Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2015 Mar;89(5):2520-9.
doi: 10.1128/JVI.02435-14. Epub 2014 Dec 10.

Timing of galectin-1 exposure differentially modulates Nipah virus entry and syncytium formation in endothelial cells

Affiliations

Timing of galectin-1 exposure differentially modulates Nipah virus entry and syncytium formation in endothelial cells

Omai B Garner et al. J Virol. 2015 Mar.

Abstract

Nipah virus (NiV) is a deadly emerging enveloped paramyxovirus that primarily targets human endothelial cells. Endothelial cells express the innate immune effector galectin-1 that we have previously shown can bind to specific N-glycans on the NiV envelope fusion glycoprotein (F). NiV-F mediates fusion of infected endothelial cells into syncytia, resulting in endothelial disruption and hemorrhage. Galectin-1 is an endogenous carbohydrate-binding protein that binds to specific glycans on NiV-F to reduce endothelial cell fusion, an effect that may reduce pathophysiologic sequelae of NiV infection. However, galectins play multiple roles in regulating host-pathogen interactions; for example, galectins can promote attachment of HIV to T cells and macrophages and attachment of HSV-1 to keratinocytes but can also inhibit influenza entry into airway epithelial cells. Using live Nipah virus, in the present study, we demonstrate that galectin-1 can enhance NiV attachment to and infection of primary human endothelial cells by bridging glycans on the viral envelope to host cell glycoproteins. In order to exhibit an enhancing effect, galectin-1 must be present during the initial phase of virus attachment; in contrast, addition of galectin-1 postinfection results in reduced production of progeny virus and syncytium formation. Thus, galectin-1 can have dual and opposing effects on NiV infection of human endothelial cells. While various roles for galectin family members in microbial-host interactions have been described, we report opposing effects of the same galectin family member on a specific virus, with the timing of exposure during the viral life cycle determining the outcome.

Importance: Nipah virus is an emerging pathogen that targets endothelial cells lining blood vessels; the high mortality rate (up to 70%) in Nipah virus infections results from destruction of these cells and resulting catastrophic hemorrhage. Host factors that promote or prevent Nipah virus infection are not well understood. Endogenous human lectins, such as galectin-1, can function as pattern recognition receptors to reduce infection and initiate immune responses; however, lectins can also be exploited by microbes to enhance infection of host cells. We found that galectin-1, which is made by inflamed endothelial cells, can both promote Nipah virus infection of endothelial cells by "bridging" the virus to the cell, as well as reduce production of progeny virus and reduce endothelial cell fusion and damage, depending on timing of galectin-1 exposure. This is the first report of spatiotemporal opposing effects of a host lectin for a virus in one type of host cell.

PubMed Disclaimer

Figures

FIG 1
FIG 1
Galectin-1 enhances infection of NiVpp in a carbohydrate binding-dependent manner. (A) Quantification of galectin-1 enhancement of NiVpp infection. NiVpp was titrated such that viral inoculum that gave luciferase activity in the linear range at 24 hpi was used. NiVpp was added to monolayers of Vero cells, and virus entry in the absence (white bars) or presence (black bars) of increasing concentrations of galectin-1 was quantified by measuring luciferase activity in infected cell lysate 24 hpi as described in Materials and Methods. The data from one of three replicate experiments are presented as mean fold increase (± the standard deviations [SD] of triplicate samples) over the virus-only (i.e., no galectin-1) condition. Significant P value determined by two-way ANOVA. (B) Galectin-1 enhancement of NiVpp infection is carbohydrate binding dependent. NiVpp (white bars) or 10 μM galectin-1 (black bars) was added to Vero cells in the presence of 100 mM lactose or 100 mM sucrose. The data are presented as in panel A as means ± the SD of triplicate samples from one of three replicate experiments is shown. (C) Galectin-1 added during spinoculation (at 4°C) shows an increase in NiVpp infection on Vero cells. NiVpp infection in the absence (white bars) or presence (black bars) of 10 μM galectin-1 added during or after spinoculation, i.e., during or after attachment of virus to cells. The data are means ± the SD of triplicate samples from one of three replicate experiments. Significant P values were determined by using the Student t test.
FIG 2
FIG 2
Galectin-1 enhances infection of NiVpp by bridging the virus to the cell through binding of viral surface and cell surface complex N-glycans. Flow cytometric analysis of L-PHA binding to 293T cells (A) or Vero cells (C) treated with kifunensine (dashed black line) shows loss of cell surface complex N-glycans compared to L-PHA binding to untreated parental cells (bold black outlined histograms). Gray-filled histogram represents negative control with secondary only. (B) Galectin-1 mediated enhancement of NiVpp infection is dependent upon complex N-glycans found on the surface of the virus. Virus infection of Vero cells in the absence (white) or presence of 20 μM galectin-1 (black) is shown for NiVpp bearing three types of envelope glycoproteins: wild-type NiV-F+-G, NiV-F3 mutant (+ wild-type G), or NiV-F+-G devoid of complex N-glycans. The data from one of three replicate experiments are presented as mean fold increases in infection (± the SD of triplicate samples) over the virus-only (no galectin-1) condition. (D) Galectin-1-mediated enhancement of NiVpp infection is dependent upon complex N-glycans on the surface of Vero cells. Virus infection in the absence (white bars) or presence (black bars) of 20 μM galectin-1 is shown for wild-type Vero cells and Vero cells devoid of complex N-glycans. The data (means ± the SD of triplicate samples) from one of three replicate experiments are presented exactly as described for panel B.
FIG 3
FIG 3
Galectin-1 enhances NiVpp and live Nipah virus infection of HUVECs. (A) NiVpp was added to monolayers of HUVECs in the absence (white bar) or presence (black bars) of 20 μM galectin-1 for 1 h, and infection was quantified by measuring Renilla luciferase activity at 24 hpi. The data are presented as the mean fold increases (± the SD of triplicate samples) in infection over the virus-only (no galectin-1) condition. The results from one of three replicate experiments are shown. (B) Quantification of galectin-1 enhancement of recombinant GLuc reporter NiV (rNiV-GLuc) infection of HUVECs. (B) rNiV-GLuc was added to monolayers of HUVECs in the absence (white bar) or the presence (black bars) of the indicated amounts of galectin-1 for 1 h, and infection was quantified by measuring the Gaussia luciferase activity. The data are presented as mean fold increases (± the SD of triplicate samples) in infection over the virus-only (no galectin-1) condition. The results of one of three replicate experiments are shown. (C) Galectin-1-mediated enhancement of rNiV-GLuc infection is dependent upon complex N-glycans on the surface of HUVECs and on the surface of the virus. Virus or HUVECs deficient in complex N-glycans were made in the presence of 20 μM kifunensine (Virus Kif+ and Cells Kif+, respectively). rNiV-GLuc infection in the absence or presence of increasing concentrations of galectin-1 is shown for wild-type rNiV-GLuc and HUVECs (Virus wt/Cells wt, black bars), wild-type rNiV-GLuc and complex N-glycan-deficient HUVECs (Virus wt/Cells Kif+, dark gray bars), complex N-glycan-deficient rNiV-GLuc and wild-type HUVECs (Virus Kif+/Cells wt, light gray bars), and complex N-glycan-deficient rNiV-GLuc and HUVECs (Virus Kif+/Cells Kif+, white bars). The data represent the average fold increases in infection determined as in panel B and are presented as means ± the SD of triplicate samples from one of three replicate experiments.
FIG 4
FIG 4
Galectin-1 can have opposing effects on Nipah virus production and syncytium formation. (A) Effect of galectin-1 added postinfection on the replicative spread of NiV. HUVECs were infected with NiVMAL for 1 h at 37°C and washed to remove excess virus, and 20 μM galectin-1 (black bars) or buffer (virus only, white bars) was added to the medium only postinfection. Virus titers (log scale) were quantified by plaque assay after 0, 12, 24, and 36 hpi. (B) Syncytium formation induced by NiV infection. HUVECs infected with live NiVMAL or NiVMAL F3 mutant virus in the absence (no galectin-1), presence of galectin-1 added before infection, or presence of galectin-1 added after infection. Cells were fixed at 24 hpi and stained with DAPI to reveal nuclei. (C) Quantitation of Fig. 4C. Nuclei per syncytium per field were enumerated for the three separate conditions.
FIG 5
FIG 5
Modeling of the glycosylated prefusion trimeric NiV-F spike and galectin-1. (A) A model of the glycosylated NiV-F ectodomain in the prefusion state was created with the Swiss-Model server (52) using the structure of the parainfluenza virus 5 F protein in the metastable, prefusion conformation (PDB accession number 2B9B) (53) as a template. The model of the NiV-F ectodomain is shown in the surface representation, with each protomer colored a different shade of gray. For each protomer, structures of complex-type glycans (54) were placed at N-linked glycosylation sites F2-F4 and oligomannose-type glycans (55) were placed at F5. Glycans at sites F2 and F4 are pink, F3 are red, and at F5 are green. Residues corresponding to the putative fusion peptide (residues 103 to 128) are colored blue. The distance between equivalent F3 glycans is ca. 100Å. (B) Homodimeric crystal structure of C2S human galectin-1 in complex with lactose (PDB accession number 1W60) (56). Galectin-1 is shown as a surface representation with the two protomers colored different shades of gray. β-Lactose is bound to both protomers and is shown as pink sticks. The distance between equivalent binding sites is ca. 50 Å. Structures and models were rendered with PyMOL (www.pymol.org).

Similar articles

Cited by

References

    1. Hsu VP, Hossain MJ, Parashar UD, Ali MM, Ksiazek TG, Kuzmin I, Neizgoda M, Ruppercht C, Bresee J, Breiman RF. 2004. Nipah virus encephalitis reemergence, Bangladesh. Emerg Infect Dis 10:2082–2087. doi:10.3201/eid1012.040701. - DOI - PMC - PubMed
    1. Stone R. 2011. Epidemiology: breaking the chain in Bangladesh. Science 331:1128–1131. doi:10.1126/science.331.6021.1128. - DOI - PubMed
    1. Nahar N, Mondal UK, Hossain MJ, Uddin Khan MS, Sultana R, Gurley ES, Luby SP. 2013. Piloting the promotion of bamboo skirt barriers to prevent Nipah virus transmission through date palm sap in Bangladesh. Glob Health Promot Int 28:378–386. doi:10.1093/heapro/das020. - DOI - PMC - PubMed
    1. Negrete OA, Levroney EL, Aguilar HC, Bertolotti-Ciarlet A, Nazarian R, Tajyar S, Lee B. 2005. EphrinB2 is the entry receptor for Nipah virus, an emergent deadly paramyxovirus. Nature 436:401–405. doi:10.1038/nature03838. - DOI - PubMed
    1. Negrete OA, Wolf MC, Aguilar HC, Enterlein S, Wang W, Muhlberger E, Su SV, Bertolotti-Ciarlet A, Flick R, Lee B. 2006. Two key residues in ephrinB3 are critical for its use as an alternative receptor for Nipah virus. PLoS Pathog 2:e7. doi:10.1371/journal.ppat.0020007. - DOI - PMC - PubMed

Publication types

LinkOut - more resources

-