It remains controversial whether human T lymphotropic virus type I (HTLV-I) coinfection leads to more rapid progression of human immunodeficiency virus (HIV) disease in dually infected individuals. To investigate whether HTLV-I infection of certain cells can modulate HIV-1 infection of surrounding cells, primary CD4+ T cells were treated with cell-free supernatants from HTLV-I–infected MT-2 cell cultures. The primary CD4+ T cells became resistant to macrophage (M)-tropic HIV-1 but highly susceptible to T cell (T)-tropic HIV-1. The CC chemokines RANTES (regulated on activation, normal T cell expressed and secreted), macrophage inflammatory protein (MIP)-1α, and MIP-1β in the MT-2 cell supernatants were identified as the major suppressive factors for M-tropic HIV-1 as well as the enhancers of T-tropic HIV-1 infection, whereas soluble Tax protein increased susceptibility to both M- and T-tropic HIV-1. The effect of Tax or CC chemokines on T-tropic HIV-1 was mediated, at least in part, by increasing HIV Env-mediated fusogenicity. Our data suggest that the net effect of HTLV-I coinfection in HIV-infected individuals favors the transition from M- to T-tropic HIV phenotype, which is generally indicative of progressive HIV disease.

During the natural course of human immunodeficiency virus (HIV) infection, a transition of HIV phenotypes has been observed (13). During primary infection and the clinically latent period, most HIV isolates are macrophage (M)-tropic (4), whereas in the advanced stage of HIV disease more cytopathic, T cell (T)-tropic viruses predominate (13). However, the host or environmental factors affecting such a transition and the reason why HIV disease progresses more rapidly in certain individuals remain unclear.

The effects of coinfection with other pathogens on the pathogenesis of HIV-1 disease have been extensively studied over the past decade. For example, a number of viral transactivators have been shown to upregulate expression from the HIV-1 LTR (58), and more recently, a human cytomegalovirus (HCMV)1–encoded chemokine receptor was found to serve as an HIV-1 entry cofactor (9). Although these in vitro studies provide important mechanistic information, the effects mediated by those pathogens required coinfection of the pathogens and HIV-1 in the same cell, a phenomenon that is considered to occur rarely in vivo.

Several laboratory and epidemiologic studies have suggested that human T lymphotropic virus type I (HTLV-I) infection exacerbates the cytopathic effects of HIV infection and accelerates the clinical progression of HIV disease in coinfected individuals (1016); however, other studies have not confirmed these observations (17). To determine the potential mechanisms whereby HTLV-I infection might modulate HIV-1 infection in dually infected individuals, we used in vitro models consisting of primary CD4+ T cells either cocultured with HTLV-I–transformed MT-2 cells in a transwell system or incubated in the presence of cell-free supernatants from MT-2 cell cultures. We demonstrate that crude supernatants from MT-2 cell cultures inhibit replication of M-tropic HIV-1, but enhance that of T- or dual-tropic HIV-1. In addition, the CC chemokines RANTES (regulated on activation normal T cell expressed and secreted), macrophage inflammatory protein (MIP)–1α, and MIP-1β in the supernatants of the MT-2 cell cultures were identified as the major suppressive factors for M-tropic HIV-1 as well as the positively regulating factors for T-tropic HIV-1. Furthermore, soluble Tax protein was shown to be a positively regulating factor for both HIV-1 phenotypes. The effect of Tax or CC chemokines is mediated, at least in part, by enhancing HIV-1 Env-mediated fusogenic activity. This study suggests that HTLV-I coinfection in HIV-infected individuals may facilitate transition from an M- to a T-tropic HIV phenotype, which is generally indicative of progression to an advanced stage of HIV disease.

Cells.

HTLV-I–transformed MT-2 (18) and HUT-102 (19) cells were provided by G. Franchini (National Cancer Institute, NIH, Bethesda, MD) and grown in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS; GIBCO BRL, Gaithersburg, MD).

JPX9 cells and JPX/M cells are Jurkat cells expressing the wild-type or mutant form, respectively, of HTLV-I Tax under the control of the metallothionein promoter (20, 21). Expression of Tax in these cells was induced by treatment of cells with 10 μM CdCl2 for 2 d.

PBMCs were isolated from healthy volunteers seronegative for both HIV and HTLV, as previously described (22), and CD4+ T cells were negatively selected by column exclusion (CD4+ subset enrichment columns; R&D Systems, Minneapolis, MN). Purity of CD4+ T cells was 95% or more, determined by flow cytometric analysis (data not shown).

Propagation of MT-2–conditioned Medium.

Crude supernatants from MT-2 cell cultures were clarified by low-speed centrifugation (3,000 rpm, 30 min) and filtered through 0.2-μm filters to remove cells. The medium (5 ml/aliquot) was incubated with either control rabbit serum (20 μl), anti-Tax antiserum (20 μl [reference 23]), or a mixture of monoclonal antibodies to RANTES, MIP-1α, and MIP-1β (50 μg/ml each; R&D Systems) at 4°C for 2 h, followed by protein A/G sepharose (UltraLink Immobilized Protein A/G; Pierce, Rockford, IL). Immune complexes bound on the sepharose were removed by extensive washing. The presence of soluble Tax protein in the medium was demonstrated by immunoprecipitation using anti-Tax serum and protein A/G sepharose, and the concentrations of CC chemokines in the medium were determined by ELISAs using commercially available kits (R&D Systems). Where indicated, the supernatants were ultracentrifuged at 20,000 rpm for 1 h to pellet HTLV-I virions. Crude supernatants from HUT-102 or A3.01 cells were also propagated in a similar manner.

Purification of Recombinant Tax Protein Expressed in Escherichia coli DH5a Strain and HTLV-I Particles from MT-2 Cells.

Tax protein was expressed in E. coli DH5a strain transformed with pGST-Tax (provided by K.T. Jeang, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD), and purified as previously described (24). As a control, glutathione S-transferase (GST) protein was also expressed and purified in the same manner. Both preparations were dialyzed and filtered, and protein concentrations were measured by colorimetric assays (Bio-Rad Laboratories, Hercules, CA). Levels of contaminated endotoxin in the preparations were <10 ng/mg protein (Limulus Amebocyte Lysate Test; BioWhittaker, Inc., Walkersville, MD). Purity and identity of the proteins were determined by SDS-PAGE followed by Coomassie blue staining and Western blotting using anti-Tax antiserum (1:2,000), respectively. In some experiments, the GST-Tax preparation was treated by anti-Tax serum followed by protein A/G sepharose to specifically remove GST–Tax fusion protein. Nuclear extracts were prepared from CD4+ T cells treated with either GST or GST-Tax as previously described (24), and were analyzed by Western blotting to monitor cellular uptake of the protein.

HTLV-I particles were purified from MT-2 cell culture supernatants as previously described (25).

Virus Strains and Infection.

The following virus stocks were propagated by transfecting 293T cells with plasmids encoding the respective molecular clones: NL4-3 (T-tropic [reference 26]); 89.6 (dual-tropic [reference 27]); and ADA8 (M-tropic [reference 28]). Approximately 2 × 105 CD4+ T cells were either pretreated with 50% MT-2–conditioned medium or control medium (A3.01-conditioned medium) or cocultured with MT-2 cells in a transwell system that separates the two cell populations by 0.2-μm pore membrane for 3 d, and then infected with the above molecular clone stock at a multiplicity of infection (MOI) of ∼0.05. Approximately half of each infected cell culture supernatant was replaced with the same medium every 4 d, and reverse transcriptase (RT) activity in the supernatants was measured as previously described (29).

Single-round Virus Replication Assay.

NL4-3-Luc-RE virus stocks pseudotyped by various Envs were generated by transfecting 293T cells with pNL4-3-Luc-RE and plasmids expressing Env from either HXB2 (T-tropic), 89.6 (dual-tropic [reference 30]), ADA (M-tropic [reference 30]) or amphotropic murine leukemia virus (AMV), as previously described (31). Approximately 105 primary CD4+ T cells were infected with the above luciferase reporter virus (5 × 105 cpm RT activity), and luciferase activity of the cell lysates was measured 4 d after infection using commercially available reagents (Promega, Madison, WI).

Fusion Assay.

Recombinant vaccinia virus (rVV)-based cell fusion assays were performed as previously described (32, 33). In brief, primary CD4+ T cells (fusion targets) were infected with vTF7-3 (expressing T7 RNA polymerase) at an MOI of 10; as fusion effectors, BSC-1 cells or CD4+ T cells were infected with vCB21R (encoding the lacZ gene driven by the T7 promoter) as well as rVV expressing the mutant HIV Env (vCB16), wild-type IIIB (T-tropic) Env (vCB41), or Ba-L (M-tropic) Env (vCB43), each at an MOI of 10. Cells were incubated at 31°C overnight, and both fusion targets and fusion effectors were mixed per well in 96-well flat-bottomed microtiter plates in the presence of 40 μg/ml of cytosine arabinoside. After 4 h at 37°C, β-galactosidase activity in the cell lysates were assayed by measuring absorbance at 570 nm using a microtiter absorbance reader (Molecular Dynamics, Sunnyvale, CA).

HTLV-I–transformed MT-2 Cells Produce Soluble Factor(s) that Inhibits Infection of Primary CD4+T Cells with M-tropic HIV-1 and that Enhance Replication of T-tropic HIV-1.

The ability of HTLV-I to modulate HIV-1 infection without coinfection of the same cell was initially evaluated by coculturing primary CD4+ T cells with MT-2 cells in a transwell system for 3 d before HIV-1 infection. In this system, the two cell populations were separated by a 0.2-μm pore membrane to avoid cell-to-cell contact, which is required for the establishment of infection with highly cell-associated HTLV-I (3436). Under these conditions, immortalization of CD4+ T cells did not occur and HTLV-I p24 antigen was not detected in the CD4+ T cell culture supernatants over 3 wk (data not shown). Primary CD4+ T cells pretreated in the MT-2 cell coculture system described above were then infected with either M-tropic HIV-1ADA or T-tropic HIV-1NL4-3, and RT activity was measured in the infected cell supernatants. CD4+ T cells cocultured with MT-2 cells before infection produced less HIV-1ADA but more HIV-1NL4-3 compared with control CD4+ T cells on day 4 after infection (Fig. 1, A and B); however, these effects were not observed beyond 8 d after infection.

The above results suggested that MT-2–conditioned medium contains soluble factor(s) that suppress M-tropic but enhance T-tropic HIV-1 infection; however, neither the positive nor the negative effects persisted beyond a few days after exposure to the supernatants. To provide continuous exposure of CD4+ T cells to the putative factors, crude cell-free supernatants were prepared from MT-2 cell cultures. Primary CD4+ T cells were pretreated for 3 d and then continuously exposed to a 1:1 dilution of the supernatants after infection with T-tropic HIV-1NL4-3, dual-tropic HIV-189.6, or M-tropic HIV-1ADA. In this setting, the enhancing effects of the supernatant on T-tropic and dual-tropic HIV-1 infection and the suppressive effects on M-tropic HIV-1 infection were sustained throughout the 12-d culture period (Fig. 2, A–C).

The effect by continuous treatment with MT-2–conditioned medium on HIV-1 infection of primary CD4+ T cells was also investigated in single-round virus replication assays. In this system, the input virus is pseudotyped by Envs of interest and expresses the luciferase gene after integration of proviral DNA into the host genome; however, it is unable to complete its life cycle because of the lack of de novo Env production. Therefore, luciferase activity in the infected cell lysates correlates well with the efficiency of virus replication during early events in the viral replicative cycle. As expected, coculture with MT-2 cells in a transwell system before infection (data not shown) or pretreatment with MT-2–conditioned medium (Fig. 3) reduced infectivity of virus pseudotyped by M-tropic Env but increased infectivity of virus pseudotyped by T-tropic HIV-1 Env, suggesting that the steps during the HIV-1 replicative cycle that are influenced by HTLV-I coinfection include early events. Similar results were obtained by using crude supernatants from another HTLV-I producing cell line, HUT-102 (data not shown).

Identification of Negative and Positive Factors Produced by MT-2 Cells.

MT-2 cells are known to produce HTLV-I virions, viral proteins such as Tax (3739), and a number of cytokines (for review see reference 40). Of note, HTLV-I–transformed CD8+ T cells were recently shown to produce the CC chemokines RANTES, MIP-1α, and MIP-1β, and to suppress infection of a CD4+ T cell line with M-tropic HIV-1 (41). In contrast, previous studies had demonstrated that mitogenic stimulation induced by HTLV-I virions increased HIV-1 replication (25), and that HTLV-I Tax protein transactivated HIV-1 LTR (8) as well as induced expression of several cytokines including CC chemokines (42). Therefore, it is likely that the net effect of crude supernatants from HTLV-I–infected cells on HIV-1 infection of adjacent cells depends upon the balance and/or accumulation of these factors. To clarify which factor(s) is responsible for the positive or negative effects on HIV-1 infection, each component (soluble Tax protein, HIV-suppressive CC chemokines, or HTLV-I virions) was removed from the crude supernatants as described in Materials and Methods. Fig. 4,A demonstrates that MT-2 cell supernatants contain soluble Tax protein (lane 2), which was successfully removed by anti-Tax antiserum followed by protein A/G sepharose treatment (lane 4). MT-2 cell supernatants also contain substantial amounts of the CC chemokines RANTES, MIP-1α, and MIP-1β, which were markedly reduced after treatment with specific antibodies and protein A/G sepharose (Fig. 4 B).

CD4+ T cells were treated with these supernatant preparations and infected with NL4-3Luc-RE virus pseudotyped by Env from either HIV-1HXB2 (T-tropic), HIV-189.6 (dual-tropic), or HIV-1ADA (M-tropic). Supernatants subtracted for the CC chemokines RANTES, MIP-1α, and MIP-1β lost their inhibitory effects on M-tropic HIV-1, and instead enhanced its replication, whereas supernatants subtracted for either Tax or HTLV-I virions retained or further augmented their inhibitory activity. In contrast, subtraction of either component (Tax, CC chemokines, or virions) reduced the ability of the supernatants to increase replication of T- (Fig. 4,C) or dual-tropic (data not shown) HIV-1. These results indicate that both soluble Tax protein and HTLV-I virions enhance HIV-1 infection of both M-tropic and T-tropic viral phenotypes, whereas the CC chemokines in the supernatants inhibit M-tropic HIV-1 infection and enhance infection with T-tropic HIV-1 infection. Therefore, supernatants from HTLV-I–infected cells invariably enhance infection with T-tropic HIV-1, since each of the identifiable factors in the supernatants (Tax, CC chemokines, and HTLV-I virions) have a positive effect on replication of viruses of these phenotypes. However, although the effect of the crude supernatant on M-tropic HIV-1 infection in the experiments shown (Figs. 1,B, 2,C, 3, and 4,C) were inhibitory, the net effect depends on the balance of enhancing and suppressing factors as demonstrated by the subtraction experiments in which individual components of the crude supernatant were removed (Fig. 4 C). In fact, when comparing different donors as sources of CD4+ T cells, we have consistently noted marked enhancement of T-tropic HIV-1 infection. In contrast, the degree of inhibition of M-tropic HIV-1 infection varied greatly among donors, suggesting differences in susceptibility among the donors to the net balance of enhancing and inhibitory factors contained in the MT-2 supernatants.

Soluble Tax Protein Increases Fusogenicity of CD4+ T Cells with Envs from HIV-1.

The role of HTLV-I Tax protein in HIV-1 infection of CD4+ T cells was further investigated in two different systems. First, we purified Tax protein from E. coli transformed with a GST–Tax fusion protein expression vector (Fig. 5,A). When added to CD4+ T cell cultures, the GST–Tax protein was taken up by the cells and transferred to the nucleus (Fig. 5,B). The effect of Tax protein on HIV-1 infection was tested and compared with that of TCR signaling induced by anti-CD3 antibody. It can be postulated from previous studies that HTLV-I–infected T cells can secrete Tax protein in the concentration (20 ng/ml) used in this study, which is sufficient to produce biological activities such as cytokine production (3739). Stimulation of CD4+ T cells with purified Tax protein alone or anti-CD3 mAb alone minimally enhanced infectivity of HIV-1 in standard infection assays (data not shown) and single-round virus replication assays; however, these two stimuli in combination markedly enhanced infectivity (Fig. 5 C). Synergy between HTLV-I Tax protein and TCR signaling has also been reported for cytokine production by T cells (43). These results suggest that Tax protein produced by HTLV-I–infected cells is alone a relatively weak enhancer of HIV-1 infection of adjacent cells; however, it synergizes with other inducers of HIV-1 infection.

To determine whether Tax protein is able to influence viral fusion/entry, we performed cell–cell fusion assays. In this system, fusion efficiency between CD4+ T cells treated with purified Tax protein, anti-CD3 antibody, or both, and fusion partner cells expressing HIV-1 Env is measured by β-galactosidase activity in the cell lysates. Similar to the single-round virus replication assays (Fig. 5,C), Tax protein alone or anti-CD3 antibody alone modestly enhanced HIV-1 Env-mediated fusogenic activity of CD4+ T cells, whereas the combination of both markedly enhanced fusogenic activity (Fig. 5 D). These effects are specific to Tax protein in the preparation, since GST protein prepared in the same manner had no effect when used as control and the Tax preparation lost its activity after treatment with anti-Tax antiserum followed by protein A/G sepharose (data not shown). These results indicate that the effect of Tax protein on HIV-1 infection is mediated, at least in part, by increasing fusogenic activity.

Induction of Tax Protein in Jurkat Cells Increases Fusogenic Activity with HIV-1 Env.

The effect of Tax protein on HIV-1 infection was investigated in another system. Jurkat cells JPX9 and JPX/M were stably transfected with the wild-type and mutant form of HTLV-I Tax protein, respectively, and Tax expression was induced by stimulation with CdCl2 (Fig. 6 A). Jurkat cells express CXCR4 (a major coreceptor for T-tropic HIV-1), but not CCR5 (a major coreceptor for M-tropic HIV-1), as well as CD4 (a receptor for HIV-1); therefore, T-tropic Env-mediated fusogenic activity was tested in these cells in the presence (+Tax) or absence (−Tax) of CdCl2.

Induction of expression of wild-type Tax protein in JPX9 cells rendered these cells more fusogenic with cells expressing HIV-1 T-tropic IIIB Env, whereas expression of the mutant Tax protein did not increase fusogenicity of JPX/M cells (Fig. 6 B). The slight decrease in fusogenic activity of JPX/M cells after CdCl2 treatment is probably due to the toxic effect of Cd2+.

CC Chemokines in Combination with anti-CD3 Enhance Infection and Fusogenicity with Envs from T-tropic HIV-1.

The role of CC chemokines in infection of CD4+ T cells with T-tropic HIV-1 was further investigated by using recombinant CC chemokines. Although stimulation of the cells with anti-CD3 increased replication of T-tropic HIV-1 by 10-fold, CC chemokines alone had no or minimal effect on infectivity of T-tropic HIV-1. In contrast, costimulation of the cells with both anti-CD3 and CC chemokines further increased replication of T-tropic HIV-1 up to an additional threefold (Fig. 7 A; data not shown). We have obtained similar results using MIP-1α or MIP-1β (data not shown).

To determine whether CC chemokines are capable of increasing fusogenic activity of the cells with HIV-1 Envs, CD4+ T cells were either untreated or treated with anti-CD3 alone, CC chemokines alone, or both, and then tested for their fusogenicity with cells expressing HIV-1 Env. CC chemokines alone did not increase HIV-1 Env-mediated fusogenic activity, whereas anti-CD3 alone did so modestly. Combination of anti-CD3 and CC chemokines further increased fusogenicity (Fig. 7 B). Thus, upregulation of T-tropic HIV-1 infection by CC chemokines is mediated, at least in part, by enhancement of fusogenic activity of the cells with HIV-1 Envs.

HTVL-I Virions in Combination with Anti-CD3 Enhance HIV-1 Infection.

The role of HTLV-I virions in HIV-1 infection of CD4+ T cells was further investigated by using purified HTLV-I particles. Stimulation of the cells with purified HTLV-I particles (1 μg/ml of protein) alone modestly enhanced infectivity of HIV-1 bearing either T- or M-tropic Env in single-round virus replication assays, whereas costimulation of the cells with both purified HTLV-I particles and anti-CD3 enhanced the infectivity markedly (Fig. 8). These results confirmed a previous study demonstrating that HTLV-I virions are able to enhance HIV-1 replication in CD4+ T cells (25).

In this study we have demonstrated in several different systems that soluble factors from HTLV-I–infected cells are able to modulate HIV-1 infection of adjacent CD4+ T cells in a positive or negative fashion. As previously reported (41), the CC chemokines RANTES, MIP-1α, and MIP-1β produced by HTLV-I–transformed cells suppressed M-tropic HIV-1 infection; however, in our studies, these chemokines were also involved in upregulation of T-tropic HIV-1 replication. We have demonstrated that direct addition of CC chemokines, in combination with anti-CD3, to CD4+ T cells renders the cells more fusogenic with HIV-1 Envs. Since CC chemokines have been demonstrated to have a variety of effects on T cells (4450), any of those activities of the CC chemokines may be involved in the enhancement of T-tropic HIV-1 replication. We are currently investigating cellular and molecular mechanisms of the CC chemokine–mediated effect. We have also confirmed a previous study showing that HTLV-I virions are able to activate T cells and enhance T-tropic HIV-1 replication (25).

The mechanisms of Tax-mediated effects may be more complex. HTLV-I Tax protein has been shown to upregulate expression of HIV-1 (8), as well as expression of various cytokines and cytokine receptors involved in T cell activation (for review see references 51, 52), thereby providing favorable circumstances for HIV-1 infection. However, expression of anti-HIV CC chemokines is also induced by direct addition of soluble Tax protein (Moriuchi, H., M. Moriuchi, and A.S. Fauci, unpublished observations). Therefore, the net effect of Tax protein may depend upon the balance or accumulation of those effects. We have demonstrated that Tax protein is able to enhance HIV-1 fusion/entry. It is likely that Tax protein transactivates expression of cellular factors that are required for viral fusion/entry. We have recently cloned the promoter regions for CXCR4 (53) and CCR5 (54), and demonstrated that Tax is able to transactivate these promoters (Moriuchi, H., M. Moriuchi, and A.S. Fauci, unpublished observations). Thus, upregulation of coreceptor expression may be responsible, at least in part, for the effect of Tax on HIV-1 fusion/entry.

Our present study also indicates that the effect of HTLV-I coinfection on the pathogenesis of HIV disease is multifactorial, and that soluble factors produced by HTLV-I–infected cells are capable of enhancing and/or suppressing HIV-1 infection of adjacent cells, depending on the balance of effects of the factors involved and the tropism of the virus. Although an increase in replication of T- and dual-tropic HIV-1 is consistently seen, the balance of enhancing and suppressing factors determines the net effect on M-tropic HIV-1 infection. Discrepancy among previous studies (1012, 1417) on the effect of HTLV-I/HIV coinfection on HIV disease progression may reflect these potentially dichotomous effects on M-tropic HIV-1. In this regard, infection with HTLV-I may favor the transition from M- to T-tropic phenotype, which is associated with HIV disease progression (3, 57–59).

In conclusion, this study provides possible mechanisms whereby coinfection of an individual with HIV-1 and HTLV-I influences the course of HIV-1 infection without necessity for actual coinfection of the same cells by the two pathogens. Further studies are required to establish the actual effects of HTLV-I coinfection on the clinical progression of HIV infection in vivo.

We thank K.T. Jeang, K. Sugamura, G. Franchini, M. Martin, R. Collman, T. Theodore, N. Landau, J. Sodroski, and E. Berger for providing reagents; J. Weddle for graphic work; and P. Walsh for editorial assistance.

GST

glutathione S-transferase

HCMV

human cytomegalovirus

HTLV-I

human T lymphotropic virus type I

MIP

macrophage inflammatory protein

MOI

multiplicity of infection

RANTES

regulated on activation, normal T cell expressed and secreted

RT

reverse transcriptase

rVV

recombinant vaccinia virus

1
Connor
RI
,
Ho
DD
Human immunodeficiency virus type 1 variants with increased replicative capacity develop during the asymptomatic stage before disease progression
J Virol
1994
68
4400
4408
[PubMed]
2
Connor
RI
,
Paxton
WA
,
Sheridan
KE
,
Koup
RA
Macrophages and CD4+ lymphocytes from two multiply exposed, uninfected individuals resist infection with primary nonsyncytium-inducing isolates of human immunodeficiency virus type 1
J Virol
1996
70
8758
8764
[PubMed]
3
Connor
RI
,
Sheridan
KE
,
Ceradini
C
,
Choe
S
,
Landau
NR
Change in coreceptor use correlates with disease progression in HIV-1 infected individuals
J Exp Med
1997
185
621
628
[PubMed]
4
Zhu
T
,
Mo
H
,
Wang
N
,
Nam
DS
,
Cao
Y
,
Koup
RA
,
Ho
DD
Genotypic and phenotypic characterization of HIV-1 patients with primary infection
Science
1993
261
1179
1181
[PubMed]
5
Gendelman
HE
,
Phelps
W
,
Feigenbaum
L
,
Ostrove
JM
,
Adachi
A
,
Howley
PM
,
Khoury
G
,
Ginsberg
HS
,
Martin
MA
Transactivation of the human immunodeficiency virus long terminal repeat sequence by DNA viruses
Proc Natl Acad Sci USA
1986
83
9759
9763
[PubMed]
6
Scala
G
,
Quinto
I
,
Ruocco
MR
,
Mallardo
M
,
Ambrosino
C
,
Squitieri
B
,
Tassone
P
,
Venuta
S
Epstein-Barr virus nuclear antigen 2 transactivates the long terminal repeat of human immunodeficiency virus type 1
J Virol
1993
67
2853
2861
[PubMed]
7
Siekevitz
M
,
Josephs
SF
,
Dukovich
M
,
Peffer
N
,
Wong-Staal
F
,
Greene
WC
Activation of the HIV-1 LTR by T cell mitogens and the trans-activator protein of HTLV-I
Science
1987
238
1575
1578
[PubMed]
8
Boehnlein
E
,
Siekevitz
M
,
Ballard
DW
,
Lowenthal
JW
,
Rimsky
L
,
Bogerd
H
,
Hoffman
J
,
Wano
Y
,
Franza
BR
,
Greene
WC
Stimulation of the HIV-1 enhancer by the HTLV-I taxgene product involves the action of inducible cellular proteins
J Virol
1988
63
1578
1586
9
Pleskoff
O
,
Treboute
C
,
Brelot
A
,
Heveker
N
,
Seman
M
,
Alizon
M
Identification of a chemokine receptor encoded by human cytomegalovirus as a cofactor for HIV-1 entry
Science
1997
276
1874
1878
[PubMed]
10
Bartholomew
C
,
Blattner
WA
,
Cleghorn
F
Progression to AIDS in homosexual men co-infected with HIV and HTLV-I in Trinidad
Lancet
1987
2
1469
[PubMed]
11
Page
JB
,
Lai
SH
,
Chitwood
DD
,
Klimas
NG
,
Smith
PC
,
Fletcher
MA
HTLV-I/II positivity and death from AIDS among HIV-1 seropositive intravenous drug users
Lancet
1990
335
1439
1441
[PubMed]
12
Pagliuca
A
,
Mufti
GJ
Co-infection with HTLV-I/II and HIV-1
Lancet
1990
336
383
13
Cleghorn
FR
,
Blattner
WA
Does human T cell lymphotropic virus type I and human immunodeficiency virus type 1 coinfection accelerate acquired immunodeficiency syndrome?
Arch Intern Med
1992
152
1372
1373
[PubMed]
14
Gotuzzo
E
,
Escamilla
J
,
Phillips
IA
,
Sanchez
J
,
Wignall
FS
,
Antigoni
J
The impact of human T-lymphotropic virus type I/II infection on the prognosis of sexually acquired cases of acquired immunodeficiency syndrome
Arch Intern Med
1992
152
1429
1432
[PubMed]
15
Schechter
M
,
Harrison
LH
,
Halsey
NA
,
Trade
G
,
Santino
M
,
Moulton
LH
,
Quinn
TC
Coinfection with human T-cell lymphotropic virus type I and HIV in Brazil. Impact on markers of HIV disease progression
JAMA (J Am Med Assoc)
1994
271
353
357
[PubMed]
16
Fantry
L
,
Dodging
E
,
Auwaerter
PG
,
Lederman
HM
Immunodeficiency and elevated CD4 lymphocyte counts in two patients coinfected with human immunodeficiency virus and human lymphotropic virus type I
Clin Infect Dis
1995
21
1446
1448
17
Harrison
LH
,
Quinn
TC
,
Schechter
M
Human T cell lymphotropic virus type I does not increase human immunodeficiency virus viral load in vivo
J Infect Dis
1997
175
438
440
[PubMed]
18
Harada
S
,
Koyanagi
Y
,
Yamamoto
N
Infection of HTLV-III/LAV in HTLV-I–carrying cells
Science
1985
229
563
566
[PubMed]
19
Popovic
M
,
Lange-Wantzin
G
,
Sarin
PS
,
Mann
D
,
Gallo
RC
Transformation of human umbilical cord blood T cells by human T cell leukemia/lymphoma virus
Proc Natl Acad Sci USA
1983
80
5402
5406
[PubMed]
20
Nagata
K
,
Ohtani
K
,
Nakamura
M
,
Sugamura
K
Activation of endogenous c-fos proto-oncogene expression by human T-cell leukemia virus type I–encoded p40taxprotein in the human T-cell line, Jurkat
J Virol
1989
68
3220
3226
[PubMed]
21
Ohtani
K
,
Nakamura
M
,
Saito
S
,
Nagata
K
,
Sugamura
K
,
Hinuma
Y
Electroporation: application to human lymphoid cell lines for stable introduction of a transactivator gene of human T-cell leukemic virus type I
Nucleic Acids Res
1989
17
1589
1604
[PubMed]
22
Moriuchi
H
,
Moriuchi
M
,
Combadiere
C
,
Murphy
PM
,
Fauci
AS
CD8+T-cell–derived factor(s), but not β-chemokines RANTES, MIP-1α, and MIP-1β, suppress HIV-1 replication in monocyte/macrophages
Proc Natl Acad Sci USA
1996
93
15341
15345
[PubMed]
23
Jeang
K-T
,
Widen
SG
,
Semmes
OJ
,
Wilson
SH
HTLV-I trans-activator protein, Tax, is a trans-repressor of the human β-polymerase gene
Science
1990
247
1082
1084
[PubMed]
24
Moriuchi
H
,
Moriuchi
M
,
Cohen
JI
Proteins and cisacting elements associated with transactivation of the varicella-zoster virus (VZV) immediate-early gene 62 promoter by VZV open reading frame 10 protein
J Virol
1995
69
4693
4701
[PubMed]
25
Zack
JA
,
Cann
AJ
,
Lugo
JP
,
Chen
ISY
HIV-1 production from infected peripheral blood T cells after HTLV-I induced mitogenic stimulation
Science
1988
240
1026
1029
[PubMed]
26
Adachi
A
,
Gendelman
HE
,
Koenig
S
,
Folks
T
,
Willey
R
,
Rabson
A
,
Martin
MA
Production of acquired immunodeficiency syndrome–associated retrovirus in human and non-human cells transfected with an infectious molecular clone
J Virol
1986
59
284
291
[PubMed]
27
Collman
R
,
Balliet
JW
,
Gregory
SA
,
Friedman
H
,
Kolson
DL
,
Nathanson
N
,
Srinivasan
A
An infectious molecular clone of unusual macrophage-tropic and highly cytopathic strain of human immunodeficiency virus type 1
J Virol
1992
66
7517
7521
[PubMed]
28
Theodore
TS
,
Englund
G
,
Buckler-White
A
,
Buckler
CE
,
Martin
MA
,
Peden
KW
Construction and characterization of a stable full-length macrophage-tropic HIV type 1 molecular clone that directs the production of high titers of progeny virions
AIDS Res Hum Retroviruses
1996
12
191
194
[PubMed]
29
Poli
G
,
Kinter
AL
,
Fauci
AS
Interleukin 1 induces expression of the human immunodeficiency virus alone and in synergy with interleukin 6 in chronically infected U1 cells: inhibition of inductive effects by the interleukin 1 receptor antagonist
Proc Natl Acad Sci USA
1994
91
108
112
[PubMed]
30
Choe
H
,
Farzan
M
,
Sun
Y
,
Sullivan
N
,
Rollins
B
,
Ponath
PD
,
Wu
L
,
Mackay
CR
,
LaRosa
G
,
Newman
W
et al
The β-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates
Cell
1996
85
1135
1148
[PubMed]
31
Deng
H
,
Liu
R
,
Ellmeier
W
,
Choe
S
,
Unutmaz
D
,
Burkhart
M
,
Di Marzio
P
,
Marmon
S
,
Sutton
RE
,
Hill
CM
et al
Identification of a major co-receptor for primary isolates of HIV-1
Nature
1996
381
661
666
[PubMed]
32
Feng
Y
,
Broder
CC
,
Kennedy
PE
,
Berger
EA
HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein–coupled receptor
Science
1996
272
872
877
[PubMed]
33
Moriuchi
H
,
Moriuchi
M
,
Arthos
J
,
Hoxie
J
,
Fauci
AS
Promonocytic U937 clones expressing CD4 and CXCR4 are resistant to infection with and cell-to-cell fusion with T-tropic HIV-1
J Virol
1997
71
9664
9671
[PubMed]
34
Chosa
TN
,
Yamamoto
Y
,
Tanaka
Y
,
Koyanagi
Y
,
Hinuma
Y
Infectivity dissociated from transforming activity in a human retrovirus adult T-cell leukemia virus
Gann
1982
73
844
847
[PubMed]
35
Miyoshi
I
,
Taguchi
H
,
Fujishita
M
,
Yoshimoto
S
,
Kubonishi
I
,
Ohtsuki
Y
,
Shiraishi
Y
,
Akagi
T
Transformation of monkey lymphocytes with adult T-cell leukemia virus
Lancet
1982
2
658
[PubMed]
36
Clapham
P
,
Nagy
K
,
Cheingsong-Popov
R
,
Weiss
RA
Productive infection and cell-free transmission of human T-cell leukemia virus in a nonlymphoid cell line
Science
1983
222
1125
1127
[PubMed]
37
Lindholm
PF
,
Marriott
SJ
,
Gitlin
SD
,
Bohan
CA
,
Brady
JN
Induction of nuclear NF-κB DNA binding activity after exposure of lymphoid cells to soluble Tax1protein
New Biol
1990
2
1034
1043
[PubMed]
38
Marriott
SJ
,
Lindholm
PF
,
Reid
RL
,
Brady
JN
Soluble HTLV-I Tax1protein stimulates proliferation of human peripheral blood lymphocytes
New Biol
1991
3
678
686
[PubMed]
39
Dhib-Jalbut
S
,
Hoffman
PM
,
Yamabe
T
,
Sun
D
,
Xia
J
,
Eisenberg
H
,
Berger
G
,
Ruscetti
FW
Extracellular human T-cell lymphotropic virus type I Tax protein induces cytokine production in adult human microglial cells
Ann Neurol
1994
36
787
790
[PubMed]
40
Hollsberg
P
,
Hafler
DA
Pathogenesis of diseases induced by human lymphotropic virus type I infection
N Engl J Med
1993
328
1173
1182
[PubMed]
41
Cocchi
F
,
DeVico
AL
,
Garzino-Demo
A
,
Arya
SK
,
Gallo
RC
,
Lusso
P
Identification of RANTES, MIP-1α, and MIP-1β as the major HIV-suppressive factors produced by CD8+T cells
Science
1995
270
1811
1815
[PubMed]
42
Baba
M
,
Imai
T
,
Yoshida
T
,
Yoshie
O
Constitutive expression of various chemokine genes in human T-cell lines infected with human T-cell leukemia virus type I: role of the viral transactivator Tax
Int J Cancer
1996
66
124
129
[PubMed]
43
Himes
SR
,
Katsikeros
R
,
Shannon
MF
Costimulation of cytokine gene expression in T cells by the human T leukemia/lymphotropic virus type I transactivator Tax
J Virol
1996
70
4001
4008
[PubMed]
44
Bacon
KB
,
Premack
BA
,
Gardner
P
,
Schall
TJ
Activation of dual T cell signaling pathways by the chemokine RANTES
Science
1995
269
1727
1730
[PubMed]
45
Taub
DD
,
Ortaldo
JR
,
Turcovski-Corrales
SM
,
Key
ML
,
Longo
DL
,
Murphy
WJ
β chemokines costimulate lymphocyte cytolysis, proliferation, and lyphokine production
J Leukocyte Biol
1996
59
81
89
[PubMed]
46
Taub
DD
,
Sayers
TJ
,
Carter
CRD
,
Ortaldo
JR
α and β chemokines induce NK cell migration and enhance NK-mediated cytolysis
J Immunol
1995
155
3877
3888
[PubMed]
47
Taub
DD
,
Turcovski-Corrales
SM
,
Key
ML
,
Longo
DL
,
Murphy
WJ
Chemokines and T lymphocyte activation. I. β chemokines costimulate human T lymphocyte activation in vitro
J Immunol
1996
156
2095
2103
[PubMed]
48
Turner
L
,
Ward
SG
,
Westwick
J
RANTES-activated human T lymphocytes. A role for phosphoinositide 3-kinase
J Immunol
1995
155
2437
2444
[PubMed]
49
Szabo
MC
,
Butcher
EC
,
McIntyre
BW
,
Schall
TJ
,
Bacon
KB
RANTES stimulation of T lymphocyte adhesion and activation: role for LFA-1 and ICAM-3
Eur J Immunol
1997
27
1061
1068
[PubMed]
50
Springer
T
Traffic signals for lymphocyte recirculation and leukocyte emigration: multistep paradigm
Cell
1994
76
301
314
[PubMed]
51
Smith
MR
,
Greene
WC
Molecular biology of the type I human T-cell leukemia virus (HTLV-I) and adult T-cell leukemia
J Clin Invest
1991
87
761
766
[PubMed]
52
Franchini
G
Molecular mechanisms of human T-cell leukemia/lymphotropic virus type I infection
Blood
1995
86
3619
3639
[PubMed]
53
Moriuchi
H
,
Moriuchi
M
,
Fauci
AS
Cloning and analysis of the promoter region for CCR5, a co-receptor for HIV-1 entry
J Immunol
1997
159
5441
5449
[PubMed]
54
Moriuchi
M
,
Moriuchi
H
,
Turner
W
,
Fauci
AS
Cloning and analysis of the promoter region for CXCR4, a co-receptor for HIV-1 entry
J Immunol
1997
159
4322
4329
[PubMed]
55
Chang-Mayer
C
,
Seto
D
,
Tateno
M
,
Levy
JA
Biological features of HIV-1 that correlate with virulence in the host
Science
1988
240
80
82
[PubMed]
56
Schuitemaker
H
,
Koot
M
,
Kootstra
NA
,
Dercksen
MW
,
de Goede
RE
,
van Steenwijk
RP
,
Lange
JM
,
Schattenkerk
JK
,
Miedema
F
,
Tersmette
M
Biological phenotype of human immunodeficiency virus type 1 clones at different stages of infection: progression of disease is associated with a shift from monocytotropic to T-cell–tropic virus populations
J Virol
1992
66
1354
1360
[PubMed]
57
Tersmette
M
,
de Goede
RE
,
Al
BJ
,
Winkel
IN
,
Gruters
RA
,
Cuypers
HT
,
Huisman
HG
,
Miedema
F
Differential syncytium-inducing capacity of human immunodeficiency virus isolates: frequent detection of syncytium-inducing isolates in patients with acquired immunodeficiency syndrome (AIDS) and AIDS-related complex
J Virol
1988
62
2026
2032
[PubMed]

H. Moriuchi and M. Moriuchi contributed equally to this project, which M. Moriuchi performed for the partial fulfillment of the requirements of the PhD program of the Department of Microbiology at Howard University, Washington, DC.

Author notes

Address correspondence to Hiroyuki Moriuchi, Laboratory of Immunoregulation, NIAID, NIH, Bldg. 10, Rm. 6A11, Bethesda, MD 20892. Phone: 301-402-2617; Fax: 301-402-4122; E-mail: hmoriuchi@atlas.niaid.nih.gov