Human cytomegalovirus (HCMV) continues to cause serious and often life-threatening disease in those with impaired or underdeveloped immune systems. This virus is able to infect and replicate in a wide range of human cell types, which enables the virus to spread to other individuals in a number of settings. Current antiviral drugs are associated with a significant toxicity profile, and there is no vaccine; these factors highlight a need to identify additional targets for the development of anti-HCMV therapies. We demonstrate for the first time that secretion of a member of the galectin family of proteins, galectin-9 (Gal-9), is upregulated during natural HCMV-reactivated infection and that this soluble cellular protein possesses a potent capacity to block HCMV infection by inhibiting virus entry into the host cell. Our findings support the possibility of harnessing the antiviral properties of Gal-9 to prevent HCMV infection and disease.
KEYWORDS: cytomegalovirus, human herpesviruses, virus-host interactions
ABSTRACT
Human cytomegalovirus (HCMV) is a ubiquitous human herpesvirus. While HCMV infection is generally asymptomatic in the immunocompetent, it can have devastating consequences in those with compromised or underdeveloped immune systems, including transplant recipients and neonates. Galectins are a widely expressed protein family that have been demonstrated to modulate both antiviral immunity and regulate direct host-virus interactions. The potential for galectins to directly modulate HCMV infection has not previously been studied, and our results reveal that galectin-9 (Gal-9) can potently inhibit HCMV infection. Gal-9-mediated inhibition of HCMV was dependent upon its carbohydrate recognition domains and thus dependent on glycan interactions. Temperature shift studies revealed that Gal-9 specific inhibition was mediated primarily at the level of virus-cell fusion and not binding. Additionally, we found that during reactivation of HCMV in hematopoietic stem cell transplant (HSCT) patients soluble Gal-9 is upregulated. This study provides the first evidence for Gal-9 functioning as a potent antiviral defense effector molecule against HCMV infection and identifies it as a potential clinical candidate to restrict HCMV infections.
IMPORTANCE Human cytomegalovirus (HCMV) continues to cause serious and often life-threatening disease in those with impaired or underdeveloped immune systems. This virus is able to infect and replicate in a wide range of human cell types, which enables the virus to spread to other individuals in a number of settings. Current antiviral drugs are associated with a significant toxicity profile, and there is no vaccine; these factors highlight a need to identify additional targets for the development of anti-HCMV therapies. We demonstrate for the first time that secretion of a member of the galectin family of proteins, galectin-9 (Gal-9), is upregulated during natural HCMV-reactivated infection and that this soluble cellular protein possesses a potent capacity to block HCMV infection by inhibiting virus entry into the host cell. Our findings support the possibility of harnessing the antiviral properties of Gal-9 to prevent HCMV infection and disease.
INTRODUCTION
Human cytomegalovirus (HCMV) infections are extremely common, with a global seroprevalence of 45% to 100% (1). Primary HCMV infection is followed by lifelong latency, during which time the virus is maintained in a nonreplicating state but with the potential to periodically reactivate to yield newly replicating virus (2, 3). While HCMV infection is generally asymptomatic in immunocompetent individuals, this virus frequently causes serious and often life-threatening disease in immunocompromised individuals, including AIDS patients and allogeneic hematopoietic stem cell transplant (HSCT) and solid organ transplant recipients, as well as in the immunonaive, including neonates (4, 5). There is no licensed vaccine against HCMV, and so anti-HCMV drugs that target components of the viral replication machinery have been the predominant therapy to treat infection in those at most risk of developing HCMV disease. Unfortunately, these drugs have a significant toxicity profile, which limits their application in a number of the most relevant clinical settings to HCMV disease, and their use is associated with the emergence of resistance (6, 7). The development of anti-HCMV drugs with a safer toxicity profile has been a focus of recent efforts, with letermovir showing promise in a recent clinical trial in HSCT recipients (8). However, there remains a pressing requirement to develop novel antiviral drugs to prevent HCMV disease. Entry of HCMV, which is dependent on membrane fusion and internalization of virions, is initially mediated by virally encoded glycoproteins found in the viral envelope, including gB, gH, and gL (9, 10). This key point in the viral replication cycle represents an attractive target to develop novel antiviral compounds, as exemplified by the potent ability of neutralizing antibodies against HCMV to efficiently inhibit infection (11).
Galectins are a family of cellular proteins characterized by the presence of a carbohydrate recognition domain (CRD), with 15 mammalian galectins identified to date (12). Classically, members of this widely expressed protein family have been studied due to their ability to influence innate and adaptive immune responses (12, 13). Galectins can bind to glycan structures expressed on the surface of both host cells and microorganisms, and as a result, specific roles for galectins in modulating both antiviral immunity and direct host-virus interactions to promote and/or inhibit viral infection and replication have recently emerged (14, 15). More specifically, galectins have been demonstrated to modulate immune responses to several other herpesvirus infections, with both galectin-1 (Gal-1) and galectin-9 (Gal-9) downregulating T cell responses to herpes simplex virus and Epstein-Barr virus both in vivo and in vitro (16–26) and Gal-9 suppressing NK cell responses during murine CMV infection (27). Galectins have been associated with directly enhancing or inhibiting viral infections, including Nipah virus, enterovirus, HIV-1, influenza virus, and dengue virus, in a virus- and cell type-specific manner (28–38); however, the functional role of Gal-9 in directly regulating any herpesvirus infection has not been investigated.
In the current study, we define Gal-9 as a cellular protein that directly inhibits HCMV infection. The results revealed that Gal-9, but not Gal-1, functions as an antiviral lectin, inhibiting HCMV infection by blocking entry into the host cell. Furthermore, we show that soluble Gal-9 concentrations in plasma increase during HCMV reactivation in HSCT recipients, consistent with a role for Gal-9 in natural HCMV infection. Together, this study provides the first evidence that Gal-9 can function as a potent inhibitor of HCMV.
RESULTS
Gal-9 inhibits HCMV infection of multiple cell types.
We sought to assess the functional consequence of Gal-9 upregulation by evaluating the impact of soluble Gal-9 on HCMV infection in permissive cells, given that exogenous galectins can both promote and inhibit a number of other viral infections (14, 15). Previous work from our laboratory has established that Gal-9 is upregulated during HCMV infection and is dependent upon interferon beta (IFN-β) induction of Gal-9 mRNA (39). We therefore sought to assess the functional consequence of Gal-9 upregulation by testing whether soluble Gal-9 could directly regulate HCMV infection. A green fluorescent protein (GFP)-expressing HCMV (Merlin-GFP) was pretreated with recombinant Gal-9 at a range of concentrations (0.25 to 100 nM) for 30 min prior to infection of human foreskin fibroblasts (HFs) at a multiplicity of infection (MOI) of 0.5. The extent of infection was assessed by flow cytometry at 72 hours postinfection (h p.i.), allowing the fold change in the percentage of infected cells to be determined. Representative scatter plots depict the percentage of GFP-positive cells in Merlin-GFP-infected cells compared to mock-infected cells (Fig. 1A). The addition of Gal-9 at each concentration tested (12.5 to 100 nM) significantly reduced the number of GFP-positive cells in a dose-dependent manner (Fig. 1C), with up to 88% inhibition at the highest concentration of Gal-9. In contrast, treatment of HCMV with Gal-1 (12.5 to 100 nM) did not significantly alter infection (Fig. 1B). These results indicate that Gal-9 is able to inhibit HCMV infection of HFs and that this is not a general property of all galectin protein family members.
Gal-9 has been associated with the induction of apoptosis in a number of cell types (40). Therefore, we assessed the ability of Gal-9 to induce cell death in HFs to determine whether the reduction in the number of HCMV-GFP-positive cells was due to the cytotoxicity of target cells, rather than infection per se. HFs were treated with Gal-9 (50 to 400 nM) for 90 min, and cell viability was determined by flow cytometry 24 h later using a LIVE/DEAD indicator dye (Zombie-NIR). In comparison to untreated cells, there was no significant difference in the percentage of live cells following Gal-9 treatment at any concentration tested (Fig. 1D). Thus, the Gal-9-mediated inhibitory effects on HCMV infection are not due to Gal-9-mediated cell death.
To determine whether the capacity of Gal-9 to inhibit HCMV infection was virus strain specific, another strain of HCMV, TB40, was assessed. TB40 was selected as it retains expression of a functional UL128-131A locus (UL128L), whereas the bacterial artificial chromosome (BAC)-derived Merlin recombinant described above has a mutated UL128L rendering it nonfunctional (41). An intact UL128L pentamer complex is nonessential for efficient HCMV infection of HFs; however, it is necessary for infection of a range of key in vivo cellular targets, including epithelial cells (42). A BAC-derived version of TB40 that had been engineered to express GFP (TB40-GFP) was used to monitor infection (43), with expression of GFP detected at 48 h p.i. by flow cytometry. Gal-9 significantly inhibited TB40 infection of HFs in a dose-dependent manner (Fig. 1E), demonstrating that the effect of Gal-9 on virus infection was not limited to the Merlin strain.
As entry mediated by the UL128L pentamer complex into epithelial cells uses a different mechanism from that with entry into HFs (endocytosis rather than direct fusion with the plasma membrane) with some viral glycoproteins key to both processes (44, 45), we expanded our analysis of Gal-9-mediated inhibition of TB40-GFP infection to retinal pigment epithelial 1 (RPE-1) cells. This analysis demonstrated a significant inhibition of TB40-GFP infection of RPE-1 cells following Gal-9 (25 to 200 nM) treatment (Fig. 1F). Taken together, these results indicate that Gal-9 can inhibit infection of HFs with low-passage-number strains of HCMV and that Gal-9 can inhibit HCMV infection of multiple clinically relevant cell types (in both an UL128L-dependent and -independent fashion).
Gal-9 lectin binding is required to inhibit HCMV infection.
To determine whether Gal-9 directly inhibited HCMV infection, Gal-9 was pretreated with a neutralizing (26) or isotype control antibody for 30 min prior to incubation with HCMV (Merlin-GFP) and infection of HFs (MOI, 0.5). Cells were then analyzed by flow cytometry at 72 h p.i. for GFP expression to assess the impact on infection efficiency. We found that pretreatment of Gal-9 with neutralizing antibody resulted in a significant reduction in its ability to restrict HCMV infection (P = 0.041, Fig. 2A), while isotype antibody treatment had no effect. As expected, direct treatment of HCMV virions with either the anti-Gal-9 or isotype antibodies had no impact on infection. Taken together, these results support a mechanism where Gal-9 binds directly to HCMV virions to inhibit their infectivity.
Galectins can mediate their effector functions by binding lectins or via protein-protein interactions. Lectin interactions are mediated by the carbohydrate recognition domain (CRD) binding to β-galactoside-containing carbohydrate ligands (46). To assess whether Gal-9-mediated inhibition of HCMV was dependent on the CRD, Gal-9 was pretreated with the disaccharide lactose (1.25 to 5 mM), which specifically blocks the Gal-9 CRD (38), for 30 min prior to incubation with HCMV (Merlin-GFP). As a control, Gal-9 was pretreated with sucrose (1.25 to 5 mM), which is a disaccharide that lacks the β-galactoside moiety required for binding to the Gal-9 CRD. Flow cytometric analysis of HCMV Merlin-GFP expression demonstrated that lactose, but not sucrose, significantly abrogated Gal-9-mediated inhibition of HCMV infection in a dose-dependent manner, and lactose or sucrose alone had no effect on HCMV infection in the absence of Gal-9 addition (Fig. 2B and C). Thus, the inhibition of HCMV infection by Gal-9 is dependent on carbohydrate binding by this lectin.
Gal-9 inhibits HCMV infection primarily through virion and not host cell binding.
As a number of cell membrane-associated ligands for galectins have been identified (47), we sought to determine whether Gal-9-mediated inhibition of HCMV infection resulted from Gal-9 binding to the host cell. HFs were pretreated with Gal-9 for 30 min, unbound Gal-9 was then removed by washing, and cells were infected with HCMV Merlin-GFP. In parallel, HCMV (Merlin-GFP) was pretreated with Gal-9 as before, and infection was assessed by flow cytometry. Pretreatment of cells with Gal-9 prior to infection had no impact on infection (Fig. 3A), while as expected, Gal-9 treatment of virions significantly decreased it (Fig. 3A). As a further test, we determined whether Gal-9 was able to inhibit HCMV when added to already-infected HFs. Cells were infected at an MOI of 0.5 for 90 min before being treated with Gal-9 for 24 h. In contrast to the inhibitory effects of Gal-9 when pretreating HCMV, incubation of preinfected cells had no significant impact on HCMV-driven GFP expression (Fig. 3B). Therefore, treatment of the virus inoculum prior to infection was at least in part necessary for the observed Gal-9-mediated inhibition of HCMV infection of HFs.
Our conclusions thus far that Gal-9 inhibited HCMV infection were based upon the detection of GFP-tagged HCMV strains where GFP is expressed from a viral IE/E promoter, and therefore, we could not report whether Gal-9 might also have an impact on infectious virus production. To assess this, we assayed the ability of Gal-9 to inhibit HCMV plaque formation. HCMV (Merlin-GFP) was pretreated with Gal-9 before infection of HFs (MOI, 0.001), and following a 90-min incubation, cells were then washed with either phosphate-buffered saline (PBS) (to remove unbound virus) or citrate buffer (to inactivate noninternalized virus). Gal-9 treatment of HCMV followed by PBS washing did result in a modest, but significant, decrease in plaque number at 12 dpi (Fig. 3C). However, washing with the more stringent citrate buffer had a dramatic impact on plaque formation, with a near-complete inhibition seen in Gal-9-treated virus (Fig. 3D).
Gal-9 inhibits HCMV by restricting viral fusion.
To elucidate at what point during viral infection Gal-9 mediated its inhibitory activity, we examined its impact on HCMV immediate early protein expression (48). HCMV (Merlin-GFP) was treated with Gal-9 for 30 min before infection of HFs seeded onto coverslips (MOI, 0.5). Cells were fixed, immunostained for immediate early protein-1 (IE-1) at 24, 48, and 72 h p.i., and visualized using fluorescence microscopy. There were no cells with IE-1 staining in the mock or isotype control samples, whereas IE-1 staining was observed in all samples infected with HCMV at 24 h p.i. (Fig. 4A). Quantification of the proportion of IE-1-positive cells revealed that Gal-9 treatment resulted in a significant decrease in the percentage of IE-1-positive cells at 24, 48, and 72 h p.i. (Fig. 4B), compared to HCMV control infection (24 h p.i, P = 0.048; 48 h p.i., P = 0.016; and 72 h p.i., P = 0.005). Thus, quantification of HCMV infection of HFs by immunofluorescence microscopy provided confirmation of Gal-9-mediated inhibition of infection and also indicated that Gal-9 inhibitory activity occurs early in HCMV infection.
The relative amount of viral DNA in infected cells, with or without Gal-9 treatment of HCMV, was then determined. Noninternalized virions were inactivated once again with citrate buffer, and whole cells were harvested at 4 h p.i. to determine the efficiency of HCMV infection/internalization but prior to the onset of viral genome replication (Fig. 4C). This analysis revealed that Gal-9 treatment significantly decreased the amount of internalized viral DNA compared to HCMV (Merlin-GFP) infection without Gal-9 treatment (n = 3, P = 0.032). Thus, the inhibitory activity of Gal-9 occurs very early in HCMV infection, prior to IE-1 expression and before internalization of the viral genome into the cell.
HCMV entry into HFs is mediated through a multistep process involving attachment of viral glycoproteins with cellular receptors, followed by direct fusion of the viral membrane with the plasma membrane (9, 44). Attachment is largely an energy-independent process, while fusion is energy dependent (49). Thus, to further explore how Gal-9 inhibited HCMV entry, temperature shift studies were performed to enable the separation of the attachment and fusion stages of infection. HCMV (Merlin-GFP) was added to HFs at 4°C for 90 min to allow virus attachment but not fusion (50). Nonadhered virus was then removed with PBS washing, Gal-9 was then added at 37°C for a further 60 min to allow for Gal-9 being present at times postbinding but prefusion, and noninternalized virus was then inactivated with citrate buffer (Fig. 5A). Notably, the addition of Gal-9 during the postbinding fusion step of infection significantly inhibited HCMV infection (Fig. 5B). In addition, lactose-mediated blockade of Gal-9 binding reversed this inhibition, with sucrose showing no impact (Fig. 5C), indicating CRD-dependent restriction of fusion.
Gal-9 secretion is upregulated in response to HCMV in vivo.
To assess the potential clinical relevance that soluble Gal-9 might have on HCMV pathogenesis in people, we assessed secretion of Gal-9 in an HCMV-infected patient cohort. In allogeneic HSCT recipients, HCMV reactivation resulting from donor (D) and/or recipient (R) HCMV seropositivity is associated with adverse outcomes posttransplantation, including severe morbidity and mortality (4). Sequential plasma samples were collected weekly from HSCT patients, and the levels of secreted/soluble Gal-9 protein were quantified. HSCT recipients were prospectively monitored for the development of HCMV reactivation by quantitative PCR (qPCR; Roche Cobas AmpliPrep CMV test). In three HSCT patients who developed posttransplant HCMV reactivation (HCMV-positive reactivators), Gal-9 concentrations were assessed by enzyme-linked immunosorbent assay (ELISA) in plasma samples taken at the initial detection of HCMV reactivation (T1), the peak of reactivation (T2), and during subsequent control of reactivation (DNAemia) following antiviral drug therapy (T3) (Table 1). In parallel, the plasma concentration of Gal-9 was assessed at matched days posttransplant from three HSCT recipients where both the donor and recipient were HCMV seronegative (D−/R−), as well as from three HSCT recipients where either the donor and/or recipient was HCMV seropositive but in whom no reactivation was detected (HCMV-positive nonreactivators).
TABLE 1.
Patient no. by group |
Serostatusa | Days posttransplant |
||
---|---|---|---|---|
Group | Start of reactivation (T1) |
Peak of reactivation (T2) |
Control of reactivation (T3) |
|
HCMV negative | ||||
1 | D−, R− | 38 | 52 | 80 |
2 | D−, R− | 39 | 53 | 83 |
3 | D−, R− | 38 | 52 | 80 |
HCMV-positive nonreactivators | ||||
4 | D+, R+ | 40 | 54 | 82 |
5 | D+, R− | 38 | 52 | 94 |
6 | D+, R− | 39 | 53 | 83 |
HCMV-positive reactivators | ||||
7 | D−, R+ | 39 | 62 | 81 |
8 | D−, R+ | 31 | 45 | 87 |
9 | D+, R+ | 44 | 51 | 86 |
D+/− and R+/− refer to the serostatus of the donor (D) and recipient (R).
Across the three time points examined, there was no significant change in plasma Gal-9 concentration in either of the two groups of HSCT recipients experiencing no HCMV reactivation (Fig. 6). In contrast, HSCT recipients who developed HCMV reactivation exhibited a significant increase in plasma Gal-9 levels which was associated with the peak of HCMV reactivation (T2, mean day 53 posttransplant). At this time point, the plasma Gal-9 concentration was significantly higher in patients with HCMV reactivation (24.4 ng/ml) than in both HCMV-negative nonreactivators (mean, 8.8 ng/ml; P = 0.0001) and HCMV-positive nonreactivators (mean, 14.1 ng/ml; P = 0.006). There was also a significant difference in Gal-9 plasma levels for HCMV-seropositive reactivators at the peak of reactivation (T2) compared to both its initiation (T1, P = 0.0015) and eventual control (T3, P = 0.0004). The concentration of Gal-9 at T3 in HCMV-seropositive reactivators was comparable to that detected at initial detection of reactivation (T1) and similar in patients with or without HCMV reactivation at these times. We also assessed the secretion of Gal-9 in response to in vitro HCMV infection of HFs. HCMV infection of HFs resulted in a significant increase in secreted Gal-9 at 48, 72, and 96 h p.i. compared to mock (Fig. 7A) and was recapitulated with interferon treatment alone, as expected (Fig. 7B) (39). Taken together, our results indicate that soluble Gal-9 has a high potential to regulate HCMV infection both in vitro and in vivo in the context of inflammatory responses and incidences of high viremia in immunosuppressed patients.
DISCUSSION
A major focus of this study aimed to examine the potential for soluble galectins to directly modulate HCMV infection. We provide the first evidence that Gal-9 has antiviral activity during HCMV infection. This inhibitory effect was not exhibited by another galectin family member, Gal-1, which has previously been demonstrated to enhance HIV-1 infection of T cells and macrophages (28–30) and promote interactions between influenza A virus and its target cell (31); in contrast, it inhibits infection of dengue virus (32), enterovirus (33), human T lymphotropic virus (34), and Nipah virus (35, 36). In addition to its role in inhibiting HCMV, Gal-9 has also been identified to modulate a wide range of other viruses. In the case of HIV infection, Gal-9 has been reported to both inhibit and promote viral infection of T cells based on their activation/glycosylation status. CRD-mediated interactions between Gal-9 and protein disulfide isomerase (PDI) specifically retained this glycoprotein on the surface of nonstimulated T cells, resulting in enhanced HIV infection (38). Conversely, Gal-9 interactions with Tim-3 on activated T cells reduced infection, resulting from Gal-9-mediated downregulation of HIV receptors on the T cell surface, including CCR5, CCR4, and alpha-4-beta-7 integrins (37). Furthermore, Gal-9 treatment of T cells latently infected with HIV resulted in reactivation of this virus (51). Collectively, these experiments demonstrate the pleiotropic effects Gal-9 can mediate throughout the life cycle of chronic viral infections. To our knowledge, the data presented here are the first to show that Gal-9 is directly involved in inhibiting any herpesvirus infection. Thus, studies to investigate whether Gal-9 can be used as an antiviral lectin in vivo and whether Gal-9 provides a barrier to infection at different stages of clinical infection (during latency and reactivation) may provide an important basis to limit HCMV disease.
Gal-9-mediated inhibition of HCMV infection appeared to be at least partially dependent on binding to the virion rather than interacting with cellular ligands, as inhibitory activity of Gal-9 was most potent when added to the virus inoculum prior to infection. Furthermore, Gal-9 was not antiviral when added postinfection, suggesting that Gal-9 does not impact postentry events during HCMV infection, at least not in the cell types tested in our studies. Gal-9 treatment of HCMV resulted in a significant reduction in immediate early (immunofluorescence assay) and late (flow cytometric analysis) gene expression, productively infected cells (plaque assay), and viral genomes entering cells (viral genome DNA qPCR). Thus, Gal-9 appears to block HCMV infection at a very early stage in the infection cycle.
Herpesvirus entry is a stepwise process, with initial virion binding mediated through heparin sulfate proteoglycans before engagement of surface entry receptors by viral glycoproteins leading to membrane fusion and entry (52). Our data show that Gal-9 acts during viral entry, blocking viral fusion, therefore inhibiting HCMV infection. Gal-9 treatment of HFs that were prebound with HCMV indicated that Gal-9 could prevent fusion of virions already attached to target cells. Furthermore, blocking of Gal-9 lectin binding activity was sufficient to prevent it from inhibiting fusion. The potential for Gal-9 to function as a lectin to mediate this inhibition indicates that HCMV entry-associated glycoproteins are key candidates for Gal-9 binding responsible for the phenotype observed. A number of glycoproteins conserved among members of the herpesviridae family, including gB, gH, and gL, are known to be associated with viral entry (9, 10, 53). In addition, HCMV encodes a number of unique glycoproteins, including UL128, UL130, and UL131A, that are also entry associated and determine viral tropism (42, 54, 55), with these proteins known to mutate during passage in primary fibroblasts (56). UL128, UL130, and UL131A associate with gH and gL to form a pentamer complex essential for infection of endothelial, epithelial, and monocytic cells (57, 58). In the absence of this pentamer complex, gH and gL complex with gO, which limits cellular tropism to fibroblast cells (59–61). To address the different pathways of HCMV entry, we used two HCMV variants that expressed either the gH-gL-gO complex (Merlin) or a functional pentamer (TB40) to enable infection of HF and RPE-1 cells, respectively. As the addition of Gal-9 inhibited infection of both fibroblasts and epithelial cells, glycoproteins gH/gL and gB are potential targets for Gal-9 binding. However, it is important to note that clinical strains of HCMV possess the capacity to encode greater than 25 glycoproteins (62, 63), the majority of which have unknown contribution to viral binding and entry. Thus, a broad-based analysis to identify specific HCMV glycoproteins that directly interact with Gal-9 will be an important focus of future studies.
Current anti-HCMV antiviral drugs in routine use (ganciclovir, foscarnet, and cidofivir) act as inhibitors of the viral DNA polymerase and are associated with adverse effects, including neutropenia and nephrotoxicity, with their use limited in certain situations (e.g., pregnancy) (64). In addition, viral mutants commonly emerge during these drug treatments that exhibit resistance to each of the current antiviral regimes predominantly due to mutations in the UL54-encoded DNA polymerase and the UL97 protein kinase (65). Thus, there is a significant requirement for novel treatments to target HCMV infection. The development of entry inhibitors has been proposed as a novel therapeutic strategy for treatment of viral infections. A number of entry inhibitors have recently been proposed as novel antiviral therapeutics, including inhibitors of attachment, such as sulfated polysaccharides (66), lactoferrin (67), and peptide-derivatized dendrimers (68), and entry inhibitors, including CFI02 (69), β-peptides (70), and CpG octadecaneuropeptides (ODNs) (71). In addition, soluble forms of host proteins that promote HCMV entry, e.g., CD90 and platelet-derived growth factor receptor alpha, have also demonstrated anti-HCMV activity (72–74).
Aside from neutralizing antibodies, few endogenous soluble factors have been identified that can directly inhibit HCMV entry, with the well-established antiviral activity of soluble cytokines, such as interferons, dependent on the activation of host gene expression to mediate their effects. We have demonstrated that Gal-9 can potently inhibit HCMV infection and therefore represents an attractive scaffold to develop antiviral compounds. Developing antiviral inhibitors based on host-encoded molecules that inhibit entry has a number of potential advantages, including a much lower possibility of an immunogenic response, development of resistance mutations in circulating strains that potentially render such variants less robust in their ability to infect cells, and that such molecules could be used in combination with currently employed antivirals due to an alternative mechanism of action. In addition, most neutralizing antibodies specifically target either UL128L-dependent or -independent mechanisms of entry (11); however, we demonstrate here that Gal-9 can inhibit HCMV infection into both fibroblasts and epithelial cells, indicating a broad spectrum of activity.
Our analyses also revealed that Gal-9 is significantly upregulated during HCMV infection both in vitro and in naturally infected individuals. We found plasma Gal-9 levels to be significantly higher in HSCT patients who underwent HCMV reactivation, and that the peak in circulating Gal-9 coincided with the peak in viral reactivation before a reduction in Gal-9 levels associated with concomitant resolution of infection, indicating that Gal-9 may play a role in HCMV pathogenesis, at least in the transplant setting. We hypothesize that increased Gal-9 levels in patients with reactivated HCMV infection are a consequence of IFN production, as both type I and II IFNs are known potent drivers of Gal-9 expression (39, 75) and can be produced directly in response to HCMV infection (39) or from immune effector cells responding to infection (76). The precise cellular source of secreted Gal-9 protein in natural HCMV infection remains to be determined, although our previous detection of upregulated Gal-9 transcript levels in peripheral blood mononuclear cells (PBMCs) from stem cell transplant recipients with HCMV reactivation (39) suggests that one or more PBMC subsets contribute to elevated Gal-9 protein in plasma.
Elevated soluble Gal-9 in plasma of individuals infected with a number of other chronic and acute viruses has also been reported, including HIV, influenza A virus, hepatitis C virus, hepatitis B virus, herpes simplex virus, Epstein-Barr virus, and dengue virus (26, 77–87), indicating an important and potentially broadly relevant role for Gal-9 in viral pathogenesis. Studies with a number of these viruses also identified a similar trend of Gal-9 returning to levels comparable to uninfected individuals following treatment or resolution of viral infection, including HIV, influenza A virus, and dengue virus (77, 78, 81). In the case of HCMV infection, we have shown that IFN-β induces Gal-9 expression and secretion (39), highlighting it as an antiviral interferon-stimulated gene (ISG). This concept is supported by the recent characterization of the “interferome” that sought to identify key genes upregulated in response to type I interferons across species (88). Gal-9 was defined as a core ISG conserved across all mammalian species tested, indicating its potential as an important conserved antiviral protein. It is also clear that Gal-9 can play a key role in promoting intracellular signaling events through immune cell surface receptors, including 4-1BB, DR3, and Tim-3 (89–91); therefore, elevated soluble Gal-9 levels detected during HCMV reactivation could also regulate immune effector function, which is key to the control of HCMV reactivation.
Recombinant Gal-9 has been demonstrated to have therapeutic potential for treatment of a range of disorders in mouse models, including allograft rejection and graft-versus-host disease (92). In turn, endogenous Gal-9 is required for the agonist activity of antibody-based drugs targeting members of the tumor necrosis factor receptor (TNFR) superfamily (89). This is reminiscent of similar requirements for Fc receptor (Fc-R) binding by agonist antibodies in several disease models, suggesting that higher-order oligomerization of cell surface receptors mediated by Gal-9 and/or Fc-Rs have the potential to regulate multiple aspects of antiviral immune cell responses. Like all herpesviruses, HCMV establishes lifelong latency; therefore, the development of antiviral therapies that target viral entry offers a promising alternative to the existing therapeutic agents available.
MATERIALS AND METHODS
Plasma from HSCT patients.
Plasma was collected from whole-blood samples drawn from adult allogeneic HSCT recipients (Westmead Hospital, Australia) and stored at −80°C. Samples were collected from each recipient at weekly intervals for the first 100 days posttransplant, with HCMV serostatuses of recipients and donors assessed in the clinic prior to transplantation. Samples were collected from both HCMV donor (D)-/recipient (R)-seronegative (D−/R−) and HCMV-seropositive (D−/R+, D+/R−, D+/R+) HSCT recipients (Table 1). Recipients were prospectively monitored for the development of HCMV reactivation by qPCR (Roche Cobas AmpliPrep CMV test). HCMV reactivation was defined as at least 150 copies of HCMV DNA per 1 ml plasma in at least two consecutive samples. All seropositive recipients (R+) were administered ganciclovir or valganciclovir prior to transplantation, and all patients with HCMV reactivation selected for this study were treated with ganciclovir or valganciclovir in the case of posttransplant HCMV reactivation. Time points posttransplant for the assessment of Gal-9 by ELISA were determined based on the three patients who developed HCMV reactivation posttransplant (HCMV-positive reactivators), with plasma Gal-9 concentrations assessed at the initial detection of HCMV reactivation (T1, mean 38 days posttransplant), the peak of reactivation (T2, mean 53 days posttransplant), and the control of reactivation (T3, mean 84 days posttransplant). Control of reactivation was defined as the first time point postreactivation at which plasma HCMV DNA returned to a level below 150 copies per ml of plasma. Plasma samples for nonreactivating groups (HCMV negative and HCMV-positive nonreactivators) were assessed at matched days posttransplant to the HCMV reactivation group. Plasma Gal-9 concentrations were determined by ELISA (R&D Systems) according to the manufacturer’s instructions. This involved a 10-fold dilution of plasma samples in the provided calibration diluent (RD6-35). This dilution was within the recommended dilution range and ensured that all concentrations detected were within the detection limit of the kit used (0.16 to 10 ng/ml).
Cell culture and virus stocks.
Human foreskin fibroblasts (HFs) and retinal pigment epithelial 1 (RPE-1) cells were obtained from the ATCC and grown at 37°C and 5% CO2 in Dulbecco’s modified Eagle medium (DMEM) supplemented with 10% fetal calf serum (FCS) and penicillin-streptomycin (100 units/ml). The UL32 GFP-expressing variant of the low-passage-number clinical isolate of Merlin was generated from a bacterial artificial chromosome (BAC) (41). The GFP-expressing TB40 used was also BAC derived (43). Virus stocks were propagated in HFs, and cell culture supernatants from virus-infected cells were centrifuged at 845 × g for 10 min to remove cellular debris before ultracentrifugation at 21,875 × g for 2 h to pellet virus (Sorvall WX+ ultracentrifuge; Thermo Fisher Scientific). Virus pellets were resuspended in fresh supplemented DMEM and stored at −80°C. Viral titers were determined by a plaque assay. Briefly, 1.5 × 105 HFs were infected in six-well plates and overlaid with a 1:1 mixture of 2× Eagle’s minimal essential medium (EMEM) supplemented with l-glutamine (4 mM), 20% FCS, and penicillin-streptomycin (200 units/ml), combined with 2% Avicel. Plates were incubated at 37°C and 5% CO2 for 12 days, plates were then washed with PBS, and plaques were enumerated under inverted light microscope (Axio, Zeiss).
Viral infections and galectin treatments.
HFs and RPE-1 cells were seeded in a 12-well plate (Corning Costar; Sigma-Aldrich) at 7.5 × 104 and 1 × 105 cells/well, respectively. For testing the inhibitory effects of galectins on HCMV, recombinant Gal-1 (R&D Systems) or Gal-9 (R&D Systems or BioLegend) was diluted in serum-free DMEM and mixed with HCMV at an appropriate MOI (0.5 for HFs, 1 for RPE-1) for 30 min at room temperature. The virus-galectin mixture was added to cells for 90 min at 37°C. Virus-containing medium was removed, cells were washed with PBS, and fresh supplemented DMEM was added to each well.
Flow cytometry.
For flow cytometric analysis of GFP expression, cells were detached with trypsin-EDTA at 72 h p.i. for Merlin-GFP infected HFs and 48 h p.i. for all TB40-GFP infected cells. Cells were washed with flow cytometry buffer (PBS with 1% FCS and 10 mM EDTA) and fixed with 1% paraformaldehyde (PFA). Flow cytometry was performed (LSRFortessa X-20; BD Biosciences), and data were analyzed with FlowJo (version 10; Tree Star, Inc., Ashland, OR). GFP expression data were normalized as the fold change in the percentage of infected cells (GFP positive) compared to untreated HCMV.
Cell viability.
HFs were seeded in a 12-well plate and treated with various concentrations of Gal-9 (25 to 400 nM) in serum-free medium for 90 min at 37°C. Medium was removed, cells were washed in PBS, and supplemented medium was added. Cell viability was assessed at 24 h posttreatment, and cells were detached by trypsin-EDTA, washed in PBS, and Zombie-NIR stained (BioLegend). Cells were washed in flow cytometry buffer and fixed in 1% PFA. Flow cytometry was performed with an LSRFortessa X-20 analyzer (BD Biosciences) and data analyzed with FlowJo (version 10; Tree Star, Inc., Ashland, OR). The percentage of live cells was determined by gating on the Zombie-NIR-negative population.
Immunofluorescence staining and microscopy.
HFs were seeded onto coverslips (5 × 104 cells/coverslip), after which cells were treated or infected. At 24, 48, and 72 h p.i., cells were washed with PBS and fixed with 4% PFA. Cells were permeabilized in 0.1% Triton X-100 (in PBS) blocked with 20% normal donkey serum (in PBS) before being stained with anti-IE1 primary antibody (clone 8B1.2; Millipore) or isotype control (mIgG2a). Primary antibodies were detected by staining with anti-mouse IgG-Alexa Fluor 549 (Invitrogen), and cells were counterstained with DAPI (Thermo Fisher Scientific) before imaging on a wide-field fluorescent light microscope (Zeiss). The percentage of infected cells was determined by quantification of IE1-postivive cells and total number of cells (DAPI positive) per image using ImageJ, with the mean taken across five images per replicate.
Quantitative PCR of viral genomes.
Gal-9-treated/infected HFs were harvested at 4 h p.i. by scraping and total DNA extracted using a Qiagen DNA extraction kit. qPCR was performed using Brilliant II SYBR green (Agilent Technologies) analyzed on a Roche LightCycler 480 at 10 min and 95°C for denaturation and then 50 amplification cycles of 15 s at 95°C and 45 s at 60°C; finally, melt curve data were generated through 1 min at 95°C, 30 s at 50°C, and 30 s at 95°C. Primers used targeted human albumin (ALB-F, 5′-TTTGCAGATGTCAGTGAAAGAGA-3′; ALB-R, 5′-TGGGGAGGCTATAGAAAATAAGG-3′) and HCMV US28 (US28-F, 5′-AGAACTCATGCTCGGTGCTT-3′; US28-R, 5′-GCTGGAGATCCATTTGAGGA-3′). The US28 primers were designed to amplify a conserved region in the US28 gene sequence (93). Viral DNA (US28) was normalized to cellular DNA (albumin) and relative changes from untreated HCMV determined by the −2ΔΔCT method.
Antibody and lactose blocking of Gal-9.
Gal-9 (50 nM) was diluted in serum-free medium and blocked by the addition of one of the following for 30 min: anti-Gal-9 monoclonal antibody (40 ng/ml, clone 9M1-3, BioLegend), isotype control (40 ng/ml, mIgG1), lactose (1.25 to 5 mM; Sigma), or sucrose (1.25 to 5 mM; Sigma). Merlin-GFP was then added for a further 30 min before addition to HFs at an MOI of 0.5. The fold change in the percentage of infected cells was assessed by flow cytometric analysis of GFP expression at 72 h p.i., as detailed above.
Gal-9 pretreatment of HCMV and HFs and Gal-9 treatment postinfection.
To pretreat virus inoculum, Gal-9 (50 nM) was diluted in serum-free medium and added to Merlin-GFP for 30 min at room temperature. The virus–Gal-9 mixture was added to HFs at an MOI of 0.5 for 90 min at 37°C. Alternatively, to pretreat cells, Gal-9 (50 nM) was added to HFs in serum-free medium for 30 min at room temperature. Gal-9-containing medium was replaced with Merlin-GFP in serum-free medium, at an MOI of 0.5, for 90 min at 37°C. Following infection, cells were washed with PBS and fresh supplemented medium added. For postinfection Gal-9 treatment, HFs were infected with Merlin-GFP at an MOI of 0.5 in serum-free medium for 90 min at 37°C, and cells were washed in PBS and serum-free medium containing Gal-9 (50 nM) added for 24 h p.i., followed by replacement with fresh supplemented medium. Merlin-GFP infection in which no Gal-9 was introduced served as the control in all experiments. The fold change in the percentage of infected cells was assessed by flow cytometric analysis of GFP expression at 72 h p.i., as detailed above.
Plaque reduction assay.
HFs were seeded into six-well plates at a density of 1.5 × 105 cells/well. Merlin-GFP was untreated or treated with Gal-9 (100 nM) for 30 min before addition to HFs at an MOI of 0.001 (150 PFU) in duplicate. Following 90 min of infection at 37°C, cells were washed with either PBS to remove unbound virus or citrate buffer (40 mM citric acid, 10 mM KCl, 135 mM NaCl [pH 3]) for 3 min at room temperature to inactivate noninternalized virus, followed by PBS washing in triplicate. Cells were overlaid with a 1:1 mixture of 2× Eagle’s minimal essential medium (2× EMEM) supplemented with l-glutamine (4 mM), 20% FCS, and penicillin-streptomycin (200 units per ml), combined with 2% Avicel. Plates were incubated at 37°C and 5% CO2 for 12 days and washed with PBS, and plaques were enumerated using an inverted light microscope (Axio; Zeiss).
Gal-9 inhibition of fusion assay.
Merlin-GFP was added to HFs at an MOI of 5 in serum-free medium for 90 min at 4°C to allow the virus to bind to cells. The viral inoculum was removed with PBS washing and replaced with serum-free medium with or without Gal-9 (50 nM). In some instances, Gal-9 was preblocked with either lactose (5 mM) or sucrose (5 mM) for 30 min, as indicated in Fig. 5C. Cells were transferred to 37°C for a further 60 min to allow virus fusion. Cells were washed in citrate buffer for 3 min at room temperature to inactivate any noninternalized virus, followed by PBS washing in triplicate (50). Cells were cultured in supplemented medium for 72 h and then harvested for flow cytometric analysis to determine the fold change in the percentage of infected cells (GFP positive).
Statistical analysis.
Statistical analyses were performed using the GraphPad Prism software. Statistically significant differences among groups were assessed using analysis of variance (ANOVA) (Bonferroni Dunn test). Statistically significant differences between pairs were assessed using Student’s t test. In all analyses, a P value of <0.05 was considered statistically significant. Data are presented as means and standard errors of the mean (SEM).
Ethics statement.
This study was approved by the institutional ethics committees of the Sydney West Area Health Service and the University of Sydney. All donors were adults and provided written informed consent in accordance with the Declaration of Helsinki.
ACKNOWLEDGMENTS
We thank Richard Stanton and Gavin Wilkinson (Cardiff University, Cardiff, Wales) for the GFP-tagged HCMV strain Merlin BAC-derived virus, and Eain Murphy (Blumberg Institute, PA, USA) for the GFP-tagged HCMV strain TB40 BAC-derived virus. We also acknowledge the assistance of the Sydney Cytometry Facility, The University of Sydney and the Advanced Microscopy Facility, The University of Sydney.
REFERENCES
- 1.Cannon MJ, Schmid DS, Hyde TB. 2010. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol 20:202–213. doi: 10.1002/rmv.655. [DOI] [PubMed] [Google Scholar]
- 2.Slobedman B, Cao JZ, Avdic S, Webster B, McAllery S, Cheung AK, Tan JC, Abendroth A. 2010. Human cytomegalovirus latent infection and associated viral gene expression. Future Microbiol 5:883–900. doi: 10.2217/fmb.10.58. [DOI] [PubMed] [Google Scholar]
- 3.Hahn G, Jores R, Mocarski ES. 1998. Cytomegalovirus remains latent in a common precursor of dendritic and myeloid cells. Proc Natl Acad Sci U S A 95:3937–3942. doi: 10.1073/pnas.95.7.3937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ariza-Heredia EJ, Nesher L, Chemaly RF. 2014. Cytomegalovirus diseases after hematopoietic stem cell transplantation: a mini-review. Cancer Lett 342:1–8. doi: 10.1016/j.canlet.2013.09.004. [DOI] [PubMed] [Google Scholar]
- 5.Cannon MJ. 2009. Congenital cytomegalovirus (CMV) epidemiology and awareness. J Clin Virol 46(Suppl 4):S6–10. doi: 10.1016/j.jcv.2009.09.002. [DOI] [PubMed] [Google Scholar]
- 6.Lurain NS, Chou S. 2010. Antiviral drug resistance of human cytomegalovirus. Clin Microbiol Rev 23:689–712. doi: 10.1128/CMR.00009-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Campos AB, Ribeiro J, Boutolleau D, Sousa H. 2016. Human cytomegalovirus antiviral drug resistance in hematopoietic stem cell transplantation: current state of the art. Rev Med Virol 26:161–182. doi: 10.1002/rmv.1873. [DOI] [PubMed] [Google Scholar]
- 8.Marty FM, Ljungman P, Chemaly RF, Maertens J, Dadwal SS, Duarte RF, Haider S, Ullmann AJ, Katayama Y, Brown J, Mullane KM, Boeckh M, Blumberg EA, Einsele H, Snydman DR, Kanda Y, DiNubile MJ, Teal VL, Wan H, Murata Y, Kartsonis NA, Leavitt RY, Badshah C. 2017. Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation. N Engl J Med 377:2433–2444. doi: 10.1056/NEJMoa1706640. [DOI] [PubMed] [Google Scholar]
- 9.Vanarsdall AL, Ryckman BJ, Chase MC, Johnson DC. 2008. Human cytomegalovirus glycoproteins gB and gH/gL mediate epithelial cell-cell fusion when expressed either in cis or in trans. J Virol 82:11837–11850. doi: 10.1128/JVI.01623-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sathiyamoorthy K, Chen J, Longnecker R, Jardetzky TS. 2017. The COMPLEXity in herpesvirus entry. Curr Opin Virol 24:97–104. doi: 10.1016/j.coviro.2017.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cui XH, Freed DC, Wang D, Qiu P, Li FS, Fu TM, Kauvar LM, McVoy MA. 2017. Impact of antibodies and strain polymorphisms on cytomegalovirus entry and spread in fibroblasts and epithelial cells. J Virol 91:e01650-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Rabinovich GA, Liu FT, Hirashima M, Anderson A. 2007. An emerging role for galectins in tuning the immune response: lessons from experimental models of inflammatory disease, autoimmunity and cancer. Scand J Immunol 66:143–158. doi: 10.1111/j.1365-3083.2007.01986.x. [DOI] [PubMed] [Google Scholar]
- 13.Rabinovich GA, Toscano MA. 2009. Turning ‘sweet’ on immunity: galectin-glycan interactions in immune tolerance and inflammation. Nat Rev Immunol 9:338–352. doi: 10.1038/nri2536. [DOI] [PubMed] [Google Scholar]
- 14.Vasta GR. 2009. Roles of galectins in infection. Nat Rev Microbiol 7:424–438. doi: 10.1038/nrmicro2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Merani S, Chen W, Elahi S. 2015. The bitter side of sweet: the role of galectin-9 in immunopathogenesis of viral infections. Rev Med Virol 25:175–186. doi: 10.1002/rmv.1832. [DOI] [PubMed] [Google Scholar]
- 16.Sloan DD, Zahariadis G, Posavad CM, Pate NT, Kussick SJ, Jerome KR. 2003. CTL are inactivated by herpes simplex virus-infected cells expressing a viral protein kinase. J Immunol 171:6733–6741. doi: 10.4049/jimmunol.171.12.6733. [DOI] [PubMed] [Google Scholar]
- 17.Gonzalez MI, Rubinstein N, Ilarregui JM, Toscano MA, Sanjuan NA, Rabinovich GA. 2005. Regulated expression of galectin-1 after in vitro productive infection with herpes simplex virus type 1: implications for T cell apoptosis. Int J Immunopathol Pharmacol 18:615–623. doi: 10.1177/039463200501800402. [DOI] [PubMed] [Google Scholar]
- 18.Rajasagi NK, Suryawanshi A, Sehrawat S, Reddy PB, Mulik S, Hirashima M, Rouse BT. 2012. Galectin-1 reduces the severity of herpes simplex virus-induced ocular immunopathological lesions. J Immunol 188:4631–4643. doi: 10.4049/jimmunol.1103063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sehrawat S, Suryawanshi A, Hirashima M, Rouse BT. 2009. Role of Tim-3/galectin-9 inhibitory interaction in viral-induced immunopathology: shifting the balance toward regulators. J Immunol 182:3191–3201. doi: 10.4049/jimmunol.0803673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shim JA, Park S, Lee ES, Niki T, Hirashima M, Sohn S. 2012. Galectin-9 ameliorates herpes simplex virus-induced inflammation through apoptosis. Immunobiology 217:657–666. doi: 10.1016/j.imbio.2011.11.002. [DOI] [PubMed] [Google Scholar]
- 21.Sehrawat S, Reddy PB, Rajasagi N, Suryawanshi A, Hirashima M, Rouse BT. 2010. Galectin-9/TIM-3 interaction regulates virus-specific primary and memory CD8 T cell response. PLoS Pathog 6:e1000882. doi: 10.1371/journal.ppat.1000882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Reddy PB, Sehrawat S, Suryawanshi A, Rajasagi NK, Mulik S, Hirashima M, Rouse BT. 2011. Influence of galectin-9/Tim-3 interaction on herpes simplex virus-1 latency. J Immunol 187:5745–5755. doi: 10.4049/jimmunol.1102105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gandhi MK, Moll G, Smith C, Dua U, Lambley E, Ramuz O, Gill D, Marlton P, Seymour JF, Khanna R. 2007. Galectin-1 mediated suppression of Epstein-Barr virus specific T-cell immunity in classic Hodgkin lymphoma. Blood 110:1326–1329. doi: 10.1182/blood-2007-01-066100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Smith C, Beagley L, Khanna R. 2009. Acquisition of polyfunctionality by Epstein-Barr virus-specific CD8+ T cells correlates with increased resistance to galectin-1-mediated suppression. J Virol 83:6192–6198. doi: 10.1128/JVI.00239-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ouyang J, Juszczynski P, Rodig SJ, Green MR, O’Donnell E, Currie T, Armant M, Takeyama K, Monti S, Rabinovich GA, Ritz J, Kutok JL, Shipp MA. 2011. Viral induction and targeted inhibition of galectin-1 in EBV+ posttransplant lymphoproliferative disorders. Blood 117:4315–4322. doi: 10.1182/blood-2010-11-320481. [DOI] [PubMed] [Google Scholar]
- 26.Klibi J, Niki T, Riedel A, Pioche-Durieu C, Souquere S, Rubinstein E, Le Moulec S, Guigay J, Hirashima M, Guemira F, Adhikary D, Mautner J, Busson P. 2009. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood 113:1957–1966. doi: 10.1182/blood-2008-02-142596. [DOI] [PubMed] [Google Scholar]
- 27.Golden-Mason L, McMahan RH, Strong M, Reisdorph R, Mahaffey S, Palmer BE, Cheng L, Kulesza C, Hirashima M, Niki T, Rosen HR. 2013. Galectin-9 functionally impairs natural killer cells in humans and mice. J Virol 87:4835–4845. doi: 10.1128/JVI.01085-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ouellet M, Mercier S, Pelletier I, Bounou S, Roy J, Hirabayashi J, Sato S, Tremblay MJ. 2005. Galectin-1 acts as a soluble host factor that promotes HIV-1 infectivity through stabilization of virus attachment to host cells. J Immunol 174:4120–4126. doi: 10.4049/jimmunol.174.7.4120. [DOI] [PubMed] [Google Scholar]
- 29.St.-Pierre C, Manya H, Ouellet M, Clark GF, Endo T, Tremblay MJ, Sato S. 2011. Host-soluble galectin-1 promotes HIV-1 replication through a direct interaction with glycans of viral gp120 and host CD4. J Virol 85:11742–11751. doi: 10.1128/JVI.05351-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.St.-Pierre C, Ouellet M, Giguere D, Ohtake R, Roy R, Sato S, Tremblay MJ. 2012. Galectin-1-specific inhibitors as a new class of compounds to treat HIV-1 infection. Antimicrob Agents Chemother 56:154–162. doi: 10.1128/AAC.05595-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chernyy ES, Rapoport EM, Andre S, Kaltner H, Gabius HJ, Bovin NV. 2011. Galectins promote the interaction of influenza virus with its target cell. Biochemistry (Mosc) 76:958–967. doi: 10.1134/S0006297911080128. [DOI] [PubMed] [Google Scholar]
- 32.Toledo KA, Fermino ML, Andrade CDC, Riul TB, Alves RT, Muller VDM, Russo RR, Stowell SR, Cummings RD, Aquino VH, Dias-Baruffi M. 2014. Galectin-1 exerts inhibitory effects during DENV-1 infection. PLoS One 9:e112474. doi: 10.1371/journal.pone.0112474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lee PH, Liu CM, Ho TS, Tsai YC, Lin C.C, Wang YF, Chen YL, Yu CK, Wang SM, Liu CC, Shiau AL, Lei HY, Chang CP. 2015. Enterovirus 71 virion-associated galectin-1 facilitates viral replication and stability. PLoS One 10:e0116278. doi: 10.1371/journal.pone.0116278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gauthier S, Pelletier I, Ouellet M, Vargas A, Tremblay MJ, Sato S, Barbeau B. 2008. Induction of galectin-1 expression by HTLV-I Tax and its impact on HTLV-I infectivity. Retrovirology 5:105. doi: 10.1186/1742-4690-5-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Levroney EL, Aguilar HC, Fulcher JA, Kohatsu L, Pace KE, Pang M, Gurney KB, Baum LG, Lee B. 2005. Novel innate immune functions for galectin-1: galectin-1 inhibits cell fusion by Nipah virus envelope glycoproteins and augments dendritic cell secretion of proinflammatory cytokines. J Immunol 175:413–420. doi: 10.4049/jimmunol.175.1.413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Garner OB, Aguilar HC, Fulcher J.A, Levroney EL, Harrison R, Wright L, Robinson LR, Aspericueta V, Panico M, Haslam SM, Morris HR, Dell A, Lee B, Baum LG. 2010. Endothelial galectin-1 binds to specific glycans on nipah virus fusion protein and inhibits maturation, mobility, and function to block syncytia formation. PLoS Pathog 6:e1000993. doi: 10.1371/journal.ppat.1000993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Elahi S, Niki T, Hirashima M, Horton H. 2012. Galectin-9 binding to Tim-3 renders activated human CD4+ T cells less susceptible to HIV-1 infection. Blood 119:4192–4204. doi: 10.1182/blood-2011-11-389585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bi S, Hong PW, Lee B, Baum LG. 2011. Galectin-9 binding to cell surface protein disulfide isomerase regulates the redox environment to enhance T-cell migration and HIV entry. Proc Natl Acad Sci U S A 108:10650–10655. doi: 10.1073/pnas.1017954108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.McSharry BP, Forbes SK, Cao JZ, Avdic S, Machala EA, Gottlieb DJ, Abendroth A, Slobedman B. 2014. Human cytomegalovirus upregulates expression of the lectin galectin 9 via induction of beta interferon. J Virol 88:10990–10994. doi: 10.1128/JVI.01259-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fujita K, Iwama H, Oura K, Tadokoro T, Samukawa E, Sakamoto T, Nomura T, Tani J, Yoneyama H, Morishita A, Himoto T, Hirashima M, Masaki T. 2017. Cancer therapy due to apoptosis: galectin-9. Int J Mol Sci 18:74. doi: 10.3390/ijms18010074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Fielding CA, Aicheler R, Stanton RJ, Wang EC, Han S, Seirafian S, Davies J, McSharry BP, Weekes MP, Antrobus PR, Prod’homme V, Blanchet FP, Sugrue D, Cuff S, Roberts D, Davison AJ, Lehner PJ, Wilkinson GW, Tomasec P. 2014. Two novel human cytomegalovirus NK cell evasion functions target MICA for lysosomal degradation. PLoS Pathog 10:e1004058. doi: 10.1371/journal.ppat.1004058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang D, Shenk T. 2005. Human cytomegalovirus UL131 open reading frame is required for epithelial cell tropism. J Virol 79:10330–10338. doi: 10.1128/JVI.79.16.10330-10338.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.O’Connor CM, Murphy EA. 2012. A myeloid progenitor cell line capable of supporting human cytomegalovirus latency and reactivation, resulting in infectious progeny. J Virol 86:9854–9865. doi: 10.1128/JVI.01278-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Compton T, Nepomuceno RR, Nowlin DM. 1992. Human cytomegalovirus penetrates host cells by pH-independent fusion at the cell surface. Virology 191:387–395. doi: 10.1016/0042-6822(92)90200-9. [DOI] [PubMed] [Google Scholar]
- 45.Ryckman BJ, Jarvis MA, Drummond DD, Nelson JA, Johnson DC. 2006. Human cytomegalovirus entry into epithelial and endothelial cells depends on genes UL128 to UL150 and occurs by endocytosis and low-pH fusion. J Virol 80:710–722. doi: 10.1128/JVI.80.2.710-722.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Drickamer K. 1988. Two distinct classes of carbohydrate-recognition domains in animal lectins. J Biol Chem 263:9557. [PubMed] [Google Scholar]
- 47.Rabinovich GA, Toscano MA, Jackson SS, Vasta GR. 2007. Functions of cell surface galectin-glycoprotein lattices. Curr Opin Struct Biol 17:513–520. doi: 10.1016/j.sbi.2007.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Torres L, Tang Q. 2014. Immediate-Early (IE) gene regulation of cytomegalovirus: IE1- and pp71-mediated viral strategies against cellular defenses. Virol Sin 29:343–352. doi: 10.1007/s12250-014-3532-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Stachowiak JC, Brodsky FM, Miller EA. 2013. A cost-benefit analysis of the physical mechanisms of membrane curvature. Nat Cell Biol 15:1019–1027. doi: 10.1038/ncb2832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Navarro D, Paz P, Tugizov S, Topp K, La Vail J, Pereira L. 1993. Glycoprotein B of human cytomegalovirus promotes virion penetration into cells, transmission of infection from cell to cell, and fusion of infected cells. Virology 197:143–158. doi: 10.1006/viro.1993.1575. [DOI] [PubMed] [Google Scholar]
- 51.Abdel-Mohsen M, Chavez L, Tandon R, Chew GM, Deng X, Danesh A, Keating S, Lanteri M, Samuels ML, Hoh R, Sacha JB, Norris PJ, Niki T, Shikuma CM, Hirashima M, Deeks SG, Ndhlovu LC, Pillai SK. 2016. Human galectin-9 is a potent mediator of HIV transcription and reactivation. PLoS Pathog 12:e1005677. doi: 10.1371/journal.ppat.1005677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Eisenberg RJ, Atanasiu D, Cairns TM, Gallagher JR, Krummenacher C, Cohen GH. 2012. Herpes virus fusion and entry: a story with many characters. Viruses 4:800–832. doi: 10.3390/v4050800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sharma S, Wisner TW, Johnson DC, Heldwein EE. 2013. HCMV gB shares structural and functional properties with gB proteins from other herpesviruses. Virology 435:239–249. doi: 10.1016/j.virol.2012.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hahn G, Revello MG, Patrone M, Percivalle E, Campanini G, Sarasini A, Wagner M, Gallina A, Milanesi G, Koszinowski U, Baldanti F, Gerna G. 2004. Human cytomegalovirus UL131-128 genes are indispensable for virus growth in endothelial cells and virus transfer to leukocytes. J Virol 78:10023–10033. doi: 10.1128/JVI.78.18.10023-10033.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Straschewski S, Patrone M, Walther P, Gallina A, Mertens T, Frascaroli G. 2011. Protein pUL128 of human cytomegalovirus is necessary for monocyte infection and blocking of migration. J Virol 85:5150–5158. doi: 10.1128/JVI.02100-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Dargan DJ, Douglas E, Cunningham C, Jamieson F, Stanton RJ, Baluchova K, McSharry BP, Tomasec P, Emery VC, Percivalle E, Sarasini A, Gerna G, Wilkinson GWG, Davison AJ. 2010. Sequential mutations associated with adaptation of human cytomegalovirus to growth in cell culture. J Gen Virol 91:1535–1546. doi: 10.1099/vir.0.018994-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wang D, Shenk T. 2005. Human cytomegalovirus virion protein complex required for epithelial and endothelial cell tropism. Proc Natl Acad Sci U S A 102:18153–18158. doi: 10.1073/pnas.0509201102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ryckman BJ, Chase MC, Johnson DC. 2008. HCMV gH/gL/UL128-131 interferes with virus entry into epithelial cells: evidence for cell type-specific receptors. Proc Natl Acad Sci U S A 105:14118–14123. doi: 10.1073/pnas.0804365105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhou M, Yu Q, Wechsler A, Ryckman BJ. 2013. Comparative analysis of gO isoforms reveals that strains of human cytomegalovirus differ in the ratio of gH/gL/gO and gH/gL/UL128-131 in the virion envelope. J Virol 87:9680–9690. doi: 10.1128/JVI.01167-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wille PT, Knoche AJ, Nelson JA, Jarvis MA, Johnson DC. 2010. A human cytomegalovirus gO-null mutant fails to incorporate gH/gL into the virion envelope and is unable to enter fibroblasts and epithelial and endothelial cells. J Virol 84:2585–2596. doi: 10.1128/JVI.02249-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Vanarsdall AL, Chase MC, Johnson DC. 2011. HCMV glycoprotein gO complexes with gH/gL promoting interference with viral entry into human fibroblasts, but not entry into epithelial cells. J Virol doi: 10.1128/JVI.05659-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Varnum SM, Streblow DN, Monroe ME, Smith P, Auberry KJ, Pasa-Tolic L, Wang D, Camp DG, Rodland K, Wiley S, Britt W, Shenk T, Smith RD, Nelson JA. 2004. Identification of proteins in human cytomegalovirus (HCMV) particles: the HCMV proteome. J Virol 78:10960–10966. doi: 10.1128/JVI.78.20.10960-10966.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rigoutsos I, Novotny J, Huynh T, Chin-Bow S.T, Parida L, Platt D, Coleman D, Shenk T. 2003. In silico pattern-based analysis of the human cytomegalovirus genome. J Virol 77:4326–4344. doi: 10.1128/JVI.77.7.4326-4344.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mareri A, Lasorella S, Iapadre G, Maresca M, Tambucci R, Nigro G. 2016. Anti-viral therapy for congenital cytomegalovirus infection: pharmacokinetics, efficacy and side effects. J Matern Fetal Neonatal Med 29:1657–1664. doi: 10.3109/14767058.2015.1058774. [DOI] [PubMed] [Google Scholar]
- 65.Gilbert C, Bestman-Smith J, Boivin G. 2002. Resistance of herpesviruses to antiviral drugs: clinical impacts and molecular mechanisms. Drug Resist Updat 5:88–114. doi: 10.1016/S1368-7646(02)00021-3. [DOI] [PubMed] [Google Scholar]
- 66.Ghosh T, Chattopadhyay K, Marschall M, Karmakar P, Mandal P, Ray B. 2009. Focus on antivirally active sulfated polysaccharides: from structure-activity analysis to clinical evaluation. Glycobiology 19:2–15. doi: 10.1093/glycob/cwn092. [DOI] [PubMed] [Google Scholar]
- 67.Andersen JH, Osbakk SA, Vorland LH, Traavik T, Gutteberg TJ. 2001. Lactoferrin and cyclic lactoferricin inhibit the entry of human cytomegalovirus into human fibroblasts. Antiviral Res 51:141–149. doi: 10.1016/S0166-3542(01)00146-2. [DOI] [PubMed] [Google Scholar]
- 68.Luganini A, Giuliani A, Pirri G, Pizzuto L, Landolfo S, Gribaudo G. 2010. Peptide-derivatized dendrimers inhibit human cytomegalovirus infection by blocking virus binding to cell surface heparan sulfate. Antiviral Res 85:532–540. doi: 10.1016/j.antiviral.2010.01.003. [DOI] [PubMed] [Google Scholar]
- 69.Jones TR, Lee SW, Johann SV, Razinkov V, Visalli RJ, Feld B, Bloom JD, O'Connell J. 2004. Specific inhibition of human cytomegalovirus glycoprotein B-mediated fusion by a novel thiourea small molecule. J Virol 78:1289–1300. doi: 10.1128/JVI.78.3.1289-1300.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.English EP, Chumanov RS, Gellman SH, Compton T. 2006. Rational development of beta-peptide inhibitors of human cytomegalovirus entry. J Biol Chem 281:2661–2667. doi: 10.1074/jbc.M508485200. [DOI] [PubMed] [Google Scholar]
- 71.Luganini A, Caposio P, Landolfo S, Gribaudo G. 2008. Phosphorothioate-modified oligodeoxynucleotides inhibit human cytomegalovirus replication by blocking virus entry. Antimicrob Agents Chemother 52:1111–1120. doi: 10.1128/AAC.00987-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Stegmann C, Hochdorfer D, Lieber D, Subramanian N, Stohr D, Sampaio KL, Sinzger C. 2017. A derivative of platelet-derived growth factor receptor alpha binds to the trimer of human cytomegalovirus and inhibits entry into fibroblasts and endothelial cells. PLoS Pathog 13:e1006273. doi: 10.1371/journal.ppat.1006273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li Q, Fischer E, Cohen JI. 2016. Cell surface THY-1 contributes to human cytomegalovirus entry via a macropinocytosis-like process. J Virol 90:9766–9781. doi: 10.1128/JVI.01092-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Li Q, Wilkie AR, Weller M, Liu X, Cohen JI. 2015. THY-1 cell surface antigen (CD90) has an important role in the initial stage of human cytomegalovirus infection. PLoS Pathog 11:e1004999. doi: 10.1371/journal.ppat.1004999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Imaizumi T, Kumagai M, Sasaki N, Kurotaki H, Mori F, Seki M, Nishi N, Fujimoto K, Tanji K, Shibata T, Tamo W, Matsumiya T, Yoshida H, Cui XF, Takanashi S, Hanada K, Okumura K, Yagihashi S, Wakabayashi K, Nakamura T, Hirashima M, Satoh K. 2002. Interferon-gamma stimulates the expression of galectin-9 in cultured human endothelial cells. J Leukoc Biol 72:486–491. [PubMed] [Google Scholar]
- 76.Gamadia LE, Remmerswaal EB, Weel JF, Bemelman F, van Lier RA, Ten Berge IJ. 2003. Primary immune responses to human CMV: a critical role for IFN-gamma-producing CD4+ T cells in protection against CMV disease. Blood 101:2686–2692. doi: 10.1182/blood-2002-08-2502. [DOI] [PubMed] [Google Scholar]
- 77.Chagan-Yasutan H, Saitoh H, Ashino Y, Arikawa T, Hirashima M, Li S, Usuzawa M, Oguma S, O’Telan EF, Obi CL, Hattori T. 2009. Persistent elevation of plasma osteopontin levels in HIV patients despite highly active antiretroviral therapy. Tohoku J Exp Med 218:285–292. doi: 10.1620/tjem.218.285. [DOI] [PubMed] [Google Scholar]
- 78.Saitoh H, Ashino Y, Chagan-Yasutan H, Niki T, Hirashima M, Hattori T. 2012. Rapid decrease of plasma galectin-9 levels in patients with acute HIV infection after therapy. Tohoku J Exp Med 228:157–161. doi: 10.1620/tjem.228.157. [DOI] [PubMed] [Google Scholar]
- 79.Jost S, Moreno-Nieves UY, Garcia-Beltran WF, Rands K, Reardon J, Toth I, Piechocka-Trocha A, Altfeld M, Addo MM. 2013. Dysregulated Tim-3 expression on natural killer cells is associated with increased galectin-9 levels in HIV-1 infection. Retrovirology 10:74. doi: 10.1186/1742-4690-10-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Tandon R, Chew GM, Byron MM, Borrow P, Niki T, Hirashima M, Barbour JD, Norris PJ, Lanteri MC, Martin JN, Deeks SG, Ndhlovu LC. 2014. Galectin-9 is rapidly released during acute HIV-1 infection and remains sustained at high levels despite viral suppression even in elite controllers. AIDS Res Hum Retroviruses 30:654–664. doi: 10.1089/AID.2014.0004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Katoh S, Ikeda M, Shimizu H, Mouri K, Obase Y, Kobashi Y, Fukushima K, Hirashima M, Oka M. 2014. Increased levels of plasma galectin-9 in patients with influenza virus infection. Tohoku J Exp Med 232:263–267. doi: 10.1620/tjem.232.263. [DOI] [PubMed] [Google Scholar]
- 82.Mengshol JA, Golden-Mason L, Arikawa T, Smith M, Niki T, McWilliams R, Randall JA, McMahan R, Zimmerman MA, Rangachari M, Dobrinskikh E, Busson P, Polyak SJ, Hirashima M, Rosen HR. 2010. A crucial role for Kupffer cell-derived galectin-9 in regulation of T cell immunity in hepatitis C infection. PLoS One 5:e9504. doi: 10.1371/journal.pone.0009504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Harwood NM, Golden-Mason L, Cheng L, Rosen HR, Mengshol JA. 2016. HCV-infected cells and differentiation increase monocyte immunoregulatory galectin-9 production. J Leukoc Biol 99:495–503. doi: 10.1189/jlb.5A1214-582R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Li H, Wu K, Tao K, Chen L, Zheng Q, Lu X, Liu J, Shi L, Liu C, Wang G, Zou W. 2012. Tim-3/galectin-9 signaling pathway mediates T-cell dysfunction and predicts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma. Hepatology 56:1342–1351. doi: 10.1002/hep.25777. [DOI] [PubMed] [Google Scholar]
- 85.J Reddy PB, Schreiber TH, Rajasagi NK, Suryawanshi A, Mulik S, Veiga-Parga T, Niki T, Hirashima M, Podack ER, Rouse BT. 2012. TNFRSF25 agonistic antibody and galectin-9 combination therapy controls herpes simplex virus-induced immunoinflammatory lesions. J Virol 86:10606–10620. doi: 10.1128/JVI.01391-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pioche-Durieu C, Keryer C, Souquere S, Bosq J, Faigle W, Loew D, Hirashima M, Nishi N, Middeldorp J, Busson P. 2005. In nasopharyngeal carcinoma cells, Epstein-Barr virus LMP1 interacts with galectin 9 in membrane raft elements resistant to simvastatin. J Virol 79:13326–13337. doi: 10.1128/JVI.79.21.13326-13337.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Keryer-Bibens C, Pioche-Durieu C, Villemant C, Souquere S, Nishi N, Hirashima M, Middeldorp J, Busson P. 2006. Exosomes released by EBV-infected nasopharyngeal carcinoma cells convey the viral latent membrane protein 1 and the immunomodulatory protein galectin 9. BMC Cancer 6:283. doi: 10.1186/1471-2407-6-283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Shaw AE, Hughes J, Gu Q, Behdenna A, Singer J.B, Dennis T, Orton RJ, Varela M, Gifford RJ, Wilson SJ, Palmarini M. 2017. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol 15:e2004086. doi: 10.1371/journal.pbio.2004086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Madireddi S, Eun S-Y, Lee S-W, Nemčovičová I, Mehta AK, Zajonc DM, Nishi N, Niki T, Hirashima M, Croft M. 2014. Galectin-9 controls the therapeutic activity of 4-1BB-targeting antibodies. J Exp Med 211:1433–1448. doi: 10.1084/jem.20132687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Madireddi S, Eun SY, Mehta AK, Birta A, Zajonc DM, Niki T, Hirashima M, Podack ER, Schreiber TH, Croft M. 2017. Regulatory T cell-mediated suppression of inflammation induced by DR3 signaling is dependent on galectin-9. J Immunol 199:2721–2728. doi: 10.4049/jimmunol.1700575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Zheng XX, Strom TB, Khoury SJ, Kuchroo VK. 2005. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol 6:1245–1252. doi: 10.1038/ni1271. [DOI] [PubMed] [Google Scholar]
- 92.Wiersma VR, de Bruyn M, Helfrich W, Bremer E. 2013. Therapeutic potential of galectin-9 in human disease. Med Res Rev 33(Suppl 1):E102–E126. doi: 10.1002/med.20249. [DOI] [PubMed] [Google Scholar]
- 93.He R, Xia C, Ruan Q, Qi Y, Ma YP, Ji YH, Guo JJ. 2011. Sequence analysis of human cytomegalovirus US28 gene in low-passage clinical isolates from children and AIDS patients. Acta Biochim Pol 58:231–236. [PubMed] [Google Scholar]