Vous êtes sur la page 1sur 10

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 12, pp.

8646 –8655, March 19, 2010


© 2010 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

A Lectin Isolated from Bananas Is a Potent Inhibitor of


HIV Replication*
Received for publication, June 24, 2009, and in revised form, January 15, 2010 Published, JBC Papers in Press, January 15, 2010, DOI 10.1074/jbc.M109.034926
Michael D. Swanson‡, Harry C. Winter§, Irwin J. Goldstein§, and David M. Markovitz‡¶储1
From the ¶Department of Internal Medicine, Division of Infectious Diseases, ‡Program in Immunology, 储Cellular and Molecular
Biology Program, and §Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109

BanLec is a jacalin-related lectin isolated from the fruit of receiving antiretroviral therapy by 2.5:1 (1). At present, it
bananas, Musa acuminata. This lectin binds to high mannose appears that an efficacious HIV vaccine is still many years away.
carbohydrate structures, including those found on viruses con- Therefore, other methods for halting the spread of HIV are
taining glycosylated envelope proteins such as human immuno- vitally needed. This has raised the possibility of developing
deficiency virus type-1 (HIV-1). Therefore, we hypothesized either intravaginally or intrarectally applied microbicides to
that BanLec might inhibit HIV-1 through binding of the glyco- halt the spread of HIV during sexual intercourse. This type of
sylated HIV-1 envelope protein, gp120. We determined that intervention is particularly needed in the developing world,
BanLec inhibits primary and laboratory-adapted HIV-1 isolates such as sub-Saharan Africa, where more than 20 million people
of different tropisms and subtypes. BanLec possesses potent are living with HIV/AIDS (1). Although abstinence has been

Downloaded from www.jbc.org by guest, on October 31, 2010


anti-HIV activity, with IC50 values in the low nanomolar to pico- suggested by some groups, campaigns to encourage this
molar range. The mechanism for BanLec-mediated antiviral method of halting transmission have not been effective (2).
activity was investigated by determining if this lectin can Although condoms are quite effective against the spread of HIV
directly bind the HIV-1 envelope protein and block entry of the and some other sexually transmitted diseases, they are only
virus into the cell. An enzyme-linked immunosorbent assay con- effective if they are used consistently and correctly, which is
firmed direct binding of BanLec to gp120 and indicated that often not the case (3, 4). This is particularly true in the devel-
BanLec can recognize the high mannose structures that are rec- oping world, where women have relatively little control over
ognized by the monoclonal antibody 2G12. Furthermore, Ban- sexual encounters and, thus, have not been able to enforce con-
Lec is able to block HIV-1 cellular entry as indicated by temper- dom usage (5), so the development of a long-lasting, self-ap-
ature-sensitive viral entry studies and by the decreased levels of plied, microbicide is very attractive. In fact, it is estimated that
the strong-stop product of early reverse transcription seen in 20% coverage with a microbicide that is only 60% effective
the presence of BanLec. Thus, our data indicate that BanLec against HIV may prevent up to 2.5 million HIV infections over
inhibits HIV-1 infection by binding to the glycosylated viral three years (6). Therefore, even modest success with microbi-
envelope and blocking cellular entry. The relative anti-HIV cides could save millions of lives.
activity of BanLec compared favorably to other anti-HIV lectins, Some of the most promising compounds for inhibiting vagi-
such as snowdrop lectin and Griffithsin, and to T-20 and mara- nal or rectal HIV transmission are agents that block HIV before
viroc, two anti-HIV drugs currently in clinical use. Based on integration of the viral genome into the target cell. Thus, the
these results, BanLec is a potential component for an anti-viral viral entry step is one potential target for a microbicide. Entry
microbicide that could be used to prevent the sexual transmis- inhibitors that have been proposed for use in a vaginal micro-
sion of HIV-1. bicide include long chain and ionic polymers (such as Pro 2000)
as well as dendrimers, lipid membrane modifiers, and anti-CD4
antibodies. HIV-binding peptides and small molecule inhibi-
Despite the development of more than 25 approved anti- tors have also been considered, including the fusion inhibitor
HIV2 drugs and improvements in the availability of antiretro- T-20 (enfuvirtide) and the CCR5 blocker maraviroc, which are
viral drugs in low and middle income countries, the rate of new already in clinical use for the treatment of HIV infection. In
HIV-1 infections is outpacing the rate of new individuals addition, lectins are a growing class of HIV-1 inhibitors under
consideration as microbicide candidates (7, 8). Lectins inhibit
HIV-1 entry by binding to carbohydrate structures found on
* This work was supported, in whole or in part, by National Institutes of Health the viral envelope. Examples of anti-HIV lectins include Cya-
Grant R01 AI062248 (of D. M. M.). This work was also supported by a Bur-
roughs Wellcome Fund Clinical Scientist Award in Translational Research.
novirin-N (CV-N) (9), Griffithsin (GRFT) (10), and snowdrop
1
To whom correspondence should be addressed: University of Michigan Medi- lectin (GNA) (11–13).
cal Center, 5220 MSRB III, 1150 West Medical Center Dr., Ann Arbor, MI 48109- The HIV-1 envelope protein gp120 contains 20 –30 possible
5640. Tel.: 734-647-1786; Fax: 734-764-0101; E-mail: dmarkov@umich.edu.
2
The abbreviations used are: HIV, human immunodeficiency virus; ELISA,
N-linked glycosylation sites. These carbohydrate structures
enzyme-linked immunosorbent assay; GRFT, Griffithsin; HEK-293 cells, make up ⬃50% of the molecular weight of the protein (14 –16).
human embryonic kidney cells; PBL, peripheral blood lymphocytes; PBMC, Glycosylation affects aspects of the viral life cycle including
peripheral blood mononuclear cell; MDM, monocyte-derived macro- protein folding (17), cellular transport, binding to cellular
phages; MTT, (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-
mide; PBS, phosphate-buffered saline; TCID50, tissue culture infective dose receptors (14, 18, 19), trans-infection by dendritic cells (19),
50%. and shielding from the immune response (20). Because glyco-

8646 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 12 • MARCH 19, 2010
Inhibition of HIV-1 Replication by BanLec
sylation is essential to the virus, it presents an attractive thera- experiments in which the products of early reverse transcrip-
peutic target. tion were assayed.
The lectin termed BanLec, isolated from the ripened fruit of HIV-1 Indicator Assays—HIV-1 infection was quantified
the banana (Musa acuminata cultivars), exists as a dimer with a using TZM-bl cells, which express a luciferase and ␤-galacto-
molecular mass of ⬃30 kDa (21). It is a member of the jacalin- sidase gene under the control of the HIV-1 LTR promoter (31,
related lectin family and can recognize high mannose struc- 38 – 40). The day before infection of 5000 TZM-bl cells/ml in
tures (22–24). Lectins in this family are characterized by the 100 ␮l of Dulbecco’s modified Eagle’s medium containing 10%
presence of a ␤-prism 1 structure composed of three Greek Key fetal bovine serum, 25 mM HEPES, and 50 ␮g/ml Geneticin
turn motifs. Greek Keys 1 and 2 are both involved in binding were added to the wells of white, opaque, 96-well tissue culture
carbohydrates and contain a GXXXD binding motif, whereas plates (Falcon). Cells were pretreated with BanLec for 30 min
Key 3 does not contain the binding motif (25, 26). However, this before infection with 100 TCID50 units of virus (⬃15,000 rela-
loop can assist ligand binding and determine lectin specificity tive luminescence units) to a final volume of 200 ␮l/well. Cells
(27). Because of its affinity for high mannose structures (15), we were exposed to virus and lectin for either 2 days with replica-
sought to investigate whether BanLec might bind the mannose- tion competent viruses or for 3 days with pseudotyped, replica-
rich envelope of HIV-1 and thereby block HIV infection. The tion defective virus. A Steady-Glo威 luciferase assay system
results presented below demonstrate that BanLec is a potent (Promega) and a plate reader containing a luminometer
inhibitor of HIV infection that markedly reduces the replica- (Tecan) were used to measure luminescence, which was indic-
tion of a range of HIV-1 isolates and has potential to be further ative of viral infection.
developed for use as a vaginal microbicide. MAGI-CCR5 cells (41, 42) were plated in 24-well tissue cul-
ture plates with 40,000 cells per well in Dulbecco’s modified

Downloaded from www.jbc.org by guest, on October 31, 2010


MATERIALS AND METHODS Eagle’s medium containing 10% fetal bovine serum, penicillin,
Proteins and Anti-HIV Compounds—BanLec was isolated and streptomycin. The cells were pretreated with lectin for 30
from bananas by modification of previously described methods min and then infected with different viral isolates at concentra-
(21, 22).3 Snowdrop lectin (GNA) was isolated from crude tions that yielded ⬃100 positively infected cells per well. Forty
extracts of snowdrop bulbs and purified over a mannose-aga- hours post-infection, cells were stained for ␤-galactosidase
rose column as described previously (28). Recombinant HIV-1 activity as described within the reagent data sheet, and positive
gp120, human monoclonal antibody 2G12, recombinant His- cells were counted visually.
tagged GRFT, T-20, maraviroc, and CD4-IgG2 were obtained Isolation and Culture of Primary Cells—PBMCs were iso-
from the NIH AIDS Research and Reference Reagent Program lated from healthy donors by venipuncture. Briefly, blood was
(10). Recombinant, glycosylated gp120 was produced in HEK- drawn into a 60-ml syringe containing 7 ml of 250 mM sodium
293 cells (human embryonic kidney cells), the recombinant citrate and 10 ml of 6% dextran solution and mixed by inver-
human antibody 2G12 was produced in Chinese hamster ovary sion. After 30 min, to allow for the sedimentation of red blood
cells, and recombinant GRFT was produced in Escherichia coli. cells, the supernatant was separated using Hypaque-Ficoll, and
The purity of all of the proteins was found to be ⬎95% as deter- the buffy coat layer was removed, washed twice with cold PBS
mined by SDS-PAGE. containing 0.2% bovine serum albumin, and centrifuged at
HIV-1 Production—The HIV-1 isolate Bru was produced in 350 ⫻ g for 10 min. The cell pellet was resuspended in RPMI
peripheral blood lymphocytes (PBL), whereas the HIV-1 BaL 1640 media at a concentration of 5 ⫻ 106 cells per ml and
(29) isolate was produced in macrophages. Primary, dual-tropic seeded into non-tissue culture-treated plates. PBL were
isolates ASM44 and ASM54 were expanded in peripheral blood removed from the adherent monocytes and washed three times
mononuclear cells (PBMCs) containing both lymphocytes and with PBS. For the differentiation of monocytes to macrophages
macrophages. Production of pseudotyped HIV-1 was per- for HIV-1 infection, the monocytes were cultured with Iscove’s
formed by transfecting HEK-293FT cells (a fast growing cell modified Dulbecco’s media containing 10% heat-inactivated
line derived from HEK-293 that contains the SV40 large T anti- human AB sera for 7 days.
gen) with plasmids coding for an HIV-1 envelope from either Infection of Monocyte-derived Macrophages (MDM)—MDM
subtype B (30) or C along with an envelope-deleted proviral were washed with PBS three times followed by the addition of
clone, pSG3⌬env (31). Proviral plasmid DNA clones pNL4-3 fresh media containing BanLec or PBS 30 min before infection.
(32), pNL(AD8) (33), p81A-4 (34 –36), and p89.6 (37) were Cells were infected with ⬃100 TCID50 of NL(AD8) for 24 h, and
transfected into HEK-293FT cells with Lipofectamine 2000 the residual virus was removed by three PBS washes followed by
(Invitrogen). The media were changed 24 h post-transfection, the addition of fresh media. Every 3 days, a sample was removed
and at 48 h post-transfection the supernatants were harvested and replaced with fresh media containing the appropriate
and frozen at ⫺80 °C. The concentration of virus in the stocks amount of BanLec for 15 days. The samples were stored at
was determined by the HIV-1 p24 Antigen Capture Assay ⫺80 °C until viral replication was determined by the HIV-1 p24
ELISA (AIDS and Cancer Virus Program) or by determining the Antigen Capture Assay ELISA (AIDS and Cancer Virus Pro-
infectious titer. HIV-1 Bru was treated with 10 units/␮l of gram). A similar experiment was done in which samples were
RNase-free DNase I (Roche Applied Science) before use in the not removed until the end of the experiment on day 7. For both
experiments, an MTT ((3-(4,5-Dimethylthiazol-2-yl)-2,5-di-
phenyltetrazolium bromide) reduction assay was performed on
3
H. C. Winter, K. A. Wearne, and I. J. Goldstein, manuscript in preparation. the final day to assess cellular viability.

MARCH 19, 2010 • VOLUME 285 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8647
Inhibition of HIV-1 Replication by BanLec
Detection of Early Products of HIV-1 Reverse Transcription
(Strong-Stop DNA) in Peripheral Blood Lymphocytes—PBL
were stimulated with phytohemagglutinin for 3 days in RPMI
media containing 10% heat-inactivated fetal bovine serum and
interleukin-2. The cells were washed with PBS and resuspended
in RPMI media containing 10% heat-inactivated fetal bovine
serum. Lectins were added 30 min before centrifuge-mediated
infection (spin-infection) with DNase I-treated HIV-1 Bru (43).
Three hours post-infection the cells were harvested, washed
with PBS, and then stored at ⫺80 °C. Cellular DNA, including
genomic and viral DNA products, was isolated using the
QIAamp DNA Blood Mini kit (Qiagen). Strong-stop DNA,
the first product of HIV-1 reverse transcription, is used for the
assessment of viral entry (13, 44). This reverse transcription
product was quantified by performing real-time PCR with
primers specific for strong-stop DNA, the DNA concentration
of the each sample was normalized, and equal DNA loading was
confirmed with primers for ␣-tubulin (44).
Determination of BanLec and Glycosylated HIV-1 gp120
Interaction by ELISA—96-Well ELISA plates were coated by

Downloaded from www.jbc.org by guest, on October 31, 2010


adding 50 ␮l of 5 ␮g/ml BanLec per well and incubated over-
night at room temperature. The next day plates were blocked
for 1.5 h at room temperature with PBS containing 1% bovine
serum albumin, 5% sucrose, and 0.05% sodium azide and then
rinsed with wash buffer (PBS containing 0.05% Tween 20, pH
FIGURE 1. BanLec has antiviral activity against multiple HIV-1 isolates
7.4) 3 times before the addition of recombinant, glycosylated with different tropisms. A, TZM-bl cells were pretreated with different con-
gp120 protein diluted in blocking buffer. After a 1-h incubation centrations of BanLec before infection with the R5 tropic isolates NL(AD8) and
at room temperature, the plates were washed 3 times before the 81A-4, dual tropic 89.6, and X4 tropic NL4-3. Forty-eight hours after exposure
to virus, luciferase activity was determined by measuring relative lumines-
addition of the detection antibodies. A sheep anti-gp120 anti- cent units (RLU). The averages from three separate experiments were used for
body (AIDS Research and Reference Reagent Program) was the calculation of IC50 values, which were determined by nonlinear regres-
sion. The IC50 for viral inhibition were as follows: NL(AD8) ⫽ 2.06 nM, 81A-4 ⫽
diluted 1:2000 in dilution buffer (wash buffer containing 0.1% 0.69 nM, 89.6 ⫽ 0.48 nM, NL4-3 ⫽ 0.49 nM. B, Magi-CCR5 indicator cells were
bovine serum albumin) and added to the wells and incubated used to determine anti-viral activity of BanLec against multiple strains of
for 1 h. The plate was washed again before a 1-h incubation with HIV-1. Forty hours after exposure to virus, infected cells were quantified by
staining for ␤-galactosidase activity. Infectivity of BanLec-treated virus is pre-
an anti-sheep antibody conjugated to alkaline phosphatase sented as a percent of positively infected cells as compared with the PBS
(Sigma) diluted 1:40,000 in dilution buffer. After the plate was control. Error bars represent S.D. from three separate experiments.
washed, p-nitrophenyl phosphate (Sigma) was added for color-
imetric analysis, and the absorbance was measured at 405 nm. Determination of Anti-HIV Activity Post-cellular Attachment—
To determine whether BanLec could block the recognition of For measuring the inhibition of HIV-1 infection post-cellular
gp120 by the anti-HIV monoclonal antibody 2G12, ELISA attachment, TZM-bl cells were plated in 96-well plates as
plates were coated overnight with 100 ␮l of a 1 ␮g/ml solution described above and spin-infected with a pseudotyped virus
of recombinant gp120 diluted in PBS. The plates were blocked with pConCgp160-opt, a consensus subtype C envelope
as described above, washed, and then treated with serial dilu- sequence, at 1250 ⫻ g for 2 h at 16 °C 1 day after plating (43, 45).
tions of BanLec in dilution buffer. After a 1-h incubation the The cells were placed on ice, the supernatant was removed, and
plates were washed to remove unbound BanLec. The 2G12 unbound virus was removed by washing 2 times with PBS con-
antibody was added at a concentration of 100 ng/ml to allow for taining 0.2% bovine serum albumin. Cell culture media con-
binding to the gp120 protein. One hour later the plates were taining inhibitors (CD4-IgG2, T-20, maraviroc, or BanLec) was
washed and incubated with biotinylated anti-human antibody added to the cells and incubated for 30 min on ice. The cells
(Jackson ImmunoResearch) followed by another series of were then moved to a 37 °C incubator, and luciferase activity
washes and the addition of streptavidin-conjugated alkaline was measured 3 days later. These results were compared with
phosphatase (Jackson ImmunoResearch). The alkaline phos- cells that were infected with virus that had been pretreated with
phatase dilution buffer contained 500 mM methyl-␣-D-man- the same inhibitors described above on ice for 30 min and then
nopyranoside, which was added to reduce any potential non- incubated at 37 °C (i.e. standard infection conditions).
specific binding of the 2G12 antibody to BanLec. The plates
were washed again, and the substrate p-nitrophenyl phosphate RESULTS
(Sigma) was added for colorimetric analysis of 2G12 binding. BanLec Is a Potent Inhibitor of Multiple HIV-1 Isolates—To
The amount of antibody that remained bound was quantified determine the anti-HIV activity of BanLec, different concentra-
by comparison to a standard curve consisting of serial 2-fold tions of the lectin were incubated with TZM-bl indicator cells
dilutions of 2G12. before infection with various HIV-1 isolates. Because a micro-

8648 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 12 • MARCH 19, 2010
Inhibition of HIV-1 Replication by BanLec
TABLE 1
Summary of the calculated IC50 values for BanLec inhibition of HIV-1
pseudotyped with HIV-1 envelopes from subtypes B and C
Subtype B Subtype C
95% Confidence 95% Confidence
Envelope IC50 Envelope IC50
intervals intervals
nM nM
SVPB5 0.30 0.27–0.34 SVPC3 0.57 0.51–0.63
SVPB6 0.85 0.74–0.98 SVPC5 0.30 0.21–0.42
SVPB11 0.71 0.65–0.77 SVPC6 0.28 0.25–0.31
SVPB17 0.33 0.26–0.41 SVPC7 2.3 1.8–2.9

Downloaded from www.jbc.org by guest, on October 31, 2010


FIGURE 2. BanLec inhibits infection of HIV-1 pseudotyped with envelopes
from multiple primary isolates. TZM-bl cells were infected with HIV-1
pseudotyped with primary HIV-1 envelope proteins from subtype B (A) and
subtype C (B) in the presence of different concentrations of BanLec. Forty-
eight hours later, luciferase activity was assessed. The IC50 values were deter-
mined as in Fig. 1 and are shown in Table 1. Results shown are the average of
three independent experiments, and error bars represent the S.D. RLU, rela-
tive luminescent units.

bicide would need to inhibit HIV-1 of different tropisms and


because glycosylation can play a role in determining viral tro-
pism (18), we tested the ability of BanLec to inhibit several
different HIV-1 isolates. The viral clones 81A-4 and NL(AD8)
are both derivatives of NL4-3 in which a portion of the envelope FIGURE 3. BanLec inhibits HIV-1 infection of MDM. A, MDM were pretreated
with BanLec for 30 min before the addition of 100 TCID50 of HIV-1 NL(AD8).
is swapped with the envelope region from either the R5 HIV-1 Twenty-four hours later, the media was removed, and the cells were washed
isolates BaL or ADA, respectively. 81A-4 and NL(AD8) use with PBS to eliminate remaining virus. Fresh media containing BanLec or PBS
CCR5 as a cellular co-receptor (R5 tropic), whereas NL4-3 uses was added to the cells. A sample of culture supernatant was taken every 3
days for p24 quantification by ELISA and replaced with new media containing
CXCR4 (X4 tropic). These isolates allow for the assessment of lectin in PBS or PBS alone as a control. On day 15, viability was assessed by an
different HIV-1 envelope sensitivity to BanLec while keeping MTT assay, which indicated no cellular toxicity (data not shown). B, MDM were
pretreated and infected with HIV-1 as described above. 24 h post-infection
the remainder of the NL4-3 viral components unchanged. The the cells were washed with PBS to remove residual virus and cultured in
dual-tropic isolate 89.6 was also assessed for susceptibility to media containing BanLec or PBS. Seven days post-infection, supernatants
BanLec. We observed dose-dependent inhibition of viral infec- were removed for determination of p24 antigen as detected by ELISA. The
concentration for a 50% reduction in p24 production was calculated to be
tion with IC50 values calculated in the low nanomolar range 9.72 nM. Cellular viability was assessed by an MTT assay, and no toxicity was
against viral isolates with different tropisms (Fig. 1A). These observed (data not shown). Results shown in panels A and B are representa-
results suggest that sensitivity of HIV-1 isolates to BanLec is tive of three and two separate experiments, respectively.
independent of viral tropism.
We further confirmed the anti-HIV activity of BanLec with experiment was generated by infection of PBMC, whereas the
the HIV-1 indicator cell line, MAGI-CCR5. With this cell line, experiment shown in Fig. 1A used virus produced by transfec-
we tested the ability of BanLec to inhibit infection by the labo- tion of HEK-293FT cells with a proviral plasmid clone. These
ratory-adapted isolates BaL (R5) and Bru (X4) and the primary results further support our initial studies by showing that
isolates ASM 44 (R5X4) and ASM 54 (R5X4) and determined BanLec can inhibit HIV isolates both independent of viral tro-
that all were inhibited by BanLec (Fig. 1B). The virus used in this pism and of the cell type used to produce virus.

MARCH 19, 2010 • VOLUME 285 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8649
Inhibition of HIV-1 Replication by BanLec
ing the media or adding additional
lectin, the IC50 value for BanLec
inhibition of HIV-1 replication was
9.72 nM (Fig. 3B), demonstrating
that BanLec remains a potent and
stable inhibitor in a long term cul-
ture system at 37 °C.
BanLec Appears to Block HIV-1
Infection at the Viral Entry Step—
Having shown that BanLec can
inhibit HIV replication in MDM, we
tested the ability of BanLec to block
cellular entry of HIV-1 in PBL. We
hypothesized that BanLec binds to
high mannose structures found on
the HIV-1 envelope, preventing
entry and, thus, infection. If so, little
or none of the strong-stop DNA
product of early HIV-1 reverse tran-
scription (see “Materials and Meth-

Downloaded from www.jbc.org by guest, on October 31, 2010


FIGURE 4. BanLec inhibits production of early HIV-1 reverse transcription products in peripheral blood
lymphocytes. Peripheral blood lymphocytes were treated with different lectin concentrations 30 min before ods”) should be detected when cells
infection with HIV-1 Bru. Three hours post-infection, cellular DNA of the infected cells was harvested, and are exposed to HIV-1 in the pres-
strong-stop DNA was quantified by real-time PCR. The number of copies was normalized to a PBS-treated
control (100%). The known anti-HIV lectin GNA (circles) was used as a positive control and to assess the relative ence of BanLec (44, 48). To test this
molar potency of BanLec (squares). hypothesis, we incubated PBL with
the HIV-1 Bru isolate in the pres-
R5 tropic viruses are the dominant form found in sexually ence of different concentrations of BanLec. As a positive con-
transmitted HIV-1 and, therefore, would have to be neutralized trol and for comparison, a similar experiment with the lectin
by microbicides. In addition, anti-HIV microbicides will need GNA was performed in parallel. Real-time PCR was used to
to inhibit infection by viral isolates from different subtypes. detect strong-stop DNA, which is a reverse transcription prod-
Although one would assume that all clades could be neutralized uct that can be detected early after viral entry before viral
due to the conservation of gp120 glycosylation, a difference in uncoating takes place (49 –51). Strong-stop DNA that may have
the susceptibility of the viral subtypes B and C to the anti-HIV, been present in the virus stock was removed by treatment with
high mannose-recognizing antibody 2G12 has been observed DNase I to eliminate false detection of reverse transcription
(46). To determine whether BanLec could inhibit additional products. Treatment with BanLec resulted in a marked de-
primary isolates from different clades, we tested BanLec for crease in strong-stop DNA at low lectin concentrations (Fig. 4)
inhibition of HIV-1 pseudotyped with envelopes derived from indicating that, in addition to inhibiting viral replication in
primary isolates of subtypes B and C. These subtypes are com- MDM, BanLec blocks HIV-1 infection in PBL. Furthermore,
monly found in North and Central America (subtype B) and this inhibition occurs at a step before early replication events,
parts of Africa and India (subtype C). We observed potent, sub-
apparently at the level of viral entry. Although it appears
nanomolar inhibition of viral replication by BanLec (Fig. 2 and
unlikely that BanLec inhibits reverse transcription, these PCR
Table 1), and no significant difference was observed when the
results do not exclude this possibility, as similar results could be
average IC50 values from the two different subtypes were com-
produced by an inhibitor of HIV-1 reverse transcriptase. This
pared by Student’s t test (p ⬍ 0.56). This suggests that BanLec
issue is addressed further below.
can effectively inhibit infection by viral isolates prominent in
regions where a microbicide would be most valuable. BanLec Binds to Glycosylated gp120—BanLec is known to
BanLec Blocks Infection of MDM—Macrophages are suscep- bind to mannose, and thus, we hypothesized that BanLec binds
tible to HIV-1 infection and can become viral reservoirs that the high mannose structures found on the glycosylated gp120
cannot be eliminated by highly active antiretroviral therapy. envelope protein and blocks entry of HIV-1 into cells. We pre-
The role of vaginal macrophages in HIV-1 pathogenesis has not pared a BanLec-based ELISA to measure binding of glycosy-
been fully characterized, but recent evidence indicates that lated HIV-1 gp120 to BanLec. We observed that BanLec does
these cells are permissive for HIV-1 infection (47). We tested indeed bind to gp120 in a concentration-dependent manner
the ability of BanLec to inhibit HIV-1 infection of MDM. As (Fig. 5A). Furthermore, a known BanLec ligand, methyl- ␣-D-
shown in Fig. 3A, nanomolar concentrations of BanLec inhib- mannopyranoside, inhibited such binding in a concentration-
ited HIV replication in MDM over a period of 15 days. Further- dependent manner (Fig. 5B). Not surprisingly, a high concen-
more, BanLec had no effect on cellular viability as determined tration of methyl-␣-D-mannopyranoside ligand was needed to
by MTT assay performed on day 15 (data not shown); therefore, compete for binding to gp120 because of the high density of
this effect was not due to cellular toxicity. When BanLec carbohydrate residues on the HIV-1 envelope protein. These
remained in the culture supernatant for 7 days without chang- results corroborate our hypothesis that BanLec inhibits HIV-1

8650 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 12 • MARCH 19, 2010
Inhibition of HIV-1 Replication by BanLec

FIGURE 6. Binding of gp120 by BanLec blocks access to the anti-HIV


monoclonal antibody 2G12. Recombinant gp120-coated ELISA plates were
treated with different concentrations of BanLec before incubation with the
2G12 antibody, which recognizes the high mannose structures found at posi-
tions Asn-295, -332, and -392 of the gp120 protein. Unbound antibody was

Downloaded from www.jbc.org by guest, on October 31, 2010


removed, and the amount of antibody remaining was determined by com-
parison to a standard curve. The results shown represent the average from
three separate experiments. Error bars represent S.E. The effect of increasing
amounts of BanLec was determined to be significant by 1-way analysis of
variance testing (p ⬍ 0.01).

decrease in the inhibitory activity of the HIV-1 attachment


inhibitor CD4-IgG2 in the post-attachment assay, whereas the
bound virus was still essentially completely susceptible to the
fusion inhibitor T-20 (Fig. 7 and Table 2). This demonstrates
FIGURE 5. BanLec binds to glycosylated gp120. A, shown is dose-depen- that the assay works as expected, with viral attachment, but not
dent binding of BanLec to glycosylated gp120. BanLec was used to coat a
96-well ELISA plate. Serial dilutions of gp120 were added in duplicate to the
fusion, taking place at 16 °C. Both the CCR5 binding inhibitor
wells. gp120 was detected with an anti-gp120 antibody. Results are repre- maraviroc and BanLec primarily blocked viral replication by
sentative of four independent experiments. B, methyl ␣-D-mannopyranoside inhibiting HIV-1 attachment, but each does appear to also have
inhibits interaction of BanLec with gp120. ELISA plates were coated with
BanLec as in panel A. Serial dilutions of methyl ␣-D-mannopyranoside a modest effect on viral fusion (Fig. 7). Importantly, when we
were added to wells along with a constant amount of gp120. The amount compare the IC50 value of BanLec to those of other anti-HIV
of gp120 bound was determined using the standard curve produced in compounds, we see that BanLec potency compares quite well to
panel A. Abs, absorbance.
the clinically approved anti-virals T-20 and maraviroc (Table
2). Thus, we conclude that BanLec potently inhibits the attach-
cellular entry by binding to high mannose structures found on ment of HIV-1 to cells and has a more modest effect on viral
the virus. fusion.
To explore the BanLec binding sites on gp120, we deter- The Anti-HIV Activity of BanLec Compares Well to Other
mined the ability of BanLec to block binding by the monoclonal Anti-HIV Lectins—Several different lectins have been found to
antibody 2G12 using the ELISA-based assay. 2G12 recognizes a inhibit HIV-1 infection. However, they vary in their degree of
cluster of N-linked glycosylation structures at positions Asn- anti-viral activity. To assess the relative molar-based potency of
295, -332, and -392 (position numbering is of the HXB2 refer- BanLec, we compared the anti-HIV activity of BanLec to that
ence sequence) that are crucial for antibody recognition (52– of two previously described anti-HIV lectins, GNA and GRFT.
54). We found that pretreatment of gp120 with BanLec Upon comparison, all three lectins showed activity in the nano-
inhibited recognition by 2G12 in a dose-dependent manner, molar range (Fig. 8). Taken together, our data suggest that
suggesting that BanLec is capable of binding to this antibody’s BanLec inhibits infection with a broad range of HIV-1 isolates
epitope consisting of high mannose structures (Fig. 6). by blocking viral entry, compares favorably with the potency of
BanLec Inhibits HIV-1 Infection before Viral Fusion—To fur- previously described lectins and clinically available anti-HIV
ther investigate at which point in the viral life cycle BanLec drugs, and is a potential component for future anti-HIV vaginal
inhibits HIV-1 infection, we tested BanLec for its ability to microbicides.
inhibit HIV-1 infection post-attachment. To do so, we tested if
BanLec could inhibit HIV-1 that was already bound to the cell DISCUSSION
but could not complete fusion due to temperature restriction The primary mechanism of inhibition by BanLec appears to
(55). As controls, we took advantage of the fact that CD4-IgG2 be blocking cellular attachment of HIV and, thus, viral entry.
inhibits HIV-1 infection by blocking attachment, whereas T-20 Our conclusion is based on the findings from our ELISA assays
works by blocking fusion. As anticipated, we observed a large that BanLec can bind to high mannose structures found on

MARCH 19, 2010 • VOLUME 285 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8651
Inhibition of HIV-1 Replication by BanLec

Downloaded from www.jbc.org by guest, on October 31, 2010


FIGURE 7. BanLec primarily inhibits binding of HIV to the cellular membrane. TZM-bl cells were spin-infected at 16 °C, a temperature that allows for
attachment of virus but does not allow fusion events to occur. The unbound virus was removed, and the cells were incubated with media containing inhibitors
(CD4-IgG2, T-20, maraviroc, or BanLec) on ice for 30 min, and then the plates were shifted to 37 °C to allow for fusion and infection to be completed (E). The
results were compared with a standard infection procedure (pre-attachment) in which the virus and inhibitors were incubated together on ice for 30 min and
then added to TZM-bl cells and incubated at 37 °C (F). The results shown are the averages from three separate experiments. Nonlinear regression analysis was
used for curve fitting and calculation of IC50 values (Table 2). RLU, relative luminescent units.

TABLE 2
Summary of the calculated IC50 values for inhibition of HIV-1
infection with the addition of drug either pre- or post-attachment to
TZM-bl cells
Post-attachment Pre-attachment
Compound 95% Confidence 95% Confidence
IC50 intervals
IC50
intervals
nM nM
CD4-IgG2 –a 1.39 0.667–2.89
T-20 2.78 1.96–3.95 1.90 0.958–3.77
Maraviroc 10.2 7.5–13.8 1.23 0.584–2.58
BanLec 13.1 9.61–17.7 0.224 0.168–0.300
a
Too high to calculate.

HIV-1 gp120, including the high mannose structures that are


FIGURE 8. Comparison of the anti-HIV activity of BanLec to the anti-HIV
recognized by the monoclonal antibody 2G12. This was corrob- lectins GNA and GRFT. TZM-bl cells were pretreated with BanLec, GRFT, or
orated by the finding that cells treated with BanLec had GNA diluted in PBS or PBS alone, as a control, for 30 min before infection by
decreased amounts of an early HIV reverse transcription prod- the R5 tropic HIV-1 virus 81-A. Forty-eight hours later, luciferase activity was
measured. The results are normalized to infected cells treated with PBS alone.
uct, strong-stop DNA, that can be detected shortly after cellular The average of three separate experiments is shown and was used to calcu-
entry of the virus and before viral uncoating. In addition, we late IC50 values by nonlinear regression. The calculated IC50 values are the
following: GNA ⫽ 34.3 nM, BanLec 3.18 nM, and GRFT 0.42 nM. RLU, relative
performed an assay that took advantage of a temperature-ar- luminescent units.
rested state (16 °C) that prevents HIV-1 fusion and compared
the inhibitory activity of BanLec and other anti-HIV drugs pre-
and post-cellular attachment of the virus, finding that most of experiments, was due to blocking viral attachment, when mara-
the inhibitory activity of BanLec comes from blocking viral viroc was added post-attachment we still observed inhibition of
attachment. Interestingly, whereas most of the inhibitory activ- HIV, albeit at a reduced level. A similar result was also seen with
ity of the CCR5 blocker maraviroc, used as a control in these BanLec, suggesting that these two compounds could have addi-

8652 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 12 • MARCH 19, 2010
Inhibition of HIV-1 Replication by BanLec
tional inhibitory activity at a post-attachment step, such as change bioavailability and reduce toxicity. This modification of
fusion of the virus to the cell. CV-N has been shown to be effective in reducing mitogenic
Our studies indicate that BanLec is a new and promising activity in vitro (70). Although BanLec has also been reported to
member of the group of lectins that are able to inhibit HIV-1 possess mitogenic activity (71), the relationship between mito-
infection through interactions with glycosylation sites found genic activity in vitro and microbicide efficacy has not been
on the viral envelope. The inhibitory activity of BanLec elucidated, so it remains possible that recombinant versions of
against HIV-1 was broad, independent of tropism, and effec- BanLec and other lectins could be developed that retain efficacy
tive against several subtype B and C envelope sequences. but have minimal mitogenic activity. The anti-HIV lectin GRFT
HIV-1 pseudotyped with envelopes derived from primary has recently been reported not to have a mitogenic effect when
isolates was inhibited by BanLec in the low nanomolar range. added to human PBMCs (72). This observation is of interest, as
BanLec was also able to inhibit HIV-1 infection of primary GRFT is in the same jacalin-related lectin family and has a sim-
cells, and thus, our results are not limited to cell lines. ilar structure to BanLec (73). GRFT has also been shown to be
Based on our findings, it is likely that BanLec will be able to non-inflammatory, non-toxic, and capable of being manufac-
inhibit other HIV-1 subtypes, as they all contain glycosylation tured on a large scale. Although clinical testing of these newer
sites in their envelope sequences. The isolates used in our lectins has yet to be performed, it appears that lectins have
experiments differed in the number of predicted N-linked gly- potential to be used as anti-HIV agents (72). Because the bind-
cosylation sites, supporting the likelihood that BanLec will be ing, toxicity, and anti-HIV activity of lectins vary, the identifi-
effective against most HIV subtypes found in both the develop- cation of novel anti-viral lectins, such as BanLec, will further
ing and developed world. Because glycosylation is not specific increase the possibility of successful development of a lectin-
to HIV-1, lectins have the potential to inhibit the replication of based anti-HIV microbicide.

Downloaded from www.jbc.org by guest, on October 31, 2010


a broad spectrum of viruses. Indeed, it has been shown that
lectins can inhibit other enveloped viruses including Ebola (56, Acknowledgments—The following reagents were obtained through the
57), Marburg (57), influenza (58), severe acute respiratory syn- AIDS Research and Reference Reagent Program, Division of AIDS,
drome coronavirus (59), and hepatitis C virus (60, 61). NIAID, National Institutes of Health: TZM-bl cells from Dr. John C.
One potential benefit of the use of lectins as anti-HIV agents is Kappes, Dr. Xiaoyun Wu, and Tranzyme Inc. (catalog #8129), MAGI-
their ability to target multiple different glycosylation sites on the CCR5 cells from Dr. Julie Overbaugh (catalog #3522), Maraviroc (cat-
virus, thus making it more difficult for resistance to develop. In alog #11580), T-20 fusion inhibitor from Roche Applied Science (cat-
support of this prediction, previous studies that determined the alog #9845), CD4-IgG2 from Progenics Pharmaceuticals (catalog
#11780), HIS-Griffithsin from Drs. Barry O’Keefe and James
resistance profiles of HIV-1 treated with lectin showed that mul-
McMahon (catalog #11610), pSG3⌬env from Drs. John C. Kappes and
tiple mutations in the envelope sequence were needed for the
Xiaoyun Wu (catalog #11051), p81A-4 from Dr. Bruce Chesebro (cat-
development of resistance (62). Furthermore, different mutations alog #11440), pAD8(NL4-3) from Dr. Eric O. Freed (catalog #11346),
in N-linked glycosylation sites are required for the development of p89.6 from Ronald G. Collman (catalog #3552), pNL4-3 from
resistance to different lectins. This suggests that the combinatorial Dr. Malcolm Martin (catalog #114), standard reference panel of Sub-
or simultaneous use of multiple lectins can reduce the likelihood of type B HIV-1 Env Clones (catalog #11227), standard reference panel
failure of a lectin-based anti-viral therapy due to resistance. If a of subtype C HIV-1 Env Clones (catalog #11326), pConC gp160-opt
population of virus develops resistance to BanLec or other anti- from Dr. Beatrice Hahn (catalog #11407), HIV-1BaL gp120 from Divi-
HIV lectins, one interesting possible consequence is that the virus sion of AIDS Acquired Immunodeficiency Syndrome, NIAID (catalog
will then be more susceptible to neutralization by the human #4961), HIV-1 gp120 monoclonal antibody (2G12) from Dr. Her-
immune response, as the carbohydrate structures found on the mann Katinger (catalog #1476).
HIV-1 envelope are thought to act as a shield against neutralizing
antibody responses (63). This glycan shield works by blocking
REFERENCES
access of epitopes to potentially neutralizing antibodies. Previously
published data demonstrate that alterations in glycosylation that 1. Executive Summary United Nations Programme on HIV/AIDS
(UNAIDS) and World Health Organization (WHO) (2008) Report on
result in resistance to lectins can make the virus vulnerable to neu- the Global AIDS Epidemic
tralizing antibody responses (18, 19). 2. Underhill, K., Montgomery, P., and Operario, D. (2007) BMJ 335, 248
Although several anti-HIV lectins have been described, it is 3. Randolph, M. E., Pinkerton, S. D., Bogart, L. M., Cecil, H., and Abramson,
highly unlikely that a majority of them can be developed for P. R. (2007) Arch. Sex. Behav. 36, 844 – 848
therapeutic use. Like all potential drugs, lectins can vary in their 4. Crosby, R., Yarber, W. L., Sanders, S. A., and Graham, C. A. (2005) J. Am.
Coll. Health 54, 143–147
degrees of potency and toxicity (64, 65). Also, it has been shown
5. Gupta, G. R. (2002) BMJ 324, 183–184
that two anti-HIV lectins can significantly differ in their ability 6. Watts, C. (2002) Microbicides, Antwerp, Belgium, May 12–15, 2002
to block attachment of HIV to epithelial cells (66). Concerns 7. Balzarini, J., and Van Damme, L. (2007) Lancet 369, 787–797
have been raised about the potential toxicity of lectins, for 8. Balzarini, J. (2005) Lancet Infect. Dis. 5, 726 –731
example CV-N. This lectin has shown success as a microbicide 9. Boyd, M. R., Gustafson, K. R., McMahon, J. B., Shoemaker, R. H., O’Keefe,
in in vivo macaque vaginal and rectal transmission models (67, B. R., Mori, T., Gulakowski, R. J., Wu, L., Rivera, M. I., Laurencot, C. M.,
Currens, M. J., Cardellina, J. H., 2nd, Buckheit, R. W., Jr., Nara, P. L.,
68), but safety concerns exist. CV-N was found to have mito-
Pannell, L. K., Sowder, R. C., 2nd, and Henderson, L. E. (1997) Antimicrob.
genic activity when PBMC cultures were exposed to the lectin Agents Chemother. 41, 1521–1530
for 3 days (65, 69). However, recombinant therapeutic proteins 10. Mori, T., O’Keefe, B. R., Sowder, R. C., 2nd, Bringans, S., Gardella, R., Berg,
can be attached to polyethylene glycol (PEG) polymer chains to S., Cochran, P., Turpin, J. A., Buckheit, R. W., Jr., McMahon, J. B., and

MARCH 19, 2010 • VOLUME 285 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8653
Inhibition of HIV-1 Replication by BanLec
Boyd, M. R. (2005) J. Biol. Chem. 280, 9345–9353 41. Chesebro, B., Wehrly, K., Metcalf, J., and Griffin, D. E. (1991) J. Infect. Dis.
11. Balzarini, J. (2007) Nat. Rev. Microbiol. 5, 583–597 163, 64 –70
12. Balzarini, J., Schols, D., Neyts, J., Van Damme, E., Peumans, W., and De 42. Chackerian, B., Long, E. M., Luciw, P. A., and Overbaugh, J. (1997) J. Virol.
Clercq, E. (1991) Antimicrob. Agents Chemother. 35, 410 – 416 71, 3932–3939
13. Balzarini, J., Hatse, S., Vermeire, K., Princen, K., Aquaro, S., Perno, C. F., 43. O’Doherty, U., Swiggard, W. J., and Malim, M. H. (2000) J. Virol. 74,
De Clercq, E., Egberink, H., Vanden Mooter, G., Peumans, W., Van 10074 –10080
Damme, E., and Schols, D. (2004) Antimicrob. Agents Chemother. 48, 44. Schmidtmayerova, H., Alfano, M., Nuovo, G., and Bukrinsky, M. (1998)
3858 –3870 J. Virol. 72, 4633– 4642
14. Matthews, T. J., Weinhold, K. J., Lyerly, H. K., Langlois, A. J., Wigzell, H., 45. Kothe, D. L., Li, Y., Decker, J. M., Bibollet-Ruche, F., Zammit, K. P., Salazar,
and Bolognesi, D. P. (1987) Proc. Natl. Acad. Sci. U.S.A. 84, 5424 –5428 M. G., Chen, Y., Weng, Z., Weaver, E. A., Gao, F., Haynes, B. F., Shaw,
15. Geyer, H., Holschbach, C., Hunsmann, G., and Schneider, J. (1988) J. Biol. G. M., Korber, B. T., and Hahn, B. H. (2006) Virology 352, 438 – 449
Chem. 263, 11760 –11767 46. Gray, E. S., Moore, P. L., Pantophlet, R. A., and Morris, L. (2007) J. Virol.
16. Allan, J. S., Coligan, J. E., Barin, F., McLane, M. F., Sodroski, J. G., Rosen, 81, 10769 –10776
C. A., Haseltine, W. A., Lee, T. H., and Essex, M. (1985) Science 228, 47. Shen, R., Richter, H. E., Clements, R. H., Novak, L., Huff, K., Bimczok, D.,
1091–1094 Sankaran-Walters, S., Dandekar, S., Clapham, P. R., Smythies, L. E., and
17. Li, Y., Luo, L., Rasool, N., and Kang, C. Y. (1993) J. Virol. 67, 584 –588 Smith, P. D. (2009) J. Virol. 83, 3258 –3267
18. Clevestig, P., Pramanik, L., Leitner, T., and Ehrnst, A. (2006) J. Gen. Virol 48. Li, B. Q., Fu, T., Dongyan, Y., Mikovits, J. A., Ruscetti, F. W., and Wang,
87, 607– 612 J. M. (2000) Biochem. Biophys. Res. Commun. 276, 534 –538
19. Geijtenbeek, T. B., Kwon, D. S., Torensma, R., van Vliet, S. J., van Duijn- 49. Warrilow, D., Stenzel, D., and Harrich, D. (2007) Retrovirology 4, 77
hoven, G. C., Middel, J., Cornelissen, I. L., Nottet, H. S., KewalRamani, 50. Warrilow, D., Meredith, L., Davis, A., Burrell, C., Li, P., and Harrich, D.
V. N., Littman, D. R., Figdor, C. G., and van Kooyk, Y. (2000) Cell 100, (2008) J. Virol. 82, 1425–1437
587–597 51. Arhel, N. J., Souquere-Besse, S., Munier, S., Souque, P., Guadagnini, S.,
20. Back, N. K., Smit, L., De Jong, J. J., Keulen, W., Schutten, M., Goudsmit, J., Rutherford, S., Prévost, M. C., Allen, T. D., and Charneau, P. (2007) EMBO

Downloaded from www.jbc.org by guest, on October 31, 2010


and Tersmette, M. (1994) Virology 199, 431– 438 J. 26, 3025–3037
21. Peumans, W. J., Zhang, W., Barre, A., Houlès, Astoul, C., Balint-Kurti, P. J., 52. Trkola, A., Purtscher, M., Muster, T., Ballaun, C., Buchacher, A., Sullivan,
Rovira, P., Rougé, P., May, G. D., Van Leuven, F., Truffa-Bachi, P., and Van N., Srinivasan, K., Sodroski, J., Moore, J. P., and Katinger, H. (1996) J. Virol.
Damme, E. J. (2000) Planta 211, 546 –554 70, 1100 –1108
22. Koshte, V. L., van Dijk, W., van der Stelt, M. E., and Aalberse, R. C. (1990) 53. Scanlan, C. N., Pantophlet, R., Wormald, M. R., Ollmann Saphire, E., Stan-
Biochem. J. 272, 721–726 field, R., Wilson, I. A., Katinger, H., Dwek, R. A., Rudd, P. M., and Burton,
23. Nakamura-Tsuruta, S., Uchiyama, N., Peumans, W. J., Van Damme, E. J., D. R. (2002) J. Virol. 76, 7306 –7321
Totani, K., Ito, Y., and Hirabayashi, J. (2008) FEBS J. 275, 1227–1239 54. Sanders, R. W., Venturi, M., Schiffner, L., Kalyanaraman, R., Katinger,
24. Mo, H., Winter, H. C., Van Damme, E. J., Peumans, W. J., Misaki, A., and H., Lloyd, K. O., Kwong, P. D., and Moore, J. P. (2002) J. Virol. 76,
Goldstein, I. J. (2001) Eur. J. Biochem. 268, 2609 –2615 7293–7305
25. Meagher, J. L., Winter, H. C., Ezell, P., Goldstein, I. J., and Stuckey, J. A. 55. Mkrtchyan, S. R., Markosyan, R. M., Eadon, M. T., Moore, J. P., Melikyan,
(2005) Glycobiology 15, 1033–1042 G. B., and Cohen, F. S. (2005) J. Virol. 79, 11161–11169
26. Singh, D. D., Saikrishnan, K., Kumar, P., Surolia, A., Sekar, K., and Vijayan, 56. Barrientos, L. G., O’Keefe, B. R., Bray, M., Sanchez, A., Gronenborn, A. M.,
M. (2005) Glycobiology 15, 1025–1032 and Boyd, M. R. (2003) Antiviral. Res. 58, 47–56
27. Jeyaprakash, A. A., Srivastav, A., Surolia, A., and Vijayan, M. (2004) J. Mol. 57. Barrientos, L. G., Lasala, F., Otero, J. R., Sanchez, A., and Delgado, R.
Biol. 338, 757–770 (2004) J. Infect. Dis. 189, 1440 –1443
28. Van Damme, E. J. M., Allen, A. K., and Peumans, W. J. (1987) FEBS Lett. 58. Smee, D. F., Bailey, K. W., Wong, M. H., O’Keefe, B. R., Gustafson, K. R.,
215, 140 –144 Mishin, V. P., and Gubareva, L. V. (2008) Antiviral Res. 80, 266 –271
29. Gartner, S., Markovits, P., Markovitz, D. M., Kaplan, M. H., Gallo, R. C., 59. Keyaerts, E., Vijgen, L., Pannecouque, C., Van Damme, E., Peumans, W.,
and Popovic, M. (1986) Science 233, 215–219 Egberink, H., Balzarini, J., and Van Ranst, M. (2007) Antiviral Res. 75,
30. Li, M., Gao, F., Mascola, J. R., Stamatatos, L., Polonis, V. R., Koutsoukos, 179 –187
M., Voss, G., Goepfert, P., Gilbert, P., Greene, K. M., Bilska, M., Kothe, 60. Helle, F., Wychowski, C., Vu-Dac, N., Gustafson, K. R., Voisset, C., and
D. L., Salazar-Gonzalez, J. F., Wei, X., Decker, J. M., Hahn, B. H., and Dubuisson, J. (2006) J. Biol. Chem. 281, 25177–25183
Montefiori, D. C. (2005) J. Virol. 79, 10108 –10125 61. Bertaux, C., Daelemans, D., Meertens, L., Cormier, E. G., Reinus, J. F.,
31. Wei, X., Decker, J. M., Liu, H., Zhang, Z., Arani, R. B., Kilby, J. M., Saag, Peumans, W. J., Van Damme, E. J., Igarashi, Y., Oki, T., Schols, D., Dragic,
M. S., Wu, X., Shaw, G. M., and Kappes, J. C. (2002) Antimicrob. Agents T., and Balzarini, J. (2007) Virology 366, 40 –50
Chemother. 46, 1896 –1905 62. Witvrouw, M., Fikkert, V., Hantson, A., Pannecouque, C., O’keefe, B. R.,
32. Adachi, A., Gendelman, H. E., Koenig, S., Folks, T., Willey, R., Rabson, A., McMahon, J., Stamatatos, L., de Clercq, E., and Bolmstedt, A. (2005) J. Vi-
and Martin, M. A. (1986) J. Virol. 59, 284 –291 rol. 79, 7777–7784
33. Freed, E. O., Englund, G., and Martin, M. A. (1995) J. Virol. 69, 3949 –3954 63. Wei, X., Decker, J. M., Wang, S., Hui, H., Kappes, J. C., Wu, X., Salazar-
34. Toohey, K., Wehrly, K., Nishio, J., Perryman, S., and Chesebro, B. (1995) Gonzalez, J. F., Salazar, M. G., Kilby, J. M., Saag, M. S., Komarova, N. L.,
Virology 213, 70 –79 Nowak, M. A., Hahn, B. H., Kwong, P. D., and Shaw, G. M. (2003) Nature
35. Chesebro, B., Nishio, J., Perryman, S., Cann, A., O’Brien, W., Chen, I. S., 422, 307–312
and Wehrly, K. (1991) J. Virol. 65, 5782–5789 64. Lis, H., and Sharon, N. (2003) Lectins, 2nd Ed., Kluwer Academic Publish-
36. Chesebro, B., Wehrly, K., Nishio, J., and Perryman, S. (1992) J. Virol. 66, ers Group, Dordrecht, Netherlands
6547– 6554 65. Huskens, D., Vermeire, K., Vandemeulebroucke, E., Balzarini, J., and
37. Collman, R., Balliet, J. W., Gregory, S. A., Friedman, H., Kolson, D. L., Schols, D. (2008) Int. J. Biochem. Cell Biol. 40, 2802–2814
Nathanson, N., and Srinivasan, A. (1992) J. Virol. 66, 7517–7521 66. Saïdi, H., Nasreddine, N., Jenabian, M. A., Lecerf, M., Schols, D., Krief, C.,
38. Derdeyn, C. A., Decker, J. M., Sfakianos, J. N., Wu, X., O’Brien, W. A., Balzarini, J., and Bélec, L. (2007) J. Transl. Med. 5, 28
Ratner, L., Kappes, J. C., Shaw, G. M., and Hunter, E. (2000) J. Virol. 74, 67. Tsai, C. C., Emau, P., Jiang, Y., Tian, B., Morton, W. R., Gustafson, K. R.,
8358 – 8367 and Boyd, M. R. (2003) AIDS Res. Hum. Retroviruses 19, 535–541
39. Platt, E. J., Wehrly, K., Kuhmann, S. E., Chesebro, B., and Kabat, D. (1998) 68. Tsai, C. C., Emau, P., Jiang, Y., Agy, M. B., Shattock, R. J., Schmidt, A.,
J. Virol. 72, 2855–2864 Morton, W. R., Gustafson, K. R., and Boyd, M. R. (2004) AIDS Res. Hum.
40. Takeuchi, Y., McClure, M. O., and Pizzato, M. (2008) J. Virol. 82, Retroviruses 20, 11–18
12585–12588 69. Buffa, V., Stieh, D., Mamhood, N., Hu, Q., Fletcher, P., and Shattock, R. J.

8654 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 12 • MARCH 19, 2010
Inhibition of HIV-1 Replication by BanLec
(2009) J. Gen. Virol. 90, 234 –243 Bakke, J., Mirsalis, J., d’Andrea, A. L., Hume, S. D., Bratcher, B., Saucedo,
70. Zappe, H., Snell, M. E., and Bossard, M. J. (2008) Adv. Drug Deliv. Rev. 60, C. J., McMahon, J. B., Pogue, G. P., and Palmer, K. E. (2009) Proc. Natl.
79 – 87 Acad. Sci. U.S.A. 106, 6099 – 6104
71. Gavrovic-Jankulovic, M., Poulsen, K., Brckalo, T., Bobic, S., Lindner, B., 73. Ziółkowska, N. E., O’Keefe, B. R., Mori, T., Zhu, C., Giomarelli, B., Vojdani,
and Petersen, A. (2008) Int. J. Biochem. Cell Biol. 40, 929 –941 F., Palmer, K. E., McMahon, J. B., and Wlodawer, A. (2006) Structure 14,
72. O’Keefe, B. R., Vojdani, F., Buffa, V., Shattock, R. J., Montefiori, D. C., 1127–1135

Downloaded from www.jbc.org by guest, on October 31, 2010

MARCH 19, 2010 • VOLUME 285 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8655

Vous aimerez peut-être aussi