Activation of Kaposi's Sarcoma-Associated Herpesvi
http://www.100md.com
病菌学杂志 2005年第21期
Departments of Laboratory Medicine
Otolaryngology, University of Washington, Seattle, Washington 98195
ABSTRACT
The oral cavity has been identified as the major site for the shedding of infectious Kaposi's sarcoma-associated herpesvirus (KSHV). While KSHV DNA is frequently detected in the saliva of KSHV seropositive persons, it does not appear to replicate in salivary glands. Some viruses employ the process of epithelial differentiation for productive viral replication. To test if KSHV utilizes the differentiation of oral epithelium as a mechanism for the activation of lytic replication and virus production, we developed an organotypic raft culture model of epithelium using keratinocytes from human tonsils. This system produced a nonkeratinized stratified squamous oral epithelium in vitro, as demonstrated by the presence of nucleated cells at the apical surface; the expression of involucrin and keratins 6, 13, 14, and 19; and the absence of keratin 1. The activation of KSHV lytic-gene expression was examined in this system using rKSHV.219, a recombinant virus that expresses the green fluorescent protein during latency from the cellular EF-1 promoter and the red fluorescent protein (RFP) during lytic replication from the viral early PAN promoter. Infection of keratinocytes with rKSHV.219 resulted in latent infection; however, when these keratinocytes differentiated into a multilayered epithelium, lytic cycle activation of rKSHV.219 occurred, as evidenced by RFP expression, the expression of the late virion protein open reading frame K8.1, and the production of infectious rKSHV.219 at the epithelial surface. These findings demonstrate that KSHV lytic activation occurs as keratinocytes differentiate into a mature epithelium, and it may be responsible for the presence of infectious KSHV in saliva.
INTRODUCTION
Since its initial description over a decade ago (14), Kaposi's sarcoma-associated herpesvirus (KSHV) has been recognized as a significant viral pathogen, particularly for immunocompromised hosts and persons infected with human immunodeficiency virus type 1 (HIV-1). KSHV is the etiologic agent of Kaposi's sarcoma (KS), having been demonstrated in biopsy specimens of all forms of KS despite distinct differences in the geographic origin, age, and gender of affected persons (29). KSHV is also associated with multicentric Castleman's disease (80) and primary effusion lymphoma (11).
The precise modes of KSHV transmission are not clearly defined; however, epidemiological data suggest that both sexual and nonsexual routes are possible. The association between KSHV transmission and sexual activity has been largely defined in North American men who have sex with men (MSM), where risk factors include sex with a partner who has KS (66), a history of sexually transmitted infections (STIs) (55), and an increased number of sexual partners (55). Other risk factors, however, such as deep kissing with an HIV-positive partner (66) and orogenital contact (25), have led to consideration of an oral source of transmission. In a cohort of MSM from San Francisco, similar prevalences of KSHV infection from 1978 to 1996 were found, despite a reduction in HIV-1 seroprevalence and the institution of "safer" sexual practices (65). This raises the issue that sexual activity may be a marker for other types of intimate contact in this population. In keeping with this idea, PCR-based studies of the male genitourinary tract have described only infrequent, low-level shedding of KSHV DNA in genital secretions (22). Infectious virions have not been demonstrated in semen. Similarly, only infrequent and low-level shedding of KSHV DNA has been observed from male urethral and anorectal secretions (66). KSHV DNA has only rarely been detected in vaginal secretions (8, 46, 93), and heterosexual sex has not been clearly associated with KSHV transmission (76).
Evidence for nonsexual transmission is supported by data derived from studies in Africa, Italy, Egypt, and Japan which document KSHV infection in groups with very low risk of STI, including children (1, 8, 42, 91, 92). In regions of Africa where KSHV is endemic, a cumulative increase in KSHV seroprevalence from birth through adolescence has been observed, with more than 40% of children over 14 years of age infected and many infants seroconverting during the first year of life (2, 30, 56). Evidence for transmission through breast milk has not been found (7), and congenital infection is thought to be rare (54). The fact that children and other groups without risk of STI are infected with KSHV implies salivary transmission, as is seen with other human herpesviruses (HHVs), such as cytomegalovirus, HHV-6, HHV-7, and Epstein-Barr virus (EBV).
KSHV DNA has been detected frequently in human saliva (6, 44), and when found, it occurs at a titer 2 to 3 log units higher than those at other anatomic sites (66). Infectious KSHV has been documented in saliva (87). For these reasons, the oral cavity is unique in its ability to support lytic replication of KSHV. Published studies report that 11% to 68% of HIV-positive MSM infected with KSHV in the absence of KS shed viral DNA in saliva on at least one occasion (9, 22, 66). One study demonstrated that 39% of KSHV-seropositive MSM shed viral DNA from the oral cavity on more than a third of the days sampled, regardless of HIV-1 status (66). Current data indicate that KSHV does not replicate in salivary glands (21, 22). Epithelial cells lining the oral cavity are a probable source, given that nucleated oral epithelial cells from nonkeratinized mucosal surfaces have been shown to support viral infection in vivo, by the detection of KSHV-specific latent and lytic mRNA transcripts (66).
Other viruses, such as EBV (31, 49, 74, 96) and human papillomavirus (HPV) (reviewed in reference 51), undergo lytic viral replication in differentiating epithelia. The organotypic raft culture model, originally developed to study keratinocyte differentiation (70), has been of great value in the study of viral replication in developing epithelium (57). Organotypic raft cultures accurately reproduce the process of epithelial differentiation in vitro, beginning with undifferentiated keratinocyte monolayers that become confluent and polarized, forming tight junctions prior to the initiation of keratinocyte differentiation. A proportion of the keratinocytes remain in the basal layer and maintain proliferative potential, while other cells leave the basal layer, lose mitotic capability, and differentiate into mature epithelial cells, forming the multilayered structure of the epithelium (39). Keratinocytes follow a programmed pattern of gene expression during differentiation, each step of which is characterized by the expression of keratin, and keratin-associated, proteins (59). This in vitro model has demonstrated that HPV productively infects cutaneous and mucosal epithelia by a process that is tightly linked to epithelial differentiation (40).
In order to test the hypothesis that activation of KSHV lytic gene expression occurs during epithelial differentiation, we employed an organotypic raft culture model that utilized keratinocytes isolated from normal tonsils. We found that the process of epithelial differentiation activated KSHV lytic-gene expression and resulted in the production of infectious virus at the epithelial surface.
MATERIALS AND METHODS
Subjects. This study was approved by the Human Subjects Review Committee of the University of Washington. Oral and written informed consent was obtained from all participants. Healthy adults planning elective tonsillectomy were eligible to donate leftover tonsil tissues not required for routine pathology. All tonsillectomy procedures were performed by members of the Department of Otolaryngology at the University of Washington. There were no modifications of routine surgical practices and procedures. Subjects with tonsillar mass or asymmetry were excluded in case they had a carcinoma or lymphoma, but otherwise, there were no specific exclusion criteria.
Tonsil-derived organotypic raft cultures. Tonsil specimens were immediately transported in Dulbecco's modified Eagle's medium (DMEM) (Gibco, Invitrogen Corporation) with 10% fetal bovine serum (FBS) (Gibco, Invitrogen Corporation) plus antibiotics (penicillin-streptomycin) at 4°C and processed on arrival. Tissue specimens were variable in size, with surface areas ranging from approximately 1 to 4 cm2. Epithelial cell sheets were separated from each specimen by careful dissection after overnight incubation in dispase II (24.0 U/ml; Gibco, Invitrogen Corporation) at 4°C. Collected materials were treated with 0.05% trypsin-EDTA (Gibco, Invitrogen Corporation) for 30 min at 37°C to form single-cell suspensions and plated at a concentration of 5 x 105 cells/ml in EpiLife medium (Ca2+; 0.06 M) supplemented with the HKGS kit (Cascade Biologics, Inc. Portland, OR) onto 60-mm culture dishes coated with mouse collagen type IV (BD Biosciences, Bedford, MA). The keratinocytes were placed in a 37°C CO2 incubator and fed every other day with EpiLife medium. The keratinocytes were expanded to 70% confluence and used for the organotypic tissue culture.
Autologous fibroblasts were recovered from each tonsil specimen by placing leftover pieces in a T75 flask and incubating them in DMEM supplemented with 10% FBS, L-glutamine, amphotericin B, ciprofloxacin, penicillin, streptomycin, and fresh vitamin C (50 μg/ml; Sigma, St. Louis, MO) at 37°C in a CO2 incubator and were fed every other day. Adherent fibroblasts were expanded and used to seed the bottoms of 12-well plates at a density of 105 cells/well for use in the raft culture model. Expanded keratinocyte cultures were trypsinized, washed, and seeded onto type I collagen-coated cell culture inserts (12-well format) at a density of 105 cells/insert in EpiLife medium. The inserts were incubated submerged at 37°C in a humidified 5% CO2 incubator for a period of 8 to 10 days until a confluent monolayer formed. The inserts were then transferred to a fresh well seeded with autologous fibroblasts at a density of 3 x 105 cells/well and submerged in FAD medium (three parts DMEM, one part Ham's F-12 [Gibco, Invitrogen Corporation], 10% FBS, 8.9 ng/ml epidermal growth factor [Sigma, St. Louis, MO], 0.45 ng/ml insulin [Sigma, St. Louis, MO], 0.45 ng/ml hydrocortisone [Sigma, St. Louis, MO], and 0.3 g/ml adenine [Sigma, St. Louis, MO]) for 24 h, followed by removal of the FAD medium from the upper chamber, exposing the keratinocyte layer to air. Once the layer was exposed, FAD medium (0.6 ml/well) was added to the lower chamber daily to ensure the insert was fed only from the bottom. The inserts were incubated at 37°C in a humidified 5% CO2 incubator for 6 days prior to harvest. Half of each insert was snap frozen fresh in Optimal Cutting Temperature medium (Sakura, Tokyo, Japan) and stored at –80°C. The remaining half was fixed in 4% paraformaldehyde for 30 min at room temperature (RT), snap frozen in Optimal Cutting Temperature medium, and stored at –80°C. A cryostat was used to cut 10-μm sections of tissue on silane-coated slides for further studies.
Three organotypic raft cultures (each derived from a separate donor in this study) were tested by PCR for the presence of herpes simplex virus types 1 and 2, varicella-zoster virus, cytomegalovirus, EBV, HHV-6, and high-risk HPV types (including types 6, 11, 16, 18, 31, 33, 35, 39, and 45) and found to be negative. Reference testing was performed by the Molecular Diagnostic Laboratory and the HPV Laboratory at the University of Washington using previously published methodology (5, 9, 43, 97).
Antibody detection of keratin proteins and viral ORF K8.1 late envelope glycoprotein. Frozen sections of organotypic raft cultures were thawed and washed in phosphate-buffered saline (PBS), pH 7.4. Tissue sections for keratin antibody staining were used directly for immunofluorescence histochemistry, whereas those used for detection of ORF K8.1 were air dried and fixed for 30 min in 4% paraformaldehyde at RT. Tissue sections were blocked with Serum-Free Protein Block (DAKO Corporation, Carpinteria, CA). Keratin antibodies were used to characterize organotypic raft cultures, including CK1, clone 34?B4; CK10, clone LHP1; CK4, clone 6B10; CK13, clone KS-1A3; CK5, clone XM26; CK14, clone LL002; CK6, clone LHK6B; CK16, clone LL025; CK19, clone b170; and involucrin, clone SY5,(all from NovoCastra Laboratories, Newcastle, United Kingdom), at dilutions recommended by the manufacturer. ORF K8.1 protein was detected with monoclonal antibody (MAb) A4 diluted 1:20 in PBS (98). Slide sections were incubated with primary antibody for 1 h at RT, washed, and detected with isotype-specific Qdot 655-conjugated goat F(ab')2 anti-mouse immunoglobulin G (IgG) secondary antibody (Quantum Dot Corporation, Hayward, CA) diluted 1:200 for 30 min at RT. Slides were mounted with Vectashield fluorescence mounting medium with DAPI (4',6'-diamidino-2-phenylindole) (Vector Laboratories, Inc., Burlingame, CA). Staining controls for keratin staining included omission of the primary antibody and normal human tonsil epithelium. Staining controls for the ORF K8.1 MAb included omission of the primary antibody, uninfected raft culture sections, and productively infected Vero cells as positive controls.
Antibody detection of viral ORF 73 (LANA) in rKSHV.219-infected oral keratinocytes. The immunofluorescence detection of KSHV ORF 73 protein was carried out with rat MAb to HHV-8 ORF 73 (ABI, Columbia, MD) diluted 1:100 in 10% normal goat serum. Alexa Fluor 647 goat anti-rat IgG (Molecular Probes, Eugene, OR) was used for fluorescence detection. Oral keratinocytes infected with recombinant KSHV.219 (rKSHV.219) for 10 days and split twice over that time were fixed for 30 min in 4% paraformaldehyde at RT, permeabilized with 0.5% Triton X-100 at RT for 15 min, and reacted with rat MAb to ORF 73. The keratinocytes were washed twice with PBS and reacted with Alexa Fluor 647 goat anti-rat IgG diluted 1:200 for 30 min, washed twice with PBS, counterstained with 1 μg/ml DAPI for 1 min, and washed in PBS.
rKSHV.219. rKSHV.219 is a JSC-derived recombinant KSHV that expresses the green fluorescent protein (GFP) during latent and lytic replication from the cellular promoter EF-1 and the red fluorescent protein (RFP) during lytic replication from the viral lytic PAN promoter. Details concerning the construction and production of rKSHV.219 have been previously published (89).
Infection of organotypic raft cultures. Epithelial cell monolayers were infected with rKSHV.219 on cell culture inserts at 70% confluence in EpiLife medium. Inserts were infected from the top with 200 μl of rKSHV.219 with Polybrene (7.5 μg/ml) and incubated at 37°C in a humidified 5% CO2 incubator for 2 h. A single preparation of virus was used for these experiments at a titer of 6 x 105 infectious units (IU)/ml as determined by titering on 293 cells and counting GFP-positive cells. Virus stock was removed, and the insert was washed with sterile PBS twice. The insert was again submerged in EpiLife medium until confluent and processed as described above. The inserts were observed daily for GFP and RFP expression by fluorescence microscopy as evidence of latent or lytic infection of cells.
Determination of IU of rKSHV.219. For the detection of infectious KSHV in the raft cultures, 0.3 ml of medium was placed in the upper chamber 5 days after air exposure and collected after overnight incubation. The determination of IU of rKSHV.219 was performed as previously described (89). Briefly, cells were removed from each supernatant by centrifugation (300 x g; 10 min) and then passed through a 0.45-μm filter and used as rKSHV.219 inocula for the infection of 293 cells. The number of IU of rKSHV.219 was then determined from the number of GFP-positive 293 cells counted 2 days postinfection. To maximize the infection, it was carried out in the presence of Polybrene (7.5 μg/ml) with centrifugation enhancement (450 x g; 20 min), and then the cells were incubated for 3 h before the medium was replaced with fresh medium without Polybrene. Medium supernatant from the rKSHV.219-infected epithelial cells was collected prior to air exposure and used as a control.
Microscopy. A Nikon Eclipse TE2000S inverted fluorescence microscope equipped with filter sets TE2000 DAPI, TE2000 FITC HYQ, TE2000 Texas Red HYQ, and Omega Optical XF305-2 was used. Images were acquired with a Photometrics CoolSNAP cf digital camera and Metavue Imaging software version 6.1.
RESULTS
Development of nonkeratinized stratified squamous epithelium in vitro. An in vitro organotypic raft culture model of oral epithelium was produced using oral keratinocytes isolated from human tonsils. The keratinocytes were expanded ex vivo and plated on cell culture inserts that served as the support for the organotypic culture. Autologous fibroblasts were incorporated into the model by seeding the bottom of each cell culture well. Confluent keratinocyte monolayers were exposed to air and observed daily prior to harvest on the sixth day. Morphological characteristics of the epithelium were assessed by phase microscopy, DAPI staining, and antibody detection of specific markers of epithelial differentiation. Raft cultures developed a multilayered epithelium 8 to 12 layers thick (Fig. 1). Nucleated cells were observed at the surface of the raft culture by DAPI staining (Fig. 1), a feature consistent with nonkeratinized epithelium (81, 85). Types of epithelia and the state of differentiation can be profiled by the expression pattern of keratins, a family of over 30 proteins expressed in specific pairings of type I (acidic) and type II (basic) members (59). To further characterize the in vitro-generated epithelium, cross sections were analyzed with monoclonal antibodies to specific keratins and involucrin. The keratin pair K4 and K13, specific for nonkeratinized oral stratified epithelia (19, 20, 59, 94), was detected in suprabasal layers (K13 is shown in Fig. 1). Conversely, the keratin pair K1 and K10, specifically found in keratinized epithelia (20, 47, 59, 75, 78), was absent (K1 is shown in Fig. 1). The keratin pair K5 and K14, which is found in all types of oral stratified epithelia (61, 67), was present in basal and suprabasal layers of the raft culture, consistent with the tissue of origin (K14 is shown in Fig. 1). Other keratin markers found in differentiated tonsil epithelium, including keratins K6 and K19 (35, 50, 58, 62), were also demonstrated in suprabasal layers of the raft culture (Fig. 1). Involucrin, a keratin-associated protein indicative of epithelial differentiation (41, 68, 83), was observed in the suprabasal layers of the raft culture (Fig. 1). These experiments demonstrated that the epithelial morphology and keratin expression pattern observed in our in vitro organotypic raft culture model was consistent with that of normal human tonsil epithelium (58).
KSHV replication in keratinocytes and in vitro epithelium. In vivo epithelium arises from keratinocytes (progenitor cells) located in the basal layer (39). Similarly, in vitro organotypic raft cultures are formed from undifferentiated keratinocyte monolayers. Therefore, oral keratinocytes, which were cultured to maintain an undifferentiated state, were infected with rKSHV.219 to examine latent and lytic replication. Figure 2A depicts undifferentiated, primary oral keratinocytes 10 days after infection with rKSHV.219 by phase (Fig. 2A, 1) and fluorescence for GFP (Fig. 2A, 2) and RFP (Fig. 2A, 3). No evidence of RFP expression was seen, indicative of latent viral infection (Fig. 2A, 3). In Fig. 2B, the same keratinocytes were examined for LANA expression by phase (Fig. 2B, 1) and by fluorescence for LANA, GFP, and DAPI (Fig. 2B, 2). GFP-expressing cells demonstrated punctate nuclear staining for LANA, while non-GFP-expressing cells did not. The absence of RFP expression and the presence of punctate nuclear LANA staining are indicative of latent viral infection in these undifferentiated keratinocyte monolayers.
Next, this model of oral epithelium was used to assess the impact of epithelial differentiation on KSHV replication. This was accomplished by infecting keratinocytes at 50 to 70% confluence on culture inserts with rKSHV.219 and using the infected keratinocytes to initiate organotypic raft cultures. The infected keratinocytes continued to grow, became confluent in 2 to 3 days, and expressed GFP but not RFP, as seen in an intact keratinocyte monolayer 1 day prior to air exposure (Fig. 3A). This was indicative of latent infection, as has been previously reported for KSHV-infected keratinocyte monolayers (10, 88). This was also similar to long-term cultures of rKSHV.219-infected keratinocytes that were passaged submerged in epithelial medium (Fig. 2). There is a single published report that describes lytic infection of primary epithelial cell monolayers in culture, but it is not currently known whether this observation was due to differences in the virus used or cell culture conditions (26). One day postconfluence, the medium from the top chamber was removed in order to expose the apical surfaces of the keratinocytes to air and to promote differentiation of the epithelium. Intact living cultures were examined daily for GFP and RFP expression. Images of phase, GFP expression, and RFP expression at 3 and 6 days after air exposure are shown (Fig. 3B and C). The expression of RFP from the lytic PAN promoter indicated that activation of lytic-gene expression occurred in the differentiating epithelium.
RFP expression in rKSHV.219-infected cells indicates early gene expression, because it is under the control of the lytic PAN promoter, which is directly activated by Rta (79). To determine if late lytic-gene expression occurred in these epithelial cells, cultures were harvested 6 days after air exposure and intact rafts were analyzed with antibody to the late viral glycoprotein K8.1. K8.1 expression was identified in a subset of cells expressing RFP (Fig. 4). In these cultures, not all cells with KSHV early gene expression progressed to late gene expression, an observation made in other cell types (18, 60, 89), although the reasons are not understood. These experiments demonstrated that the process of epithelial differentiation activated KSHV lytic gene expression. Observations of the intact organotypic raft culture suggested that this was predominately occurring in cells at the apical surface; therefore, the location of lytic-gene expression within the rafts was examined.
To identify the location of cells with lytic-gene expression within the epithelial tissue, rafts were sectioned and examined by fluorescence microscopy for GFP and RFP expression. On cross section, GFP was distributed throughout all layers of the raft culture epithelium but was expressed most intensely at the surface, where squames had become flattened and more densely compacted (Fig. 5B). RFP was observed primarily in cells at the apical surface of the raft culture, indicating that lytic-gene expression was occurring predominantly in differentiated epithelial cells (Fig. 5C). In order to compare the localization of late lytic-protein expression with that of early lytic-gene expression (indicated by RFP), sections of the raft cultures were stained with antibody to the ORF K8.1 protein. As seen for the RFP-positive cells, K8.1 was detected in cells at the apical surface of the epithelium (Fig. 6).
Production of infectious KSHV. Because the eventual outcome of the activation of KSHV lytic-gene expression is the generation of infectious virus, the organotypic raft cultures were tested for the production of infectious KSHV. This was carried out by adding medium to the upper chamber of the cultures 5 days after air exposure. The medium was removed after overnight incubation and used to inoculate 293 cells to determine the presence of IU of rKSHV.219 by examination for GFP-positive 293 cells 2 days postinoculation. Figure 7 shows an image of 293 cells infected with supernatant from one of the rKSHV.219-infected raft cultures. Three raft cultures derived from one tonsil and two from a second tonsil were tested, and all were found to produce infectious virus, ranging from 44 to 165 IU per 0.9-cm2 insert. Medium removed from the upper chamber at the time of air exposure was also used to inoculate 293 cells and was found to be negative. This demonstrated that differentiating keratinocytes activated KSHV lytic replication and became permissive for the production of infectious KSHV.
DISCUSSION
Saliva has been the only readily transmitted bodily fluid proven to contain infectious KSHV (87), and epidemiological data suggest saliva as a likely route of transmission (2, 30, 56), but the source of KSHV in saliva has not been well characterized. Unlike human cytomegalovirus or human herpesvirus 6, current data suggest that KSHV does not replicate in salivary glands (22, 66). EBV and HPV activate lytic replication in developing epithelia, suggesting that KSHV might also replicate in epithelial cells lining the oral cavity. In order to study KSHV replication in oral epithelia, we developed an organotypic raft culture model, derived from human tonsil keratinocytes, that recapitulated the characteristics of a nonkeratinized stratified squamous oral epithelium. Using this model, we examined the impact of epithelial differentiation on KSHV replication. We found that undifferentiated primary keratinocytes maintained latent infection with KSHV; however, the process of epithelial differentiation resulted in the activation of both early and late lytic viral gene expression, with the release of infectious KSHV at the apical surface of the epithelium.
The production of a nonkeratinized organotypic raft culture model of oral epithelium was a critical component of these studies. The organotypic raft culture model is a well-established system for the production of epithelia in vitro. Early raft culture models, which utilized skin keratinocytes to produce multilayered epithelial tissue, exhibited differentiation patterns that resembled the tissue from which the keratinocytes originated (3, 4, 95). Keratinocytes were cultured on dermal equivalents composed of type I collagen and fibroblasts and raised to the air-medium interface to promote differentiation. This model has also been applied to keratinocytes derived from the oral cavity, including buccal (16, 17, 33, 34, 73), lingual (38), and peritonsillar (62) surfaces. Several of these studies underscored the importance of fibroblasts located in adjacent connective tissue, which provide permissive and instructive signals to differentiating keratinocytes (64, 77). In our system, primary keratinocytes isolated from nonkeratinized tonsil epithelium were seeded onto semipermeable cell culture inserts, and a fibroblast feeder layer derived from the tonsil dermis was established in the cell culture well beneath the insert. This format allowed excellent visualization and photography of cultures during development and provided a stable support for histological sectioning of harvested tissue. Raft cultures derived from primary tonsil keratinocytes had the expected morphological characteristics of nonkeratinized oral epithelium (58). An advanced stage of epithelial differentiation was achieved in suprabasal layers of the raft culture, as evidenced by expression of keratins 4 and 13 (59, 94) and involucrin (68). In addition, the presence of nucleated cells at the apical surface, the absence of specific markers of keratinization (keratins 1 and 10), and suprabasal expression of specific markers for nonkeratinized stratified squamous epithelium (keratins 4 and 13) (47) showed this to be an appropriate system to examine KSHV replication in oral epithelia.
We used our in vitro raft culture model of oral epithelium to test the hypothesis that basal keratinocytes, latently infected with KSHV, would activate viral lytic cycle gene expression during the process of epithelial differentiation, as suggested for the activation of EBV in the oral epithelia (49, 96). EBV, a gammaherpesvirus related to KSHV, is found in B cells and epithelial cells and is transmitted in saliva. Although the role of epithelial cells in primary EBV infection remains controversial (37, 63, 74), suprabasal epithelial cells in oral hairy leukoplakia lesions have been shown to replicate viral genomes and to express both lytic (36, 96) and latent (86) cycle antigens. Of value in our study, KSHV readily infected primary oral keratinocytes in culture and established a latent infection, which allowed us to initiate the process of epithelial differentiation using rKSHV.219-infected primary keratinocytes. The use of rKSHV.219 allowed the daily observation of raft cultures during differentiation for the identification of infected cells by GFP expression and for the activation of lytic gene expression indicated by the expression of RFP. While no RFP was seen in the basal layer of the epithelium, the process of keratinocyte differentiation resulted in the expression of RFP by many cells at the upper layer of the culture. While the expression of RFP from the lytic PAN promoter indicates early gene expression, a subset of RFP-positive cells was found to have late lytic gene expression as well, indicated by the presence of the late lytic envelope glycoprotein K8.1. The expression of K8.1 requires viral lytic DNA synthesis and therefore signifies that keratinocyte differentiation activates lytic DNA replication (13). The result of lytic replication is the production of infectious virus. In this in vitro model of oral epithelium, infectious virus was recovered from the apical surface of the raft culture, demonstrating that the complete KSHV lytic replication cycle can be completed as latently infected keratinocytes differentiate into a mature epithelium. Unlike in vitro epithelial models of EBV (49) and HPV (57) infection, which require stimulation with a phorbol ester to induce viral lytic replication, the activation of KSHV lytic-gene expression in differentiating primary keratinocytes is brought about by a natural cellular process.
KSHV lytic-cycle activation is initiated by Rta, the immediate-early transcriptional activator, expressed from open reading frame (ORF) 50 (53, 84, 90). Other agents known to induce KSHV lytic-cycle activation include phorbol esters (12, 69) and DNA-demethylating agents, such as azacytidine (15). Unlike KSHV culture systems that rely on the chemical treatment of cells, activation of KSHV lytic-gene expression in raft culture epithelium is brought about by a natural cellular process. Although the mechanism is unknown, one possible explanation is chromatin remodeling. Chromatin rearrangement around the late promoter region during epithelial differentiation has been described for HPV (23) and suggests that DNA binding of differentiation-specific cellular transcription factors is involved. Transcriptional regulation in eukaryotic cells is heavily dependent upon histone acetylation and methylation (reviewed in reference 24). Keratinocyte differentiation and growth arrest can be induced by inhibitors of histone deacetylase, such as sodium butyrate (72, 82) and tricostatin A (71). The KSHV Rta promoter can also be induced by these histone deacetylase inhibitors (52, 89), and Rta itself is involved in the recruitment of cellular factors (32, 35) that influence chromatin remodeling and access to cellular transcription factors (52). Another possible mechanism is the recruitment of cellular transcription factors at gene promoter sites by regulation of AP1, Sp1, and CCATT/enhancer-binding proteins (27, 28). ORF 50 utilizes a GC box initiator element and binds Sp family transcription factors (52), as do other cellular and viral promoters (48). Sp1 proteins are critical transactivators of regulated keratinocyte gene expression (27), and promoters containing GC box initiator elements may be more active in keratinocytes. For example, the EBV early protein BMRF2, which utilizes a GC box initiator element and is highly expressed in oral hairy leukoplakia lesions, exhibits 10-fold-greater activity in epithelial cells than in lymphocytes (45). The mechanisms of activation (chromatin remodeling and recruitment of cellular transcription factors) are not mutually exclusive, and other factors could be involved. Further experiments will be required to determine the mechanisms that are important for viral activation in differentiating cells.
In summary, this work demonstrates that the process of oral epithelial differentiation can activate KSHV lytic-gene expression. Moreover, because differentiated oral keratinocytes are competent to support productive viral replication with the release of infectious viral particles, oral epithelial cells are a likely source of infectious KSHV present in saliva. The switch between latent and lytic replication is the hallmark of herpesvirus biology, allowing the permanent infection of the host, and is critical to the pathogenesis and transmission of herpesviruses. The results presented here illustrate an in vitro model of a natural cellular process causing the switch from latent to lytic replication and will be of value in unraveling the cellular and viral mechanisms involved in activation of KSHV from latency.
ACKNOWLEDGMENTS
We thank Bala Chandran for antibodies to KSHV K8.1 viral protein; Meei Li Huang, Nancy Kiviat, and Donna Kenney for viral PCR testing; and Negin Nowbarn-Nekahi for critical reading of the manuscript.
This work was supported by Public Health Service Grants DE14149-04 to J.V. and AI51946-02 to A.S.J.
REFERENCES
Andreoni, M., G. El-Sawaf, G. Rezza, B. Ensoli, E. Nicastri, L. Ventura, L. Ercoli, L. Sarmati, and G. Rocchi. 1999. High seroprevalence of antibodies to human herpesvirus-8 in Egyptian children: evidence of nonsexual transmission. J. Natl. Cancer Inst. 91:465-469.
Andreoni, M., L. Sarmati, E. Nicastri, G. El Sawaf, M. El Zalabani, I. Uccella, R. Bugarini, S. G. Parisi, and G. Rezza. 2002. Primary human herpesvirus 8 infection in immunocompetent children. JAMA 287:1295-1300.
Asselineau, D., and M. Prunieras. 1984. Reconstruction of ‘simplified’ skin: control of fabrication. Br. J. Dermatol. 111(Suppl. 27):219-222.
Bell, E., S. Sher, B. Hull, C. Merrill, S. Rosen, A. Chamson, D. Asselineau, L. Dubertret, B. Coulomb, C. Lapiere, B. Nusgens, and Y. Neveux. 1983. The reconstitution of living skin. J. Investig. Dermatol. 81:2S-10S.
Boeckh, M., M. Huang, J. Ferrenberg, T. Stevens-Ayers, L. Stensland, W. G. Nichols, and L. Corey. 2004. Optimization of quantitative detection of cytomegalovirus DNA in plasma by real-time PCR. J. Clin. Microbiol. 42:1142-1148.
Boldogh, I., P. Szaniszlo, W. A. Bresnahan, C. M. Flaitz, M. C. Nichols, and T. Albrecht. 1996. Kaposi's sarcoma herpesvirus-like DNA sequences in the saliva of individuals infected with human immunodeficiency virus. Clin. Infect. Dis. 23:406-407.
Brayfield, B. P., C. Kankasa, J. T. West, J. Muyanga, G. Bhat, W. Klaskala, C. D. Mitchell, and C. Wood. 2004. Distribution of Kaposi sarcoma-associated herpesvirus/human herpesvirus 8 in maternal saliva and breast milk in Zambia: implications for transmission. J. Infect. Dis. 189:2260-2270.
Calabro, M. L., J. R. Fiore, A. Favero, A. Lepera, A. Saracino, G. Angarano, T. F. Schulz, and L. Chieco-Bianchi. 1999. Detection of human herpesvirus 8 in cervicovaginal secretions and seroprevalence in human immunodeficiency virus type 1-seropositive and -seronegative women. J. Infect. Dis. 179:1534-1537.
Casper, C., M. Redman, M. L. Huang, J. Pauk, T. M. Lampinen, S. E. Hawes, C. W. Critchlow, R. A. Morrow, L. Corey, N. Kiviat, and A. Wald. 2004. HIV infection and human herpesvirus-8 oral shedding among men who have sex with men. J. Acquir. Immune Defic. Syndr. 35:233-238.
Cerimele, F., F. Curreli, S. Ely, A. E. Friedman-Kien, E. Cesarman, and O. Flore. 2001. Kaposi's sarcoma-associated herpesvirus can productively infect primary human keratinocytes and alter their growth properties. J. Virol. 75:2435-2443.
Cesarman, E., Y. Chang, P. S. Moore, J. W. Said, and D. M. Knowles. 1995. Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas. N. Engl. J. Med. 332:1186-1191.
Cesarman, E., P. S. Moore, P. H. Rao, G. Inghirami, D. M. Knowles, and Y. Chang. 1995. In vitro establishment and characterization of two acquired immunodeficiency syndrome-related lymphoma cell lines (BC-1 and BC-2) containing Kaposi's sarcoma-associated herpesvirus-like (KSHV) DNA sequences. Blood 86:2708-2714.
Chandran, B., C. Bloomer, S. R. Chan, L. Zhu, E. Goldstein, and R. Horvat. 1998. Human herpesvirus-8 ORF K8.1 gene encodes immunogenic glycoproteins generated by spliced transcripts. Virology 249:140-149.
Chang, Y., E. Cesarman, M. S. Pessin, F. Lee, J. Culpepper, D. M. Knowles, and P. S. Moore. 1994. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science 266:1865-1869.
Chen, J., K. Ueda, S. Sakakibara, T. Okuno, C. Parravicini, M. Corbellino, and K. Yamanishi. 2001. Activation of latent Kaposi's sarcoma-associated herpesvirus by demethylation of the promoter of the lytic transactivator. Proc. Natl. Acad. Sci. USA 98:4119-4124.
Chinnathambi, S., A. Tomanek-Chalkley, N. Ludwig, E. King, R. DeWaard, G. Johnson, P. W. Wertz, and J. R. Bickenbach. 2003. Recapitulation of oral mucosal tissues in long-term organotypic culture. Anat. Rec. A 270:162-174.
Chung, J. H., K. H. Cho, D. Y. Lee, O. S. Kwon, M. W. Sung, K. H. Kim, and H. C. Eun. 1997. Human oral buccal mucosa reconstructed on dermal substrates: a model for oral epithelial differentiation. Arch. Dermatol. Res. 289:677-685.
Ciufo, D. M., J. S. Cannon, L. J. Poole, F. Y. Wu, P. Murray, R. F. Ambinder, and G. S. Hayward. 2001. Spindle cell conversion by Kaposi's sarcoma-associated herpesvirus: formation of colonies and plaques with mixed lytic and latent gene expression in infected primary dermal microvascular endothelial cell cultures. J. Virol. 75:5614-5626.
Clausen, H., P. Vedtofte, D. Moe, E. Dabelsteen, T. T. Sun, and B. Dale. 1986. Differentiation-dependent expression of keratins in human oral epithelia. J. Investig. Dermatol. 86:249-254.
Cooper, D., A. Schermer, and T. T. Sun. 1985. Classification of human epithelia and their neoplasms using monoclonal antibodies to keratins: strategies, applications, and limitations. Lab. Investig. 52:243-256.
Corbellino, M., L. Poirel, G. Bestetti, M. Pizzuto, J. T. Aubin, M. Capra, C. Bifulco, E. Berti, H. Agut, G. Rizzardini, M. Galli, and C. Parravicini. 1996. Restricted tissue distribution of extralesional Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS patients with Kaposi's sarcoma. AIDS Res. Hum. Retrovir. 12:651-657.
Corey, L., S. Brodie, M. L. Huang, D. M. Koelle, and A. Wald. 2002. HHV-8 infection: a model for reactivation and transmission. Rev. Med. Virol. 12:47-63.
del Mar Pena, L. M., and L. A. Laimins. 2001. Differentiation-dependent chromatin rearrangement coincides with activation of human papillomavirus type 31 late gene expression. J. Virol. 75:10005-10013.
de Ruijter, A. J., A. H. van Gennip, H. N. Caron, S. Kemp, and A. B. van Kuilenburg. 2003. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem. J. 370:737-749.
Dukers, N. H., N. Renwick, M. Prins, R. B. Geskus, T. F. Schulz, G. J. Weverling, R. A. Coutinho, and J. Goudsmit. 2000. Risk factors for human herpesvirus 8 seropositivity and seroconversion in a cohort of homosexual men. Am. J. Epidemiol. 151:213-224.
Duus, K. M., V. Lentchitsky, T. Wagenaar, C. Grose, and J. Webster-Cyriaque. 2004. Wild-type Kaposi's sarcoma-associated herpesvirus isolated from the oropharynx of immune-competent individuals has tropism for cultured oral epithelial cells. J. Virol. 78:4074-4084.
Eckert, R. L., J. F. Crish, E. B. Banks, and J. F. Welter. 1997. The epidermis: genes on—genes off. J. Investig. Dermatol. 109:501-509.
Eckert, R. L., J. F. Crish, T. Efimova, S. R. Dashti, A. Deucher, F. Bone, G. Adhikary, G. Huang, R. Gopalakrishnan, and S. Balasubramanian. 2004. Regulation of involucrin gene expression. J. Investig. Dermatol. 123:13-22.
Ganem, D. 1998. Human herpesvirus 8 and its role in the genesis of Kaposi's sarcoma. Curr. Clin. Top. Infect. Dis. 18:237-251.
Gessain, A., P. Mauclere, M. van Beveren, S. Plancoulaine, A. Ayouba, J. L. Essame-Oyono, P. M. Martin, and G. de The. 1999. Human herpesvirus 8 primary infection occurs during childhood in Cameroon, Central Africa. Int. J. Cancer 81:189-192.
Greenspan, J. S., D. Greenspan, E. T. Lennette, D. I. Abrams, M. A. Conant, V. Petersen, and U. K. Freese. 1985. Replication of Epstein-Barr virus within the epithelial cells of oral "hairy" leukoplakia, an AIDS-associated lesion. N. Engl. J. Med. 313:1564-1571.
Gwack, Y., H. Byun, S. Hwang, C. Lim, and J. Choe. 2001. CREB-binding protein and histone deacetylase regulate the transcriptional activity of Kaposi's sarcoma-associated herpesvirus open reading frame 50. J. Virol. 75:1909-1917.
Hansson, A., B. K. Bloor, Y. Haig, P. R. Morgan, J. Ekstrand, and R. C. Grafstrom. 2001. Expression of keratins in normal, immortalized and malignant oral epithelia in organotypic culture. Oral Oncol. 37:419-430.
Hansson, A., B. K. Bloor, Z. Sarang, Y. Haig, P. R. Morgan, H. J. Stark, N. E. Fusenig, J. Ekstrand, and R. C. Grafstrom. 2003. Analysis of proliferation, apoptosis and keratin expression in cultured normal and immortalized human buccal keratinocytes. Eur. J. Oral Sci. 111:34-41.
Hassan, A. H., P. Prochasson, K. E. Neely, S. C. Galasinski, M. Chandy, M. J. Carrozza, and J. L. Workman. 2002. Function and selectivity of bromodomains in anchoring chromatin-modifying complexes to promoter nucleosomes. Cell 111:369-379.
Henderson, S., D. Huen, M. Rowe, C. Dawson, G. Johnson, and A. Rickinson. 1993. Epstein-Barr virus-coded BHRF1 protein, a viral homologue of Bcl-2, protects human B cells from programmed cell death. Proc. Natl. Acad. Sci. USA 90:8479-8483.
Herrmann, K., P. Frangou, J. Middeldorp, and G. Niedobitek. 2002. Epstein-Barr virus replication in tongue epithelial cells. J. Gen. Virol. 83:2995-2998.
Hildebrand, H. C., L. Hakkinen, C. B. Wiebe, and H. S. Larjava. 2002. Characterization of organotypic keratinocyte cultures on de-epithelialized bovine tongue mucosa. Histol. Histopathol. 17:151-163.
Hume, W. J., and C. S. Potten. 1983. Proliferative units in stratified squamous epithelium. Clin. Exp. Dermatol. 8:95-106.
Hummel, M., J. B. Hudson, and L. A. Laimins. 1992. Differentiation-induced and constitutive transcription of human papillomavirus type 31b in cell lines containing viral episomes. J. Virol. 66:6070-6080.
Kalinin, A. E., A. V. Kajava, and P. M. Steinert. 2002. Epithelial barrier function: assembly and structural features of the cornified cell envelope. Bioessays 24:789-800.
Kikuta, H., O. Itakura, T. Ariga, and K. Kobayashi. 1997. Detection of human herpesvirus 8 DNA sequences in peripheral blood mononuclear cells of children. J. Med. Virol. 53:81-84.
Kimura, H., M. Morita, Y. Yabuta, K. Kuzushima, K. Kato, S. Kojima, T. Matsuyama, and T. Morishima. 1999. Quantitative analysis of Epstein-Barr virus load by using a real-time PCR assay. J. Clin. Microbiol. 37:132-136.
Koelle, D. M., M. L. Huang, B. Chandran, J. Vieira, M. Piepkorn, and L. Corey. 1997. Frequent detection of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) DNA in saliva of human immunodeficiency virus-infected men: clinical and immunologic correlates. J. Infect. Dis. 176:94-102.
Lagenaur, L. A., and J. M. Palefsky. 1999. Regulation of Epstein-Barr virus promoters in oral epithelial cells and lymphocytes. J. Virol. 73:6566-6572.
Lampinen, T. M., S. Kulasingam, J. Min, M. Borok, L. Gwanzura, J. Lamb, K. Mahomed, G. B. Woelk, K. B. Strand, M. L. Bosch, D. C. Edelman, N. T. Constantine, D. Katzenstein, and M. A. Williams. 2000. Detection of Kaposi's sarcoma-associated herpesvirus in oral and genital secretions of Zimbabwean women. J. Infect. Dis. 181:1785-1790.
Lane, E. B., J. Bartek, P. E. Purkis, and I. M. Leigh. 1985. Keratin antigens in differentiating skin. Ann. N. Y. Acad. Sci. 455:241-258.
Lania, L., B. Majello, and P. De Luca. 1997. Transcriptional regulation by the Sp family proteins. Int. J. Biochem. Cell Biol. 29:1313-1323.
Li, Q. X., L. S. Young, G. Niedobitek, C. W. Dawson, M. Birkenbach, F. Wang, and A. B. Rickinson. 1992. Epstein-Barr virus infection and replication in a human epithelial cell system. Nature 356:347-350.
Lindberg, K., and J. G. Rheinwald. 1990. Three distinct keratinocyte subtypes identified in human oral epithelium by their patterns of keratin expression in culture and in xenografts. Differentiation 45:230-241.
Longworth, M. S., and L. A. Laimins. 2004. Pathogenesis of human papillomaviruses in differentiating epithelia. Microbiol. Mol. Biol Rev. 68:362-372.
Lu, F., J. Zhou, A. Wiedmer, K. Madden, Y. Yuan, and P. M. Lieberman. 2003. Chromatin remodeling of the Kaposi's sarcoma-associated herpesvirus ORF50 promoter correlates with reactivation from latency. J. Virol. 77:11425-11435.
Lukac, D. M., R. Renne, J. R. Kirshner, and D. Ganem. 1998. Reactivation of Kaposi's sarcoma-associated herpesvirus infection from latency by expression of the ORF 50 transactivator, a homolog of the EBV R protein. Virology 252:304-312.
Mantina, H., C. Kankasa, W. Klaskala, B. Brayfield, J. Campbell, Q. Du, G. Bhat, F. Kasolo, C. Mitchell, and C. Wood. 2001. Vertical transmission of Kaposi's sarcoma-associated herpesvirus. Int. J. Cancer 94:749-752.
Martin, J. N., D. E. Ganem, D. H. Osmond, K. A. Page-Shafer, D. Macrae, and D. H. Kedes. 1998. Sexual transmission and the natural history of human herpesvirus 8 infection. N. Engl. J. Med. 338:948-954.
Mayama, S., L. E. Cuevas, J. Sheldon, O. H. Omar, D. H. Smith, P. Okong, B. Silvel, C. A. Hart, and T. F. Schulz. 1998. Prevalence and transmission of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) in Ugandan children and adolescents. Int. J. Cancer 77:817-820.
Meyers, C., M. G. Frattini, J. B. Hudson, and L. A. Laimins. 1992. Biosynthesis of human papillomavirus from a continuous cell line upon epithelial differentiation. Science 257:971-973.
Mischke, D., T. Genka, G. Wille, H. Lobeck, and A. G. Wild. 1991. Keratins as molecular markers of epithelial differentiation: differential expression in crypt epithelium of human palatine tonsils. Ann. Otol. Rhinol. Laryngol. 100:372-377.
Moll, R., W. W. Franke, D. L. Schiller, B. Geiger, and R. Krepler. 1982. The catalog of human cytokeratins: patterns of expression in normal epithelia, tumors and cultured cells. Cell 31:11-24.
Moses, A. V., K. N. Fish, R. Ruhl, P. P. Smith, J. G. Strussenberg, L. Zhu, B. Chandran, and J. A. Nelson. 1999. Long-term infection and transformation of dermal microvascular endothelial cells by human herpesvirus 8. J. Virol. 73:6892-6902.
Nelson, W. G., and T. T. Sun. 1983. The 50- and 58-kdalton keratin classes as molecular markers for stratified squamous epithelia: cell culture studies. J. Cell Biol. 97:244-251.
Neugebauer, P., B. Bonnekoh, A. Wevers, O. Michel, G. Mahrle, T. Krieg, and E. Stennert. 1996. Human keratinocyte culture from the peritonsillar mucosa. Eur. Arch. Otorhinolaryngol. 253:245-251.
Niedobitek, G., A. Agathanggelou, N. Steven, and L. S. Young. 2000. Epstein-Barr virus (EBV) in infectious mononucleosis: detection of the virus in tonsillar B lymphocytes but not in desquamated oropharyngeal epithelial cells. Mol. Pathol. 53:37-42.
Okazaki, M., K. Yoshimura, Y. Suzuki, and K. Harii. 2003. Effects of subepithelial fibroblasts on epithelial differentiation in human skin and oral mucosa: heterotypically recombined organotypic culture model. Plast. Reconstr. Surg. 112:784-792.
Osmond, D. H., S. Buchbinder, A. Cheng, A. Graves, E. Vittinghoff, C. K. Cossen, B. Forghani, and J. N. Martin. 2002. Prevalence of Kaposi sarcoma-associated herpesvirus infection in homosexual men at beginning of and during the HIV epidemic. JAMA 287:221-225.
Pauk, J., M. L. Huang, S. J. Brodie, A. Wald, D. M. Koelle, T. Schacker, C. Celum, S. Selke, and L. Corey. 2000. Mucosal shedding of human herpesvirus 8 in men. N. Engl. J. Med. 343:1369-1377.
Purkis, P. E., J. B. Steel, I. C. Mackenzie, W. B. Nathrath, I. M. Leigh, and E. B. Lane. 1990. Antibody markers of basal cells in complex epithelia. J. Cell Sci. 97:39-50.
Reibel, J., H. Clausen, B. A. Dale, and S. M. Thacher. 1989. Immunohistochemical analysis of stratum corneum components in oral squamous epithelia. Differentiation 41:237-244.
Renne, R., W. Zhong, B. Herndier, M. McGrath, N. Abbey, D. Kedes, and D. Ganem. 1996. Lytic growth of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) in culture. Nat. Med. 2:342-346.
Rheinwald, J. G., and H. Green. 1975. Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell 6:331-343.
Saunders, N., A. Dicker, C. Popa, S. Jones, and A. Dahler. 1999. Histone deacetylase inhibitors as potential anti-skin cancer agents. Cancer Res 59:399-404.
Schmidt, R., C. Cathelineau, M. T. Cavey, V. Dionisius, S. Michel, B. Shroot, and U. Reichert. 1989. Sodium butyrate selectively antagonizes the inhibitory effect of retinoids on cornified envelope formation in cultured human keratinocytes. J. Cell Physiol. 140:281-287.
Selvaratnam, L., A. T. Cruchley, H. Navsaria, P. W. Wertz, E. P. Hagi-Pavli, I. M. Leigh, C. A. Squier, and D. M. Williams. 2001. Permeability barrier properties of oral keratinocyte cultures: a model of intact human oral mucosa. Oral Dis. 7:252-258.
Sixbey, J. W., J. G. Nedrud, N. Raab-Traub, R. A. Hanes, and J. S. Pagano. 1984. Epstein-Barr virus replication in oropharyngeal epithelial cells. N. Engl. J. Med. 310:1225-1230.
Smedts, F., F. Ramaekers, R. E. Leube, K. Keijser, M. Link, and P. Vooijs. 1993. Expression of keratins 1, 6, 15, 16, and 20 in normal cervical epithelium, squamous metaplasia, cervical intraepithelial neoplasia, and cervical carcinoma. Am. J. Pathol. 142:403-412.
Smith, N. A., C. A. Sabin, R. Gopal, D. Bourboulia, W. Labbet, C. Boshoff, D. Barlow, B. Band, B. S. Peters, A. de Ruiter, D. W. Brown, R. A. Weiss, J. M. Best, and D. Whitby. 1999. Serologic evidence of human herpesvirus 8 transmission by homosexual but not heterosexual sex. J. Infect. Dis. 180:600-606.
Smola, H., G. Thiekotter, and N. E. Fusenig. 1993. Mutual induction of growth factor gene expression by epidermal-dermal cell interaction. J. Cell Biol. 122:417-429.
Sondell, B., P. Dyberg, G. K. Anneroth, P. O. Ostman, and T. Egelrud. 1996. Association between expression of stratum corneum chymotryptic enzyme and pathological keratinization in human oral mucosa. Acta Derm. Venereol. 76:177-181.
Song, M. J., H. J. Brown, T. T. Wu, and R. Sun. 2001. Transcription activation of polyadenylated nuclear RNA by Rta in human herpesvirus 8/Kaposi's sarcoma-associated herpesvirus. J. Virol. 75:3129-3140.
Soulier, J., L. Grollet, E. Oksenhendler, P. Cacoub, D. Cazals-Hatem, P. Babinet, M. F. d'Agay, J. P. Clauvel, M. Raphael, and L. Degos. 1995. Kaposi's sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman's disease. Blood 86:1276-1280.
Squier, C. A., and M. J. Kremer. 2001. Biology of oral mucosa and esophagus. J. Natl. Cancer Inst. Monogr. 2001:7-15.
Staiano-Coico, L., R. E. Helm, C. K. McMahon, I. Pagan-Charry, A. LaBruna, V. Piraino, and P. J. Higgins. 1989. Sodium-N-butyrate induces cytoskeletal rearrangements and formation of cornified envelopes in cultured adult human keratinocytes. Cell Tissue Kinet. 22:361-375.
Sumitomo, S., S. Kumasa, Y. Iwai, and M. Mori. 1986. Involucrin expression in epithelial tumors of oral and pharyngeal mucosa and skin. Oral Surg. Oral Med. Oral Pathol. 62:155-163.
Sun, R., S. F. Lin, L. Gradoville, Y. Yuan, F. Zhu, and G. Miller. 1998. A viral gene that activates lytic cycle expression of Kaposi's sarcoma-associated herpesvirus. Proc. Natl. Acad. Sci. USA 95:10866-10871.
Ten Cate, A. 2003. Ten Cates oral histology, 6th ed. The C.V. Mosby Co., St. Louis, Mo.
Thomas, J. A., D. H. Felix, D. Wray, J. C. Southam, H. A. Cubie, and D. H. Crawford. 1991. Epstein-Barr virus gene expression and epithelial cell differentiation in oral hairy leukoplakia. Am. J. Pathol. 139:1369-1380.
Vieira, J., M. L. Huang, D. M. Koelle, and L. Corey. 1997. Transmissible Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) in saliva of men with a history of Kaposi's sarcoma. J. Virol. 71:7083-7087.
Vieira, J., P. O'Hearn, L. Kimball, B. Chandran, and L. Corey. 2001. Activation of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) lytic replication by human cytomegalovirus. J. Virol. 75:1378-1386.
Vieira, J., and P. M. O'Hearn. 2004. Use of the red fluorescent protein as a marker of Kaposi's sarcoma-associated herpesvirus lytic gene expression. Virology 325:225-240.
West, J. T., and C. Wood. 2003. The role of Kaposi's sarcoma-associated herpesvirus/human herpesvirus-8 regulator of transcription activation (RTA) in control of gene expression. Oncogene 22:5150-5163.
Whitby, D., M. R. Howard, M. Tenant-Flowers, N. S. Brink, A. Copas, C. Boshoff, T. Hatzioannou, F. E. Suggett, D. M. Aldam, A. S. Denton, et al. 1995. Detection of Kaposi sarcoma associated herpesvirus in peripheral blood of HIV-infected individuals and progression to Kaposi's sarcoma. Lancet 346:799-802.
Whitby, D., M. Luppi, C. Sabin, P. Barozzi, A. R. Di Biase, F. Balli, F. Cucci, R. A. Weiss, C. Boshoff, and G. Torelli. 2000. Detection of antibodies to human herpesvirus 8 in Italian children: evidence for horizontal transmission. Br. J. Cancer 82:702-704.
Whitby, D., N. A. Smith, S. Matthews, S. O'Shea, C. A. Sabin, R. Kulasegaram, C. Boshoff, R. A. Weiss, A. de Ruiter, and J. M. Best. 1999. Human herpesvirus 8: seroepidemiology among women and detection in the genital tract of seropositive women. J. Infect. Dis. 179:234-236.
Wild, G. A., and D. Mischke. 1986. Variation and frequency of cytokeratin polypeptide patterns in human squamous non-keratinizing epithelium. Exp. Cell Res. 162:114-126.
Wilson, J. L., S. C. Dollard, L. T. Chow, and T. R. Broker. 1992. Epithelial-specific gene expression during differentiation of stratified primary human keratinocyte cultures. Cell Growth Differ. 3:471-483.
Young, L. S., R. Lau, M. Rowe, G. Niedobitek, G. Packham, F. Shanahan, D. T. Rowe, D. Greenspan, J. S. Greenspan, A. B. Rickinson, et al. 1991. Differentiation-associated expression of the Epstein-Barr virus BZLF1 transactivator protein in oral hairy leukoplakia. J. Virol. 65:2868-2874.
Zerr, D. M., D. Gupta, M. L. Huang, R. Carter, and L. Corey. 2002. Effect of antivirals on human herpesvirus 6 replication in hematopoietic stem cell transplant recipients. Clin. Infect. Dis. 34:309-317.
Zhu, L., V. Puri, and B. Chandran. 1999. Characterization of human herpesvirus-8 K8.1A/B glycoproteins by monoclonal antibodies. Virology 262:237-249.(Andrew S. Johnson, Nicole)
濠电姷鏁告慨鐑藉极閹间礁纾块柟瀵稿Т缁躲倝鏌﹀Ο渚&婵炲樊浜濋崑鎰版偣閸ヮ亜鐨烘い锔诲幖閳规垿鎮╃紒妯婚敪濠电偛鐪伴崐婵嬨€佸鑸电劶鐎广儱妫涢崢鍗炩攽閻愭潙鐏ョ€规洦鍓熷鎼佸Χ閸℃劒绨婚梺鍐叉惈閸燁偊宕㈤幘顔界厸閻忕偛澧介埥澶愭煃閽樺妯€鐎规洩绻濋幃娆忣啅椤斾粙鐛庨梻鍌欑閹碱偊藝椤愶箑鐤炬繝濠傚悩濞戙垹绀冩い鏃囧閹芥洖鈹戦悙鏉戠仸閼裤倝鏌$€n亪鍙勯柣鎿冨亰瀹曞爼濡搁敃鈧惃鎴︽⒑缁嬫鍎愰柟姝屽吹缁骞掗弬鍝勪壕闁挎繂绨肩花浠嬫煕閺冩挾鐣辨い顏勫暣婵″爼宕卞Δ鈧ḿ鎴︽⒑缁嬫鍎愰柟鐟版喘瀵鈽夐姀鈥充汗閻庤娲栧ú銏ゅ焵椤掍礁绗氬ǎ鍥э躬瀹曪絾寰勬繝鍌ゆ綋闂備礁鐤囬~澶愬垂閸喚鏆﹂柛顐f礀閻撴﹢鏌i幇闈涘濞存粍鐟╁铏规嫚閺屻儺鈧绱掗悩鑼х€规洘娲熷畷锟犳倷瀹ュ棛鈽夐柍钘夘樀婵偓闁绘﹢娼ф慨锔戒繆閻愵亜鈧牜鏁幒妞濆洭顢涢悙鏉戔偓鍫曟煥閺冨倻鎽傛繛鍫滅矙閺岋綁骞囬浣叉灆闂佺ǹ瀛╁褰掋€冮妷鈺傚€烽柛娆忣槸閺嬬姴顪冮妶鍐ㄧ仾婵☆偄鍟悾鐑藉Ω閳哄﹥鏅i梺缁樺姍濞佳囧箹缁嬪簱鏀介柣妯虹仛閺嗏晠鏌涚€n剙鈻堟鐐存崌椤㈡棃宕卞Δ鍐摌闂備浇顕栭崹濂告倶閹邦優娲敂閸曨収鍚呴梻浣虹帛閿氶柛姘e亾缂備焦顨呴ˇ闈涱潖濞差亜绠伴幖杈剧悼閻g敻姊洪悷鏉挎Щ妞ゆ垵顦悾鐑芥偨缁嬪潡鍞跺┑鐘焺娴滄繂螞閸愵喖鏋侀柛灞剧矌妞规娊鎮楅敐搴濈盎濠碘€炽偢濮婄粯鎷呴崨濠冨枑闂佺ǹ顑囬崑銈夌嵁閹版澘绠柦妯侯槼閹芥洟姊洪崫鍕偍闁搞劌缍婇悰顕€濮€閵堝棛鍘搁梺鎼炲劗閺呮稑鐡┑鐘殿暯閳ь剝灏欓惌娆撴煛瀹€瀣М濠殿喒鍋撻梺瀹犳〃閼宠埖绂掗埡鍐=闁稿本姘ㄥ瓭闂佹寧娲︽禍顏堟偘椤旂晫绡€闁搞儯鍔嶅▍銏ゆ⒑缂佹〒鍦焊濞嗘挻鍋柍褜鍓氭穱濠囨倷椤忓嫧鍋撻弽顓炵濡わ絽鍟壕濠氭煟閺傛寧鎲告い鈺冨厴閺岀喖骞嶉纰辨毉闂佹娊鏀遍崹鍧楀蓟濞戙垺鏅滈悹鍥ㄥ絻缁楀矂姊洪懡銈呮瀾闁革絾娼欑叅妞ゅ繐瀚€瑜旈弻娑㈠Ψ椤旀儳甯ュ┑鐐跺亹閸犳牕顫忓ú顏勭闁告瑥顦伴崕鎾绘⒑閻熸澘鏆辨慨姗堝閹广垹顫濋懜纰樻嫽婵炶揪绲介幊娆掋亹閹烘垵鐝樺銈嗗笒閸婂鎯屽▎鎾寸厵闁绘垶锕╁▓鏇㈡煕婵犲倻浠涢柕鍥у瀵剟宕归瑙勫瘱缂備胶鍋撻崕鎶藉Χ閹间礁钃熼柨鐔哄Т缁€鍐煏婵炲灝鍔楅柛瀣尵缁瑩骞愭惔銏㈡婵犵數濮烽弫鍛婃叏椤撱垹绠柛鎰靛枛缁€瀣煕椤垵浜為柡澶婄秺閺岋絾鎯旈姀鈺佹櫛闂佸摜濮甸悧鐘诲箖閵夆晜鍋傞幖杈剧稻濞堟儳鈹戦濮愪粶闁稿鎸搁埞鎴﹀焺閸愵亝鎲欏銈忛檮婵炲﹪寮诲☉銏犵闁哄鍨规禒鍝ョ磽娴h櫣甯涚紒瀣崌閸┾偓妞ゆ帊鑳堕埊鏇炵暆閿濆懏鍋ラ柟顖氭湰缁绘繈宕戦懞銉︻棃闁诡喒鏅犲畷锝嗗緞鐎n亝鐎鹃梻鍌欐祰濡椼劎娆㈤敓鐘查棷闁挎繂顦埀顑跨窔瀵挳濮€閳╁啯鐝曢梻浣藉Г閿氭い锔跨矙閸┾偓妞ゆ帊鑳堕埥澶愭煃鐟欏嫬鐏撮柡浣哥Ч瀹曠喖顢曢敍鍕礋婵犵數濮幏鍐川椤旂晫褰х紓鍌欑贰閸犳鎮烽敂鐐床婵犻潧顑呴悙濠囨煏婵炑冨暙缁犵増绻濋悽闈浶為柛銊у帶閳绘柨鈽夐姀鈩冩珖闂侀潧鐗嗛ˇ顖毿ч弻銉︾厸闁搞儮鏅涢弸鎴︽煕濞嗗繒绠婚柡宀嬬秮閹垽鎮℃惔婵嗘瀳婵犵妲呴崹顏堝磻閹剧粯鈷掑ù锝堝Г閵嗗啰绱掗埀顒佹媴閾忓湱鐓嬮梺鍦檸閸犳宕戦埡鍌滅瘈闂傚牊渚楅崕蹇曠磼閳ь剟宕橀埞澶哥盎濡炪倖鎸鹃崰搴ㄦ倶閿濆鐓欓悗鐢登归崢瀛樻叏婵犲嫮甯涢柟宄版噽缁數鈧綆浜濋悾浼存⒒娴e憡鎲稿┑顔炬暬楠炴垿宕惰閺嗭附绻濋棃娑冲姛闁汇倐鍋撴繝鐢靛仦閸ㄥ吋銇旈幖浣哥柧闁挎繂娲ㄧ壕浠嬫煕鐏炲墽鎳呴柛鏂跨Т閻f繈鏁愰崨顔间淮闂佺硶鏂傞崹钘夘嚕椤曗偓瀹曞ジ鎮㈤崫鍕闂傚倷鑳堕幊鎾活敋椤撱垹纾块柟瀵稿仧閻熻绻涢幋娆忕仾闁绘挻娲熼弻锟犲磼濮樺彉铏庨梺鍝勬4缂嶄線寮婚垾宕囨殕閻庯綆鍓涢惁鍫ユ倵鐟欏嫭绀冮悽顖涘浮閿濈偛鈹戠€e灚鏅i梺缁橈耿濞佳囩嵁閳ь剛绱撻崒姘偓鎼佸磹閻戣姤鍊块柨鏇炲€堕埀顒€鍟村畷銊ヮ潰閵堝懏鍠橀柡灞芥椤撳ジ宕ㄩ姘曞┑锛勫亼閸婃牜鏁幒妤€绐楁慨姗嗗厳缂傛岸鏌熼柇锕€鏋ょ痪鎯у悑閵囧嫰寮崶褌姹楃紓浣哄Т椤兘寮婚妸銉㈡斀闁割偅绻€濡叉劙姊烘潪鎵槮闁哥喐娼欓悾鐑藉箳閹搭厽鍍甸梺鍛婎殘閸嬬偤骞夐姀銈嗏拻濞达綀妫勯崥鐟扳攽閻愨晛浜剧紓鍌氬€搁崐褰掓嚌妤e啫鐓濈€广儱顦伴弲鏌ユ煕閵夈垺娅囬柣鎾愁儔濮婃椽宕滈懠顒€甯ラ梺鍝ュУ閹瑰洭宕洪埀顒併亜閹哄秶顦︾紒妤佸笚閵囧嫰顢曢敐鍥╃杽濡炪們鍨洪敃銏ゅ箖閳哄拋鏁冮柨婵嗗缂嶅矂姊婚崒姘偓椋庢濮橆兗缂氱憸宥囧弲闂侀潧鐗嗛幃鍨洪鍕庘晠鏌嶆潪鎷屽厡闁告棑绠戦—鍐Χ閸℃鐟ㄩ梺绋款儏濡繂顕i妸鈺傚亱闁割偁鍨婚鏇熺節閵忥絾纭炬い鎴濇搐鐓ら悗鐢电《閸嬫挾鎲撮崟顒傦紭闂佹悶鍔忔慨銈嗙┍婵犲洤閱囬柡鍥╁仧閸婄偤姊洪棃娴ㄥ綊宕曢悜妯侯嚤闁哄洢鍨洪悡鐔兼煟閺傛寧鎲搁柣顓炶嫰椤儻顦虫い銊ョ墦瀵偊顢欓崲澶嬫瀹曨亪宕橀鍡忔(婵犵數濮伴崹濂稿春閺嶎厼绀夐柡宥庡幖绾惧鏌ㄥ┑鍡╂Ч闁绘挻娲熼弻鏇熷緞濡櫣浠╁銈傛櫓閸撴瑩鍩ユ径鎰闁荤喐澹嗗В銏㈢磽娴d粙鍝洪悽顖涱殔宀h儻绠涘☉妯溾晝鎲歌箛娑欏仼濠靛倸鎲¢埛鎴犳喐閻楀牆淇俊顐e灥閳规垿鎮欓埡浣峰闂傚倷娴囬鏍窗濡ゅ啫鍨濋柟鎹愵嚙缁犳澘鈹戦悩鎻掓殭缂佸墎鍋涢…璺ㄦ崉閾忓湱浼囧┑顔硷功閸嬫挾鎹㈠┑瀣仺闂傚牊绋愮划璺侯渻閵堝棙澶勯柛妯圭矙楠炲牓濡搁敂鍝勪簼闂佸憡鍔戦崝宥呂i鍕拺闁告繂瀚埢澶愭煕濡亽鍋㈢€规洖缍婂畷鎺楁倷鐎电ǹ甯惧┑鐘垫暩閸嬬喖宕戦幘鏂ユ瀺闁糕剝绋掗悡鏇㈡倵閿濆啫濡煎┑鈥炽偢濡焦寰勯幇顓犲弳濠电娀娼уΛ娆撳闯瑜版帗鐓涢柛鈩冪懃閺嬫垵菐閸パ嶈含妞ゃ垺绋戦~婵嬪础閻愨晛寰嶅┑鐘愁問閸犳牠鏁冮妷銉富濞寸姴顑冮埀顑跨窔瀵挳濮€閳╁啯鐝抽梻浣虹《閸撴繈鎮烽敃鍌ゆ晣濠电姵纰嶉埛鎺懨归敐鍫燁仩閻㈩垱鐩弻鐔哄枈閸楃偘绨介梺鐟扮畭閸ㄥ綊鍩為幋鐘亾閿濆骸澧紒渚婄畵閺岋絾鎯旈婊呅i梺鍝ュУ閻楁粎鍒掓繝姘櫜闁糕剝鐟ч惁鍫熺節閻㈤潧孝闁稿﹥鎮傞、鏃堫敃閿旂晫鍘甸梺鍝勵儛閸嬪嫭鎱ㄩ崒娑欏弿濠电姴鍟妵婵囶殽閻愭潙濮堥柟顖涙閺佹劙宕掑☉杈ㄧ秾闂傚倸鍊风粈渚€骞栭锔藉亱婵犲﹤鐗嗙粈鍫ユ煟閺冨牜妫戦柡鍡畵閺岋綁鎮㈢粙娆炬闂佸壊鍋掓禍顏堝蓟濞戙垹鍗抽柕濞垮劚鐎涳絽鈹戦悙瀛樼稇閻庢凹鍓熼垾鏃堝礃椤斿槈褔鏌涢埄鍐炬畼闁荤喐鍔欏铏圭磼濡椽鍤嬬紓浣哄У閹告悂顢氶敐澶婄缂佹稑顑嗛弲婊堟⒑閸撴彃浜為柛鐘虫崌瀹曘垽鏌嗗鍡忔嫼闂佽崵鍠撴晶妤呭箚閸垻纾煎璺侯儐缂嶆垿鏌i敐鍥ㄦ毈鐎规洖宕埥澶娾枎閹存繂绗氶梺鑽ゅ枑缁秶鍒掗幘宕囨殾婵犲﹤鍠氬ḿ鈺傘亜閹烘埈妲归柛宥囨暬濮婃椽妫冨ù銉ョ墦瀵彃饪伴崼婵堬紱闂佺ǹ鐬奸崑鐐烘偂閵夆晜鐓熼柡鍌涘閹牏鈧稒绻堥弻锝夋偄閸濄儲鍤傜紓浣哄У閹瑰洭鎮伴鈧浠嬵敇閻愭鍟囨俊鐐€栭幐楣冨磻閻斿摜顩锋い鏍ㄧ矌绾捐棄霉閿濆棗绲诲ù婊呭亾缁绘繈濮€閿濆棛銆愰梺鎰佸灡濞叉繈濡甸幇鏉跨闁瑰濯Σ顖炴⒒娴e懙鍦崲濡ゅ懎纾婚柟閭﹀厴閺嬫棃鏌曢崼婵愭Ч闁抽攱鍨圭槐鎾存媴鐠愵垳绱扮紓浣哄У閻楃娀寮婚敐澶婄厸濠电姴鍊绘禒鈺呮⒑娴兼瑧鎮奸柛蹇旓耿瀵偊骞樼紒妯轰汗闂佽偐鈷堥崜锕€危娴煎瓨鈷掑ù锝堟娴滃綊鏌嶅畡鎵ⅵ鐎规洘绮岄~婵囷紣濠靛洦娅撻梻浣侯攰閹活亪姊介崟顖氱厱闁硅揪闄勯崑锝夋煕閵夘垳宀涢柛瀣崌閹煎綊顢曢妶鍕寜闂傚倸鍊风粈渚€骞夐敓鐘茶摕闁靛⿵瀵屽▓浠嬫煙闂傚顦﹂柣鎾寸箞閺岀喖骞戦幇闈涙缂備胶濯寸紞渚€寮婚敐澶婄疀妞ゆ挾鍠撶粙鍥ь渻閵堝懎顒㈤柟鍛婃倐閸╃偤骞嬮敂钘変汗闁荤姴娉ч崘褏鐭楅梻鍌欑閹芥粓宕抽妷鈺佸瀭闁割偅娲滃畵渚€鏌涢埄鍐槈缁炬儳鍚嬮幈銊╂晲鎼粹€崇缂傚倸绉撮敃顏堢嵁閸愵喖鐓涢柛娑卞幘椤斿矂姊洪崷顓炲妺缂佽鍊规穱濠囨煥鐎n剛鐦堥梺姹囧灲濞佳嗏叴闂備胶枪椤戝棝骞戦崶顒€钃熼柣鏂跨殱閺嬫棃鏌涢…鎴濇灍闁诲繑鎸剧槐鎺撱偅閸愵亞鏆紓浣哄У閻楃姴顕f繝姘亜閻炴稈鈧厖澹曢梺姹囧灮濞呫儵鎮烽悧鍫熺槑闂備浇宕甸崰鎰垝鎼淬垺娅犳俊銈呮噺閸嬪倿鏌ㄩ悢鍝勑㈤柦鍐枑缁绘盯骞嬪▎蹇曚患缂備胶濮垫繛濠囧蓟閿熺姴纾兼慨妯块哺閻ㄦ垿姊洪崫鍕靛剰闁瑰啿閰i崺鐐哄箣閿旇棄鈧兘鏌涘▎蹇fЦ婵炲拑缍佸缁樻媴缁嬭法鐩庣紓浣藉皺閸嬫挾绮氭潏銊х瘈闁搞儜鍜佸晪婵$偑鍊栧Λ浣规叏閵堝應鏋嶉柕蹇嬪€栭埛鎴︽偣閹帒濡兼繛鍛姍閺岀喖宕欓妶鍡楊伓Otolaryngology, University of Washington, Seattle, Washington 98195
ABSTRACT
The oral cavity has been identified as the major site for the shedding of infectious Kaposi's sarcoma-associated herpesvirus (KSHV). While KSHV DNA is frequently detected in the saliva of KSHV seropositive persons, it does not appear to replicate in salivary glands. Some viruses employ the process of epithelial differentiation for productive viral replication. To test if KSHV utilizes the differentiation of oral epithelium as a mechanism for the activation of lytic replication and virus production, we developed an organotypic raft culture model of epithelium using keratinocytes from human tonsils. This system produced a nonkeratinized stratified squamous oral epithelium in vitro, as demonstrated by the presence of nucleated cells at the apical surface; the expression of involucrin and keratins 6, 13, 14, and 19; and the absence of keratin 1. The activation of KSHV lytic-gene expression was examined in this system using rKSHV.219, a recombinant virus that expresses the green fluorescent protein during latency from the cellular EF-1 promoter and the red fluorescent protein (RFP) during lytic replication from the viral early PAN promoter. Infection of keratinocytes with rKSHV.219 resulted in latent infection; however, when these keratinocytes differentiated into a multilayered epithelium, lytic cycle activation of rKSHV.219 occurred, as evidenced by RFP expression, the expression of the late virion protein open reading frame K8.1, and the production of infectious rKSHV.219 at the epithelial surface. These findings demonstrate that KSHV lytic activation occurs as keratinocytes differentiate into a mature epithelium, and it may be responsible for the presence of infectious KSHV in saliva.
INTRODUCTION
Since its initial description over a decade ago (14), Kaposi's sarcoma-associated herpesvirus (KSHV) has been recognized as a significant viral pathogen, particularly for immunocompromised hosts and persons infected with human immunodeficiency virus type 1 (HIV-1). KSHV is the etiologic agent of Kaposi's sarcoma (KS), having been demonstrated in biopsy specimens of all forms of KS despite distinct differences in the geographic origin, age, and gender of affected persons (29). KSHV is also associated with multicentric Castleman's disease (80) and primary effusion lymphoma (11).
The precise modes of KSHV transmission are not clearly defined; however, epidemiological data suggest that both sexual and nonsexual routes are possible. The association between KSHV transmission and sexual activity has been largely defined in North American men who have sex with men (MSM), where risk factors include sex with a partner who has KS (66), a history of sexually transmitted infections (STIs) (55), and an increased number of sexual partners (55). Other risk factors, however, such as deep kissing with an HIV-positive partner (66) and orogenital contact (25), have led to consideration of an oral source of transmission. In a cohort of MSM from San Francisco, similar prevalences of KSHV infection from 1978 to 1996 were found, despite a reduction in HIV-1 seroprevalence and the institution of "safer" sexual practices (65). This raises the issue that sexual activity may be a marker for other types of intimate contact in this population. In keeping with this idea, PCR-based studies of the male genitourinary tract have described only infrequent, low-level shedding of KSHV DNA in genital secretions (22). Infectious virions have not been demonstrated in semen. Similarly, only infrequent and low-level shedding of KSHV DNA has been observed from male urethral and anorectal secretions (66). KSHV DNA has only rarely been detected in vaginal secretions (8, 46, 93), and heterosexual sex has not been clearly associated with KSHV transmission (76).
Evidence for nonsexual transmission is supported by data derived from studies in Africa, Italy, Egypt, and Japan which document KSHV infection in groups with very low risk of STI, including children (1, 8, 42, 91, 92). In regions of Africa where KSHV is endemic, a cumulative increase in KSHV seroprevalence from birth through adolescence has been observed, with more than 40% of children over 14 years of age infected and many infants seroconverting during the first year of life (2, 30, 56). Evidence for transmission through breast milk has not been found (7), and congenital infection is thought to be rare (54). The fact that children and other groups without risk of STI are infected with KSHV implies salivary transmission, as is seen with other human herpesviruses (HHVs), such as cytomegalovirus, HHV-6, HHV-7, and Epstein-Barr virus (EBV).
KSHV DNA has been detected frequently in human saliva (6, 44), and when found, it occurs at a titer 2 to 3 log units higher than those at other anatomic sites (66). Infectious KSHV has been documented in saliva (87). For these reasons, the oral cavity is unique in its ability to support lytic replication of KSHV. Published studies report that 11% to 68% of HIV-positive MSM infected with KSHV in the absence of KS shed viral DNA in saliva on at least one occasion (9, 22, 66). One study demonstrated that 39% of KSHV-seropositive MSM shed viral DNA from the oral cavity on more than a third of the days sampled, regardless of HIV-1 status (66). Current data indicate that KSHV does not replicate in salivary glands (21, 22). Epithelial cells lining the oral cavity are a probable source, given that nucleated oral epithelial cells from nonkeratinized mucosal surfaces have been shown to support viral infection in vivo, by the detection of KSHV-specific latent and lytic mRNA transcripts (66).
Other viruses, such as EBV (31, 49, 74, 96) and human papillomavirus (HPV) (reviewed in reference 51), undergo lytic viral replication in differentiating epithelia. The organotypic raft culture model, originally developed to study keratinocyte differentiation (70), has been of great value in the study of viral replication in developing epithelium (57). Organotypic raft cultures accurately reproduce the process of epithelial differentiation in vitro, beginning with undifferentiated keratinocyte monolayers that become confluent and polarized, forming tight junctions prior to the initiation of keratinocyte differentiation. A proportion of the keratinocytes remain in the basal layer and maintain proliferative potential, while other cells leave the basal layer, lose mitotic capability, and differentiate into mature epithelial cells, forming the multilayered structure of the epithelium (39). Keratinocytes follow a programmed pattern of gene expression during differentiation, each step of which is characterized by the expression of keratin, and keratin-associated, proteins (59). This in vitro model has demonstrated that HPV productively infects cutaneous and mucosal epithelia by a process that is tightly linked to epithelial differentiation (40).
In order to test the hypothesis that activation of KSHV lytic gene expression occurs during epithelial differentiation, we employed an organotypic raft culture model that utilized keratinocytes isolated from normal tonsils. We found that the process of epithelial differentiation activated KSHV lytic-gene expression and resulted in the production of infectious virus at the epithelial surface.
MATERIALS AND METHODS
Subjects. This study was approved by the Human Subjects Review Committee of the University of Washington. Oral and written informed consent was obtained from all participants. Healthy adults planning elective tonsillectomy were eligible to donate leftover tonsil tissues not required for routine pathology. All tonsillectomy procedures were performed by members of the Department of Otolaryngology at the University of Washington. There were no modifications of routine surgical practices and procedures. Subjects with tonsillar mass or asymmetry were excluded in case they had a carcinoma or lymphoma, but otherwise, there were no specific exclusion criteria.
Tonsil-derived organotypic raft cultures. Tonsil specimens were immediately transported in Dulbecco's modified Eagle's medium (DMEM) (Gibco, Invitrogen Corporation) with 10% fetal bovine serum (FBS) (Gibco, Invitrogen Corporation) plus antibiotics (penicillin-streptomycin) at 4°C and processed on arrival. Tissue specimens were variable in size, with surface areas ranging from approximately 1 to 4 cm2. Epithelial cell sheets were separated from each specimen by careful dissection after overnight incubation in dispase II (24.0 U/ml; Gibco, Invitrogen Corporation) at 4°C. Collected materials were treated with 0.05% trypsin-EDTA (Gibco, Invitrogen Corporation) for 30 min at 37°C to form single-cell suspensions and plated at a concentration of 5 x 105 cells/ml in EpiLife medium (Ca2+; 0.06 M) supplemented with the HKGS kit (Cascade Biologics, Inc. Portland, OR) onto 60-mm culture dishes coated with mouse collagen type IV (BD Biosciences, Bedford, MA). The keratinocytes were placed in a 37°C CO2 incubator and fed every other day with EpiLife medium. The keratinocytes were expanded to 70% confluence and used for the organotypic tissue culture.
Autologous fibroblasts were recovered from each tonsil specimen by placing leftover pieces in a T75 flask and incubating them in DMEM supplemented with 10% FBS, L-glutamine, amphotericin B, ciprofloxacin, penicillin, streptomycin, and fresh vitamin C (50 μg/ml; Sigma, St. Louis, MO) at 37°C in a CO2 incubator and were fed every other day. Adherent fibroblasts were expanded and used to seed the bottoms of 12-well plates at a density of 105 cells/well for use in the raft culture model. Expanded keratinocyte cultures were trypsinized, washed, and seeded onto type I collagen-coated cell culture inserts (12-well format) at a density of 105 cells/insert in EpiLife medium. The inserts were incubated submerged at 37°C in a humidified 5% CO2 incubator for a period of 8 to 10 days until a confluent monolayer formed. The inserts were then transferred to a fresh well seeded with autologous fibroblasts at a density of 3 x 105 cells/well and submerged in FAD medium (three parts DMEM, one part Ham's F-12 [Gibco, Invitrogen Corporation], 10% FBS, 8.9 ng/ml epidermal growth factor [Sigma, St. Louis, MO], 0.45 ng/ml insulin [Sigma, St. Louis, MO], 0.45 ng/ml hydrocortisone [Sigma, St. Louis, MO], and 0.3 g/ml adenine [Sigma, St. Louis, MO]) for 24 h, followed by removal of the FAD medium from the upper chamber, exposing the keratinocyte layer to air. Once the layer was exposed, FAD medium (0.6 ml/well) was added to the lower chamber daily to ensure the insert was fed only from the bottom. The inserts were incubated at 37°C in a humidified 5% CO2 incubator for 6 days prior to harvest. Half of each insert was snap frozen fresh in Optimal Cutting Temperature medium (Sakura, Tokyo, Japan) and stored at –80°C. The remaining half was fixed in 4% paraformaldehyde for 30 min at room temperature (RT), snap frozen in Optimal Cutting Temperature medium, and stored at –80°C. A cryostat was used to cut 10-μm sections of tissue on silane-coated slides for further studies.
Three organotypic raft cultures (each derived from a separate donor in this study) were tested by PCR for the presence of herpes simplex virus types 1 and 2, varicella-zoster virus, cytomegalovirus, EBV, HHV-6, and high-risk HPV types (including types 6, 11, 16, 18, 31, 33, 35, 39, and 45) and found to be negative. Reference testing was performed by the Molecular Diagnostic Laboratory and the HPV Laboratory at the University of Washington using previously published methodology (5, 9, 43, 97).
Antibody detection of keratin proteins and viral ORF K8.1 late envelope glycoprotein. Frozen sections of organotypic raft cultures were thawed and washed in phosphate-buffered saline (PBS), pH 7.4. Tissue sections for keratin antibody staining were used directly for immunofluorescence histochemistry, whereas those used for detection of ORF K8.1 were air dried and fixed for 30 min in 4% paraformaldehyde at RT. Tissue sections were blocked with Serum-Free Protein Block (DAKO Corporation, Carpinteria, CA). Keratin antibodies were used to characterize organotypic raft cultures, including CK1, clone 34?B4; CK10, clone LHP1; CK4, clone 6B10; CK13, clone KS-1A3; CK5, clone XM26; CK14, clone LL002; CK6, clone LHK6B; CK16, clone LL025; CK19, clone b170; and involucrin, clone SY5,(all from NovoCastra Laboratories, Newcastle, United Kingdom), at dilutions recommended by the manufacturer. ORF K8.1 protein was detected with monoclonal antibody (MAb) A4 diluted 1:20 in PBS (98). Slide sections were incubated with primary antibody for 1 h at RT, washed, and detected with isotype-specific Qdot 655-conjugated goat F(ab')2 anti-mouse immunoglobulin G (IgG) secondary antibody (Quantum Dot Corporation, Hayward, CA) diluted 1:200 for 30 min at RT. Slides were mounted with Vectashield fluorescence mounting medium with DAPI (4',6'-diamidino-2-phenylindole) (Vector Laboratories, Inc., Burlingame, CA). Staining controls for keratin staining included omission of the primary antibody and normal human tonsil epithelium. Staining controls for the ORF K8.1 MAb included omission of the primary antibody, uninfected raft culture sections, and productively infected Vero cells as positive controls.
Antibody detection of viral ORF 73 (LANA) in rKSHV.219-infected oral keratinocytes. The immunofluorescence detection of KSHV ORF 73 protein was carried out with rat MAb to HHV-8 ORF 73 (ABI, Columbia, MD) diluted 1:100 in 10% normal goat serum. Alexa Fluor 647 goat anti-rat IgG (Molecular Probes, Eugene, OR) was used for fluorescence detection. Oral keratinocytes infected with recombinant KSHV.219 (rKSHV.219) for 10 days and split twice over that time were fixed for 30 min in 4% paraformaldehyde at RT, permeabilized with 0.5% Triton X-100 at RT for 15 min, and reacted with rat MAb to ORF 73. The keratinocytes were washed twice with PBS and reacted with Alexa Fluor 647 goat anti-rat IgG diluted 1:200 for 30 min, washed twice with PBS, counterstained with 1 μg/ml DAPI for 1 min, and washed in PBS.
rKSHV.219. rKSHV.219 is a JSC-derived recombinant KSHV that expresses the green fluorescent protein (GFP) during latent and lytic replication from the cellular promoter EF-1 and the red fluorescent protein (RFP) during lytic replication from the viral lytic PAN promoter. Details concerning the construction and production of rKSHV.219 have been previously published (89).
Infection of organotypic raft cultures. Epithelial cell monolayers were infected with rKSHV.219 on cell culture inserts at 70% confluence in EpiLife medium. Inserts were infected from the top with 200 μl of rKSHV.219 with Polybrene (7.5 μg/ml) and incubated at 37°C in a humidified 5% CO2 incubator for 2 h. A single preparation of virus was used for these experiments at a titer of 6 x 105 infectious units (IU)/ml as determined by titering on 293 cells and counting GFP-positive cells. Virus stock was removed, and the insert was washed with sterile PBS twice. The insert was again submerged in EpiLife medium until confluent and processed as described above. The inserts were observed daily for GFP and RFP expression by fluorescence microscopy as evidence of latent or lytic infection of cells.
Determination of IU of rKSHV.219. For the detection of infectious KSHV in the raft cultures, 0.3 ml of medium was placed in the upper chamber 5 days after air exposure and collected after overnight incubation. The determination of IU of rKSHV.219 was performed as previously described (89). Briefly, cells were removed from each supernatant by centrifugation (300 x g; 10 min) and then passed through a 0.45-μm filter and used as rKSHV.219 inocula for the infection of 293 cells. The number of IU of rKSHV.219 was then determined from the number of GFP-positive 293 cells counted 2 days postinfection. To maximize the infection, it was carried out in the presence of Polybrene (7.5 μg/ml) with centrifugation enhancement (450 x g; 20 min), and then the cells were incubated for 3 h before the medium was replaced with fresh medium without Polybrene. Medium supernatant from the rKSHV.219-infected epithelial cells was collected prior to air exposure and used as a control.
Microscopy. A Nikon Eclipse TE2000S inverted fluorescence microscope equipped with filter sets TE2000 DAPI, TE2000 FITC HYQ, TE2000 Texas Red HYQ, and Omega Optical XF305-2 was used. Images were acquired with a Photometrics CoolSNAP cf digital camera and Metavue Imaging software version 6.1.
RESULTS
Development of nonkeratinized stratified squamous epithelium in vitro. An in vitro organotypic raft culture model of oral epithelium was produced using oral keratinocytes isolated from human tonsils. The keratinocytes were expanded ex vivo and plated on cell culture inserts that served as the support for the organotypic culture. Autologous fibroblasts were incorporated into the model by seeding the bottom of each cell culture well. Confluent keratinocyte monolayers were exposed to air and observed daily prior to harvest on the sixth day. Morphological characteristics of the epithelium were assessed by phase microscopy, DAPI staining, and antibody detection of specific markers of epithelial differentiation. Raft cultures developed a multilayered epithelium 8 to 12 layers thick (Fig. 1). Nucleated cells were observed at the surface of the raft culture by DAPI staining (Fig. 1), a feature consistent with nonkeratinized epithelium (81, 85). Types of epithelia and the state of differentiation can be profiled by the expression pattern of keratins, a family of over 30 proteins expressed in specific pairings of type I (acidic) and type II (basic) members (59). To further characterize the in vitro-generated epithelium, cross sections were analyzed with monoclonal antibodies to specific keratins and involucrin. The keratin pair K4 and K13, specific for nonkeratinized oral stratified epithelia (19, 20, 59, 94), was detected in suprabasal layers (K13 is shown in Fig. 1). Conversely, the keratin pair K1 and K10, specifically found in keratinized epithelia (20, 47, 59, 75, 78), was absent (K1 is shown in Fig. 1). The keratin pair K5 and K14, which is found in all types of oral stratified epithelia (61, 67), was present in basal and suprabasal layers of the raft culture, consistent with the tissue of origin (K14 is shown in Fig. 1). Other keratin markers found in differentiated tonsil epithelium, including keratins K6 and K19 (35, 50, 58, 62), were also demonstrated in suprabasal layers of the raft culture (Fig. 1). Involucrin, a keratin-associated protein indicative of epithelial differentiation (41, 68, 83), was observed in the suprabasal layers of the raft culture (Fig. 1). These experiments demonstrated that the epithelial morphology and keratin expression pattern observed in our in vitro organotypic raft culture model was consistent with that of normal human tonsil epithelium (58).
KSHV replication in keratinocytes and in vitro epithelium. In vivo epithelium arises from keratinocytes (progenitor cells) located in the basal layer (39). Similarly, in vitro organotypic raft cultures are formed from undifferentiated keratinocyte monolayers. Therefore, oral keratinocytes, which were cultured to maintain an undifferentiated state, were infected with rKSHV.219 to examine latent and lytic replication. Figure 2A depicts undifferentiated, primary oral keratinocytes 10 days after infection with rKSHV.219 by phase (Fig. 2A, 1) and fluorescence for GFP (Fig. 2A, 2) and RFP (Fig. 2A, 3). No evidence of RFP expression was seen, indicative of latent viral infection (Fig. 2A, 3). In Fig. 2B, the same keratinocytes were examined for LANA expression by phase (Fig. 2B, 1) and by fluorescence for LANA, GFP, and DAPI (Fig. 2B, 2). GFP-expressing cells demonstrated punctate nuclear staining for LANA, while non-GFP-expressing cells did not. The absence of RFP expression and the presence of punctate nuclear LANA staining are indicative of latent viral infection in these undifferentiated keratinocyte monolayers.
Next, this model of oral epithelium was used to assess the impact of epithelial differentiation on KSHV replication. This was accomplished by infecting keratinocytes at 50 to 70% confluence on culture inserts with rKSHV.219 and using the infected keratinocytes to initiate organotypic raft cultures. The infected keratinocytes continued to grow, became confluent in 2 to 3 days, and expressed GFP but not RFP, as seen in an intact keratinocyte monolayer 1 day prior to air exposure (Fig. 3A). This was indicative of latent infection, as has been previously reported for KSHV-infected keratinocyte monolayers (10, 88). This was also similar to long-term cultures of rKSHV.219-infected keratinocytes that were passaged submerged in epithelial medium (Fig. 2). There is a single published report that describes lytic infection of primary epithelial cell monolayers in culture, but it is not currently known whether this observation was due to differences in the virus used or cell culture conditions (26). One day postconfluence, the medium from the top chamber was removed in order to expose the apical surfaces of the keratinocytes to air and to promote differentiation of the epithelium. Intact living cultures were examined daily for GFP and RFP expression. Images of phase, GFP expression, and RFP expression at 3 and 6 days after air exposure are shown (Fig. 3B and C). The expression of RFP from the lytic PAN promoter indicated that activation of lytic-gene expression occurred in the differentiating epithelium.
RFP expression in rKSHV.219-infected cells indicates early gene expression, because it is under the control of the lytic PAN promoter, which is directly activated by Rta (79). To determine if late lytic-gene expression occurred in these epithelial cells, cultures were harvested 6 days after air exposure and intact rafts were analyzed with antibody to the late viral glycoprotein K8.1. K8.1 expression was identified in a subset of cells expressing RFP (Fig. 4). In these cultures, not all cells with KSHV early gene expression progressed to late gene expression, an observation made in other cell types (18, 60, 89), although the reasons are not understood. These experiments demonstrated that the process of epithelial differentiation activated KSHV lytic gene expression. Observations of the intact organotypic raft culture suggested that this was predominately occurring in cells at the apical surface; therefore, the location of lytic-gene expression within the rafts was examined.
To identify the location of cells with lytic-gene expression within the epithelial tissue, rafts were sectioned and examined by fluorescence microscopy for GFP and RFP expression. On cross section, GFP was distributed throughout all layers of the raft culture epithelium but was expressed most intensely at the surface, where squames had become flattened and more densely compacted (Fig. 5B). RFP was observed primarily in cells at the apical surface of the raft culture, indicating that lytic-gene expression was occurring predominantly in differentiated epithelial cells (Fig. 5C). In order to compare the localization of late lytic-protein expression with that of early lytic-gene expression (indicated by RFP), sections of the raft cultures were stained with antibody to the ORF K8.1 protein. As seen for the RFP-positive cells, K8.1 was detected in cells at the apical surface of the epithelium (Fig. 6).
Production of infectious KSHV. Because the eventual outcome of the activation of KSHV lytic-gene expression is the generation of infectious virus, the organotypic raft cultures were tested for the production of infectious KSHV. This was carried out by adding medium to the upper chamber of the cultures 5 days after air exposure. The medium was removed after overnight incubation and used to inoculate 293 cells to determine the presence of IU of rKSHV.219 by examination for GFP-positive 293 cells 2 days postinoculation. Figure 7 shows an image of 293 cells infected with supernatant from one of the rKSHV.219-infected raft cultures. Three raft cultures derived from one tonsil and two from a second tonsil were tested, and all were found to produce infectious virus, ranging from 44 to 165 IU per 0.9-cm2 insert. Medium removed from the upper chamber at the time of air exposure was also used to inoculate 293 cells and was found to be negative. This demonstrated that differentiating keratinocytes activated KSHV lytic replication and became permissive for the production of infectious KSHV.
DISCUSSION
Saliva has been the only readily transmitted bodily fluid proven to contain infectious KSHV (87), and epidemiological data suggest saliva as a likely route of transmission (2, 30, 56), but the source of KSHV in saliva has not been well characterized. Unlike human cytomegalovirus or human herpesvirus 6, current data suggest that KSHV does not replicate in salivary glands (22, 66). EBV and HPV activate lytic replication in developing epithelia, suggesting that KSHV might also replicate in epithelial cells lining the oral cavity. In order to study KSHV replication in oral epithelia, we developed an organotypic raft culture model, derived from human tonsil keratinocytes, that recapitulated the characteristics of a nonkeratinized stratified squamous oral epithelium. Using this model, we examined the impact of epithelial differentiation on KSHV replication. We found that undifferentiated primary keratinocytes maintained latent infection with KSHV; however, the process of epithelial differentiation resulted in the activation of both early and late lytic viral gene expression, with the release of infectious KSHV at the apical surface of the epithelium.
The production of a nonkeratinized organotypic raft culture model of oral epithelium was a critical component of these studies. The organotypic raft culture model is a well-established system for the production of epithelia in vitro. Early raft culture models, which utilized skin keratinocytes to produce multilayered epithelial tissue, exhibited differentiation patterns that resembled the tissue from which the keratinocytes originated (3, 4, 95). Keratinocytes were cultured on dermal equivalents composed of type I collagen and fibroblasts and raised to the air-medium interface to promote differentiation. This model has also been applied to keratinocytes derived from the oral cavity, including buccal (16, 17, 33, 34, 73), lingual (38), and peritonsillar (62) surfaces. Several of these studies underscored the importance of fibroblasts located in adjacent connective tissue, which provide permissive and instructive signals to differentiating keratinocytes (64, 77). In our system, primary keratinocytes isolated from nonkeratinized tonsil epithelium were seeded onto semipermeable cell culture inserts, and a fibroblast feeder layer derived from the tonsil dermis was established in the cell culture well beneath the insert. This format allowed excellent visualization and photography of cultures during development and provided a stable support for histological sectioning of harvested tissue. Raft cultures derived from primary tonsil keratinocytes had the expected morphological characteristics of nonkeratinized oral epithelium (58). An advanced stage of epithelial differentiation was achieved in suprabasal layers of the raft culture, as evidenced by expression of keratins 4 and 13 (59, 94) and involucrin (68). In addition, the presence of nucleated cells at the apical surface, the absence of specific markers of keratinization (keratins 1 and 10), and suprabasal expression of specific markers for nonkeratinized stratified squamous epithelium (keratins 4 and 13) (47) showed this to be an appropriate system to examine KSHV replication in oral epithelia.
We used our in vitro raft culture model of oral epithelium to test the hypothesis that basal keratinocytes, latently infected with KSHV, would activate viral lytic cycle gene expression during the process of epithelial differentiation, as suggested for the activation of EBV in the oral epithelia (49, 96). EBV, a gammaherpesvirus related to KSHV, is found in B cells and epithelial cells and is transmitted in saliva. Although the role of epithelial cells in primary EBV infection remains controversial (37, 63, 74), suprabasal epithelial cells in oral hairy leukoplakia lesions have been shown to replicate viral genomes and to express both lytic (36, 96) and latent (86) cycle antigens. Of value in our study, KSHV readily infected primary oral keratinocytes in culture and established a latent infection, which allowed us to initiate the process of epithelial differentiation using rKSHV.219-infected primary keratinocytes. The use of rKSHV.219 allowed the daily observation of raft cultures during differentiation for the identification of infected cells by GFP expression and for the activation of lytic gene expression indicated by the expression of RFP. While no RFP was seen in the basal layer of the epithelium, the process of keratinocyte differentiation resulted in the expression of RFP by many cells at the upper layer of the culture. While the expression of RFP from the lytic PAN promoter indicates early gene expression, a subset of RFP-positive cells was found to have late lytic gene expression as well, indicated by the presence of the late lytic envelope glycoprotein K8.1. The expression of K8.1 requires viral lytic DNA synthesis and therefore signifies that keratinocyte differentiation activates lytic DNA replication (13). The result of lytic replication is the production of infectious virus. In this in vitro model of oral epithelium, infectious virus was recovered from the apical surface of the raft culture, demonstrating that the complete KSHV lytic replication cycle can be completed as latently infected keratinocytes differentiate into a mature epithelium. Unlike in vitro epithelial models of EBV (49) and HPV (57) infection, which require stimulation with a phorbol ester to induce viral lytic replication, the activation of KSHV lytic-gene expression in differentiating primary keratinocytes is brought about by a natural cellular process.
KSHV lytic-cycle activation is initiated by Rta, the immediate-early transcriptional activator, expressed from open reading frame (ORF) 50 (53, 84, 90). Other agents known to induce KSHV lytic-cycle activation include phorbol esters (12, 69) and DNA-demethylating agents, such as azacytidine (15). Unlike KSHV culture systems that rely on the chemical treatment of cells, activation of KSHV lytic-gene expression in raft culture epithelium is brought about by a natural cellular process. Although the mechanism is unknown, one possible explanation is chromatin remodeling. Chromatin rearrangement around the late promoter region during epithelial differentiation has been described for HPV (23) and suggests that DNA binding of differentiation-specific cellular transcription factors is involved. Transcriptional regulation in eukaryotic cells is heavily dependent upon histone acetylation and methylation (reviewed in reference 24). Keratinocyte differentiation and growth arrest can be induced by inhibitors of histone deacetylase, such as sodium butyrate (72, 82) and tricostatin A (71). The KSHV Rta promoter can also be induced by these histone deacetylase inhibitors (52, 89), and Rta itself is involved in the recruitment of cellular factors (32, 35) that influence chromatin remodeling and access to cellular transcription factors (52). Another possible mechanism is the recruitment of cellular transcription factors at gene promoter sites by regulation of AP1, Sp1, and CCATT/enhancer-binding proteins (27, 28). ORF 50 utilizes a GC box initiator element and binds Sp family transcription factors (52), as do other cellular and viral promoters (48). Sp1 proteins are critical transactivators of regulated keratinocyte gene expression (27), and promoters containing GC box initiator elements may be more active in keratinocytes. For example, the EBV early protein BMRF2, which utilizes a GC box initiator element and is highly expressed in oral hairy leukoplakia lesions, exhibits 10-fold-greater activity in epithelial cells than in lymphocytes (45). The mechanisms of activation (chromatin remodeling and recruitment of cellular transcription factors) are not mutually exclusive, and other factors could be involved. Further experiments will be required to determine the mechanisms that are important for viral activation in differentiating cells.
In summary, this work demonstrates that the process of oral epithelial differentiation can activate KSHV lytic-gene expression. Moreover, because differentiated oral keratinocytes are competent to support productive viral replication with the release of infectious viral particles, oral epithelial cells are a likely source of infectious KSHV present in saliva. The switch between latent and lytic replication is the hallmark of herpesvirus biology, allowing the permanent infection of the host, and is critical to the pathogenesis and transmission of herpesviruses. The results presented here illustrate an in vitro model of a natural cellular process causing the switch from latent to lytic replication and will be of value in unraveling the cellular and viral mechanisms involved in activation of KSHV from latency.
ACKNOWLEDGMENTS
We thank Bala Chandran for antibodies to KSHV K8.1 viral protein; Meei Li Huang, Nancy Kiviat, and Donna Kenney for viral PCR testing; and Negin Nowbarn-Nekahi for critical reading of the manuscript.
This work was supported by Public Health Service Grants DE14149-04 to J.V. and AI51946-02 to A.S.J.
REFERENCES
Andreoni, M., G. El-Sawaf, G. Rezza, B. Ensoli, E. Nicastri, L. Ventura, L. Ercoli, L. Sarmati, and G. Rocchi. 1999. High seroprevalence of antibodies to human herpesvirus-8 in Egyptian children: evidence of nonsexual transmission. J. Natl. Cancer Inst. 91:465-469.
Andreoni, M., L. Sarmati, E. Nicastri, G. El Sawaf, M. El Zalabani, I. Uccella, R. Bugarini, S. G. Parisi, and G. Rezza. 2002. Primary human herpesvirus 8 infection in immunocompetent children. JAMA 287:1295-1300.
Asselineau, D., and M. Prunieras. 1984. Reconstruction of ‘simplified’ skin: control of fabrication. Br. J. Dermatol. 111(Suppl. 27):219-222.
Bell, E., S. Sher, B. Hull, C. Merrill, S. Rosen, A. Chamson, D. Asselineau, L. Dubertret, B. Coulomb, C. Lapiere, B. Nusgens, and Y. Neveux. 1983. The reconstitution of living skin. J. Investig. Dermatol. 81:2S-10S.
Boeckh, M., M. Huang, J. Ferrenberg, T. Stevens-Ayers, L. Stensland, W. G. Nichols, and L. Corey. 2004. Optimization of quantitative detection of cytomegalovirus DNA in plasma by real-time PCR. J. Clin. Microbiol. 42:1142-1148.
Boldogh, I., P. Szaniszlo, W. A. Bresnahan, C. M. Flaitz, M. C. Nichols, and T. Albrecht. 1996. Kaposi's sarcoma herpesvirus-like DNA sequences in the saliva of individuals infected with human immunodeficiency virus. Clin. Infect. Dis. 23:406-407.
Brayfield, B. P., C. Kankasa, J. T. West, J. Muyanga, G. Bhat, W. Klaskala, C. D. Mitchell, and C. Wood. 2004. Distribution of Kaposi sarcoma-associated herpesvirus/human herpesvirus 8 in maternal saliva and breast milk in Zambia: implications for transmission. J. Infect. Dis. 189:2260-2270.
Calabro, M. L., J. R. Fiore, A. Favero, A. Lepera, A. Saracino, G. Angarano, T. F. Schulz, and L. Chieco-Bianchi. 1999. Detection of human herpesvirus 8 in cervicovaginal secretions and seroprevalence in human immunodeficiency virus type 1-seropositive and -seronegative women. J. Infect. Dis. 179:1534-1537.
Casper, C., M. Redman, M. L. Huang, J. Pauk, T. M. Lampinen, S. E. Hawes, C. W. Critchlow, R. A. Morrow, L. Corey, N. Kiviat, and A. Wald. 2004. HIV infection and human herpesvirus-8 oral shedding among men who have sex with men. J. Acquir. Immune Defic. Syndr. 35:233-238.
Cerimele, F., F. Curreli, S. Ely, A. E. Friedman-Kien, E. Cesarman, and O. Flore. 2001. Kaposi's sarcoma-associated herpesvirus can productively infect primary human keratinocytes and alter their growth properties. J. Virol. 75:2435-2443.
Cesarman, E., Y. Chang, P. S. Moore, J. W. Said, and D. M. Knowles. 1995. Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas. N. Engl. J. Med. 332:1186-1191.
Cesarman, E., P. S. Moore, P. H. Rao, G. Inghirami, D. M. Knowles, and Y. Chang. 1995. In vitro establishment and characterization of two acquired immunodeficiency syndrome-related lymphoma cell lines (BC-1 and BC-2) containing Kaposi's sarcoma-associated herpesvirus-like (KSHV) DNA sequences. Blood 86:2708-2714.
Chandran, B., C. Bloomer, S. R. Chan, L. Zhu, E. Goldstein, and R. Horvat. 1998. Human herpesvirus-8 ORF K8.1 gene encodes immunogenic glycoproteins generated by spliced transcripts. Virology 249:140-149.
Chang, Y., E. Cesarman, M. S. Pessin, F. Lee, J. Culpepper, D. M. Knowles, and P. S. Moore. 1994. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science 266:1865-1869.
Chen, J., K. Ueda, S. Sakakibara, T. Okuno, C. Parravicini, M. Corbellino, and K. Yamanishi. 2001. Activation of latent Kaposi's sarcoma-associated herpesvirus by demethylation of the promoter of the lytic transactivator. Proc. Natl. Acad. Sci. USA 98:4119-4124.
Chinnathambi, S., A. Tomanek-Chalkley, N. Ludwig, E. King, R. DeWaard, G. Johnson, P. W. Wertz, and J. R. Bickenbach. 2003. Recapitulation of oral mucosal tissues in long-term organotypic culture. Anat. Rec. A 270:162-174.
Chung, J. H., K. H. Cho, D. Y. Lee, O. S. Kwon, M. W. Sung, K. H. Kim, and H. C. Eun. 1997. Human oral buccal mucosa reconstructed on dermal substrates: a model for oral epithelial differentiation. Arch. Dermatol. Res. 289:677-685.
Ciufo, D. M., J. S. Cannon, L. J. Poole, F. Y. Wu, P. Murray, R. F. Ambinder, and G. S. Hayward. 2001. Spindle cell conversion by Kaposi's sarcoma-associated herpesvirus: formation of colonies and plaques with mixed lytic and latent gene expression in infected primary dermal microvascular endothelial cell cultures. J. Virol. 75:5614-5626.
Clausen, H., P. Vedtofte, D. Moe, E. Dabelsteen, T. T. Sun, and B. Dale. 1986. Differentiation-dependent expression of keratins in human oral epithelia. J. Investig. Dermatol. 86:249-254.
Cooper, D., A. Schermer, and T. T. Sun. 1985. Classification of human epithelia and their neoplasms using monoclonal antibodies to keratins: strategies, applications, and limitations. Lab. Investig. 52:243-256.
Corbellino, M., L. Poirel, G. Bestetti, M. Pizzuto, J. T. Aubin, M. Capra, C. Bifulco, E. Berti, H. Agut, G. Rizzardini, M. Galli, and C. Parravicini. 1996. Restricted tissue distribution of extralesional Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS patients with Kaposi's sarcoma. AIDS Res. Hum. Retrovir. 12:651-657.
Corey, L., S. Brodie, M. L. Huang, D. M. Koelle, and A. Wald. 2002. HHV-8 infection: a model for reactivation and transmission. Rev. Med. Virol. 12:47-63.
del Mar Pena, L. M., and L. A. Laimins. 2001. Differentiation-dependent chromatin rearrangement coincides with activation of human papillomavirus type 31 late gene expression. J. Virol. 75:10005-10013.
de Ruijter, A. J., A. H. van Gennip, H. N. Caron, S. Kemp, and A. B. van Kuilenburg. 2003. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem. J. 370:737-749.
Dukers, N. H., N. Renwick, M. Prins, R. B. Geskus, T. F. Schulz, G. J. Weverling, R. A. Coutinho, and J. Goudsmit. 2000. Risk factors for human herpesvirus 8 seropositivity and seroconversion in a cohort of homosexual men. Am. J. Epidemiol. 151:213-224.
Duus, K. M., V. Lentchitsky, T. Wagenaar, C. Grose, and J. Webster-Cyriaque. 2004. Wild-type Kaposi's sarcoma-associated herpesvirus isolated from the oropharynx of immune-competent individuals has tropism for cultured oral epithelial cells. J. Virol. 78:4074-4084.
Eckert, R. L., J. F. Crish, E. B. Banks, and J. F. Welter. 1997. The epidermis: genes on—genes off. J. Investig. Dermatol. 109:501-509.
Eckert, R. L., J. F. Crish, T. Efimova, S. R. Dashti, A. Deucher, F. Bone, G. Adhikary, G. Huang, R. Gopalakrishnan, and S. Balasubramanian. 2004. Regulation of involucrin gene expression. J. Investig. Dermatol. 123:13-22.
Ganem, D. 1998. Human herpesvirus 8 and its role in the genesis of Kaposi's sarcoma. Curr. Clin. Top. Infect. Dis. 18:237-251.
Gessain, A., P. Mauclere, M. van Beveren, S. Plancoulaine, A. Ayouba, J. L. Essame-Oyono, P. M. Martin, and G. de The. 1999. Human herpesvirus 8 primary infection occurs during childhood in Cameroon, Central Africa. Int. J. Cancer 81:189-192.
Greenspan, J. S., D. Greenspan, E. T. Lennette, D. I. Abrams, M. A. Conant, V. Petersen, and U. K. Freese. 1985. Replication of Epstein-Barr virus within the epithelial cells of oral "hairy" leukoplakia, an AIDS-associated lesion. N. Engl. J. Med. 313:1564-1571.
Gwack, Y., H. Byun, S. Hwang, C. Lim, and J. Choe. 2001. CREB-binding protein and histone deacetylase regulate the transcriptional activity of Kaposi's sarcoma-associated herpesvirus open reading frame 50. J. Virol. 75:1909-1917.
Hansson, A., B. K. Bloor, Y. Haig, P. R. Morgan, J. Ekstrand, and R. C. Grafstrom. 2001. Expression of keratins in normal, immortalized and malignant oral epithelia in organotypic culture. Oral Oncol. 37:419-430.
Hansson, A., B. K. Bloor, Z. Sarang, Y. Haig, P. R. Morgan, H. J. Stark, N. E. Fusenig, J. Ekstrand, and R. C. Grafstrom. 2003. Analysis of proliferation, apoptosis and keratin expression in cultured normal and immortalized human buccal keratinocytes. Eur. J. Oral Sci. 111:34-41.
Hassan, A. H., P. Prochasson, K. E. Neely, S. C. Galasinski, M. Chandy, M. J. Carrozza, and J. L. Workman. 2002. Function and selectivity of bromodomains in anchoring chromatin-modifying complexes to promoter nucleosomes. Cell 111:369-379.
Henderson, S., D. Huen, M. Rowe, C. Dawson, G. Johnson, and A. Rickinson. 1993. Epstein-Barr virus-coded BHRF1 protein, a viral homologue of Bcl-2, protects human B cells from programmed cell death. Proc. Natl. Acad. Sci. USA 90:8479-8483.
Herrmann, K., P. Frangou, J. Middeldorp, and G. Niedobitek. 2002. Epstein-Barr virus replication in tongue epithelial cells. J. Gen. Virol. 83:2995-2998.
Hildebrand, H. C., L. Hakkinen, C. B. Wiebe, and H. S. Larjava. 2002. Characterization of organotypic keratinocyte cultures on de-epithelialized bovine tongue mucosa. Histol. Histopathol. 17:151-163.
Hume, W. J., and C. S. Potten. 1983. Proliferative units in stratified squamous epithelium. Clin. Exp. Dermatol. 8:95-106.
Hummel, M., J. B. Hudson, and L. A. Laimins. 1992. Differentiation-induced and constitutive transcription of human papillomavirus type 31b in cell lines containing viral episomes. J. Virol. 66:6070-6080.
Kalinin, A. E., A. V. Kajava, and P. M. Steinert. 2002. Epithelial barrier function: assembly and structural features of the cornified cell envelope. Bioessays 24:789-800.
Kikuta, H., O. Itakura, T. Ariga, and K. Kobayashi. 1997. Detection of human herpesvirus 8 DNA sequences in peripheral blood mononuclear cells of children. J. Med. Virol. 53:81-84.
Kimura, H., M. Morita, Y. Yabuta, K. Kuzushima, K. Kato, S. Kojima, T. Matsuyama, and T. Morishima. 1999. Quantitative analysis of Epstein-Barr virus load by using a real-time PCR assay. J. Clin. Microbiol. 37:132-136.
Koelle, D. M., M. L. Huang, B. Chandran, J. Vieira, M. Piepkorn, and L. Corey. 1997. Frequent detection of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) DNA in saliva of human immunodeficiency virus-infected men: clinical and immunologic correlates. J. Infect. Dis. 176:94-102.
Lagenaur, L. A., and J. M. Palefsky. 1999. Regulation of Epstein-Barr virus promoters in oral epithelial cells and lymphocytes. J. Virol. 73:6566-6572.
Lampinen, T. M., S. Kulasingam, J. Min, M. Borok, L. Gwanzura, J. Lamb, K. Mahomed, G. B. Woelk, K. B. Strand, M. L. Bosch, D. C. Edelman, N. T. Constantine, D. Katzenstein, and M. A. Williams. 2000. Detection of Kaposi's sarcoma-associated herpesvirus in oral and genital secretions of Zimbabwean women. J. Infect. Dis. 181:1785-1790.
Lane, E. B., J. Bartek, P. E. Purkis, and I. M. Leigh. 1985. Keratin antigens in differentiating skin. Ann. N. Y. Acad. Sci. 455:241-258.
Lania, L., B. Majello, and P. De Luca. 1997. Transcriptional regulation by the Sp family proteins. Int. J. Biochem. Cell Biol. 29:1313-1323.
Li, Q. X., L. S. Young, G. Niedobitek, C. W. Dawson, M. Birkenbach, F. Wang, and A. B. Rickinson. 1992. Epstein-Barr virus infection and replication in a human epithelial cell system. Nature 356:347-350.
Lindberg, K., and J. G. Rheinwald. 1990. Three distinct keratinocyte subtypes identified in human oral epithelium by their patterns of keratin expression in culture and in xenografts. Differentiation 45:230-241.
Longworth, M. S., and L. A. Laimins. 2004. Pathogenesis of human papillomaviruses in differentiating epithelia. Microbiol. Mol. Biol Rev. 68:362-372.
Lu, F., J. Zhou, A. Wiedmer, K. Madden, Y. Yuan, and P. M. Lieberman. 2003. Chromatin remodeling of the Kaposi's sarcoma-associated herpesvirus ORF50 promoter correlates with reactivation from latency. J. Virol. 77:11425-11435.
Lukac, D. M., R. Renne, J. R. Kirshner, and D. Ganem. 1998. Reactivation of Kaposi's sarcoma-associated herpesvirus infection from latency by expression of the ORF 50 transactivator, a homolog of the EBV R protein. Virology 252:304-312.
Mantina, H., C. Kankasa, W. Klaskala, B. Brayfield, J. Campbell, Q. Du, G. Bhat, F. Kasolo, C. Mitchell, and C. Wood. 2001. Vertical transmission of Kaposi's sarcoma-associated herpesvirus. Int. J. Cancer 94:749-752.
Martin, J. N., D. E. Ganem, D. H. Osmond, K. A. Page-Shafer, D. Macrae, and D. H. Kedes. 1998. Sexual transmission and the natural history of human herpesvirus 8 infection. N. Engl. J. Med. 338:948-954.
Mayama, S., L. E. Cuevas, J. Sheldon, O. H. Omar, D. H. Smith, P. Okong, B. Silvel, C. A. Hart, and T. F. Schulz. 1998. Prevalence and transmission of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) in Ugandan children and adolescents. Int. J. Cancer 77:817-820.
Meyers, C., M. G. Frattini, J. B. Hudson, and L. A. Laimins. 1992. Biosynthesis of human papillomavirus from a continuous cell line upon epithelial differentiation. Science 257:971-973.
Mischke, D., T. Genka, G. Wille, H. Lobeck, and A. G. Wild. 1991. Keratins as molecular markers of epithelial differentiation: differential expression in crypt epithelium of human palatine tonsils. Ann. Otol. Rhinol. Laryngol. 100:372-377.
Moll, R., W. W. Franke, D. L. Schiller, B. Geiger, and R. Krepler. 1982. The catalog of human cytokeratins: patterns of expression in normal epithelia, tumors and cultured cells. Cell 31:11-24.
Moses, A. V., K. N. Fish, R. Ruhl, P. P. Smith, J. G. Strussenberg, L. Zhu, B. Chandran, and J. A. Nelson. 1999. Long-term infection and transformation of dermal microvascular endothelial cells by human herpesvirus 8. J. Virol. 73:6892-6902.
Nelson, W. G., and T. T. Sun. 1983. The 50- and 58-kdalton keratin classes as molecular markers for stratified squamous epithelia: cell culture studies. J. Cell Biol. 97:244-251.
Neugebauer, P., B. Bonnekoh, A. Wevers, O. Michel, G. Mahrle, T. Krieg, and E. Stennert. 1996. Human keratinocyte culture from the peritonsillar mucosa. Eur. Arch. Otorhinolaryngol. 253:245-251.
Niedobitek, G., A. Agathanggelou, N. Steven, and L. S. Young. 2000. Epstein-Barr virus (EBV) in infectious mononucleosis: detection of the virus in tonsillar B lymphocytes but not in desquamated oropharyngeal epithelial cells. Mol. Pathol. 53:37-42.
Okazaki, M., K. Yoshimura, Y. Suzuki, and K. Harii. 2003. Effects of subepithelial fibroblasts on epithelial differentiation in human skin and oral mucosa: heterotypically recombined organotypic culture model. Plast. Reconstr. Surg. 112:784-792.
Osmond, D. H., S. Buchbinder, A. Cheng, A. Graves, E. Vittinghoff, C. K. Cossen, B. Forghani, and J. N. Martin. 2002. Prevalence of Kaposi sarcoma-associated herpesvirus infection in homosexual men at beginning of and during the HIV epidemic. JAMA 287:221-225.
Pauk, J., M. L. Huang, S. J. Brodie, A. Wald, D. M. Koelle, T. Schacker, C. Celum, S. Selke, and L. Corey. 2000. Mucosal shedding of human herpesvirus 8 in men. N. Engl. J. Med. 343:1369-1377.
Purkis, P. E., J. B. Steel, I. C. Mackenzie, W. B. Nathrath, I. M. Leigh, and E. B. Lane. 1990. Antibody markers of basal cells in complex epithelia. J. Cell Sci. 97:39-50.
Reibel, J., H. Clausen, B. A. Dale, and S. M. Thacher. 1989. Immunohistochemical analysis of stratum corneum components in oral squamous epithelia. Differentiation 41:237-244.
Renne, R., W. Zhong, B. Herndier, M. McGrath, N. Abbey, D. Kedes, and D. Ganem. 1996. Lytic growth of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) in culture. Nat. Med. 2:342-346.
Rheinwald, J. G., and H. Green. 1975. Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell 6:331-343.
Saunders, N., A. Dicker, C. Popa, S. Jones, and A. Dahler. 1999. Histone deacetylase inhibitors as potential anti-skin cancer agents. Cancer Res 59:399-404.
Schmidt, R., C. Cathelineau, M. T. Cavey, V. Dionisius, S. Michel, B. Shroot, and U. Reichert. 1989. Sodium butyrate selectively antagonizes the inhibitory effect of retinoids on cornified envelope formation in cultured human keratinocytes. J. Cell Physiol. 140:281-287.
Selvaratnam, L., A. T. Cruchley, H. Navsaria, P. W. Wertz, E. P. Hagi-Pavli, I. M. Leigh, C. A. Squier, and D. M. Williams. 2001. Permeability barrier properties of oral keratinocyte cultures: a model of intact human oral mucosa. Oral Dis. 7:252-258.
Sixbey, J. W., J. G. Nedrud, N. Raab-Traub, R. A. Hanes, and J. S. Pagano. 1984. Epstein-Barr virus replication in oropharyngeal epithelial cells. N. Engl. J. Med. 310:1225-1230.
Smedts, F., F. Ramaekers, R. E. Leube, K. Keijser, M. Link, and P. Vooijs. 1993. Expression of keratins 1, 6, 15, 16, and 20 in normal cervical epithelium, squamous metaplasia, cervical intraepithelial neoplasia, and cervical carcinoma. Am. J. Pathol. 142:403-412.
Smith, N. A., C. A. Sabin, R. Gopal, D. Bourboulia, W. Labbet, C. Boshoff, D. Barlow, B. Band, B. S. Peters, A. de Ruiter, D. W. Brown, R. A. Weiss, J. M. Best, and D. Whitby. 1999. Serologic evidence of human herpesvirus 8 transmission by homosexual but not heterosexual sex. J. Infect. Dis. 180:600-606.
Smola, H., G. Thiekotter, and N. E. Fusenig. 1993. Mutual induction of growth factor gene expression by epidermal-dermal cell interaction. J. Cell Biol. 122:417-429.
Sondell, B., P. Dyberg, G. K. Anneroth, P. O. Ostman, and T. Egelrud. 1996. Association between expression of stratum corneum chymotryptic enzyme and pathological keratinization in human oral mucosa. Acta Derm. Venereol. 76:177-181.
Song, M. J., H. J. Brown, T. T. Wu, and R. Sun. 2001. Transcription activation of polyadenylated nuclear RNA by Rta in human herpesvirus 8/Kaposi's sarcoma-associated herpesvirus. J. Virol. 75:3129-3140.
Soulier, J., L. Grollet, E. Oksenhendler, P. Cacoub, D. Cazals-Hatem, P. Babinet, M. F. d'Agay, J. P. Clauvel, M. Raphael, and L. Degos. 1995. Kaposi's sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman's disease. Blood 86:1276-1280.
Squier, C. A., and M. J. Kremer. 2001. Biology of oral mucosa and esophagus. J. Natl. Cancer Inst. Monogr. 2001:7-15.
Staiano-Coico, L., R. E. Helm, C. K. McMahon, I. Pagan-Charry, A. LaBruna, V. Piraino, and P. J. Higgins. 1989. Sodium-N-butyrate induces cytoskeletal rearrangements and formation of cornified envelopes in cultured adult human keratinocytes. Cell Tissue Kinet. 22:361-375.
Sumitomo, S., S. Kumasa, Y. Iwai, and M. Mori. 1986. Involucrin expression in epithelial tumors of oral and pharyngeal mucosa and skin. Oral Surg. Oral Med. Oral Pathol. 62:155-163.
Sun, R., S. F. Lin, L. Gradoville, Y. Yuan, F. Zhu, and G. Miller. 1998. A viral gene that activates lytic cycle expression of Kaposi's sarcoma-associated herpesvirus. Proc. Natl. Acad. Sci. USA 95:10866-10871.
Ten Cate, A. 2003. Ten Cates oral histology, 6th ed. The C.V. Mosby Co., St. Louis, Mo.
Thomas, J. A., D. H. Felix, D. Wray, J. C. Southam, H. A. Cubie, and D. H. Crawford. 1991. Epstein-Barr virus gene expression and epithelial cell differentiation in oral hairy leukoplakia. Am. J. Pathol. 139:1369-1380.
Vieira, J., M. L. Huang, D. M. Koelle, and L. Corey. 1997. Transmissible Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) in saliva of men with a history of Kaposi's sarcoma. J. Virol. 71:7083-7087.
Vieira, J., P. O'Hearn, L. Kimball, B. Chandran, and L. Corey. 2001. Activation of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) lytic replication by human cytomegalovirus. J. Virol. 75:1378-1386.
Vieira, J., and P. M. O'Hearn. 2004. Use of the red fluorescent protein as a marker of Kaposi's sarcoma-associated herpesvirus lytic gene expression. Virology 325:225-240.
West, J. T., and C. Wood. 2003. The role of Kaposi's sarcoma-associated herpesvirus/human herpesvirus-8 regulator of transcription activation (RTA) in control of gene expression. Oncogene 22:5150-5163.
Whitby, D., M. R. Howard, M. Tenant-Flowers, N. S. Brink, A. Copas, C. Boshoff, T. Hatzioannou, F. E. Suggett, D. M. Aldam, A. S. Denton, et al. 1995. Detection of Kaposi sarcoma associated herpesvirus in peripheral blood of HIV-infected individuals and progression to Kaposi's sarcoma. Lancet 346:799-802.
Whitby, D., M. Luppi, C. Sabin, P. Barozzi, A. R. Di Biase, F. Balli, F. Cucci, R. A. Weiss, C. Boshoff, and G. Torelli. 2000. Detection of antibodies to human herpesvirus 8 in Italian children: evidence for horizontal transmission. Br. J. Cancer 82:702-704.
Whitby, D., N. A. Smith, S. Matthews, S. O'Shea, C. A. Sabin, R. Kulasegaram, C. Boshoff, R. A. Weiss, A. de Ruiter, and J. M. Best. 1999. Human herpesvirus 8: seroepidemiology among women and detection in the genital tract of seropositive women. J. Infect. Dis. 179:234-236.
Wild, G. A., and D. Mischke. 1986. Variation and frequency of cytokeratin polypeptide patterns in human squamous non-keratinizing epithelium. Exp. Cell Res. 162:114-126.
Wilson, J. L., S. C. Dollard, L. T. Chow, and T. R. Broker. 1992. Epithelial-specific gene expression during differentiation of stratified primary human keratinocyte cultures. Cell Growth Differ. 3:471-483.
Young, L. S., R. Lau, M. Rowe, G. Niedobitek, G. Packham, F. Shanahan, D. T. Rowe, D. Greenspan, J. S. Greenspan, A. B. Rickinson, et al. 1991. Differentiation-associated expression of the Epstein-Barr virus BZLF1 transactivator protein in oral hairy leukoplakia. J. Virol. 65:2868-2874.
Zerr, D. M., D. Gupta, M. L. Huang, R. Carter, and L. Corey. 2002. Effect of antivirals on human herpesvirus 6 replication in hematopoietic stem cell transplant recipients. Clin. Infect. Dis. 34:309-317.
Zhu, L., V. Puri, and B. Chandran. 1999. Characterization of human herpesvirus-8 K8.1A/B glycoproteins by monoclonal antibodies. Virology 262:237-249.(Andrew S. Johnson, Nicole)