12
Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of leucine residues on Vpr-mediated transactivation and virus replication Dineshkumar Thotala a , Elizabeth A. Schafer a , Biswanath Majumder a , Michelle L. Janket a , Marc Wagner a , Alagarsamy Srinivasan b , Simon Watkins c , Velpandi Ayyavoo a, * a Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, United States b Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, United States c Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15261, United States Received 13 July 2004; accepted 13 July 2004 Abstract HIV-1 Vpr has been shown to transactivate LTR-directed expression through its interaction with several proteins of cellular origin including the glucocorticoid receptor (GR). Upon activation, steroid receptors bind to proteins containing the signature motif LxxLL, translocate into the nucleus, bind to their response element, and activate transcription. The presence of such motifs in HIV-1 Vpr has prompted us to undertake the analysis of the role of specific leucine residue(s) involved in Vpr–GR interaction, subcellular localization and its effect on Vpr–GR-mediated transactivation. The individual leucine residues present in H I, II, and III were mutated in the Vpr molecule and evaluated for their ability to interact with GR, transactivate GRE and HIV-1 LTR promoters, and their colocalization with GR. While Vpr mutants L42 and L67 showed reduced activation, substitutions at L20, L23, L26, L39, L64, and L68 exhibited a similar and slightly higher level of activation compared to Vpr wt . Interestingly, a substitution at residue L22 resulted in a significantly higher GRE and HIV-1 LTR transactivation (8- to 11-fold higher) in comparison to wild type. Confocal microscopy indicated that Vpr L22A exhibited a distinct condensed nuclear localization pattern different from the nuclear/perinuclear pattern noted with Vpr wt . Further, electrophoretic mobility shift assay (EMSA) revealed that the VprL22A–GR complex had higher DNA-binding activity when compared to the wild type Vpr–GR complex. These results suggest a contrasting role for the leucine residues on HIV-1 LTR-directed transactivation dependent upon their location in Vpr. D 2004 Elsevier Inc. All rights reserved. Keywords: Glucocorticoid receptor; Transactivation; Replication Introduction Human immunodeficiency virus type-1 (HIV-1) Vpr is a 14-kDa virion-associated non-structural protein. While Vpr has been shown to be important for viral replication in non- dividing cells (Balliet et al., 1994; Connor et al., 1995; Heinzinger et al., 1994; Subbramanian et al., 1998), increased viral replication in T-cell lines as well as activation in latently infected cells have also been reported (Levy et al., 1995; Nakamura et al., 2002). Vpr transactivates HIV-1 LTR and increases virus replication in target cells (Agostini et al., 1996; Cohen et al., 1990; Wang et al., 1995). Recently, we and others have shown that Vpr, either in the context of virus infection or as exogenous protein, transactivates HIV-1 LTR and upregulates viral replication before the synthesis of Tat in infected cells. Also, Vpr and Tat together transactivate HIV-1 LTR in an additive manner (Hrimech et al., 1999; Sawaya et al., 2000; Vanitharani et al., 2001). Specifically, HIV-1 Vpr-mediated transactivation is shown to occur through steroid receptors and other coactivators (Kino et al., 1999; Sherman et al., 2000; Vanitharani et al., 2001). 0042-6822/$ - see front matter D 2004 Elsevier Inc. All rights reserved. doi:10.1016/j.virol.2004.07.013 * Corresponding author. Department of Infectious Diseases & Micro- biology, University of Pittsburgh, 130 DeSoto Street, Pittsburgh, PA 15261. Fax: +1 412 383 8926. E-mail address: [email protected] (V. Ayyavoo). Virology 328 (2004) 89– 100 www.elsevier.com/locate/yviro

Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

www.elsevier.com/locate/yviro

Virology 328 (2

Structure–functional analysis of human immunodeficiency

virus type 1 (HIV-1) Vpr: role of leucine residues on

Vpr-mediated transactivation and virus replication

Dineshkumar Thotalaa, Elizabeth A. Schafera, Biswanath Majumdera, Michelle L. Janketa,

Marc Wagnera, Alagarsamy Srinivasanb, Simon Watkinsc, Velpandi Ayyavooa,*

aDepartment of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, United StatesbDepartment of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, United States

cCenter for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15261, United States

Received 13 July 2004; accepted 13 July 2004

Abstract

HIV-1 Vpr has been shown to transactivate LTR-directed expression through its interaction with several proteins of cellular origin

including the glucocorticoid receptor (GR). Upon activation, steroid receptors bind to proteins containing the signature motif LxxLL,

translocate into the nucleus, bind to their response element, and activate transcription. The presence of such motifs in HIV-1 Vpr has

prompted us to undertake the analysis of the role of specific leucine residue(s) involved in Vpr–GR interaction, subcellular localization and

its effect on Vpr–GR-mediated transactivation. The individual leucine residues present in H I, II, and III were mutated in the Vpr molecule

and evaluated for their ability to interact with GR, transactivate GRE and HIV-1 LTR promoters, and their colocalization with GR. While Vpr

mutants L42 and L67 showed reduced activation, substitutions at L20, L23, L26, L39, L64, and L68 exhibited a similar and slightly higher

level of activation compared to Vprwt. Interestingly, a substitution at residue L22 resulted in a significantly higher GRE and HIV-1 LTR

transactivation (8- to 11-fold higher) in comparison to wild type. Confocal microscopy indicated that Vpr L22A exhibited a distinct

condensed nuclear localization pattern different from the nuclear/perinuclear pattern noted with Vprwt. Further, electrophoretic mobility shift

assay (EMSA) revealed that the VprL22A–GR complex had higher DNA-binding activity when compared to the wild type Vpr–GR complex.

These results suggest a contrasting role for the leucine residues on HIV-1 LTR-directed transactivation dependent upon their location in Vpr.

D 2004 Elsevier Inc. All rights reserved.

Keywords: Glucocorticoid receptor; Transactivation; Replication

Introduction

Human immunodeficiency virus type-1 (HIV-1) Vpr is a

14-kDa virion-associated non-structural protein. While Vpr

has been shown to be important for viral replication in non-

dividing cells (Balliet et al., 1994; Connor et al., 1995;

Heinzinger et al., 1994; Subbramanian et al., 1998),

increased viral replication in T-cell lines as well as activation

0042-6822/$ - see front matter D 2004 Elsevier Inc. All rights reserved.

doi:10.1016/j.virol.2004.07.013

* Corresponding author. Department of Infectious Diseases & Micro-

biology, University of Pittsburgh, 130 DeSoto Street, Pittsburgh, PA 15261.

Fax: +1 412 383 8926.

E-mail address: [email protected] (V. Ayyavoo).

in latently infected cells have also been reported (Levy et al.,

1995; Nakamura et al., 2002). Vpr transactivates HIV-1 LTR

and increases virus replication in target cells (Agostini et al.,

1996; Cohen et al., 1990; Wang et al., 1995). Recently, we

and others have shown that Vpr, either in the context of virus

infection or as exogenous protein, transactivates HIV-1 LTR

and upregulates viral replication before the synthesis of Tat

in infected cells. Also, Vpr and Tat together transactivate

HIV-1 LTR in an additive manner (Hrimech et al., 1999;

Sawaya et al., 2000; Vanitharani et al., 2001). Specifically,

HIV-1 Vpr-mediated transactivation is shown to occur

through steroid receptors and other coactivators (Kino et

al., 1999; Sherman et al., 2000; Vanitharani et al., 2001).

004) 89–100

Page 2: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

D. Thotala et al. / Virology 328 (2004) 89–10090

Steroid receptors belong to a super family of ligand-

dependent transcription factors that are known to interact

with other proteins containing the signature motif, LxxLL

(Feng et al., 1998; Heery et al., 1997). Upon binding to their

ligands, these receptors translocate into the nucleus and

activate/repress transcription depending upon the signal

(Northrop et al., 1992; Paliogianni and Boumpas, 1995;

Schmidt et al., 1994). HIV-1 Vpr has been shown to interact

with glucocorticoid receptor (GR) and p300/CBP and

activate transcription through the glucocorticoid response

element (GRE) (Felzien et al., 1998; Kino et al., 2002a).

HIV-1 Vpr is a pleiotropic protein, with diverse functions

that include cell cycle arrest, apoptosis, nuclear import of

the preintegration complex, transcriptional activation/repres-

sion, and interaction with viral and several cellular proteins

(Levy et al., 1993; Subbramanian et al., 1998; Zhao et al.,

1994). Structure–function studies have indicated that

nuclear localization, virion incorporation, cell cycle arrest,

and transcriptional regulation are mediated by different

functional domains of Vpr (Di Marzio et al., 1995;

Mahalingam et al., 1997; Sawaya et al., 2000; Singh et

al., 2001). Vpr-mediated cell cycle arrest is linked to an

increase in virus production (Goh et al., 1998; Levy et al.,

1995; Yao et al., 1998); however, Vpr–GR transcriptional

coactivation function is reported to be independent of cell

cycle arrest (Kino et al., 2002b; Sherman et al., 2000).

Previous studies have shown that Vpr mimics glucocorti-

coids and induces similar functions through its interaction

with GR (Ayyavoo et al., 1997). GR is a member of the

steroid hormone receptor family of transcription factors that

regulates a large number of genes in a hormone or ligand-

dependent manner. Under normal conditions, GR is present

in the cytosol as an inactive complex with two molecules of

heat shock protein 90 (HSP 90). Upon binding of the

ligands, GR dissociates from the HSP molecules and

translocates into the nucleus, binds to the consensus

palindromic sequences known as GRE and activates/

represses transcription based on the stimuli (Cadepond et

al., 1992; Li et al., 2002).

Recent structural studies utilizing NMR and CD spectro-

scopy of the synthetic full-length Vpr showed three helical

domains and three turn regions with slightly different

boundaries in comparison to the structures noted with the

N- and C-terminal fragments of Vpr (Morellet et al., 2003;

Wecker et al., 2002). The structural features of the protein

include a well-designed g-turn (14–16)-a helix (17–33)—

turn (34–36) followed by an a helix (40–48)—loop (49–

54)-a helix (55–83) domain and a flexible C-terminal

sequence. Vpr contains multiple leucine-rich domains

(LxxL) in addition to the LxxLL motif in the C- and N-

terminal region of the protein. The predicted amino acid

sequence analysis of Vpr from viruses of different clades

has shown conserved leucine motifs in helices I and III,

suggesting an important role for these domains in patho-

genesis. Vpr, being a virion-associated molecule, supports

the notion that Vpr might be available as a viral protein

during early infection (before the appearance of Tat and

Rev) to enhance gene expression by directly binding to the

viral and cellular promoters (Sawaya et al., 2000; Vanithar-

ani et al., 2001; Zhao et al., 1994). Though previous studies

have addressed the Vpr-GR coactivator functions, these

studies pertain to the mutational analysis of residues 64–68

corresponding to the LxxLL motif. To precisely map and

understand the role of leucine residues present in the a-

helices, we have constructed mutants of Vpr using site-

specific mutagenesis. All the leucine mutants were analyzed

for their ability to transactivate a reporter gene under the

control of GRE and HIV-1 LTR. Next, we evaluated

whether the subcellular localization of Vpr altered GR

translocation as well as its effect on GRE and HIV-1 LTR

transactivation in the absence of hormone stimulation. The

results generated here suggest that the leucine residues can

be functionally categorized into three groups: alterations of

residues can lead to a reduced, minimal or no effect, or an

enhanced level of transactivation in comparison to the wild-

type Vpr. Interestingly, the Vpr mutant L22A translocated

GR with a distinct condensed nuclear localization pattern

resulting in a significantly higher transactivation effect when

compared to Vprwt. Additionally, the increased DNA-

binding activity of VprL22A-GR directly correlates with

Vpr coactivator function.

Results

Generation and characterization of leucine mutants of Vpr

Structure analysis by NMR and CD spectroscopy show

that Vpr contains three helical domains with a basic amino

acid-enriched C-terminus. These helical domains are rich in

leucine residues with classical steroid signature motifs

(LxxLL) in the helical domains. Steroid receptors interact

with proteins through these classical motifs. To identify the

specific leucine domains involved in Vpr-GR interaction,

we have substituted individual leucine (L) residues in helix

I, II, and III to alanine (A) by site-directed mutagenesis (Fig.

1). To enable the detection of Vpr, sequences corresponding

to an eight amino acid Flag epitope (DYKDDDDK) were

fused in frame to the 3V end of the Vpr coding sequence. To

confirm that all of these Vpr mutants expressed a stable

protein in vivo, 293T cells were transfected with pVpr

expression plasmids. Cells were lysed 48 h after transfection

and subjected to immunoblot analysis with the M2 mono-

clonal antibody. Protein expression analysis indicated that

all mutant Vpr molecules showed an appropriate band

corresponding to 14 kDa size protein similar to wild type

Vpr (Fig. 1B). Additionally, the specificity of the antibody

was further confirmed by using the lysate of vector

(pcDNA3.1) transfected 293T cells, which did not recognize

any band. These results indicate that the steady state level of

all the single leucine mutant Vpr molecules is similar to that

of wild-type Vpr.

Page 3: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

Fig. 1. (A) Schematic representation of HIV-1 Vpr leucine mutants: Vpr leucine mutants were generated by site directed mutagenesis. Vpr wild type represents

the vpr gene derived from pNL43 proviral DNA and the change in residues at a particular position is marked for each mutant. LxxL motifs are underlined and

the helical domains are marked as I, II, and III HD above the amino acid sequence. (B) Immunoblot analysis of expression of Vpr mutant molecules: 293T cells

were transfected with 5 Ag of Vpr expression plasmids with a Flag tag using Lipofectamine. Forty-eight hours posttransfection cells were lysed and equal

amounts of protein containing total cell lysates were resolved in a 12% SDS-PAGE and immunoblotted with M2-Flag monoclonal antibody (1:250 dilution).

Vpr mutant constructs representing the residues are marked on top of each lane. The vector represents 293T cells transfected with the pcDNA3.1 vector

backbone.

D. Thotala et al. / Virology 328 (2004) 89–100 91

Transcriptional activation of GRE and HIV-1 LTR-driven

reporter gene by HIV-1 Vpr mutants

To precisely determine the role of specific leucine

residues involved in GRE transactivation, we cotransfected

Vpr mutant plasmids with the pGRE-luciferase reporter

construct in HeLa cells, which express GR endogenously.

Forty-eight-hour posttransfection, cells were lysed and cell

lysates were normalized for transfection efficiency by h-gallevel and assessed for luciferase activity. The fold difference

was calculated considering the activity observed with Vprwt

as 1 (Fig. 2A). Individual substitution mutations at L42 and

L67 significantly (P b 0.01) reduced the GRE-luciferase

activity when compared to Vprwt, whereas mutations at L23,

L39, and L68 exhibited similar activity and L20, L26, and

L64 slightly higher (N5-fold in case of L26 and N3 in case of

L64) levels of transactivation activity. Interestingly, the

mutation at L22 increased the activity by 8- to 11-fold over

wild type. Next, we addressed the question regarding the

requirement of GR for Vpr-mediated transactivation using

CV-1 cells, which are negative for GR expression. CV-1

cells were transfected with pVpr expression plasmid and

pGRE-directed luciferase reporter activity was measured.

CV-1 cells transfected with pVpr and pGRE plasmids alone

did not show any transcriptional activity (data not shown),

whereas cotransfection of the GR expression plasmid with

the reporter and Vpr expression constructs resulted in high

luciferase activity similar to the levels noted in HeLa cells

(Fig. 2B). The mutation at L22 consistently showed the

highest activity both in HeLa and CV-1 cells; however, the

presence of GR was necessary for Vpr-mediated GRE-

activation in CV-1 cells.

In an effort to assess the relevance of the transactivation

function of Vpr for virus replication, HeLa cells were

cotransfected with pVpr mutant molecules and the pHIV-1

LTR-luciferase reporter construct and were monitored for

luciferase activity (Fig. 2C). HIV-1 Vprwt plasmid increased

the transactivation activity by 4- to 6-fold when compared to

the vector backbone-transfected cells. Considering the Vprwt

activity as 1, mutations in L42 and L67 completely

abolished the activity, whereas L22A showed a 17-fold

higher activity than Vprwt. It is interesting to note that L22A

showed high transcriptional activity on both HIV-1 LTR and

GRE promoters in HeLa cells. To confirm Vpr-mediated

HIV-1 LTR transactivation is through the GRE domain in

LTR sequences, we transfected CV-1 cells with HIV-1 LTR

and Vpr mutants with or without the GR expression

plasmid, and assessed the reporter activity (Fig. 2D). Results

indicated that HIV-1 LTR-mediated transactivation by Vpr

plasmids is slightly lower (data not shown) in the absence of

GR, whereas cotransfection of pGRa plasmid increased the

transactivation efficiency similar to that observed in HeLa

cells (RLU). These results differ from the GRE-mediated

transactivation, where presence of GR is absolutely neces-

sary suggesting that HIV-1 Vpr might bind to other

promoter elements present in the HIV-1 LTR. However,

Page 4: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

Fig. 2. Effect of HIV-1 Vpr leucine mutants on pGRE and HIV-1 LTR transactivation: HeLa cells were transfected with HIV-1 LTR luciferase or GRE-

luciferase reporter plasmid in the presence and absence of Vpr mutants using Lipofectamine. Forty-eight hours posttransfection cells were lysed and luciferase

activity was measured. Fold activation was calculated as one, representing Vpr wild type activity. Panel A, HeLa cells transfected with pGRE-luciferase and

pVpr mutant molecules; Panel B, CV-1 cells transfected with pGRE-luciferase, pGR and pVpr mutants; Panel C, HeLa cells transfected with pHIV-1 LTR-

luciferase and pVpr mutant plasmids; Panel D, CV-1 cells transfected with pHIV-1 LTR-luciferase, pGR and pVpr mutant plasmids. The fold activation SD was

derived from three independent experiments. * designates the P value of b0.01.

D. Thotala et al. / Virology 328 (2004) 89–10092

presence of GR enhances Vpr-mediated transactivation of

both autologous and heterologous promoters.

Role of the leucine residues in Vpr–GR interaction

GR has been shown to interact with Vprwt directly to

enhance the coactivator effect. To determine the specific

leucine residue involved in GR interaction, a GST-pull

down assay was performed using 35S-labeled in vitro

translated mutant Vpr proteins and recombinant GR that

was expressed in Escherichia coli BL-21 cells and purified

using GST beads. As a control, GST beads as well as in

vitro translated vif gene product were used (Fig. 3A).

Results indicated that GST-GR pulled all our leucine mutant

Vpr molecules, whereas the GST control did not show any

interaction. Additionally, the use of in vitro translated Vif

product was not pulled by GST-GR suggesting that the pull

down assay shows specificity. Next, to ensure that our in

vitro translated Vpr mutants expressed the appropriate size,

we loaded 1/10th of the in vitro translated product in SDS-

PAGE and it was then autoradiographed (Fig. 3A-input).

Results from these analyses suggest that the individual

leucine substitution of Vpr did not alter the Vpr–GR

interactions and that the observed interaction is very

specific.

Since the pull-down assay utilizes a prokaryotic

expressed protein, we wished to confirm this interaction in

eukaryotic cells. HeLa cells were transfected with phGRa

and Vpr mutant expression plasmids, and immunoprecipi-

tation was performed followed by Western blot analysis

(Fig. 3B). Transfected cell lysates were immunoprecipitated

with anti-GR antibody, resolved on SDS-PAGE, and

immunoblotted for Vpr using M2-flag specific antibody

(Fig. 3B). An interaction between Vpr mutants and GR was

observed as noted in the in vitro GST-GR pull down assay,

whereas vector-transfected cells did not show any specific

bands at 14 kDa size. Similarly, cell lysates were immuno-

precipitated with anti-Vpr antibody (M2-Flag) and immu-

noblotted with anti-GR antibody. Results indicate that cells

transfected with pVpr expression plasmids pulled down GR,

whereas vector-transfected cell lysate did not show any

detectable band (data not shown) suggesting that single

point Vpr leucine mutant did not interfere with GR

interaction and this interaction is specific.

Effect of subcellular distribution of Vpr mutants on GR

translocation

GR-mediated transactivation is generally correlated with

the translocation of GR from the cytoplasm to the nucleus

followed by binding to GRE sequences (DeFranco, 2002;

Htun et al., 1996). Upon expression, HIV-1 Vprwt localizes

at the nuclear membrane/nucleus of the cells (Kamata and

Aida, 2000). To examine whether the subcellular distribu-

tion of Vpr alters GR translocation and its ability to bind to

GRE sequences, we transfected HeLa cells with pVprwt and

assessed the localization pattern of Vpr and GR by indirect

immunofluorescence using Vpr- and GR-specific antibodies

(Fig. 4A). Vprwt and GR colocalized at the nuclear rim and

nucleus of the Vprwt-transfected cells (a–d), whereas control

Page 5: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

Fig. 3. (A) In vitro interaction of Vpr and GR by GST pull-down assay:

Bacterially expressed GST or GST-GR were bound to glutathione-

sepharose beads and incubated with in vitro-translated Vpr mutants protein

labeled with [35S]methionine in 250 Al binding buffer (20 mM Tris–Hcl, pH

7.8, 100 mM NaCl, 10% Glycerol, 1 mM dithiothreitol, 1 mM EDTA, 1

mM phenylmethanesulfonyl fluoride, 1 mM leupeptin, 1 mM pepstatin).

Beads were washed extensively with binding buffer and bound proteins

were eluted by boiling them in reducing sample buffer, followed by SDS-

PAGE and autoradiography. Pull: GST-GR represents in vitro translated

Vpr mutants pulled using GST-GR beads; Pull: GST represents in vitro

translated Vpr mutants pulled using GST beads; and INPUT, represents in

vitro translated Vpr and Vpr products used in pull-down assays. (B) In vivo

interactions between GR and Vpr mutants: HeLa cells were transfected with

GR and Flag-tagged Vpr mutant expression plasmids. Immunoprecipitates

containing GR and Vpr were prepared with anti-GR antibody and applied to

a 12% SDS-PAGE gel. Immunoprecipitated complex was transferred to a

membrane and immunoblotted with anti-Flag M2 antibody followed by

chemiluminescence’s detection. All the mutants are designated on top of the

lane with their respective changes. Vector lane represents lysates from

pcDNA 3.1 vector-transfected cells.

D. Thotala et al. / Virology 328 (2004) 89–100 93

cells exhibited a weak cytoplasmic GR distribution and

when exposed to 10�6 AM of dexamethasone, GR trans-

located into the nucleus (e–h). Since dexamethasone is a

high affinity ligand for GR and participates in the trans-

location of GR into the nucleus, we compared the local-

ization pattern of Vpr and GR in the absence of additional

hormone exposure using charcoal-stripped serum containing

medium (a–d). It is interesting to note that HIV-1 Vprwt and

GR colocalized at the nuclear rim as well as in the nucleus.

Structure–function analysis of Vpr has shown that in

comparison to wild type Vpr, individual substitution

mutations in Vpr (especially in the helices) altered the

subcellular distribution (Kamata and Aida, 2000; Mahalin-

gam et al., 1997). First to examine the sub-cellular local-

ization of Vprwt and leucine mutants in the absence of

endogenous GR, we transfected CV-1 cells with Vpr

expression plasmids and performed indirect immunofluor-

escence using M2-Flag antibody (Table 1). Results indicate

that Vprwt, L42, and L67 exhibit nuclear membrane

distribution, whereas, L22, L23, L26, and L68 exhibited a

cytoplasmic pattern. Mutants L20, L39, and L64 showed

both cytoplasmic and perinuclear expression. Next, to

evaluate how the subcellular distribution of Vpr mutants

impacted GR localization, CV-1 cells were cotransfected

with pVpr leucine mutants and pGR and assessed for the

subcellular distribution of Vpr and GR (Suppl. Figs. 1A and

B; Table 1). Vpr mutants L23, L26, and L68 predominantly

exhibited a cytoplasmic pattern, whereas Vprwt, L20, L64,

and L67 localized in the nuclear membrane and cytoplasm

upon expression. Vpr L22A displayed a distinct nuclear

distribution in the presence of GR expression (Fig. 4B). Vpr

mutant L22A and GR colocalized into the nucleus in a

condensed form. To further establish the pattern of L22A

and GR, we performed confocal microscopy using cells

transfected with L22A and GR plasmids (Fig. 4C). Our

results indicate a distinct condensed pattern of L22A and

GR suggesting that L22A and the GR coactivator complex

might have a different localization pattern in the nucleus.

Additionally, it is interesting to note that none of this

complex localizes within the nucleolus compartment.

Together, these data suggest that the increase in GR-

VprL22A-mediated transactivation might be due partly to

the ability of GR and L22A to colocalize into the nucleus.

DNA binding affinity of GR–Vpr complex

Vpr has been shown to exert its transactivation effect

with GR through GRE sites, gel shift assay was performed

using GRE consensus sequences to assess the DNA binding

of Vpr–GR complex (Tseng et al., 2001). Cell lysates

derived from HeLa cells transfected with pVprwt resulted in

a specific complex with the labeled GRE sequences (Fig.

5A). This DNA–protein complex induced by Vpr was

similar to the complex induced by dexamethasone (1 AM),

whereas cell lysate from vector-transfected cells did not

show any distinct band at the corresponding position. Next

to test whether presence of both Vpr and GR is required for

GRE-binding, we transfected CV-1 cells with pVpr alone or

pVpr and pGR and extracted the lysates and performed

electrophoretic mobility shift assay (EMSA). Results

indicate that lysate from pVpr-transfected cells did not

show any binding, whereas Vpr–GR complex containing

lysates showed specific GRE binding (data not shown),

suggesting that Vpr–GR complex is required for GRE

binding activity in the absence of hormonal stimulation.

HIV-1 LTR and GRE reporter assays indicated that Vpr

leucine mutants exhibited differential transactivation. For

example, mutant L22A exhibited higher transactivation

activity and a distinct nuclear localization pattern. These

results suggest that GR-VprL22A might have significant

DNA-binding ability. To measure the ability of GR-L22A to

bind to the GRE domain in comparison to Vpr wild type, we

employed a competition gel-shift mobility assay using p32-

labeled double-stranded GRE oligonucleotides (Fig. 5B).

Different fold excess of cold GRE oligonucleotide (ranging

from 2.5- to 4-fold) was used. Competition assays revealed

Page 6: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

Fig. 4. (A) Expression and cellular colocalization of GR and pVpr mutant molecules: HeLa cells were transfected with Flag tagged pVprwt and pGR expression

plasmids and grown in a glass chamber slide. Forty-eight hours posttransfection cells were fixed and incubated with anti-GR and anti-Flag antibodies. Cells

were washed five times with PBS and incubated with secondary antibody conjugated to TRITC or FITC for 90 min at room temperature. Cells were washed

five times in PBS before mounting with Vectashield containing DAPI. Fluorescence was visualized with a Nikon microscope equipped with a digital camera

and images were captured as Photoshop files. Vprwt (a–d), represents cells cotransfected with pVprwt and phGRa expression plasmids. Panel Dex (e–h),

represents HeLa cells transfected with pcDNA3.1 vector control and phGRa expression plasmids and treated with 10 nM of dexamethosone and stained with

anti-GR antibody. (B and C) Immunofluorescence and Confocal visualization of VprL22A and GR colocalization in HeLa cells: HeLa cells were transfected

with Flag-tagged pVprL22A and phGRa expression plasmid and stained with anti-Flag and anti-GR antibody as before. B represents the immunofluorescence

studies and Panel C represents the confocal image analysis.

D. Thotala et al. / Virology 328 (2004) 89–10094

that Vpr wild type could be competed off with 2-fold excess

of cold GRE oligonucleotide (Lane 4), whereas a 4-fold

higher concentration was required to compete the GR-L22A

mutant (Lane 10), suggesting that nuclear extract containing

GR-VprL22A might have higher DNA-binding affinity to

GRE. This could partly explain the increase in Vpr–GR-

mediated transactivation in the case of VprL22A mutant.

Correlation of Vpr-mediated transactivation to virus

replication

To functionally correlate the transactivation effect of Vpr

in the context of virus replication, we performed a single

round infection assay using virus particles containing Vprwt

and Vpr mutants molecules by complementing pNL43 R-E-

plasmid with Vpr mutants and HxB2-Env plasmids. We

chose the single round infection system to precisely

quantitate the effect of virion-associated Vpr on HIV-1

replication. The ability of Vpr mutants to incorporate into

the virus particles was determined as described (Singh et

al., 2001) and presented in Table 1. Vpr mutant L39 and

L42 was not included in the single round infection due to

their deficiency in virion incorporation. The quantitation of

virus particles released into the media of the infected cells

by p24 antigen assay was used for evaluation. Virus

particles containing Vpr L22A showed maximum virus

Page 7: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

Fig. 5. (A) Electro Mobility Shift Assays: EMSAwas performed using labeled GRE oligonucleotide with HeLa cell nuclear extract. Lane oligo, labeled GRE

oligonucleotide in the absence of nuclear extract; Lane vector, GRE labeled oligonucleotide in the presence of the nuclear extract of HeLa cell transfected with

the vector plasmid; Lane Vpr-wt, GRE-labeled oligonucleotide in the presence of the nuclear extract of HeLa cells transfected with pVpr wt and phGRa

expression plasmids; Lane Dex, HeLa cell lysates with over expression of phGRa and stimulated with 10 nM dexamethasone. (B) Competition assays:

competition of GRE binding to HeLa cell nuclear extracts by molar excess (1- to 4-fold) of the cold GRE (lanes 3–10). Lane 1, labeled GRE in the absence of

nuclear extract; lane 2, labeled oligonucleotide in the presence of nuclear extract of cells alone; Lanes 3–6, nuclear extracts from cells transfected with pVprwt

in the presence of labeled GRE and 1-, 2-, 3-, and 4-fold excess of cold GRE oligonucleotide, respectively; and Lanes 7–10, nuclear extract from cells

transfected with pVprL22A in the presence of labeled GRE and 1-, 2-, 3-, and 4-fold excess of cold GRE oligonucleotide, respectively. Arrow indicates the

major complex formed.

D. Thotala et al. / Virology 328 (2004) 89–100 95

replication (5 ng/ml) followed by Vpr wild type (4 ng/ml)

on day 5, whereas mutants L23A and L68A resulted in

reduced levels (b2.5 ng/ml) of replication (Fig. 6). Similar

results were observed in multiple experiments using

PBMCs isolated from different healthy donors (data not

shown). Similarly, using the single round infection assay

with Vpr mutant incorporated virion molecules, we infected

CEM-GFP cells containing a plasmid encoding the green

fluorescent protein (humanized S65T GFP) driven by the

HIV-1 long terminal repeat and measured the virus

Table 1

Functional characteristics of HIV-1 Vpr mutants

Vpr mutants Expression Cell

cycle arrest

Virion

incorporation

Vpr wt ++ +++ yes

L20A ++ ++ yes

L22A ++ ++ yes

L23A + � yes

L26A ++ � yes

L39G ++ +++ no

L42G ++ � no

L64A ++ ++ yes

L67A ++ ++ yes

L68A ++ +/� yes

N.D., not determined.

infection by measuring the GFP in flow cytometry analysis

(data not shown). Vpr L22A mutant incorporated virus

showed an increase in number of cell number as well as

MFI compared to Vprwt incorporated virus particles,

suggesting that L22A increase virus replication during

early infection. These results indicate that an increase in

viral replication kinetics by Vpr is directly correlated with

the ability to transactivate HIV-1 LTR and GRE. However,

it is important to note that this affect is only due to the

virion-associated Vpr mutant molecules that are presented

Subcellular distribution of

Vpr in the absence of GR

Subcellular distribution of

Vpr in the presence of GR

Nuclear membrane Nuclear membrane

Cytoplasm + nuclear

membrane

Cytoplasm + nuclear

membrane

Cytoplasm Nucleus

Cytoplasm Cytoplasm

Cytoplasm Cytoplasm

Cytoplasm + nucleus N.D.

Nuclear membrane N.D.

Cytoplasm + nuclear

membrane

Cytoplasm + nuclear

membrane

Nuclear membrane Nuclear membrane +

cytoplasm

Cytoplasm Cytoplasm

Page 8: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

Fig. 6. Effect of virion-associated Vpr on virus replication during early

infection: Ficoll-Hypaque fractionated normal human PBMCs were

stimulated with PHA (5 Ag/ml) and infected with Vpr and Env trans-

complemented pNL43 R�E�. Cell free supernatants were collected at 24 h

intervals postinfection and assayed for the release of p24 antigen in the

medium. These experiments were repeated using PBMCs from different

seronegative donors and similar results were observed.

D. Thotala et al. / Virology 328 (2004) 89–10096

only in the virus particles. These results suggest that

presence of L22A Vpr mutant molecule in the virus

particles might increase virus replication during early

infection. Though we have observed an 8-fold increase in

transactivation assays, we did not see similar fold increase

in the single round virus replication assay. This could be

due to the presence of Vpr as virion-associated molecule s

and is absent in the proviral genome.

Discussion

AIDS pathogenesis is considered to be the result of the

interplay between the virus and the target cells of the host.

The cytopathic effects mediated by both the viral and

cellular genes have been suggested to be responsible for the

pathogenesis. Vpr has been implicated as one of the viral

genes contributing to AIDS pathogenesis along with HIV-1

env, tat, and nef. The features that qualify Vpr in this regard

are the following: (i) Vpr is incorporated into the virus

particles despite being a non-structural protein and the

amount of Vpr present in the virus particles is reported to be

in the range of few to ~2500 copies (Singh et al., 2001;

Tungaturthi et al., 2003), (ii) the presence of Vpr in cells

following virus infection, and (iii) the presence of free Vpr

(cell-free and virus-free) in the body fluids (serum and

CSF). Vpr has been reported to regulate multiple cellular

and viral functions. Vpr regulates both autologous and

heterologous promoters to enhance the transcription of viral

and cellular promoters. Additionally, Vpr-mediated cellular

effects have been linked to an increase in virus replication

and disease progression (Goh et al., 1998; Jowett et al.,

1995). Furthermore, the highly conserved nature of Vpr in

vivo suggests that Vpr plays an important role during early

infection and pathogenesis. Structure–function analyses of

Vpr have indicated that nuclear localization, cell cycle

arrest, and virion incorporation are mediated by distinct

domains of Vpr (Mahalingam et al., 1997). Recently, it has

been established that Vpr functions as a coactivator of

nuclear receptors through the signature motif LxxLL and

this function is distinct from its cell cycle regulation (Kino

et al., 1999; Sherman et al., 2000). Although recent mapping

studies suggest that the LxxLL domain in HIII of Vpr is the

interaction domain of nuclear receptors, there are also

related sequences in HI and HII. The role of these specific

amino acids involved in Vpr–GR interaction residues is

unclear. Here, we seek to identify the specific leucine

residues within the LxxLL domains involved in this

function by introducing individual leucine mutations. To

keep the structure and charge compatible we mutated the

leucine (L) residues into alanine (A) with the exception of

residues at L39 and L42, which were mutated to glycine

(G).

Our analysis of Vpr-mediated transactivation of GRE-

luciferase and HIV-1 LTR-luciferase revealed that leucine

residues are important for transactivation mediated by the

Vpr–GR complex. Disruption of leucine at these sites by

substitution with alanine/glycine significantly alters both

HIV-1 LTR and GRE-mediated transactivation in spite of

GR-Vpr mutants interaction. It is likely that the substitution

may interfere at the structure level. Such an effect could

result in disruption of specific residues that may participate

in Vpr’s structure, confirmation, and folding. Leucine

residues in the helical structure have been shown to

participate in protein–protein, protein–nucleic acid, and

protein–lipid interactions (Mahalingam et al., 1995; Tacke

et al., 1993). However, our results on GR and Vpr mutant

interactions show that GR physically interacts with all of the

Vpr mutants, suggesting that a single amino acid change in

these domains does not alter the binding. Additionally, it is

also possible that changes caused by a single leucine residue

could also compensated by other compensatory residues.

Previous studies by Kino et al. (2002a) and Sherman et al.

(2000) suggest that leucine mutations in these motifs abolish

the GR interaction. However, the Vpr mutant they used has

multiple leucine mutations within a single construct, thus

the discrepancy could be due the variation in these

constructs compared to ours. This could be due to the

changes in multiple leucine residues that might affect

structure and function. It is also possible that the adjacent

compensatory residues can rescue the loss of a single

leucine residue on Vpr–GR interaction. Though the Vpr–

GR interaction is not altered, changes in a leucine resides

such as L22 could alter other biophysical properties that

could result in altered subcellular distribution and DNA-

binding activity as observed.

Transcription of HIV-1 is mainly dependent on DNA-

binding proteins, transcription factors and viral transactiva-

tors. It is likely that Vpr transactivates HIV-1 LTR and/or

heterologous promoters through its interaction with GR and

therefore increases the ability of GR to bind to the

Page 9: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

D. Thotala et al. / Virology 328 (2004) 89–100 97

transcriptions factors. In this regard, it has been shown that

Vpr also has DNA-binding ability that is linked to the C-

terminal basic residues (Hogan et al., 2003; Zhang et al.,

1998). It is not known whether Vpr binds directly to the

GRE sequence. Our results indicate that Vpr-mediated

DNA-binding activity requires GR, suggesting that Vpr-

interacting proteins might mediate similar functions. It is

interesting to note that a mutation in the L22 residue results

in significantly higher transactivation (6- to 10-fold more)

suggesting that L22A may not alter the Vpr–GR interaction

but instead enhance the Vpr–GR–GRE binding affinity. The

later is supported by the subcellular localization studies

where L22A and GR colocalize in the nucleus. Our study

focuses on the effect of Vpr–GR interaction in the absence

of dexamethasone. Though dexamethasone increases Vpr-

mediated GRE transactivation, hormones present in culture

media are sufficient for Vpr–GR interaction. Sherman et al.

(2001) observed similar results, whereas Kino et al. (2002a)

suggested that dexamethasone is necessary for Vpr–GR

binding. However, our results are in agreement with

Sherman et al. (2001) indicating that the presence or

absence of dexamethasone does not alter the ability of GR

to bind with Vpr mutant molecules.

Vpr is a multi-phenotypic protein, with distinct domains

to control-specific functions. Vpr-mediated cell cycle arrest

has been linked to virus replication (Goh et al., 1998;

Gummuluru and Emerman, 1999); however, recent studies

suggest that the transactivation by Vpr is not directly

correlated with cell cycle function (Sherman et al. 2001).

Transactivation of autologous/heterologous promoters and

cell-cycle arrest functions are regulated by distinct domains

of Vpr and are independent of each other (Sawaya et al.,

2000; Sherman et al., 2000). Our studies on L22A and

L39G (single point mutation) mutants positive for cell cycle

arrest function with opposing transactivation activity further

confirms that the transactivation activity of HIV-1 Vpr

through GR is independent of cell cycle arrest function in

the context of leucine residues. Additional studies using

mutants from other regions are needed to further confirm

this observation.

It is interesting to note that L22A has significant trans-

activation effects in the absence of other external stimuli such

as glucocorticoids suggesting that a mutation at L22 might

alter some other properties of Vpr such as altered localization

and/or DNA-binding ability. Confocal microscopic analysis

and further competition assays using GRE-binding by EMSA

further confirms that the increased transactivation of L22 is

due its ability to bind the GRE DNAwith GR. It is interesting

to note that many Vpr-GR induced functions can be mediated

in the absence of dexamethasone, further confirming our

previous studies of Vpr mimicking GC activity (Ayyavoo et

al., 1997). These studies support that Vpr functions can be

suppressed by a new class of antivirals targeting these

pathways. Further studies are needed to identify new targets

that could block virus replication without interfering with the

host cell functions.

Materials and methods

Cells

HeLa, CV-1, and 293T cells were grown in DMEM

supplemented with 10% FCS, 1% glutamine and 1%

penicillin-streptomycin. HeLa and HeLa-T4 cells were

obtained from NIH, AIDS reagent program. CV-1 cells

were obtained from Dr. Frank Jenkins at the University of

Pittsburgh and 239T cells were a generous gift from Dr.

Michelle Calos, Stanford University CA. CEM-GFP cells

were obtained through AIDS Research and Reference

Reagent Program, DAIDS, HIH from Dr. Jacaques Corbeil

and maintained as described (Gervaix et al., 1997). Blood

from HIV-1-negative healthy donors was used to isolate

peripheral blood mononuclear cells (PBMCs) by Ficoll-

Hypaque (Pharmacia, USA) gradient centrifugation. Puri-

fied PBMCs were resuspended in RPMI 1640 supplemented

with 10% heat-inactivated FCS, stimulated with phytohe-

moagglutinin (PHA) (5 Ag/ml) for 2 days, and cultured in

IL-2 (5 U/ml) containing medium.

Plasmids

Mutants of HIV-1 vpr were generated using overlap PCR

and/or by Quick-change mutagenesis (Singh et al., 2001)

and were cloned under the control of the CMV promoter in

pCDNA3.1 (Invitrogen, CA) with a flag epitope. All the

Vpr mutant constructs were sequenced to verify the integrity

of the mutations. Proviral constructs pNL43 R+E�, pNL43

R�E�, pIIIB Env and HIV-1 LTR-luciferase reporter

constructs were obtained from NIH ARRRP, contributed

by Dr. Landau and Drs. Jeeninga and Berkhort, respectively.

pGRE (5X)-luciferase reporter plasmid was constructed by

PCR amplifying the 5XGRE consensus sequence from

pGRE5/EBV vector (USB, CA) using specific forward

(5VATACGCGGATCCTCTAGA AGATCCGCT3V) and

reverse (5VATCATACTCGAGGGCCCTCGCAGACA3V)primers. Amplified PCR product (290 bp) was cloned in

the upstream of a firefly luciferase gene reporter construct.

The human glucocorticoid receptor gene alpha (hGRa) was

PCR amplified using specific forward (5VATCGGGGATCCGATGGACTCCAAAGAATCA3 V) and reverse (5 VGTGGTCCTCGAGCTTTTGATGAAACAGAAG3V) pri-

mers and cloned into pcDNA 3.1 (Invitrogen) and pET41b

(+) (Novagen, CA) vectors for further eukaryotic and

prokaryotic expression studies, respectively. The integrity

of the plasmid DNA was tested by DNA sequence analysis

(ABI 7700, CA).

In vitro pull-down assay

HIV-1 GST-Vprwt and GST-hGRa were expressed

following 1 mM IPTG induction in BL21 cells. Protein

was purified using the BugBuster system (Novagen)

according to the manufacturer’s instruction. The integrity

Page 10: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

D. Thotala et al. / Virology 328 (2004) 89–10098

and purity of the proteins were analyzed by SDS-poly-

acrylamide gel electrophoresis followed by Coomassie blue

staining. For in vitro pull-down assays, GST beads

containing GR or Vpr were used. Vpr mutants were in

vitro translated using T7 in vitro transcription/translation

system (Promega, WI) as per the manufacturer’s instruction.

Equal amounts of in vitro translated products were added to

50 Al of GST-GR beads in 500 Al of GST bind/wash buffer

(Novagen) and rocked at 4 8C for 90 min. GST beads were

then washed with wash buffer three times and resuspended

in protein sample buffer. Proteins bound to GST beads were

resolved on a 12% SDS-PAGE and autoradiographed.

Transfection and luciferase assay

HeLa and CV-1 cells were transfected with HIV-1 LTR

luciferase or GRE5X-luciferase reporter plasmid (1 Ag) inthe presence and absence of Vpr mutants (0.5 Ag), pGR and

pCMVh-Gal using lipofectamine (Invitrogen). Forty-eight

hours posttransfection, cells were lysed in 500 Al of 1�reporter lysis buffer and luciferase activity was measured

following the manufacturer’s protocol (Promega). Trans-

fection efficiency was normalized by transfecting with

CMV h-gal plasmid and analyzing the h-gal activity

(Promega).

In vivo interaction studies

HeLa cells were transfected with Vpr mutant plasmids

using lipofectamine. Forty-eight hours posttransfection, the

cells were washed with PBS and lysed in RIPA buffer (50

mM Tris–HCl, [pH 7.6], 150 mM NaCl, 0.5% Triton X-100,

0.5% deoxycholic acid, 0.1% sodium dodecyl sulfate (SDS),

1 mM phenylmethylsulfonyl fluoride (PMSF) and PI

cocktail). Cell lysate was centrifuged to remove the cell

debris. Protein estimation of the cell lysate was carried out

using Bradford reagent (Bio-Rad, Richmond, CA). Equiv-

alent amount (150 Ag) of cellular protein was subjected to

immunoprecipitation using anti-GR antibodies. Immunopre-

cipitated complex was analyzed on SDS-PAGE and

immunoblotted for Vpr using M2 anti-FLAG antibody

(1:200) from Sigma, MO.

Immunofluorescence and confocal microscopy

HeLa cells were seeded in four-well chamber slides and

transfected with Vpr or GR expression plasmid DNA using

Lipofectamine. Forty-eight hours posttransfection, cells

were washed with PBS and fixed in methanol at room

temperature for 10 min. After washing three times with

PBS, cells were incubated with anti-GR antibody (Affinity

BioReagents, CO) at 37 8C in a humidified incubator for 90

min. Primary antibody was washed three times with PBS

and then incubated with anti-Flag M2 monoclonal antibody-

FITC (Sigma) to detect Vpr, and anti rabbit-TRITC to detect

GR. Following several washes with PBS, cells were

mounted with Vectashield containing DAPI (Vector Labo-

ratories, CA). Immunofluorescence was detected using

Nikon inverted fluorescence microscope with appropriate

filters. Confocal microscopy was performed using a Leica

TCS NT confocal tri-laser scanning inverted microscope

(Wetzlar, Germany).

Electrophoretic mobility shift assay

Oligonucleotide containing the GRE consensus sequence

(5VGACCCTAGAGGATCTGTACAGGATGTTCTAGAT

3V) was either used as a probe or competitor. One picomolar

of GRE oligonucleotide was end-labeled with 2 Al of p32ATP using T4-polynucleotide kinase (Promega) in a 50-Alreaction. The labeled oligonucleotide was purified to

remove unincorporated labels using a Qiaquick nucleotide

removal kit (Qiagen, Valencia, CA). HeLa cells were

transfected with Vpr wild type or Vpr mutants as described

above. Forty-eight hours posttransfection, cells were washed

with PBS and nuclear extracts were prepared using the NE-

PER nuclear and cytoplasmic extraction kit following the

manufacturer’s instructions (Pierce, IL). DNA binding

reactions were carried out with approximately 1 pM of

p32 labeled GR consensus double-strand oligonucleotide

(50,000 cpm) and 5.0 Ag nuclear extract (Tseng et al., 2001).Briefly, nuclear extract was pre-incubated in a binding

buffer [50 mM Tris–HCl (pH 7.6), 0.5 mM EDTA (pH 8.0),

0.5 mM DTT, 50% Glycerol, 2 Ag of poly: dIdC] in a final

volume of 30 Al at room temperature for 10 min. The

labeled oligo was then added and incubated at room

temperature for 30 min. For competition experiments, a

cold probe was added to the nuclear extract 20 min before

the addition of labeled probe. The samples were analyzed on

a 5% non-denaturing polyacrylamide gel (5% polyacryla-

mide gel, 50 mM Tris–Glycine) and autoradiographed.

Single round infection assay

To assess the effect of Vpr as a virion-associated

molecule, we have used pseudotype viruses containing

Vprwt or Vpr mutant molecules. A single round infection

assay was used to study the effect of different Vpr mutants

on HIV-1 replication by transfecting 293T cells with

proviral DNA pNL43 R�E� with pEnv and Vpr expression

plasmid produced pseudotype viruses. Seventy-two hours

posttransfection culture supernatant was collected and

assayed for virus production by quantitating the p24 by

ELISA. PBMCs (5 � 106) were infected with 10 ng of p24

antigen-equivalent viruses for 4 h, washed thoroughly with

RPMI medium, and resuspended in appropriate growth

medium. Supernatant was collected at 24-h intervals to

measure virus replication by analyzing p24 by ELISA

(AIDS Reagent program, NIH). Additionally, to measure

the effect of virion-associated Vpr molecules during early

infection, CEM-GFP cells were infected with the leucine

mutant complemented pNL43 R-E-/VSV-G-Env viruses at

Page 11: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

D. Thotala et al. / Virology 328 (2004) 89–100 99

0.01 multiplicity of infection (MOI) for 6 h at 37 8C as

described (Gervaix et al., 1997). The cells then were washed

three times with PBS, resuspended in culture medium, and

incubated at 378C in 5% CO2. Aliquots of cells were taken

at days 2 and 4 and fixed in 2% paraformaldehyde. The

cells were analyzed by flow cytometry followed by cell

quest software to measure the MFI and percent positive

cells.

Acknowledgments

HIV-1 LTR-luciferase construct, pNL43 R+E� and

pNL43 R�E�, pHXB2-Env, pHIV-1 LTR-luciferase plas-

mids and CEM-GFP were obtained from the AIDS Research

and reference reagent Program, Division of AIDS, NIAID.

We thank Dr. Pavlakis for his generous gift of RU486 for

our initial experiments. We thank Dr. Frank Jenkins at the

University of Pittsburgh for CV-1 cells and Dr. Michelle

Calos, at Stanford University, for 293T cells. We thank Dr.

Indranil Mukhopadhyay, University of Pittsburgh for his

help with the statistical analysis. This work was supported

by AI50463 from NIAID, NIH to VA.

Appendix A. Supplementary data

Supplementary data associated with this article can be

found, in the online version, at doi:10.1016/j.virol.2004.07.

013.

References

Agostini, I., Navarro, J.M., Rey, F., Bouhamdan, M., Spire, B., Vigne, R.,

Sire, J., 1996. The human immunodeficiency virus type 1 Vpr

transactivator: cooperation with promoter-bound activator domains

and binding to TFIIB. J. Mol. Biol. 261, 599–606.

Ayyavoo, V., Mahboubi, A., Mahalingam, S., Ramalingam, R.,

Kudchodkar, S., Williams, W.V., Green, D.R., Weiner, D.B., 1997.

HIV-1 Vpr suppresses immune activation and apoptosis through

regulation of nuclear factor kB. Nat. Med. 3, 1117–1123.

Balliet, J.W., Kolson, D.L., Eiger, G., Kim, F.M., McGann, K.A.,

Srinivasan, A., Collman, R., 1994. Distinct effects in primary macro-

phages and lymphocytes of the human immunodeficiency virus type 1

accessory genes vpr, vpu, and nef: mutational analysis of a primary

HIV-1 isolate. Virology 200, 623–631.

Cadepond, F., Gasc, J.M., Delahaye, F., Jibard, N., Schweizer-Groyer, G.,

Segard-Maurel, I., Evans, R., Baulieu, E.E., 1992. Hormonal regulation

of the nuclear localization signals of the human glucocorticosteroid

receptor. Exp. Cell Res. 201, 99–108.

Cohen, E.A., Dehni, G., Sodroski, J.G., Haseltine, W.A., 1990. Human

immunodeficiency virus vpr product is a virion-associated regulatory

protein. J. Virol. 64, 3097–3099.

Connor, R.I., Chen, B.K., Choe, S., Landau, N.R., 1995. Vpr is required for

efficient replication of human immunodeficiency virus type-1 in

mononuclear phagocytes. Virology 206, 935–944.

DeFranco, D.B., 2002. Navigating steroid homone receptors through the

nuclear compartment. Mol. Endocrinol. 16, 1449–1455.

Di Marzio, P., Choe, S., Ebright, M., Knoblauch, R., Landau, N.R., 1995.

Mutational analysis of cell cycle arrest, nuclear localization, and virion

packaging of human immunodeficiency virus type 1 Vpr. J. Virol. 69,

7909–7916.

Felzien, L.K., Woffendin, C., Hottiger, M.O., Subbramanian, R.A., Cohen,

E.A., Nabel, G.J., 1998. HIV transcriptional activation by the accessory

protein, VPR is mediated by the p300 co-activator. Proc. Natl. Acad.

Sci. U.S.A. 95, 5281–5286.

Feng, W., Ribeiro, R.C., Wagner, R.L., Nguyen, H., Apriletti, J.W.,

Fletterick, R.J., Baxter, J.D., Kushner, P.J., West, B.L., 1998. Hormone

dependent coactivator binding to a hydrophobic cleft on nuclear

receptors. Science 280, 1747–1749.

Gervaix, A., West, D., Leoni, L.M., Richman, D.D., Wong-Staal, F.,

Corbeil, J.A., 1997. A new reporter cell line to monitor HIV infection

and drug susceptibility in vitro. Proc. Natl. Acad. Sci. U.S.A. 94,

4653–4658.

Goh, W.C., Rogel, M.E., Kinsey, C.M., Michael, S.F., Fultz, P.N., Nowak,

M.A., Hahn, B.H., Emerman, M., 1998. HIV-1 Vpr increases viral

expression by manipulation of the cell cycle: a mechanism for selection

of Vpr in vivo. Nat. Med. 4, 65–71.

Gummuluru, S., Emerman, M., 1999. Cell cycle- and Vpr-mediated

regulation of human immunodeficiency virus type 1 expression in

primary and transformed T-cell lines. J. Virol. 73, 5422–5430.

Heery, D.M., Kalkhoven, E., Hoare, S., Parker, M.G., 1997. A signature

motif in transcriptional co-activators mediates binding to nuclear

receptors. Nature 387, 733–736.

Heinzinger, N.K., Bukinsky, M.I., Haggerty, S.A., Kewalramani, V., Lee,

M.A., Gendelman, H.E., Ratner, L., Stevenson, M., Emerman, M.,

1994. The Vpr protein of human immunodeficiency virus type 1

influences nuclear localization of viral nucleic acids in non-dividing

host cells. Proc. Natl. Acad. Sci. U.S.A. 91, 7311–7315.

Hogan, T.H., Nonnemacher, M.R., Krebs, F.C., Henderson, A., Wigdahl,

B., 2003. HIV-1 Vpr binding to HIV-1 LTR C/EBP cis-acting elements

and adjacent regions is sequence-specific. Biomed. Pharmacother. 57,

41–48.

Hrimech, M., Yao, X.J., Bachand, F., Rougeau, N., Cohen, E.A., 1999.

Human immunodeficiency virus type 1 (HIV-1) Vpr functions as

an immediate early protein during HIV-1 infection. J. Virol. 73,

4101–4109.

Htun, H., Barsony, J., Reni, I., Gould, D.L., Hager, G.L., 1996.

Visualization of glucocorticoid receptor translocation and intranuclear

organization in living cells with a green fluorescent protein chimers.

Proc. Natl. Acad. Sci. U.S.A. 93, 4845–4850.

Jowett, J.B., Planelles, V., Poon, B., Shah, N.P., Chen, M.L., Chen, I.S.,

1995. The human immunodeficiency virus type 1 Vpr gene arrests

infected T cells in the G2+M phase of the cell cycle. J. Virol. 69,

6304–6313.

Kamata, M., Aida, Y., 2000. Two putative a-helical domains of human

immunodeficiency virus type 1 Vpr mediate nuclear localization by at

least two mechanisms. J. Virol. 74, 7179–7186.

Kino, T., Gragerov, A., Kopp, J.B., Stauber, H., Pavalakis, G.N., Chrousos,

G.P., 1999. The HIV-1 virion-associated protein vpr is a coactivator of

the human glucocorticoid receptor. J. Exp. Med. 189, 51–62.

Kino, T., Gragerov, A., Slobodskaya, O., Tsopananomichalou, M.,

Chrouusos, G.P., Paylakis, G.N., 2002a. Human immunodeficiency

virus type 1 (HIV-1) accessory protein Vpr induces transcription of the

HIV-1 and glucocorticoid-responsive promoters by binding directly to

p300/CBP coactivators. J. Virol. 76, 9724–9734.

Kino, T., Tsukamoto, M., Chrousos, G.P., 2002b. Transcription factor

TFIIH components enhance the GR coactivator activity but not the cell

cycle-arresting activity of the human immunodeficiency virus type-1

protein Vpr. Biochem. Biophys. Res. Commun. 298, 17–23.

Levy, D.N., Fernandes, L.S., Williams, W.V., Weiner, D.B., 1993. Induction

of cell differentiation by human immunodeficiency virus 1 vpr. Cell 72,

541–550.

Levy, D.N., Rafaeli, Y., Weiner, D.B., 1995. Extracellular vpr protein

increases cellular permissiveness of human immunodeficiency virus

replication and reactivates virus from latency. J. Virol. 69, 1243–1252.

Page 12: Structure–functional analysis of human immunodeficiency virus … · 2017. 2. 20. · Structure–functional analysis of human immunodeficiency virus type 1 (HIV-1) Vpr: role of

D. Thotala et al. / Virology 328 (2004) 89–100100

Li, D.P., Periyasamy, S., Jones, T.J., Sanchez, R., 2002. Heat and chemical

shock potential of glucocorticoid receptor transactivation requires heat

shock factor (HSF) activity. J. Biol. Chem. 275, 26058–26065.

Mahalingam, S., Khan, S.A., Jabbar, M.A., Monken, C.E., Collman, R.,

Srinivasan, A., 1995. Mutagenesis of the putative alpha helical domain

of the Vpr protein of human immunodeficiency virus type 1: effect on

stability and virion incorporation. Proc. Natl. Acad. Sci. U.S.A. 92,

3794–3798.

Mahalingam, S., Ayyavoo, V., Patel, M., Kieber-Emmons, T., Weiner, D.B.,

1997. Nuclear import, virion incorporation, and cell cycle arrest/

differentiation are mediated by distinct functional domains of human

immunodeficiency virus type 1 Vpr. J. Virol. 71, 6339–6347.

Morellet, N., Bouaziz, S., Petitjean, P., Roques, B.P., 2003. NMR structure

of the HIV-1 regulatory protein VPR. J. Mol. Biol. 327, 215–227.

Nakamura, T., Suzuki, H., Okamoto, T., Kotani, S., Atsuji, Y., Tanaka, T.,

Ito, Y., 2002. Recombinant Vpr (rVpr) causes augmentation of HIV-1

p24 Ag level in U1 cells through its ability to induce the secretion of

TNF. Virus Res. 90, 263–268.

Northrop, J.P., Crabtree, G.R., Mattila, P.S., 1992. Negative regulation of

interleukin 2 transcription by the glucocorticoid receptor. J. Exp. Med.

175 (5), 1235–1245.

Paliogianni, F., Boumpas, D.T., 1995. Glucocorticoids regulate calcineurin-

dependent trans-activating pathways for interleukin-2 gene transcription

in human T lymphocytes. Transplantation 59 (9), 1333–1339.

Sawaya, B.E., Khalili, K., Gordon, J., Taube, R., Amini, S., 2000.

Cooperative interaction between HIV-1 regulatory proteins Tat and

Vpr modulates transcription of the viral genome. J. Biol. Chem. 275,

35209–35214.

Schmidt, J., Fleissner, S., Heimann-Weitschat, I., Lindstaedt, R., Pomberg,

B., Werner, U., Szelenyi, I., 1994. Effect of corticosteroids, cyclosporin

A and methotrexate on cytokine release from monocytes and T-cell

subsets. Immunopharmacology 27 (3), 173–179.

Sherman, M.P., de Noronha, C.M., Pearce, D., Greene, W.C., 2000. Human

immunodeficiency virus type 1 vpr contains two leucine rich helices

that mediate glucocorticoid receptor co-activation independently of its

effects on cell cycle arrest. J. Virol. 74, 8159–8165.

Singh, S.P., Tungaturthi, P., Cartas, M., Tomkowicz, B., Rizyi, T.A., Khan,

S.A., Kalyanaraman, V.S., Srinivasan, A., 2001. Virion-associated HIV-

1 Vpr: variable amount in virus particles derived from cells upon virus

infection or proviral DNA transfection. Virology 283, 78–83.

Subbramanian, R.A., Kessous-Elbaz, A., Lodge, R., Forget, J., Yao,

X.J., Bergeron, D., Cohen, E.A., 1998. Human immunodeficiency

virus type 1 Vpr is a positive regulator of viral transcription and

infectivity in primary human macrophages. J. Exp. Med. 187,

1103–1111.

Tacke, E., Schmitz, J., Prufer, D., Rohde, W., 1993. Mutational analysis of

the nucleic acid-binding 17-kDa phosphoprotein of potato leafroll

luteovirus identifies an amphipathic alpha helix as the domain for

protein/protein interactions. Virology 197, 274–282.

Tseng, Y.T., Stabila, J.P., Nguyen, T.T., McGonnigal, B.G., Waschek, J.A.,

Padbury, J.F., 2001. A novel glucocorticoid regulatory unit mediates the

hormone responsiveness of the beta1-adrenergic receptor gene. Mol.

Cell. Endocrinol. 181, 165–178.

Tungaturthi, P.K., Sawaya, B.E., Singh, S.P., Tomkowicz, B., Ayyavoo, V.,

Khalili, K., Collman, R.G., Amini, S., Srinivasan, A., 2003. Role of

HIV-1 Vpr in AIDS pathogenesis: relevance and implications of

intravirion, intracellular and free Vpr. Biomed. Pharmacother. 57,

20–24.

Vanitharani, R., Mahalingam, S., Rafaeli, Y., Singh, S.P., Srinivasan, A.,

Weiner, D.B., Ayyavoo, V., 2001. HIV-1 vpr regulates viral tran-

scription in part through the GRE and upstream region (�176 to �670)

of HIV-1 LTR. Virology 289, 334–342.

Wang, L., Mukherjee, S., Jia, F., Narayan, O., Zhao, L.J., 1995. Interaction

of virion protein Vpr of human immunodeficiency virus type 1 with

cellular transcription factor Sp1 and trans-activation of viral long

terminal repeat. J. Biol. Chem. 270, 25564–25569.

Wecker, K., Morellet, N., Bouaziz, S., Roques, B.P., 2002. NMR structure

of the HIV-1 regulatory protein Vpr in H20/trifluoroethanol. Compar-

ison with the Vpr N-terminal (1–51) and C-terminal (52–96) domains.

Eur. J. Biochem. 269, 3779–3788.

Yao, X.J., Mouland, A.J., Subbramanian, R.A., Forget, J., Rougeau, N.,

Bergeron, D., Cohen, E.A., 1998. Vpr stimulates viral expression and

induces cell killing in human immunodeficiency virus type-1 infected

dividing Jurkat T cells. J. Virol. 72, 4686–4693.

Zhang, S., Pointer, D., Singer, G., Feng, Y., Park, K., Zhao, L.J., 1998.

Direct binding to nucleic acids by Vpr of human immunodeficiency

virus type 1. Gene 212, 157–166.

Zhao, L.J., Mukherjee, S., Narayan, O., 1994. Biochemical mechanism of

HIV-1 Vpr function: specific interaction with a cellular protein. J. Biol.

Chem. 269, 15577–15582.