS1 mAb. See also Figure S4.

(D) Gas6 enhances DV binding to TYRO3- and AXL-expressing cells. Cells were incubated for 90 min at 4°C with DV2-JAM in serum-free medium containing Gas6 (1 μg/ml) or PBS (mock). Mean fluorescent intensity was measured by flow cytometry and normalized to that in noninfected cells.

(E) 293T cells expressing WT AXL or a molecule mutated in the Gas6-binding site (E66R/T84R) were incubated with DV2-JAM in serum-free medium containing Gas6 (1 μg/ml) or PBS (mock). After 3 hr, medium was replaced by medium supplemented with 10% FBS. The inset displays wild-type and mutant AXL surface expression.

(F) Plastic-coated DV2-JAM (107 FIU) was incubated with Gas6 or Gas6ΔGla (2 μg/ml) for 1 hr. Bound Gas6 was detected by ELISA using a goat polyclonal anti-Gas6 (10 μg/ml) Ab.

(G) Plastic-coated DV2-JAM particles (107 FIU) were incubated with the indicated Fc-chimeras (2 μg/ml) in the presence or absence of Gas6 or Gas6ΔGla (2 μg/ml). Bound Fc-chimeras were detected using an HRP-conjugated anti-human IgG.

(H) Cells were incubated with Gas6ΔGla (1 μg/ml) for 30 min before and during a 3 hr-incubation with DV2-JAM. Data are represented as mean ± SEM of at least three independent experiments. **p < 0.001, ***p < 0.0001. See also Figure S5.

We found that DV did not interact directly with TYRO3 or AXL, since we were unable to immunoprecipitate virus particles with TAM-Fc constructs (Figure S4G). However, incubation of DV particles with purified Gas6 (Figure 4D) or with fetal bovine serum (FBS) (Figure S4H), in which ProS is present at ~300 nM (Fernández-Fernández et al., 2008), potentiated virus binding to TAM-expressing cells. Thus, DV attachment to TYRO3 and AXL likely occurs only in the presence of TAM ligands. Moreover, AXL-mediated enhancement of DV infection was no longer detectable in serum-free medium, but was restored after preincubating DV particles with Gas6 (Figure 4E) or FBS (Figure S4I). Similar results were observed with TYRO3 (Figure S4J). Consistent with these findings, an AXL receptor mutated in the Gas6 binding site (AXL E66R/T84R) (Sasaki et al., 2006) did not enhance infection of DV complexed to Gas6, even though it was expressed at the cell surface (Figure 4E). To further investigate the importance of viral PtdSer in TAM-mediated enhancement of DV infection, we used Gas6ΔGla, a Gas6 variant impaired for PtdSer binding (Figure S4K). Gas6ΔGla did not interact with DV particles coated onto plate wells (Figure 4F) and failed to bridge DV virions to TYRO3-Fc and AXL-Fc (Figure 4G). Pretreatment of TAM-expressing 293T cells with Gas6ΔGla abrogated DV infection (Figure 4H). As for TIM receptors, ANX5 inhibited DV infection of 293T cells expressing TYRO3 or AXL (Figure S4L), strongly suggesting that the TAM effect is PtdSer-dependent. Together, these data support a tripartite model, whereby TAM ligands bind to PtdSer associated with DV particles and bridge virions to TAM receptors.

TIM-1 and AXL Expression Correlate with DV Susceptibility

We next investigated the relationship between TIM and TAM expression on the cell surface and the efficiency of DV infection in various cell lines. We detected high levels of TIM-1 and/or AXL expression on the surface of A549, Vero, Cos-7, and Huh7 5.1 cells (Figure 5A), all of which are readily infected by DV2-JAM (Figure 5B). 293T, U937, or RAJI cells, which are poorly susceptible to DV2-JAM (Figure 5B), lack TIM-1 and AXL (Figure 5A). To investigate whether enhanced infectivity associated with TIM and TAM expression correlate with increased virus binding, we performed binding studies using the A549 cell line that endogenously expresses TIM-1 and AXL (Figure 5A). A549 were also chosen because they express low levels of heparan sulfate proteoglycan, which has been described as an attachment receptor for DV (Chen et al., 1997) and thus may interfere with the binding studies. We found that DV2 interacts efficiently with A549 cells. Interestingly, we observed that anti-TIM1 Ab significantly inhibits DV2 binding, while anti-AXL polyclonal has only a modest effect (Figure 5C). These data suggest that TIM-1 is an important receptor that mediates DV attachment to A549 cells.

Figure 5. TIM-1 and AXL Expression and DV Infection in Cell Lines.

Figure 5

(A) Surface level of TIM-1 and AXL on cell lines was monitored by flow cytometry. Gray shading represents cell staining with a control Ab.

(B) The indicated cells were challenged with DV2-JAM.

(C) A549 cells were incubated with DV2-JAM for 1 hr at 4°C in the presence of goat polyclonal anti-TIM-1, anti-AXL, or normal goat IgG as control. Mean fluorescent intensity was measured by flow cytometry and normalized to binding in presence of control IgG. Data are shown as one representative flow cytometry analysis (left panel) and are represented as mean ± SEM of at least three independent experiments (right panel).

Endogenous TIM-1 and AXL Mediate DV Infection

We then analyzed the effects of neutralizing anti-TIM1 and/or anti-AXL Abs on DV infection of different cell lines. The Huh7 5.1 cell line expresses only TIM-1 (Figure 5A). An anti-TIM-1 Ab inhibited DV2 infection but not HSV-1 infection (Figure 6A). The A549 cell line expresses both TIM-1 and AXL (Figure 5A). DV2 infection was partly reduced with an anti-TIM-1 or anti-AXL Ab administrated alone, while the two Abs in combination fully inhibited DV2 (Figures 6B and 6C) and DV3 (Figure S5A) but not HSV-1 infection. Similar results were obtained on Vero cells, which express TIM-1 and AXL (Figure S5B). We then silenced TIM-1 or AXL expression by RNA interference in A549 cells (Figure 6D). DV infection was reduced in AXL-silenced cells and almost totally inhibited in TIM-1 silenced cells (Figure 6D), indicating an important role of TIM-1 in DV infection of A549. Notably, as for TIM- and TAM-293T-transfected cells, ANX5 blocked DV infection of A549 cells (Figure 6E). Altogether, these results show that TIM and TAM receptors may naturally cooperate to promote DV infection and that viral PtdSer mediates infection in cells endogenously expressing the receptors.

Figure 6. Endogenous TIM-1 and AXL Molecules Mediate DV Infection.

Figure 6

(A and B) Huh7.5.1 (A) and A549 (B) cells were infected with the indicated DV strains or HSV-1 in the presence of polyclonal goat anti-TIM-1, anti-AXL, or normal goat IgG as control. The levels of infected cells were normalized to infection in presence of control IgG.

(C) Representative immunofluorescence analysis of A549 infected with DV2-JAM in the presence of the indicated Ab. Green anti-prM 2H2, Blue DAPI. Scale bar: 100 μm.

(D) A549 cells were transfected by the indicated siRNA, and TIM-1 and AXL expression was assessed by flow cytometry after 2 days, at the time of infection. Cells were infected with DV2-JAM or HSV-1. Infection was normalized to infection in nontargeting (siNT) siRNA-transfected cells.

(E) A549 cells were infected with DV2-JAM or HSV-1 preincubated with different concentrations of ANX5.

(F) and (G) Human primary kidney epithelial cells (F) or astrocytes (G) were infected with DV in the presence of control IgG or an anti-AXL Ab. Insets display AXL cellular expression at the time of infection. Data are represented as mean ± SEM of at least three independent experiments. **p < 0.001, ***p < 0.0001. See also Figure S6.

Epithelial cells and astrocytes are DV targets in vivo (Balsitis et al., 2009; Ramos et al., 1998; Limon-Flores et al., 2005). Primary kidney epithelial cells and astrocytes express AXL (Figures 6F and 6G) but not TYRO3, TIM-1 or TIM-4 (not shown). DV infection was significantly reduced by an anti-AXL Ab in both cell types (Figures 6F and 6G). Silencing AXL in astrocytes also significantly decreased DV3 infection (Figure S5C). Therefore, as illustrated for AXL, the PtdSer receptors identified in our screening are involved in the infection of human primary cells, an observation that should be relevant for DV pathogenesis.

DISCUSSION

The present study adds significant insights into the molecular interactions that occur between DV and the host cell during viral entry. We show that TIM and TAM proteins, two receptor families involved in apoptotic cell recognition and clearance, mediate DV infection. TIM- and TAM-mediated enhancement of virus infection is dependent on PtdSer associated with DV particles. This supports a model by which DV use a strategy of “apoptotic mimicry” to infect target cells (Laliberte and Moss, 2009; Mercer and Helenius, 2008).

We provide strong evidence indicating that TIM and TAM receptors are cellular factors mediating DV binding and infection. First, ectopic expression of TIM and TAM rendered human cells susceptible to DV infection. Second, Ab against TIM and TAM receptors inhibited DV infection in cells naturally expressing these proteins. Third, in analyses of DV infectivity, evidence of loss-of-function was obtained using RNA interference. Finally, binding studies revealed that TIM and TAM molecules interact with DV particles and enhance virus endocytosis. However, our data suggest that TIM and TAM proteins do not directly promote DV internalization. We observed that the TIM-1 cytoplasmic tail is dispensable for DV entry, indicating that TIM-mediated enhancement of infection relies mainly on the receptor ectodomain. We propose that TIM-1 may act primarily as a tethering/attachment receptor that binds and enhances DV infection without any obligatory role in virus endocytosis. This is consistent with the proposed role of TIM receptors during the engulfment of apoptotic cells (Park et al., 2009; Toda et al., 2012). Our results also suggest that TIM-mediated enhancement of DV infection relies on still uncharacterized cellular receptors. It is possible that TIM receptors pass the virus in cis to another molecule that is ultimately responsible for virus endocytosis, as we proposed for DC-SIGN-mediated DV entry (Lozach et al., 2005). Alternatively, TIMs could also directly associate with unknown molecules to form an entry complex that coordinates virus endocytosis. Our data suggest that TIM and TAM receptors may play distinct roles during DV infection. We found that tailless and kinase-dead AXL variants are still able to enhance DV endocytosis. However, and as shown for Ebola and Lassa virus entry (Shimojima et al., 2007, 2012), both mutated AXL molecules are unable to enhance DV infection, indicating that tyrosine kinase activity is important for AXL potentiation of viral infection. These data suggest that AXL function is not restricted to enhance virus endocytosis but also to initiate signaling that facilitates infection of the target cells. Of note, Gas6 binding to TAM receptors has been shown to inhibit innate inflammatory immune responses (Lemke and Rothlin, 2008), including signaling downstream of TLRs (Rothlin et al., 2007). It is thus tempting to speculate that TAM ligation by DV during entry may initiate signal transduction events that may modulate host immune responses to potentiate a postendocytic step of the DV life cycle.

In cells expressing both TIM-1 and AXL (such as A549 and Vero), DV infection was partly reduced by anti-TIM-1 or anti-AXL Ab administrated alone and completely inhibited by the two Ab in combination. Thus, TIM-1 and AXL may cooperate to mediate optimal DV infection. TAM receptors are known to physically associate with non-TAM receptors by heterotypic dimerization (Linger et al., 2008; Rothlin et al., 2007). One can speculate that they may recruit TIM and/or additional receptors to form a viral entry complex required for DV uptake and infection (Figure 7A). DVs use the clathrin-mediated endocytosis as a major entry route for internalization (Krishnan et al., 2007; van der Schaar et al., 2008). It will be worth further assessment of the role of the clathrin-dependent pathway and other endocytic routes during TIM- and TAM-mediated enhancement of DV infection.

Figure 7. Model of PtdSer Acquisition by DV Particles and the TIM and TAM Receptor-Mediated Enhancement of DV Infection.

Figure 7

(A) Proposed model of direct and indirect (bridging) recognition of DV by the TIM and TAM receptors.

(B) Schematic representation of DV particle acquiring phosphatidylserine (PdtSer) upon budding into the ER lumen.

The role played by TIM and TAM proteins during DV pathogenesis in vivo is currently unknown. In vivo, TIM and TAM receptors are expressed in many cell types relevant to DV infection. AXL is widely expressed (Lemke and Rothlin, 2008) with prominent expression in macrophages, dendritic cells, and vascular endothelia, and we show here that DV infection of cultured epithelial cells and astrocytes is AXL-dependent. TIM-1 is abundant on Th-2 T cells, mucosal epithelial cells, and mast cells; the last have been recently identified as important DV target cells (Freeman et al., 2010; Kondratowicz et al., 2011; St John et al., 2011). TIM-4 is present exclusively on antigen-presenting cells (Kobayashi et al., 2007; Wong et al., 2010), which are sensitive to DV infection and thought to play an important role in DV pathogenesis. Our data indicate that TIM and TAM usage is not sufficient to explain DV tropism. Indeed, we have shown that DVs can infect cells lacking these proteins, suggesting that other cellular receptors that dictate virus tropism remain to be characterized. However, our results suggest that TIM and TAM receptors contribute to enhance infection of relevant target cells, which may have important consequences for virus spread.

We reveal an unexpected role for PtdSer during DV infection. Our data suggest that PtdSer is displayed on the surface of DV particles and is important for TIM- and TAM-mediated infection. In agreement with the binding of apoptotic cells to PtdSer receptors (Ravichandran, 2011; Ravichandran and Lorenz, 2007), we describe a dual mode of recognition of DV by TIM and TAM receptors. TIM molecules directly interact with DV. Our muta-genesis study of TIM strongly suggests that PtdSer associated with DV particles binds to the MILIBS of the molecule. Indirect recognition of DV is mediated by a TAM ligand such as Gas6, which recognizes through its Gla domain viral PtdSer and bridges DV virions to AXL and TYRO3. Together, these observations suggest that, by mimicking apoptotic bodies, DVs subvert the apoptotic clearance function of TIM and TAM receptors to enhance infection.

TIM and TAM proteins mediate the entry of other viruses (Feigelstock et al., 1998; Kondratowicz et al., 2011; Morizono et al., 2011; Shimojima et al., 2012, 2006). AXL and TIM-1 are receptors for Ebola virus, which, like DV, infects a broad range of cell types and causes hemorrhagic fever (Kondratowicz et al., 2011; Shimojima et al., 2006). TIM-1-mediated Ebola infection depends on a direct interaction between the viral glycoprotein GP through residues outside the MILIBS (Kondratowicz et al., 2011), indicating that DV and Ebola virus may use distinct TIM-1 regions. A recent study also identified Gas6 and ProS as “1bridging factors” that link PtdSer expressed on the viral envelope of lentiviral pseudotypes and vaccinia virus to AXL expressed on the target cell (Morizono et al., 2011). Collectively, these data suggest that TIM and TAM facilitation of viral infection may represent a general mechanism exploited by viruses that incorporate PtdSer in their membrane for optimal infection.

An interesting direction for future work is to identify how DVs acquire this lipid and how PtdSer becomes accessible to TIM and TAM receptors. The DV membrane is derived from the ER of the infected cell upon budding. The ER membrane is known to contain PtdSer in the lumenal leaflet (Leventis and Grinstein, 2010), which suggests an obvious mechanism through which PtdSer may be incorporated into DV virions (Figure 7B). However, the viral membrane does not appear to be readily exposed to receptors at the virion surface. This is shown by structural studies, which have revealed that a closed icosahedral shell of the envelope protein completely encloses the viral membrane in mature DV particles (Kuhn et al., 2002). It is plausible that TIM and TAM molecules or other yet-to-be identified receptors may display weak interactions with the E protein that trigger opening of the icosahedral shell, leading to exposure of viral membrane, as recently suggested by studies with Ab complexes (Cockburn et al., 2012). Also, recent studies have indicated that the majority of the DV particles released from infected cells are partially mature virions, which display a mixture of immature and mature surfaces (Junjhon et al., 2010; Pierson and Diamond, 2012; Plevka et al., 2011). These immature-like regions could expose membrane patches, such that PtdSer would be accessible for TIM and TAM interactions. Partially mature virions result in an incomplete cleavage of the envelope glycoprotein prM. Additional studies are thus required to evaluate whether modulation of the efficiency of the prM cleavage impacts TIM and TAM receptor usage.

EXPERIMENTAL PROCEDURES

cDNA Library Screening

We used sequence databases (SWISS-PROT, Uniprot, Human Protein Reference Database) and selected 1728 full-length cDNAs encoding plasma membrane receptors from an arrayed cDNA library cloned into the vector pCMVSPORT6 (Porcel et al., 2004). In a first round of screening, 293T cells were transfected for 4 hr with 216 pools of 8 cDNAs (1 μg each) in a 24-well plate format following Lipofectamine LTX protocol. An equal amount of a DC-SIGN cDNA dilution (diluted 1/8 in empty plasmid) was used as a positive control. Empty vector was used as a negative control. Transfected cells were challenged with the DV2-JAM primary strain (moi = 2) and infection was quantified 48 hr later by flow cytometry using the 2H2 anti-prM mAb. Pools of cDNA that rendered 293T cells positive for prM protein intracellular staining entered the second round of screening, in which single cDNAs from each positive pool were tested as described above.

Viruses and Cells

The DV-1-TVP5175 strain, DV2-JAM strain (Jamaica), DV2-New Guinea C strain, DV2-16881 strain, DV3-PAH881/88 strain (Thailand), DV4-1086 strain and WNV (Israeli IS-98-STI strain) were propagated in AP61 cell monolayers after having undergone limited cell passages. Virus titers were assessed by flow cytometry analysis (FACS) on C6/36 cells and were expressed as FACS infectious units (FIU). YFV was grown and titrated on Vero cells. HSV-1(F) was propagated and titrated on Vero cells as described elsewhere (Taddeo et al., 2004). HEK293T, CHO745 cells, Cos-7, A549, VERO, and Huh7 5.1 cells (a gift of C.Rice, New York, USA) were maintained in DMEM supplemented with 10% FBS, 1% penicillin/streptomycin. Human primary astrocytes and epithelial cells were purchased from LONZA and cultured according to the manufactured conditions.

Virus Pull-Down

DV particles (107 FIU) were incubated overnight at 4° C with 2 μg of Fc-chimera proteins in TBS, 10 mM CaCl2. BSA-saturated Protein G Sepharose beads (GE Healthcare) were added and incubated for 4 hr at 4°C. Beads were washed with TBS, 10 mM CaCl2, 0.05% Tween, and bound material was resolved in 1× Laemmli buffer in nonreducing conditions. Nitrocellulose-bound E envelope glycoprotein was detected with the 4G2 mAb and HRP-conjugated rabbit anti-mouse IgG antibody (Sigma-Aldrich).

Cell-Binding Assay

Cells (4 × 105) were incubated with DV for 90 min at 4°C in binding buffer (DMEM, NaN3 0.05%) containing either 2% BSA or 5% FBS. Cells were treated with 100 U heparin for 30 min at room temperature, before incubation with the virus. The cells were washed twice with cold binding buffer and fixed in PBS-PFA 2% at 4° C. Cell surface-absorbed DV particles were stained with the 4G2 Ab, then analyzed by flow cytometry. For bridging assays, cells were simultaneously incubated with virus and Gas6 (10 μg/ml).

ELISA Binding

For detecting direct interactions between TIM-Fc and DV, Fc-fused proteins were first coated (400 ng/well) in TBS supplemented with 10 mM CaCl2 on 96-well plates overnight at 4°C. Wells were washed with TBS 10 mM CaCl2 and saturated for 2 hr at 37°C with TBS 10 mM CaCl2, 2% BSA. DV particles (5.106 FIU/well) were added and incubated for 2 hr at 4°C. Bound particles were detected with the biotinylated 4G2 antibody (1 μg/ml) and Horseradish peroxydase (HRP)-conjugated Streptavidine (R&D systems). For Gas6 bridging experiments, DV particles were incubated with Gas6 proteins (2 μg/ml) and Fc-chimera proteins (2 μg/ml) for 1 hr at 37°C in TBS 10 mM CaCl2, 0.05% Tween. Bound Fc-chimeras were detected with HRP-conjugated rabbit anti-human IgG antibody. For Gas6 binding experiments, DV particles (107 FIU) or PtdSer (3-sn-Phosphatidyl-L-serine from bovine brain) were coated overnight at 4°C. Wells were incubated with Gas6 proteins (2 μg/ml) and extensively washed. Bound Gas6 proteins were labeled with a goat anti-Gas6 polyclonal antibody and detected with a HRP-conjugated donkey anti-goat IgG antibody. PtdSer was detected on coated DV particles (107 FIU) using anti-PtdSer 1H6 mAb (10 μg/ml) and a HRP-conjugated rabbit anti-mouse IgG antibody in PBS BSA 2%.

Immunofluorescence Assay

Cells were cultured on Lab-Tek II-CC2 Chamber Slide (Nunc, Roskilde, Denmark) and infected with DV2-JAM. Cells were fixed with PBS-PFA 4%, permeabilized with 0.05% saponin in PBS, and incubated with the anti-prM Ab 2H2. Slides were mounted with ProLong Gold antifade reagent containing 4,6-diamidino-2-phenylindole (DAPI) for nuclei staining. For internalization assay, prechilled CHO745 cells were incubated with viral particles at 4°C for 1 hr. Cells were washed with cold PBS to remove unbound particles and shifted at 37°C for 30 min. Cells were fixed with PBS-PFA 4% before immunostaining with 4G2 anti-E antibody under permeabilized and unpermeabilized conditions.

RNA Interference

Cells were transiently transfected using the Lipofectamine RNAiMax protocol (Life Technologies) with 10 nM final siRNAs as described previously. After 48 hr, cells were infected at the indicated moi, and infected cell percentages were quantified 24 hr postinfection by flow cytometry. Pools of siRNAs (ON-TARGETplus SMARTpool) used in this study were from Dharmacon: TIM-1 (L-019856-00), AXL (L-003104-00). Nontargeting negative control (NT) was used as control.

Flow Cytometry Analysis

Flow cytometry analysis was performed as previously described (Lozach et al., 2005). DV infection was detected using the anti-prM 2H2 mAb or an NS1 mAb. HSV-1 infection was detected using the anti-ICP4 mAb (clone 10F1, Santa Cruz Biotechnology).

Inhibition of Infection Assay

Cells were incubated for 30 min prior to infection with media containing the indicated quantities of goat anti-TIM and/or anti-TAM polyclonal antibodies. Identical concentrations of purified normal goat IgG were used as control. Cells were then infected for 3 hr incubation in the presence of inhibitors, washed, and incubated with culture medium. Infection was quantified by flow cytometry as indicated above.

RNA Purification, Reverse Transcription, and Real-Time qPCR

Total RNA was extracted from infected cells, using an RNeasy Mini Kit (QIAGEN, Courtaboeuf, France) with on-column DNase digestion, and stored at −20°C. cDNA was synthesized from 500 ng total isolated RNA by random priming-reverse transcription with the SuperScript VILO cDNA Synthesis Kit (Invitrogen by Life Technologies). Real-time quantitative PCR (qPCR) was performed using the Fast SYBR Green Master Mix Kit (Invitrogen by Life Technologies) on an Applied Biosystems 7500 Fast real-time PCR system (Life Technologies). The primers for viral RNA quantification were designed with PrimerExpress software V2.0 (Life Technologies) within the conserved region of the capsid gene of DV2-JAM (Forward primer 5′-TTCTCACTTGGAATGCTGCAA-3′; Reverse primer 5′-GCCACAAGGGCCATGAAC-3′) and QuantiTect primers for GAPDH were purchased from QIAGEN. Relative expression quantification was performed based on the comparative CT Method, using GAPDH as endogenous reference control.

Statistical Analyses

Graphical representation and statistical analyses were performed using Prism5 software (GraphPad Software). Unless otherwise stated, results are shown as means ± standard deviation (SD) from three independent experiments. Differences were tested for statistical significance using the paired two-tailed t test.

Supplementary Material

Supp Figures

ACKNOWLEDGMENTS

We thank Mike Diamond (Washington University School of Medicine St. Louis, USA) for generously supplying us with the anti-DV NS1 mAb, DV1, and DV4 strains; Kodi Ravichandran (University of Virginia, Charlottesville, VA, USA) for the BAl1 cDNA; Gabor Gyapay (Genoscope, Evry, France) and Olivier Allibert (CEA, Evry, France) for the cDNA collection and bioinformatic analysis. We also thank Félix Rey, Jean Claude Gluckman, Hugues de Thé, and Alessia Zamborlini, for critical reading of the manuscript and helpful discussions, and the Imagery Department of the Institut Universitaire d'Hématologie, which is supported by grants from the Conseil Regional d'Ile-de-France and the Ministère de la Recherche. A.A. is supported by the Fondation pour la Recherche Médicale, Pediatric Dengue Vaccine Initiative, Institut Universitaire d'Hématologie, Institut National de la Recherche Médicale (INSERM), and ANR DENtry. O.S. is supported by the ANR and Institut Pasteur. G.L. is supported by grants from the National Institutes of Health (R01 AI077058 and R01 AI101400). X.C., R.M.R., M.P., and E.L. are supported by fellowships from Fondation pour la Recherche Médicale, Marie Curie (EU program CARMUSYS), TOTAL Oil and Gas Venezuela and Instituto Venezolano de Investigaciones Cientificas (IVIC), and the Leukemia and Lymphoma Society, respectively.

Footnotes

SUPPLEMENTAL INFORMATION Supplemental Information includes five figures and Supplemental Experimental Procedures and can be found with this article online at http://dx.doi.org/10.1016/j.chom.2012.08.009.

REFERENCES

  1. Anderson R. Manipulation of cell surface macromolecules by flaviviruses. Adv. Virus Res. 2003;59:229–274. doi: 10.1016/S0065-3527(03)59007-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ansarah-Sobrinho C, Nelson S, Pierson TC. Flaviviruses. In: Pöhlmann S, Simmons G, editors. Viral Entry into Host Cells. Landes Bioscience; Austin, TX: 2007. pp. 1–14. [Google Scholar]
  3. Balsitis SJ, Coloma J, Castro G, Alava A, Flores D, McKerrow JH, Beatty PR, Harris E. Tropism of dengue virus in mice and humans defined by viral nonstructural protein 3-specific immunostaining. Am. J. Trop. Med. Hyg. 2009;80:416–424. [PubMed] [Google Scholar]
  4. Bressanelli S, Stiasny K, Allison SL, Stura EA, Duquerroy S, Lescar J, Heinz FX, Rey FA. Structure of a flavivirus envelope glycoprotein in its low-pH-induced membrane fusion conformation. EMBO J. 2004;23:728–738. doi: 10.1038/sj.emboj.7600064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Cabrera-Hernandez A, Thepparit C, Suksanpaisan L, Smith DR. Dengue virus entry into liver (HepG2) cells is independent of hsp90 and hsp70. J. Med. Virol. 2007;79:386–392. doi: 10.1002/jmv.20786. [DOI] [PubMed] [Google Scholar]
  6. 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;3:866–871. doi: 10.1038/nm0897-866. [DOI] [PubMed] [Google Scholar]
  7. Cockburn JJ, Navarro Sanchez ME, Fretes N, Urvoas A, Staropoli I, Kikuti CM, Coffey LL, Arenzana Seisdedos F, Bedouelle H, Rey FA. Mechanism of dengue virus broad cross-neutralization by a monoclonal antibody. Structure. 2012;20:303–314. doi: 10.1016/j.str.2012.01.001. [DOI] [PubMed] [Google Scholar]
  8. Couvelard A, Marianneau P, Bedel C, Drouet MT, Vachon F, Hénin D, Deubel V. Report of a fatal case of dengue infection with hepatitis: demonstration of dengue antigens in hepatocytes and liver apoptosis. Hum. Pathol. 1999;30:1106–1110. doi: 10.1016/s0046-8177(99)90230-7. [DOI] [PubMed] [Google Scholar]
  9. Dejnirattisai W, Webb AI, Chan V, Jumnainsong A, Davidson A, Mongkolsapaya J, Screaton G. Lectin switching during dengue virus infection. J. Infect. Dis. 2011;203:1775–1783. doi: 10.1093/infdis/jir173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Esko JD, Stewart TE, Taylor WH. Animal cell mutants defective in glycosaminoglycan biosynthesis. Proc. Natl. Acad. Sci. USA. 1985;82:3197–3201. doi: 10.1073/pnas.82.10.3197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Feigelstock D, Thompson P, Mattoo P, Zhang Y, Kaplan GG. The human homolog of HAVcr-1 codes for a hepatitis A virus cellular receptor. J. Virol. 1998;72:6621–6628. doi: 10.1128/jvi.72.8.6621-6628.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Fernández-Fernández L, Bellido-Martín L, García de Frutos P. Growth arrest-specific gene 6 (GAS6). An outline of its role in haemostasis and inflammation. Thromb. Haemost. 2008;100:604–610. doi: 10.1160/th08-04-0253. [DOI] [PubMed] [Google Scholar]
  13. Fernandez-Garcia MD, Mazzon M, Jacobs M, Amara A. Pathogenesis of flavivirus infections: using and abusing the host cell. Cell Host Microbe. 2009;5:318–328. doi: 10.1016/j.chom.2009.04.001. [DOI] [PubMed] [Google Scholar]
  14. Fernandez-Garcia MD, Meertens L, Bonazzi M, Cossart P, Arenzana-Seisdedos F, Amara A. Appraising the roles of CBLL1 and the ubiquitin/proteasome system for flavivirus entry and replication. J. Virol. 2011;85:2980–2989. doi: 10.1128/JVI.02483-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Freeman GJ, Casasnovas JM, Umetsu DT, DeKruyff RH. TIM genes: a family of cell surface phosphatidylserine receptors that regulate innate and adaptive immunity. Immunol. Rev. 2010;235:172–189. doi: 10.1111/j.0105-2896.2010.00903.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Halstead SB. Dengue. Lancet. 2007;370:1644–1652. doi: 10.1016/S0140-6736(07)61687-0. [DOI] [PubMed] [Google Scholar]
  17. Jessie K, Fong MY, Devi S, Lam SK, Wong KT. Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. J. Infect. Dis. 2004;189:1411–1418. doi: 10.1086/383043. [DOI] [PubMed] [Google Scholar]
  18. Jindadamrongwech S, Thepparit C, Smith DR. Identification of GRP 78 (BiP) as a liver cell expressed receptor element for dengue virus serotype 2. Arch. Virol. 2004;149:915–927. doi: 10.1007/s00705-003-0263-x. [DOI] [PubMed] [Google Scholar]
  19. Junjhon J, Edwards TJ, Utaipat U, Bowman VD, Holdaway HA, Zhang W, Keelapang P, Puttikhunt C, Perera R, Chipman PR, et al. Influence of pr-M cleavage on the heterogeneity of extracellular dengue virus particles. J. Virol. 2010;84:8353–8358. doi: 10.1128/JVI.00696-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Kobayashi N, Karisola P, Peña-Cruz V, Dorfman DM, Jinushi M, Umetsu SE, Butte MJ, Nagumo H, Chernova I, Zhu B, et al. TIM-1 and TIM-4 glycoproteins bind phosphatidylserine and mediate uptake of apoptotic cells. Immunity. 2007;27:927–940. doi: 10.1016/j.immuni.2007.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Kondratowicz AS, Lennemann NJ, Sinn PL, Davey RA, Hunt CL, Moller-Tank S, Meyerholz DK, Rennert P, Mullins RF, Brindley M, et al. T-cell immunoglobulin and mucin domain 1 (TIM-1) is a receptor for Zaire Ebolavirus and Lake Victoria Marburgvirus. Proc. Natl. Acad. Sci. USA. 2011;108:8426–8431. doi: 10.1073/pnas.1019030108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Krishnan MN, Sukumaran B, Pal U, Agaisse H, Murray JL, Hodge TW, Fikrig E. Rab 5 is required for the cellular entry of dengue and West Nile viruses. J. Virol. 2007;81:4881–4885. doi: 10.1128/JVI.02210-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Kuhn RJ, Zhang W, Rossmann MG, Pletnev SV, Corver J, Lenches E, Jones CT, Mukhopadhyay S, Chipman PR, Strauss EG, et al. Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell. 2002;108:717–725. doi: 10.1016/s0092-8674(02)00660-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Kyle JL, Harris E. Global spread and persistence of dengue. Annu. Rev. Microbiol. 2008;62:71–92. doi: 10.1146/annurev.micro.62.081307.163005. [DOI] [PubMed] [Google Scholar]
  25. Laliberte JP, Moss B. Appraising the apoptotic mimicry model and the role of phospholipids for poxvirus entry. Proc. Natl. Acad. Sci. USA. 2009;106:17517–17521. doi: 10.1073/pnas.0909376106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Lemke G, Rothlin CV. Immunobiology of the TAM receptors. Nat. Rev. Immunol. 2008;8:327–336. doi: 10.1038/nri2303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Leventis PA, Grinstein S. The distribution and function of phosphatidylserine in cellular membranes. Annu Rev Biophys. 2010;39:407–427. doi: 10.1146/annurev.biophys.093008.131234. [DOI] [PubMed] [Google Scholar]
  28. Limon-Flores AY, Perez-Tapia M, Estrada-Garcia I, Vaughan G, Escobar-Gutierrez A, Calderon-Amador J, Herrera-Rodriguez SE, Brizuela-Garcia A, Heras-Chavarria M, Flores-Langarica A, et al. Dengue virus inoculation to human skin explants: an effective approach to assess in situ the early infection and the effects on cutaneous dendritic cells. Int. J. Exp. Pathol. 2005;86:323–334. doi: 10.1111/j.0959-9673.2005.00445.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Linger RM, Keating AK, Earp HS, Graham DK. TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv. Cancer Res. 2008;100:35–83. doi: 10.1016/S0065-230X(08)00002-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Lozach PY, Burleigh L, Staropoli I, Navarro-Sanchez E, Harriague J, Virelizier JL, Rey FA, Desprès P, Arenzana-Seisdedos F, Amara A. Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN)-mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals. J. Biol. Chem. 2005;280:23698–23708. doi: 10.1074/jbc.M504337200. [DOI] [PubMed] [Google Scholar]
  31. Mercer J, Helenius A. Vaccinia virus uses macropinocytosis and apoptotic mimicry to enter host cells. Science. 2008;320:531–535. doi: 10.1126/science.1155164. [DOI] [PubMed] [Google Scholar]
  32. Miller JL, de Wet BJ, Martinez-Pomares L, Radcliffe CM, Dwek RA, Rudd PM, Gordon S. The mannose receptor mediates dengue virus infection of macrophages. PLoS Pathog. 2008a;4:e17. doi: 10.1371/journal.ppat.0040017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Modis Y, Ogata S, Clements D, Harrison SC. Structure of the dengue virus envelope protein after membrane fusion. Nature. 2004;427:313–319. doi: 10.1038/nature02165. [DOI] [PubMed] [Google Scholar]
  34. Morizono K, Xie Y, Olafsen T, Lee B, Dasgupta A, Wu AM, Chen IS. The soluble serum protein Gas6 bridges virion envelope phosphatidylserine to the TAM receptor tyrosine kinase Axl to mediate viral entry. Cell Host Microbe. 2011;9:286–298. doi: 10.1016/j.chom.2011.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Navarro-Sanchez E, Altmeyer R, Amara A, Schwartz O, Fieschi F, Virelizier JL, Arenzana-Seisdedos F, Desprès P. Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Rep. 2003;4:723–728. doi: 10.1038/sj.embor.embor866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Park D, Tosello-Trampont AC, Elliott MR, Lu M, Haney LB, Ma Z, Klibanov AL, Mandell JW, Ravichandran KS. BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature. 2007;450:430–434. doi: 10.1038/nature06329. [DOI] [PubMed] [Google Scholar]
  37. Park D, Hochreiter-Hufford A, Ravichandran KS. The phosphatidylserine receptor TIM-4 does not mediate direct signaling. Curr. Biol. 2009;19:346–351. doi: 10.1016/j.cub.2009.01.042. [DOI] [PubMed] [Google Scholar]
  38. Pierson TC, Diamond MS. Degrees of maturity: the complex structure and biology of flaviviruses. Curr Opin Virol. 2012;2:168–175. doi: 10.1016/j.coviro.2012.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Plevka P, Battisti AJ, Junjhon J, Winkler DC, Holdaway HA, Keelapang P, Sittisombut N, Kuhn RJ, Steven AC, Rossmann MG. Maturation of flaviviruses starts from one or more icosahedrally independent nucleation centres. EMBO Rep. 2011;12:602–606. doi: 10.1038/embor.2011.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Porcel BM, Delfour O, Castelli V, De Berardinis V, Friedlander L, Cruaud C, Ureta-Vidal A, Scarpelli C, Wincker P, Schächter V, et al. Numerous novel annotations of the human genome sequence supported by a 5′-end-enriched cDNA collection. Genome Res. 2004;14:463–471. doi: 10.1101/gr.1481104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Ramos C, Sánchez G, Pando RH, Baquera J, Hernández D, Mota J, Ramos J, Flores A, Llausás E. Dengue virus in the brain of a fatal case of hemorrhagic dengue fever. J. Neurovirol. 1998;4:465–468. doi: 10.3109/13550289809114548. [DOI] [PubMed] [Google Scholar]
  42. Ravichandran KS. Beginnings of a good apoptotic meal: the find-me and eat-me signaling pathways. Immunity. 2011;35:445–455. doi: 10.1016/j.immuni.2011.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ravichandran KS, Lorenz U. Engulfment of apoptotic cells: signals for a good meal. Nat. Rev. Immunol. 2007;7:964–974. doi: 10.1038/nri2214. [DOI] [PubMed] [Google Scholar]
  44. Robinson MJ, Sancho D, Slack EC, LeibundGut-Landmann S, Reis e Sousa C. Myeloid C-type lectins in innate immunity. Nat. Immunol. 2006;7:1258–1265. doi: 10.1038/ni1417. [DOI] [PubMed] [Google Scholar]
  45. Rothlin CV, Ghosh S, Zuniga EI, Oldstone MB, Lemke G. TAM receptors are pleiotropic inhibitors of the innate immune response. Cell. 2007;131:1124–1136. doi: 10.1016/j.cell.2007.10.034. [DOI] [PubMed] [Google Scholar]
  46. Santiago C, Ballesteros A, Martínez-Muñoz L, Mellado M, Kaplan GG, Freeman GJ, Casasnovas JM. Structures of T cell immunoglobulin mucin protein 4 show a metal-Ion-dependent ligand binding site where phosphatidylserine binds. Immunity. 2007a;27:941–951. doi: 10.1016/j.immuni.2007.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Santiago C, Ballesteros A, Tami C, Martínez-Muñoz L, Kaplan GG, Casasnovas JM. Structures of T Cell immunoglobulin mucin receptors 1 and 2 reveal mechanisms for regulation of immune responses by the TIM receptor family. Immunity. 2007b;26:299–310. doi: 10.1016/j.immuni.2007.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Sasaki T, Knyazev PG, Clout NJ, Cheburkin Y, Göhring W, Ullrich A, Timpl R, Hohenester E. Structural basis for Gas6-Axl signalling. EMBO J. 2006;25:80–87. doi: 10.1038/sj.emboj.7600912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Shimojima M, Takada A, Ebihara H, Neumann G, Fujioka K, Irimura T, Jones S, Feldmann H, Kawaoka Y. Tyro3 family-mediated cell entry of Ebola and Marburg viruses. J. Virol. 2006;80:10109–10116. doi: 10.1128/JVI.01157-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Shimojima M, Ikeda Y, Kawaoka Y. The mechanism of Axl-mediated Ebola virus infection. J. Infect. Dis. 2007;196(Suppl 2):S259–S263. doi: 10.1086/520594. [DOI] [PubMed] [Google Scholar]
  51. Shimojima M, Ströher U, Ebihara H, Feldmann H, Kawaoka Y. Identification of cell surface molecules involved in dystroglycan-independent Lassa virus cell entry. J. Virol. 2012;86:2067–2078. doi: 10.1128/JVI.06451-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. St John AL, Rathore AP, Yap H, Ng ML, Metcalfe DD, Vasudevan SG, Abraham SN. Immune surveillance by mast cells during dengue infection promotes natural killer (NK) and NKT-cell recruitment and viral clearance. Proc. Natl. Acad. Sci. USA. 2011;108:9190–9195. doi: 10.1073/pnas.1105079108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Taddeo B, Zhang W, Lakeman F, Roizman B. Cells lacking NF-kappaB or in which NF-kappaB is not activated vary with respect to ability to sustain herpes simplex virus 1 replication and are not susceptible to apoptosis induced by a replication-incompetent mutant virus. J. Virol. 2004;78:11615–11621. doi: 10.1128/JVI.78.21.11615-11621.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Tassaneetrithep B, Burgess TH, Granelli-Piperno A, Trumpfheller C, Finke J, Sun W, Eller MA, Pattanapanyasat K, Sarasombath S, Birx DL, et al. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J. Exp. Med. 2003;197:823–829. doi: 10.1084/jem.20021840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Thepparit C, Smith DR. Serotype-specific entry of dengue virus into liver cells: identification of the 37-kilodalton/67-kilodalton high-affinity laminin receptor as a dengue virus serotype 1 receptor. J. Virol. 2004;78:12647–12656. doi: 10.1128/JVI.78.22.12647-12656.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Toda S, Hanayama R, Nagata S. Two-step engulfment of apoptotic cells. Mol. Cell. Biol. 2012;32:118–125. doi: 10.1128/MCB.05993-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Umetsu DT, Umetsu SE, Freeman GJ, DeKruyff RH. TIM gene family and their role in atopic diseases. Curr. Top. Microbiol. Immunol. 2008;321:201–215. doi: 10.1007/978-3-540-75203-5_10. [DOI] [PubMed] [Google Scholar]
  58. van der Schaar HM, Rust MJ, Chen C, van der Ende-Metselaar H, Wilschut J, Zhuang X, Smit JM. Dissecting the cell entry pathway of dengue virus by single-particle tracking in living cells. PLoS Pathog. 2008;4:e1000244. doi: 10.1371/journal.ppat.1000244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Wong K, Valdez PA, Tan C, Yeh S, Hongo JA, Ouyang W. Phosphatidylserine receptor Tim-4 is essential for the maintenance of the homeostatic state of resident peritoneal macrophages. Proc. Natl. Acad. Sci. USA. 2010;107:8712–8717. doi: 10.1073/pnas.0910929107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Wu SJ, Grouard-Vogel G, Sun W, Mascola JR, Brachtel E, Putvatana R, Louder MK, Filgueira L, Marovich MA, Wong HK, et al. Human skin Langerhans cells are targets of dengue virus infection. Nat. Med. 2000;6:816–820. doi: 10.1038/77553. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp Figures

RESOURCES