Indinavir (IDV) is a potent and selective human immunodeficiency virus type 1 (HIV-1) protease inhibitor (PI) widely used in antiretroviral therapy for suppression of HIV, but its effects on the immune system are relatively unknown. Recently, it has been reported that PIs inhibit lymphocyte apoptosis. In the present study we have investigated the effects of ex vivo addition of IDV on lymphocyte activation and apoptosis in cells from HIV-infected children (n = 18) and from healthy uninfected individuals (controls, n = 5) as well as in Jurkat and PM1 T-cell lines. Pretreatment of control peripheral blood mononuclear cell (PBMC) cultures with IDV resulted in a dose-dependent inhibition of lymphoproliferative responses to different activation stimuli. Additionally, this treatment led to cell-cycle arrest in G0/G1 phase in anti-CD3 monoclonal antibody–stimulated PBMC cultures in controls and in 15 of 18 HIV-infected children. Spontaneous- or activation-induced apoptosis of PBMCs from HIV-infected or uninfected individuals or of Fas-induced apoptosis in Jurkat and PM1 T cell lines were not inhibited by IDV. Moreover, IDV did not inhibit activation of caspases-1, -3, -4, -5, -9, and -8 in lysates of Jurkat T cells undergoing Fas-induced apoptosis. The findings indicate that IDV interferes with cell-cycle progression in primary cells but does not directly affect apoptosis. It is concluded that IDV may prolong cell survival indirectly by inhibiting their entry into cell cycle. In individuals on PI therapy, PI-mediated effects could potentially modulate immunologic responses independently of antiviral activity against HIV.

Immunodeficiency is a hallmark of human immunodeficiency virus type 1 (HIV-1) disease and is characterized by a progressive decrease in CD4 T cells. The introduction of combination therapies (eg, with drugs targeting the viral-specific enzymes reverse transcriptase and protease1-3) have had a dramatic effect in slowing disease progression. In the majority of patients, drug regimens containing HIV-1 protease inhibitors (PIs) and reverse transcriptase inhibitors (RTIs) are effective in reducing plasma viral burden, with concomitant increase in CD4 lymphocyte counts.4,5 When therapy fails, the virologic rebound is associated with a decline in CD4 T cells. Curiously, in a subset of patients, the immunologic benefit persists despite virologic failure.6,7 The mechanism of this “discordant” response is not clearly understood, and the observed effects have been ascribed to virologic factors as well as to direct effects of the PIs on the immune system, distinct from their antiviral effects.

Mechanisms of CD4 T-cell loss in HIV disease have been under debate for several years. Loss of CD4 T cells by apoptosis has been put forth as one mechanism for the immune deficiency in HIV infection. Increased lymphocyte apoptosis is recognized as a feature of HIV infection and has been attributed to direct and indirect effects of HIV on the immune system.8,9 Therapy-induced decrease in plasma HIV RNA has, in fact, been shown to result in reduction in lymphocyte apoptosis.10,11 The antiviral effect of PI drugs is mediated by the inhibition of the HIV protease, the enzyme that is required for the processing of HIV polypeptides into smaller peptides and maturation of viral particles.12-14 It has been argued that the similarity between HIV protease (an aspartyl protease) and the cellular caspases (cysteine proteases) involved in apoptosis could result in PI-mediated anticaspase activity.13-16 Some recent studies with the PI Ritonavir have implicated a direct antiapoptotic effect of the drug on T cells17-19 as a possible mechanism for CD4 T-cell increase following therapy. However, in another study, the in vitro addition of the PI Indinavir (IDV) to cells of HIV-infected patients did not result in antiapoptotic effects.20 The mechanism by which PI therapy results in immunologic benefit independently of its antiviral effects thus remains unclear.

A characteristic feature of PIs is their ability to modulate proteosomes21,22 that are involved in a variety of cellular functions and constitute an important component of the biological system. It has been shown that this effect of PIs on proteosomes impairs antigen presentation and processing by antigen-presenting cells.21 In addition, PIs have also been shown to influence lymphocyte proliferation20,23; the reported effects, however, have been contradictory, presumably because the experimental systems have been different as have the doses of drugs used, which have varied widely in these studies.

In the present study we have tested the hypothesis that PIs mediate immunomodulatory effects that can influence host immunity in a manner distinct from the benefits attributable to control of viral replication. Here we have examined the effect of therapeutic concentrations of IDV on lymphocyte activation and on spontaneous and activation-induced lymphocyte apoptosis in peripheral blood mononuclear cell (PBMC) cultures of healthy volunteers and HIV-infected patients and in T cell lines.

Donors

This study was conducted in healthy volunteers (n = 5) and in HIV-infected children (n = 18) during regularly scheduled visits to the Pediatric Immunology Clinic at North Shore University Hospital (Manhasset, NY) by protocols approved by the hospital's institutional review board. The children ranged in age from 0.4 to 13.9 years (median, 8.7) and had absolute CD4 counts ranging from 17 to 2371 cells/μL (median, 595). Plasma virus load ranged from 1.6 to 5.5 log10 HIV RNA copies/mL (median, 3 log10). Most infected children were on antiretroviral therapy that included the RTIs Zidovudine, Stavudine, Zalcitabine, and Didanosine and the PIs Nelfinavir or Saquinavir plus Ritonavir. Nine of 18 patients were on one or 2 PIs in combination with 2 RTIs, whereas the others were on 2 (n = 4) or 3 RTIs (n = 5).

Isolation of cells

Peripheral venous blood was collected by venipuncture into tubes containing acid citrate dextrose as an anticoagulant. PBMCs were isolated by Ficoll-Hypaque (Lymphoprep; Nycomed, Oslo, Norway) density gradient centrifugation and suspended at a density of 1 × 106 cells/mL in RPMI 1640 (Whittaker Biproducts, Walkersville, MD), 10% fetal calf serum (Hyclone Laboratories, Logan, UT), 2 mM L-glutamine (Whittaker Biproducts), penicillin G (100 U/mL), and streptomycin (100 μg/mL) (Whittaker Biproducts).

Cell lines and other reagents

Two different T-cell lines, Jurkat (clone E6-1; ATCC, American Type Culture Collection, Manassas, VA) and PM124 (AIDS Research and Reference Reagent Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD) were maintained and cultured in RPMI 1640, 10% fetal bovine serum, 2 mM L-glutamine, and antibiotics penicillin and streptomycin. Antihuman Fas (CD95) monoclonal antibody (mAb) CH11 and fluorogenic substrates Ac-YVAD-AFC, Ac-DEVD-AFC, Ac-LEHD-AFC, and Ac-IETD-AFC of caspase-1; caspase-3; caspases-4, -5, and -9; and caspase-8, respectively, and their specific inhibitors (Ac-YVAD-CHO, Z-DEVD-FMK, Z-LEHD-FMK, and Z-IETD-FMK, respectively) were purchased from Kamiya Biomedical Company (Seattle, WA). Purified mouse antihuman CD3 mAb was purchased from BD Pharmingen (San Diego, CA). A gift of IDV sulfate powder was generously given by Merck Research Laboratories (Rahway, NJ). RNAse, phytohemagglutinin (PHA), concanavalin A (Con A), phorbol 12-myristate 13-acetate (PMA), and Ionomycin were purchased from Sigma (St Louis, MO), and propidium iodide was obtained from Molecular Probes (Eugene, OR).

Culture conditions and measurement of lymphocyte apoptosis and cell-cycle profiles

Test PBMCs were cultured for 3 days with anti-CD3 mAb (0.5 μg/mL) or with control isotype-matched antibody to study spontaneous- and activation-induced lymphocyte apoptosis. To study the effect of IDV, the drug was added ex vivo to PBMC cultures for 18 hours in 24-well culture plates (Becton Dickinson Labware, Franklin Lakes, NJ). Different concentrations of IDV were prepared from IDV powder dissolved in dimethyl sulfoxide (DMSO) and added to the cultures at a final dilution of 1 μL/mL. Thereafter, the cells were cultured with anti-CD3 mAb (0.5 μg/mL) or with control isotype-matched antibody for an additional 48 hours. At termination of cultures, cells were harvested by centrifugation, washed, and fixed in 70% ethanol for 1 hour and incubated with propidium iodide (50 μg/mL) and RNAse (100 μg/ml). Samples were kept in the dark at room temperature for 30 minutes and stored at 4°C until analyzed for cell-cycle profile and apoptosis. In healthy volunteers, for the study of IDV effect on lymphocyte apoptosis, additional experiments were performed to simulate the state of in vivo lymphocyte activation, which is a characteristic feature of the HIV-infected state. PBMCs were first activated with PHA (2 μg/mL) for 4 days, washed, and then treated as described above with different concentrations of IDV for 18 hours, followed by activation with anti-CD3 mAb for 48 hours.

Apoptosis in Jurkat and PM1 T cells was induced through Fas death pathway by culturing the cells with anti-Fas mAb CH11 (100 ng/mL) in the presence of different concentrations of IDV for 2, 6, and 18 hours. In other experiments, Jurkat and PM1 T cells were precultured with different concentrations of IDV for 18 hours followed by treatment with CH11 for 6 and 18 hours. To investigate the effect of IDV on the cell cycle, Jurkat and PM1 T cells were cultured with different concentrations of IDV over periods of 24, 48, and 72 hours and stained with propidium iodide.

Lymphocyte apoptosis was investigated as described in propidium iodide-stained cells using the Coulter Epics Elite ESP (Coulter, Hialeah, FL) instrument after careful gating for lymphocytes.25 For cell-cycle studies, DNA content was determined by using listmode files with the multicycle flow cytometry software (Phoenix Flow System, San Diego, CA). In healthy volunteers, results of cycling cells (in S + G2/M phase) or of cells undergoing apoptosis were expressed as a percentage of total cells for that culture condition. In patients, cycling cells and apoptotic cells in IDV-treated cultures are expressed as a percentage of cycling cells or apoptotic cells in cultures without IDV (ie, cells precultured with 1 μL/mL DMSO).

Lymphoproliferation studies

PBMCs from healthy human volunteers were precultured in medium alone or with different concentrations of IDV for 18 hours in 96-well plates (Becton Dickinson Labware), followed by the addition of stimuli in triplicate [viz, anti-CD3 mAb (0.5 μg/mL), PHA (2 μg/mL), Con A (2 μg/mL), PMA (10 ng/mL) plus Ionomycin (1 μM)], isotype-matched antibodies, or DMSO (control) for an additional 48 hours. Cultures were pulsed with [14C]-thymidine (1 μCi/well) for the last 18 hours, and uptake of [14C]-thymidine in cells was quantitated by scintillation counting.26Lymphoproliferative responses of IDV-treated cultures are expressed as the percentage of [14C]-thymidine incorporated in cultures without IDV treatment.

Study of caspase activity

Cell lysates from CH11-treated Jurkat CD4 T cells was used to quantitate activity of caspase-1, -3, -4, -5, -9, and -8. In brief, 1 × 106 Jurkat CD4 T cells/mL were cultured with 100 ng/mL anti-Fas antibody CH11 for 3 hours. Cells were washed once with ice-cold Hanks balanced salt solution, followed by suspension in cell lysis buffer [20 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 1% Triton X-100, 2.5 mM sodium pyrophosphate, 1 mM sodium orthovanadate, 1 mM phenylmethylsulfonyl fluoride, 1 mg/mL leupeptin, and 20 mg/mL aprotinin] for 20 minutes. Cells were incubated on ice for 20 minutes and then disrupted by passing through a 20-gauge needle for 10 times. Cell extracts were clarified by centrifugation at 16 000g for 15 minutes at 4°C. To study the effect of IDV on caspase activity, cell extracts were incubated with different concentrations of IDV (12.5 μM to 100 μM) or with specific caspase inhibitors (Ac-YVAD-CHO, Z-DEVD-FMK, Z-LEHD-FMK, and Z-IETD-FMK) followed by addition of caspase-specific fluorogenic substrates (Ac-YVAD-AFC, Ac-DEVD-AFC, Ac-LEHD-AFC, and Ac-IETD-AFC) for caspase-1; caspase-3; caspases-4, -5, and -9; and caspase-8, respectively. Activity of each caspase was estimated as a release of 7-amino-4-trifluoromethyl coumarin (AFC) from the caspase-specific substrate and measured by dual scanning microplate spectrofluorometer (SPECTRAmax Gemini XS; Molecular Devices, Sunnyvale, CA). Kinetics of individual reactions were monitored for 2 hours at 37°C in the microplate spectrofluorometer.

Statistical analysis

SigmaStat 2.0 statistical software (Jandel Scientific Software, San Rafel, CA) was used for all statistical analysis.

IDV blocks lymphocyte cell cycle in G0/G1 phase in cells from healthy volunteers and HIV-infected children

PBMCs from healthy volunteers and from HIV-infected children were incubated in the absence or presence of different concentrations of IDV for 18 hours followed by stimulation with anti-CD3 or control antibodies for an additional 48 hours. The effect of IDV on anti-CD3 mAb–induced cell-cycle progression in healthy volunteers is shown in Figure 1A. Preincubation of PBMCs with IDV blocked anti-CD3–induced cell-cycle progression in a dose-dependent manner. IDV was found to block cells in the G0/G1 phase, prior to their entry into synthesis phase (S). At the lowest concentration of IDV (5 μM), 36.2% ± 3.5% cells were in cell cycle compared with 47.2 ± 1.2 cycling cells in PBMCs cultured with anti-CD3 mAb without prior exposure to IDV (P < .05). At the highest concentration of IDV (50 μM), only 15% ± 1.3% cells were in cycling phase. The effect of IDV on anti-CD3–induced cell-cycle progression in PBMCs of HIV-infected children is shown in Figure 1B. Pretreatment of PBMCs with IDV was found to inhibit anti-CD3–induced cell entry into S phase in a dose-dependent manner in 15 of 18 HIV-infected children. The ex vivo blocking effect of IDV on anti-CD3–induced cell cycling was noted to occur in patients regardless of whether they were receiving PI therapy or not. Addition of IDV at the same time as anti-CD3 mAb did not inhibit cell-cycle progression. Viability of cells was equivalent in cultures with or without IDV pretreatment.

Fig. 1.

Effect of IDV on anti-CD3–induced cell-cycle progression.

(A) PBMCs from control volunteers (n = 5). *, P < .005. (B) PBMCs from HIV-infected children (n = 15). PBMCs from healthy volunteers and HIV-infected children were preincubated without or with different concentrations of IDV for 18 hours followed by activation with anti-CD3 mAb (0.5 μg/mL) for an additional 48 hours, and cells were stained with propidium iodide for cell-cycle profile analysis by multicycle flow cytometry software. In healthy volunteers, cycling cells in S + G2/M phase are represented as percentage (mean ± SD) of total cells for each test condition. For patients, cells in S + G2/M phase in IDV-treated cultures are expressed as a percentage (mean ± SD) of cells in S + G2/M phase in cultures without IDV treatment. *, P < .05.

Fig. 1.

Effect of IDV on anti-CD3–induced cell-cycle progression.

(A) PBMCs from control volunteers (n = 5). *, P < .005. (B) PBMCs from HIV-infected children (n = 15). PBMCs from healthy volunteers and HIV-infected children were preincubated without or with different concentrations of IDV for 18 hours followed by activation with anti-CD3 mAb (0.5 μg/mL) for an additional 48 hours, and cells were stained with propidium iodide for cell-cycle profile analysis by multicycle flow cytometry software. In healthy volunteers, cycling cells in S + G2/M phase are represented as percentage (mean ± SD) of total cells for each test condition. For patients, cells in S + G2/M phase in IDV-treated cultures are expressed as a percentage (mean ± SD) of cells in S + G2/M phase in cultures without IDV treatment. *, P < .05.

Close modal

IDV impairs lymphoproliferative responses

Pretreatment of PBMC cultures of healthy volunteers with IDV (0-50 μM) for 18 hours, followed by stimulation with anti-CD3 mAb (0.5 μg/mL) for 48 hours resulted in dose-dependent reduction of lymphoproliferative responses (Figure 2). This antiproliferative effect of IDV was also noted for cultures stimulated with PHA, PMA plus Ionomycin, or Con A. A similar pattern of dose-dependent inhibition was noted in cultures of unstimulated cells, with more than 50% inhibition of [14C]-thymidine uptake at a 5 to 10 μM dose range of IDV. At 18 hours after preincubation of PBMCs with IDV, no significant difference in [14C]-thymidine uptake and cell viability was noted between cells cultured with and without different concentrations of IDV (data not shown).

Fig. 2.

Effect of IDV on lymphoproliferation.

PBMCs from healthy volunteers were preincubated without or with different concentrations of IDV for 18 hours followed by activation with different stimuli for an additional 48 hours. Proliferation was measured with [14C]-thymidine incorporation added at 1 μCi/well for the last 48 hours. Results of [14C]-thymidine incorporation in IDV-treated cultures are expressed as the percentage of responses of cells cultured without IDV. Data represents mean ± SD for 1 of 5 representative experiments: ♦, medium alone; ●, anti-CD3 (0.5 μg/mL); ▴, PHA (2 μg/mL); ▪, PMA (10 ng/mL); + Ionomycin (1 μM); and ○, Con A (2 μg/mL).

Fig. 2.

Effect of IDV on lymphoproliferation.

PBMCs from healthy volunteers were preincubated without or with different concentrations of IDV for 18 hours followed by activation with different stimuli for an additional 48 hours. Proliferation was measured with [14C]-thymidine incorporation added at 1 μCi/well for the last 48 hours. Results of [14C]-thymidine incorporation in IDV-treated cultures are expressed as the percentage of responses of cells cultured without IDV. Data represents mean ± SD for 1 of 5 representative experiments: ♦, medium alone; ●, anti-CD3 (0.5 μg/mL); ▴, PHA (2 μg/mL); ▪, PMA (10 ng/mL); + Ionomycin (1 μM); and ○, Con A (2 μg/mL).

Close modal

Spontaneous and anti-CD3–induced lymphocyte apoptosis in HIV-infected children

Spontaneous lymphocyte apoptosis was investigated in 3-day PBMC cultures of HIV-infected children. As shown in Figure3, spontaneous lymphocyte apoptosis was not significantly different in HIV-infected children who were on PI drug regimens (range, 4.4% to 34%; median, 10%) from those not on PI-containing drug regimens (range, 4.3% to 33%; median, 20%). Activation-induced lymphocyte apoptosis following anti-CD3 stimulation, however, was significantly increased over spontaneous apoptosis only in children on non-PI therapies, whereas the percentage of lymphocyte apoptosis remained unaffected following anti-CD3 activation in children who were on PI regimens. Anti-CD3–induced lymphocyte apoptosis was also significantly higher in children on non-PI drug regimens (range, 7.6% to 47%; median, 25.1%) as compared with those on PI-containing drug regimens (range, 6.5% to 34%; median, 12.2%;P < .05).

Fig. 3.

Spontaneous and anti-CD3–induced apoptosis in PBMCs from HIV-infected children.

PBMCs of HIV-infected children were cultured in medium for 3 days without anti-CD3 mAb to determine spontaneous lymphocyte apoptosis and with anti-CD3 mAb (0.5 μg/mL) for 48 hours to determine activation-induced lymphocyte apoptosis. Data shown represents percentage of lymphocyte apoptosis, estimated by propidium iodide staining. Symbols represent individual patients who were on non-PI– (left) or on PI- (right) containing antiretroviral therapy regimens. Mean value in each data set is represented by a solid line.

Fig. 3.

Spontaneous and anti-CD3–induced apoptosis in PBMCs from HIV-infected children.

PBMCs of HIV-infected children were cultured in medium for 3 days without anti-CD3 mAb to determine spontaneous lymphocyte apoptosis and with anti-CD3 mAb (0.5 μg/mL) for 48 hours to determine activation-induced lymphocyte apoptosis. Data shown represents percentage of lymphocyte apoptosis, estimated by propidium iodide staining. Symbols represent individual patients who were on non-PI– (left) or on PI- (right) containing antiretroviral therapy regimens. Mean value in each data set is represented by a solid line.

Close modal

IDV does not inhibit lymphocyte apoptosis

Preincubation of PBMCs from HIV-infected children with IDV (5-50 μM) did not influence spontaneous or anti-CD3–induced apoptosis (Figure 4A); this response was not different between patients receiving PI- or non–PI-containing antiretroviral therapies (data not shown).

Fig. 4.

Effect of IDV on spontaneous and anti-CD3–induced apoptosis in PBMCs.

The PBMCs were from HIV-infected children (A, n = 15) and from healthy volunteers (B). PBMCs were preincubated without and with different concentrations of IDV followed by culture in the absence (■) or presence (▪) of anti-CD3 mAb (0.5 μg/mL) for an additional 48 hours. Lymphocyte apoptosis was estimated by propidium iodide staining. In healthy volunteers, cultures with 50 μM caspase inhibitor Ac-DEVD-CHO were also established. In patients, apoptosis in IDV-treated cultures are expressed as a percentage of cells undergoing apoptosis in cultures without IDV pretreatment. In healthy volunteers, lymphocytes undergoing apoptosis are expressed as a percentage of total cells for each condition. * and **, P < .05.

Fig. 4.

Effect of IDV on spontaneous and anti-CD3–induced apoptosis in PBMCs.

The PBMCs were from HIV-infected children (A, n = 15) and from healthy volunteers (B). PBMCs were preincubated without and with different concentrations of IDV followed by culture in the absence (■) or presence (▪) of anti-CD3 mAb (0.5 μg/mL) for an additional 48 hours. Lymphocyte apoptosis was estimated by propidium iodide staining. In healthy volunteers, cultures with 50 μM caspase inhibitor Ac-DEVD-CHO were also established. In patients, apoptosis in IDV-treated cultures are expressed as a percentage of cells undergoing apoptosis in cultures without IDV pretreatment. In healthy volunteers, lymphocytes undergoing apoptosis are expressed as a percentage of total cells for each condition. * and **, P < .05.

Close modal

In PBMCs of healthy individuals, the above protocol of 18 hours' preincubation with IDV followed by anti-CD3 activation yielded very few cells undergoing apoptosis. These studies were extended to examine the effect of IDV in apoptosis of PBMC cultures following prior activation with PHA (Figure 4B). PBMCs from healthy volunteers were cultured with PHA (2 μg/mL) for 4 days followed by incubation with different concentrations of IDV for 18 hours. These activated cells were cultured with anti-CD3 or control mAb (0.5 μg/mL) for an additional 48 hours to study activation-induced apoptosis. No significant difference in apoptosis was noted in PBMC cultures that were treated or not treated with different concentrations of IDV, whereas anti-CD3–induced apoptosis was significantly blocked by Ac-DEVD-CHO, a known inhibitor of caspases.

IDV does not influence cell cycle or apoptosis of Jurkat and PM1 T-cell lines

IDV at concentrations of 5 to 50 μM did not influence the cell-cycle profile of Jurkat (Figure 5A) or PM1 (Figure 5B) T-cell lines over incubation periods of 24, 48, or 72 hours. Fas-induced apoptosis was not inhibited in cells precultured with different concentrations of IDV or in cultures simultaneously incubated with different concentrations of IDV and anti-Fas antibody CH11, either in Jurkat (Figure 5C) or PM1 T cells (Figure 5D) over a period of 2 to 18 hours.

Fig. 5.

Effect of IDV on cell-cycle progression.

Cell-cycle progression of Jurkat (A) and PM1 T (B) cells and apoptosis in Jurkat (C) and PM1 (D) T-cell lines. For cell-cycle analysis Jurkat and PM1 T cells were cultured with different concentrations of IDV for 24 (⋄), 48 (♦), and 72 (×) hours. Cell-cycle progression was determined by propidium iodide staining and is represented as the percentage of cells in S + G2/M phase by using multicycle flow cytometry software. To study the effect of IDV on anti-Fas–induced apoptosis in Jurkat and PM1 T cells, cells were cultured without or with anti-Fas mAb CH11 (100 ng/mL) in presence of different concentrations of IDV, and apoptosis was determined at 2 (▵), 6 (■), and 18 (●) hours. Cells were also precultured with different concentrations of IDV for 18 hours, and apoptosis was induced by culturing these cells with CH11 for 6 (▪) and 18 (●) hours. Data are shown for mean ± SD of the percentage of cells undergoing apoptosis as estimated by propidium iodide staining.

Fig. 5.

Effect of IDV on cell-cycle progression.

Cell-cycle progression of Jurkat (A) and PM1 T (B) cells and apoptosis in Jurkat (C) and PM1 (D) T-cell lines. For cell-cycle analysis Jurkat and PM1 T cells were cultured with different concentrations of IDV for 24 (⋄), 48 (♦), and 72 (×) hours. Cell-cycle progression was determined by propidium iodide staining and is represented as the percentage of cells in S + G2/M phase by using multicycle flow cytometry software. To study the effect of IDV on anti-Fas–induced apoptosis in Jurkat and PM1 T cells, cells were cultured without or with anti-Fas mAb CH11 (100 ng/mL) in presence of different concentrations of IDV, and apoptosis was determined at 2 (▵), 6 (■), and 18 (●) hours. Cells were also precultured with different concentrations of IDV for 18 hours, and apoptosis was induced by culturing these cells with CH11 for 6 (▪) and 18 (●) hours. Data are shown for mean ± SD of the percentage of cells undergoing apoptosis as estimated by propidium iodide staining.

Close modal

IDV does not inhibit caspase activity

The activity of caspases-1, -3, -4, -5, -9, and -8 over 120 minutes in lysates of CH11 mAb-treated Jurkat T cells as demonstrated by release of AFC from caspase-specific fluorogenic substrates is illustrated in Figure 6. IDV at concentrations of 12.5 to 100 μM did not inhibit the activity of any of the caspases, whereas addition of caspase-specific inhibitors resulted in the expected inhibition of caspase activity.

Fig. 6.

Effect of IDV on caspases-1, -3, -4, -5, -8, and -9.

Cell lysates from apoptotic Jurkat T cells were preincubated with different concentrations of IDV (▿, 12.5 μM; ■, 25 μM; ▪, 50 μM; and ♦, 100 μM) or with 100 μM of specific caspase inhibitors (▾) followed by assay for caspase activity as described in the “Materials and methods” section by the release of 7-amino-4-trifluoromethyl coumarin, expressed as relative fluorescence units (RFU), from caspase-specific fluorogenic substrates over a 2-hour period. Controls consisted of wells without lysates (●) and of lysates without inhibitors (○).

Fig. 6.

Effect of IDV on caspases-1, -3, -4, -5, -8, and -9.

Cell lysates from apoptotic Jurkat T cells were preincubated with different concentrations of IDV (▿, 12.5 μM; ■, 25 μM; ▪, 50 μM; and ♦, 100 μM) or with 100 μM of specific caspase inhibitors (▾) followed by assay for caspase activity as described in the “Materials and methods” section by the release of 7-amino-4-trifluoromethyl coumarin, expressed as relative fluorescence units (RFU), from caspase-specific fluorogenic substrates over a 2-hour period. Controls consisted of wells without lysates (●) and of lysates without inhibitors (○).

Close modal

Combination therapies with HIV PIs have had a dramatic effect on the survival of HIV-infected patients.27,28 An intriguing observation is that, in a subset of patients, the increase in CD4 T cells and the decrease in activation status of T cells can be sustained, despite virologic relapse after development of drug resistance.6,7,29 It has recently been suggested that PIs can directly inhibit lymphocyte apoptosis and that this effect may contribute to an immunologic benefit independently of an antiviral effect.17-19 In the present study we demonstrate that the effects of the PI, IDV, are broad ranging and that at therapeutic concentrations IDV can inhibit entry of cells into cell cycle. The drug was found to inhibit cell proliferation but had no direct antiapoptotic activity nor did it inhibit cellular caspases involved in apoptosis. These findings, and the known effects of HIV proteases and HIV PIs on cellular proteases and proteasomes,21,22,30-32 implicate PIs in potentially modulating a wide range of immunologic responses and provide insight into a mechanism for promoting cell survival, without directly influencing cell death.

A novel finding in this study was that IDV blocks cells from entering into cell cycle. This effect was reflected in the inhibition of cell proliferation following activation stimuli. In fact, IDV was found to inhibit 14C-thymidine uptake in cells activated through 3 different pathways: direct T-cell receptor cross-linking using anti-CD3 mAbs, indirect T-cell activation induced by mitogens such as PHA and ConA, as well as activation induced by pharmacologic means using PMA plus Ionomycin. These findings are in agreement with the previously reported antiproliferative effects of IDV reported for myeloblastic and promyelocytic leukemia cell lines.23 The broad-based antiproliferative effect favors the likelihood of inhibition of proteasomal activity,21,22 or modulation of cytokine production.33-36 Proteasomes are the main proteolytic complexes operating in the cytosol and nucleus and are involved in many biological and degradative processes.37,38 The PI, Ritonavir, has been shown to block the chymotrypsin-like activity of murine 20S proteasome.21,22 Proteasomes control the cell cycle by proteolytic degradation of several cell-cycle regulatory proteins such as cyclins, cyclin-dependent kinases, and their inhibitors.37,38 Additionally, PIs have been reported to influence cytokines that regulate lymphocyte proliferation.33-36,39,40 It has been shown that the serum levels of interleukin 16 (IL-16) are increased in patients on IDV-containing drug regimens.35,36 IL-16 is known to inhibit anti-CD3–induced lymphocyte activation and lymphoproliferation.39,40 These observations, together with our findings, suggest that IDV may be blocking cell-cycle progression in lymphocytes, possibly by enhancing the levels of IL-16 production in association with the blocking of proteasomal activity.

Recent clinical investigations, including our own studies and those of several other groups, have shown that following potent antiretroviral therapy a decrease in virus load is accompanied by an increase in CD4 T-cell counts and decreased T-cell apoptosis.10,11,41-43Accelerated lymphocyte apoptosis is a well-known feature of HIV disease.8,9,44,45 The increased apoptosis involving CD4 and CD8 T cells in HIV infection has been explained on the basis of host and viral factors.9,44 Host factors implicated include imbalance in cytokine production and immune activation following HIV infection that results in increased expression of death receptors and ligands.45 Viral factors include viral proteins (gp120, vpr, tat, nef, and protease), all of which have been shown to influence lymphocyte apoptosis in vitro.46-50Thus, the decrease in lymphocyte apoptosis in patients on antiretroviral therapy has been attributed to the rescue of cells from the direct and indirect effects of HIV. Additionally, the PIs themselves have been ascribed to have antiapoptotic properties. Although the activity of PIs is aimed at viral proteases, it has been hypothesized that they can influence host cellular proteases (viz, caspases) because of the similarity shared by these proteases at their active sites. Ritonavir has been reported to inhibit apoptosis in PBMCs cultured from HIV donors,19 from HIV+ donors,17 and in CD34 cells18 and also has been shown to inhibit caspase activity.17-19 In our studies reported herein, the in vitro addition of IDV did not inhibit the spontaneous or anti-CD3–induced apoptosis in PBMCs of healthy volunteers and of HIV-infected children or the apoptosis induced through Fas pathway in Jurkat and PM1 T-cell lines. We also did not find evidence for inhibition of specific caspases involved in the apoptosis cascade. In agreement with these findings, Lu and Andrieu20 have reported that neither T-cell receptor or CD3 ligation nor Fas-triggered apoptosis was affected by IDV in PBMCs from healthy volunteers or HIV-infected donors.

The differences in our findings from the reported inhibition of lymphocyte apoptosis and of caspase activity by PIs17-19can be explained on the basis of the difference in experimental systems and in data interpretation. A major difference is that we used short-term exposure of lymphocytes to IDV (18 hours), whereas others17-19 have used longer culture periods (3-15 days) with Ritonavir prior to lymphocyte activation. Thus, in cells cultured for longer duration with a PI, fewer cells would be cycling because PIs block their entry into cell cycle. It has been shown that cells that have undergone multiple rounds of cell division are more susceptible to apoptosis51 (Chavan et al, unpublished observations, 2000). Thus, cells cultured for prolonged periods with a PI would be expected to exhibit apparently reduced apoptosis as compared with cells cultured in the absence of the PI. These cells would also be expected to show less caspase activity, as reported by Sloand et al17,18 and Weichold et al19 even without a direct effect of the PI on caspases, as shown in our study. The cell-cycle inhibitory effect of PIs could also be a factor responsible for the smaller increase of apoptosis following anti-CD3 stimulation in cells of patients on PI therapy as compared with patients not on PI therapy. Collectively, these studies indicate that IDV may not have a direct effect on apoptosis but may prolong cell survival by inhibition of cell-cycle progression. The difference in the molecular structure of IDV and Ritonavir is a less likely explanation for differential effects on lymphocyte apoptosis. In agreement with this contention we have found that another PI, Nelfinavir, has effects similar to IDV on cell-cycle progression without direct antiapoptotic activity (Chavan et al, unpublished observations, 2000).

The inhibition of cell-cycle progression and antiproliferative property of IDV could affect the host/virus relationship in various ways, as well as modulate immune responses in general. Potential adverse effects of IDV include inhibition of clonal expansion of the cells of the immune system.52 PIs are known to have unexplained toxicities such as abnormal fat distribution and lipodystrophy, insulin-resistant diabetes, and hypercholesteremia.53-56Inhibition of cell-cycle progression by IDV may be a potential factor contributing to the increased differentiation of lipid-producing adipose tissues,53,54 as cell-cycle blocking is essential for differentiation of adipose cells. Interestingly, the effect of IDV on cell cycle was seen only in primary PBMC cultures, and IDV was unable to block continuously cycling cells such as Jurkat and PM1. Thus, inhibition of immune activation may be restricted to newly recruited cells. This property of PIs may in fact be beneficial to the host in terms of host/virus interaction; a decrease in activation of the immune system may prevent the spread of the virus and T-cell–mediated immunopathogenesis in HIV infection. It has been well documented that HIV replication is supported mainly in cycling cells.57,58 HIV-specific proteins such as vpr block the cells in G2 phase that the virus uses to its advantage to increase the synthesis of viral proteins, and thereby increases its replication.59 Blocking of the cells in G0/G1 phase by PIs could result in reduced viral production of wild-type as well as drug-resistant mutant virus strains, thereby contributing to persistent benefit despite development of viral resistance mutations observed in different clinical cohorts.31 Additionally, as reported by Ikezoe et al,23 this property might be useful for treatment of patients with acute promyelocytic leukemia with PIs. Elucidation of mechanisms by which PIs block cell-cycle progression and exert antiproliferative activity will provide insight into the full effect of PI therapy on the immunopathogenesis of HIV disease and open new vistas for disease targets for this class of agents.

Supported by grants AI28281 and DA05161 from the National Institutes of Health, Bethesda, MD.

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 U.S.C. section 1734.

1
Gazzard
B
The picture of future therapy.
Int J Clin Pract Suppl.
103
1999
45
48
2
Clumeck
N
Choosing the best initial therapy for HIV-1 infection [editorial; comment].
N Engl J Med.
341
1999
1925
1926
3
Kiso
Y
Protease inhibitors [editorial].
Biopolymers.
51
1999
1
4
Mitsuyasu
R
HIV protease inhibitors: immunological insights.
AIDS.
13(suppl 1)
1999
S19
S27
5
Pradier
C
Pesce
A
Carrieri
P
et al
Effect of indinavir and higher CD4+ T-cell count on viral load response after 6 months of highly active antiretroviral therapy.
Clin Ther.
21
1999
1313
1320
6
Mezzaroma
I
Carlesimo
M
Pinter
E
et al
Clinical and immunologic response without decrease in virus load in patients with AIDS after 24 months of highly active antiretroviral therapy.
Clin Infect Dis.
29
1999
1423
1430
7
Belec
L
Piketty
C
Si-Mohamed
A
et al
High levels of drug-resistant human immunodeficiency virus variants in patients exhibiting increasing CD4+ T cell counts despite virologic failure of protease inhibitor-containing antiretroviral combination therapy.
J Infect Dis.
181
2000
1808
1812
8
Famularo
G
De Simone
C
Marcellini
S
Apoptosis: mechanisms and relation to AIDS.
Med Hypotheses.
48
1997
423
429
9
Cotton
MF
Cassella
C
Rapaport
EL
Tseng
PO
Marschner
S
Finkel
TH
Apoptosis in HIV-1 infection.
Behring Inst Mitt.
1996
220
231
10
Chavan
SJ
Tamma
SL
Kaplan
M
Gersten
M
Pahwa
SG
Reduction in T cell apoptosis in patients with HIV disease following antiretroviral therapy.
Clin Immunol.
93
1999
24
33
11
Aries
SP
Weyrich
K
Schaaf
B
Hansen
F
Dennin
RH
Dalhoff
K
Early T-cell apoptosis and Fas expression during antiretroviral therapy in individuals infected with human immunodeficiency virus-1.
Scand J Immunol.
48
1998
86
91
12
Phenix
BN
Angel
JB
Mandy
F
et al
Decreased HIV-associated T cell apoptosis by HIV protease inhibitors.
AIDS Res Hum Retroviruses.
16
2000
559
567
13
Gulnik
S
Erickson
JW
Xie
D
HIV protease: enzyme function and drug resistance.
Vitam Horm.
58
2000
213
256
14
Navia
MA
McKeever
BM
A role for the aspartyl protease from the human immunodeficiency virus type 1 (HIV-1) in the orchestration of virus assembly.
Ann N Y Acad Sci.
616
1990
73
85
15
Stennicke
HR
Salvesen
GS
Catalytic properties of the caspases.
Cell Death Differ.
6
1999
1054
1059
16
Popov
ME
Kashparov
IV
Rumsh
LD
Popov
EM
Mechanism of action of aspartic proteases, III: conformational characteristics of HIV-1 protease inhibitor JG-365.
Bioorg Khim.
25
1999
418
422
17
Sloand
EM
Kumar
PN
Kim
S
Chaudhuri
A
Weichold
FF
Young
NS
Human immunodeficiency virus type 1 protease inhibitor modulates activation of peripheral blood CD4(+) T cells and decreases their susceptibility to apoptosis in vitro and in vivo.
Blood.
94
1999
1021
1027
18
Sloand
EM
Maciejewski
J
Kumar
P
Kim
S
Chaudhuri
A
Young
N
Protease inhibitors stimulate hematopoiesis and decrease apoptosis and ICE expression in CD34(+) cells.
Blood.
96
2000
2735
2739
19
Weichold
FF
Bryant
JL
Pati
S
Barabitskaya
O
Gallo
RC
Reitz
MS
Jr
HIV-1 protease inhibitor ritonavir modulates susceptibility to apoptosis of uninfected T cells.
J Hum Virol.
2
1999
261
269
20
Lu
W
Andrieu
JM
HIV protease inhibitors restore impaired T-cell proliferative response in vivo and in vitro: a viral-suppression-independent mechanism.
Blood.
96
2000
250
258
21
Andre
P
Groettrup
M
Klenerman
P
et al
An inhibitor of HIV-1 protease modulates proteasome activity, antigen presentation, and T cell responses.
Proc Natl Acad Sci U S A.
95
1998
13120
13124
22
Schmidtke
G
Holzhutter
HG
Bogyo
M
et al
How an inhibitor of the HIV-I protease modulates proteasome activity.
J Biol Chem.
274
1999
35734
35740
23
Ikezoe
T
Daar
ES
Hisatake
JI
Taguchi
H
Koeffler
HP
HIV-1 protease inhibitors decrease proliferation and induce differentiation of human myelocytic leukemia cells.
Blood.
96
2000
3553
3559
24
Lusso
P
Cocchi
F
Balotta
C
et al
Growth of macrophage-tropic and primary human immunodeficiency virus type 1 (HIV-1) isolates in a unique CD4+ T-cell clone (PM1): failure to downregulate CD4 and to interfere with cell-line-tropic HIV-1.
J Virol.
69
1995
3712
3720
25
McCloskey
TW
Chavan
S
Lakshmi Tamma
SM
Pahwa
S
Comparison of seven quantitative assays to assess lymphocyte cell death during HIV infection: measurement of induced apoptosis in anti-Fas-treated Jurkat cells and spontaneous apoptosis in peripheral blood mononuclear cells from children infected with HIV.
AIDS Res Hum Retroviruses.
14
1998
1413
1422
26
Bluming
AZ
Lynch
MJ
Kavanah
M
Khiroya
R
Transformation antigens on stimulated lymphocytes.
J Immunol.
114
1975
717
721
27
Rizzardi
GP
Pantaleo
G
Therapeutic perspectives in HIV-1 infection from recent advances in HIV-1 pathogenesis: it is time to move on.
J Biol Regul Homeost Agents.
13
1999
151
157
28
Bright
DC
Antiretroviral therapy for HIV infection: a review of current medications and therapies.
J Am Optom Assoc.
70
1999
355
383
29
Lempicki
RA
Kovacs
JA
Baseler
MW
et al
Impact of HIV-1 infection and highly active antiretroviral therapy on the kinetics of CD4+ and CD8+ T cell turnover in HIV-infected patients.
Proc Natl Acad Sci U S A.
97
2000
13778
13783
30
Snasel
J
Pichova
I
The cleavage of host cell proteins by HIV-1 protease.
Folia Biol (Praha).
42
1996
227
230
31
Korant
BD
Strack
P
Frey
MW
Rizzo
CJ
A cellular anti-apoptosis protein is cleaved by the HIV-1 protease.
Adv Exp Med Biol.
436
1998
27
29
32
Zhang
D
Zhang
N
Wick
MM
Byrn
RA
HIV type 1 protease activation of NF-kappa B within T lymphoid cells.
AIDS Res Hum Retroviruses.
11
1995
223
230
33
Imami
N
Antonopoulos
C
Hardy
GA
Gazzard
B
Gotch
FM
Assessment of type 1 and type 2 cytokines in HIV type 1-infected individuals: impact of highly active antiretroviral therapy.
AIDS Res Hum Retroviruses.
20
15
1999
1499
1508
34
Stylianou
E
Aukrust
P
Kvale
D
Muller
F
Froland
SS
IL-10 in HIV infection: increasing serum IL-10 levels with disease progression—down-regulatory effect of potent anti-retroviral therapy.
Clin Exp Immunol.
116
1999
115
120
35
De Paoli
P
Zanussi
S
Caggiari
L
et al
Kinetics of lymphokine production in HIV+ patients treated with highly active antiretroviral therapy and interleukin 2.
J Clin Immunol.
19
1999
317
325
36
Bisset
LR
Rothen
M
Joller-Jemelka
HI
Dubs
RW
Grob
PJ
Opravil
M
Change in circulating levels of the chemokines macrophage inflammatory proteins 1 alpha and 11 beta, RANTES, monocyte chemotactic protein-1 and interleukin-16 following treatment of severely immunodeficient HIV-infected individuals with indinavir.
AIDS.
11
1997
485
491
37
Kornitzer
D
Ciechanover
A
Modes of regulation of ubiquitin-mediated protein degradation.
J Cell Physiol.
182
2000
1
11
38
Hershko
A
Roles of ubiquitin-mediated proteolysis in cell-cycle control.
Curr Opin Cell Biol.
9
1997
788
799
39
Ogasawara
H
Takeda-Hirokawa
N
Sekigawa
I
Hashimoto
H
Kaneko
Y
Hirose
S
Inhibitory effect of interleukin-16 on interleukin-2 production by CD4+ T cells.
Immunology.
96
1999
215
219
40
Cruikshank
WW
Lim
K
Theodore
AC
et al
IL-16 inhibition of CD3-dependent lymphocyte activation and proliferation.
J Immunol.
157
1996
5240
5248
41
Roger
PM
Breittmayer
JP
Arlotto
C
et al
Highly active anti-retroviral therapy (HAART) is associated with a lower level of CD4+ T cell apoptosis in HIV-infected patients.
Clin Exp Immunol.
118
1999
412
416
42
Hazenberg
MD
Stuart
JW
Otto
SA
et al
T-cell division in human immunodeficiency virus (HIV)-1 infection is mainly due to immune activation: a longitudinal analysis in patients before and during highly active antiretroviral therapy (HAART).
Blood.
95
2000
249
255
43
Gougeon
ML
Montagnier
L
Programmed cell death as a mechanism of CD4 and CD8 T cell deletion in AIDS: molecular control and effect of highly active anti-retroviral therapy.
Ann N Y Acad Sci.
887
1999
199
212
44
Ahsan
N
Langhoff
E
Immunopathogenesis of human immunodeficiency virus.
Semin Nephrol.
18
1998
422
435
45
Peter
ME
Ehret
A
Berndt
C
Krammer
PH
AIDS and the death receptors.
Br Med Bull.
53
1997
604
616
46
Chirmule
N
Pahwa
S
Envelope glycoproteins of human immunodeficiency virus type 1: profound influences on immune functions.
Microbiol Rev.
60
1996
386
406
47
Shostak
LD
Ludlow
J
Fisk
J
et al
Roles of p53 and caspases in the induction of cell cycle arrest and apoptosis by HIV-1 vpr.
Exp Cell Res.
251
1999
156
165
48
McCloskey
TW
Ott
M
Tribble
E
et al
Dual role of HIV Tat in regulation of apoptosis in T cells.
J Immunol.
158
1997
1014
1019
49
Zauli
G
Gibellini
D
Secchiero
P
et al
Human immunodeficiency virus type 1 Nef protein sensitizes CD4(+) T lymphoid cells to apoptosis via functional upregulation of the CD95/CD95 ligand pathway.
Blood.
93
1999
1000
1010
50
Korant
BD
Strack
P
Frey
MW
Rizzo
CJ
A cellular anti-apoptosis protein is cleaved by the HIV-1 protease.
Adv Exp Med Biol.
436
1998
27
29
51
Renno
T
Attinger
A
Locatelli
S
Bakker
T
Vacheron
S
MacDonald
HR
Cutting edge: apoptosis of superantigen-activated T cells occurs preferentially after a discrete number of cell divisions in vivo.
J Immunol.
162
1999
6312
6315
52
Halloran
PF
Molecular mechanisms of new immunosuppressants.
Clin Transplant.
10
1996
118
123
53
Wentworth
JM
Burris
TP
Chatterjee
VK
HIV protease inhibitors block human preadipocyte differentiation, but not via the PPARgamma/RXR heterodimer.
J Endocrinol.
164
2000
R7
R10
54
Berthold
HK
Parhofer
KG
Ritter
MM
et al
Influence of protease inhibitor therapy on lipoprotein metabolism.
J Intern Med.
246
1999
567
575
55
Safrin
S
Grunfeld
C
Fat distribution and metabolic changes in patients with HIV infection [editorial].
AIDS.
13
1999
2493
2505
56
Mathe
G
Human obesity and thinness, hyperlipidemia, hyperglycemia, and insulin resistance associated with HIV1 protease inhibitors: prevention by alternating several antiproteases in short sequences [editorial].
Biomed Pharmacother.
53
1999
449
451
57
Gummuluru
S
Emerman
M
Cell cycle- and Vpr-mediated regulation of human immunodeficiency virus type 1 expression in primary and transformed T-cell lines.
J Virol.
73
1999
5422
5430
58
Dornadula
G
Zhang
H
Shetty
S
Pomerantz
RJ
HIV-1 virions produced from replicating peripheral blood lymphocytes are more infectious than those from nonproliferating macrophages due to higher levels of intravirion reverse transcripts: implications for pathogenesis and transmission.
Virology.
253
1999
10
16
59
Goh
WC
Rogel
ME
Kinsey
CM
et al
HIV-1 Vpr increases viral expression by manipulation of the cell cycle: a mechanism for selection of Vpr in vivo.
Nat Med.
4
1998
65
71

Author notes

Savita Pahwa, Division of Allergy/Immunology, North Shore University Hospital, NYU School of Medicine, 350 Community Dr, Research Bldg #303, Manhasset, NY 11030; e-mail: spahwa@nshs.edu.

Sign in via your Institution