Urokinase-type plasminogen activator (uPA) signaling via its receptor uPAR inhibits late events in HIV-1 replication in acutely infected primary monocyte-derived macrophages (MDMs) and promonocytic U937 cells. Here we show that U937-derived, chronically infected U1 cells stimulated with phorbol 12-myristate 13-acetate (PMA) express integrins, uPA, and soluble uPAR at levels similar to those of MDMs. uPA inhibited HIV expression in U1 cells incubated with either PMA or tumor necrosis factor-α (TNF-α), but not with other HIV-inductive cytokines or lipopolysaccharide. Of interest, only PMA and TNF-α, but not other HIV-inductive stimuli, induced surface expression of the αM chain CD11b in U1 cells constitutively expressing CD18, the β2 chain of the Mac-1 integrin. Like uPA, fibrinogen, a Mac-1 (CD11b/CD18) ligand, and M25, a peptide homologous to a portion of the β-propeller region of CD11b preventing its association with uPAR, inhibited HIV virion release in PMA-stimulated U1 cells. Both uPAR small-interference RNA (siRNA) and soluble anti-β1/-β2 monoclonal antibodies abolished the anti-HIV effects of uPA, whereas CD11b siRNA reversed the anti-HIV effect of M25, but not that induced by uPA. Thus, either uPA/uPAR interaction, Mac-1 activation, or prevention of its association with uPAR triggers a signaling pathway leading to the inefficient release of HIV from monocytic cells.

Urokinase-type plasminogen activator (uPA), a serine protease that activates plasminogen to plasmin,1  is synthesized as an inactive precursor (pro-uPA) that undergoes a rapid proteolytic activation. uPA binds to a specific glycosyl-phospatidyl-inositol (GPI)–anchored receptor, uPAR, localized at the cell surface.2  Both uPA and uPAR are expressed by inflammatory cells, including neutrophils, monocytes, macrophages, and activated T lymphocytes,2  in which they play important roles in cell activation, adhesion, and migration.3,4  In addition to localizing the enzymatic activity of uPA on the leading edge of migrating cells, uPAR mediates signaling by uPA.5  The binding of uPA to uPAR induces migration, adhesion, and proliferation of different cell types, independent of the catalytic activity of uPA.6,7  As a GPI receptor lacking an intracellular domain, uPAR requires the interaction with transduction-competent receptors, such as the G-protein–coupled receptor formyl peptide receptor-like–1 (FPRL1),5  the gp130 signal–transducing chain of the interleukin-6 (IL-6) receptor family,8  or integrins such as α5β1 in epidermal cancer cells and CD11b/CD18 (Mac-1) in monocyte-macrophages.5,6,9,10 

High serum and cerebrospinal spinal fluid levels of soluble uPAR (suPAR) have been correlated with the severity of HIV-1 disease independent of CD4+ T-cell counts or viremia levels.11-14  Furthermore, uPA expression has been observed in the brains of HIV+ individuals whose brains stained negatively for both HIV-1 p24 Gag antigen and uPAR,15  suggesting a potential role of uPA as a negative regulator of HIV-1 expression. In vitro, uPA inhibits HIV-1 replication in lymphoid histocultures, primary monocyte-derived macrophages (MDM), promonocytic U937 cells acutely infected with HIV, and chronically infected promonocytic U1 cells stimulated with the differentiating agent phorbol 12-myristate 13-acetate (PMA) or tumor necrosis factor-α (TNF-α).16,17  In particular, uPA was shown to promote the sequestration of HIV particles in cytoplasmic vacuoles, likely belonging to multivescicular bodies,18-20  an effect that was fully accounted for by the signaling-competent amino-terminal fragment (ATF) of uPA.5  More recently, we showed that vitronectin (VN)–dependent cell adhesion is crucial for uPA-mediated inhibition of virus replication in MDM and in PMA-stimulated U1 cells.17  A better definition of this signaling pathway and of its determinants may be relevant for understanding the dynamics of tissue seeding by infected leukocytes that may affect their ability or efficiency to establish HIV reservoirs in sanctuary sites21  and cause organ/tissue-specific pathology, such as HIV-associated dementia, interstitial lung disease, nephropathy, enteropathy, and wasting syndrome.22,23 

In the present study we investigated which among the known uPAR-associated signaling-competent receptors mediate its inhibitory signal on HIV-1 expression in monocytic cells. Our findings indicate that such an inhibitory effect is mediated by β1 and/or β2 integrins, but does not require the expression of CD11b. In addition, we demonstrated that stimulation of the Mac-1 integrin by fibrinogen (FNG) or prevention of the association between CD11b and uPAR fully mimicked uPA/uPAR-dependent inhibition of late events in virus expression. These findings reinforce the hypothesis of a common pathway controlling the late phase of HIV assembly and release from infected monocytic cells.

Reagents

Lipopolysaccharide (LPS)–free (< 2 × 10−5 EU/IU, corresponding to < 10−10 EU/mg) human pro-uPA (52 kDa) was provided by Dr Jack Henkin (Abbott Laboratories, Abbott Park, IL). The ATF peptide was purchased from American Diagnostica (Stamford, CT). Pro-uPA and ATF were used at 10 nM. FNG, phosphatidylinositol-specific phospholipase C (PIPLC) from Bacillus cereus, crystal violet, goat anti–mouse fluorescein isothiocyanate (FITC) antibody (Ab), rabbit anti–goat FITC Ab, and PMA were purchased from Sigma-Aldrich (St Louis, MO). Interleukin-6 (IL-6) and interferon-γ (IFN-γ) were purchased from R&D Systems (Minneapolis, MN). PMA, IL-6, and IFN-γ were resuspended as recommended by the manufacturers and used at final concentrations of 6, 10, and 50 ng/mL, respectively, based on previous studies. PIPLC was resuspended at 20 U/mL in culture medium and used at the final concentration of 10 U/mL. M25 (PRYQHIGLVAMFRQNTG) and its scrambled peptide (scM25, HQIPGAYRGVNQRFTML) were purchased from PRIMM (Milan, Italy) and dissolved at 20 mM in dimethylsulfoxide (DMSO) and used at the indicated concentrations. A list of antibodies used and their sources is provided in Table S1 (available on the Blood website; see the Supplemental Materials link at the top of the online article). All experiments were performed in 96-well microtiter plates (Falcon; BD Biosciences, Bedford, MA) unless otherwise specified.

Quantification of uPA and suPAR secretion

Both molecules were measured in culture supernatants of U1 cells and MDM. uPA concentrations were measured by a commercial kit (IMUBIND uPA ELISA kit no. 894; American Diagnostica, Greenwich, CT) with a lower detection limit of 10 pg/mL. Concentrations of suPAR and the uPA-suPAR complex were determined using an in-house sandwich ELISA. In this assay, suPAR is captured using a monoclonal Ab (mAb) as capture Ab and the associated uPA is detected using a polyclonal Ab directed against uPA. This assay does not detect suPAR and uPA alone but displays a linear dose-response to complexes between suPAR and uPA, as previously described.14,15 

Chronically HIV-infected U1 cell line

The promonocytic U1 cell line contains 2 copies of integrated X4 HIV-1LAI/IIIB provirus per cell, and it is characterized by a constitutive state of relative viral latency.24  High levels of virus expression are rapidly induced by stimulation of U1 cells with either PMA or cytokines.25,26  U1 cells were stimulated at the concentration of 2 × 105 cells/mL in RPMI 1640 containing 10% of heat-inactivated fetal bovine serum (FBS), and were first incubated with either uPA or peptides 30 minutes before the addition of PMA or HIV-inductive stimuli.

U1 cell adhesion assay

U1 cells were resuspended in culture medium containing 10% FBS and left either unstimulated or stimulated and seeded in triplicates into 96-well tissue-culture plates. At the indicated time points, cells were washed with warm culture medium, fixed for 15 minutes with 3% paraphormaldehyde in phosphate-buffered saline (PBS), stained for 10 minutes with 0.5% crystal violet (in 20% methanol/80% water), washed 3 times and lysed with 100 μL 1% sodium dodecyl sulfate (SDS) in water. The results were obtained by measuring the absorbance at 570 nm.

Acute CCR5-dependent infection of primary human monocyte-derived macrophages

Circulating monocytes were isolated from Ficoll-Hypaque purified peripheral blood mononuclear cells (PBMC) of several independent, healthy, HIV-1–seronegative donors by iso-osmotic Percoll gradient.27  Their purity was approximately 90% based on morphology and CD14 staining (data not shown). Monocytes were allowed to differentiate for 7 days in culture in Dulbecco modified eagle medium (DMEM) enriched with 10% FBS and 10% human AB serum before infection.17  Thus, human MDMs were treated for 30 minutes with uPA or ATF and then infected with the CCR5-dependent (R5) laboratory-adapted HIV-1BaL strain at the multiplicity of infection (MOI) of 0.1.17  MDM cultures were carried on for 3 to 4 weeks after infection and supernatants were collected every 3 to 4 days and stored at −20°C for determination of their reverse transcriptase (RT) activity content. Collected culture supernatants were replenished with fresh culture medium, with uPA or ATF for treated cultures.

Acute X4 HIV-1 infection of the human promonocytic U937 cell line

U937 cells were resuspended in RPMI 1640 plus 10% FBS and infected with the laboratory-adapted X4 HIV-1LAI/IIIB strain at the MOI of 1 after 30 minutes' preincubation with FNG. Cell cultures were carried on for 3 to 4 weeks after infection and supernatants were collected every 2 to 3 days and stored at −20°C for determination of their RT activity content. Collected culture supernatants were replenished with fresh culture medium, with FNG for treated cultures.

HIV-1 quantification by RT activity assay

HIV-1 expression was monitored by determination of Mg++-dependent RT activity in culture supernatants, reflecting the production of new progeny virions.28  Release of cell-associated virions by 5 consecutive cycles of cell freezing and thawing was performed as previously described.16 

Cytofluorimetric analysis

U1 cells were washed twice in 2% FBS/PBS and stained by using the indicated Ab at 1 μg/105 cells unless otherwise specified. After 30 minutes of incubation on ice, U1 cells were centrifuged twice in 2% FBS/PBS at 390g for 5 minutes at +4°C; the cells were then incubated with 1 μg of the secondary anti–mouse-FITC or anti–rabbit-FITC Ab. The cells were spun again after 30 minutes of incubation on ice and resuspended in 2% formaldehyde/PBS. Fifteen thousand cells were acquired using a FACScan (Becton Dickinson, Franklin Lakes, NJ) flow cytometry apparatus and analyzed by CellQuest software (Becton Dickinson).

Cell proliferation

Cell proliferation was assessed by the uptake of [3H]-thymidine. [3H]-thymidine (3.7 × 107 Bq) was added to 4 × 104 cells in 200 μL medium and incubated 16 hours at 37°C, 5% CO2. Cells were harvested and the β emission was counted in a 1450 β-counter (1450 Microbeta Plus; PerkinElmer Life and Analytical Sciences, Waltham, MA).

siRNA transient transfection

Three pairs of validated stealth small-interference RNA (siRNA; 25 nt) for each target antigen were purchased from Invitrogen (Paisley, United Kingdom) and resuspended at the final concentration of 20 μM in diethyl pyrocarbonate (DEPC)–treated water, as detailed in Table S1. Typically, 2 × 106 cells were transfected with 1 μM siRNA, then resuspended at 106 cells/mL and finally diluted 4 hours later at 2 × 105 cells/mL before incubation with uPA and stimuli. Cells transiently transfected with irrelevant stealth siRNA duplex containing 45% to 55% GC (Mock siRNA; Invitrogen) were used as negative controls.

Statistical analysis

Results are reported as mean values plus or minus standard deviation (SD), and the analysis of variance was performed by a 2-tailed Student t test to calculate P values for paired observations (unless otherwise specified). The observed differences were considered significant if the P value obtained was below .05.

The anti-HIV activity of uPA depends on its ATF–receptor- binding domain and the cell-surface expression of uPAR

Exogenously added pro-uPA is immediately cleaved in vitro into its active form, uPA, by the plasmin present in the cell-culture serum-enriched medium.29  The anti-HIV effect of uPA on stimulated U1 cells was already shown to be dependent on its receptor-interacting component, ATF, and not on its catalytic activity.16,17  This finding was confirmed here, in that only uPA peptides maintaining an ATF growth factor-like domain,30  including the so-called Omega loop,30  but not the enzymatically competent low molecular weight fragment, inhibited HIV expression in chronically infected U1 cells stimulated with either PMA (Figure 1A) or TNF-α (data not shown).

Figure 1

UPA inhibits HIV expression in chronically infected U1 cells via binding of its ATF domain to uPAR. (A) U1 cells were preincubated with different concentrations of either uPA or peptides representing the low molecular weight (LMW), ATF, growth factor domain (GFD), or omega loop components of uPA. Cells were then stimulated with PMA and followed for virus expression in culture supernatants. All peptides significantly inhibited (P < .001) virus expression in the same range of concentrations, except for LMW, which was ineffective at all tested concentrations. (B) U1 cells were analyzed for uPAR expression 2 and 24 hours after incubation with PIPLC (10 U/mL). Down-modulation of uPAR expression was clearly detectable at both time points. Geometric mean fluorescence intensities (MFI) are shown: isotype, MFI: 4; Nil, unstimulated cells, MFI: 31; PIPLC, MFI: 14. (C) U1 cells were incubated with PIPLC for 2 hours and then stimulated with PMA in the presence or absence of uPA (10 nM). The RT activity levels were measured in the culture supernatants at the peak of virus expression (day 4 after stimulation). PIPLC abolished the inhibitory effect of uPA on virus expression without affecting the inductive effect of PMA (*P < .001). (D) U1 cells were stimulated with PMA in the presence or absence of suPAR and uPA, and RT activity was determined in the culture supernatants after 3 days of culture (*P < .001);a similar pattern of virus expression was observed even at days 2 and 4 of culture (data not shown). The error bars indicate the SD of duplicate samples. All the described experiments were performed in duplicate wells and repeated 3 times, and provided identical results. cpm indicates counts per minute.

Figure 1

UPA inhibits HIV expression in chronically infected U1 cells via binding of its ATF domain to uPAR. (A) U1 cells were preincubated with different concentrations of either uPA or peptides representing the low molecular weight (LMW), ATF, growth factor domain (GFD), or omega loop components of uPA. Cells were then stimulated with PMA and followed for virus expression in culture supernatants. All peptides significantly inhibited (P < .001) virus expression in the same range of concentrations, except for LMW, which was ineffective at all tested concentrations. (B) U1 cells were analyzed for uPAR expression 2 and 24 hours after incubation with PIPLC (10 U/mL). Down-modulation of uPAR expression was clearly detectable at both time points. Geometric mean fluorescence intensities (MFI) are shown: isotype, MFI: 4; Nil, unstimulated cells, MFI: 31; PIPLC, MFI: 14. (C) U1 cells were incubated with PIPLC for 2 hours and then stimulated with PMA in the presence or absence of uPA (10 nM). The RT activity levels were measured in the culture supernatants at the peak of virus expression (day 4 after stimulation). PIPLC abolished the inhibitory effect of uPA on virus expression without affecting the inductive effect of PMA (*P < .001). (D) U1 cells were stimulated with PMA in the presence or absence of suPAR and uPA, and RT activity was determined in the culture supernatants after 3 days of culture (*P < .001);a similar pattern of virus expression was observed even at days 2 and 4 of culture (data not shown). The error bars indicate the SD of duplicate samples. All the described experiments were performed in duplicate wells and repeated 3 times, and provided identical results. cpm indicates counts per minute.

Close modal

To demonstrate the essential role of the GPI-anchored protein uPAR31  in uPA-mediated HIV inhibitory signals, U1 cells were incubated with PIPLC from B cereus that specifically removes GPI anchors.32  PIPLC indeed reduced the expression of uPAR on U1 cell surfaces 2 to 24 hours after incubation, as measured by cytofluorimetric analysis (Figure 1B). Concomitantly, PIPLC promoted the release of suPAR in the culture supernatant, at concentrations from 33 pM (Table 1) to 220 pM (not shown), while it did not modulate the cell-surface expression of non–GPI-anchored receptors including integrins, as measured by FACS analysis (data not shown). PIPLC did not interfere with HIV expression in either unstimulated or PMA-stimulated U1 cells (Figure 1C), suggesting that neither uPAR per se nor endogenous uPA play a significant role in HIV-1 expression in U1 cells. In contrast, PIPLC abrogated the anti-HIV activity of exogenous uPA (Figure 1C) without any cytotoxic or cytostatic effects, as tested by [3H]-thymidine uptake (data not shown).

Table 1

Concentrations of uPA, suPAR, and the uPA-suPAR complex in culture supernatants of U1 cells and of primary MDMs acutely infected or not infected with HIV-1

Cell type/ Stimulus/infectionRT activity, cpm/μLsuPAR, pMuPA, pMsuPAR/uPA ratiosuPAR-uPA complex, pM
U1 
    Nil < 100 33 ± 0 2.9 ± 0.6 11.38 2.9 ± 0.5 
    PMA 1657 ± 221† 49 ± 1‡ 2.5 ± 1 19.6§ 2.4 ± 0.4 
    TNF-α 2021 ± 310† 34 ± 1 4.6 ± 1 7.4‖ 4.5 ± 0.3‖ 
    IL-6 2421 ± 212† 33 ± 1 2.7 ± 0.6 12.2 2.6 ± 0.2 
MDM 
    Control* NA 217 ± 102 8 ± 2 27.2 7 ± 1 
    HIV 6967 ± 882† 182 ± 93 8 ± 2 22.8 7 ± 1 
Cell type/ Stimulus/infectionRT activity, cpm/μLsuPAR, pMuPA, pMsuPAR/uPA ratiosuPAR-uPA complex, pM
U1 
    Nil < 100 33 ± 0 2.9 ± 0.6 11.38 2.9 ± 0.5 
    PMA 1657 ± 221† 49 ± 1‡ 2.5 ± 1 19.6§ 2.4 ± 0.4 
    TNF-α 2021 ± 310† 34 ± 1 4.6 ± 1 7.4‖ 4.5 ± 0.3‖ 
    IL-6 2421 ± 212† 33 ± 1 2.7 ± 0.6 12.2 2.6 ± 0.2 
MDM 
    Control* NA 217 ± 102 8 ± 2 27.2 7 ± 1 
    HIV 6967 ± 882† 182 ± 93 8 ± 2 22.8 7 ± 1 

The levels of expression of the analytes were measured 48 hours after stimulation of U1 cells and at the peak of viral replication in MDMs. Because MDMs were seeded at twice the concentration of U1 cells, the results shown have been normalized by dividing by a factor of 2. Results are expressed as mean plus or minus SD of the concentrations determined from 3 independent experiments with U1 cells and from MDM cultures from 5 independent donors (Figure 2).

Significantly higher levels of HIV production were observed in PMA or cytokine-stimulated U1 cells versus unstimulated cells (Nil; †P = .001), as well as in infected versus uninfected MDMs (†P = .001). Furthermore, increased levels of suPAR were observed after U1 cell stimulation with PMA, but not with cytokines (‡P = .001 vs Nil). There were also increases in the suPAR/uPA ratio in PMA and TNF-α–stimulated U1 cells (§P = .0045 and ‖P = .01, respectively) and the suPAR-uPA concentrations in cells stimulated with TNF-α (‖P = .01).

NA indicates not applicable.

*

Control: uninfected MDM.

We next determined the concentrations of uPA, suPAR, and of their complex in culture supernatants of both unstimulated and stimulated U1 cells (Table 1). Most of the uPA released from stimulated U1 cells was indeed complexed with suPAR (Table 1), as previously reported in vivo.15  Addition of suPAR did not alter HIV expression per se from either unstimulated or PMA-stimulated U1 cells (Figure 1D). Furthermore, suPAR did not interfere with HIV-1 replication in acutely infected U937 cells (data not shown, n = 3) reinforcing the concept that its role in virus production is strictly related to mediating uPA-dependent cell signaling. In contrast, a 20:1 molar excess of suPAR to uPA abrogated the anti-HIV activity of uPA (Figure 1D), whereas, vice versa, a 5:1 excess of uPA to suPAR overcame the inhibitory effect of suPAR (data not shown).

ATF inhibits HIV replication in primary human MDMs

We have previously reported that uPA inhibits virus replication in primary MDM17  and we here report that exogenously added ATF exerts an anti-HIV activity in vitro–infected MDMs established from several donors (Figure 2A).

Figure 2

ATF inhibits acute R5 HIV-1 replication in MDMs. (A). The anti-HIV activity of uPA and ATF were tested on primary MDMs infected with an R5 HIV-1. The compounds were added to the cell cultures at the time of infection and every 3 days after infection. The results of a single experiment, representative of 5 independently performed experiments with cells from different donors, are shown. The error bars indicate the SD of triplicate samples, whereas asterisks at the indicated time points indicate statistical significance (P = .001) among Nil and uPA or ATF treated cells; no significant differences were observed in the inhibition levels induced by uPA or ATF. (B) Culture supernatants of uninfected MDMs and in vitro–infected MDMs (C) were analyzed for the levels of uPA and suPAR at the day corresponding to the peak of viral replication (RT activity) in 5 independent experiments performed with cells from different donors. Differences in the levels of suPAR, uPA, the uPA-suPAR complex, and the suPAR/uPA ratio between uninfected and infected cells tested were not significant. R2  was calculated by linear regression and P values by Spearman rank test. cpm indicates counts per minute.

Figure 2

ATF inhibits acute R5 HIV-1 replication in MDMs. (A). The anti-HIV activity of uPA and ATF were tested on primary MDMs infected with an R5 HIV-1. The compounds were added to the cell cultures at the time of infection and every 3 days after infection. The results of a single experiment, representative of 5 independently performed experiments with cells from different donors, are shown. The error bars indicate the SD of triplicate samples, whereas asterisks at the indicated time points indicate statistical significance (P = .001) among Nil and uPA or ATF treated cells; no significant differences were observed in the inhibition levels induced by uPA or ATF. (B) Culture supernatants of uninfected MDMs and in vitro–infected MDMs (C) were analyzed for the levels of uPA and suPAR at the day corresponding to the peak of viral replication (RT activity) in 5 independent experiments performed with cells from different donors. Differences in the levels of suPAR, uPA, the uPA-suPAR complex, and the suPAR/uPA ratio between uninfected and infected cells tested were not significant. R2  was calculated by linear regression and P values by Spearman rank test. cpm indicates counts per minute.

Close modal

Both uninfected and infected MDMs released approximately 3-fold more uPA and 6-fold more suPAR than U1 cells on a per cell basis (Table 1). However, as observed in U1 cells, most soluble uPA was complexed by suPAR in MDMs, with a suPAR/uPA ratio always greater than 20 (Table 1). There was not a significant correlation between the levels of released uPA and suPAR in infected MDMs (Figure 2B), or between the levels of released uPA or suPAR and peak HIV replication in MDMs, although there was a trend toward higher levels of (complexed) uPA and more virus production (Figure 2C).

PMA and TNF-α, but not other HIV-inductive stimuli, up-regulate CD11b expression in U1 cells

Unstimulated U1 cells showed constitutive cell-surface expression of uPAR and CD18, but negligible presence of CD11b (Table 2; Figure 3A). PMA stimulation up-regulated the levels of cell-surface uPAR (reaching a plateau approximately 20 hours after stimulation, as previously reported in both U93733  and MCF-7 cells34 ) and induced the expression of CD11b, thus allowing the formation of Mac-1 (Figure 3A top panels). Superimposable results, although inducing lower levels of CD11b expression, were observed by TNF-α stimulation (Figure 3A middle panels). In contrast, IL-6 strongly up-regulated both HIV-1 and uPAR expression, but failed to induce detectable levels of CD11b (Figure 3A bottom panels). Of interest, uPA inhibited HIV expression in U1 cells stimulated with both PMA and TNF-α, but not with IL-6 (Figure 3B). U1 cells were also stimulated with IFN-γ or granulocyte-macrophage colony-stimulating factor (GM-CSF) plus LPS,35-37  which up-regulated virus expression to comparable levels, in the presence and absence of uPA.26  However, both IFN-γ and GM-CSF plus PLS did not induce expression of CD11b (data not shown); concomitantly, uPA failed to inhibit HIV production induced by these stimuli.

Table 2

Expression of integrin chains and cell-surface receptors in U1 cells

NilPMAIL-6
Integrin chain 
    αIIb (CD41) − − − 
    αL (CD11a) 
    αE (CD103) − − − 
    αM (CD11b) − +* − 
    αX (CD11c) − +* − 
    αV (CD51) − − − 
    αVβ3 (VN-R) − − − 
    αVβ5 (FN-R, VN-R) 
    α5β1 (FN-R) 
    α1 (VLA1) − − − 
    α2 (CD49b) − − − 
    α3 (CD49c) − − − 
    α4 (CD49d) 
    α5 (CD49e) 
    α6 (CD49f) − +* − 
    β1 (CD29) 
    β2 (CD18) ++* 
    β3 (CD61) − − − 
    β4 (CD104) 
Receptors 
    uPAR ++* ++* 
    FPRL1 
    EGFR − − − 
NilPMAIL-6
Integrin chain 
    αIIb (CD41) − − − 
    αL (CD11a) 
    αE (CD103) − − − 
    αM (CD11b) − +* − 
    αX (CD11c) − +* − 
    αV (CD51) − − − 
    αVβ3 (VN-R) − − − 
    αVβ5 (FN-R, VN-R) 
    α5β1 (FN-R) 
    α1 (VLA1) − − − 
    α2 (CD49b) − − − 
    α3 (CD49c) − − − 
    α4 (CD49d) 
    α5 (CD49e) 
    α6 (CD49f) − +* − 
    β1 (CD29) 
    β2 (CD18) ++* 
    β3 (CD61) − − − 
    β4 (CD104) 
Receptors 
    uPAR ++* ++* 
    FPRL1 
    EGFR − − − 

Cell-surface expression of the indicated molecules was evaluated by cytofluorimetric analysis after 24 hours of stimulation (n = 4 for all antigens except uPAR and CD11b, which were tested in 9 independent experiments). An identical pattern of expression was observed up to 72 hours after stimulation. Up-regulation of CD11b and CD11c in U937 cells stimulated with PMA was previously reported.63 

VN-R indicates vitronectin receptor; FN-R, fibronectin receptor; −, negative cells; and +, positive cells.

*

P < .05 versus Nil.

Cells testing positive with a geometric mean fluorescence intensity greater than 2-fold that of unstimulated cells.

Figure 3

Induction of CD11b expression in U1 cells and uPA-mediated inhibition of HIV-1 expression. (A) U1 cells were gated based on their light scatter and analyzed for both uPAR (R4 mAb) and CD11b (αM44 mAb) expression 20 hours after stimulation with PMA, TNF-α, or IL-6; the integrin β2 chain CD18 is constitutively expressed on U1 cells (Table 1). All stimuli up-regulated the levels of uPAR on the cell surface, but only PMA and, to a lesser extent, TNF-α, promoted the expression of CD11b on U1 cells. (B) uPA inhibits HIV expression in U1 cells stimulated with PMA and TNF-α, but not with IL-6, IFN-γ, or GM-CSF plus LPS. The results were obtained from a single experiment representative or 5 or more independently performed experiments. Error bars indicate the SD of duplicate samples; and cpm, counts per minute.

Figure 3

Induction of CD11b expression in U1 cells and uPA-mediated inhibition of HIV-1 expression. (A) U1 cells were gated based on their light scatter and analyzed for both uPAR (R4 mAb) and CD11b (αM44 mAb) expression 20 hours after stimulation with PMA, TNF-α, or IL-6; the integrin β2 chain CD18 is constitutively expressed on U1 cells (Table 1). All stimuli up-regulated the levels of uPAR on the cell surface, but only PMA and, to a lesser extent, TNF-α, promoted the expression of CD11b on U1 cells. (B) uPA inhibits HIV expression in U1 cells stimulated with PMA and TNF-α, but not with IL-6, IFN-γ, or GM-CSF plus LPS. The results were obtained from a single experiment representative or 5 or more independently performed experiments. Error bars indicate the SD of duplicate samples; and cpm, counts per minute.

Close modal

Among other uPA coreceptors and integrin chains expressed by U1 cells, neither FPRL1, nor α5β1, α6, and αX integrin chains were involved in uPA-mediated inhibition of HIV expression (see Table S1 and Figures S1,S2).

The Mac-1 ligand FNG inhibits late events of HIV expression in U1 cells and in acutely infected U937 cells

We next evaluated whether known Mac-1 ligands could also affect HIV expression in either PMA or TNF-α stimulated U1 cells in the absence of uPA. U1 cells were incubated with FNG, known to bind to both Mac-138  and the VN receptor αVβ3 (which, however, was not expressed by U1 cells, as shown in Table 2)17 ; the cells were then stimulated by either PMA or HIV-inductive cytokines. Incubation of U1 cells with FNG in the absence of other stimuli did not activate HIV expression (data not shown); however, FNG inhibited HIV expression in U1 cells stimulated with PMA (Figure 4A), but not with IL-6 (data not shown). As with uPA inhibition,16  FNG inhibition of HIV expression in PMA-stimulated U1 cells was reversed by cell disruption (Figure 4B). Furthermore, FNG inhibited the acute replication of X4 HIV-1 in U937 cells (Figure 4C). As observed with U1 cells, U937 cell disruption at time points before and after the peak of HIV replication restored the levels of RT activity to those of control cells (Figure 4D).

Figure 4

FNG inhibits HIV expression in PMA-stimulated U1 cells and in U937 cells acutely infected with an X4 HIV-1. (A) U1 cells were incubated with the indicated concentrations of FNG and then stimulated with PMA. (B) Cell disruption rescues the RT activity levels of U1 cells stimulated with PMA in the presence of FNG, as detected after 4 days of culture (arrowhead in panel A). (C) U937 cells were incubated with FNG (100 μg/mL) and then infected with the X4 strain HIV-1LAI/IIIB; FNG was added to the cell cultures every 72 hours, when 50% of the culture supernatant was replaced with culture medium. (D) Cell disruption rescued the RT activity levels of acutely infected U937 cells in the presence of FNG at the indicated days after infection (arrowheads in panel C; *P < .001). The results shown were derived from 1 experiment, representative of 3 (A,B) and 2 (C,D) independently performed experiments. cpm indicates counts per minute.

Figure 4

FNG inhibits HIV expression in PMA-stimulated U1 cells and in U937 cells acutely infected with an X4 HIV-1. (A) U1 cells were incubated with the indicated concentrations of FNG and then stimulated with PMA. (B) Cell disruption rescues the RT activity levels of U1 cells stimulated with PMA in the presence of FNG, as detected after 4 days of culture (arrowhead in panel A). (C) U937 cells were incubated with FNG (100 μg/mL) and then infected with the X4 strain HIV-1LAI/IIIB; FNG was added to the cell cultures every 72 hours, when 50% of the culture supernatant was replaced with culture medium. (D) Cell disruption rescued the RT activity levels of acutely infected U937 cells in the presence of FNG at the indicated days after infection (arrowheads in panel C; *P < .001). The results shown were derived from 1 experiment, representative of 3 (A,B) and 2 (C,D) independently performed experiments. cpm indicates counts per minute.

Close modal

Preventing association of CD11b/CD18 from uPAR in the absence of receptor ligands inhibits HIV-1 expression

To investigate the role of Mac-1 interaction with uPAR in terms of inhibition of HIV expression, we tested the potential effect of M2, a peptide known to prevent or disrupt the integrin association with uPAR.39  Surprisingly, M25 alone, but not its control scrambled peptide, inhibited HIV expression in PMA-stimulated U1 cells in the absence of uPA in a concentration-dependent fashion, reaching more than 90% inhibition at 80 μM (Figure 5A). Furthermore, M25 inhibited the residual levels of RT activity in the presence of uPA (Figure 5A); identical results were obtained in U1 cells stimulated with TNF-α (data not shown). The M25 anti-HIV mechanism resembled that induced by both uPA and FNG in that it was fully reverted by cell disruption (Figure 5B), suggesting that prevention of the association between Mac-1 and uPAR triggers an inhibitory pathway of HIV-1 particle release.

Figure 5

M25 inhibits HIV expression in PMA-stimulated U1 cells both in the absence and presence of uPA. (A) U1 cells were incubated with different concentrations (5-80 μM) of the M25 peptide or its scrambled form (M25scr) in the presence or absence of uPA (10 nM) and were then stimulated with PMA. A concentration-dependent inhibition of HIV expression was observed in the presence of M25, but not of its control peptide, both in the presence and absence of uPA. (B) Cell disruption rescued HIV expression from the inhibitory effect of M25 (40 μM) or uPA (10 nM) after 5 days of culture (*P < .001; **P < .001). The results shown were derived from one experiment representative of 6 (A) and 3 (B) independently performed experiments. cpm indicates counts per minute.

Figure 5

M25 inhibits HIV expression in PMA-stimulated U1 cells both in the absence and presence of uPA. (A) U1 cells were incubated with different concentrations (5-80 μM) of the M25 peptide or its scrambled form (M25scr) in the presence or absence of uPA (10 nM) and were then stimulated with PMA. A concentration-dependent inhibition of HIV expression was observed in the presence of M25, but not of its control peptide, both in the presence and absence of uPA. (B) Cell disruption rescued HIV expression from the inhibitory effect of M25 (40 μM) or uPA (10 nM) after 5 days of culture (*P < .001; **P < .001). The results shown were derived from one experiment representative of 6 (A) and 3 (B) independently performed experiments. cpm indicates counts per minute.

Close modal

The anti-HIV effect triggered by uPA/uPAR interaction is independent of CD11b

Experiments were next carried out after transfection of U1 cells with siRNA directed to either uPAR or CD11b. As shown earlier, the constitutive levels of expression of uPAR in U1 cells were enhanced by 24-hour stimulation with PMA (Figure 6A top left panel), while CD11b, which was not expressed by unstimulated cells, was clearly induced by PMA (Figure 6A top right panel). Transfection with their respective siRNA partially reduced the expression of uPAR (Figure 6A bottom left panel) while it abrogated PMA-induced CD11b expression (Figure 6A bottom right panel); these suppressive effects were stable for at least 3 days after cell stimulation (data not shown).

Figure 6

CD11b is required for the anti-HIV activity of M25 but not of uPA. (A) Expression of both uPAR and CD11b in unstimulated and PMA-stimulated U1 cells before (top panels) and after (bottom panels) transfection with their respective siRNA and control mock siRNA. The uPAR siRNA showed only a partial inhibitory effect on its target, whereas the CD11b siRNA abolished the expression of the integrin chain. (B) CD11b siRNA abolished the M25 inhibitory effects on HIV expression but did not affect uPA-dependent inhibition, whereas decreased uPAR expression abrogated the inhibitory effects of both agents. (C) Lack of CD11b expression did not interfere with uPA/uPAR–dependent adhesion of PMA-stimulated U1 cells to the plastic substrate, an adhesion that was otherwise inhibited by uPAR siRNA (*P < .001). No effect of M25 on the adhesion of PMA-stimulated U1 cells was observed. The results shown were derived from one experiment representative of 4 independently performed experiments. cpm indicates counts per minute.

Figure 6

CD11b is required for the anti-HIV activity of M25 but not of uPA. (A) Expression of both uPAR and CD11b in unstimulated and PMA-stimulated U1 cells before (top panels) and after (bottom panels) transfection with their respective siRNA and control mock siRNA. The uPAR siRNA showed only a partial inhibitory effect on its target, whereas the CD11b siRNA abolished the expression of the integrin chain. (B) CD11b siRNA abolished the M25 inhibitory effects on HIV expression but did not affect uPA-dependent inhibition, whereas decreased uPAR expression abrogated the inhibitory effects of both agents. (C) Lack of CD11b expression did not interfere with uPA/uPAR–dependent adhesion of PMA-stimulated U1 cells to the plastic substrate, an adhesion that was otherwise inhibited by uPAR siRNA (*P < .001). No effect of M25 on the adhesion of PMA-stimulated U1 cells was observed. The results shown were derived from one experiment representative of 4 independently performed experiments. cpm indicates counts per minute.

Close modal

Mock siRNA did not alter the inhibitory effects of either uPA or M25 on HIV production, while CD11b siRNA abolished the anti-HIV activity of M25 but did not affect the inhibitory capacity of uPA (Figure 6B left and right panels, respectively). In contrast, the reduced expression of uPAR abolished the anti-HIV activity of both uPA and M25 peptide (Figure 6B middle panel).

Given the dependence of uPA anti-HIV effect on VN-mediated cell adhesion,40  we next investigated the potential interference of these siRNAs on U1 cell adhesion to the plastic substrate in the presence to either uPA or M25. As expected, M25 did not affect plastic adhesion of both unstimulated (not shown) and PMA-stimulated U1 cells (Figure 6C). Transfection with CD11b siRNA did not alter the ability of uPA to induce U1 cell adhesion, whereas this effect was lost in U1 cells transfected with uPAR siRNA (Figure 6C).

Cell adhesion–dependent uPA inhibition of HIV expression involves integrin β chains

We then investigated the potential contribution of β1 and β2 integrin chains. Different anti-integrin blocking Abs were used to coat the plastic of tissue-culture microwells, thereby inducing integrin-dependent cell adhesion.40  In addition, we tested the same Abs in suspension to prevent cell clustering by homotypic interactions.41 

Plates coated with Abs recognizing antigens not expressed by unstimulated U1 cells, such as CD3 and CD11b (Table 2), failed to induce cell adhesion (Figure 7 top panel). In contrast, Abs recognizing β1, but not β2, integrin chains induced cell adhesion in both unstimulated and uPA-stimulated cells (Figure 7 top panel). PMA stimulation enhanced the levels of cell adhesion in plates coated with anti-CD11a, anti-CD11b, and anti-β1, but not anti-β2, integrin chains (Figure 7 top panel). Consistently, uPA increased the levels of cell adhesion in PMA-stimulated cells in plates coated with Abs directed against CD11a, CD11b, β1, but not β2, integrin chains. As expected, when Abs were added in suspension, either no adhesion or reduced levels of adhesion were observed in comparison to what was observed in the analogous Ab-coated plates (Figure 7 top panel).

Figure 7

Differential effect of soluble and coated Ab directed against α and β integrin chains on U1 cell adhesion and HIV-1 expression. Microtiter wells were coated with different Ab (1 μg/well) before seeding and stimulation of U1 cells (2 × 105 cells/mL). In parallel, U1 cells were resuspended at 2 × 105 cells/mL in medium enriched with the indicated Ab (10 μg/mL) before seeding and stimulation with PMA in the presence or absence of uPA. The same concentration of Ab was supplemented after 24 hours of cell culture. Cell adhesion (top panel) and virus expression (bottom panel) were measured 48 hours after cell seeding. The results shown were derived from 1 experiment representative of 3 independently performed experiments. Significantly enhanced cell adhesion was observed in all PMA-stimulated cells versus unstimulated cells (Nil; *P < .001). Furthermore, uPA increased PMA-stimulated cell adhesion (**P < .001 vs PMA alone). Significantly increased levels of cell adhesion were also observed when otherwise unstimulated U1 cells were incubated in wells coated with anti-CD29 mAb (°P < .001 vs Nil). Virus expression was significantly inhibited by uPA in all conditions (⋀P < .001 vs PMA), but not in uPA-treated, PMA-stimulated cells incubated with either soluble anti-CD29 or soluble anti-CD18 Ab- versus BSA-treated cells (*P < .001). Finally, significantly lower levels of HIV production were observed in PMA-stimulated U1 cells incubated in wells coated with anti-CD11b mAb (***P = .004).

Figure 7

Differential effect of soluble and coated Ab directed against α and β integrin chains on U1 cell adhesion and HIV-1 expression. Microtiter wells were coated with different Ab (1 μg/well) before seeding and stimulation of U1 cells (2 × 105 cells/mL). In parallel, U1 cells were resuspended at 2 × 105 cells/mL in medium enriched with the indicated Ab (10 μg/mL) before seeding and stimulation with PMA in the presence or absence of uPA. The same concentration of Ab was supplemented after 24 hours of cell culture. Cell adhesion (top panel) and virus expression (bottom panel) were measured 48 hours after cell seeding. The results shown were derived from 1 experiment representative of 3 independently performed experiments. Significantly enhanced cell adhesion was observed in all PMA-stimulated cells versus unstimulated cells (Nil; *P < .001). Furthermore, uPA increased PMA-stimulated cell adhesion (**P < .001 vs PMA alone). Significantly increased levels of cell adhesion were also observed when otherwise unstimulated U1 cells were incubated in wells coated with anti-CD29 mAb (°P < .001 vs Nil). Virus expression was significantly inhibited by uPA in all conditions (⋀P < .001 vs PMA), but not in uPA-treated, PMA-stimulated cells incubated with either soluble anti-CD29 or soluble anti-CD18 Ab- versus BSA-treated cells (*P < .001). Finally, significantly lower levels of HIV production were observed in PMA-stimulated U1 cells incubated in wells coated with anti-CD11b mAb (***P = .004).

Close modal

None of the Abs tested modified the levels of HIV expression in unstimulated cells or cells incubated with uPA alone (Figure 7 bottom panel). When U1 cells were stimulated with PMA, no effects were observed in the presence of all tested Abs, except in plates coated with anti-CD11b that inhibited virus production, while no effects were observed when this Ab was diluted in cell culture supernatants. UPA maintained its anti-HIV activity in all tested conditions except in the presence of soluble anti-β1 and anti-β2 integrin chains that abolished its antiviral effect, without affecting uPA-mediated enhancement of PMA-induced cell adhesion.

Thus, the cell adhesion–dependent antiviral signal triggered by uPA binding to uPAR requires downstream activation of β integrin chains.

In the present study we investigated the role of potential signaling partners mediating uPA/uPAR-dependent inhibition of late events in HIV-1 replication in monocytic cells such as primary MDMs, and promonocytic U1 and U937 cells lines. UPA anti-HIV activity in U1 cells was restricted to PMA and TNF-α stimulation, but was ineffective when these cells were stimulated with other cytokines or LPS. This restricted pattern of the uPA-mediated anti-HIV effect was paralleled by the up-regulation of CD11b on the surface of U1 cells which constitutively expressed the β2 chain (CD18) to form Mac-1 (also known as complement receptor-3) known to provide signaling competence to the uPA/uPAR complex in myelomonocytic cells. Independent of uPA/uPAR, FNG binding to Mac-1 inhibited late events in HIV production in both stimulated U1 cells and acutely infected U937 cells. Furthermore, prevention of uPAR association with CD11b by the M25 peptide showed similar HIV-inhibitory effects in PMA-stimulated U1 cells. siRNA targeting uPAR reversed the inhibitory effect of both uPA and M25, whereas siRNA-mediated down-regulation of CD11b abolished M25 but did not affect uPA-dependent antiviral effect. Soluble Ab directed against β1 and/or β2 integrin chains, but not against the α chains of CD11a-b-c, prevented uPA interference with HIV expression. These findings suggest a model whereby a similar, if not identical, anti-HIV signal, interfering with late events in the virus life cycle, is triggered by uPA binding to uPAR and by uPAR-independent ligation of Mac-1 or prevention of the interaction between CD11b and uPAR.

As observed in cancer,42  the uPA/uPAR system plays a relevant role in HIV disease. Soluble levels of suPAR represent a potent predictor of HIV disease progression, independent of the levels of peripheral CD4+ T-cell counts and viremia.11  These observations were extended and confirmed in patients with HIV-associated dementia.14,15  Cultivation and infection of both primary MDMs and promonocytic cell lines, such as U1 and U937, revealed that both uPA and suPAR levels are expressed in vitro at levels similar to those observed in vivo.15  Furthermore, in vitro–released uPA was mostly complexed with suPAR (Table 1), as described in vivo.15  This observation likely accounts for the lack of evidence of an autocrine/paracrine role of endogenous uPA on HIV replication in our model systems. Because free uPA inhibits acute HIV-1 replication in primary MDMs and U937 cells as well as virus expression in chronically infected U1 cells,16  its sequestration by suPAR to form a biologically inactive uPA-suPAR complex may indeed prevent the anti-HIV effects of uPA. These findings provide a potential explanation for the CD4 and viremia independent prognostic value of increased suPAR concentrations in HIV disease progression.

Furthermore, HIV-1 infection of primary MDMs did not alter levels of expression of uPA and suPAR (Table 1). This observation may account for the lack of correlation between circulating levels of uPA and suPAR and viremia.11  In contrast, suPAR levels were found to be strictly correlated with the state of immune activation in HIV-infected individuals, as determined by the serum levels of TNF-α,43  sTNFrII,12,44  CCL2, CCL4, and CCL5.45  suPAR levels also correlate with lipid and glucose metabolism in HIV-infected individuals,43,46  as was reflected in vitro by a trend of association among the levels of HIV-1 replication in acutely infected MDMs and the levels of suPAR and uPA-suPAR complexes (Figure 2).

uPA-dependent inhibition of virus expression in U1 cells was restricted to PMA and TNF-α stimulation, while other HIV-inductive stimuli (including IL-6, IFN-γ, and GM-CSF plus LPS) were unaffected. Of interest, both PMA and TNF-α up-regulate HIV expression via activation of NF-κB,47  whereas the other stimuli act by activation of an ERK-1/-2 AP-1–dependent pathway up-regulating virus transcription.26,48  Since the inhibitory effect of uPA occurs at a posttranscriptional/posttranslational level resulting in the accumulation of virions in intracellular vacuoles,16,49  the selectivity of the anti-HIV effect of uPA was unlikely related to interference with virus transcription, as previously described.16,17,49 

We previously demonstrated that the catalytic component of uPA was dispensable for its anti-HIV effects, unlike its signaling component ATF,16,49  as here confirmed in stimulated U1 cells with smaller ATF peptides and primary MDMs. Removal of uPAR, by either cleavage of its GPI anchor or reduction of its expression by means of siRNA, resulted in the abolition of the antiviral effect of uPA. Because uPAR is a GPI-anchored receptor, signal transduction is usually mediated by other receptors and, in monocytic cells, either by FPRLI50  or by integrins such as Mac-1.51  Indeed, both FPRLI and some integrins, including αVβ5, α5β1, and other chains, were constitutively expressed by U1 cells and not affected by HIV-inductive stimuli. However, no evidence of interference with uPA-dependent anti-HIV effects was obtained by incubating U1 cells with agonists of either FPRL1 or these integrins.

Unstimulated U1 cells express abundant levels of the integrin β chain CD18, but not of the α chains CD11b or CD11c. Only PMA and TNF-α up-regulated the expression of CD11b and CD11c, as reported,52  suggesting that the uPA anti-HIV effect could be mediated by either Mac-1 (CD11b/CD18) or p150/95 (CD11c/CD18). However, prevention of CD11c expression by means of siRNA did not affect HIV expression in PMA-stimulated cells (data not shown), while different CD11b ligands, including FNG and anti-CD11b mAb (on coated plates) triggered an HIV-inhibitory signal in PMA-stimulated U1 cells. Like uPA, FNG interfered with the late stage of virus production both in U1 and acutely infected U937 cells. Before our observation, Mac-1 was shown to enhance CD4/CCR5-dependent HIV entry in monocytes, macrophages, and dendritic cells,53-55  whereas, conversely, several Mac-1 ligands, including FNG, have shown inhibitory effects on the enhancement of opsonized HIV infection.53-55  Of interest, another Mac-1 ligand, soluble CD16 (sCD16), was reported to be significantly decreased in HIV disease progression.53,56  In addition, intercellular adhesion molecule-1, a ligand of Mac-1, is incorporated into the virion envelope.57  A confounding aspect of the role of Mac-1 in HIV replication is the observation that ligation of the β2 integrin chain CD18 by immobilized Ab has been shown to increase HIV transcriptional activation in THP1 monocytic cells.58,59  Similar results were described in primary monocytes and U937 cells in the presence of FNG,52,54  in U1 cells cocultivated with HUVEC,60  and in chronically infected promyelocytic OM10.1 cells in which an autocrine release of TNF-α was involved in the up-regulation of virus expression.58,59  However, the adhesion of LPS-stimulated THP1 cells to tissue-culture plastic has been reported to down-regulate the release of HIV virions in both stimulated THP1 cell lines and primary MDMs.58  In agreement with these earlier observations, we have previously reported that the abolition of uPA inhibitory effects and up-regulation of virus production was observed in both acutely infected MDMs and PMA-stimulated U1 cells cultivated in the absence of VN or in nonadherent conditions obtained on Teflon-coated plates.17 

Quite surprisingly, incubation of U1 cells with M25, a peptide derived from the β propeller region of CD11b known to disrupt its association with uPAR,61  also inhibited late events in HIV expression in PMA-stimulated U1 cells. Since CD11b is not expressed in unstimulated U1 cells, M25 likely prevents its association with uPAR after PMA stimulation. Thus, expression of Mac-1 uncoupled from uPAR leads to an inhibitory effect on HIV expression independent of the exogenous addition of either uPA or integrin ligands other than those present in the culture medium (such as heat-inactivated C3). In this regard, disruption of uPAR-integrin interaction has been previously shown to prevent the association between β integrin and Src kinases, thereby affecting adhesion-dependent signal transduction.62  This effect has been linked to the ability of uPAR to stabilize complexes of caveolin, integrins, and Src kinases ultimately responsible for adhesion-dependent signaling.62 

Interference with either uPAR or CD11b expression by siRNA resulted in different biologic effects. UPAR targeting siRNA, as well as PIPLC treatment, only partially decreased the levels of uPAR expression in PMA-stimulated U1 cells, yet it abolished uPA and M25 anti-HIV effects. Conversely, almost complete prevention of CD11b expression by siRNA resulted in the expected loss of the M25 anti-HIV effect, but was irrelevant for uPA-dependent effects on HIV expression and cell adhesion. These results suggest that uPAR may functionally replace CD11b in terms of triggering a β chain–dependent anti-HIV signal, as previously suggested.63  In this regard, uPA may promote an indirect activation of β1 and β2 integrin chains after the recruitment of VN to form a uPA/uPAR/VN supercomplex competent of mediating cell adhesion and mechanotransduction.64  In support of this interpretation, soluble anti-β1 and anti-β2 Ab did not prevent uPA induced cell adhesion, as recently reported,65  while they fully prevented the anti-HIV effect of uPA. Overall these results suggest that an interplay between integrins and uPAR may occur in lipid rafts known to play a fundamental role in both viral entry47  and in the exit of new progeny virions from infected cells.66 

In addition to uPA/uPAR and Mac-1 and its ligands, other extracellular stimuli, including IFN-γ67  and CCL2/MCP-1,68  have been previously shown to inhibit virion release by favoring intracellular virion accumulation. In this regard, assembly and release of HIV virions in multivescicular bodies belonging to the hexosomal pathway69  are crucial aspects characterizing the infection of macrophages.69  Although the relevance of intracellular virion accumulation in macrophages has been recently questioned,70,71  different monocytic cells, including U937, HL-60, THP1, and MonoMac cells,72  primary monocytes,73,74  and macrophages derived from CD34+ bone marrow precursors75  show accumulation and release of HIV virions in intracellular compartments for several weeks, in addition to their generation at the plasma membrane. Even more important, these morphogenetic features of virion assembly in subcellular vacuolar compartments have been documented in vivo in the brain of individuals with HIV encephalitis.76 

In conclusion, our results suggest a general model whereby interacting, yet distinct, ligands can trigger a signaling pathway resulting in the inhibition of HIV spread by interfering with late- stage virion assembly and release in macrophages. At least 3 converging signals leading to this common final pathway can be postulated based on the present results: (1) ligation of uPAR by its natural ligand uPA via its ATF domain; (2) ligand binding and cross-linking of Mac-1; and (3) prevention of uPAR interaction with CD11b resulting in the assembly of Mac-1 dissociated from uPAR. As for other examples of biologic redundancy, these observations suggest that control of HIV assembly, maturation, and virion release are crucial checkpoints for efficient viral spreading from macrophages to other susceptible cell types.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Nicolai Sidenius for testing levels of suPAR and uPA-suPAR complex; Massimo Resnati and Elisabetta Ferrero (San Raffaele Scientific Institute, Milan, Italy) for providing Abs against FPRL1, CD11b, and α1 and α3 integrin chains; and Ji-Ming Wang (National Cancer Institute, Frederick, MD) for kind provision of the W peptide.

This study was supported in part by a grant from the VI National Program of Research against AIDS of the Istituto Superiore di Sanità (Rome, Italy; G.P.) and by National Institutes of Health grant 1-R21-MH075670-01A1 (M.A., F.B., and G.P.).

S.A.M. participated in this study as partial fulfillment of her PhD course in Molecular and Cellular Biology of the Vita-Salute San Raffaele University in Milan and of the Open University of London, United Kingdom. Her PhD course is sustained by a fellowship from the Europrise Network of Excellence funded by the Sixth Framework Programme of the European Commission.

National Institutes of Health

Contribution: M.A. designed and performed experiments, analyzed and interpreted the results, and contributed to the drafting of the manuscript; S.A.M. and C.E. performed experiments and interpreted the results; R.P. and F.B. designed experiments and contributed to the interpretation of the results and drafting of the manuscript; and G.P. designed experiments, analyzed and interpreted the results, and contributed to the drafting of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Massimo Alfano, P2/P3 Laboratories, DIBIT, Via Olgettina n. 58, 20132, Milan, Italy; e-mail: alfano.massimo@hsr.it.

1
Dano
 
K
Andreasen
 
PA
Grondahl-Hansen
 
J
Kristensen
 
P
Nielsen
 
LS
Skriver
 
L
Plasminogen activators, tissue degradation, and cancer.
Adv Cancer Res
1985
, vol. 
44
 (pg. 
139
-
266
)
2
Blasi
 
F
Vassalli
 
JD
Dano
 
K
Urokinase-type plasminogen activator: proenzyme, receptor, and inhibitors.
J Cell Biol
1987
, vol. 
104
 (pg. 
801
-
804
)
3
Gyetko
 
MR
Libre
 
EA
Fuller
 
JA
Chen
 
GH
Toews
 
G
Urokinase is required for T lymphocyte proliferation and activation in vitro.
J Lab Clin Med
1999
, vol. 
133
 (pg. 
274
-
288
)
4
May
 
AE
Kanse
 
SM
Lund
 
LR
Gisler
 
RH
Imhof
 
BA
Preissner
 
KT
Urokinase receptor (CD87) regulates leukocyte recruitment via beta 2 integrins in vivo.
J Exp Med
1998
, vol. 
188
 (pg. 
1029
-
1037
)
5
Blasi
 
F
Carmeliet
 
P
uPAR: a versatile signalling orchestrator.
Nat Rev Mol Cell Biol
2002
, vol. 
3
 (pg. 
932
-
943
)
6
Ossowski
 
L
Aguirre-Ghiso
 
JA
Urokinase receptor and integrin partnership: coordination of signaling for cell adhesion, migration and growth.
Curr Opin Cell Biol
2000
, vol. 
12
 (pg. 
613
-
620
)
7
Blasi
 
F
uPA, uPAR, PAI-1: key intersection of proteolytic, adhesive and chemotactic highways?
Immunol Today
1997
, vol. 
18
 (pg. 
415
-
417
)
8
Shushakova
 
N
Tkachuk
 
N
Dangers
 
M
, et al. 
Urokinase-induced activation of the gp130/Tyk2/Stat3 pathway mediates a pro-inflammatory effect in human mesangial cells via expression of the anaphylatoxin C5a receptor.
J Cell Sci
2005
, vol. 
118
 (pg. 
2743
-
2753
)
9
Chapman
 
HA
Plasminogen activators, integrins, and the coordinated regulation of cell adhesion and migration.
Curr Opin Cell Biol
1997
, vol. 
9
 (pg. 
714
-
724
)
10
Preissner
 
KT
Kanse
 
SM
May
 
AE
Urokinase receptor: a molecular organizer in cellular communication.
Curr Opin Cell Biol
2000
, vol. 
12
 (pg. 
621
-
628
)
11
Sidenius
 
N
Sier
 
CF
Ullum
 
H
, et al. 
Serum level of soluble urokinase-type plasminogen activator receptor is a strong and independent predictor of survival in human immunodeficiency virus infection.
Blood
2000
, vol. 
96
 (pg. 
4091
-
4095
)
12
Ostrowski
 
SR
Katzenstein
 
TL
Pedersen
 
M
, et al. 
Plasma levels of intact and cleaved urokinase receptor decrease in HIV-1-infected patients initiating highly active antiretroviral therapy.
Scand J Immunol
2006
, vol. 
63
 (pg. 
478
-
486
)
13
Sporer
 
B
Koedel
 
U
Popp
 
B
Paul
 
R
Pfister
 
HW
Evaluation of cerebrospinal fluid uPA, PAI-1, and soluble uPAR levels in HIV-infected patients.
J Neuroimmunol
2005
, vol. 
163
 (pg. 
190
-
194
)
14
Cinque
 
P
Nebuloni
 
M
Santovito
 
ML
, et al. 
The urokinase receptor is overexpressed in the AIDS dementia complex and other neurological manifestations.
Ann Neurol
2004
, vol. 
55
 (pg. 
687
-
694
)
15
Sidenius
 
N
Nebuloni
 
M
Sala
 
S
, et al. 
Expression of the urokinase plasminogen activator and its receptor in HIV-1-associated central nervous system disease.
J Neuroimmunol
2004
, vol. 
157
 (pg. 
133
-
139
)
16
Alfano
 
M
Sidenius
 
N
Panzeri
 
B
Blasi
 
F
Poli
 
G
Urokinase-urokinase receptor interaction mediates an inhibitory signal for HIV-1 replication.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 (pg. 
8862
-
8867
)
17
Elia
 
C
Cassol
 
E
Sidenius
 
N
, et al. 
Inhibition of HIV replication by the plasminogen activator is dependent on vitronectin-mediated cell adhesion.
J Leukoc Biol
2007
, vol. 
82
 (pg. 
1212
-
1220
)
18
Granowitz
 
EV
Saget
 
BM
Wang
 
MZ
Dinarello
 
CA
Skolnik
 
PR
Interleukin 1 induces HIV-1 expression in chronically infected U1 cells: blockade by interleukin 1 receptor antagonist and tumor necrosis factor binding protein type 1.
Mol Med
1995
, vol. 
1
 (pg. 
667
-
677
)
19
Folks
 
T
Benn
 
S
Rabson
 
A
, et al. 
Characterization of a continuous T-cell line susceptible to the cytopathic effects of the acquired immunodeficiency syndrome (AIDS)-associated retrovirus.
Proc Natl Acad Sci U S A
1985
, vol. 
82
 (pg. 
4539
-
4543
)
20
Folks
 
TM
Clouse
 
KA
Justement
 
J
, et al. 
Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone.
Proc Natl Acad Sci U S A
1989
, vol. 
86
 (pg. 
2365
-
2368
)
21
Maslin
 
CL
Kedzierska
 
K
Webster
 
NL
Muller
 
WA
Crowe
 
SM
Transendothelial migration of monocytes: the underlying molecular mechanisms and consequences of HIV-1 infection.
Curr HIV Res
2005
, vol. 
3
 (pg. 
303
-
317
)
22
Sankaran
 
S
George
 
MD
Reay
 
E
, et al. 
Rapid onset of intestinal epithelial barrier dysfunction in primary human immunodeficiency virus infection is driven by an imbalance between immune response and mucosal repair and regeneration.
J Virol
2008
, vol. 
82
 (pg. 
538
-
545
)
23
Weeks
 
BS
The role of HIV-1 activated leukocyte adhesion mechanisms and matrix metalloproteinase secretion in AIDS pathogenesis.
Int J Mol Med
1998
, vol. 
1
 (pg. 
361
-
366
)
24
Pomerantz
 
RJ
Feinberg
 
MB
Trono
 
D
Baltimore
 
D
Lipopolysaccharide is a potent monocyte/macrophage-specific stimulator of human immunodeficiency virus type 1 expression.
J Exp Med
1990
, vol. 
172
 (pg. 
253
-
261
)
25
Poli
 
G
Orenstein
 
JM
Kinter
 
A
Folks
 
TM
Fauci
 
AS
Interferon-alpha but not AZT suppresses HIV expression in chronically infected cell lines.
Science
1989
, vol. 
244
 (pg. 
575
-
577
)
26
Rizzi
 
C
Crippa
 
MP
Jeeninga
 
RE
, et al. 
Pertussis toxin B-oligomer suppresses IL-6 induced HIV-1 and chemokine expression in chronically infected U1 cells via inhibition of activator protein 1.
J Immunol
2006
, vol. 
176
 (pg. 
999
-
1006
)
27
Mengozzi
 
M
De Filippi
 
C
Transidico
 
P
, et al. 
Human immunodeficiency virus replication induces monocyte chemotactic protein-1 in human macrophages and U937 promonocytic cells.
Blood
1999
, vol. 
93
 (pg. 
1851
-
1857
)
28
Fernie
 
BF
Poli
 
G
Fauci
 
AS
Alpha interferon suppresses virion but not soluble human immunodeficiency virus antigen production in chronically infected T-lymphocytic cells.
J Virol
1991
, vol. 
65
 (pg. 
3968
-
3971
)
29
Cubellis
 
MV
Nolli
 
ML
Cassani
 
G
Blasi
 
F
Binding of single-chain prourokinase to the urokinase receptor of human U937 cells.
J Biol Chem
1986
, vol. 
261
 (pg. 
15819
-
15822
)
30
Ploug
 
M
Structure-function relationships in the interaction between the urokinase-type plasminogen activator and its receptor.
Curr Pharm Des
2003
, vol. 
9
 (pg. 
1499
-
1528
)
31
Koshelnick
 
Y
Ehart
 
M
Stockinger
 
H
Binder
 
BR
Mechanisms of signaling through urokinase receptor and the cellular response.
Thromb Haemost
1999
, vol. 
82
 (pg. 
305
-
311
)
32
Skretting
 
G
Torgersen
 
ML
van Deurs
 
B
Sandvig
 
K
Endocytic mechanisms responsible for uptake of GPI-linked diphtheria toxin receptor.
J Cell Sci
1999
, vol. 
112
 
Pt 22
(pg. 
3899
-
3909
)
33
Picone
 
R
Kajtaniak
 
EL
Nielsen
 
LS
, et al. 
Regulation of urokinase receptors in monocytelike U937 cells by phorbol ester phorbol myristate acetate.
J Cell Biol
1989
, vol. 
108
 (pg. 
693
-
702
)
34
Stillfried
 
GE
Saunders
 
DN
Ranson
 
M
Plasminogen binding and activation at the breast cancer cell surface: the integral role of urokinase activity.
Breast Cancer Res
2007
, vol. 
9
 pg. 
R14
 
35
Clouse
 
KA
Powell
 
D
Washington
 
I
, et al. 
Monokine regulation of human immunodeficiency virus-1 expression in a chronically infected human T cell clone.
J Immunol
1989
, vol. 
142
 (pg. 
431
-
438
)
36
Goletti
 
D
Kinter
 
AL
Biswas
 
P
Bende
 
SM
Poli
 
G
Fauci
 
AS
Effect of cellular differentiation on cytokine-induced expression of human immunodeficiency virus in chronically infected promonocytic cells: dissociation of cellular differentiation and viral expression.
J Virol
1995
, vol. 
69
 (pg. 
2540
-
2546
)
37
Goletti
 
D
Kinter
 
AL
Hardy
 
EC
Poli
 
G
Fauci
 
AS
Modulation of endogenous IL-1 beta and IL-1 receptor antagonist results in opposing effects on HIV expression in chronically infected monocytic cells.
J Immunol
1996
, vol. 
156
 (pg. 
3501
-
3508
)
38
Simon
 
DI
Ezratty
 
AM
Francis
 
SA
Rennke
 
H
Loscalzo
 
J
Fibrin(ogen) is internalized and degraded by activated human monocytoid cells via Mac-1 (CD11b/CD18): a nonplasmin fibrinolytic pathway.
Blood
1993
, vol. 
82
 (pg. 
2414
-
2422
)
39
Kugler
 
MC
Wei
 
Y
Chapman
 
HA
Urokinase receptor and integrin interactions.
Curr Pharm Des
2003
, vol. 
9
 (pg. 
1565
-
1574
)
40
Feldhaus
 
MJ
Kessel
 
JM
Zimmerman
 
GA
McIntyre
 
TM
Engagement of ICAM-3 activates polymorphonuclear leukocytes: aggregation without degranulation or beta 2 integrin recruitment.
J Immunol
1998
, vol. 
161
 (pg. 
6280
-
6287
)
41
Graham
 
IL
Gresham
 
HD
Brown
 
EJ
An immobile subset of plasma membrane CD11b/CD18 (Mac-1) is involved in phagocytosis of targets recognized by multiple receptors.
J Immunol
1989
, vol. 
142
 (pg. 
2352
-
2358
)
42
Binder
 
BR
Mihaly
 
J
Prager
 
GW
uPAR-uPA-PAI-1 interactions and signaling: a vascular biologist's view.
Thromb Haemost
2007
, vol. 
97
 (pg. 
336
-
342
)
43
Andersen
 
O
Eugen-Olsen
 
J
Kofoed
 
K
Iversen
 
J
Haugaard
 
SB
Soluble urokinase plasminogen activator receptor is a marker of dysmetabolism in HIV-infected patients receiving highly active antiretroviral therapy.
J Med Virol
2008
, vol. 
80
 (pg. 
209
-
216
)
44
Ostrowski
 
SR
Katzenstein
 
TL
Piironen
 
T
Gerstoft
 
J
Pedersen
 
BK
Ullum
 
H
Soluble urokinase receptor levels in plasma during 5 years of highly active antiretroviral therapy in HIV-1-infected patients.
J Acquir Immune Defic Syndr
2004
, vol. 
35
 (pg. 
337
-
342
)
45
Pawlak
 
K
Mysliwiec
 
M
Pawlak
 
D
The urokinase-type plasminogen activator/its soluble receptor system is independently related to carotid atherosclerosis and associated with CC-chemokines in uraemic patients.
Thromb Res
2008
, vol. 
122
 (pg. 
328
-
335
)
46
Andersen
 
O
Eugen-Olsen
 
J
Kofoed
 
K
Iversen
 
J
Haugaard
 
SB
suPAR associates to glucose metabolic aberration during glucose stimulation in HIV-infected patients on HAART.
J Infect
2008
, vol. 
57
 (pg. 
55
-
63
)
47
Alfano
 
M
Poli
 
G
Role of cytokines and chemokines in the regulation of innate immunity and HIV infection.
Mol Immunol
2005
, vol. 
42
 (pg. 
161
-
182
)
48
Yang
 
X
Chen
 
Y
Gabuzda
 
D
ERK MAP kinase links cytokine signals to activation of latent HIV-1 infection by stimulating a cooperative interaction of AP-1 and NF-kappa B.
J Biol Chem
1999
, vol. 
274
 (pg. 
27981
-
27988
)
49
Wada
 
M
Wada
 
NA
Shirono
 
H
Taniguchi
 
K
Tsuchie
 
H
Koga
 
J
Amino-terminal fragment of urokinase-type plasminogen activator inhibits HIV-1 replication.
Biochem Biophys Res Commun
2001
, vol. 
284
 (pg. 
346
-
351
)
50
Resnati
 
M
Pallavicini
 
I
Wang
 
JM
, et al. 
The fibrinolytic receptor for urokinase activates the G protein-coupled chemotactic receptor FPRL1/LXA4R.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 (pg. 
1359
-
1364
)
51
Ross
 
GD
Role of the lectin domain of Mac-1/CR3 (CD11b/CD18) in regulating intercellular adhesion.
Immunol Res
2002
, vol. 
25
 (pg. 
219
-
227
)
52
Sitrin
 
RG
Pan
 
PM
Srikanth
 
S
Todd
 
RF
Fibrinogen activates NF-kappa B transcription factors in mononuclear phagocytes.
J Immunol
1998
, vol. 
161
 (pg. 
1462
-
1470
)
53
Bouhlal
 
H
Galon
 
J
Kazatchkine
 
MD
Fridman
 
WH
Sautes-Fridman
 
C
Haeffner Cavaillon
 
N
Soluble CD16 inhibits CR3 (CD11b/CD18)-mediated infection of monocytes/macrophages by opsonized primary R5 HIV-1.
J Immunol
2001
, vol. 
166
 (pg. 
3377
-
3383
)
54
Thieblemont
 
N
Haeffner-Cavaillon
 
N
Haeffner
 
A
Cholley
 
B
Weiss
 
L
Kazatchkine
 
MD
Triggering of complement receptors CR1 (CD35) and CR3 (CD11b/CD18) induces nuclear translocation of NF-kappa B (p50/p65) in human monocytes and enhances viral replication in HIV-infected monocytic cells.
J Immunol
1995
, vol. 
155
 (pg. 
4861
-
4867
)
55
Stoiber
 
H
Speth
 
C
Dierich
 
MP
Role of complement in the control of HIV dynamics and pathogenesis.
Vaccine
2003
, vol. 
21
 
Suppl 2
(pg. 
S77
-
S82
)
56
Khayat
 
D
Soubrane
 
C
Andrieu
 
JM
, et al. 
Changes of soluble CD16 levels in serum of HIV-infected patients: correlation with clinical and biologic prognostic factors.
J Infect Dis
1990
, vol. 
161
 (pg. 
430
-
435
)
57
Stoiber
 
H
Frank
 
I
Spruth
 
M
, et al. 
Inhibition of HIV-1 infection in vitro by monoclonal antibodies to the complement receptor type 3 (CR3): an accessory role for CR3 during virus entry?
Mol Immunol
1997
, vol. 
34
 (pg. 
855
-
863
)
58
Shattock
 
RJ
Friedland
 
JS
Griffin
 
GE
Release of human immunodeficiency virus by THP-1 cells and human macrophages is regulated by cellular adherence and activation.
J Virol
1993
, vol. 
67
 (pg. 
3569
-
3575
)
59
Shattock
 
RJ
Griffin
 
GE
Cellular adherence enhances HIV replication in monocytic cells.
Res Virol
1994
, vol. 
145
 (pg. 
139
-
145
)
60
Fan
 
ST
Hsia
 
K
Edgington
 
TS
Upregulation of human immunodeficiency virus-1 in chronically infected monocytic cell line by both contact with endothelial cells and cytokines.
Blood
1994
, vol. 
84
 (pg. 
1567
-
1572
)
61
Simon
 
DI
Wei
 
Y
Zhang
 
L
, et al. 
Identification of a urokinase receptor-integrin interaction site: promiscuous regulator of integrin function.
J Biol Chem
2000
, vol. 
275
 (pg. 
10228
-
10234
)
62
Wei
 
Y
Yang
 
X
Liu
 
Q
Wilkins
 
JA
Chapman
 
HA
A role for caveolin and the urokinase receptor in integrin-mediated adhesion and signaling.
J Cell Biol
1999
, vol. 
144
 (pg. 
1285
-
1294
)
63
Todd
 
RF
Petty
 
HR
Beta 2 (CD11/CD18) integrins can serve as signaling partners for other leukocyte receptors.
J Lab Clin Med
1997
, vol. 
129
 (pg. 
492
-
498
)
64
Madsen
 
CD
Sarra Ferraris
 
GM
Andolfo
 
A
Cunningham
 
O
Sidenius
 
N
uPAR-induced cell adhesion and migration: vitronectin provides the key.
J Cell Biol
2007
, vol. 
177
 (pg. 
927
-
939
)
65
Petzinger
 
J
Saltel
 
F
Hersemeyer
 
K
, et al. 
Urokinase receptor (CD87) clustering in detergent-insoluble adhesion patches leads to cell adhesion independently of integrins.
Cell Commun Adhes
2007
, vol. 
14
 (pg. 
137
-
155
)
66
Wilflingseder
 
D
Stoiber
 
H
Float on: lipid rafts in the lifecycle of HIV.
Front Biosci
2007
, vol. 
12
 (pg. 
2124
-
2135
)
67
Biswas
 
P
Poli
 
G
Kinter
 
AL
, et al. 
Interferon gamma induces the expression of human immunodeficiency virus in persistently infected promonocytic cells (U1) and redirects the production of virions to intracytoplasmic vacuoles in phorbol myristate acetate-differentiated U1 cells.
J Exp Med
1992
, vol. 
176
 (pg. 
739
-
750
)
68
Fantuzzi
 
L
Spadaro
 
F
Vallanti
 
G
, et al. 
Endogenous CCL2 (monocyte chemotactic protein-1) modulates human immunodeficiency virus type-1 replication and affects cytoskeleton organization in human monocyte-derived macrophages.
Blood
2003
, vol. 
102
 (pg. 
2334
-
2337
)
69
Ono
 
A
Freed
 
EO
Cell-type-dependent targeting of human immunodeficiency virus type 1 assembly to the plasma membrane and the multivesicular body.
J Virol
2004
, vol. 
78
 (pg. 
1552
-
1563
)
70
Deneka
 
M
Pelchen-Matthews
 
A
Byland
 
R
Ruiz-Mateos
 
E
Marsh
 
M
In macrophages, HIV-1 assembles into an intracellular plasma membrane domain containing the tetraspanins CD81, CD9, and CD53.
J Cell Biol
2007
, vol. 
177
 (pg. 
329
-
341
)
71
Welsch
 
S
Keppler
 
OT
Habermann
 
A
Allespach
 
I
Krijnse-Locker
 
J
Krausslich
 
HG
HIV-1 buds predominantly at the plasma membrane of primary human macrophages.
PLoS Pathog
2007
, vol. 
3
 pg. 
e36
 
72
Cassol
 
E
Alfano
 
M
Biswas
 
P
Poli
 
G
Monocyte-derived macrophages and myeloid cell lines as targets of HIV-1 replication and persistence.
J Leukoc Biol
2006
, vol. 
80
 (pg. 
1018
-
1030
)
73
Gartner
 
S
Markovits
 
P
Markovitz
 
DM
Kaplan
 
MH
Gallo
 
RC
Popovic
 
M
The role of mononuclear phagocytes in HTLV-III/LAV infection.
Science
1986
, vol. 
233
 (pg. 
215
-
219
)
74
Gendelman
 
HE
Orenstein
 
JM
Martin
 
MA
, et al. 
Efficient isolation and propagation of human immunodeficiency virus on recombinant colony-stimulating factor 1-treated monocytes.
J Exp Med
1988
, vol. 
167
 (pg. 
1428
-
1441
)
75
Folks
 
TM
Justement
 
J
Kinter
 
A
, et al. 
Characterization of a promonocyte clone chronically infected with HIV and inducible by 13-phorbol-12-myristate acetate.
J Immunol
1988
, vol. 
140
 (pg. 
1117
-
1122
)
76
Orenstein
 
JM
Jannotta
 
F
Human immunodeficiency virus and papovavirus infections in acquired immunodeficiency syndrome: an ultrastructural study of three cases.
Hum Pathol
1988
, vol. 
19
 (pg. 
350
-
361
)
Sign in via your Institution