40
1 Timing of galectin-1 exposure differentially modulates Nipah virus entry and syncytia formation in 1 endothelial cells 2 3 Running Title: Nipah virus infection regulated by galectin-1 4 5 Omai B. Garner 1 , Tatyana Yun 2 , Olivier Pernet 3 , Hector C. Aguilar 4 , Arnold Park 3 , Thomas A. 6 Bowden 5 , Alexander N. Freiberg 2 , Benhur Lee 3,6* , Linda G. Baum 1* 7 8 1 Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los 9 Angeles, CA, USA 10 2 Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA 11 3 Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of 12 Medicine at UCLA, Los Angeles, CA, USA 13 4 Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State 14 University, Pullman, WA, USA 15 5 Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, 16 Roosevelt Drive, Oxford OX3 7BN, United Kingdom 17 6 Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA 18 *These authors contributed equally 19 Corresponding authors: [email protected] and [email protected] 20 21 JVI Accepts, published online ahead of print on 10 December 2014 J. Virol. doi:10.1128/JVI.02435-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved. on April 5, 2018 by guest http://jvi.asm.org/ Downloaded from

Timing of galectin-1 exposure differentially modulates Nipah virus

  • Upload
    buiminh

  • View
    226

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Timing of galectin-1 exposure differentially modulates Nipah virus

  1

Timing of galectin-1 exposure differentially modulates Nipah virus entry and syncytia formation in 1 

endothelial cells 2 

Running Title: Nipah virus infection regulated by galectin-1 4 

Omai B. Garner1, Tatyana Yun2, Olivier Pernet3, Hector C. Aguilar4, Arnold Park3, Thomas A. 6 

Bowden5, Alexander N. Freiberg2, Benhur Lee3,6*, Linda G. Baum1* 7 

1Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los 9 

Angeles, CA, USA 10 

2Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA 11 

3Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of 12 

Medicine at UCLA, Los Angeles, CA, USA 13 

4Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State 14 

University, Pullman, WA, USA 15 

5Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, 16 

Roosevelt Drive, Oxford OX3 7BN, United Kingdom 17 

6Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA 18 

*These authors contributed equally 19 

Corresponding authors: [email protected] and [email protected] 20 

21 

JVI Accepts, published online ahead of print on 10 December 2014J. Virol. doi:10.1128/JVI.02435-14Copyright © 2014, American Society for Microbiology. All Rights Reserved.

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 2: Timing of galectin-1 exposure differentially modulates Nipah virus

  2

Abstract 22 

Nipah virus (NiV) is a deadly emerging enveloped paramyxovirus that primarily targets human 23 

endothelial cells. Endothelial cells express the innate immune effector galectin-1 that we have 24 

previously shown can bind to specific N-glycans on the NiV envelope fusion glycoprotein (F). NiV-F 25 

mediates fusion of infected endothelial cells into syncytia, resulting in endothelial disruption and 26 

hemorrhage. Galectin-1 is an endogenous carbohydrate binding protein that binds to specific glycans 27 

on NiV-F to reduce endothelial cell fusion, an effect that may reduce pathophysiologic sequelae of 28 

NiV infection. However, galectins play multiple roles in regulating host-pathogen interactions; for 29 

example, galectins can promote attachment of HIV to T cells and macrophages and attachment of 30 

HSV-1 to keratinocytes, but can also inhibit influenza entry into airway epithelial cells. Using live 31 

Nipah virus, in the present study, we demonstrate that galectin-1 can enhance NiV attachment to and 32 

infection of primary human endothelial cells by bridging glycans on the viral envelope to host cell 33 

glycoproteins. In order to exhibit an enhancing effect, galectin-1 must be present during the initial 34 

phase of virus attachment; in contrast, addition of galectin-1 post-infection results in reduced 35 

production of progeny virus and syncytia formation. Thus, galectin-1 can have dual and opposing 36 

effects on NiV infection of human endothelial cells. While various roles for galectin family members 37 

in microbial-host interactions have been described, we report opposing effects of the same galectin 38 

family member on a specific virus, with the timing of exposure during the viral life cycle determining 39 

the outcome. 40 

41 

Importance 42 

Nipah virus is an emerging pathogen that targets endothelial cells lining blood vessels; the high 43 

mortality rate (up to 70%) in Nipah virus infections results from destruction of these cells and resulting 44 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 3: Timing of galectin-1 exposure differentially modulates Nipah virus

  3

catastrophic hemorrhage. Host factors that promote or prevent Nipah virus infection are not well 45 

understood. Endogenous human lectins, such as galectin-1, can function as pattern recognition 46 

receptors to reduce infection and initiate immune responses; however, lectins can also be exploited by 47 

microbes to enhance infection of host cells. We found that galectin-1, which is made by inflamed 48 

endothelial cells, can both promote Nipah virus infection of endothelial cells by “bridging” the virus to 49 

the cell, as well as reduce production of progeny virus and reduce endothelial cell fusion and damage, 50 

depending on timing of galectin-1 exposure. This is the first report of spatiotemporal opposing effects 51 

of a host lectin for a virus in one type of host cell. 52 

53 

54 

55 

56 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 4: Timing of galectin-1 exposure differentially modulates Nipah virus

  4

Introduction 57 

Nipah virus (NiV) is an emerging zoonotic paramyxovirus that targets endothelial and neural 58 

cells. Infection with NiV can result in a severe encephalitic or respiratory syndrome with case fatality 59 

rates ranging from 40-100% in humans [1, 2]. While initial outbreaks involved transmission from bats-60 

to-pigs, and then from pigs-to-humans, more recent outbreaks have been shown to involve human-to-61 

human virus transmission [2]. There are currently no approved vaccines or anti-viral agents targeting 62 

NiV for human cases, and quarantine has been the predominant measure to restrain the spread of the 63 

virus[3]. 64 

NiV preferentially infects microvasculature endothelial cells that express the entry receptor 65 

ephrinB2 [4]. The NiV attachment protein NiV-G binds to ephrinB2 or ephrinB3 [5]on the host cell 66 

plasma membrane, triggering the NiV fusion protein, NiV-F, to execute fusion of the viral envelope 67 

with the host cell membrane. Infected microvascular endothelial cells produce NiV-F and NiV-G 68 

which results in fusion of the cells into syncytia[6]; this syncytia formation contributes to vascular 69 

compromise and hemorrhage, two hallmarks of Nipah virus infection [7]. 70 

Endothelial cells respond to viral infection by producing inflammatory mediators including 71 

cytokines, which regulate leukocyte trafficking into adjacent tissues, and by presenting viral antigens 72 

[8]. Among the inflammatory mediators produced by endothelial cells are the galectins, a family of 73 

mammalian lectins implicated in immune regulation. Vascular endothelial cells express several 74 

galectin family members, including galectins-1 and -9. For example, infection of vascular endothelial 75 

cells with Ebola virus results in endothelial cell activation [9] and increased expression of galectin-1 76 

[10, 11]. Infection of endothelial cells with Dengue virus induces expression of galectin-9 [12], as does 77 

binding of double stranded RNA to TLR-3 on endothelial cells [13]. 78 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 5: Timing of galectin-1 exposure differentially modulates Nipah virus

  5

Galectins play several roles in modulating host-pathogen interactions. Galectins can participate 79 

in innate immune recognition and clearance of pathogens, however pathogens can also use galectins as 80 

attachment receptors. Galectins specifically recognize a subset of microbes based on binding to 81 

pathogen specific polysaccharides [14]. For example, galectin-4 and galectin-8 bind to and directly kill 82 

human blood-group B expressing Escherichia coli [15] and galectin-3 binds to and directly kills 83 

Candida albicans [16] in the absence of immunoglobulin or complement. In contrast, Pseudomonas 84 

aeruginosa and Trypanosoma cruzi use galectin-3 to bind to and infect human cells, and galectin-1 on 85 

human cervical epithelial cells is an attachment factor for Trichomonas vaginalis [17-19]. Similarly, 86 

galectin-1, which preferentially recognizes LacNAc (Gal1,4GlcNAc) containing glycans, increases 87 

human immunodeficiency virus (HIV) infection of monocyte-derived macrophages through 88 

stabilization of virus attachment [20-22] and galectin-9 promotes HIV infection of T cells by 89 

regulating the cell surface redox status [23]. Thus, different galectins can have pleiotropic and 90 

sometimes opposing roles in the etiology and pathophysiology of microbial infections. However, 91 

opposing effects of a single galectin on a specific virus have not been reported. 92 

Using NiV-F/G pseudotyped particles, our labs previously demonstrated that galectin-1 93 

produced by endothelial cells can inhibit endothelial cell syncytia formation mediated by NiV-G and 94 

NiV-F [11, 24]. In these studies, galectin-1 reduced NiV-F mediated fusion of endothelial cells by 95 

interfering with NiV-F maturation, reducing lateral mobility of NiV-F on the plasma membrane, and 96 

inhibiting the conformational change in NiV-F triggered by receptor binding to NiV-G that is 97 

necessary for cell-cell fusion [11]. While galectin-1 produced by endothelial cells can clearly inhibit 98 

post-infection cell-cell fusion triggered by NiV-F, our prior studies did not address the effect of 99 

galectin-1 on NiV attachment and entry into host cells. In the present study, we found that galectin-1 100 

can enhance live NiV attachment to and infection of endothelial cells by bridging glycans on the viral 101 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 6: Timing of galectin-1 exposure differentially modulates Nipah virus

  6

envelope to cell surface glycoproteins. While the presence of galectin-1 enhances NiV entry, galectin-1 102 

also inhibits syncytia formation of infected cells. In both cases, a specific N-linked glycan site on NiV-103 

F plays a critical role in mediating galectin-1’s effects. Our results highlight the complex roles 104 

galectin-1 may play in viral pathogenesis. 105 

106 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 7: Timing of galectin-1 exposure differentially modulates Nipah virus

  7

Materials and Methods 107 

Cells, virus, and reagents 108 

Vero cells (ATCC) were maintained in MEM alpha (Invitrogen) with 10% FBS (Hyclone) and 109 

2mM Glutamax in 5%CO2 at 37C. 293T cells were maintained in DMEM (Invitrogen) with 10% FBS 110 

(Hyclone) and 2mM Glutamax. VeroCCL-81 cells were maintained in DMEM (Gibco) with 10% FBS 111 

(Hyclone). HUVECs (BD Biosciences) were maintained in MDCB-131 complete media with fetal 112 

bovine serum and antibiotics (VEC Technologies, INC). HUVECs (LONZA) were maintained in 113 

Endothelial Basal Medium-2 (EGM-2) with EGM-2 SingleQuots. 114 

Recombinant human galectin-1 was expressed in E. coli and purified by affinity 115 

chromatography on lactosyl-Sepharose, as described [25]. Galectin-1 has unpaired thiols in the 116 

carbohydrate recognition domain that can form disulfide bonds and inactivate the lectin, so galectin-1 117 

is prepared and stored in PBS with dithiothreitol (DTT) and, in all assays, buffer controls are the stock 118 

PBS with 1.2mM DTT at the appropriate dilution [26]. 119 

NiV-F and G were pseudotyped onto a reporter vesicular stomatitis virus (VSV) expressing 120 

Renilla luciferase as in [27]. Nipah virus (NiV) strain Malaysia (NiV-MAL) was kindly provided by the 121 

Special Pathogens Branch, CDC, Atlanta, GA, and propagated in VeroE6 cells. All work with 122 

infectious NiV was carried out under Biosafety Level 4 (BSL-4) conditions in the Robert E. Shope 123 

BSL-4 and in the Galveston National Laboratory BSL-4 laboratories at the University of Texas 124 

Medical Branch (UTMB). 125 

126 

Quantitation of viral entry 127 

Cells plated in 48-well plates were infected with pseudotyped virions in PBS plus 1% FBS for 128 

2 hours (hrs) at 25oC at 2000rpm (spinoculation). After 2 hr, cells were washed, appropriate growth 129 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 8: Timing of galectin-1 exposure differentially modulates Nipah virus

  8

medium was added, and cells were incubated at 37oC. At 24 hr post-infection, cells were lysed and 130 

luciferase activity was measured as relative light units (RLU) using a Renilla luciferase detection 131 

system (Promega, Madison, WI) and a Veritas microplate luminometer (Turner Biosystems, 132 

Sunnyvale, CA). Galectin-1, buffer control, 100mM lactose and 100mM sucrose were added just prior 133 

to spinoculation. 134 

135 

Reduction in complex N-glycans 136 

Cells (Vero, 293T, HUVECs) were treated with kifunensine (2g/ml) for 24 hrs. To confirm 137 

loss of complex N-glycans on the cell surface, cells were suspended in PBS with 5mM 138 

ethylenediaminetetraacetic acid (EDTA), washed, and resuspended in biotinlyated L-PHA in PBS 139 

(1:1000) plus 1% bovine serum albumin (BSA) (Gemini Scientific) for 1 hr at 4oC. Cells were washed 140 

in PBS and bound PHA detected with a FITC-conjugated streptavidin (SA) (1:60) in PBS plus 1% 141 

BSA, for 1 hr at 4C. Cells were washed in PBS with 1% BSA and flow cytometric analysis was 142 

performed on a FACScan, using CellQuest software (Becton-Dickenson). Pseudotyped VSV 143 

expressing NiV-G and NiV-F were produced by infecting kifunensinee-treated or control cells with 144 

VSV-rLuc virions for 24 hr. Supernatents were collected and virions purified by centrifugation through 145 

20% sucrose in NTE buffer. To examine loss of N-glycan processing by immunoblot, NiV-G and 146 

NiV-F were separated and detected as previously described [24]. 147 

148 

Rescue of recombinant Nipah virus (rNiV) and in vitro luciferase assay 149 

Recombinant Nipah virus expressing secreted Gaussia luciferase (GLuc) (rNiV-GLuc) was rescued at 150 

BSL-4 [28]. Briefly, HEK 293T cells were transfected with the following plasmids: T7-driven 151 

positive-sense rNiV antigenome (3.5g), codon-optomized pCAGGS/T7 (1g), and the T7-driven 152 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 9: Timing of galectin-1 exposure differentially modulates Nipah virus

  9

support plasmids encoding NiV nucleoprotein (1g), phosphoprotein (0.2g) and polymerase (0.4g). 153 

Transfection was performed using TransIT-LT1 transfection reagent (Mirus) following the 154 

manufacturer’s instructions. After 4 days post-transfection, Gaussia luciferase expression was 155 

confirmed using cell supernatant and the BioLux Gaussia Luciferase assay kit (New England Biolabs). 156 

Collected supernatant was used for subsequent virus preparation in Vero CCL-81 cells. HUVECs were 157 

infected with rNiV-GLuc at different multiplicity of infections (MOIs) and cells supernatants were 158 

collected at the indicated intervals. Luciferase assay were performed using BioLux Gaussia Luciferase 159 

assay kit (New England BioLabs) according to the manufacturer’s protocol and expression was 160 

measured on Modulus Luminometer (Turner BioSystems, Inc). 161 

162 

Nipah virus plaque assay 163 

For titrations, confluent monolayers of VeroCCL-81 cells were infected with 100μl of serial 164 

tenfold dilutions of virus-containing cell supernatant. After 1 h incubation at 37°C and 5% CO2, the 165 

inocula were removed and wells overlaid with a mixture of one part 1.0% methylcellulose (Fisher 166 

Scientific) and one part 2xMEM (Gibco, Invitrogen) supplemented with 2% FBS and 2% 167 

penicillin/streptomycin. The plates were incubated at 37°C and 5% CO2 for 3 days and stained with 168 

0.25% crystal violet in 10% buffered formalin. Plates were washed and plaques enumerated. 169 

170 

Wild-type Nipah virus, galectin-1 experiments 171 

HUVECs were infected with NiVMAL at a MOI of 0.1 for 1hr at 37oC. Infections were 172 

performed either in the presence of various galectin-1 concentrations (1, 7, and 20µM) or cells were 173 

treated with galectin-1 post-infection. In the case of infections performed in the presence of galectin-1, 174 

the inoculum was removed 1 hpi and cells were washed three times with 0.1M β-Lactose, followed by 175 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 10: Timing of galectin-1 exposure differentially modulates Nipah virus

  10

incubation in complete EGM-2 media. Cells infected only with NiV were washed three times with 176 

media and incubated in complete EGM-2 media containing different concentrations of galectin-1 (1, 7 177 

or 20µM). Supernatant aliquots were harvested at 1, 12, 24 and 36 hpi and stored at -800C until 178 

titration. DTT was used as negative control and experiments were performed in triplicates. 179 

For DAPI staining, HUVECs were grown on 12-well plate and incubated with NiV for 1 h at 180 

37°C. Galectin-1 treatment was performed as described above. Twenty-four hpi, cells were fixed in 181 

10% buffered formalin, nuclei stained with DAPI (Sigma) 20µg/ml and analyzed by fluorescence 182 

microscopy. 183 

184 

185 

186 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 11: Timing of galectin-1 exposure differentially modulates Nipah virus

  11

Results 187 

188 

Galectin-1 increases viral infection of endothelial cells 189 

Galectin-1 inhibits endothelial cell syncytia formation mediated by NiV envelope 190 

glycoproteins[11]. Although galectin-1 can bind to both NiV-F and –G, we showed that inhibition of 191 

cell-cell fusion was mediated largely by galectin-1 binding to abundant and accessible lactosamine 192 

moieties on complex N-glycans at the “F3” N-glycan site (93NNT101) on NiV-F[11] [24]. The F3 N-193 

glycan site is close to the cathepsin L cleavage site 103DLVGDVR109. Binding of dimeric galectin-1 to 194 

F3 N-glycans on different NiV-F protomers likely accounts for the suite of inhibitory activities that 195 

galectin-1 exerts on NiV-F mediated fusion[11]. We therefore expected galectin-1 to inhibit NiV 196 

infection just as potently. Surprisingly, when we examined the function of galectin-1in Nipah virus 197 

entry using NiV-F/G pseudotyped particles (NiVpp) [29], we found that addition of recombinant 198 

galectin-1 significantly increased infection of target cells in a concentration dependent manner (Fig. 199 

1A). 200 

We confirmed that the galectin-1 mediated increase in infection was carbohydrate dependent. 201 

Lactose, but not sucrose, specifically abrogated the galectin-1 mediated increase in infection, while 202 

lactose alone in the absence of galectin-1 had no effect (Fig. 1B). Thus, exogenous galectin-1 increased 203 

viral infection in a dose-dependent and carbohydrate binding-dependent manner. 204 

205 

Galectin-1 increases viral attachment to host cells 206 

Entry of NiV into host cells requires two steps, viral attachment to the target cell surface 207 

mediated by NiV-G followed by membrane fusion mediated by NiV-F. Viral attachment is largely 208 

energy-independent while virus-cell fusion is an energy-dependent process [30]. To examine the point 209 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 12: Timing of galectin-1 exposure differentially modulates Nipah virus

  12

at which galectin-1 enhancement occurs, NiVpp was centrifuged onto target cells in the presence or 210 

absence of galectin-1 for 2 hours at 4oC, to allow viral attachment but not virus-target cell fusion. 211 

Alternatively, virus was centrifuged onto target cells in the absence of galectin-1, and then incubated 212 

with or without galectin-1 for 1 hr at 37oC to permit fusion. As shown in Fig. 1C, galectin-1 present 213 

during the 4oC spinoculation step clearly enhanced viral entry. In contrast, galectin-1 added after 214 

spinoculation at 4oC and present only during the 37oC fusion step, did not increase viral entry. These 215 

experiments suggest that galectin-1 increased infection by enhancing viral attachment to host cells. 216 

Moreover, that no inhibition was seen suggests that there are functional NiV-F envelope spikes on the 217 

virion particles that are not bound by galectin-1 and are still capable of being triggered by receptor 218 

binding to NiV-G. 219 

220 

Galectin-1 bridges N-glycans on viral envelope glycoproteins to glycoproteins on target cells 221 

Galectin-1 stabilizes HIV-1 attachment and adsorption to host cells by binding specifically to 222 

clustered complex N-glycans on gp120 and CD4, bridging the virus to the target cell [20-22]. We 223 

asked if complex N-glycans, that bear glycan ligands recognized by galectin-1, were involved in the 224 

galectin-1 mediated increase in viral entry of NiVpp. To eliminate complex N-glycans from viral 225 

envelope glycoproteins, virus was produced in 293T cells treated with kifunensine, to block processing 226 

of high-mannose N-glycan precursors into complex N-glycans [31]. We detected equivalent expression 227 

levels of NiV-F by immunoblot from cells treated with and without kifunensine (data not shown). We 228 

confirmed that kifunensine blocked N-glycan processing in 293T cells by flow cytometry with the 229 

lectin L-PHA (Fig. 2A); loss of L-PHA binding indicates loss of complex N-glycans. 230 

Vero cells were infected with NiVpp virus, with NiVpp virus lacking complex N-glycans from 231 

kifunensine-treated 293T cells, or with NiVpp virus specifically lacking the NiV-F3 N-glycan in the 232 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 13: Timing of galectin-1 exposure differentially modulates Nipah virus

  13

presence or absence of exogenous galectin-1 (Fig. 2B). As expected, addition of galectin-1 resulted in 233 

a 12.6 fold increase in target cell infection of virus with a full complement of complex N-glycans (Fig. 234 

2B “wt NiVpp”. Remarkably, the loss of the F3 N-glycan site on NiV-F resulted in a >80% reduction 235 

in galectin-1 mediated enhancement (3.2 fold vs 12.6 fold enhancement for NiV-F3pp vs wt NiVpp 236 

respectively), underscoring the contribution of the complex N-glycan at the F3 site in the enhancement 237 

of virus entry as well as fusogenicity [11, 27]. However, the galectin-1 mediated increase was further 238 

reduced (~95% reduction) when cells were infected with virus lacking complex N-glycans (Fig. 2B, 239 

“Complex N-glycanless NiVpp”), indicating that other complex N-glycans on NiV-F or NiV-G also 240 

contributed to the galectin-1 enhancement effect. 241 

To examine the role of host cell complex N-glycans in galectin-1 mediated enhancement of 242 

viral infection, we treated uninfected Vero cells with kifunensine to reduce complex N-glycans on the 243 

target cells. A reduction of complex N-glycans was confirmed by flow cytometry with L-PHA, as 244 

described above (Fig. 2C). A greater than ten-fold reduction in complex N-glycans from target Vero 245 

cells reduced galectin-1 mediated enhancement of infection by approximately 30% (Fig. 2D). Thus, 246 

complex N-glycans on host cell glycoproteins participate in the galectin-1 enhancement of viral 247 

infection; however, as ten-fold reduction of complex N-glycans on kifunensine treated host cells 248 

partially reduced the galectin-1 enhancement effect, other host cell glycans, likely O-glycans, may also 249 

play a role on the host cell side [32]. Collectively, the data in Fig. 2 demonstrate that glycans on both 250 

the viral envelope and on the target cell surface are important for galectin-1 enhancement. Our results 251 

support a model in which the enhancement effect results from dimeric galectin-1 acting as a bridge or 252 

bivalent attachment factor between the virus and the target cell, with complex N-glycans contributing 253 

to attachment on both the virus and the host cell. 254 

255 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 14: Timing of galectin-1 exposure differentially modulates Nipah virus

  14

Galectin-1 enhances Nipah virus infection of human endothelial cells 256 

Endothelial cells are primary targets of NiV infection in vivo, due to expression of the viral 257 

attachment receptor ephrinB2 [4]. To determine if galectin-1 would enhance Nipah virus infection of 258 

physiologically relevant target cells, we infected primary human umbilical vein endothelial cells 259 

(HUVECs) with NiVpp (Fig. 3A) in the presence or absence of exogenous galectin-1. The addition of 260 

galectin-1 resulted in a 6-fold increase in infection of HUVECs, similar to the effect we had observed 261 

with Vero cells as targets of infection. 262 

Thus far, we used NiVpp, rather than replication competent, wild-type infectious NiV. NiVpp 263 

is a VSV-based pseudotyped virus capable only of single cycle of infection, and the bullet-shaped 264 

VSV-based particles may not fully recapitulate the morphology or envelope glycoprotein densities 265 

present on native pleomorphic paramyxovirus particles [33]. To determine if galectin-1 modulates 266 

infection by replication competent NiV, we first generated and rescued, under BSL4 conditions, a 267 

recombinant NiV expressing secreted Gaussia luciferase (rNiV-GLuc) (see Materials and Methods). 268 

To facilitate the generation of additional recombinant NiVs such as the one bearing the F3 N-glycan, 269 

we also developed an efficient and robust reverse genetics system for henipaviruses [28]. 270 

271 

This rNiV-GLuc replicated to the same end-point titers as the parental NiV Malaysia strain 272 

(data not shown) and was pathogenic in a lethal hamster challenge model [34]; using rNiV-GLuc 273 

allowed us to accurately quantify enhancement or inhibition of live virus entry by sampling the 274 

infected cell culture supernatant for GLuc activity. We first asked if rNiV-GLuc would also exhibit 275 

enhanced infection of endothelial cells in the presence of galectin-1. HUVEC cells infected at an MOI 276 

of 0.05 with rNiV-GLuc showed a dose-dependent enhancement of infection in the presence of 277 

galectin-1 at 24 hours post infection (hpi). The enhancement effect plateaued at 7-10 M galectin-1, 278 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 15: Timing of galectin-1 exposure differentially modulates Nipah virus

  15

with a 6-fold increase in infection compared to controls lacking exogenous galectin-1 (Fig. 3B). This 279 

level of enhancement was similar to that observed for NiVpp in HUVECs, albeit in the presence of 280 

20M galectin-1. 281 

Next we reasoned that, if galectin-1 was bridging virions to the cell surface, as suggested by the 282 

data in Fig. 2, then the relative level of enhancement should be increased when the amount of viral 283 

inoculum was decreased. That is, when the number of infectious viral particles is more limiting, the 284 

effect of galectin-1 on bridging viruses to the cell surface should become more apparent. Thus, we 285 

reduced the viral inoculum by 5-fold and quantified the corresponding effect of galectin-1 on virus 286 

infection. Indeed, at a MOI of 0.01, 10M of galectin-1 enhanced rNiV-GLuc infection by up to 40-287 

fold (Fig. 3C, black bars) compared to the 6-fold enhancement seen with an equivalent dose of 288 

galectin-1 when a higher viral inoculum (MOI of 0.05) was used (Fig 3B). 289 

290 

Galectin-1 enhances rNiV-GLuc virus infection by bridging complex N-Glycans 291 

To confirm that galectin-1 enhances rNiV-GLuc infection by bridging complex N-glycans 292 

present on virions and the endothelial cell surface, as we had seen in infection of Vero cells with 293 

NiVpp (Fig. 2), we generated either HUVECs or viruses deficient in complex N-glycans by treating 294 

HUVECs with kifunensin (Cells Kif+) or growing rNiV-GLuc in the presence of kifunensin (Virus 295 

Kif+). Complex N-glycanless Nipah virus (Virus Kif+) was equally infectious when tittered on Vero 296 

cells (data not shown). As shown in Fig. 3C, depletion of complex N-glycans on target cells (Cells 297 

Kif+) moderately reduced galectin-1 enhanced rNiV-GLuc infection of HUVECs (black bars versus 298 

dark grey bars), as we had seen with Vero cells. Depleting virus of complex N-glycans (Virus Kif+) 299 

also reduced galectin-1 enhancement of infection (light grey bars) more significantly, as we had seen 300 

with NiVpp. When both virus and HUVECs lack complex N-glycans (Virus Kif+/Cells Kif+), 301 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 16: Timing of galectin-1 exposure differentially modulates Nipah virus

  16

galectin-1 had the least enhancement effect (white bars). In toto, our data reveal that galectin-1 302 

enhanced infection of both rNiV-GLuc and NiVpp by the same mechanism, i.e. the interaction 303 

between galectin-1 and Nipah virus results primarily from galectin-1 binding to complex N-glycans on 304 

NiV envelope glycoproteins, and that enhancement of infection involves galectin-1-mediated bridging 305 

of viral glycans to glycans on the host cell surface. The remaining degree of galectin-1 mediated 306 

enhancement seen with kifunensine treated cells and virus suggest that lactosamine residues on O-307 

linked glycans might also serve as bridging receptors for galectin-1’s infection enhancement effect. 308 

However, the role of these putative O-glycans may only be unmasked when cognate complex N-309 

glycans are limiting. 310 

311 

Timing of galectin-1 exposure has opposing effects in Nipah virus replication 312 

Previous work in our labs has shown that galectin-1 inhibited syncytia formation mediated by 313 

NiV-F+G expressed by endothelial cells [11, 24]. Galectin-1 reduced NiV-F mediated fusion of 314 

endothelial cells by retarding NiV-F maturation, reducing the lateral mobility of NiV-F in the plasma 315 

membrane, and inhibiting triggering of NiV-F that results in fusion of the virus with the host cell 316 

plasma membrane. Moreover, endogenous galectin-1 produced by endothelial cells was sufficient to 317 

inhibit syncytia formation [11]. Importantly, all of the galectin-1 mediated effects that we had 318 

previously analyzed would occur following Nipah virus infection, and was studied only in a cell-cell 319 

fusion context. Thus, while the data presented demonstrate that galectin-1 enhances viral entry when 320 

present at initial infection, we asked if downstream events, i.e. syncytia formation and viral replication 321 

in human endothelial cells infected with NiVMAL, were affected by the presence of galectin-1. 322 

To examine the effect on viral replication, HUVECs infected with the Nipah virus strain 323 

Malaysia (NiVMAL) were incubated post-infection with galectin-1 or buffer control. The presence of 324 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 17: Timing of galectin-1 exposure differentially modulates Nipah virus

  17

exogenous galectin-1 post-infection at early time points (12 and 24 hpi) inhibited virus replication 325 

titers below the limit of detection, effectively reducing viral titers by >3 logs at 24 hpi. This inhibitory 326 

effect of galectin-1 persisted at 36 hpi as a 15-fold reduction in viral titers (Fig. 4A). 327 

To examine the effect of galectin-1 on syncytia formation, wild type NiVMAL-infected 328 

HUVECs were analyzed microscopically in three conditions; no exogenous galectin-1 added, galectin-329 

1 added before infection, or galectin-1 added post-infection. Addition of exogenous galectin-1 before 330 

infection enhanced syncytia formation (Fig. 4B, panel 2), consistent with the effect of galectin-1 on 331 

increasing NiV infection of endothelial cells (Fig. 3). In contrast, addition of exogenous galectin-1 332 

after NiV infection essentially abrogated syncytia formation (Fig. 4B, panel 3). This inhibitory effect 333 

of galectin-1 on endothelial cell fusion (quantified in Fig. 4C) is consistent with our previous 334 

observation using pseudotyped virus [11]. 335 

To determine if the NiV-F3 N-glycan contributes to the differential effect of galectin-1 on 336 

syncytia formation in the context of live virus infection, we generated and rescued a rNiV-GLuc 337 

bearing the NiV-F3 N-glycan mutant. Infection with the rNiV-GLuc F3 N-glycan mutant virus resulted 338 

in an overall increase in syncytia formation, compared to wild-type NiV-MAL, consistent with the 339 

hyperfusogenic phenotype previously observed for the NiV-F3 mutant [27]. However, addition of 340 

galectin-1 before infection no longer enhanced syncytia formation, in contrast to the enhancement seen 341 

with the wild-type virus in the presence of galectin-1 (Fig. 4C). Galectin-1 was still able to inhibit 342 

syncytia formation by the mutant virus when added after infection, although lack of N-glycans at the 343 

F3 site made the mutant virus more resistant to the inhibitory effect of galectin-1 (Fig. 4C). 344 

Collectively, our data indicate that the timing of galectin-1 exposure differentially modulates 345 

Nipah virus entry, syncytia formation and replicative spread in endothelial cells. Galectin-1 present at 346 

the attachment/entry step of viral infection enhanced NiV entry into endothelial cells and also 347 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 18: Timing of galectin-1 exposure differentially modulates Nipah virus

  18

enhanced syncytia formation, likely due to increased virus envelope production in initially infected 348 

cells. In contrast, galectin-1 added after NiV infection of endothelial cells inhibited both syncytia 349 

formation and the replicative spread of progeny virus such that viral titers at 24hpi and 36hpi lagged by 350 

at least 4 logs and 1 log respectively. 351 

352 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 19: Timing of galectin-1 exposure differentially modulates Nipah virus

  19

Discussion 353 

Innate immune mediators such as galectins have many functions, including triggering 354 

endothelial activation, inducing cytokine production and release, promoting leukocyte infiltration of 355 

infected tissues, and mediating pathogen clearance [10, 14, 16, 35, 36]. As galectin-1 can recognize 356 

both self (mammalian) and non-self (pathogen) glycans, depending on factors such as glycan ligand 357 

density and presentation [37], galectin-1 has been proposed to be both a damage-associated molecular 358 

pattern (DAMP) that is released from injured cells, and a pattern recognition receptor (PRR), 359 

recognizing pathogen associated molecular patterns (PAMPs), such as specific viral glycans [11]. 360 

Thus, galectin-1 can have dual functions in innate immunity [38]. Our current data also demonstrate 361 

that galectin-1 has complex and dichotomous roles in NiV infection. Galectin-1 enhanced initial NiV 362 

attachment to and entry into human endothelial target cells (Fig. 3), while subsequently inhibiting cell 363 

syncytia formation (Fig. 4) [11, 24], a key sequel of viral replicative spread. The ability of galectin-1 to 364 

produce these divergent effects depends on when it is present during the viral replicative life cycle. 365 

There are several examples of galectin-1 having complex or opposing roles in different viral 366 

infections, via binding to glycan ligands on both the virus and the target cell. Galectin-1 bound to 367 

influenza virus reduced virus entry into host cells; moreover, intranasal administration of galectin-1 368 

enhanced survival of mice infected with a lethal dose of influenza virus, and galectin-1 null mice were 369 

more susceptible to influenza virus infection than wildtype mice [39]. In contrast, galectin-1 increased 370 

HIV-1 infection of monocyte derived macrophages through stabilization of virus attachment and 371 

adsorption [20, 21]. That galectin-1 stabilized HIV adsorption to macrophages suggests that dimeric 372 

galectin-1 can act as a bridge between virus glycans and host cell surface glycans, although this was 373 

not directly demonstrated in those studies. 374 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 20: Timing of galectin-1 exposure differentially modulates Nipah virus

  20

Here we examined whether galectin-1 formed a bridge between viral particles and host cells to 375 

enhance infection. The enhancement effect was clearly glycan-dependent, as the specific inhibitor 376 

lactose, but not the non-cognate disaccharide sucrose, blocked the enhancement of viral infection (Fig. 377 

1B). To specifically address the step at which galectin-1 binding promoted infection, we bound virus to 378 

host cells at 4oC and initiated viral entry at 37oC (Fig. 1C). Galectin-1 added only during the 379 

attachment step at 4oC enhanced infection, while galectin-1 added after viral attachment, but during 380 

fusion at 37oC, did not enhance infection. These data support a model whereby galectin-1 enhancement 381 

occurs as a result of bridging glycoproteins on the virus to glycoproteins on the host cell. To determine 382 

whether the essential glycans recognized by galectin-1 were on the virus, the host cell, or both, we 383 

eliminated complex N-glycans on viral particles and on the surface of the target cells by treatment with 384 

the -mannosidase I inhibitor kifunensine. Complex N-glycans on viral glycoproteins were clearly 385 

required for galectin-1 enhancement of infection (Fig. 2B and 3C). We have previously demonstrated 386 

that galectin-1 specifically interacts with the F3 N-glycan on the NiV-F glycoprotein, and that the F3 387 

N-glycan site is occupied by complex N-glycans that bears cognate ligands for galectin-1 [11, 27]. In 388 

the present report, we show that this F3 glycan was the most significant viral determinant of galectin-1 389 

mediated enhancement of infection (Fig. 2B and Fig. 4C). On the target cells, both complex N-glycans 390 

and O-glycans appear to contribute to galectin-1 enhancement of infection (Fig. 2D and 3C) [40]. 391 

While several endothelial cell surface glycoprotein receptors for galectin-1 have been identified [41], 392 

the entire repertoire of cell surface glycoproteins that could bind to galectin-1 to facilitate viral 393 

“bridging” and increase virus attachment is not known. In addition, little is known about the N- or O-394 

glycan composition of the Nipah cellular receptor ephrinB2 [42] and whether galectin-1 can bind to 395 

ephrinB2 on the surface of endothelial cells. 396 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 21: Timing of galectin-1 exposure differentially modulates Nipah virus

  21

Lectin-glycan interactions play complex roles in virus-host pathogenesis, and other types of 397 

innate immune lectins have apparently dichotomous roles in viral pathogenesis. For example, mannan-398 

binding lectin (MBL), a soluble innate immune lectin, binds to influenza A virus and leads directly to 399 

virus inactivation [43]. In contrast, another mannose-binding human lectin, macrophage mannose 400 

receptor (MMR) potentiates HIV binding to target cells [44-46], and MMR can also potentiate 401 

infection of macrophages by influenza virus [47], so that different mannose-binding lectins can bind to 402 

viruses to either reduce or potentiate infection. Considering the long-standing evolutionary relationship 403 

between virus and host, it is not entirely surprising that viruses have learned to co-opt host innate 404 

immune defenses for increased target cell entry and replication. 405 

In the present study, we demonstrate that galectin-1 enhanced infection of endothelial cells by 406 

live NiV (Fig. 3C); as inflammation is known to increase endothelial cell production of galectin-1 [11], 407 

we speculate that, in vivo, galectin-1 produced by endothelial cells may increase target cell infection by 408 

increasing viral attachment to host target cells. However, galectin-1 can also inhibit syncytia formation 409 

among endothelial cells infected with NiV in vitro (Fig. 4). This effect may limit the extent or severity 410 

of NiV pathogenicity in vivo, potentially reducing endothelial cell damage and the resulting 411 

hemorrhagic diathesis. Moreover, the opposing effects of increased viral entry by facilitating viral 412 

attachment without affecting viral fusion with the endothelial cell membrane versus inhibition of 413 

membrane fusion during syncytia formation of infected cells suggests that these two types of fusion 414 

events are not identical. 415 

It is remarkable that complex N-glycans on a specific site in NiV-F (F3) can account for the 416 

majority of galectin-1’s effect. To gain further insight into how galectin-1 binding to F3 N-glycan 417 

mediates its dichotomous role in NiV infection and spread, we modeled a trimeric spike with the 418 

cognate N-glycans at the relvant sites. Fig 5A shows that the F3 N-glycan (red) is positioned right 419 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 22: Timing of galectin-1 exposure differentially modulates Nipah virus

  22

“above” the fusion peptide (blue), and proximal to the putative target cell membrane. The distance 420 

between glycan binding sites on the dimeric galectin-1 is ~50Å (Fig. 5B), which is too short to span 421 

the F3 N-glycans between any two F protomers on the same timeric NiV-F spike. Thus, galectin-1 422 

likely binds to the F3 N-glycans on NiVF protomers from distinct F spikes, forming a glycoprotein 423 

lattice that blocks the triggering of the fusion peptide (Fig. 5A, legend). We posit that glycoprotein 424 

lattice formation predominates when galectin-1 binds to NiV-F expressed on infected cells, thereby 425 

inhibiting syncytia formation [11]. However, on the virion surface that is tightly packed with envelope 426 

glycoproteins, glycoprotein lattice formation may be less efficient, and the ability of galectin-1 to 427 

bridge virus predominates. The latter can lead to an enhancement of infection when receptor binding 428 

triggers fusion by the other virion-associated F spikes that are not bound by galectin-1. 429 

As mentioned above, while endothelial cells express abundant galectins, inflammation in 430 

general and viral infection specifically are known to significantly increase endothelial cell expression 431 

of several galectins, including galectin-1. Moreover, as secreted galectins bind back to glycoproteins 432 

on the plasma membrane as well as to glycans on extracellular matrix glycoproteins, galectins can be 433 

highly concentrated at the cell surface. Thus, the galectin-1 concentration used in these studies may 434 

reflect the physiologic concentration of galectin-1 encountered by a virus at the endothelial cell 435 

membrane [48-51]. 436 

This study further characterizes the complex interaction between galectin-1 and NiV. While 437 

other lectins have been shown to have effects both beneficial and detrimental to the host in microbial 438 

infections [20, 21, 36, 39], we report these opposing effects for a single virus, NiV, and a single lectin, 439 

galectin-1, in the same host cell. These studies may help to explain the range of clinical outcomes of 440 

NiV infection, as some patients recover fully after infection, while other patients succumb. Perhaps 441 

differential expression of galectin-1 in different individuals at different time points during infection 442 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 23: Timing of galectin-1 exposure differentially modulates Nipah virus

  23

either promotes virus-endothelial cell interactions or prevents endothelial cell damage. The present 443 

work suggests a complex spatiotemporal interplay between an immune regulator and a virus that may 444 

ultimately determine the outcome of infection. 445 

446 

Acknowledgements 447 

This study was supported by NIH grant R01AI060694 (to L.G.B. and B.L.) a Ruth L. Kirschstein 448 

National Research Service Award T32HL69766 (to O.B.G.), NIH grant R01AI109022 (to H.A.C.), and 449 

Medical Research Council Grant MR/L009528/1 (to T.A.B.). A.N.F. acknowledges partial support by 450 

a grant from the NIAID/NIH through the Western Regional Center of Excellence for Biodefense and 451 

Emerging Infectious Disease Research (U54 AI057156). 452 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 24: Timing of galectin-1 exposure differentially modulates Nipah virus

  24

453 

References 454 

1. Hsu, V.P., M.J. Hossain, U.D. Parashar, M.M. Ali, T.G. Ksiazek, I. Kuzmin, M. 455 

Neizgoda, C. Ruppercht, J. Bresee, and R.F Breiman. 2004. Nipah virus encephalitis 456 

reemergence, Bangladesh. Emerg. Infect. Dis. 10:2082-2087. 457 

2. Stone, R. 2011. Epidemiology. Breaking the chain in Bangladesh. Science. 331:1128-1131. 458 

3. Nahar, N., U.K. Mondal, M.J. Hossain, M.S. Uddin Khan, R. Sultana, E.S. Gurley, and 459 

S.P. Luby. 2014. Piloting the promotion of bamboo skirt barriers to prevent Nipah virus 460 

transmission through date palm sap in Bangladesh. Glob. Health. Promot. Epub ahead of print. 461 

4. Negrete, O.A., E.L Levroney, H.C. Aguilar, A. Bertolotti-Ciarlet, R. Nazarian, S. Tajyar, 462 

and B. Lee. 2005. EphrinB2 is the entry receptor for Nipah virus, an emergent deadly 463 

paramyxovirus. Nature. 436:401-405. 464 

5. Negrete, O.A., M.C. Wolf, H.C. Aguilar, S. Enterlein, W. Wang, E. Muhlberger, S.V. Su, 465 

A. Bertolotti-Ciarlet, R. Flick, and B. Lee. 2006. Two key residues in ephrinB3 are critical 466 

for its use as an alternative receptor for Nipah virus. PLoS Pathog. 2:e7. 467 

6. Aguilar, H.C. and R.M. Iorio. 2012. Henipavirus membrane fusion and viral entry. Curr. Top. 468 

Microbiol. Immunol. 359:79-94. 469 

7. Maisner, A., J. Neufeld, and H. Weingartl. 2009. Organ- and endotheliotropism of Nipah 470 

virus infections in vivo and in vitro. Thromb. Haemost. 102:1014-1023. 471 

8. Behling-Kelly, E. and C.J. Czuprynski. 2007. Endothelial cells as active participants in 472 

veterinary infections and inflammatory disorders. Anim. Health Res. Rev. 8:47-58. 473 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 25: Timing of galectin-1 exposure differentially modulates Nipah virus

  25

9. Wahl-Jensen, V.M., T.A. Afanasieva, J. Seebach, U. Stroher, H. Feldmann, and H.J. 474 

Schnittler. 2005. Effects of Ebola virus glycoproteins on endothelial cell activation and barrier 475 

function. J. Virol. 79:10442-10450. 476 

10. Thijssen, V.L., R. Postel, R.J. Brandwijk, R.P. DIngs, I. Nesmelova, S. Satijn, N. 477 

Verhofstad, Y. Nakabeppu, L.G Baum, J. Bakkers, K.H. Mayo, F. Poirier, and A.W. 478 

Griffioen. 2006. Galectin-1 is essential in tumor angiogenesis and is a target for 479 

antiangiogenesis therapy. Proc. Natl. Acad. Sci. USA. 103:15975-15980. 480 

11. Garner, O.B., H.C. Aguilar, J.A. FUlcher, E.L Levroney, R. Harrison, L. Wright, L.R. 481 

Robinson, V. Aspericueta, M. Panico, S.M. Haslam, H.R. Morris, A. Dell, B. Lee, and 482 

L.G. Baum. 2010. Endothelial galectin-1 binds to specific glycans on nipah virus fusion 483 

protein and inhibits maturation, mobility, and function to block syncytia formation. PLoS 484 

Pathog. 6:e1000993. 485 

12. Warke, R.V., K. Xhaja, K.J. Martin, M.F. Fournier, S.K. Shaw, N. Brizuela, N. de Bosch, 486 

D. Lapointe, F.A. Ennis, A.L. Rothman, and I. Bosch. 2003. Dengue virus induces novel 487 

changes in gene expression of human umbilical vein endothelial cells. J. Virol. 77:11822-488 

11832. 489 

13. Imaizumi, T., H. Yoshida, N. Nishi, H. Sashinami, T. Nakamura, M. Hirashima, C. 490 

Ohyama, K Itoh, and K. Satoh. 2007. Double-stranded RNA induces galectin-9 in vascular 491 

endothelial cells: involvement of TLR3, PI3K, and IRF3 pathway. Glycobiology. 17:12C-5C. 492 

14. Stowell, S.R., C.M. Arthur, R. McBride, O. Berger, N. Razi, J. Heimburg-Molinaro, L.C. 493 

Rodrigues, J.P. Gourdine, A.J. Noll, S. von Gunten, D.F. Smith, Y.A. Knirel, J.C. 494 

Paulson, and R.D. Cummings. 2014. Microbial glycan microarrays define key features of 495 

host-microbial interactions. Nat. Chem. Biol. 10:470-476. 496 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 26: Timing of galectin-1 exposure differentially modulates Nipah virus

  26

15. Stowell, S.R., C.M. Arthur, M. Dias-Baruffi, L.C. Rodrigues, J.P. Gourdine, J. Heimburg-497 

Molinaro, T. Ju, R.J. Molinaro, C. Rivera-Marrero, B. Xia, D.F. Smith, and R.D 498 

Cummings. 2010. Innate immune lectins kill bacteria expressing blood group antigen. Nat. 499 

Med. 16:295-301. 500 

16. Kohatsu, L., D.K. Hsu, A.G. Jegalian, F.T. Liu, and L.G. Baum. 2006. Galectin-3 induces 501 

death of Candida species expressing specific beta-1,2-linked mannans. J. Immunol. 177:4718-502 

4726. 503 

17. Gupta, S.K., S. Masinick, M. Garrett, and L.D. Hazlett. 1997. Pseudomonas aeruginosa 504 

lipopolysaccharide binds galectin-3 and other human corneal epithelial proteins. Infect. Immun. 505 

65:2747-2753. 506 

18. Moody, T.N., J. Ochieng, and F. Villalta. 2000. Novel mechanism that Trypanosoma cruzi 507 

uses to adhere to the extracellular matrix mediated by human galectin-3. FEBS Lett. 470:305-508 

308. 509 

19. Kleshchenko, Y.Y., T.N. Moody, V.A. Furtak, J. Ochieng, M.F. Lima, and F. Villalta. 510 

2004. Human galectin-3 promotes Trypanosoma cruzi adhesion to human coronary artery 511 

smooth muscle cells. Infect. Immun. 72:6717-6721. 512 

20. Ouellet, M., S. Mercier, I. Pelletier, S. Bounou, J. Roy, J. Hirabayashi, S. Sato, and M.J. 513 

Tremblay. 2005. Galectin-1 acts as a soluble host factor that promotes HIV-1 infectivity 514 

through stabilization of virus attachment to host cells. J. Immunol. 174:4120-4126. 515 

21. Mercier, S., C. St-Pierre, I. Pelletier, M. Oullet, M.J. Tremblay, and S. Sato. 2008. 516 

Galectin-1 promotes HIV-1 infectivity in macrophages through stabilization of viral adsorption. 517 

Virology. 371:121-129. 518 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 27: Timing of galectin-1 exposure differentially modulates Nipah virus

  27

22. St-Pierre, C., H. Manya, M. Oullet, G.F. Clark, T. Endo, M.J. Tremblay, and S. Sato. 519 

2011. Host-soluble galectin-1 promotes HIV-1 replication through a direct interaction with 520 

glycans of viral gp120 and host CD4. J. Virol. 85:11742-11751. 521 

23. Bi, S., P.W. Hong, B. Lee, and L.G. Baum. 2011. Galectin-9 binding to cell surface protein 522 

disulfide isomerase regulates the redox environment to enhance T-cell migration and HIV 523 

entry. Proc. Natl. Acad. Sci. U S A. 108:10650-10655. 524 

24. Levroney, E.L., H.C. Aguilar, J.A. Fulcher, L. Kohatsu, K.E. Pace, M. Pang, K. B. 525 

Gurney, L.G. Baum, and B. Lee. 2005. Novel innate immune functions for galectin-1: 526 

galectin-1 inhibits cell fusion by Nipah virus envelope glycoproteins and augments dendritic 527 

cell secretion of proinflammatory cytokines. J. Immunol. 175:413-420. 528 

25. Pace, K.E., C. Lee, P.L. Stewart, and L.G. Baum. 1999. Restricted receptor segregation into 529 

membrane microdomains occurs on human T cells during apoptosis induced by galectin-1. J. 530 

Immunol. 163:3801-3811. 531 

26. Perillo, N.L., C.H. Uittenbogaart, J.T. Nguyen, and L.G. Baum. 1997. Galectin-1, an 532 

endogenous lectin produced by thymic epithelial cells, induces apoptosis of human thymocytes. 533 

J. Exp. Med. 185:1851-1858. 534 

27. Aguilar, H.C., K.A. Matreyek, C.M. Filone, S.T. Hashimi, E.L. Levroney, O.A. Negrete, 535 

A. Bertolotti-Ciarlet, D.Y. Choi, I. McHardy, J.A. Fulcher, S.V. Su, M.C. Wolf, L. 536 

Kohatsu, L.G. Baum, and B. Lee. 2006. N-glycans on Nipah virus fusion protein protect 537 

against neutralization but reduce membrane fusion and viral entry. J. Virol. 80:4878-4889. 538 

28. Yun, T., A. Park, T.E. Hill, O. Pernet, S.M. Beaty, T. L. Juelich, J.K. Smith, L. Zhang, 539 

Y.E. Wang, F. Vigant, J. Gao, P. Wu, B. Lee, and A.N. Freiberg. 2014. Efficient reverse 540 

genetics reveals genetic determinants of budding and fusogenic differences between Nipah and 541 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 28: Timing of galectin-1 exposure differentially modulates Nipah virus

  28

Hendra virus and enables real-time monitoring of viral spread in small animal models of 542 

henipavirus infection. J. Virol. pii:JVI02583-14. 543 

29. Tamin, A., B.H Harcourt, M.K. Lo, J.A. Roth, M.C. Wolf, B. Lee, H. Weingartl, J.C. 544 

Audonnet, W.J. Bellini, and P.A. Rota. 2009. Development of a neutralization assay for 545 

Nipah virus using pseudotype particles. J. Virol. Methods. 160:1-6. 546 

30. Stachowiak, J.C., F.M. Brodsky, and E.A. Miller. 2013. A cost-benefit analysis of the 547 

physical mechanisms of membrane curvature. Nat. Cell. Biol. 15:1019-10127. 548 

31. Elbein, A.D., J.E. Tropea, M. Mitchell, and G.P. Kaushal. 1990. Kifunensine, a potent 549 

inhibitor of the glycoprotein processing mannosidase I. J. Biol. Chem. 265:15599-155605. 550 

32. Nguyen, J.T., D.P. Evans, M. Galvan, K.E, Pae, D. Leitenberg, T.N. Bui, and L.G. Baum. 551 

2001. CD45 modulates galectin-1-induced T cell death: regulation by expression of core 2 O-552 

glycans. J. Immunol. 167:5697-56707. 553 

33. Wolf, M.C., Y. Wang, A.N. Freiberg, H.C. Aguilar, M.R. Holbrook, and B. Lee. 2009. A 554 

catalytically and genetically optimized beta-lactamase-matrix based assay for sensitive, 555 

specific, and higher throughput analysis of native henipavirus entry characteristics. Virol. J. 556 

6:119. 557 

34. Pernet, O., B.S. Schneider, S.M. Beaty, M. LeBreton, T.E. Yun, A. Park, T.T. Zachariah, 558 

T.A. Bowden, P. Hitchens, C.M. Ramirez, P. Daszak, J. Mazet, A.N. Freiberg, N.D. Wolfe, 559 

and B. Lee. 2014. Evidence for henipavirus spillover into human populations in Africa. Nat. 560 

Commun. 5:5342. 561 

35. Liu, F.T., R.Y. Yang, and D.K. Hsu. 2012. Galectins in acute and chronic inflammation. Ann. 562 

N Y Acad. Sci. 1253:80-91. 563 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 29: Timing of galectin-1 exposure differentially modulates Nipah virus

  29

36. Cerliani, J.P., S.R. Stowell, I.D. Mascanfroni, C.M. Arthur, R.D. Cummings, and G.A. 564 

Rabinovich. 2011. Expanding the universe of cytokines and pattern recognition receptors: 565 

galectins and glycans in innate immunity. J. Clin. Immunol. 31:10-21. 566 

37. Dam, T.K. and C.F. Brewer. 2010. Lectins as pattern recognition molecules: the effects of 567 

epitope density in innate immunity. Glycobiology. 20:270-279. 568 

38. Sato, S., C. St-Pierre, P. Bhaumik, and J. Nieminen. 2009. Galectins in innate immunity: 569 

dual functions of host soluble beta-galactoside-binding lectins as damage-associated molecular 570 

patterns (DAMPs) and as receptors for pathogen-associated molecular patterns (PAMPs). 571 

Immunol. Rev. 230:172-187. 572 

39. Yang, M.L., Y.H. Chen, S.W. Wang, Y.J. Huang, C.H. Leu, N.C. Yeh, C.Y. CHu, C.C. 573 

Lin, G.S. Shieh, Y.L. Chen, J.R. Wang, C.H. Wang, C.L. Wu, and A.L. Shiau. 2011. 574 

Galectin-1 binds to influenza virus and ameliorates influenza virus pathogenesis. J. Virol. 575 

85:10010-10020. 576 

40. Earl, L.A., S. Bi, and L.G. Baum. 2010. N- and O-glycans modulate galectin-1 binding, 577 

CD45 signaling, and T cell death. J. Biol. Chem. 285:2232-2244. 578 

41. Croci, D.O., J.P. Cerliani, N.A. Pinto, L.G. Morosi, and G.A. Rabinovich. 2014. 579 

Regulatory role of glycans in the control of hypoxia-driven angiogenisis and sensitivity to anti-580 

angiogenic treatment. Glycobiology. 24:1283-1290. 581 

42. Xu, K., Y.P. Chan, K.R. Rajashankar, D. Khetawat, L. Yan, M.V. Kolev, C.C. Broder, 582 

and D.B. Nikolov. 2012. New insights into the Hendra virus attachment and entry process 583 

from structures of the virus G glycoprotein and its complex with Ephrin-B2. PLoS One. 584 

7:e48742. 585 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 30: Timing of galectin-1 exposure differentially modulates Nipah virus

  30

43. Kase, T., Y. Suzuki, T. Kawai, T. Sakamoto, K. Ohtani, S. Eda, A. Maeda, T. Okuno, T. 586 

Kurimura, and N. Wakamiya. 1999. Human mannan-binding lectin inhibits the infection of 587 

influenza A virus without complement. Immunology. 97:385-392. 588 

44. Nguyen, D.G. and J.E. Hildreth. 2003. Involvement of macrophage mannose receptor in the 589 

binding and transmission of HIV by macrophages. Eur. J. Immunol. 33:483-493. 590 

45. Liu, Y., H. Liu, B.O. Kim, V.H. Gattone, J. Li, A. Nath, J. Blum, and J.J. He. 2004. CD4-591 

independent infection of astrocytes by human immunodeficiency virus type 1: requirement for 592 

the human mannose receptor. J. Virol. 78:4120-4133. 593 

46. Lai, J., O.K. Bernhard, S.G. Turville, A.N. Harman, J. Wilkinson, and A.L. Cunningham. 594 

2009. Oligomerization of the macrophage mannose receptor enhances gp120-mediated binding 595 

of HIV-1. J. Biol. Chem. 284:11027-11038. 596 

47. Upham, J.P., D. Pickett, T. Irimura, E.M. Anders, and P.C. Reading. 2010. Macrophage 597 

receptors for influenza A virus: role of the macrophage galactose-type lectin and mannose 598 

receptor in viral entry. J. Virol. 84:3730-3737. 599 

48. Darrow, A.L., R.V. Shohet, and J.G. Maresh. 2011. Transcriptional analysis of the 600 

endothelial response to diabetes reveals a role for galectin-3. Physiol. Genomics. 20:1144-601 

1152. 602 

49. Bi, S., L.A. Earl, L. Jacobs, and L.G. Baum. 2008. Structural features of galectin-9 and 603 

galectin-1 that determine distinct T cell death pathways. J. Biol. Chem. 283:12248-12258. 604 

50. Stillman, B.N., D.K. Hsu, M. Pang, C.F. Brewer, P. Johnson, F.T. Liu, and L.G. Baum. 605 

2006. Galectin-3 and galectin-1 bind distinct cell surface glycoprotein receptors to induce T 606 

cell death. 176:778-789. 607 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 31: Timing of galectin-1 exposure differentially modulates Nipah virus

  31

51. He, J., and L.G. Baum. 2004. Presentation of galectin-1 by extracellular matrix triggers T cell 608 

death. J. Biol. Chem. 279:4705-4712. 609 

52. Arnold, K., L. Bordoli, J. Kopp, and T. Schwede. 2006. The SWISS-MODEL workspace: a 610 

web-based environment for protein structure homology modelling. Bioinformatics. 22:195-201. 611 

53. Yin, H.S., X. Wen, R.G. Paterson, R.A. Lamb, and T.S. Jardetzky. 2006. Structure of the 612 

parainfluenza virus 5 F protein in its metastable, prefusion conformation. Nature. 439:38-44. 613 

54. Crispin, M., X. Yu, and T.A. Bowden. 2013. Crystal structure of sialylated IgG Fc: 614 

implications for the mechanism of intravenous immunoglobulin therapy. Proc. Natl. Acad. Sci. 615 

U S A. 110:E3544-3546. 616 

55. Crispin, M., T.A. Bowden, C.H. Coles, K. Harlos, A.R. Aricescu, D.J. Harvey, D.I. Stuart, 617 

and E.Y. Jones. 2009. Carbohydrate and domain architecture of an immature antibody 618 

glycoform exhibiting enhanced effector functions. J. Mol. Biol. 387:1061-1066. 619 

56. Lopez-Lucendo, M.F., D. Solis, S. Andre, J. Hirabayashi, K. Kasai, H. Kaltner, H.J. 620 

Gabius, and A. Romero. 2004. Growth-regulatory human galectin-1: crystallographic 621 

characterisation of the structural changes induced by single-site mutations and their impact on 622 

the thermodynamics of ligand binding. J. Mol. Biol. 343:957-970. 623 

 624 

625 

626 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 32: Timing of galectin-1 exposure differentially modulates Nipah virus

  32

Figure Legends 627 

Figure 1. Galectin-1 enhances infection of NiVpp in a carbohydrate binding dependent manner. 628 

(A) Quantification of galectin-1 enhancement of NiVpp infection. NiVpp was titrated such that viral 629 

inoculum that gave luciferase activity in the linear range at 24hpi was used. NiVpp was added to 630 

monolayers of Vero cells, and virus entry in the absence (white bars) and presence (black bars) of 631 

increasing concentrations of galectin-1 was quantified by measuring luciferase activity in infected cell 632 

lysate 24 hpi as described in Experimental Procedures. Data from one of three replicate experiments 633 

are presented as mean fold-increase (+/- SD of triplicate samples) over the virus only (no galectin-1) 634 

condition. Significant p value determined by two-way ANOVA. (B) Galectin-1 enhancement of NiVpp 635 

infection is carbohydrate binding dependent. NiVpp (white bars) and 10 M galectin-1 (black bars) 636 

was added to Vero cells in the presence of 100mM lactose or 100mM sucrose. Data are presented as in 637 

(A); mean +/- SD of triplicate samples from one of three replicate experiments is shown. (C) Galectin-638 

1 added during spinoculation (at 4C) shows an increase in NiVpp infection on Vero cells. NiVpp 639 

infection in the absence (white bars) or presence (black bars) of 10M galectin-1 added during or after 640 

spinoculation, i.e. during or after attachment of virus to cells. Data are mean +/- SD of triplicate 641 

samples from one of three replicate experiments. Significant p value determined by Student’s t test. 642 

643 

Figure 2. Galectin-1 enhances infection of NiVpp by bridging the virus to the cell through 644 

binding of viral surface and cell surface complex N-glycans. Flow cytometric analysis of L-PHA 645 

binding to 293T cells (A) or Vero cells (C) treated with kifunensine (dashed black line) shows loss of 646 

cell surface complex N-glycans when compared to L-PHA binding to untreated parental cells (bold 647 

black outlined histograms). Gray filled histogram represents negative control with secondary only. 648 

(B) Galectin-1 mediated enhancement of NiVpp infection is dependent upon complex N-glycans found 649 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 33: Timing of galectin-1 exposure differentially modulates Nipah virus

  33

on the surface of the virus. Virus infection of Vero cells in the absence (white) or presence of 20M 650 

galectin-1 (black) is shown for NiVpp bearing three types of envelope glycoproteins; wild type NiV-F 651 

+ -G, NiV-F3 mutant (+ wild type G), or NiV-F + -G devoid of complex N-glycans. Data from one of 652 

three replicate experiments are presented as mean fold-increase in infection (+/- SD of triplicate 653 

samples) over the virus only (no galectin-1) condition. (D) Galectin-1 mediated enhancement of 654 

NiVpp infection is dependent upon complex N-glycans on the surface of Vero cells. Virus infection in 655 

the absence (white bars) or presence (black bars) of 20 M galectin-1 is shown for wild-type Vero 656 

cells and Vero cells devoid of complex N-glycans. Data (mean +/- SD of triplicate samples) from one 657 

of three replicate experiments are presented exactly as described for (B). 658 

659 

Figure 3. Galectin-1 enhances NiVpp and live Nipah virus infection of HUVECs. 660 

(A) NiVpp was added to monolayers of HUVECS in the absence (white bar) and presence (black bars) 661 

of 20M of galectin-1 for 1 hour, and infection was quantified by measuring Renilla luciferase activity 662 

at 24hpi. Data are presented as mean fold-increase (+/- SD of triplicate samples) in infection over the 663 

virus only (no galectin-1) condition. One of three replicate experiments is shown. (B) Quantification of 664 

galectin-1 enhancement of recombinant GLuc reporter NiV (rNiV-GLuc) infection of HUVECs. (B) 665 

rNiV-GLuc was added to monolayers of HUVECs in the absence (white bar) and presence (black bars) 666 

of the indicated amounts of galectin-1 for 1 hour, and infection was quantified by measuring Gaussia 667 

luciferase activity. Data are presented as mean fold-increase (+/- SD of triplicate samples) in infection 668 

over the virus only (no galectin-1) condition. One of three replicate experiments is shown. (C) 669 

Galectin-1 mediated enhancement of rNiV-GLuc infection is dependent upon complex N-glycans on 670 

the surface of HUVECs and on the surface of the virus. Virus or HUVECs deficient in complex N-671 

glycans were made in the presence of 20M Kifunensine (Virus Kif+ and Cells Kif+, respectively). 672 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 34: Timing of galectin-1 exposure differentially modulates Nipah virus

  34

rNiV-GLuc infection in the absence or presence of increasing concentrations of galectin-1 is shown for 673 

wild type rNiV-GLuc and HUVECs (Virus wt/Cells wt, black bars), wild type rNiV-GLuc and 674 

complex N-glycan deficient HUVECs (Virus wt/Cells Kif+, dark grey bars), complex N-glycan 675 

deficient rNiV-GLuc and wild type HUVECs (Virus Kif+/Cells wt, light grey bars), and complex N-676 

glycan deficient rNiV-GLuc and HUVECs (Virus kif+/Cells Kif+, white bars). Data represent the 677 

average fold- increase in infection determined as in (B), and presented as mean +/- SD of triplicate 678 

samples from one of three replicate experiments. 679 

680 

Figure 4. Galectin-1 can have opposing effects on Nipah virus production and syncytia formation 681 

(A) Effect of galectin-1 added post-infection on the replicative spread of Nipah virus. HUVECs were 682 

infected with NiVMAL for 1 hour at 37 C, washed to remove excess virus, and 20M galectin-1 (black 683 

bars) or buffer (virus only, white bars) added to the media only post-infection. Viral titers (log scale) 684 

were quantified by plaque assay after 0, 12, 24, and 36 hpi. (B) Syncytia formation induced by NiV 685 

infection. HUVECs infected with live NiVMAL or NiVMAL F3 mutant virus in the absence (no galectin-686 

1), presence of galectin-1 added before infection, or presence of galectin-1 added after infection. Cells 687 

were fixed at 24 hpi and stained with DAPI to reveal nuclei. (C) Quantitation of Figure 4C. Nuclei per 688 

syncytium per field were enumerated for the three separate conditions. 689 

690 

Figure 5. Modeling of the glycosylated pre-fusion trimeric NiV-F spike and galectin-1 691 

(A) A model of the glycosylated NiV-F ectodomain in the prefusion state was created with the SWISS-692 

MODEL server [52] using the structure of the parainfluenza virus 5 F protein in the metastable, 693 

prefusion conformation (PDB accession number 2B9B) [53] as a template. The model of the NiV-F 694 

ectodomain is shown in the surface representation, with each protomer colored a different shade of 695 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 35: Timing of galectin-1 exposure differentially modulates Nipah virus

  35

gray. For each protomer, structures of complex-type glycans [54] were placed at N-linked 696 

glycosylation sites F2-F4 and oligomannose-type glycans [55] were placed at F5. Glycans at sites F2 697 

and F4 are colored pink, F3 are colored red, and at F5 are colored green. Residues corresponding to the 698 

putative fusion peptide (residues 103-128) are colored blue. The distance between equivalent F3 699 

glycans is approximately 100Å. (B) Homo-dimeric crystal structure of C2S human galectin-1 in 700 

complex with lactose (PDB accession number 1W60) [56]. Galectin-1 is shown as a surface 701 

representation with the two protomers colored different shades of gray. -lactose is bound to both 702 

protomers and is shown as pink sticks. The distance between equivalent binding sites is approximately 703 

50Å. Structures and models were rendered with pymol (www.pymol.org). 704 

705 

706 

707 

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 36: Timing of galectin-1 exposure differentially modulates Nipah virus

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 37: Timing of galectin-1 exposure differentially modulates Nipah virus

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 38: Timing of galectin-1 exposure differentially modulates Nipah virus

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 39: Timing of galectin-1 exposure differentially modulates Nipah virus

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from

Page 40: Timing of galectin-1 exposure differentially modulates Nipah virus

on April 5, 2018 by guest

http://jvi.asm.org/

Dow

nloaded from