Human Immunodeficiency Virus Type 1 ... - Journal of Virology

0 downloads 0 Views 283KB Size Report
Jun 17, 2004 - Schools of Dentistry1 and Medicine,2 University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Received ... ml); and targeted viral entry through high-affinity interactions ...... well-Wild, H. Hale-Donze, G. McGrady, X. Y. Song, and S. M. Wahl. .... Jin, F. Y., C. Nathan, D. Radzioch, and A. Ding.
JOURNAL OF VIROLOGY, May 2005, p. 6432–6440 0022-538X/05/$08.00⫹0 doi:10.1128/JVI.79.10.6432–6440.2005 Copyright © 2005, American Society for Microbiology. All Rights Reserved.

Vol. 79, No. 10

Human Immunodeficiency Virus Type 1 Stimulates the Expression and Production of Secretory Leukocyte Protease Inhibitor (SLPI) in Oral Epithelial Cells: a Role for SLPI in Innate Mucosal Immunity N. K. Jana,1 L. R. Gray,2 and D. C. Shugars1,2* Schools of Dentistry1 and Medicine,2 University of North Carolina at Chapel Hill, Chapel Hill, North Carolina Received 17 June 2004/Accepted 5 January 2005

The innate immune response is a key barrier against pathogenic microorganisms such as human immunodeficiency virus type 1 (HIV-1). Because HIV-1 is rarely transmitted orally, we hypothesized that oral epithelial cells participate in the innate immune defense against this virus. We further hypothesized that secretory leukocyte protease inhibitor (SLPI), a 12-kDa mucosal antiviral protein, is a component of the host immune response to this virus. Here we demonstrated constitutive expression and production of SLPI in immortalized human oral keratinocytes. Brief exposure of cells to HIV-1 BaL and HXB2 significantly increased SLPI mRNA and protein production compared to that in mock-exposed cells (P < 0.01), as evaluated by real-time quantitative reverse transcription-PCR and enzyme-linked immunosorbent assay. HIV-1-mediated stimulation of SLPI occurred at the transcriptional level, was dose and time dependent, was elicited by heat-inactivated and infectious viruses, and did not depend on cellular infection. Experiments with purified retroviral proteins showed that the stimulatory effect was induced specifically by external envelope glycoproteins from HIV-1 and simian immunodeficiency virus. SLPI responsiveness to HIV-1 was also observed in an unrelated oral epithelial cell line and in normal (nonimmortalized) human oral epithelial cells isolated from healthy uninfected gingival tissues. In this first report of SLPI regulation by HIV-1, we show that the expression and production of the antimicrobial and anti-inflammatory protein can be stimulated in oral epithelial cells by the virus through interactions with gp120 in the absence of direct infection. These findings indicate that SLPI is a component of the oral mucosal response to HIV-1. promoting fertility (39); and enhancing wound healing in skin (4, 62). SLPI anti-HIV-1 activity was first described by McNeely et al. (33) in studies demonstrating that saliva-derived and recombinant forms of SLPI protected human monocyte-derived macrophages and CD4⫹ T cells against infection with HIV-1 BaL. Inhibition was dose, pH, and temperature dependent; occurred at physiological concentrations in saliva (1 to 10 ␮g/ ml); and targeted viral entry through high-affinity interactions with a cell surface molecule (33, 34, 58) recently identified as annexin II (30). Reports of no or inconsistent inhibition by two research groups (25, 55) prompted a reexamination of SLPI antiviral activity. SLPI inhibition in vitro was subsequently confirmed by several groups (19, 48, 51), including one group that initially reported negative findings (47) with various cell culture models and HIV-1 isolates having different subtypes and chemokine coreceptor usage patterns. Additional evidence for SLPI antiviral activity came from African studies of perinatal HIV-1 transmission in which high transmission rates were associated with low levels (below 0.1 ␮g/ml, the MIC required in vitro) (33) of vaginal SLPI in infected mothers (40) and salivary SLPI in breast-fed babies (11). Thus, SLPI does indeed possess anti-HIV-1 properties and likely plays a key role in protecting mucosal surfaces against viral infection. Little is known of SLPI function and regulation in oral tissues. Most studies have been conducted with cells and tissues derived from the lungs (1–3, 21, 32, 43, 44, 57), the reproductive tract (10, 39, 60), and skin (4, 52, 62). These studies have revealed that the human SLPI gene is nonpolymorphic (23) and is expressed in a tissue- and mucosa-specific

Oral tissues are naturally resistant to infection with human immunodeficiency virus type 1 (HIV-1), unlike genital and anorectal tissues (7). The mechanisms contributing to the protective effect in the oral cavity are incompletely understood. The innate immune response is a key defense against HIV-1, particularly at mucosal surfaces (27). Mucosal epithelium participates in this response by providing a physical barrier to infection and producing anti-HIV-1 peptides and proteins that serve as key effector molecules. Oral antimicrobial factors having anti-HIV-1 activity at physiologic concentrations include secretory leukocyte protease inhibitor (SLPI), human beta defensins (hBDs), salivary agglutinin, thrombospondin 1, and mucins (33, 42, 49). Knowledge gained by studying the oral innate defense against this virus will shed light on new strategies for reducing transmission at genital and anorectal sites, where most infections occur in vivo. SLPI, a member of the trappin gene family that includes elafin (45), is a 107-kDa protein produced and secreted primarily from epithelial cells lining mucosal surfaces (12) and skin (52), neutrophils (44), and lipopolysaccharide-stimulated macrophages (22). Originally described for its activity against serine proteases (e.g., leukocyte elastase and cathepsin G) (53), SLPI participates in the mucosal defense by reducing inflammation (16); suppressing matrix metalloproteinase production and activity (61); blocking the in vitro growth of selected bacteria (17), fungi (54), and non-HIV-1 viruses (5); * Corresponding author. Mailing address: 311 Dental Research Center, CB 7455, Chapel Hill, NC 27599-7455. Phone: (919) 966-5310. Fax: (919) 966-6761. E-mail: [email protected]. 6432

VOL. 79, 2005

HIV-1 STIMULATION OF SLPI IN ORAL EPITHELIAL CELLS

manner (3). Regulation of the antiprotease activity occurs primarily at the transcriptional level, although gene expression can also be modulated posttranscriptionally (3, 32). Treatment of cells with the proinflammatory mediators interleukin 1␤ and tumor necrosis factor alpha (24, 43, 50, 52), phorbol ester (32), neutrophil elastase (1, 57), corticosteroids (2), and progesterone (24) increased SLPI mRNA and protein production. SLPI expression was also induced by lipopolysaccharide and lipoteichoic acid in macrophages (22) and by insulin-like growth factor I and transforming growth factor alpha in skin keratinocytes (52). Studies of SLPI expression in oral tissues have been limited to the major and minor salivary glands, where SLPI mRNA and protein production is localized to serous and ductal epithelial cells (12, 37, 59). In salivary glands, inhibitor production is anatomically distinct from areas of active HIV-1 replication in the stroma (59). SLPI expression in other tissues of the oral cavity has not been reported. Because HIV-1 transmission rarely occurs through the oral route, we hypothesized that oral keratinocytes participate in the innate immune response to the virus by stimulating local SLPI production and secretion. In this study, we used reverse transcription (RT)-PCR and immunohistochemical analyses to demonstrate constitutive expression of SLPI in oral epithelial cells. Real-time quantitative RT-PCR (QRT-PCR) and enzyme-linked immunosorbent assay (ELISA) showed that SLPI mRNA production and protein production were increased 100-fold and 3-fold, respectively, after brief contact of oral epithelial cells with HIV-1 compared to controls. The stimulatory effect occurred in a dose- and time-dependent manner, was controlled at the transcriptional level, did not require cellular infection, and was mediated through interactions with viral external envelope glycoprotein gp120. The effect was observed in two immortalized oral keratinocyte cell lines and in normal (nonimmortalized) oral keratinocytes. HIV-1-mediated regulation of SLPI represents a novel protective mechanism in the oral cavity and suggests unique strategies for reducing HIV-1 infection at nonoral mucosal sites. (This work was presented by D. C. Shugars and N. K. Jana in part at the 82nd General Section of the International Association of Dental Research, Honolulu, Hawaii, 10 to 13 March 2004 [abstr. 3854].) MATERIALS AND METHODS Cells. The human oral epithelial cell line GMSM-K (a generous gift from V. A. Murrah and E. P. Gilchrist, University of North Carolina School of Dentistry) was derived from normal oral buccal mucosa of a stillborn fetus and immortalized with simian virus 40 T antigen (14). GMSM-K cells exhibit differentiation markers characteristic of normal keratinocytes, including the expression of lowand high-molecular-weight keratins, squamous cell-specific keratin 14, and epithelial cell-specific antigen Ber-EP4. As determined by immunoblotting and immunohistochemical analysis, GMSM-K cells no longer express the T antigen (14). Cells were grown in keratinocyte serum-free medium (SFM) with supplements (epidermal growth factor, bovine pituitary extract, and 1% penicillinstreptomycin) (Invitrogen, Carlsbad, CA) (growth medium). Normal human gingival epithelial (HGE) cells were isolated from uninflamed gingival tissues overlying partially impacted third molars (26). Tissue was obtained from non-HIV-1-infected healthy donors according to procedures recommended by the University of North Carolina Institutional Review Board. Briefly, excised tissues were rinsed twice in phosphate-buffered saline (PBS) containing 1% penicillin-streptomycin and 1 mg/ml amphotericin B (all from Invitrogen) (PBS⫹), cut into small blocks (2 mm by 2 mm), and incubated overnight at 4°C with dipase (6 mg/ml; Becton Dickinson, Franklin Lakes, NJ) in PBS⫹. Single-

6433

cell suspensions were generated by trypsinizing the epithelial layer and plating in T-25 flasks containing growth medium further supplemented with hydrocortisone, bovine insulin, gentamicin sulfate, amphotericin B, and 0.15 mM CaCl2 (Invitrogen) (growth⫹ medium). Cells were grown to near confluence, passaged once, and cryopreserved (5 ⫻ 105 cells/vial) by a standard procedure. Frozen HGE cells were thawed and cultured in growth⫹ medium without further passage prior to use. HGE cultures contained primarily (⬎98%) oral epithelial cells based on morphology determined by light microscopy. The human H9 T-cell line was obtained from the AIDS Research and Reference Reagent Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (31). H9 cells were grown in RPMI 1640 medium with 2 mM L-glutamine, 1% penicillin-streptomycin, and 10% fetal bovine serum. OKF6/TERT-1 cells, hTERT-immortalized human oral epithelial cells (8), were obtained from J. G. Rheinwald (Harvard Institutes of Medicine, Boston, MA). The cells were propagated in growth⫹ medium further supplemented with 0.4 mM calcium chloride. Viruses and recombinant viral proteins. The following reagents were obtained through the AIDS Research and Reference Reagent Program: HIV-1 BaL (13) and recombinant HIV-1 proteins LAV p55 Gag, Rev (38), NL4-3 integrase (F185H/C280S; from Robert Craigie), HXB2 reverse transcriptase p51 (46), simian immunodeficiency virus (SIV) mac239 gp130 (18), and BaL gp120. HIV-1 HXB2 was purchased from Advanced Biotechnologies (Columbia, MD). Recombinant gp120 from HIV-1 MN and IIIB was obtained from ImmunoDiagnostics (Woburn, MA). All recombinant proteins were generated in bacteria and purified according to standard protocols (13, 18, 38, 46). Cell-free virus stocks were prepared and titers were determined with phytohemagglutinin-stimulated human peripheral blood mononuclear cells (PBMCs) by standard procedures (9). RT-PCR analysis. Total RNA (2 ␮g) was purified from cell lysates (RNeasy; Qiagen, Valencia, CA) and subjected to RT-PCR amplification (Titan One Tube RT-PCR system; Roche, Branchburg, NJ) in 35 cycles with published PCR primers specific for SLPI (50) or the ␤-actin housekeeping gene (Promega, Madison, WI). Controls included reactions lacking RT or template RNA. RTPCR products were separated by 2% agarose gel electrophoresis, visualized by ethidium bromide staining, and verified by sequencing. Immunocytochemical analysis. Unstimulated GMSM-K cells were cytospun onto ProbeOnPlus glass microscopic slides (Fisher Scientific, Pittsburgh, PA), air dried, fixed with methanol-acetone, permeabilized with PBS containing 0.05% Triton X-100 (Sigma-Aldrich, St. Louis, MO), and blocked in 15-min roomtemperature incubations sequentially with peroxide (3% in PBS; Sigma-Aldrich) avidin (Vector, Burlingame, CA), biotin (Vector), and Power Block (BioGenex, San Ramon, CA). Cells were incubated for 1 h at 37°C with goat anti-human SLPI polyclonal antibody (R & D Systems, Minneapolis, MN) diluted 1:10 in common antibody diluent (BioGenex), washed four times with automation buffer (Biomeda, Foster City, CA), and incubated for 20 min at 37°C with biotinylated donkey anti-goat secondary antibody diluted 1:2,500 in PBS containing 0.05% Tween 20 (Jackson ImmunoResearch, West Grove, PA). Cells were washed with automation buffer and treated with ABC Elite reagents and NovaRed peroxidase substrate (both from Vector) to identify SLPI-producing cells. Cell preparations were immunostained with the secondary antibody only (no primary antibody) to assess background staining. SLPI stimulation assay. In triplicate, cells (105 cells/well in a 24-well tissue culture dish) were plated in growth medium. When confluence reached 80 to 90%, the cells were washed with PBS and incubated with 1 ml of SFM for 3 h to minimize the potential effects of epidermal growth factor and pituitary extract on SLPI expression. After the medium was removed, cell-free virus (multiplicity of infection [MOI] of 0.1 in 200 ␮l SFM) or purified recombinant protein (200 ng/200 ␮l SFM) was added to the wells and incubated for 30 min at 37°C. Because the virus stocks and recombinant proteins were diluted a minimum of 1:30 in SFM to obtain the desired concentrations, mock cultures were treated with SFM only (200 ␮l/well). After the incubation period, growth medium (1 ml) was added, and the cells were incubated for 4 days without a medium change. Every 24 h, an aliquot of culture supernatant was removed and analyzed for SLPI protein content by an ELISA (R & D Systems). At 96 h, the cells were washed and either centrifuged onto glass slides for immunocytochemical evaluation or harvested in RNAlater (Ambion, Austin, TX) for RT-PCR-based analyses. Cell viability was monitored by trypan blue dye exclusion at the end of each experiment. Results were verified in at least two independent experiments for each treatment condition. QRT-PCR analysis. SLPI mRNA in cell lysates was quantitated by real-time RT-PCR with TaqMan technology (Roche). TaqMan-specific SLPI forward (5⬘GCTGTGGAAGGCTCTGGAAAGT-3⬘) and reverse (5⬘-GCATTTGATGCC

6434

JANA ET AL.

J. VIROL. ACAAGTGTCA-3⬘) primers and a 5⬘-VIC (6-carboxyrhodamine)/3⬘-minor groove binding-labeled SLPI probe (5⬘-AGAAACCTGAGTGCCAGAGT-3⬘) were designed by using Primer Express computer software (Applied Biosystems) to span across introns and prevent amplification of contaminating genomic DNA. TaqMan-specific primers and a 6-carboxyfluorescein-labeled probe for ␤-actin were obtained from Applied Biosystems. Briefly, first-strand cDNA was generated from purified RNA with RT (SuperScript II reverse transcriptase; Invitrogen) and the TaqMan-specific SLPI reverse primer. The cDNA was amplified by real-time PCR with an ABI prism 7000 PCR and detection instrument (Applied Biosystems, Foster City, CA). Each 50-␮l reaction mixture contained 10 ng of cDNA, 25 ␮l of 2⫻ TaqMan universal PCR master mix with UNG AmpErase (final concentrations of 3 mM MgCl2, 0.2 mM each deoxynucleoside triphosphate, and 1.25 U of Amplitaq Gold), 200 nM each probe, and 300 nM each primer (all from Applied Biosystems). After a 2-min incubation at 50°C and a 10-min incubation at 95°C, reaction mixtures were subjected to 40 amplification cycles of denaturation (95°C for 15 s) and annealing/extension (60°C for 1 min). Data were acquired during the annealing/extension phase and analyzed by using the SDS program (Applied Biosystems). The concentrations of SLPI primers and probe in the PCR mixtures were optimized and validated by using serial dilutions of total RNA from unstimulated GMSM-K cells. A control PCR containing the SLPI primers (but excluding the probe) were run under identical conditions, and the products were evaluated by 2% agarose gel electrophoresis to confirm the amplification of a single 60-bp DNA fragment (data not shown). The fold change in SLPI expression over time was determined by comparing the quantity of SLPI RNA to that of ␤-actin RNA (internal control) for each reaction. Statistical analysis. SLPI concentrations (␮g/ml) were expressed as the mean ⫾ standard error (SE) and analyzed with regard to treatment conditions by using Statview statistical analysis software (Abacus Concepts). Differences in test conditions were evaluated by using the unpaired Student t test (significance was set at a P value of ⬍0.05).

RESULTS Human oral keratinocytes constitutively express SLPI. The first cells that HIV-1 likely encounters in the mouth are epithelial cells lining intraoral mucosal surfaces. To date, studies of SLPI gene expression in the oral cavity have been limited to salivary glands (37, 59). To determine whether oral epithelial cells also express the antiprotease, we analyzed SLPI expression and production in transformed GMSM-K oral keratinocytes and normal human gingival cells established from healthy gingival tissues of non-HIV-1-infected donors. As shown in Fig. 1A (top gel), the SLPI gene was constitutively expressed in unstimulated GMSM-K and HGE cells but not in the negative control H9 T cells. RT-PCR specificity was verified in reactions lacking RT or RNA and by sequencing the SLPI product generated from GMSM-K cells. Amplification of the housekeeping ␤-actin gene confirmed the RNA integrity in the reactions (Fig. 1A, bottom gel). Immunohistochemical staining with an anti-SLPI antibody demonstrated a punctate cytoplasmic pattern of SLPI production consistent with protein secretion (Fig. 1B). As determined by an ELISA, SLPI levels in the medium of unstimulated triplicate cultures of GMSM-K cells were (without a medium change) 63.4 ⫾ 3.8 pg/ml at 24 h, 139.3 ⫾ 0.9 pg/ml at 48 h, 427.0 ⫾ 11.8 pg/ml at 72 h, and

FIG. 1. SLPI is constitutively expressed in oral keratinocytes. (A) Total RNA from unstimulated GMSM-K (lanes 1 to 3), HGE (lanes 4 to 6; positive control), and H9 (lanes 7 to 9; negative control) cells was analyzed by RT-PCR with primers for human SLPI (top gel) or ␤-actin (bottom gel). Reaction mixtures containing no RNA and no RT served

as additional negative controls. The sizes of molecular mass markers (in base pairs) are indicated on the left of each gel. (B) Unstimulated GMSM-K cells were cytospun onto glass slides and immunostained with anti-SLPI polyclonal antibody and secondary detection antibody (SLPI-stained cells; magnification, ⫻200). Cells were reacted with the secondary antibody alone to evaluate background staining (negative control).

VOL. 79, 2005

HIV-1 STIMULATION OF SLPI IN ORAL EPITHELIAL CELLS

FIG. 2. HIV-1 stimulates SLPI production in GMSM-K cells. Triplicate cultures of GMSM-K cells were incubated with HIV-1 BaL (■), HIV-1 HXB2 (Œ), or SFM only ({) for 30 min at 37°C. Growth medium was added, and the cells were incubated for 4 days. At 24-h intervals, culture supernatants were collected and analyzed for SLPI content by an ELISA. Data are presented as the mean ⫾ SE for each time point.

1,216.1 ⫾ 17.7 pg/ml at 96 h. SLPI production in the cells was confirmed by immunoblotting (data not shown). SLPI expression and secretion in GMSM-K cells increase in response to HIV-1. To evaluate whether exposure to HIV-1 affected SLPI mRNA and protein production in oral epithelial cells, GMSM-K cells were incubated with R5 (CCR5 utilizing) HIV-1 BaL, X4 (CXCR4 utilizing) HIV-1 HXB2, or SFM only (mock) for 30 min at 37°C and then cultured for 4 days without a medium change. Because the virus stocks (generated in phytohemagglutinin-stimulated PBMCs) were diluted at least 1:30 in SFM to obtain the desired MOI, control cells were treated with SFM rather than supernatant from stimulated PBMCs. At 24-h intervals, culture supernatants were collected and analyzed for secreted SLPI by an ELISA. As shown in Fig. 2, SLPI production significantly increased threefold at 48, 72, and 96 h after treatment in virus-exposed cells compared to mock-exposed cells (P ⬍ 0.05). Cells treated with the R5 and X4 viruses showed equivalent SLPI levels (P ⬎ 0.05), suggesting that the stimulatory effect was independent of chemokine coreceptor usage. The effect was not due to a stimulation of cellular proliferation by HIV-1, as virus-treated and mock cultures exhibited similar growth kinetics (data not shown). Cell viability remained greater than 95% throughout the culture period. No cellular infection was detected under these experimental conditions, as assessed with a PCR-based assay to detect inte-

6435

grated proviral DNA and viral p24 quantitation of culture supernatants (unpublished data). Expression of SLPI mRNA after HIV-1 exposure in GMSMK cells. SLPI mRNA expression in oral keratinocytes in response to HIV-1 was examined by real-time QRT-PCR. Here, GMSM-K cells were incubated with BaL for various durations prior to RNA isolation, RT, and quantitative PCR amplification. Change in SLPI expression over time was measured by comparing the fold change in SLPI cDNA relative to ␤-actin cDNA (internal control) for each lysate. Compared to mocktreated cells, SLPI mRNA level rapidly increased 50-fold at 1 h after virus addition, peaked at a 100-fold level at 2 h after virus addition, and was subsequently maintained at an approximately 60-fold-higher level for the remainder of the observation period (Fig. 3). These results indicate that virus-cell contact leads to a rapid and sustained increase in SLPI expression and that the stimulatory effect occurs at the transcriptional level. HIV-1-induced SLPI stimulation in oral epithelial cells is dose dependent, does not require infectious virus, and is induced by brief exposure to virus. To further characterize the effect, the SLPI stimulation assay was repeated under various conditions. In one experiment, GMSM-K cells were incubated with various concentrations of BaL (serial dilutions; MOIs from 1.0 to 0.0075), infectious BaL (MOI, 0.1), BaL heat inactivated for 1 h at 56°C (MOI, 0.1), or SFM only for 30 min at 37°C prior to the addition of growth medium. For clarity, results from the dose-response conditions are presented in Fig. 4A, while results from the infectious/heat-inactivated conditions are presented in Fig. 4B. The mock treatment data are reproduced in Fig. 4B for comparison. Significant dose-dependent elevations in SLPI secretion were first detected at 24 h after virus exposure in cells treated with MOIs of 0.0075, 0.03, 0.1, and 1.0 compared to controls (Fig. 4A) (P ⬍ 0.05). At 48, 72, and 96 h after exposure, all virus-treated cultures exhibited

FIG. 3. Real-time QRT-PCR analysis. In triplicate, GMSM-K cells were incubated with HIV-1 BaL for either 0 min (negative control) or 30 min at 37°C prior to the addition of growth medium. Total RNA was purified from cell lysates collected at the indicated times and analyzed by real-time QRT-PCR. The change in SLPI expression after HIV-1 exposure was measured by comparing the quantity of SLPI RNA to that of ␤-actin RNA (internal control) for each lysate. Data are presented as the mean ⫾ SE fold change in SLPI RNA relative to ␤-actin RNA over time.

6436

JANA ET AL.

J. VIROL.

FIG. 4. SLPI secretion in GMSM-K cells exposed to HIV-1 BaL at various concentrations and under various conditions. (A) Triplicate cultures of cells were incubated for 30 min with serial dilutions of BaL or SFM only (MOI, 0) prior to 4-day culture and assayed for SLPI. (B) Triplicate cultures were incubated with heat-inactivated BaL (■; MOI, 0.1), infectious BaL (䊐; MOI, 1.0), or SFM only ({; MOI, 0) (B) prior to the addition of growth medium. Culture supernatants were collected at 24-h intervals after virus exposure and tested for SLPI. The SFM-only control data shown in panel B are the same as the data shown in panel A and are reproduced in panel B for comparison. Data are presented as the mean ⫾ SE.

significantly higher SLPI levels than controls (P ⬍ 0.05). Similar results were observed in cells exposed to serial dilutions of HIV-1 HXB2 (data not shown). As shown in Fig. 4B, SLPI production was similar in cells treated with infectious or heatinactivated virus, indicating that infectious virus is not essential for SLPI stimulation. This finding suggests that a component of the virus particle is responsible for enhanced SLPI secretion. To determine the minimum stimulation period required for the effect, the stimulation assay was repeated using GMSM-K cells incubated with infectious BaL for 0, 10, 20, or 30 min and then washed to remove unbound virus prior to medium addition. Culture supernatants were collected 96 h later and assayed for SLPI content. An increase in SLPI secretion was detected after virus exposures of 10 min or longer compared to controls (Fig. 5A). To determine whether shorter periods of virus contact can also induce SLPI production, the assay was repeated using cells incubated with virus for 0, 1, or 5 min prior

to virus removal. Incubation of cells with BaL for 5 min but not 1 min led to a significant increase in SLPI stimulation compared to the control (P ⬍ 0.05). In experiments not shown, exposure of cells to virus for up to 2 h prior to medium addition did not further enhance SLPI secretion compared to cells treated with virus for 30 min. Thus, only a brief (5-min) exposure of HIV-1 is needed to induce SLPI secretion from oral keratinocytes. Also, continuous exposure to virus is not required for the effect. SLPI stimulation is induced specifically by HIV-1 and SIV external envelope glycoproteins. The component of the HIV-1 particle likely to elicit the effect is the external envelope glycoprotein gp120, since this protein is located on the exterior of the viral envelope and provides initial contact with target cells. Other viral proteins, however, might be responsible for the effect. To test this possibility, GMSM-K cells were incubated for 30 min with either SFM alone or a panel of purified re-

FIG. 5. SLPI production in GMSM-K cells exposed to HIV-1 BaL for various durations. (A) GMSM-K cells were incubated with HIV-1 BaL (MOI, 0.1) for 0 (mock), 10, 20, or 30 min. Cells were washed, incubated with growth medium, and assayed for SLPI secretion. (B) The experiment was repeated as described for panel A with BaL exposure limited to 0 (mock), 1, or 5 min prior to virus removal and addition of growth medium. Data are presented as the mean ⫾ SE at 96 h after virus exposure of triplicate cultures.

VOL. 79, 2005

HIV-1 STIMULATION OF SLPI IN ORAL EPITHELIAL CELLS

6437

FIG. 6. SLPI stimulation is mediated by the external envelope glycoproteins of diverse retroviruses. (A) SLPI production in triplicate GMSM-K cultures was assessed after incubation with a panel of purified bacterially expressed HIV-1 proteins. Shown are the SLPI levels in culture supernatants collected at 24, 48, 72, and 96 h after the addition of purified recombinant protein. IN, integrase. (B) The SLPI stimulation experiment was repeated with purified bacterially expressed external envelope glycoproteins from HIV-1 or SIV as the stimulant. Data are presented as the mean ⫾ SE of triplicate cultures.

combinant (bacterially expressed) HIV-1 proteins that included BaL gp120, Rev, Gag, integrase, and RT. SLPI secretion was measured at 24-h intervals as described above. Exposure of cells to recombinant gp120 from BaL significantly increased SLPI secretion compared to mock-treated cultures and cultures treated with the other viral proteins (P ⬍ 0.01). The level of SLPI stimulation with BaL gp120 was similar to levels seen using intact infectious and heat-inactivated BaL (compare Fig. 6A with Fig. 2, 4A and B, and 5A), indicating that gp120 alone was likely responsible for the effect. The nonenvelope viral proteins as well as human soluble CD4 (data not shown) failed to stimulate SLPI secretion. To assess whether SLPI stimulation is a property shared by retroviruses, the SLPI stimulation experiment was repeated with recombinant gp120 from HIV-1 MN, IIIB, and BaL and gp130 from SIV mac239 as inducers (Fig. 6B). All of the

HIV-1-derived gp120 proteins as well as the SIV gp130 protein stimulated SLPI production to similar levels. SLPI secretion is stimulated by HIV-1 in an unrelated oral keratinocyte cell line and in normal HGE cells. The SLPI stimulation experiments described above utilized an oral epithelial cell line originally immortalized with simian virus 40 T antigen but no longer expresses the oncogene (14). To determine whether SLPI regulation by HIV-1 also occurred in other keratinocytes, we repeated the assay with two cell sources: oral epithelial OKF6/TERT-1 cells immortalized by the human TERT catalytic telomerase subunit (8) and normal HGE cells established from healthy gingival tissues of a non-HIV-1-infected donor. The cultures were exposed to infectious BaL or HXB2 (MOI, 0.1), recombinant BaL gp120 (200 ng/well), or SFM alone for 30 min prior to the addition of growth medium. As shown in Fig. 7A and B, the infectious viruses and recom-

FIG. 7. Increased SLPI stimulation by HIV-1 in an immortalized oral keratinocyte cell line (OFK6/TERT-1) and normal (nonimmortalized) HGE cells. OFK6/TERT-1 (A) and HGE (B) cultures were treated for 30 min with infectious HIV-1 (BaL ■ or HXB2 Œ; MOI, 0.1), purified bacterially expressed BaL gp120 (䊐; 200 ng), or SFM alone ({; mock). SLPI content in culture supernatants was measured at 24-h intervals by an ELISA.

6438

JANA ET AL.

J. VIROL.

binant gp120 induced SLPI secretion in OFK6/TERT-1 (Fig. 7A) and normal HGE (Fig. 7B) cultures. Similar results were obtained with normal HGE cells from another uninfected healthy donor (data not shown). DISCUSSION The innate immune defense appears to be particularly adept at protecting the oral cavity against HIV-1 infection, as evidenced by the paucity of HIV-1 transmission through the oral route compared to genital and anorectal routes. Here, we demonstrate that SLPI, an antiviral mucosal protein, is constitutively expressed in immortalized and normal oral keratinocytes. Brief exposure of cells to HIV-1 led to a significant increase in SLPI mRNA and protein secretion that was dose and time dependent, occurred rapidly after virus contact, did not require productive cellular infection, and was elicited specifically by the external envelope glycoproteins of HIV-1 and SIV strains. In this first in-depth characterization of SLPI regulation by a virus, our findings suggest that SLPI stimulation by HIV-1 represents a previously unrecognized mechanism that contributes to the oral host immune defense against this virus. SLPI has been recognized as an important component of the innate mucosal immunity to microbial pathogens. Previous studies have confirmed its anti-inflammatory, antimicrobial, and wound-healing properties and its role in the maintaining the integrity of mucosal surfaces by inactivating destructive serine proteases during inflammation (reviewed in reference 16). The natural abundance of SLPI in oral secretions (33) has led to the speculation that the antiprotease is a major inhibitor of HIV-1 in saliva. Here, we show that oral keratinocytes are a previously unrecognized source of the inhibitor. In the time course experiment, SLPI stimulation was observed in GMSM-K cells exposed to virus for as little as 5 min. This time frame has biological relevance in that it mirrors the brief duration that oral epithelial cells are likely to be exposed to virus in vivo during receptive oral sex. Oropharyngeal tissues of infants are likely exposed to virus in infected milk for longer, multiple periods during nursing. In our experiments, the number of SLPI transcripts rose 50-fold within 1 h after exposure to HIV-1 (Fig. 3), while a significant increase in SLPI protein secretion were first detected 48 h after virus exposure in most experiments (Fig. 2, 4, 5B, and 7). The lag period between inhibitor expression and secretion likely reflects the time needed for the activation of SLPI expression and the translation, intracellular transport, secretion and accumulation of the protein in the culture supernatant. Based on the time lag, it is unlikely that virusmediated SLPI upregulation protects target cells and tissues against virus acquisition during initial infection. Rather, antiviral inhibitors (including SLPI) already present in salivary fluids likely serve this purpose. When initial protective oral defenses are overwhelmed by a bolus of virus (e.g., during breastfeeding, receptive oral sex, and experimentally infected rhesus macaques; reviewed in reference 49), virus can penetrate the mucosa via transcytosis and possibly direct infection (29, 35), and create an initial focus of viral replication. Over time, subsequent rounds of replication spread the virus through the tissues and draining lymph nodes, and systemic infection occurs. We propose that HIV-1-induced SLPI from

oral keratinocytes may help to limit virus spread through tissues rather than prevent initial infection. In addition, distal regions of the oral mucosa may be protected by the elevated SLPI level in oral fluids that ensue after virus stimulation. In support of this latter hypothesis, Lin et al. (28) recently reported higher mean SLPI concentrations in saliva from submandibular/sublingual glands of HIV-1-infected individuals compared to uninfected individuals. The anti-inflammatory property of the inhibitor may also help to control and/or limit viral spread by dampening the inflammatory response and suppressing the activation of susceptible and/or infected cells. Our data suggest that interactions between HIV-1 gp120 and component(s) of the cellular membrane activate a signaling pathway that ultimately “turns on” SLPI expression. The mechanism of action may include NF-␬␤, activated protein 1 (AP-1), and CAAT enhancer binding protein (C/EBP), nuclear transcription factors that activate an array of cytokines and chemokines in response to inflammation, microbial infections, and stress. The human SLPI promoter contains binding sites for NF-␬␤, AP-1, and C/EBP (3, 36). HIV-1 binding to cell surface CD4 during infection activates the binding of these transcription factors to the promoters of HIV-1, inflammatory cytokines and chemokines (41), thus creating an inflammatory milieu conducive to viral replication. SLPI appears to be a previously unrecognized cellular gene that is also activated by gp120-CD4 interactions. Given its anti-inflammatory property, SLPI upregulation in virus-exposed keratinocytes may serve as a mechanism to dampen the local inflammatory response to infection, as exogenous SLPI attenuated NF-␬␤-dependent inflammatory responses in human endothelial cells and macrophages after atherogenic stimuli (15). Another possibility is that SLPI stimulation is induced by virus interactions with other cell surface molecules such as Toll-like receptors (TLRs), evolutionarily conserved pattern recognition receptors engaged by specific pathogens on the surfaces of immune and epithelial cells (20). Pathogen-TLR interactions also lead to activated inflammatory gene expression through AP-1 and NF-␬␤-mediated pathways. By RTPCR, we have detected mRNAs for TLRs 1 to 9 in unstimulated GMSM-K cells (unpublished data). Preincubation of GMSM-K cells with antibodies to TLRs 2 and 4 blocked SLPI induction by BaL in GMSM-K cells (unpublished data), suggesting a role for the TLR pathway in the stimulatory effect. Many viruses, including respiratory syncytial virus, measles virus, cytomegalovirus, and influenza virus, modulate host immune responses through TLR activation by viral envelope glycoproteins, core proteins, and replicative nucleic acid intermediates (e.g., single- or double-stranded RNAs) (56). Thus, engagement of TLR-mediated pathways may represent a common mechanism by which cells detect and respond to viruses. Alternatively, interactions between HIV-1 gp120 and a nonCD4 cell surface protein involved in viral entry, such as sulfatide, heparans, galactosyl ceramide (GalCer), and intercellular adhesion molecule 1 (6), may initiate the effect. Future investigations will clarify the mechanism of HIV-1-induced SLPI upregulation and identify the signaling pathway involved in the effect. Our finding of virus-mediated induction of SLPI mirrors a recent report (42) describing HIV-1 induction of hBD-2 and hBD-3, cationic antimicrobial peptides also produced in epi-

VOL. 79, 2005

HIV-1 STIMULATION OF SLPI IN ORAL EPITHELIAL CELLS

thelial cells. In that study, normal oral epithelial cells exposed to X4 and R5 viruses expressed higher levels of hBD-2 and hBD-3 (but not hBD-1) mRNA compared to controls. The active peptides were specific for X4 viruses, had little effect against R5 viruses, and blocked viral replication through two mechanisms: direct binding to virions and down-modulation of cell surface CXCR4 expression. In contrast, SLPI antiviral activity involves a cellular rather than a viral target (30) and is active against both R5 and X4 viruses (33, 48, 51), although activity is diminished slightly with HIV-1 isolates having broad coreceptor usage patterns (e.g., CCR5, CXCR4, and either CCR2 or CCR3) (51). Thus, in its role as “gatekeeper” of the body, the oral cavity has evolved complementary strategies involving at least two distinct antimicrobial molecules in its quest to protect the body against HIV-1. Under our experimental conditions, no productive HIV-1 infection was detected in oral keratinocytes, as evaluated by p24 secretion or PCR for newly generated proviral DNA (unpublished data). This result is in agreement with that of Quin ˜ones-Mateu et al. (42), who found no productive infection in normal oral keratinocytes inoculated with either X4 or R5 viruses. The findings, however, contrast with those of Liu et al. (29), who showed infection of oral keratinocytes using high doses of X4 and R5/X4 viruses but not an R5 virus through a CD4-independent mechanism that may involve GalCer and/or CXCR4 as cellular receptors. In another study, Moore et al. (35) showed infection of oral keratinocytes with cell-free R5 viruses but not X4 viruses. The discrepancies may be due to variations in experimental conditions, such as the use of Polybrene during virus exposure by Liu et al. (29) and differences in virus dosages and/or levels of cell surface GalCer, CCR5, and/or CXCR4 expression in the cell models. It is not known whether HIV-1 stimulates SLPI production at nonoral mucosal sites and what effects enhanced inhibitor production has on the susceptibility of mucosal tissues to virus infection. Oral and genital mucosae differ in several aspects that may alter their susceptibility to this virus, including the architecture of the epithelium, composition and viscosity of the fluids bathing the sites, and local immune responses. In addition, tissue-specific differences in SLPI regulation by HIV-1 may contribute to tissue susceptibility, as treatment with tumor growth factor ␤1 suppressed SLPI mRNA levels in respiratory epithelial cells (21) but had the opposite effect in endometrial epithelial cells (50). Characterization of the SLPI response to HIV-1 in nonoral mucosal tissues will shed light on this important issue. In summary, our study reveals that oral epithelial cells constitutively express SLPI. Furthermore, SLPI expression can be manipulated by HIV-1 through infection-independent interactions initiated at the cell surface. Given the anti-inflammatory and antiviral properties of SLPI, the induction of SLPI in virus-stimulated cells represents a tug-of-war between the virus and the host immune response, as the virus attempts to stimulate the local inflammatory response while the inhibitor tries to dampen the response and/or protect neighboring cells against infection. An imbalance between the opposing responses may dictate whether virus exposure ultimately results in productive infection or protection. It will be important to identify other immune response genes whose expression is modified after contact of virus with the oral epithelium. These

6439

studies will lead to a greater understanding of the mechanisms through which the host immune response protects against mucosal HIV-1 infection. ACKNOWLEDGMENTS We thank Danny R. Gray for technical assistance and helpful discussions. This work was supported by grant R21DE15055 from the National Institutes of Health. REFERENCES 1. Abbinante-Nissen, J. M., L. G. Simpson, and G. D. Leikauf. 1993. Neutrophil elastase increases secretory leukocyte protease inhibitor transcript levels in airway epithelial cells. Am. J. Physiol. 265:L286–L292. 2. Abbinante-Nissen, J. M., L. G. Simpson, and G. D. Leikauf. 1995. Corticosteroids increase secretory leukocyte protease inhibitor transcript levels in airway epithelial cells. Am. J. Physiol. 268:L601–L606. 3. Abe, T., N. Kobayashi, K. Yoshimura, B. C. Trapnell, H. Kim, R. C. Hubbard, M. T. Brewer, R. C. Thompson, and R. G. Crystal. 1991. Expression of the secretory leukoprotease inhibitor gene in epithelial cells. J. Clin. Investig. 87:2207–2215. 4. Ashcroft, G. S., K. Lei, W. Jin, G. Longenecker, A. B. Kulkarni, T. Greenwell-Wild, H. Hale-Donze, G. McGrady, X. Y. Song, and S. M. Wahl. 2000. Secretory leukocyte protease inhibitor mediates non-redundant functions necessary for normal wound healing. Nat. Med. 6:1147–1153. 5. Beppu, Y., Y. Imamura, M. Tashiro, T. Towatari, H. Ariga, and H. Kido. 1997. Human mucus protease inhibitor in airway fluids is a potential defensive compound against infection with influenza A and Sendai viruses. J. Biochem. (Tokyo) 121:309–316. 6. Clapham, P. R., and A. McKnight. 2002. Cell surface receptors, virus entry and tropism of primate lentiviruses. J. Gen. Virol. 83:1809–1829. 7. Cohen, M. S., D. C. Shugars, and S. A. Fiscus. 2000. Limits on oral transmission of HIV-1. Lancet 356:272. 8. Dickson, M. A., W. C. Hahn, Y. Ino, V. Ronfard, J. Y. Wu, R. A. Weinberg, D. N. Louis, F. P. Li, and J. G. Rheinwald. 2000. Human keratinocytes that express hTERT and also bypass a p16INK4a-enforced mechanism that limits life span become immortal yet retain normal growth and differentiation characteristics. Mol. Cell. Biol. 20:1436–1447. 9. Division of AIDS, National Institute of Allergy and Infectious Diseases. 1994. ACTG virology manual for HIV laboratories. Publication NIH-943828. National Institutes of Health, Bethesda, Md. 10. Draper, D. L., D. V. Landers, M. A. Krohn, S. L. Hillier, H. C. Weisenfeld, and R. P. Heine. 2000. Levels of vaginal secretory leukocyte protease inhibitor are decreased in women with lower reproductive tract infections. Am. J. Obstet. Gynecol. 183:1243–1248. 11. Farquhar, C., T. C. VanCott, D. A. Mbori-Ngacha, L. Horani, R. K. Bosire, J. K. Kreiss, B. A. Richardson, and G. C. John-Stewart. 2002. Salivary secretory leukocyte protease inhibitor is associated with reduced transmission of human immunodeficiency virus type 1 through breast milk. J. Infect. Dis. 186:1173–1176. 12. Franken, C., C. J. Meijer, and J. H. Dijkman. 1989. Tissue distribution of antileukoprotease and lysozyme in humans. J. Histochem. Cytochem. 37: 493–498. 13. Gartner, S., P. Markovits, D. M. Markovitz, M. H. Kaplan, R. C. Gallo, and M. Popovic. 1986. The role of mononuclear phagocytes in HTLV-III/LAV infection. Science 233:215–219. 14. Gilchrist, E. P., M. P. Moyer, E. J. Shillitoe, N. Clare, and V. A. Murrah. 2000. Establishment of a human polyclonal oral epithelial cell line. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 90:340–347. 15. Henriksen, P. A., M. Hitt, Z. Xing, J. Wang, C. Haslett, R. A. Riemersma, D. J. Webb, Y. V. Kotelevtsev, and J. M. Sallenave. 2004. Adenoviral gene delivery of elafin and secretory leukocyte protease inhibitor attenuates NF␬␤-dependent inflammatory responses of human endothelial cells and macrophages to atherogenic stimuli. J. Immunol. 172:4535–4544. 16. Hiemstra, P. S. 2002. Novel roles of protease inhibitors in infection and inflammation. Biochem. Soc. Trans. 30:116–120. 17. Hiemstra, P. S., R. J. Maassen, J. Stolk, R. Heinzel-Wieland, G. J. Steffens, and J. H. Dijkman. 1996. Antibacterial activity of antileukoprotease. Infect. Immun. 64:4520–4524. 18. Hill, C. M., H. Deng, D. Unutmaz, V. N. Kewalramani, L. Bastiani, M. K. Gorny, S. Zolla-Pazner, and D. R. Littman. 1997. Envelope glycoproteins from human immunodeficiency virus types 1 and 2 and simian immunodeficiency virus can use human CCR5 as a coreceptor for viral entry and make direct CD4-dependent interactions with this chemokine receptor. J. Virol. 71:6296–6304. 19. Hocini, H., P. Becquart, H. Bouhlal, H. Adle-Biassette, M. D. Kazatchkine, and L. Belec. 2000. Secretory leukocyte protease inhibitor inhibits infection of monocytes and lymphocytes with human immunodeficiency virus type 1 but does not interfere with transcytosis of cell-associated virus across tight epithelial barriers. Clin. Diagn. Lab. Immunol. 7:515–518.

6440

JANA ET AL.

20. Janssens, S., and R. Beyaert. 2003. Role of Toll-like receptors in pathogen recognition. Clin. Microbiol. Rev. 16:637–646. 21. Jaumann, F., A. Elssner, G. Mazur, S. Dobmann, and C. Vogelmeier, for the Munich Lung Transplant Group. 2000. Transforming growth factor-beta1 is a potent inhibitor of secretory leukoprotease inhibitor expression in a bronchial epithelial cell line. Eur. Respir. J. 15:1052–1057. 22. Jin, F. Y., C. Nathan, D. Radzioch, and A. Ding. 1997. Secretory leukocyte protease inhibitor: a macrophage product induced by and antagonistic to bacterial lipopolysaccharide. Cell 88:417–426. 23. Kikuchi, T., T. Abe, S. Hoshi, N. Matsubara, Y. Tominaga, K. Satoh, and T. Nukiwa. 1998. Structure of the murine secretory leukoprotease inhibitor (Slpi) gene and chromosomal localization of the human and murine SLPI genes. Am. J. Respir. Cell Mol. Biol. 19:875–880. 24. King, A. E., K. Morgan, J. M. Sallenave, and R. W. Kelly. 2003. Differential regulation of secretory leukocyte protease inhibitor and elafin by progesterone. Biochem. Biophys. Res. Commun. 310:594–599. 25. Konopka, K., N. Shine, E. Pretzer, and N. Du ¨zgu ¨nes. 1999. Secretory leukocyte protease inhibitor (SLPI): oxidation of SLPI does not explain its variable anti-HIV activity. J. Dent. Res. 78:1773–1776. 26. Krisanaprakornkit, S., A. Weinberg, C. N. Perez, and B. A. Dale. 1998. Expression of the peptide antibiotic human ␤-defensin 1 in cultured gingival epithelial cells and gingival tissue. Infect. Immun. 66:4222–4228. 27. Levy, J. A. 2001. The importance of the innate immune system in controlling HIV infection and disease. Trends Immunol. 22:312–316. 28. Lin, A. L., D. A. Johnson, K. T. Stephan, and C. K. Yeh. 2004. Salivary secretory leukocyte protease inhibitor increases in HIV infection. J. Oral Pathol. Med. 33:410–416. 29. Liu, X., J. Zha, H. Chen, J. Nishitani, P. Camargo, S. W. Cole, and J. A. Zack. 2003. Human immunodeficiency virus type 1 infection and replication in normal human oral keratinocytes. J. Virol. 77:3470–3476. 30. Ma, G., T. Greenwell-Wild, K. Lei, W. Jin, J. Swisher, N. Hardegen, C. T. Wild, and S. M. Wahl. 2004. Secretory leukocyte protease inhibitor binds to annexin II, a cofactor for macrophage HIV-1 infection. J. Exp. Med. 200: 1337–1346. 31. Mann, D. L., S. J. O’Brien, D. A. Gilbert, Y. Reid, M. Popovic, E. ReadConnole, R. C. Gallo, and A. F. Gazdar. 1989. Origin of the HIV-susceptible human CD4⫹ cell line H9. AIDS Res. Hum. Retrovir. 5:253–255. 32. Maruyama, M., J. G. Hay, K. Yoshimura, C. S. Chu, and R. G. Crystal. 1994. Modulation of the secretory leukocyte protease inhibitor gene expression in human bronchial epithelial cells by phorbol ester. J. Clin. Investig. 94:368– 375. 33. McNeely, T. B., M. Dealy, D. J. Dripps, J. M. Orenstein, S. P. Eisenberg, and S. M. Wahl. 1995. Secretory leukocyte protease inhibitor: a human saliva protein exhibiting anti-human immunodeficiency virus 1 activity in vitro. J. Clin. Investig. 96:456–464. 34. McNeely, T. B., D. C. Shugars, M. Rosendahl, C. Tucker, S. P. Eisenberg, and S. M. Wahl. 1997. Inhibition of human immunodeficiency virus type 1 infectivity by secretory leukocyte protease inhibitor occurs prior to viral reverse transcription. Blood 90:1141–1149. 35. Moore, J. S., F. Rahemtulla, L. W. Kent, S. D. Hall, M. R. Ikizler, P. F. Wright, H. H. Nguyen, and S. Jackson. 2003. Oral epithelial cells are susceptible to cell-free and cell-associated HIV-1 infection in vitro. Virology 313:343–353. 36. Mulligan, M. S., A. B. Lentsch, M. Huber-Lang, R-F. Guo, V. Sarma, C. D. Wright, T. R. Ulich, and P. A. Ward. 2000. Anti-inflammatory effects of human forms of secretory leukocyte protease inhibitor. Am. J. Pathol. 156: 1033–1039. 37. Ohlsson, M., U. Fryksmark, A. Polling, H. Tegner, and K. Ohlsson. 1984. Localization of antileukoprotease in the parotid and the submandibular salivary glands. Acta Otolaryngol. 98:147–151. 38. Orsini, M. J., A. N. Thakur, W. W. Andrews, M. L. Hammarskjo ¨ld, and D. Rekosh. 1995. Expression and purification of the HIV type 1 Rev protein produced in Escherichia coli and its use in the generation of monoclonal antibodies. AIDS Res. Hum. Retrovir. 11:945–953. 39. Ota, Y., K. Shimoya, Q. Zhang, A. Moriyama, R. Chin, K. Tenma, T. Kimura, M. Koyama, C. Azuma, and Y. Murata. 2002. The expression of secretory leukocyte protease inhibitor (SLPI) in the fallopian tube: SLPI protects the acrosome reaction of sperm from inhibitory effects of elastase. Hum. Reprod. 17:2517–2522. 40. Pillay, K., A. Coutsoudis, A. K. Agadzi-Naqvi, L. Kuhn, H. M. Coovadia, and E. N. Janoff. 2001. Secretory leukocyte protease inhibitor in vaginal fluids and perinatal human immunodeficiency virus type 1 transmission. J. Infect. Dis. 183:653–656. 41. Popik, W., J. E. Hesselgeser, and P. M. Pitha. 1998. Binding of human immunodeficiency virus type 1 to CD4 and CXCR4 receptors differentially regulates expression of inflammatory genes and activates the MEK/ERK signaling pathway. J. Virol. 72:6406–6413.

J. VIROL. 42. Quin ˜ ones-Mateu, M. E., M. M. Lederman, Z. Feng, B. Chakraborty, J. Weber, H. R. Rangel, M. L. Marotta, M. Mirza, B. Jiang, P. Kiser, K. Medvik, S. F. Sieg, and A. Weinberg. 2004. Human epithelial ␤-defensins 2 and 3 inhibit HIV-1 replication. AIDS 17:F39–F48. 43. Sallenave, J. M., J. Shulmann, J. Crossley, M. Jordana, and J. Gauldie. 1994. Regulation of secretory leukocyte proteinase inhibitor (SLPI) and elastase-specific inhibitor (ESI/elafin) in human airway epithelial cells by cytokines and neutrophilic enzymes. Am. J. Respir. Cell Mol. Biol. 11:733– 741. 44. Sallenave, J. M., M. Si-Tahar, G. Cox, M. Chignard, and J. Gauldie. 1997. Secretory leukocyte proteinase inhibitor is a major leukocyte elastase in human neutrophils. J. Leukoc. Biol. 61:695–702. 45. Schalkwijk, J., O. Wiedow, and S. Hirose. 1999. The trappin gene family: proteins defined by an N-terminal transglutaminase substrate domain and a C-terminal four-disulphide core. Biochem. J. 340:569–577. 46. Schatz, O., J. Mous, and S. F. Le Grice. 1990. HIV-1 RT-associated ribonuclease H displays both endonuclease and 3⬘-5⬘ exonuclease activity. EMBO J. 9:1171–1176. 47. Shine, N. R., S. C. Wang, K. Konopka, E. A. Burks, N. Du ¨zgu ¨nes, and C. P. Whitman. 2002. Secretory leukocyte protease inhibitor: inhibition of human immunodeficiency virus-1 infection of monocytic THP-1 cells by a newly cloned protein. Bioorg. Chem. 30:249–263. 48. Shugars, D. C., D. L. Sauls, and J. B. Weinberg. 1997. Secretory leukocyte protease inhibitor blocks infectivity of primary monocytes and mononuclear cells with both monocytotropic and lymphocytotropic strains of human immunodeficiency virus type 1. Oral Dis. 3(Suppl. 1):S70–S72. 49. Shugars, D. C., S. P. Sweet, D. Malamud, S. H. Kazmi, K. Page-Shafer, and S. J. Challacombe. 2002. Saliva and inhibition of HIV infection: molecular mechanisms. Oral Dis. 8(Suppl. 2):169–175. 50. Si-Tahar, M., D. Merlin, S. Sitaraman, and J. L. Madara. 2000. Constitutive and regulated secretion of secretory leukocyte protease inhibitor by human intestinal epithelial cells. Gastroenterology 118:1061–1071. 51. Skott, P., E. Lucht, M. Ehnlund, and E. Bjo ¨rling. 2002. Inhibitory function of secretory leukocyte proteinase inhibitor (SLPI) in human saliva is HIV-1 specific and varies with virus tropism. Oral Dis. 8:160–167. 52. Sørensen, O. E., J. B. Cowland, K. Theilgaard-Mo ¨nch, L. Liu, T. Ganz, and N. Borregaard. 2003. Wound healing and expression of antimicrobial peptides/polypeptides in human keratinocytes, a consequence of common growth factors. J. Immunol. 170:5583–5589. 53. Thompson, R. C., and K. Ohlsson. 1986. Isolation, properties, and complete amino acid sequences of human secretory leukocyte protease inhibitor, a potent inhibitor of leukocyte elastase. Proc. Natl. Acad. Sci. USA 83:6692– 6696. 54. Tomee, J. F., P. S. Heimstra, R. Heinzel-Wieland, and H. F. Kauffman. 1997. Antileukoprotease: an endogenous protein in the innate mucosal defense against fungi. J. Infect. Dis. 176:740–747. 55. Turpin, J. A., C. A. Schaeffer, M. Bu, L. Graham, R. W. Buckheit, Jr., D. Clanton, and W. G. Rice. 1996. Human immunodeficiency virus type-1 (HIV-1) replication is unaffected by human secretory leukocyte protease inhibitor. Antiviral Res. 29:269–277. 56. Vaidya, S. A., and G. Cheng. 2003. Toll-like receptors and innate antiviral responses. Curr. Opin. Immunol. 15:402–407. 57. van Wetering, S., A. C. van der Linden, M. A. van Sterkenburg, K. F. Rabe, J. Schalkwijk, and P. S. Hiemstra. 2000. Regulation of secretory leukocyte protease inhibitor (SLPI) production by human bronchial epithelial cells: increase of cell-associated SLPI by neutrophil elastase. J. Investig. Med. 48:359–366. 58. Wahl, S. M., T. B. McNeely, E. N. Janoff, D Shugars, P. Worley, C. Tucker, and J. M. Orenstein. 1997. Secretory leukocyte protease inhibitor (SLPI) in mucosal fluids inhibits HIV-1. Oral Dis. 3(Suppl. 1):S64–S69. 59. Wahl, S. M., P. Worley, W. Jin, T. B. McNeely, S. Eisenberg, C. Fasching, J. M. Orenstein, and E. N. Janoff. 1997. Anatomic dissociation between HIV-1 and its endogenous inhibitor in mucosal tissues. Am. J. Pathol. 150: 1275–1284. 60. Zhang, D., R. C. Simmen, F. J. Michel, G. Zhao, D. Vale-Cruz, and F. A. Simmen. 2002. Secretory leukocyte protease inhibitor mediates proliferation of human endometrial epithelial cells by positive and negative regulation of growth-associated genes. J. Biol. Chem. 277:29999–30009. 61. Zhang, Y., D. L. DeWitt, T. B. McNeely, S. M. Wahl, and L. M. Wahl. 1997. Secretory leukocyte protease inhibitor suppresses the production of monocyte prostaglandin H synthase-2, prostaglandin E2, and matrix metalloproteinases. J. Clin. Investig. 99:894–900. 62. Zhu, J., C. Nathan, W. Jin, D. Sim, G. S. Ashcroft, S. M. Wahl, L. Lacomis, H. Erdjument-Bromage, P. Tempst, C. D. Wright, and A. Ding. 2002. Conversion of proepithelin to epithelins: roles of SLPI and elastase in host defense and wound repair. Cell 111:867–878.