SlideShare a Scribd company logo
1 of 7
Download to read offline
Determinants in V2C2 region of HIV-1 clade C primary envelopes
conferred altered neutralization susceptibilities to IgG1b12 and
PG9 monoclonal antibodies in a context-dependent manner
Shilpa Patil a,1
, Ipsita Choudhary b,1
, Nakul K. Chaudhary a
, Rajesh Ringe b,2
, Manish Bansal a
,
Brihaspati Narayan Shukla a
, Saikat Boliar a
, Bimal K. Chakrabarti a
, Jayanta Bhattacharya a,n
a
HIV Vaccine Translational Research Laboratory, THSTI-IAVI HIV Vaccine Design Program, Translational Health Science and Technology Institute,
450, Udyog Vihar, Phase-III, Gurgaon 122016, Haryana, India
b
National AIDS Research Institute, Pune, Maharashtra, India
a r t i c l e i n f o
Article history:
Received 31 March 2014
Returned to author for revisions
14 April 2014
Accepted 16 June 2014
Keywords:
HIV-1
Clade C
Envelope
Neutralizing antibody
CD4bs
IgG1b12
VRC01
PG9
PG16
a b s t r a c t
In the present study by examining pseudoviruses expressing patient chimeric envelopes (Envs) made
between an IgG1b12 (b12)-sensitive (2-5.J3) and a b12-resistant (4.J22) HIV-1 clade C envelope, we
identified determinants in the V2C2 region that governed susceptibility to b12 monoclonal antibody, but
not to other CD4 binding site antibodies. Interestingly, when the V2C2 sequence of the 2-5.J3 Env was
transferred to other b12-resistant primary clade C Envs, their susceptibility to b12 varied, indicating that
this effect was context dependent. In addition, we identified determinants within the V2 region in the
b12-resistant envelope that significantly modulated the neutralization of Env-pseudotyped viruses
to PG9/PG16 MAbs. The enhanced neutralization susceptibilities of Envs to b12 and PG9 MAbs were
correlated with increased exposure of their corresponding epitopes highlighting vulnerabilities in the
V2C2 region that altered Env conformation necessary for the efficient accessibility of b12 and PG9
antibodies.
& 2014 Elsevier Inc. All rights reserved.
Introduction
The entry of the Human Immunodeficiency Virus Type 1 (HIV-1)
into the target cells depends primarily on the interaction of gp120 of
trimeric envelope (Env) glycoprotein (gp160) with CD4 and subse-
quently with a coreceptor, usually CCR5 or CXCR4, which facilitates
fusion of viral membrane to cell membrane. In recent years, a
number of broad and potent neutralizing human monoclonal
antibodies were isolated from slow progressing individuals
(Huang et al., 2012; Walker et al., 2011; Walker et al., 2009; Wu
et al., 2010) with unique specificities which has provided clues of
several vulnerabilities associated with immune evasion by HIV-1.
The CD4 binding sites (CD4bs) and quaternary epitopes (QNE) on
the HIV-1 Env trimers are important targets for vaccine develop-
ment. b12 is the first identified neutralizing monoclonal antibody
(MAb) (Burton et al., 1994) whose epitopes in HIV-1 Env overlap
the CD4 binding domain of gp120 (Burton et al., 1994; Mo et al.,
1997). The b12-mediated neutralization has been shown to be
influenced by the mutations in V2 and C3 of gp120 (Mo et al.,
1997). Additionally, altered conformation of Env due to subtle
mutation in variable loops changed the sensitivity of neutraliza-
tion of virus to b12 (Duenas-Decamp et al., 2008; Ly and
Stamatatos, 2000; Utachee et al., 2010). By alanine scanning
mutagenesis, Pantophlet et al. (2003) demonstrated several resi-
dues in gp120 that are important for gp120 recognition by
neutralizing and non-neutralizing MAbs to CD4bs. Other
structure-based studies also demonstrated distinct vulnerable
sites in gp120 conferring b12 susceptibility and/or resistance
(Chen et al., 2009; Liu et al., 2008; Wu et al., 2009; Zhou et al.,
2007). In addition, in monkey models, using SHIV-HXBc2 and
SHIV-89.6P viruses, specific amino acid changes in V2 region
(SHIV-89.6P) and V1, V2, V3, and gp41 regions (SHIV-HXBc2) that
were associated with b12 resistance were documented (Etemad-
Moghadam et al., 1999; Si et al., 2001). Recently, we have also
demonstrated a single amino acid substitution at the 681 position
in the gp41 membrane proximal external region (MPER) enhanced
binding and neutralization of HIV-1 clade A, B and CRF02-AG-235
envelopes to CD4-Ig and b12 MAb, indicating gp41 as referred
Contents lists available at ScienceDirect
journal homepage: www.elsevier.com/locate/yviro
Virology
http://dx.doi.org/10.1016/j.virol.2014.06.018
0042-6822/& 2014 Elsevier Inc. All rights reserved.
n
Corresponding author.
E-mail addresses: JBhattacharya@iavi.org,
JBhattacharya@thsti.res.in (J. Bhattacharya).
1
Equal contribution.
2
Current address: Weill Cornell Medical College, New York, USA.
Virology 462-463 (2014) 266–272
above can also modulate HIV-1 to b12 susceptibility (Ringe and
Bhattacharya, 2012). Very recently Utachee et al. (2014) demon-
strated that the presence of aspartic acid at the position 185 in V2
loop confers HIV-1A/E subtype envelopes with enhanced suscept-
ibility to b12 monoclonal antibodies and that this effect was found
to be not dependent on glycan residues present in 186 and 197
positions previously described to be regulating envelope suscept-
ibility to b12 (Utachee et al., 2010). On the other hand, the variable
loops particularly V1, V2 and V3 regions have also been shown to
be important for the recognition of recently discovered broad and
potent MAbs, such as PG9, PG16, PGT121 and PGT145 (Doores and
Burton, 2010; Moore et al., 2011; Ringe et al., 2012; Thenin et al.,
2012; Walker et al., 2011; Walker et al., 2009). Interestingly, HIV-1
clade C has been widely shown to be refractory to neutralization
by b12 MAb in different geographic settings (Gray et al., 2006;
Kulkarni et al., 2009; Ringe et al., 2010; Zhang et al., 2010).
In natural infection, the antibodies usually develop within the first
few weeks of infection (Moore et al., 1994; Tomaras et al., 2008).
However, the antibodies which neutralize autologous viruses were
found to be elicited after several weeks of initial infection (Montefiori
et al., 2007) and have been demonstrated to exert immune selection
pressure on autologous viral variants (Bunnik et al., 2008; Frost et al.,
2005; Gray et al., 2007; Li et al., 2006; Richman et al., 2003; Wei et al.,
2003). It has been demonstrated earlier that HIV-1 variants that
become resistant to autologous neutralizing antibodies in the course
of infection by virtue of their escape strategies developed different
susceptibilities to monoclonal antibodies and/or heterologous serum
or plasma antibodies (Mascola, 2009; Mascola and Montefiori, 2010).
We previously reported that env clones obtained at different time
points from an Indian patient (NARI-IVC2) with early infection was
unusually sensitive to b12, an MAb that targets CD4 binding site
(CD4bs) on HIV-1 Env (Ringe et al., 2010). This observation prompted
us to investigate the determinants in Env responsible for enhanced
sensitivity of these Envs to b12. Since, pseudoviruses expressing all the
envs obtained from the NARI-IVC2 patient at different time points
showed comparable sensitivity to b12, we examined the b12-sensitive
Env, 2-5.J3 with that of 4.J22, also a clade C but b12-resistant Env
obtained from a different patient (NARI-IVC4) towards mapping
determinants responsible for altered sensitivity to b12. Constructing
the chimeric Envs taking both sensitive (2-5.J3) and resistant (4.J22)
isolates and subsequently by fine mapping, we identified amino acid
residues in V2C2 regions that conferred reciprocal susceptibilities to
b12 and PG9/16 MAbs.
Results and discussion
V2C2 region modulated susceptibility of primary patient-derived Envs
to b12 MAb
We previously showed that HIV-1 clade C env clones obtained
at different time points from an Indian patient (IVC-2) with recent
infection showed enhanced susceptibility to b12 MAb (Ringe et al.,
2010). In order to decipher the enhanced sensitivity of IVC-2 Envs
to b12, we prepared chimeric Env constructs by exchanging
different Env fragments between a b12-sensitive (2-5.J3) Env and
a b12-resistant (4.J22) Env (Ringe et al., 2010) which was obtained
from an unrelated patient (Fig. 1). Since all the env clones that we
obtained from this particular patient showed enhanced b12
sensitivity, we used the most sensitive env (2-5.J3 with IC50 of
0.29 mg/ml) along with an env (4.J22 with IC50 of 450 mg/ml)
obtained from an unrelated patient (NARI-IVC4) to prepare chi-
meric Env constructs. Pseudotyped viruses expressing chimeric
Envs were tested for their sensitivities to b12 MAb, which revealed
that residues within V2 and proximal half of the C2 regions of the
2-5.J3 Env conferred enhanced susceptibility of 2-5.J3 Env to b12
MAb (Fig. 2, top figure). Thus, when the V2C2 region of the 4.J22
Env was substituted with that of 2–5.J3 Env [V1V2C2 (2–5.J3)], the
chimeric virus was shown to become sensitive. In the process of
fine mapping, we ruled out that the V1 and other regions besides
Fig. 1. Construction of chimeric HIV-1 clade C Envs. Env chimeras were prepared by swapping different fragments of the b12-sensitive (2-5.J3) and -resistant (4.J22) Envs. In
addition, specific point substitution of amino acid residues in both the b12-sensitive and -resistant Envs were made. The V2C2 sequence (including the six amino acid
residues modulating PG9/16 sensitivity) that modulated the susceptibility of Env-pseudotyped viruses to b12 MAb is highlighted. The dashes indicate identical amino acid
sequences while dots represent missing sequences.
S. Patil et al. / Virology 462-463 (2014) 266–272 267
V2C2 region (Fig. 1) played any role in altering b12 sensitivity.
Interestingly, apart from having difference in V2 loop lengths,
between 2-5.J3 and 4.J22 Envs, only six amino acid differences
were observed in the V2C2 region. To understand whether these
six residues (P183/D185/N186/N192 in V2 and V207/N215 in C2
regions) were responsible for enhanced susceptibility of 2-5.J3 Env
to b12 MAb, we substituted these residues in 2-5.J3 Env with their
corresponding ones from b12-resistant 4.J22 Env to give rise to
P183S/D185E/N186K/N192K/V207I/N215T [referred to as 2–5.J3 (4.
J22-V2C2/SEKKIT)] and examined their sensitivity to b12 MAb. As
shown in Fig. 2 (middle figure) 2-5.J3 (4.J22-V2C2/SEKKIT) dis-
played comparable b12 sensitivity as 2-5.J3 wild type Env, sug-
gesting that these residues did not play any role in b12
susceptibility and indicated that enhanced susceptibility of 2-5.J3
Env was due to the coordinated effects of residues in the V2
region.
We next examined whether increased sensitivity of 2-5.J3 Env
to b12 was due to increased shedding of gp120. Thus, pseudotyped
viruses expressing 2-5.J3 and V1V2C2 (2-5.J3) Envs were incu-
bated with b12 at a concentration that provided 50% neutraliza-
tion (0.3 mg/ml) and those expressing 4.J22 Env with 10 mg/ml at
37 1C in a CO2 incubator for different times (Fig. S1). The relative
gp120 shedding was assessed by measuring the infectivity of
pseudotyped virus-b12 complex at indicated times in TZM-bl cells.
We found that the reduction in infectivity was comparable
between the b12-sensitive and -resistant variants. Our data
indicated that enhanced sensitivity of 2-5.J3 Env was not due to
b12-induced gp120 shedding.
Substitution of 2-5.J3 V2C2 sequence in unrelated b12-resistant
primary clade C Envs displayed varied b12 and VRC01 susceptibilities
We next examined the ability of the 2-5.J3 V2C2 sequence to
alter b12 susceptibility of few unrelated primary clade C Envs. For
this, we selected four b12-resistant clade C Envs: 3-5.J25, 5.J41, 7.
J20 and 16055-2.3, and substituted their V2C2 with that of 2-5.J3
Env. As shown in Fig. 2 (bottom figure), all the four Envs contain-
ing 2–5.J3 V2C2 sequence differed in their degree of susceptibility
to b12 MAb. While 3-5.J25 and 16055-2.3 Envs containing 2-5.J3
V2C2 sequence displayed substantial and modest increase in b12
susceptibilities respectively compared to their corresponding wild
types, 7.J20 (2-5.J3 V2C2) showed evidence of marginal increase in
b12 susceptibility. Conversely, 2-5.J3 V2C2 sequence was not found
to influence 5.J41 Env in modulating its susceptibility to b12 MAb.
Interestingly, presence of aspartic acid at the position of 185
(D185) (Fig. 1) was not found to affect the neutralization of these
Fig. 2. Neutralization susceptibilities of wild type, chimeric and mutant pseudotyped viruses expressing 2-5.J3 and 4.J22 Envs (top and middle panel) and other unrelated
primary Envs expressing 2-5.J3 V2C2 sequence (bottom panel) to b12 MAb. The neutralization assays were carried out in duplicates in at least three independent time points.
S. Patil et al. / Virology 462-463 (2014) 266–272268
four Envs by b12 MAb, which is in contrast to the observation
reported very recently by Utachee et al. (2014).
Since b12 MAb recognizes residues in gp120 that overlaps
CD4bs, we next examined whether 2-5.J3 V2C2 sequence altered
Env sensitivities to VRC01 (which is another CD4bs recognizing
MAb), QS1420 (anti-CD4 MAb) and sCD4. As shown in Fig. 3, while
all the wild-type Envs and their chimeras (containing 2-5.J3 V2C2
sequence) showed comparable sensitivities to VRC01 MAb, sCD4
and QS1420 MAb, only 4.J22 and 16055-2.3 Envs containing 2-5.J3
V2C2 displayed modest reduction in their sensitivities to VRC01
MAb as compared to their respective wild types. Overall, our data
indicate that the effect of presence of 2-5.J3 V2C2 sequence on
susceptibility of Envs to b12 and VRC01 is context dependent and
likely would vary between Envs.
Determinants in V2C2 region modulated neutralization of env-
pseudotyped viruses by PG9/PG16 MAbs
Since the V2C2 sequences of 2-5.J3 and 4.J22 differ by six
residues (although 4.J22 has shorter V2 loop length compared to
2-5.J3 Env) as described above, we next examined whether these
residues play any role in modulating the sensitivities of these two
Envs to PG and PG16 MAbs as a small change in V2 loop was
previously reported to significantly modulate Env susceptibility to
Envs to PG9/16 MAbs which targets QNE in V2 loop (Doores and
Burton, 2010; Moore et al., 2011; Ringe et al., 2012). As shown in
Fig. 4, we found that introduction of S183/E185/K186/K192/I207/
T215 in 2-5.J3 Env conferred 2-5.J3 (4.J22-V2C2/SEKKIT) pseudo-
virus with a significant increase in susceptibility to PG9 and PG16
MAbs. Conversely, a significant reduction in the sensitivity of 4.J22
Env was observed when specific substitutions of P183/D185/N186/
N192/V207/N215 were made in 4.J22 Env. This effect was found to
be comparable when substitution of only P183/D185/N186/N192
was made in the V2 loop of the 4.J22 Env, suggesting these
minimal changes were sufficient in modulating the exposure of
PG9/16 epitopes that resulted in the reduction in sensitivity of 4.
J22 Env to PG9 and PG16 MAbs and further indicated that N186
and N192 abrogate access of QNE by PG9/16 MAbs. It is to be noted
in this context that substitution of 4.J22 V2C2 with that of 2-5.J3
Env displayed significant reduction in the susceptibility of
PG9/PG16 MAbs, which is likely due to the increase in V2 loop
length as described earlier.
Since an extra glycan residue at the position 192 is present in
the V2 loop of 2-5.J3 Env compared to the 4.J22 Env, we sought to
examine whether this glycan (N192) has any role in modulating
PG9/16 sensitivity. Hence, we substituted N192A in 2-5.J3 Env and
introduced a glycan (K192NSS) in 4.J22 Env and examined its
susceptibility to PG9 and PG16 MAbs. As shown in Fig. 4 (bottom
rows), introduction of glycan in 4.J22 Env (K192NSS) conferred 4.
J22 (K192NSS) Env-pseudotyped virus with significant reduction
in its susceptibility to PG9 and PG16 MAbs; on the contrary, N192A
substitution in 2-5.J3 Env showed marginal increment (though not
significant) in PG9 susceptibility but not to PG16 MAb. These
observations suggest that the presence of glycan at position 192 in
the V2 loop interferes with efficient recognition by PG9/16 MAbs
and likely is dependent on loop length.
In summary, in the present study, using chimeric Envs and
through site-directed mutagenesis, we demonstrated that V2C2
altered b12 susceptibility of HIV-1 clade C Envs in a context-
dependent manner. In addition, we identified minimal residues
in V2 loop that modulated Env susceptibility to PG9/PG16 MAbs.
The neutralization susceptibility of Envs to b12 and PG9/16 MAbs
was found to be associated with exposure of their corresponding
epitopes as determined as wash-out experiments (Fig. 5). We
ruled out that the presence of aspartic acid at the position 185
(D185) in V2 loop as recently demonstrated (Utachee et al., 2014)
is not absolutely required for enhanced susceptibility of Envs to
b12 MAb as 7.J20 wild-type Env although naturally containing
D185, it is resistant to b12 MAb. In addition, 5.J41 (2-5.J3-V2C2)
and 7.J20 (2-5.J3-V2C2) chimeric Envs expressing D185 did not
show any reasonable susceptibility to b12 MAbs. In this paper, we
also demonstrated that the presence of glycan residues at the
positions 186 (N186) and 192 (N192) significantly abrogated access
of PG9/16 MAbs to QNE in the V2 loop of the b12-sensitive 2-5.J3
Env. Our data was substantiated by the fact that N192A alone did
not show any change in improvement of access of PG9/16 MAbs in
2-5.J3 Env. However, in 4.J22 Env, addition of a glycan at the
position 192 (K192NSS) conferred significant reduction in access of
QNE by PG9/16 MAbs, indicating this effect was likely to be
associated with shorter V2 loop length.V1V2 domain in Env plays
a pivotal role in either recognizing the neutralizing antibodies or
Fig. 3. Neutralization susceptibilities of wild type and chimeric Env-pseudotyped viruses to VRC01 MAb (top panel) and to sCD4 and anti-CD4 MAb (QS4120) (bottom panel).
The neutralization assays were carried out in duplicates in at least three independent time points.
S. Patil et al. / Virology 462-463 (2014) 266–272 269
facilitating the escape of HIV-1 variants to the neutralizing anti-
bodies including autologous antibodies, possibly due to its con-
formation as well as glycosylation pattern (Overbaugh and Morris,
2012; Pinter et al., 2004; Rong et al., 2007). Historically, the V1V2
sequences have been shown to undergo changes earlier than that
of the other regions of the Env (Burns and Desrosiers, 1991;
Johnson et al., 1991; Laird et al., 2008; Moore et al., 2011; Moore
et al., 2009; Overbaugh and Rudensey, 1992; Overbaugh et al.,
1991). Our data suggest that the V2C2 region of the 2-5.J3 Env
possibly affected the spike apex region, which in turn influenced
Env to undergo conformational changes resulting in differential
modulation of exposure of epitopes targeted by b12 MAb. Because
conformational change is a complex phenomenon and is often
context dependent, this may possibly explain why despite contain-
ing the same 2-5.J3 V2C2 sequence, other Envs displayed distinct
susceptibilities to b12 MAb.
Materials and methods
HIV-1 Envs and antibodies
The HIV-1 clade C envelopes 2-5.J3, 3-5.J25 and 4.J22 obtained
from Indian patients with recent infection were described pre-
viously (Ringe et al., 2010). 7.J20 is also an HIV-1 clade C Env
cloned in pcDNA3.1D/V5-His-TOPO (Invitrogen Inc.) obtained from
an Indian patient with recent infection, while 16055-2.3 was
kindly provided by Dr. David Montefiori. b12 monoclonal antibody
was kindly provided by Dr. Dennis Burton, and was also obtained
from the NIH AIDS Research Reagent and Reference Program.
VRC01 and sCD4 were obtained from NIH AIDS Research Reagent
and Reference Program. PG9 and PG16 MAbs were kindly provided
by Dr. Wayne Koff, International AIDS Vaccine Initiative (IAVI),
New York. QS4120 MAb was purchased from Calbiochem Inc.
Construction of Env chimera and point mutations
Chimeric Env constructs were made (Fig. 1) by swapping different
variable and constant domains of b12-sensitive Env (2-5.J3) into
resistant Env (4.J22) by blunt ligation of PCR products using either
In-Fusion Dry Down PCR kit (Clontech Inc.) as described before
(Ringe and Bhattacharya, 2012) following the manufacturer's protocol
or by two rounds of overlapping PCR in which first-round amplicons
were used as primers in the second round. Site-directed mutagenesis
to substitute specific residues in V2 and C2 regions were carried out
using tailor-made primers by PCR using high fidelity polymerase,
Platinum Taq (Invitrogen Inc.) or Phusion high fidelity polymerase
(New England Biolabs Inc.). The purified PCR products were then
digested with DpnI joined with backbone plasmid by annealing in the
presence of pox DNA polymerase as described before (Ringe et al.,
2011).
Preparation of Env-pseudotyped viruses and infectivity assay
Env-pseudotyped viruses were prepared by co-transfection of
pSVIIIenv containing patient Envs and pSG3Δenv in 293T cells
using calcium phosphate (Promega, Inc.) as described before
(Ringe et al., 2010). The infectivity assay was carried out in TZM-
bl cells expressing CD4, CCR5 and CXCR4.
Neutralization assay
Neutralization assays were carried out using TZM-bl cells as
described before (Ringe et al., 2010). Env-pseudotyped viruses were
incubated with varying dilutions of plasma (heat-inactivated) and
varying concentrations of b12 and PG9/16 MAbs for 1 h at 37 1C, and
subsequently 1 Â 104
TZM-bl cells were added into the mixture in the
presence of 25 μg/ml DEAE dextran (Sigma Inc.). The plates were
further incubated in a CO2 incubator at 37 1C for two days and the
degree of virus neutralization was assessed by measuring relative
luminescence units (RLU) in a Luminometer (Victor 4, PerkinElmer
Inc.).
Virus-antibody washout assay
In order to assess the degree of exposure of neutralizing
epitopes recognized by b12 and PG9 MAbs, virus antibody wash-
out assay was carried out as essentially described by Chakrabarti
et al. (2011). Briefly, MAbs (b12 and PG9) were serially 5-fold
diluted in 250 ml of 10% DMEM media to which 250 ml of Env-
pseudoviruses were added such that the final concentrations of
Fig. 4. Effect of glycan modification and specific amino acid substitutions in 2–5.J3 and 4.J22 Envs towards modulation of their susceptibilities to PG9 and PG16 MAbs.
The point substitutions were made by site-directed mutagenesis. The data are representation of three independent experiments done in duplicates.
S. Patil et al. / Virology 462-463 (2014) 266–272270
antibodies became 50À0.08 mg/ml in a total volume of 500 ml,
while in the “no inhibitor” control, the same volume of media was
added instead of antibody. The reaction mixture was incubated for
30 min at 37 1C following which 250 ml of reaction mixture was
added in ultra-clear centrifuge tubes (Beckman Coulter, Inc.),
which was diluted to 4 ml by complete DMEM and centrifuged
at 35,000 rpm in an SW60 Ti swinging rotor (Beckman Optima-XE-
100, Inc.) for 2 h at 4 1C. The virus pellet was washed twice with
4 ml of media by centrifuging at 40,000 rpm for 30 min at 4 1C.
After the final wash, 250 ml of DMEM was added to the virus pellet
and suspended by gentle shaking at 4 1C for 30 min. 100 ml of the
suspended virus was used to infect 100 ml of TZM-b cells
(0.2 Â 106
/ml) in duplicate. From the remaining 250 ml of reaction
mixture, an equal volume of the antibody virus mixture was used
as a “no washout” control. Plates were incubated at 37 1C in a CO2
incubator for 2 days, following which the degree of virus neu-
tralization was assessed by measuring the reduction of infection in
TZM-bl cells.
GenBank accession number
The GenBank accession number of HIV-1 clade C Env, 7.J20 is
EU908223.1.
Acknowledgments
This work was supported in part by a grant from the Depart-
ment of Biotechnology, Government of India, to JB (BT/PR12853/
MED/29/141/2009) and by the intramural support from the IAVI-
THSTI HIV Vaccine Design Program. IAVI's work was made possible
by generous support from many donors including: the Bill &
Melinda Gates Foundation; the Ministry of Foreign Affairs of
Denmark; Irish Aid; the Ministry of Finance of Japan; the Ministry
of Foreign Affairs of the Netherlands; the Norwegian Agency for
Development Cooperation (NORAD); the United Kingdom
Department for International Development (DFID); and the United
States Agency for International Development (USAID). The full list
of IAVI donors is available at www.iavi.org. This study was made
possible by the generous support of the American people through
USAID. The contents are the responsibility of the authors and do
not necessarily reflect the views of USAID or the United States
Government. We thank Dr. Dennis Burton, Scripps Research
Institute, La Jolla, California, and Dr. Wayne Koff, International
AIDS Vaccine Initiative, New York, for help with obtaining b12, PG9
and PG16 monoclonal antibodies, respectively. We sincerely
acknowledge the support of the members of the HVTR laboratory,
Prof. G Balakrish Nair, Prof. Sudhanshu Vrati, Dr. Rick King and Dr.
Rajat Goyal. We thank the Director, National AIDS Research
Institute, Pune, for making available the HIV-1 clade C envelope
clones.
Appendix A. Supporting information
Supplementary data associated with this article can be found in
the online version at http://dx.doi.org/10.1016/j.virol.2014.06.018.
References
Bunnik, E.M., Pisas, L., van Nuenen, A.C., Schuitemaker, H., 2008. Autologous
neutralizing humoral immunity and evolution of the viral envelope in the
course of subtype B human immunodeficiency virus type 1 infection. J. Virol.
82, 7932–7941.
Burns, D.P., Desrosiers, R.C., 1991. Selection of genetic variants of simian immuno-
deficiency virus in persistently infected rhesus monkeys. J. Virol. 65,
1843–1854.
Burton, D.R., Pyati, J., Koduri, R., Sharp, S.J., Thornton, G.B., Parren, P.W., Sawyer, L.S.,
Hendry, R.M., Dunlop, N., Nara., P.L., 1994. Efficient neutralization of primary
isolates of HIV- 1 by a recombinant human monoclonal antibody. Science 266,
1024–1027.
Chakrabarti, B.K., Walker, L.M., Guenaga, J.F., Ghobbeh, A., Poignard, P., Burton, D.R.,
Wyatt, R.T., 2011. Direct antibody access to the HIV-1 membrane-proximal
external region positively correlates with neutralization sensitivity. J. Virol. 85,
8217–8226.
Fig. 5. The effect of presence of the 2-5.J3 V2C2 sequence in 4.J22 Env (top panel) and substitution of P183S/D185E/N186K/N192K/V207I/N215T in 2-5.J3 Env (bottom panel)
on exposure of b12 and PG9 epitopes, respectively. Virus-MAb washout assay was carried out using an ultracentrifuge to remove unbound b12 and PG9 MAbs from the
Env-pseudotyped viruses. MAb bound Env-pseudotyped viruses were mixed with TZM-bl cells and reduction in virus infection was assessed in a dose-dependent manner.
S. Patil et al. / Virology 462-463 (2014) 266–272 271
Chen, L., Kwon, Y.D., Zhou, T., Wu, X., O'Dell, S., Cavacini, L., Hessell, A.J., Pancera, M.,
Tang, M., Xu, L., Yang, Z.Y., Zhang, M.Y., Arthos, J., Burton, D.R., Dimitrov, D.S.,
Nabel, G.J., Posner, M.R., Sodroski, J., Wyatt, R., Mascola, J.R., Kwong, P.D., 2009.
Structural basis of immune evasion at the site of CD4 attachment on HIV-1
gp120. Science 326, 1123–1127.
Doores, K.J., Burton, D.R., 2010. Variable loop glycan dependency of the broad and
potent HIV-1-neutralizing antibodies PG9 and PG16. J. Virol. 84, 10510–10521.
Duenas-Decamp, M.J., Peters, P., Burton, D., Clapham, P.R., 2008. Natural resistance of
human immunodeficiency virus type 1 to the CD4bs antibody b12 conferred by a
glycan and an arginine residue close to the CD4 binding loop. J. Virol. 82, 5807–5814.
Etemad-Moghadam, B., Sun, Y., Nicholson, E.K., Karlsson, G.B., Schenten, D.,
Sodroski, J., 1999. Determinants of neutralization resistance in the envelope
glycoproteins of a simian-human immunodeficiency virus passaged in vivo.
J. Virol. 73, 8873–8879.
Frost, S.D., Wrin, T., Smith, D.M., Kosakovsky Pond, S.L., Liu, Y., Paxinos, E., Chappey,
C., Galovich, J., Beauchaine, J., Petropoulos, C.J., Little, S.J., Richman, D.D., 2005.
Neutralizing antibody responses drive the evolution of human immunodefi-
ciency virus type 1 envelope during recent HIV infection. Proc. Natl. Acad. Sci.
U. S. A. 102, 18514–18519.
Gray, E.S., Meyers, T., Gray, G., Montefiori, D.C., Morris, L., 2006. Insensitivity of
paediatric HIV-1 subtype C viruses to broadly neutralising monoclonal anti-
bodies raised against subtype B. PLoS Med. 3, e255.
Gray, E.S., Moore, P.L., Choge, I.A., Decker, J.M., Bibollet-Ruche, F., Li, H., Leseka, N.,
Treurnicht, F., Mlisana, K., Shaw, G.M., Karim, S.S., Williamson, C., Morris, L.,
2007. Neutralizing antibody responses in acute human immunodeficiency virus
type 1 subtype C infection. J. Virol. 81, 6187–6196.
Huang, J., Ofek, G., Laub, L., Louder, M.K., Doria-Rose, N.A., Longo, N.S., Imamichi, H.,
Bailer, R.T., Chakrabarti, B., Sharma, S.K., Alam, S.M., Wang, T., Yang, Y., Zhang, B.,
Migueles, S.A., Wyatt, R., Haynes, B.F., Kwong, P.D., Mascola, J.R., Connors, M.,
2012. Broad and potent neutralization of HIV-1 by a gp41-specific human
antibody. Nature 491, 406–412.
Johnson, P.R., Hamm, T.E., Goldstein, S., Kitov, S., Hirsch, V.M., 1991. The genetic fate
of molecularly cloned simian immunodeficiency virus in experimentally
infected macaques. Virology 185, 217–228.
Kulkarni, S.S., Lapedes, A., Tang, H., Gnanakaran, S., Daniels, M.G., Zhang, M.,
Bhattacharya, T., Li, M., Polonis, V.R., McCutchan, F.E., Morris, L., Ellenberger,
D., Butera, S.T., Bollinger, R.C., Korber, B.T., Paranjape, R.S., Montefiori, D.C.,
2009. Highly complex neutralization determinants on a monophyletic lineage
of newly transmitted subtype C HIV-1 Env clones from India. Virology 385,
505–520.
Laird, M.E., Igarashi, T., Martin, M.A., Desrosiers, R.C., 2008. Importance of the V1/V2
loop region of simian-human immunodeficiency virus envelope glycoprotein
gp120 in determining the strain specificity of the neutralizing antibody
response. J. Virol. 82, 11054–11065.
Li, B., Decker, J.M., Johnson, R.W., Bibollet-Ruche, F., Wei, X., Mulenga, J., Allen, S.,
Hunter, E., Hahn, B.H., Shaw, G.M., Blackwell, J.L., Derdeyn, C.A., 2006. Evidence
for potent autologous neutralizing antibody titers and compact envelopes in
early infection with subtype C human immunodeficiency virus type 1. J. Virol.
80, 5211–5218.
Liu, J., Bartesaghi, A., Borgnia, M.J., Sapiro, G., Subramaniam, S., 2008. Molecular
architecture of native HIV-1 gp120 trimers. Nature 455, 109–113.
Ly, A., Stamatatos, L., 2000. V2 loop glycosylation of the human immunodeficiency
virus type 1 SF162 envelope facilitates interaction of this protein with CD4 and
CCR5 receptors and protects the virus from neutralization by anti-V3 loop and
anti-CD4 binding site antibodies. J. Virol. 74, 6769–6776.
Mascola, J.R., 2009. The cat and mouse of HIV-1 antibody escape. PLoS Pathog. 5,
e1000592.
Mascola, J.R., Montefiori, D.C., 2010. The role of antibodies in HIV vaccines. Annu.
Rev. Immunol. 28, 413–444.
Mo, H., Stamatatos, L., Ip, J.E., Barbas, C.F., Parren, P.W., Burton, D.R., Moore, J.P., Ho,
D.D., 1997. Human immunodeficiency virus type 1 mutants that escape
neutralization by human monoclonal antibody IgG1b12. off. J. Virol. 71,
6869–6874.
Montefiori, D.C., Morris, L., Ferrari, G., Mascola, J.R., 2007. Neutralizing and other
antiviral antibodies in HIV-1 infection and vaccination. Curr. Opin. HIV AIDS 2,
169–176.
Moore, J.P., Cao, Y., Ho, D.D., Koup, R.A., 1994. Development of the anti-gp120
antibody response during seroconversion to human immunodeficiency virus
type 1. J. Virol. 68, 5142–5155.
Moore, P.L., Gray, E.S., Sheward, D., Madiga, M., Ranchobe, N., Lai, Z., Honnen, W.J.,
Nonyane, M., Tumba, N., Hermanus, T., Sibeko, S., Mlisana, K., Abdool Karim, S.
S., Williamson, C., Pinter, A., Morris, L., 2011. Potent and broad neutralization of
HIV-1 subtype C by plasma antibodies targeting a quaternary epitope including
residues in the V2 loop. J. Virol. 85, 3128–3141.
Moore, P.L., Ranchobe, N., Lambson, B.E., Gray, E.S., Cave, E., Abrahams, M.R.,
Bandawe, G., Mlisana, K., Abdool Karim, S.S., Williamson, C., Morris, L., 2009.
Limited neutralizing antibody specificities drive neutralization escape in early
HIV-1 subtype C infection. PLoS Pathog. 5, e1000598.
Overbaugh, J., Morris, L., 2012. The Antibody Response against HIV-1. Cold Spring
Harb. Perspect. Med. 2, a007039.
Overbaugh, J., Rudensey, L.M., 1992. Alterations in potential sites for glycosylation
predominate during evolution of the simian immunodeficiency virus envelope
gene in macaques. J. Virol. 66, 5937–5948.
Overbaugh, J., Rudensey, L.M., Papenhausen, M.D., Benveniste, R.E., Morton, W.R.,
1991. Variation in simian immunodeficiency virus env is confined to V1 and V4
during progression to simian AIDS. J. Virol. 65, 7025–7031.
Pantophlet, R., Ollmann Saphire, E., Poignard, P., Parren, P.W., Wilson, I.A., Burton, D.
R., 2003. Fine mapping of the interaction of neutralizing and nonneutralizing
monoclonal antibodies with the CD4 binding site of human immunodeficiency
virus type 1 gp120. J. Virol. 77, 642–658.
Pinter, A., Honnen, W.J., He, Y., Gorny, M.K., Zolla-Pazner, S., Kayman, S.C., 2004. The
V1/V2 domain of gp120 is a global regulator of the sensitivity of primary
human immunodeficiency virus type 1 isolates to neutralization by antibodies
commonly induced upon infection. J. Virol. 78, 5205–5215.
Richman, D.D., Wrin, T., Little, S.J., Petropoulos, C.J., 2003. Rapid evolution of the
neutralizing antibody response to HIV type 1 infection. Proc. Natl. Acad. Sci. U.
S. A. 100, 4144–4149.
Ringe, R., Bhattacharya, J., 2012. Association of enhanced HIV-1 neutralization by a
single Y681H substitution in gp41 with increased gp120-CD4 interaction and
macrophage Infectivity. PLoS One 7, e37157.
Ringe, R., Phogat, S., Bhattacharya, J., 2012. Subtle alteration of residues including
N-linked glycans in V2 loop modulates HIV-1 neutralization by PG9 and PG16
monoclonal antibodies. Virology 426, 34–41.
Ringe, R., Sharma, D., Zolla-Pazner, S., Phogat, S., Risbud, A., Thakar, M., Paranjape,
R., Bhattacharya, J., 2011. A single amino acid substitution in the C4 region in
gp120 confers enhanced neutralization of HIV-1 by modulating CD4 binding
sites and V3 loop. Virology 418, 123–132.
Ringe, R., Thakar, M., Bhattacharya, J., 2010. Variations in autologous neutralization
and CD4 dependence of b12 resistant HIV-1 clade C env clones obtained at
different time points from antiretroviral naive Indian patients with recent
infection. Retrovirology 7, 76.
Rong, R., Bibollet-Ruche, F., Mulenga, J., Allen, S., Blackwell, J.L., Derdeyn, C.A., 2007.
Role of V1V2 and other human immunodeficiency virus type 1 envelope
domains in resistance to autologous neutralization during clade C infection.
J. Virol. 81, 1350–1359.
Si, Z., Cayabyab, M., Sodroski, J., 2001. Envelope glycoprotein determinants of
neutralization resistance in a simian-human immunodeficiency virus (SHIV-
HXBc2P 3.2) derived by passage in monkeys. J. Virol. 75, 4208–4218.
Thenin, S., Roch, E., Samleerat, T., Moreau, T., Chaillon, A., Moreau, A., Barin, F.,
Braibant, M., 2012. Naturally occurring substitutions of conserved residues in
HIV-1 variants of different clades are involved in PG9 and PG16 resistance to
neutralization. J. Gen. Virol. 93, 1495–1505.
Tomaras, G.D., Yates, N.L., Liu, P., Qin, L., Fouda, G.G., Chavez, L.L., Decamp, A.C.,
Parks, R.J., Ashley, V.C., Lucas, J.T., Cohen, M., Eron, J., Hicks, C.B., Liao, H.X., Self,
S.G., Landucci, G., Forthal, D.N., Weinhold, K.J., Keele, B.F., Hahn, B.H., Greenberg,
M.L., Morris, L., Karim, S.S., Blattner, W.A., Montefiori, D.C., Shaw, G.M.,
Perelson, A.S., Haynes, B.F., 2008. Initial B-cell responses to transmitted human
immunodeficiency virus type 1: virion-binding immunoglobulin M (IgM) and
IgG antibodies followed by plasma anti-gp41 antibodies with ineffective
control of initial viremia. J. Virol. 82, 12449–12463.
Utachee, P., Isarangkura-na-ayuthaya, P., Tokunaga, K., Ikuta, K., Takeda, N.,
Kameoka, M., 2014. Impact of amino acid substitutions in the V2 and C2
regions of human immunodeficiency virus type 1 CRF01_AE envelope glyco-
protein gp120 on viral neutralization susceptibility to broadly neutralizing
antibodies specific for the CD4 binding site. Retrovirology 11, 32.
Utachee, P., Nakamura, S., Isarangkura-Na-Ayuthaya, P., Tokunaga, K., Sawanpanya-
lert, P., Ikuta, K., Auwanit, W., Kameoka, M., 2010. Two N-linked glycosylation
sites in the V2 and C2 regions of human immunodeficiency virus type
1 CRF01_AE envelope glycoprotein gp120 regulate viral neutralization suscept-
ibility to the human monoclonal antibody specific for the CD4 binding domain.
J. Virol. 84, 4311–4320.
Walker, L.M., Huber, M., Doores, K.J., Falkowska, E., Pejchal, R., Julien, J.P., Wang, S.K.,
Ramos, A., Chan-Hui, P.Y., Moyle, M., Mitcham, J.L., Hammond, P.W., Olsen, O.A.,
Phung, P., Fling, S., Wong, C.H., Phogat, S., Wrin, T., Simek, M.D., 2011. Broad
neutralization coverage of HIV by multiple highly potent antibodies (Principal
Investigators, P.G., Koff, W.C., Wilson, I.A., Burton, D.R., Poignard, P.). Nature 477,
466–470.
Walker, L.M., Phogat, S.K., Chan-Hui, P.Y., Wagner, D., Phung, P., Goss, J.L., Wrin, T.,
Simek, M.D., Fling, S., Mitcham, J.L., Lehrman, J.K., Priddy, F.H., Olsen, O.A., Frey,
S.M., Hammond, P.W., Kaminsky, S., Zamb, T., Moyle, M., Koff, W.C., Poignard, P.,
Burton, D.R., 2009. Broad and potent neutralizing antibodies from an African
donor reveal a new HIV-1 vaccine target. Science 326, 285–289.
Wei, X., Decker, J.M., Wang, S., Hui, H., Kappes, J.C., Wu, X., Salazar-Gonzalez, J.F.,
Salazar, M.G., Kilby, J.M., Saag, M.S., Komarova, N.L., Nowak, M.A., Hahn, B.H.,
Kwong, P.D., Shaw, G.M., 2003. Antibody neutralization and escape by HIV-1.
Nature 422, 307–312.
Wu, X., Yang, Z.Y., Li, Y., Hogerkorp, C.M., Schief, W.R., Seaman, M.S., Zhou, T., Schmidt,
S.D., Wu, L., Xu, L., Longo, N.S., McKee, K., O'Dell, S., Louder, M.K., Wycuff, D.L., Feng,
Y., Nason, M., Doria-Rose, N., Connors, M., Kwong, P.D., Roederer, M., Wyatt, R.T.,
Nabel, G.J., Mascola, J.R., 2010. Rational design of envelope identifies broadly
neutralizing human monoclonal antibodies to HIV-1. Science 329, 856–861.
Wu, X., Zhou, T., O'Dell, S., Wyatt, R.T., Kwong, P.D., Mascola, J.R., 2009. Mechanism
of human immunodeficiency virus type 1 resistance to monoclonal antibody
B12 that effectively targets the site of CD4 attachment. J. Virol. 83,
10892–10907.
Zhang, H., Rola, M., West, J.T., Tully, D.C., Kubis, P., He, J., Kankasa, C., Wood, C., 2010.
Functional properties of the HIV-1 subtype C envelope glycoprotein associated
with mother-to-child transmission. Virology 400, 164–174.
Zhou, T., Xu, L., Dey, B., Hessell, A.J., Van Ryk, D., Xiang, S.H., Yang, X., Zhang, M.Y.,
Zwick, M.B., Arthos, J., Burton, D.R., Dimitrov, D.S., Sodroski, J., Wyatt, R., Nabel,
G.J., Kwong, P.D., 2007. Structural definition of a conserved neutralization
epitope on HIV-1 gp120. Nature 445, 732–737.
S. Patil et al. / Virology 462-463 (2014) 266–272272

More Related Content

What's hot

Coreceptor usage
Coreceptor usageCoreceptor usage
Coreceptor usagemakarandptl
 
In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus
In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus
In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus Yordan Penev
 
Test 1 article
Test 1 articleTest 1 article
Test 1 articleGreg
 
In vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconIn vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconBinodGupta27
 
Beltwide poster main
Beltwide poster mainBeltwide poster main
Beltwide poster mainSameer Khanal
 
Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06Kimberly Schafer
 
Protein engineering presentation
Protein engineering presentationProtein engineering presentation
Protein engineering presentationHuzaifa Ummar
 
2016.05.19 KromTiD web powerpoint
2016.05.19 KromTiD web powerpoint2016.05.19 KromTiD web powerpoint
2016.05.19 KromTiD web powerpointGretchen Pratt
 
Recombinant Origin of the Retrovirus XMRV
Recombinant Origin of the Retrovirus XMRVRecombinant Origin of the Retrovirus XMRV
Recombinant Origin of the Retrovirus XMRVdegarden
 
TransVax Phase 2 Trial Results
TransVax Phase 2 Trial ResultsTransVax Phase 2 Trial Results
TransVax Phase 2 Trial ResultsVicalInc
 
Seminario biomol
Seminario biomolSeminario biomol
Seminario biomolLala Duque
 
J. Virol.-2009-Wang-7166-75
J. Virol.-2009-Wang-7166-75J. Virol.-2009-Wang-7166-75
J. Virol.-2009-Wang-7166-75Morteza Loghmani
 

What's hot (18)

abbas morovvati
abbas morovvatiabbas morovvati
abbas morovvati
 
Coreceptor usage
Coreceptor usageCoreceptor usage
Coreceptor usage
 
In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus
In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus
In Vitro Characterization of a Novel Cis-acting Element (NCE) in the Cd4 Locus
 
Test 1 article
Test 1 articleTest 1 article
Test 1 article
 
In vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic repliconIn vitro transcription and transfection of HCV genomic replicon
In vitro transcription and transfection of HCV genomic replicon
 
Beltwide poster main
Beltwide poster mainBeltwide poster main
Beltwide poster main
 
Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06
 
Protein engineering presentation
Protein engineering presentationProtein engineering presentation
Protein engineering presentation
 
2016.05.19 KromTiD web powerpoint
2016.05.19 KromTiD web powerpoint2016.05.19 KromTiD web powerpoint
2016.05.19 KromTiD web powerpoint
 
FINAL PUBLISHED PAPER SEP_2015
FINAL PUBLISHED PAPER SEP_2015FINAL PUBLISHED PAPER SEP_2015
FINAL PUBLISHED PAPER SEP_2015
 
Science
ScienceScience
Science
 
Recombinant Origin of the Retrovirus XMRV
Recombinant Origin of the Retrovirus XMRVRecombinant Origin of the Retrovirus XMRV
Recombinant Origin of the Retrovirus XMRV
 
Homology directed repair (HDR) Knock-in
Homology directed repair (HDR) Knock-inHomology directed repair (HDR) Knock-in
Homology directed repair (HDR) Knock-in
 
TransVax Phase 2 Trial Results
TransVax Phase 2 Trial ResultsTransVax Phase 2 Trial Results
TransVax Phase 2 Trial Results
 
Seminario biomol
Seminario biomolSeminario biomol
Seminario biomol
 
Sigma Xi
Sigma XiSigma Xi
Sigma Xi
 
Delta 32[1]
Delta 32[1]Delta 32[1]
Delta 32[1]
 
J. Virol.-2009-Wang-7166-75
J. Virol.-2009-Wang-7166-75J. Virol.-2009-Wang-7166-75
J. Virol.-2009-Wang-7166-75
 

Similar to Patil S et al 2014 Virology

Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016SHILPA PATIL
 
Senior Thesis_UC Davis_Lauren Drayer
Senior Thesis_UC Davis_Lauren DrayerSenior Thesis_UC Davis_Lauren Drayer
Senior Thesis_UC Davis_Lauren DrayerLauren Drayer
 
Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfstudy help
 
Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfstudy help
 
Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)SHILPA PATIL
 
Covid, lo studio sugli anticorpi di guariti e obesi
Covid, lo studio sugli anticorpi di guariti e obesiCovid, lo studio sugli anticorpi di guariti e obesi
Covid, lo studio sugli anticorpi di guariti e obesiilfattoquotidiano.it
 
SARS vaccine development.pdf
SARS vaccine development.pdfSARS vaccine development.pdf
SARS vaccine development.pdfssuserdd2585
 
Molecular characterisation, attenuation and inactivation of very virulent inf...
Molecular characterisation, attenuation and inactivation of very virulent inf...Molecular characterisation, attenuation and inactivation of very virulent inf...
Molecular characterisation, attenuation and inactivation of very virulent inf...Majed Mohammed
 
Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...
Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...
Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...USTC, Hefei, PRC
 
SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...
SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...
SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...Henok Abebe
 
how immunity generated from covid 19 vaccines differs from
how immunity generated from covid 19 vaccines differs fromhow immunity generated from covid 19 vaccines differs from
how immunity generated from covid 19 vaccines differs fromDr-Ajay Tripathi
 
Choose one of the psychological disorders discusse
Choose   one   of   the   psychological   disorders   discusseChoose   one   of   the   psychological   disorders   discusse
Choose one of the psychological disorders discusseJinElias52
 
Sars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamosSars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamosJauru Freitas
 
HIV-1 PATHOGENESIS AND GENETIC FORMS
HIV-1 PATHOGENESIS AND GENETIC FORMSHIV-1 PATHOGENESIS AND GENETIC FORMS
HIV-1 PATHOGENESIS AND GENETIC FORMSdivyabms
 
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Dr. Md. Ehsanul Haque
 

Similar to Patil S et al 2014 Virology (20)

Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016
 
Senior Thesis_UC Davis_Lauren Drayer
Senior Thesis_UC Davis_Lauren DrayerSenior Thesis_UC Davis_Lauren Drayer
Senior Thesis_UC Davis_Lauren Drayer
 
GP120 pdf
GP120 pdfGP120 pdf
GP120 pdf
 
COVID-19.pptx
COVID-19.pptxCOVID-19.pptx
COVID-19.pptx
 
Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdf
 
Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdf
 
Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)
 
Deshpande et al.,Retrovirology 2016
Deshpande et al.,Retrovirology 2016Deshpande et al.,Retrovirology 2016
Deshpande et al.,Retrovirology 2016
 
Covid, lo studio sugli anticorpi di guariti e obesi
Covid, lo studio sugli anticorpi di guariti e obesiCovid, lo studio sugli anticorpi di guariti e obesi
Covid, lo studio sugli anticorpi di guariti e obesi
 
ISMB2010_Poster
ISMB2010_PosterISMB2010_Poster
ISMB2010_Poster
 
SARS vaccine development.pdf
SARS vaccine development.pdfSARS vaccine development.pdf
SARS vaccine development.pdf
 
Molecular characterisation, attenuation and inactivation of very virulent inf...
Molecular characterisation, attenuation and inactivation of very virulent inf...Molecular characterisation, attenuation and inactivation of very virulent inf...
Molecular characterisation, attenuation and inactivation of very virulent inf...
 
Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...
Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...
Protective ZIKV Vaccines Engineered to eliminate enhancement of dengue infect...
 
SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...
SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...
SEROPREVALENTSTATUSANDGENETICDIVERSITYOFINFECTIOUSBURSALDISEASEIBDVVIRUSOFPOU...
 
how immunity generated from covid 19 vaccines differs from
how immunity generated from covid 19 vaccines differs fromhow immunity generated from covid 19 vaccines differs from
how immunity generated from covid 19 vaccines differs from
 
Choose one of the psychological disorders discusse
Choose   one   of   the   psychological   disorders   discusseChoose   one   of   the   psychological   disorders   discusse
Choose one of the psychological disorders discusse
 
Jiy302
Jiy302Jiy302
Jiy302
 
Sars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamosSars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamos
 
HIV-1 PATHOGENESIS AND GENETIC FORMS
HIV-1 PATHOGENESIS AND GENETIC FORMSHIV-1 PATHOGENESIS AND GENETIC FORMS
HIV-1 PATHOGENESIS AND GENETIC FORMS
 
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
 

Patil S et al 2014 Virology

  • 1. Determinants in V2C2 region of HIV-1 clade C primary envelopes conferred altered neutralization susceptibilities to IgG1b12 and PG9 monoclonal antibodies in a context-dependent manner Shilpa Patil a,1 , Ipsita Choudhary b,1 , Nakul K. Chaudhary a , Rajesh Ringe b,2 , Manish Bansal a , Brihaspati Narayan Shukla a , Saikat Boliar a , Bimal K. Chakrabarti a , Jayanta Bhattacharya a,n a HIV Vaccine Translational Research Laboratory, THSTI-IAVI HIV Vaccine Design Program, Translational Health Science and Technology Institute, 450, Udyog Vihar, Phase-III, Gurgaon 122016, Haryana, India b National AIDS Research Institute, Pune, Maharashtra, India a r t i c l e i n f o Article history: Received 31 March 2014 Returned to author for revisions 14 April 2014 Accepted 16 June 2014 Keywords: HIV-1 Clade C Envelope Neutralizing antibody CD4bs IgG1b12 VRC01 PG9 PG16 a b s t r a c t In the present study by examining pseudoviruses expressing patient chimeric envelopes (Envs) made between an IgG1b12 (b12)-sensitive (2-5.J3) and a b12-resistant (4.J22) HIV-1 clade C envelope, we identified determinants in the V2C2 region that governed susceptibility to b12 monoclonal antibody, but not to other CD4 binding site antibodies. Interestingly, when the V2C2 sequence of the 2-5.J3 Env was transferred to other b12-resistant primary clade C Envs, their susceptibility to b12 varied, indicating that this effect was context dependent. In addition, we identified determinants within the V2 region in the b12-resistant envelope that significantly modulated the neutralization of Env-pseudotyped viruses to PG9/PG16 MAbs. The enhanced neutralization susceptibilities of Envs to b12 and PG9 MAbs were correlated with increased exposure of their corresponding epitopes highlighting vulnerabilities in the V2C2 region that altered Env conformation necessary for the efficient accessibility of b12 and PG9 antibodies. & 2014 Elsevier Inc. All rights reserved. Introduction The entry of the Human Immunodeficiency Virus Type 1 (HIV-1) into the target cells depends primarily on the interaction of gp120 of trimeric envelope (Env) glycoprotein (gp160) with CD4 and subse- quently with a coreceptor, usually CCR5 or CXCR4, which facilitates fusion of viral membrane to cell membrane. In recent years, a number of broad and potent neutralizing human monoclonal antibodies were isolated from slow progressing individuals (Huang et al., 2012; Walker et al., 2011; Walker et al., 2009; Wu et al., 2010) with unique specificities which has provided clues of several vulnerabilities associated with immune evasion by HIV-1. The CD4 binding sites (CD4bs) and quaternary epitopes (QNE) on the HIV-1 Env trimers are important targets for vaccine develop- ment. b12 is the first identified neutralizing monoclonal antibody (MAb) (Burton et al., 1994) whose epitopes in HIV-1 Env overlap the CD4 binding domain of gp120 (Burton et al., 1994; Mo et al., 1997). The b12-mediated neutralization has been shown to be influenced by the mutations in V2 and C3 of gp120 (Mo et al., 1997). Additionally, altered conformation of Env due to subtle mutation in variable loops changed the sensitivity of neutraliza- tion of virus to b12 (Duenas-Decamp et al., 2008; Ly and Stamatatos, 2000; Utachee et al., 2010). By alanine scanning mutagenesis, Pantophlet et al. (2003) demonstrated several resi- dues in gp120 that are important for gp120 recognition by neutralizing and non-neutralizing MAbs to CD4bs. Other structure-based studies also demonstrated distinct vulnerable sites in gp120 conferring b12 susceptibility and/or resistance (Chen et al., 2009; Liu et al., 2008; Wu et al., 2009; Zhou et al., 2007). In addition, in monkey models, using SHIV-HXBc2 and SHIV-89.6P viruses, specific amino acid changes in V2 region (SHIV-89.6P) and V1, V2, V3, and gp41 regions (SHIV-HXBc2) that were associated with b12 resistance were documented (Etemad- Moghadam et al., 1999; Si et al., 2001). Recently, we have also demonstrated a single amino acid substitution at the 681 position in the gp41 membrane proximal external region (MPER) enhanced binding and neutralization of HIV-1 clade A, B and CRF02-AG-235 envelopes to CD4-Ig and b12 MAb, indicating gp41 as referred Contents lists available at ScienceDirect journal homepage: www.elsevier.com/locate/yviro Virology http://dx.doi.org/10.1016/j.virol.2014.06.018 0042-6822/& 2014 Elsevier Inc. All rights reserved. n Corresponding author. E-mail addresses: JBhattacharya@iavi.org, JBhattacharya@thsti.res.in (J. Bhattacharya). 1 Equal contribution. 2 Current address: Weill Cornell Medical College, New York, USA. Virology 462-463 (2014) 266–272
  • 2. above can also modulate HIV-1 to b12 susceptibility (Ringe and Bhattacharya, 2012). Very recently Utachee et al. (2014) demon- strated that the presence of aspartic acid at the position 185 in V2 loop confers HIV-1A/E subtype envelopes with enhanced suscept- ibility to b12 monoclonal antibodies and that this effect was found to be not dependent on glycan residues present in 186 and 197 positions previously described to be regulating envelope suscept- ibility to b12 (Utachee et al., 2010). On the other hand, the variable loops particularly V1, V2 and V3 regions have also been shown to be important for the recognition of recently discovered broad and potent MAbs, such as PG9, PG16, PGT121 and PGT145 (Doores and Burton, 2010; Moore et al., 2011; Ringe et al., 2012; Thenin et al., 2012; Walker et al., 2011; Walker et al., 2009). Interestingly, HIV-1 clade C has been widely shown to be refractory to neutralization by b12 MAb in different geographic settings (Gray et al., 2006; Kulkarni et al., 2009; Ringe et al., 2010; Zhang et al., 2010). In natural infection, the antibodies usually develop within the first few weeks of infection (Moore et al., 1994; Tomaras et al., 2008). However, the antibodies which neutralize autologous viruses were found to be elicited after several weeks of initial infection (Montefiori et al., 2007) and have been demonstrated to exert immune selection pressure on autologous viral variants (Bunnik et al., 2008; Frost et al., 2005; Gray et al., 2007; Li et al., 2006; Richman et al., 2003; Wei et al., 2003). It has been demonstrated earlier that HIV-1 variants that become resistant to autologous neutralizing antibodies in the course of infection by virtue of their escape strategies developed different susceptibilities to monoclonal antibodies and/or heterologous serum or plasma antibodies (Mascola, 2009; Mascola and Montefiori, 2010). We previously reported that env clones obtained at different time points from an Indian patient (NARI-IVC2) with early infection was unusually sensitive to b12, an MAb that targets CD4 binding site (CD4bs) on HIV-1 Env (Ringe et al., 2010). This observation prompted us to investigate the determinants in Env responsible for enhanced sensitivity of these Envs to b12. Since, pseudoviruses expressing all the envs obtained from the NARI-IVC2 patient at different time points showed comparable sensitivity to b12, we examined the b12-sensitive Env, 2-5.J3 with that of 4.J22, also a clade C but b12-resistant Env obtained from a different patient (NARI-IVC4) towards mapping determinants responsible for altered sensitivity to b12. Constructing the chimeric Envs taking both sensitive (2-5.J3) and resistant (4.J22) isolates and subsequently by fine mapping, we identified amino acid residues in V2C2 regions that conferred reciprocal susceptibilities to b12 and PG9/16 MAbs. Results and discussion V2C2 region modulated susceptibility of primary patient-derived Envs to b12 MAb We previously showed that HIV-1 clade C env clones obtained at different time points from an Indian patient (IVC-2) with recent infection showed enhanced susceptibility to b12 MAb (Ringe et al., 2010). In order to decipher the enhanced sensitivity of IVC-2 Envs to b12, we prepared chimeric Env constructs by exchanging different Env fragments between a b12-sensitive (2-5.J3) Env and a b12-resistant (4.J22) Env (Ringe et al., 2010) which was obtained from an unrelated patient (Fig. 1). Since all the env clones that we obtained from this particular patient showed enhanced b12 sensitivity, we used the most sensitive env (2-5.J3 with IC50 of 0.29 mg/ml) along with an env (4.J22 with IC50 of 450 mg/ml) obtained from an unrelated patient (NARI-IVC4) to prepare chi- meric Env constructs. Pseudotyped viruses expressing chimeric Envs were tested for their sensitivities to b12 MAb, which revealed that residues within V2 and proximal half of the C2 regions of the 2-5.J3 Env conferred enhanced susceptibility of 2-5.J3 Env to b12 MAb (Fig. 2, top figure). Thus, when the V2C2 region of the 4.J22 Env was substituted with that of 2–5.J3 Env [V1V2C2 (2–5.J3)], the chimeric virus was shown to become sensitive. In the process of fine mapping, we ruled out that the V1 and other regions besides Fig. 1. Construction of chimeric HIV-1 clade C Envs. Env chimeras were prepared by swapping different fragments of the b12-sensitive (2-5.J3) and -resistant (4.J22) Envs. In addition, specific point substitution of amino acid residues in both the b12-sensitive and -resistant Envs were made. The V2C2 sequence (including the six amino acid residues modulating PG9/16 sensitivity) that modulated the susceptibility of Env-pseudotyped viruses to b12 MAb is highlighted. The dashes indicate identical amino acid sequences while dots represent missing sequences. S. Patil et al. / Virology 462-463 (2014) 266–272 267
  • 3. V2C2 region (Fig. 1) played any role in altering b12 sensitivity. Interestingly, apart from having difference in V2 loop lengths, between 2-5.J3 and 4.J22 Envs, only six amino acid differences were observed in the V2C2 region. To understand whether these six residues (P183/D185/N186/N192 in V2 and V207/N215 in C2 regions) were responsible for enhanced susceptibility of 2-5.J3 Env to b12 MAb, we substituted these residues in 2-5.J3 Env with their corresponding ones from b12-resistant 4.J22 Env to give rise to P183S/D185E/N186K/N192K/V207I/N215T [referred to as 2–5.J3 (4. J22-V2C2/SEKKIT)] and examined their sensitivity to b12 MAb. As shown in Fig. 2 (middle figure) 2-5.J3 (4.J22-V2C2/SEKKIT) dis- played comparable b12 sensitivity as 2-5.J3 wild type Env, sug- gesting that these residues did not play any role in b12 susceptibility and indicated that enhanced susceptibility of 2-5.J3 Env was due to the coordinated effects of residues in the V2 region. We next examined whether increased sensitivity of 2-5.J3 Env to b12 was due to increased shedding of gp120. Thus, pseudotyped viruses expressing 2-5.J3 and V1V2C2 (2-5.J3) Envs were incu- bated with b12 at a concentration that provided 50% neutraliza- tion (0.3 mg/ml) and those expressing 4.J22 Env with 10 mg/ml at 37 1C in a CO2 incubator for different times (Fig. S1). The relative gp120 shedding was assessed by measuring the infectivity of pseudotyped virus-b12 complex at indicated times in TZM-bl cells. We found that the reduction in infectivity was comparable between the b12-sensitive and -resistant variants. Our data indicated that enhanced sensitivity of 2-5.J3 Env was not due to b12-induced gp120 shedding. Substitution of 2-5.J3 V2C2 sequence in unrelated b12-resistant primary clade C Envs displayed varied b12 and VRC01 susceptibilities We next examined the ability of the 2-5.J3 V2C2 sequence to alter b12 susceptibility of few unrelated primary clade C Envs. For this, we selected four b12-resistant clade C Envs: 3-5.J25, 5.J41, 7. J20 and 16055-2.3, and substituted their V2C2 with that of 2-5.J3 Env. As shown in Fig. 2 (bottom figure), all the four Envs contain- ing 2–5.J3 V2C2 sequence differed in their degree of susceptibility to b12 MAb. While 3-5.J25 and 16055-2.3 Envs containing 2-5.J3 V2C2 sequence displayed substantial and modest increase in b12 susceptibilities respectively compared to their corresponding wild types, 7.J20 (2-5.J3 V2C2) showed evidence of marginal increase in b12 susceptibility. Conversely, 2-5.J3 V2C2 sequence was not found to influence 5.J41 Env in modulating its susceptibility to b12 MAb. Interestingly, presence of aspartic acid at the position of 185 (D185) (Fig. 1) was not found to affect the neutralization of these Fig. 2. Neutralization susceptibilities of wild type, chimeric and mutant pseudotyped viruses expressing 2-5.J3 and 4.J22 Envs (top and middle panel) and other unrelated primary Envs expressing 2-5.J3 V2C2 sequence (bottom panel) to b12 MAb. The neutralization assays were carried out in duplicates in at least three independent time points. S. Patil et al. / Virology 462-463 (2014) 266–272268
  • 4. four Envs by b12 MAb, which is in contrast to the observation reported very recently by Utachee et al. (2014). Since b12 MAb recognizes residues in gp120 that overlaps CD4bs, we next examined whether 2-5.J3 V2C2 sequence altered Env sensitivities to VRC01 (which is another CD4bs recognizing MAb), QS1420 (anti-CD4 MAb) and sCD4. As shown in Fig. 3, while all the wild-type Envs and their chimeras (containing 2-5.J3 V2C2 sequence) showed comparable sensitivities to VRC01 MAb, sCD4 and QS1420 MAb, only 4.J22 and 16055-2.3 Envs containing 2-5.J3 V2C2 displayed modest reduction in their sensitivities to VRC01 MAb as compared to their respective wild types. Overall, our data indicate that the effect of presence of 2-5.J3 V2C2 sequence on susceptibility of Envs to b12 and VRC01 is context dependent and likely would vary between Envs. Determinants in V2C2 region modulated neutralization of env- pseudotyped viruses by PG9/PG16 MAbs Since the V2C2 sequences of 2-5.J3 and 4.J22 differ by six residues (although 4.J22 has shorter V2 loop length compared to 2-5.J3 Env) as described above, we next examined whether these residues play any role in modulating the sensitivities of these two Envs to PG and PG16 MAbs as a small change in V2 loop was previously reported to significantly modulate Env susceptibility to Envs to PG9/16 MAbs which targets QNE in V2 loop (Doores and Burton, 2010; Moore et al., 2011; Ringe et al., 2012). As shown in Fig. 4, we found that introduction of S183/E185/K186/K192/I207/ T215 in 2-5.J3 Env conferred 2-5.J3 (4.J22-V2C2/SEKKIT) pseudo- virus with a significant increase in susceptibility to PG9 and PG16 MAbs. Conversely, a significant reduction in the sensitivity of 4.J22 Env was observed when specific substitutions of P183/D185/N186/ N192/V207/N215 were made in 4.J22 Env. This effect was found to be comparable when substitution of only P183/D185/N186/N192 was made in the V2 loop of the 4.J22 Env, suggesting these minimal changes were sufficient in modulating the exposure of PG9/16 epitopes that resulted in the reduction in sensitivity of 4. J22 Env to PG9 and PG16 MAbs and further indicated that N186 and N192 abrogate access of QNE by PG9/16 MAbs. It is to be noted in this context that substitution of 4.J22 V2C2 with that of 2-5.J3 Env displayed significant reduction in the susceptibility of PG9/PG16 MAbs, which is likely due to the increase in V2 loop length as described earlier. Since an extra glycan residue at the position 192 is present in the V2 loop of 2-5.J3 Env compared to the 4.J22 Env, we sought to examine whether this glycan (N192) has any role in modulating PG9/16 sensitivity. Hence, we substituted N192A in 2-5.J3 Env and introduced a glycan (K192NSS) in 4.J22 Env and examined its susceptibility to PG9 and PG16 MAbs. As shown in Fig. 4 (bottom rows), introduction of glycan in 4.J22 Env (K192NSS) conferred 4. J22 (K192NSS) Env-pseudotyped virus with significant reduction in its susceptibility to PG9 and PG16 MAbs; on the contrary, N192A substitution in 2-5.J3 Env showed marginal increment (though not significant) in PG9 susceptibility but not to PG16 MAb. These observations suggest that the presence of glycan at position 192 in the V2 loop interferes with efficient recognition by PG9/16 MAbs and likely is dependent on loop length. In summary, in the present study, using chimeric Envs and through site-directed mutagenesis, we demonstrated that V2C2 altered b12 susceptibility of HIV-1 clade C Envs in a context- dependent manner. In addition, we identified minimal residues in V2 loop that modulated Env susceptibility to PG9/PG16 MAbs. The neutralization susceptibility of Envs to b12 and PG9/16 MAbs was found to be associated with exposure of their corresponding epitopes as determined as wash-out experiments (Fig. 5). We ruled out that the presence of aspartic acid at the position 185 (D185) in V2 loop as recently demonstrated (Utachee et al., 2014) is not absolutely required for enhanced susceptibility of Envs to b12 MAb as 7.J20 wild-type Env although naturally containing D185, it is resistant to b12 MAb. In addition, 5.J41 (2-5.J3-V2C2) and 7.J20 (2-5.J3-V2C2) chimeric Envs expressing D185 did not show any reasonable susceptibility to b12 MAbs. In this paper, we also demonstrated that the presence of glycan residues at the positions 186 (N186) and 192 (N192) significantly abrogated access of PG9/16 MAbs to QNE in the V2 loop of the b12-sensitive 2-5.J3 Env. Our data was substantiated by the fact that N192A alone did not show any change in improvement of access of PG9/16 MAbs in 2-5.J3 Env. However, in 4.J22 Env, addition of a glycan at the position 192 (K192NSS) conferred significant reduction in access of QNE by PG9/16 MAbs, indicating this effect was likely to be associated with shorter V2 loop length.V1V2 domain in Env plays a pivotal role in either recognizing the neutralizing antibodies or Fig. 3. Neutralization susceptibilities of wild type and chimeric Env-pseudotyped viruses to VRC01 MAb (top panel) and to sCD4 and anti-CD4 MAb (QS4120) (bottom panel). The neutralization assays were carried out in duplicates in at least three independent time points. S. Patil et al. / Virology 462-463 (2014) 266–272 269
  • 5. facilitating the escape of HIV-1 variants to the neutralizing anti- bodies including autologous antibodies, possibly due to its con- formation as well as glycosylation pattern (Overbaugh and Morris, 2012; Pinter et al., 2004; Rong et al., 2007). Historically, the V1V2 sequences have been shown to undergo changes earlier than that of the other regions of the Env (Burns and Desrosiers, 1991; Johnson et al., 1991; Laird et al., 2008; Moore et al., 2011; Moore et al., 2009; Overbaugh and Rudensey, 1992; Overbaugh et al., 1991). Our data suggest that the V2C2 region of the 2-5.J3 Env possibly affected the spike apex region, which in turn influenced Env to undergo conformational changes resulting in differential modulation of exposure of epitopes targeted by b12 MAb. Because conformational change is a complex phenomenon and is often context dependent, this may possibly explain why despite contain- ing the same 2-5.J3 V2C2 sequence, other Envs displayed distinct susceptibilities to b12 MAb. Materials and methods HIV-1 Envs and antibodies The HIV-1 clade C envelopes 2-5.J3, 3-5.J25 and 4.J22 obtained from Indian patients with recent infection were described pre- viously (Ringe et al., 2010). 7.J20 is also an HIV-1 clade C Env cloned in pcDNA3.1D/V5-His-TOPO (Invitrogen Inc.) obtained from an Indian patient with recent infection, while 16055-2.3 was kindly provided by Dr. David Montefiori. b12 monoclonal antibody was kindly provided by Dr. Dennis Burton, and was also obtained from the NIH AIDS Research Reagent and Reference Program. VRC01 and sCD4 were obtained from NIH AIDS Research Reagent and Reference Program. PG9 and PG16 MAbs were kindly provided by Dr. Wayne Koff, International AIDS Vaccine Initiative (IAVI), New York. QS4120 MAb was purchased from Calbiochem Inc. Construction of Env chimera and point mutations Chimeric Env constructs were made (Fig. 1) by swapping different variable and constant domains of b12-sensitive Env (2-5.J3) into resistant Env (4.J22) by blunt ligation of PCR products using either In-Fusion Dry Down PCR kit (Clontech Inc.) as described before (Ringe and Bhattacharya, 2012) following the manufacturer's protocol or by two rounds of overlapping PCR in which first-round amplicons were used as primers in the second round. Site-directed mutagenesis to substitute specific residues in V2 and C2 regions were carried out using tailor-made primers by PCR using high fidelity polymerase, Platinum Taq (Invitrogen Inc.) or Phusion high fidelity polymerase (New England Biolabs Inc.). The purified PCR products were then digested with DpnI joined with backbone plasmid by annealing in the presence of pox DNA polymerase as described before (Ringe et al., 2011). Preparation of Env-pseudotyped viruses and infectivity assay Env-pseudotyped viruses were prepared by co-transfection of pSVIIIenv containing patient Envs and pSG3Δenv in 293T cells using calcium phosphate (Promega, Inc.) as described before (Ringe et al., 2010). The infectivity assay was carried out in TZM- bl cells expressing CD4, CCR5 and CXCR4. Neutralization assay Neutralization assays were carried out using TZM-bl cells as described before (Ringe et al., 2010). Env-pseudotyped viruses were incubated with varying dilutions of plasma (heat-inactivated) and varying concentrations of b12 and PG9/16 MAbs for 1 h at 37 1C, and subsequently 1 Â 104 TZM-bl cells were added into the mixture in the presence of 25 μg/ml DEAE dextran (Sigma Inc.). The plates were further incubated in a CO2 incubator at 37 1C for two days and the degree of virus neutralization was assessed by measuring relative luminescence units (RLU) in a Luminometer (Victor 4, PerkinElmer Inc.). Virus-antibody washout assay In order to assess the degree of exposure of neutralizing epitopes recognized by b12 and PG9 MAbs, virus antibody wash- out assay was carried out as essentially described by Chakrabarti et al. (2011). Briefly, MAbs (b12 and PG9) were serially 5-fold diluted in 250 ml of 10% DMEM media to which 250 ml of Env- pseudoviruses were added such that the final concentrations of Fig. 4. Effect of glycan modification and specific amino acid substitutions in 2–5.J3 and 4.J22 Envs towards modulation of their susceptibilities to PG9 and PG16 MAbs. The point substitutions were made by site-directed mutagenesis. The data are representation of three independent experiments done in duplicates. S. Patil et al. / Virology 462-463 (2014) 266–272270
  • 6. antibodies became 50À0.08 mg/ml in a total volume of 500 ml, while in the “no inhibitor” control, the same volume of media was added instead of antibody. The reaction mixture was incubated for 30 min at 37 1C following which 250 ml of reaction mixture was added in ultra-clear centrifuge tubes (Beckman Coulter, Inc.), which was diluted to 4 ml by complete DMEM and centrifuged at 35,000 rpm in an SW60 Ti swinging rotor (Beckman Optima-XE- 100, Inc.) for 2 h at 4 1C. The virus pellet was washed twice with 4 ml of media by centrifuging at 40,000 rpm for 30 min at 4 1C. After the final wash, 250 ml of DMEM was added to the virus pellet and suspended by gentle shaking at 4 1C for 30 min. 100 ml of the suspended virus was used to infect 100 ml of TZM-b cells (0.2 Â 106 /ml) in duplicate. From the remaining 250 ml of reaction mixture, an equal volume of the antibody virus mixture was used as a “no washout” control. Plates were incubated at 37 1C in a CO2 incubator for 2 days, following which the degree of virus neu- tralization was assessed by measuring the reduction of infection in TZM-bl cells. GenBank accession number The GenBank accession number of HIV-1 clade C Env, 7.J20 is EU908223.1. Acknowledgments This work was supported in part by a grant from the Depart- ment of Biotechnology, Government of India, to JB (BT/PR12853/ MED/29/141/2009) and by the intramural support from the IAVI- THSTI HIV Vaccine Design Program. IAVI's work was made possible by generous support from many donors including: the Bill & Melinda Gates Foundation; the Ministry of Foreign Affairs of Denmark; Irish Aid; the Ministry of Finance of Japan; the Ministry of Foreign Affairs of the Netherlands; the Norwegian Agency for Development Cooperation (NORAD); the United Kingdom Department for International Development (DFID); and the United States Agency for International Development (USAID). The full list of IAVI donors is available at www.iavi.org. This study was made possible by the generous support of the American people through USAID. The contents are the responsibility of the authors and do not necessarily reflect the views of USAID or the United States Government. We thank Dr. Dennis Burton, Scripps Research Institute, La Jolla, California, and Dr. Wayne Koff, International AIDS Vaccine Initiative, New York, for help with obtaining b12, PG9 and PG16 monoclonal antibodies, respectively. We sincerely acknowledge the support of the members of the HVTR laboratory, Prof. G Balakrish Nair, Prof. Sudhanshu Vrati, Dr. Rick King and Dr. Rajat Goyal. We thank the Director, National AIDS Research Institute, Pune, for making available the HIV-1 clade C envelope clones. Appendix A. Supporting information Supplementary data associated with this article can be found in the online version at http://dx.doi.org/10.1016/j.virol.2014.06.018. References Bunnik, E.M., Pisas, L., van Nuenen, A.C., Schuitemaker, H., 2008. Autologous neutralizing humoral immunity and evolution of the viral envelope in the course of subtype B human immunodeficiency virus type 1 infection. J. Virol. 82, 7932–7941. Burns, D.P., Desrosiers, R.C., 1991. Selection of genetic variants of simian immuno- deficiency virus in persistently infected rhesus monkeys. J. Virol. 65, 1843–1854. Burton, D.R., Pyati, J., Koduri, R., Sharp, S.J., Thornton, G.B., Parren, P.W., Sawyer, L.S., Hendry, R.M., Dunlop, N., Nara., P.L., 1994. Efficient neutralization of primary isolates of HIV- 1 by a recombinant human monoclonal antibody. Science 266, 1024–1027. Chakrabarti, B.K., Walker, L.M., Guenaga, J.F., Ghobbeh, A., Poignard, P., Burton, D.R., Wyatt, R.T., 2011. Direct antibody access to the HIV-1 membrane-proximal external region positively correlates with neutralization sensitivity. J. Virol. 85, 8217–8226. Fig. 5. The effect of presence of the 2-5.J3 V2C2 sequence in 4.J22 Env (top panel) and substitution of P183S/D185E/N186K/N192K/V207I/N215T in 2-5.J3 Env (bottom panel) on exposure of b12 and PG9 epitopes, respectively. Virus-MAb washout assay was carried out using an ultracentrifuge to remove unbound b12 and PG9 MAbs from the Env-pseudotyped viruses. MAb bound Env-pseudotyped viruses were mixed with TZM-bl cells and reduction in virus infection was assessed in a dose-dependent manner. S. Patil et al. / Virology 462-463 (2014) 266–272 271
  • 7. Chen, L., Kwon, Y.D., Zhou, T., Wu, X., O'Dell, S., Cavacini, L., Hessell, A.J., Pancera, M., Tang, M., Xu, L., Yang, Z.Y., Zhang, M.Y., Arthos, J., Burton, D.R., Dimitrov, D.S., Nabel, G.J., Posner, M.R., Sodroski, J., Wyatt, R., Mascola, J.R., Kwong, P.D., 2009. Structural basis of immune evasion at the site of CD4 attachment on HIV-1 gp120. Science 326, 1123–1127. Doores, K.J., Burton, D.R., 2010. Variable loop glycan dependency of the broad and potent HIV-1-neutralizing antibodies PG9 and PG16. J. Virol. 84, 10510–10521. Duenas-Decamp, M.J., Peters, P., Burton, D., Clapham, P.R., 2008. Natural resistance of human immunodeficiency virus type 1 to the CD4bs antibody b12 conferred by a glycan and an arginine residue close to the CD4 binding loop. J. Virol. 82, 5807–5814. Etemad-Moghadam, B., Sun, Y., Nicholson, E.K., Karlsson, G.B., Schenten, D., Sodroski, J., 1999. Determinants of neutralization resistance in the envelope glycoproteins of a simian-human immunodeficiency virus passaged in vivo. J. Virol. 73, 8873–8879. Frost, S.D., Wrin, T., Smith, D.M., Kosakovsky Pond, S.L., Liu, Y., Paxinos, E., Chappey, C., Galovich, J., Beauchaine, J., Petropoulos, C.J., Little, S.J., Richman, D.D., 2005. Neutralizing antibody responses drive the evolution of human immunodefi- ciency virus type 1 envelope during recent HIV infection. Proc. Natl. Acad. Sci. U. S. A. 102, 18514–18519. Gray, E.S., Meyers, T., Gray, G., Montefiori, D.C., Morris, L., 2006. Insensitivity of paediatric HIV-1 subtype C viruses to broadly neutralising monoclonal anti- bodies raised against subtype B. PLoS Med. 3, e255. Gray, E.S., Moore, P.L., Choge, I.A., Decker, J.M., Bibollet-Ruche, F., Li, H., Leseka, N., Treurnicht, F., Mlisana, K., Shaw, G.M., Karim, S.S., Williamson, C., Morris, L., 2007. Neutralizing antibody responses in acute human immunodeficiency virus type 1 subtype C infection. J. Virol. 81, 6187–6196. Huang, J., Ofek, G., Laub, L., Louder, M.K., Doria-Rose, N.A., Longo, N.S., Imamichi, H., Bailer, R.T., Chakrabarti, B., Sharma, S.K., Alam, S.M., Wang, T., Yang, Y., Zhang, B., Migueles, S.A., Wyatt, R., Haynes, B.F., Kwong, P.D., Mascola, J.R., Connors, M., 2012. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491, 406–412. Johnson, P.R., Hamm, T.E., Goldstein, S., Kitov, S., Hirsch, V.M., 1991. The genetic fate of molecularly cloned simian immunodeficiency virus in experimentally infected macaques. Virology 185, 217–228. Kulkarni, S.S., Lapedes, A., Tang, H., Gnanakaran, S., Daniels, M.G., Zhang, M., Bhattacharya, T., Li, M., Polonis, V.R., McCutchan, F.E., Morris, L., Ellenberger, D., Butera, S.T., Bollinger, R.C., Korber, B.T., Paranjape, R.S., Montefiori, D.C., 2009. Highly complex neutralization determinants on a monophyletic lineage of newly transmitted subtype C HIV-1 Env clones from India. Virology 385, 505–520. Laird, M.E., Igarashi, T., Martin, M.A., Desrosiers, R.C., 2008. Importance of the V1/V2 loop region of simian-human immunodeficiency virus envelope glycoprotein gp120 in determining the strain specificity of the neutralizing antibody response. J. Virol. 82, 11054–11065. Li, B., Decker, J.M., Johnson, R.W., Bibollet-Ruche, F., Wei, X., Mulenga, J., Allen, S., Hunter, E., Hahn, B.H., Shaw, G.M., Blackwell, J.L., Derdeyn, C.A., 2006. Evidence for potent autologous neutralizing antibody titers and compact envelopes in early infection with subtype C human immunodeficiency virus type 1. J. Virol. 80, 5211–5218. Liu, J., Bartesaghi, A., Borgnia, M.J., Sapiro, G., Subramaniam, S., 2008. Molecular architecture of native HIV-1 gp120 trimers. Nature 455, 109–113. Ly, A., Stamatatos, L., 2000. V2 loop glycosylation of the human immunodeficiency virus type 1 SF162 envelope facilitates interaction of this protein with CD4 and CCR5 receptors and protects the virus from neutralization by anti-V3 loop and anti-CD4 binding site antibodies. J. Virol. 74, 6769–6776. Mascola, J.R., 2009. The cat and mouse of HIV-1 antibody escape. PLoS Pathog. 5, e1000592. Mascola, J.R., Montefiori, D.C., 2010. The role of antibodies in HIV vaccines. Annu. Rev. Immunol. 28, 413–444. Mo, H., Stamatatos, L., Ip, J.E., Barbas, C.F., Parren, P.W., Burton, D.R., Moore, J.P., Ho, D.D., 1997. Human immunodeficiency virus type 1 mutants that escape neutralization by human monoclonal antibody IgG1b12. off. J. Virol. 71, 6869–6874. Montefiori, D.C., Morris, L., Ferrari, G., Mascola, J.R., 2007. Neutralizing and other antiviral antibodies in HIV-1 infection and vaccination. Curr. Opin. HIV AIDS 2, 169–176. Moore, J.P., Cao, Y., Ho, D.D., Koup, R.A., 1994. Development of the anti-gp120 antibody response during seroconversion to human immunodeficiency virus type 1. J. Virol. 68, 5142–5155. Moore, P.L., Gray, E.S., Sheward, D., Madiga, M., Ranchobe, N., Lai, Z., Honnen, W.J., Nonyane, M., Tumba, N., Hermanus, T., Sibeko, S., Mlisana, K., Abdool Karim, S. S., Williamson, C., Pinter, A., Morris, L., 2011. Potent and broad neutralization of HIV-1 subtype C by plasma antibodies targeting a quaternary epitope including residues in the V2 loop. J. Virol. 85, 3128–3141. Moore, P.L., Ranchobe, N., Lambson, B.E., Gray, E.S., Cave, E., Abrahams, M.R., Bandawe, G., Mlisana, K., Abdool Karim, S.S., Williamson, C., Morris, L., 2009. Limited neutralizing antibody specificities drive neutralization escape in early HIV-1 subtype C infection. PLoS Pathog. 5, e1000598. Overbaugh, J., Morris, L., 2012. The Antibody Response against HIV-1. Cold Spring Harb. Perspect. Med. 2, a007039. Overbaugh, J., Rudensey, L.M., 1992. Alterations in potential sites for glycosylation predominate during evolution of the simian immunodeficiency virus envelope gene in macaques. J. Virol. 66, 5937–5948. Overbaugh, J., Rudensey, L.M., Papenhausen, M.D., Benveniste, R.E., Morton, W.R., 1991. Variation in simian immunodeficiency virus env is confined to V1 and V4 during progression to simian AIDS. J. Virol. 65, 7025–7031. Pantophlet, R., Ollmann Saphire, E., Poignard, P., Parren, P.W., Wilson, I.A., Burton, D. R., 2003. Fine mapping of the interaction of neutralizing and nonneutralizing monoclonal antibodies with the CD4 binding site of human immunodeficiency virus type 1 gp120. J. Virol. 77, 642–658. Pinter, A., Honnen, W.J., He, Y., Gorny, M.K., Zolla-Pazner, S., Kayman, S.C., 2004. The V1/V2 domain of gp120 is a global regulator of the sensitivity of primary human immunodeficiency virus type 1 isolates to neutralization by antibodies commonly induced upon infection. J. Virol. 78, 5205–5215. Richman, D.D., Wrin, T., Little, S.J., Petropoulos, C.J., 2003. Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proc. Natl. Acad. Sci. U. S. A. 100, 4144–4149. Ringe, R., Bhattacharya, J., 2012. Association of enhanced HIV-1 neutralization by a single Y681H substitution in gp41 with increased gp120-CD4 interaction and macrophage Infectivity. PLoS One 7, e37157. Ringe, R., Phogat, S., Bhattacharya, J., 2012. Subtle alteration of residues including N-linked glycans in V2 loop modulates HIV-1 neutralization by PG9 and PG16 monoclonal antibodies. Virology 426, 34–41. Ringe, R., Sharma, D., Zolla-Pazner, S., Phogat, S., Risbud, A., Thakar, M., Paranjape, R., Bhattacharya, J., 2011. A single amino acid substitution in the C4 region in gp120 confers enhanced neutralization of HIV-1 by modulating CD4 binding sites and V3 loop. Virology 418, 123–132. Ringe, R., Thakar, M., Bhattacharya, J., 2010. Variations in autologous neutralization and CD4 dependence of b12 resistant HIV-1 clade C env clones obtained at different time points from antiretroviral naive Indian patients with recent infection. Retrovirology 7, 76. Rong, R., Bibollet-Ruche, F., Mulenga, J., Allen, S., Blackwell, J.L., Derdeyn, C.A., 2007. Role of V1V2 and other human immunodeficiency virus type 1 envelope domains in resistance to autologous neutralization during clade C infection. J. Virol. 81, 1350–1359. Si, Z., Cayabyab, M., Sodroski, J., 2001. Envelope glycoprotein determinants of neutralization resistance in a simian-human immunodeficiency virus (SHIV- HXBc2P 3.2) derived by passage in monkeys. J. Virol. 75, 4208–4218. Thenin, S., Roch, E., Samleerat, T., Moreau, T., Chaillon, A., Moreau, A., Barin, F., Braibant, M., 2012. Naturally occurring substitutions of conserved residues in HIV-1 variants of different clades are involved in PG9 and PG16 resistance to neutralization. J. Gen. Virol. 93, 1495–1505. Tomaras, G.D., Yates, N.L., Liu, P., Qin, L., Fouda, G.G., Chavez, L.L., Decamp, A.C., Parks, R.J., Ashley, V.C., Lucas, J.T., Cohen, M., Eron, J., Hicks, C.B., Liao, H.X., Self, S.G., Landucci, G., Forthal, D.N., Weinhold, K.J., Keele, B.F., Hahn, B.H., Greenberg, M.L., Morris, L., Karim, S.S., Blattner, W.A., Montefiori, D.C., Shaw, G.M., Perelson, A.S., Haynes, B.F., 2008. Initial B-cell responses to transmitted human immunodeficiency virus type 1: virion-binding immunoglobulin M (IgM) and IgG antibodies followed by plasma anti-gp41 antibodies with ineffective control of initial viremia. J. Virol. 82, 12449–12463. Utachee, P., Isarangkura-na-ayuthaya, P., Tokunaga, K., Ikuta, K., Takeda, N., Kameoka, M., 2014. Impact of amino acid substitutions in the V2 and C2 regions of human immunodeficiency virus type 1 CRF01_AE envelope glyco- protein gp120 on viral neutralization susceptibility to broadly neutralizing antibodies specific for the CD4 binding site. Retrovirology 11, 32. Utachee, P., Nakamura, S., Isarangkura-Na-Ayuthaya, P., Tokunaga, K., Sawanpanya- lert, P., Ikuta, K., Auwanit, W., Kameoka, M., 2010. Two N-linked glycosylation sites in the V2 and C2 regions of human immunodeficiency virus type 1 CRF01_AE envelope glycoprotein gp120 regulate viral neutralization suscept- ibility to the human monoclonal antibody specific for the CD4 binding domain. J. Virol. 84, 4311–4320. Walker, L.M., Huber, M., Doores, K.J., Falkowska, E., Pejchal, R., Julien, J.P., Wang, S.K., Ramos, A., Chan-Hui, P.Y., Moyle, M., Mitcham, J.L., Hammond, P.W., Olsen, O.A., Phung, P., Fling, S., Wong, C.H., Phogat, S., Wrin, T., Simek, M.D., 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies (Principal Investigators, P.G., Koff, W.C., Wilson, I.A., Burton, D.R., Poignard, P.). Nature 477, 466–470. Walker, L.M., Phogat, S.K., Chan-Hui, P.Y., Wagner, D., Phung, P., Goss, J.L., Wrin, T., Simek, M.D., Fling, S., Mitcham, J.L., Lehrman, J.K., Priddy, F.H., Olsen, O.A., Frey, S.M., Hammond, P.W., Kaminsky, S., Zamb, T., Moyle, M., Koff, W.C., Poignard, P., Burton, D.R., 2009. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326, 285–289. Wei, X., Decker, J.M., Wang, S., Hui, H., Kappes, J.C., Wu, X., Salazar-Gonzalez, J.F., Salazar, M.G., Kilby, J.M., Saag, M.S., Komarova, N.L., Nowak, M.A., Hahn, B.H., Kwong, P.D., Shaw, G.M., 2003. Antibody neutralization and escape by HIV-1. Nature 422, 307–312. Wu, X., Yang, Z.Y., Li, Y., Hogerkorp, C.M., Schief, W.R., Seaman, M.S., Zhou, T., Schmidt, S.D., Wu, L., Xu, L., Longo, N.S., McKee, K., O'Dell, S., Louder, M.K., Wycuff, D.L., Feng, Y., Nason, M., Doria-Rose, N., Connors, M., Kwong, P.D., Roederer, M., Wyatt, R.T., Nabel, G.J., Mascola, J.R., 2010. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329, 856–861. Wu, X., Zhou, T., O'Dell, S., Wyatt, R.T., Kwong, P.D., Mascola, J.R., 2009. Mechanism of human immunodeficiency virus type 1 resistance to monoclonal antibody B12 that effectively targets the site of CD4 attachment. J. Virol. 83, 10892–10907. Zhang, H., Rola, M., West, J.T., Tully, D.C., Kubis, P., He, J., Kankasa, C., Wood, C., 2010. Functional properties of the HIV-1 subtype C envelope glycoprotein associated with mother-to-child transmission. Virology 400, 164–174. Zhou, T., Xu, L., Dey, B., Hessell, A.J., Van Ryk, D., Xiang, S.H., Yang, X., Zhang, M.Y., Zwick, M.B., Arthos, J., Burton, D.R., Dimitrov, D.S., Sodroski, J., Wyatt, R., Nabel, G.J., Kwong, P.D., 2007. Structural definition of a conserved neutralization epitope on HIV-1 gp120. Nature 445, 732–737. S. Patil et al. / Virology 462-463 (2014) 266–272272