Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Gene Expression Profiling of Human Vaginal Cells In Vitro Discriminates Compounds with Pro-Inflammatory and Mucosa-Altering Properties: Novel Biomarkers for Preclinical Testing of HIV Microbicide Candidates

  • Irina A. Zalenskaya ,

    ZalensIA@EVMS.edu (IAZ); DoncelGF@EVMS.edu (GFD)

    Affiliation CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America

  • Theresa Joseph,

    Current address: Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America

    Affiliation CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America

  • Jasmin Bavarva,

    Current address: Biospecimen Research Group, Leidos Biomedical Research, Inc. for the Frederick National Laboratory for Cancer Research, Rockville, Maryland, United States of America

    Affiliation Virginia Bioinformatics Institute, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America

  • Nazita Yousefieh,

    Affiliation CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America

  • Suzanne S. Jackson,

    Affiliation CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America

  • Titilayo Fashemi,

    Affiliation Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America

  • Hidemi S. Yamamoto,

    Affiliation Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America

  • Robert Settlage,

    Affiliation Virginia Bioinformatics Institute, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America

  • Raina N. Fichorova,

    Affiliation Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America

  • Gustavo F. Doncel

    ZalensIA@EVMS.edu (IAZ); DoncelGF@EVMS.edu (GFD)

    Affiliation CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America

Abstract

Background

Inflammation and immune activation of the cervicovaginal mucosa are considered factors that increase susceptibility to HIV infection. Therefore, it is essential to screen candidate anti-HIV microbicides for potential mucosal immunomodulatory/inflammatory effects prior to further clinical development. The goal of this study was to develop an in vitro method for preclinical evaluation of the inflammatory potential of new candidate microbicides using a microarray gene expression profiling strategy.

Methods

To this end, we compared transcriptomes of human vaginal cells (Vk2/E6E7) treated with well-characterized pro-inflammatory (PIC) and non-inflammatory (NIC) compounds. PICs included compounds with different mechanisms of action. Gene expression was analyzed using Affymetrix U133 Plus 2 arrays. Data processing was performed using GeneSpring 11.5 (Agilent Technologies, Santa Clara, CA).

Results

Microarraray comparative analysis allowed us to generate a panel of 20 genes that were consistently deregulated by PICs compared to NICs, thus distinguishing between these two groups. Functional analysis mapped 14 of these genes to immune and inflammatory responses. This was confirmed by the fact that PICs induced NFkB pathway activation in Vk2 cells. By testing microbicide candidates previously characterized in clinical trials we demonstrated that the selected PIC-associated genes properly identified compounds with mucosa-altering effects. The discriminatory power of these genes was further demonstrated after culturing vaginal cells with vaginal bacteria. Prevotella bivia, prevalent bacteria in the disturbed microbiota of bacterial vaginosis, induced strong upregulation of seven selected PIC-associated genes, while a commensal Lactobacillus gasseri associated to vaginal health did not cause any changes.

Conclusions

In vitro evaluation of the immunoinflammatory potential of microbicides using the PIC-associated genes defined in this study could help in the initial screening of candidates prior to entering clinical trials. Additional characterization of these genes can provide further insight into the cervicovaginal immunoinflammatory and mucosal-altering processes that facilitate or limit HIV transmission with implications for the design of prevention strategies.

Introduction

According to the latest global estimates from UNAIDS, there were about 35 million people living with HIV in 2013 (UNAIDS. GAP report; 2014, www.unaids.org). During the same time, there were around 2.1 million new HIV infections, and 1.5 million people died of HIV-related causes. Women represent slightly more than half of the infected population worldwide and approximately 60% in sub-Saharan Africa, where gender inequalities increase women’s vulnerability to HIV. In some regions young women and adolescent girls account for a disproportionate number of new infections among young people and those living with HIV (UNAIDS. GAP report, 2014, www.unaids.org).

The main route of male-to-female HIV transmission is through the epithelium of the female genital tract exposed to virus-containing semen [1]. Vaginal topical antiviral microbicides represent a novel female-controlled method of prevention of sexual transmission of HIV. During the past 20 years, numerous microbicide products have been developed, with a handful of them reaching advanced clinical trials [25]. Thus far, only the nucleotide reverse transcriptase inhibitor (NRTI), tenofovir, tested in the CAPRISA 004 study demonstrated a moderate reduction in HIV acquisition [6]. All other candidate microbicides were ineffective in preventing primary HIV infection. Moreover, one of the first candidate microbicides, a non-ionic detergent nonoxynol-9 (N-9), increased the risk of HIV infection among frequent users. It was later demonstrated that this compound altered epithelial integrity and induced a mucosal immunoinflammatory response leading to recruitment of immune cells, which are potential targets for HIV infection [7]. Two other microbicide candidates, C31G (Savvy) and cellulose sulfate (CS) which failed in phase III clinical trials also proved to alter the mucosal microenvironment [8,9].

Currently, there are multiple candidate microbicides in preclinical development and a handful of products undergoing clinical studies [3,10]. The new candidates have diverse modes of action, including inhibition of HIV attachment, fusion and entry, replication, integration, and other less defined mechanisms. Preclinical studies of products with multiple mechanisms of action are also in progress [3,10]. Lessons learned from the failures of the first generation microbicides underscore the need for early testing of candidate microbicides for their possible adverse effects on the cervicovaginal and rectal mucosae.

The healthy female genital epithelium presents a strong barrier to viral invasion. HIV transmission is estimated to be 1–2 cases per 1000 coital acts [11,12]. However, affected by complex host and viral factors, transmission rates can be much higher [11]. Increased HIV-1 cervicovaginal transmission is strongly associated with inflammation and general immune activation of the cervicovaginal mucosa—conditions that stimulate influx of HIV target immune cells to the mucosal surface, increase the receptivity of these cells to HIV-1, and are often accompanied by epithelial lesions, which facilitate HIV-1 access to its target cells [1324]. To avoid these adverse effects, the proinflammatory potential of microbicide candidates should be evaluated early during their primary screening [25,26]. Currently, cell-based, explant-based and animal-based models are used to define and preclinically assess potential biomarkers of inflammation, which include cytokines, chemokines and some other molecules [25,2732]. Biomarker selection in these assays relies on prior knowledge of their involvement in inflammatory responses.

In this study, we set out to identify novel biomarkers for preclinical in vitro assessment of the pro-inflammatory/immunomodulatory potential of microbicides using microarray hybridization technology (gene expression profiling). Our goal was to generate unique gene expression profiles for compounds that were known to cause mucosal inflammatory/immunomodulatory response in order to use these profiles for the characterization of microbicide candidates. To this end, transcriptomes of immortalized human vaginal epithelial (Vk2/E6E7) cells exposed to well-known pro-inflammatory/immunomodulatory compounds (PIC) and non-inflammatory compounds (NIC) were compared. Using microarray technology we identified 20 genes that were consistently deregulated by treatment with PICs but not with NICs, thus presenting a signature specific to compounds with potential pro-inflammatory/ immunomodulatory effects on the cervicovaginal mucosa.

Materials and Methods

Materials

TLR ligands, Pam3CSK4 (Pam), imiquimod (IMQ) were purchased from Invivogen (San Diego, CA), macrophage activating lipopeptide 2 (MALP2) was purchased from Alexis Biochemicals (Enzo Life Sciences, Plymouth Meeting, PA). TNF-α was purchased from R&D systems (Minneapolis, MN). N-9 was a gift from OrthoMcNeil Corporation. Other compounds/candidate microbicides were acquired as follows: hydroxyethyl cellulose (HEC)—from Hercules (Hopewell, VA), cellulose sulfate (CS) and dextran sulfate (DS)—from Dextran Products (Scarborough, Ontario), PRO2000—from Indevus Pharmaceuticals (Lexington, MA), tenofovir (TFV)—from Gilead (Foster City, CA), C31G from Biosyn Inc. (Huntington Valley, PA), UC-781—from Regis (Morton Grove, IL), emtricibine (FTC)—gift from Dr. Parang (Keykavous Parang, University of Rhode Island).

Doses of compounds (Table 1) have been defined in our previous studies such that their cytotoxicity, if any, would not exceed 15% [33,34].

Cell culture, treatment with compounds

The vaginal keratinocyte cell line Vk2/E6E7 is a gift from Dr. Fichorova (Brigham and Women’s Hospital [35]. This cell line closely mimics the morphological and functional characteristics of native human vaginal tissue and primary epithelial cells of origin [29,3641]. Cells were maintained in keratinocyte serum-free medium (Gibco, Invitrogen, Grand Island, NY) supplemented with bovine pituitary extract (50 μg/ml), epidermal growth factor (0.1 ng/ml), penicillin-streptomycin (1%), and CaCl2 (0.4 mM). Cells were grown to ~70–80% confluence and treated for 6 h with agents presented in Table 1. Antibiotics were omitted in bacterial colonization experiments. Cells were collected and RNA extracted for gene expression analysis as described below.

Bacterial colonization

Bacterial colonization of Vk2/E6E7 cells was performed in the Laboratory of Genital Tract Biology, Brigham and Women’s Hospital (BWH) as described in detail elsewhere [41].

Lactobacillus isolate and Prevotella bivia were obtained from vaginal swabs and phenotypically characterized by classic microbiology techniques as described before [42]. Because conventional methods of phenotyping cannot identify Lactobacilli to the species level, sequencing of the 16S rRNA gene of Lactobacillus was performed at the Center for Clinical and Translational Metagenomics at BWH. The genetic analysis classified the Lactobacillus isolate as L. gasseri with 99% identity. Vaginal epithelial cultures (Vk2/E6E7 cells) were colonized with L. gasseri, representing the Gram-positive facultative anaerobes typical for the normal vaginal flora [4346], or Prevotella (P.) bivia representing the anaerobic Gram-negative rods associated with bacterial vaginosis (BV) [4749].

RNA isolation

Prior to RNA isolation, Vk2 cells were rinsed three times with cold PBS. Total RNA was then extracted with Trizol (Invitrogen Life Technologies, Carlsbad, CA) and purified using RNeasy mini kit columns from Qiagen Sciences (Qiagen, Valencia, CA) according to the manufacturer’s instructions.

The integrity of total RNA was qualified by Agilent Bioanalyzer 2100 capillary electrophoresis and input amount quantified by Nanodrop ND-1000 Spectrophotometer.

Microarray expression profiling

Microarray mRNA expression profiling was performed by Asuragen, Inc. (Austin, TX). The mRNA was amplified into cRNA and biotin-labeled using modified MessageAmp-based protocols (Ambion Inc., Austin, TX). Labeled cRNA was fragmented, and hybridized to Affymetrix HG-U133 Plus 2.0 arrays (Affymetrix) according to the standard Affymetrix protocol. The U133 Plus 2.0 chip contains more than 56,000 probesets and includes 38,500 well characterized human genes and expressed sequence tags. Affymetrix raw data were acquired using GeneChip operating software (GCOS 1.3) to yield CEL files.

Data normalization and statistical analysis

Data were processed and analyzed using GeneSpring 11.5 (Agilent Technologies, Santa Clara, CA). The background subtraction, normalization, and log base 2 transformation of gene signals were carried out using the Robust Multi-array Analysis (RMA) summarization algorithm [50]

For statistical analysis, one-way ANOVA was used for multiple group comparison, followed by multiple testing correction setting the false discovery rate (FDR) at 0.05 using the Benjamini and Hochberg method [51]. Genes/probesets with FDR corrected p-values < 0.05 were considered statistically significant. Next, pair-wise comparisons were performed on probesets having statistical significance to detect how these probesets differ in a treatment condition versus control. Probesets showing fold change differences >2 were considered as differentially expressed. Only those probes that were consistently detected as differential and statistically significant were considered as altered.

Quantitative real-time PCR (qPCR)

Microarray data for selected genes were validated using quantitative real-time PCR. For the cDNA synthesis 1 μg of total RNA was reverse transcribed using the Reverse Transcription System kit from Promega Corp. (Madison, WI USA). Reverse transcription was primed with Oligo(dT)15 in a total volume of 20 μl according to the manufacturer’s protocol.

Quantitative real-time PCR (qPCR) was performed on Roche LightCycler Carousel-based system using 4.05 software. PCR was performed in 20 μl reaction volume containing 1 μl cDNA using LightCycler FastStart DNA Master SYBR Green I (Roche, Indianapolis, IN) according to manufacturer’s instructions. The sequences of the primers used for PCR are presented in S1 Table.

Thermocycler parameters were 95°C for 10 min followed by 45 cycles at 95°C for 10 s, 55°C for 5 s, 72°C for 15 s. Each sample was run in triplicates, and normalized to GAPDH RNA used as the endogenous control. The threshold cycle (Ct) of GAPDH was used to normalize target gene expression (ΔCt). The relative change in gene expression was calculated using the 2-ΔΔCt method [52].

Experiments were performed at least three times, the mean and SD were calculated using Graph Pad software (version 5.01).

Nuclear and cytoplasmic fractions separation, NFkB activation assay

Vk2/E6E7 cells were exposed to PICs and NICs and harvested within 60 min followed the treatment with 10–15 min interval. Nuclear and cytoplasmic fractions were obtained as described [53] with slight modifications. Cells were rinsed twice with ice-cold PBS, scraped, collected by centrifugation at 1000 x g for 5 min, resuspended in STC buffer (0.25 M sucrose, 10 mM tris-HCl, pH 7.5, 3 mM CaCl2) supplemented with protease inhibitor cocktail (BD Bioscience, Bedford, MA). Equal volume of STC buffer, containing 1% Triton X-100 was added to the cell suspension, followed by incubation on ice for 10 min. Nuclear and cytoplasmic fractions were separated by centrifugation at 500 x g for 7 min. Nuclear pellet was resuspended in STC, equal volume of 2 x SDS loading buffer was added to the nuclear and cytoplasmic fractions. Cytoplasmic and nuclear proteins were separated by 10% SDS polyacrylamide gel and transferred to PVDF membrane. IκB-α (degradation in cytoplasm) and NFκB/p 65 (nuclear translocation) were assayed by immunoblotting, as described earlier [34] using corresponding antibodies, both at 1:1000 dilution. Rabbit anti-human polyclonal antibodies, NFκB/p65 and IκB-α, were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA)

Results

Identification of genes discriminating between PIC and NIC by gene profiling of Vk2 cells

To develop biomarkers that may discriminate pro-inflammatory/ immunomodulatory compounds (PICs) from non-inflammatory compounds (NICs) in in vitro studies, we employed comparative transcriptional profiling using a microarray technique (Affymetrix U133 Plus 2.0). Well-characterized PICs and NICs (Table 1) were applied to human immortalized vaginal epithelial cells (Vk2/E6E7) that have been selected as an in vitro test model. Vk2 cells are similar in characteristics to the cells of the tissue of origin, and have proved to be an adequate model to study vaginal epithelial responses to topical agents [29,35,40]. With the goal of identifying genes that are consistently and significantly changed by PIC as compared to NIC treatments, Vk2 cells were exposed to the following compounds. PICs included a pleiotropic proinflammatory cytokine TNF-α, TLR ligands (TLR-L) such as Pam3CSK4, MALP2, and imiquimod, and the detergent N-9 (for a total of 33 arrays), while NICs were represented by HEC and CMC (18 arrays) as well as growth medium control (GM) (54 arrays) (Table 1). A total of 105 Affymetrix U133 Plus 2.0 arrays were processed and analyzed as described in the Materials and Methods section.

The numbers of differentially expressed probesets (fold change cut off = 2, FDR adjusted p-value < 0.05) revealed by microarray analysis in the PIC group were: 215 for TNF-α, 332 for Pam3CSK4, 629 for MALP2, 774 for imiquimod, and 957 for N-9. In the NIC group deregulation was much more modest: 80 probesets for HEC and 20 probesets for CMC (Fig 1, Table 2; S2S8 Tables).

thumbnail
Fig 1. Diagrams showing the number of significantly altered probesets indentified by microarray gene profiling of Vk2 cells exposed to PIC and NIC.

Total number of the altered probeserts for each treatment/category is shown in brackets (a gene can be represented by more than one probeset).

https://doi.org/10.1371/journal.pone.0128557.g001

thumbnail
Table 2. Number of differentially expressed probesets in treatment groups compared to control (growth medium).

https://doi.org/10.1371/journal.pone.0128557.t002

Furthermore, the fold changes induced by NICs did not exceed 3.3, while the fold changes in the PIC group reached values >25 for TNF- α, >30 for Pam3CK4, >48 for MALP2, >22 for imiquimod, and >23 for N-9 (S2S8 Tables).

With the aim of identifying a group of shared deregulated genes, we first analyzed differentially expressed genes within the TLR-L group. There were 116 differentially expressed probesets in common among Pam3CSK4, MALP2 and imiquimod (Fig 1A; S9 Table).

Next, we compared gene expression after all PIC treatments, TNF-a, TLR-L, and N-9. As a result, 27 probesets with significantly altered expression in all PIC treatments were identified (Fig 1B, S10 Table). These 27 probesets encompass 22 genes (some genes are represented by more than one probeset). Next, the expression profile of PICs was compared against that of NICs. Overlap between the PIC-common probesets and probesets differentially expressed by NICs revealed two genes from the PIC list that were also differentially expressed in NICs (Fig 1C, S10 Table). Upregulated by PICs, MMP1 was also upregulated in HEC treatments (fold change 3.3). COL8A1 was downregulted by PICs and both NICs, HEC and CMC (fold change -3.0 and -2.3 respectively). Therefore these genes were excluded from the final list of the PIC-specific probesets. The final list of the probesets altered by PIC but not NIC treatments contains 25 probesets mapped to 20 PIC discriminatory genes (PIC-DG) (Table 3, S10 Table).

thumbnail
Table 3. Genes differenially expressed in VK2 cells treated with proinflammatory/immunomodulatory compounds.

https://doi.org/10.1371/journal.pone.0128557.t003

Of these genes, 19 genes are upregulated, and one is downregulated. A heatmap and hierarchical clustering generated from expression profiles of 20 discriminatory genes common to all PIC treatments of Vk2 cells clearly demonstrates segregation of PICs from NICs (Fig 2).

thumbnail
Fig 2. Hierarchical clustering of the 20 discriminatory genes was performed using GeneSpring 11.5 as described in Materials and Methods.

Columns represent treatments, rows represent genes. Gene expression levels are indicated by color: red is for upregulation and green is for downregulation. Expression data are averages from at least six experiments/microarrays for each treatment. Hierarchical clustering based on the discriminatory genes demonstrates sharp segregation of the PIC vs NIC treatments.

https://doi.org/10.1371/journal.pone.0128557.g002

Functional analysis of the discriminatory genes

Functional analysis of the PIC-DGs was performed using Ingenuity Pathway Analysis (IPA). All 20 genes were mapped and found to be network-eligible; 18 genes were function/pathway eligible. The top biofunctions for PIC-DGs, as revealed by IPA, are presented in Table 4. Inflammatory response was indicated as the most significant biofunction (p value 3.05E-07–2.05E-0.3), thus supporting pro-inflammatory activity as the principal common feature for the selected PICs, in spite of belonging to diverse molecular and mechanistic categories. Inflammatory/immune modulating response function was attributed by IPA to twelve genes from the PIC-DG list.

thumbnail
Table 4. Functional categories of the PIC/NIC discriminatory genesa.

https://doi.org/10.1371/journal.pone.0128557.t004

We extended the IPA-generated list of genes belonging to this category by including PIC-upregulated MAFB and SERPINB2 (see below and Discussion) based on recently published studies indicating their link to the inflammatory/ immunomodulatory processes [54,55]. Four genes in the category of inflammatory/immune response, CCL20, IL8, CXCL2, CXCL3, encode for chemokines that are involved in immune cells trafficking. IL8 is also known as one of the major mediators of inflammatory responses. Other genes in this category include OLR1, PTGS2, TNFAIP3, CYLD and NFKBIZ. OLR1 is a receptor for oxidatively modified low density lipoprotein (oxLDL) that upon binding by its ligand induces activation of NFκB (nuclear factor kappa B), the master complex in immunoinflammatory response. PTGS2 (prostaglandin synthase-2) also called COX-2 (cyclooxygenase-2) is one of the key enzymes involved in inflammatory processes [56,57]. TNFAIP3 (A20) and CYLD are deubiquitinating enzymes that play a prominent role in inflammatory signaling by regulating NFkB activation. Another molecule that is tightly associated with NFκB regulation is NFKBIZ, NFκB inhibitor-zeta, also known as I kappa B zeta.

Other important top biofunctions/diseases include immune cell trafficking, cellular movement, cellular growth and proliferation, cancer, and cell death (Table 4).

Functional analysis (by IPA) of relationships between PIC-DGs highlighted inflammatory response and cellular movement as the highest scored hypothetical gene networks (Fig 3).

thumbnail
Fig 3. Top network of Vk2 PIC/NIC discriminatory genes generated by Ingenuity Pathway Analysis.

Red/pink color indicates upregulation of the genes (microarray data). Connections of NFkB complex with other genes is shown in blue color.

https://doi.org/10.1371/journal.pone.0128557.g003

Products of six of these genes are localized to the extracellular space, with functions involved in communication with other cells including chemotaxis of immune cells (Fig 3). Of importance, a factor assigned by IPA as a central mediator for most of the PIC-DGs is nuclear factor kB (NFκB), which is a key molecule in the control of inflammatory and immune responses [5862].We have earlier demonstrated that N-9 activates NFκB signaling pathway in Vk2 cells [34]. In the present study we validate the IPA-identified central position of NFκB in gene deregulation induced by TNF-α and TLR ligands in human vaginal cells. We demonstrate that following the treatments, fast degradation of IκB-α takes place in the cytoplasm accompanied by release and translocation of p65/NFκB to the nucleus indicating NFkB activation (Fig 4).

thumbnail
Fig 4. NFκB activation in Vk2 cells in response to PIC treatments.

Fast degradation of IκB-α in the cytoplasm and translocation of p65/NFkB to the nucleus following Vk2 cells exposure to PICs was detected in cytoplasmic and nuclear fractions using corresponding antibodies.

https://doi.org/10.1371/journal.pone.0128557.g004

Real-time qPCR analysis of selected genes

Based on their functions and level of expression, eight genes, PTGS2, CCL20, IL8, CXCL2, CXCL3, TNFAIP3, OLR1 and CYLD, were selected for validation by qPCR. All selected genes, except for CYLD showed significant upregulation (Fig 5), thus confirming the microarray data.

thumbnail
Fig 5. Real-time qPCR validation of changes in expression of eight selected genes observed in microarray analysis.

Bars represent the mean ± SD of fold change relative to growth medium control. At least 3 independent experiments were performed for each treatment. All genes were normalized to GAPDH. Asterisks placed vertically denote p values for each PIC treatment relative to HEC used as a reference. (***p<0.0005, **p<0.005, *p< 0.05; Student t-test)

https://doi.org/10.1371/journal.pone.0128557.g005

Generally, most of the genes tested demonstrated higher fold changes with qPCR compared to microarray. A similar trend was also described by others [63,64].

Evaluation of candidate microbicides using discriminatory genes

We used a panel of PIC-DG to test their discriminatory power and to evaluate the pro-inflammatory potential of several microbicides /active ingredients: non-ionic detergent C31G (an active ingredient of Savvy), non-ionic polymers—PRO2000, cellulose sulfate (CS), dextran sulfate (DS), reverse transcriptase inhibitors (RTI)—nucleoside/nucleotide RTI (NRTI)—tenofovir (TFV) and emtricitabine (FTC), and non-NRTI (NNRTI)—UC781. The heatmap of cluster analysis of microarray data demonstrates that C31G clusters together with PICs, while the other candidate microbicides proved to be not pro-inflammatory by this gene expression profile (Fig 6).

thumbnail
Fig 6. Transcription profile of the 20 discriminatory genes expression in Vk2 cells exposed to candidate microbicides and selected PICs and NICs.

Columns represent treatments, rows represent genes. Gene expression levels are indicated by color: red is for upregulation and green is for downregulation. Expression data are averages from at least six experiments/microarrays for each treatment. Clustering based on 20 PIC/NIC discriminatory genes places C31G (known as causing inflammatory response) to the PIC category, while dextran sulfate (DS) and cellulose sulfate (CS)—into the NIC group.

https://doi.org/10.1371/journal.pone.0128557.g006

It should be noted, however, that although UC781 clustered with the NIC group, it caused upregulation of several of the PIC-DGs belonging to inflammation/immune response category which was confirmed by real-time RT-PCR analysis (Fig 7).

thumbnail
Fig 7. Real-time qPCR validation of changes in expression of eight selected genes observed in the microarray analysis of microbicide candidates.

Experimental details are as in Fig 5. HEC and TNF-α are added as references.

https://doi.org/10.1371/journal.pone.0128557.g007

Panel of selected PIC-DGs clearly discriminates normal vaginal flora- from BV-related bacteria in the Vk2 cell test model

Earlier we demonstrated that P.bivia, bacteria associated with BV caused significant NFkB activation and increase in IL-8 level in Vk2 cells [41]. In the study reported herein, vaginal epithelial expression of seven selected PIC-DGs, PTGS2, CCL20, IL8, CXCL2, CXCL3, TNFAIP3, OLR1, following exposure to P. bivia and beneficial commensal Lactobacillus (L.) gasseri was compared using qPCR. Strong induction of all seven PIC-DG by P. bivia (3 to 75 fold increase), but not by L. gasseri, was observed (Fig 8), thus validating the discriminatory power of these genes in pathologically relevant conditions of the cervicovaginal tract.

thumbnail
Fig 8. PIC-DG expression following bacterial colonization of Vk2 cells as revealed by quantitative real time RT-PCR.

P. bivia (right) induced strong upregulation of all seven PIC-DEGs, while L. gasseri (left) did not cause any changes. Results are presented as mean ±SD of three experiments.

https://doi.org/10.1371/journal.pone.0128557.g008

Discussion

The molecular and cellular mechanisms underpinning HIV-1 vaginal transmission are not yet fully understood. Numerous studies indicate that the initial targets of HIV-1 infection could primarily be CD4+ T cells, with dendritic cells (DC), Langerhans cells (LC), and/or macrophages also playing a role (reviewed in [18,65]). Inflammation and immune activation of cervicovaginal mucosa are considered factors that increase susceptibility to HIV infection due to recruitment and activation of HIV-target immune cells and increased production of immunostimulatory factors that paradoxically intensify HIV replication at the sites of viral exposure. [1318]. In addition, cervicovaginal inflammation may cause disruption of the epithelial barrier, thus providing a portal for viral entry [66,67]. Therefore, it is critical to screen candidate microbicides for potential mucosal immunomodulatory/inflammatory effects. The goal of this study was to develop an in vitro method for preclinical evaluation of pro-inflammatory/immunomodulatory potential of new candidate microbicides and identify new biomarkers of mucosal alteration. To this end, we compared gene expression profiles of Vk2 cells treated with PICs and NICs in order to get a panel of genes that are consistently altered by PICs compared to NICs, thus distinguishing between these two groups.

To make the discriminatory genes more encompassing we selected PICs that belong to different molecular and mechanistic groups. TNF-α is a well characterized pleiotropic proiflammatory cytokine [68,69]. N-9, a nonionic cell membrane disrupting surfactant, is a microbicide that increased the probability of HIV-1 transmission in clinical trials, supposedly, due to vaginal mucosa irritation and inflammation [70,71]. Furthermore, cervicovaginal lavages collected from women who had used N-9 for three days enhanced HIV replication in vitro [40]. The other pro-inflammatory group included different TLR-ligands (TLR-L) (Table 1). TLRs are signaling transmembrane proteins that recognize and rapidly respond to various conserved microbial pathogen-associated molecular patterns (PAMPs). The interaction of TLRs with their corresponding PAMPs leads to initiation of innate immune response and triggers pro-inflammatory pathways, in which activation of NFκB plays a central role (see reviews [58,59,61,62]. There are 10 TLRs identified in the human thus far. The female lower genital tract mucosa was found to express most of them [37,7275]. There is strong evidence supporting the link between the presence of infectious conditions in the lower genital tract, including sexually transmitted infections (STI) and BV, and increased risk of HIV-1 acquisition [1921]. It is possible that stimulation of innate immune and inflammatory signaling by the PAMP-activated cervicovaginal TLRs through cytokine and chemokine induction increases availability of HIV-1 target cells in the mucosal epithelium, facilitating primary HIV-1 infection.

In this study we identified a set of 20 genes of which 19 genes are consistently activated and one gene is downregulated in human vaginal epithelial cells exposed to various compounds that shared proinflammatory/immunomodulatory activity. Our selection of the stimuli was confirmed by the functional analysis of these discriminatory genes using the IPA program and published data, which revealed that out of the 20 selected genes, 14 genes are known to be involved in inflammatory and immune responses. For most of our inflammation-related genes, their role in inflammatory responses is largely understood, albeit primarily in tissues other than the cervicovaginal mucosa. At the same time, novel features that might link them to HIV susceptibility have recently emerged (discussed below).

Although in vivo models have obvious limitations compared to the in vitro ones, the set of genes or biomarkers reported herein can be tested early in the process of drug discovery or product development to identify compounds with properties that may result in undesirable mucosal effects. In an attempt to validate this set of biomarkers, we tested microbicide candidates previously characterized in clinical studies with known cervicovaginal mucosal effects [2] and we were able to properly qualify those compounds. In clinical trials, CS, PRO2000 and DS appeared to be safe and did not cause adverse mucosal effects/epithelial disruption, while C31G might have caused vaginal alterations comparable to the effect observed with N-9 [2,7680]. Clustering analysis of the microarray data classified C31G as PIC, while CS, DS, PRO2000 aligned with the NIC group in good correlation with the results from clinical trials. Tenofovir, currently completing a phase III confirmatory trial in the form of 1% vaginal gel, did not show a pro-inflammatory profile. This is in agreement with results from numerous safety studies including the Phase IIb trial, CAPRISA 004 [8183]. Interestingly, although clustering with NICs, UC781 showed upregulation of certain PIC-associated genes. Product development of UC781 as a vaginal microbicide, which had reached clinical stage, was discontinued due to pharmaceutical and safety issues. High concentrations of UC781 revealed significant changes in the mucosa of macaques and rabbits [84].

In addition, the discriminatory power of seven selected PIC-DGs was confirmed in the bacterial exposure experiment using commensal and pathogenic microorganisms. We found strong induction of these selected PIC-DGs in Vk2 cells by P. bivia (3 to 75 fold increase), bacteria associated with BV [4749]. By contrast, L gasseri, one of the dominant beneficial Lactobacillus species in the vagina of healthy women [43,44,46,85] did not cause any changes (Fig 8). Upregulation of inflammation-related genes by the BV-related bacteria may have pathogenic implications and provide further insight on the mechanisms underpinning increased rates of HIV-1 transmission in women with BV [86,87].

Clearly these new biomarkers do not identify every potential adverse effect on the cervicovaginal mucosa or environment. CS, which failed to protect women from HIV and may have even increased their susceptibility, did not show inflammatory effects, neither in this model nor in clinical studies. However, it has been reported that it might downregulate epithelial junctional proteins or alter the vaginal microbiome facilitating HIV infection [9,88].

In addition to generating a group of new biomarkers of vaginal mucosal alteration, the reported discriminatory genes may also provide clues to better understanding of the mucosal changes that coexist with and may facilitate HIV transmission. For instance, these genes include several widely known chemokines. IL8, is one of the major mediators of the inflammatory response [89,90]. This chemokine, also known as CXCL8, attracts T cells and neutrophils, stimulates adhesion of monocytes to endothelial cells via interaction with its receptors CXCR1 and CXCR2 [91]. High vaginal IL-8 levels were observed at BV [92] and in response to some pro-inflammatory compounds [29]. IL-8 was found to stimulate HIV-1 replication in T cells and macrophages and increase HIV-1 transmission in cervical explants tissues [93,94]. Much less is known about CXCL2 (MIP-2α, GRO-2, GRO-β) and CXCL3 (MIP-2β, GRO-3, GRO-γ) that are structurally related to IL-8. Like IL-8, they also interact with CXCR2 and are involved in inflammatory and immune responses by attracting and activating immune cells [95].

Another chemokine which is significantly upregulated by PICs is CCL20, or macrophage inflammatory protein 3α (MIP-3α). CCL20 is the only chemokine that interacts with CCR6 receptor [96] which is expressed by Th17 lymphocytes and by LCs. Several studies indicate that the CD4+ Th17 cells are early HIV/SIV preferential targets and are implicated in HIV pathogenesis [97101]. Recruitment of CCR6-expressing Th17 cells through CCL20-CCR6 interactions was demonstrated in diverse tissues [102104]. In the vaginal epithelium these interactions may bring more HIV-target cells to the site of viral exposure. CCL20 was shown to be the main chemoattractant for LCs in human vaginal epithelial cells [105]. Besides CCR6, mucosal LCs express HIV major receptors, CD4 and CCR5, and are capable of internalizing virions. It is still debated whether new virions can be produced by LCs and DCs, however, LCs are able to migrate from the mucosa to the lymph nodes where virions can be transmitted to lymphocytes for productive infection. In in vivo and ex vivo studies, involvement of LCs in HIV sexual infection has been clearly demonstrated [65,106110]. In ex vivo experiments, inflammatory stimuli, TNF-α and Pam3CSK4 (a ligand for TLR1/TLR2 heterodimer) strongly increased HIV-1 transmission by LCs [107]. Conversely, suppression of CCL20 production and possible prevention of LCs and Th17 attraction and activation correlated with restriction of mucosal transmission of SIV, as demonstrated by glycerol monolaureate studies in non-human primates [13].

Upregulation of PTGS2 by all PICs observed in this study is a strong assertion of the proinflammatory potential of the selected PICs. The protein encoded by PTGS2, most often called COX-2 (cyclooxygenase-2), is an inducible enzyme that is expressed in response to various pathophysiological stimuli. It is one of the key enzymes involved in inflammation. It catalyzes the first steps of conversion of arachidonic acid into prostaglandins (PG) that play an important role in inflammatory and immunomodulatory processes [111113]. One of the major COX-2 products, PGE2 was shown to directly enhance HIV-1 long terminal repeat (LTR) transcription in human T cells [114]. We have earlier demonstrated that PGE2 levels were elevated in the vaginal epithelial cells following COX-2 induction in response to diverse pro-inflammatory/immunomodulatory stimuli [33,34]. Activation of PTGS2 gene resulting in PGE2 elevation could be a factor contributing to HIV sexual transmission in inflammatory conditions.

TNFAIP3 (tumor necrosis factor, alpha-induced protein 3) is another gene that plays an important role in regulation of inflammation and immunity and regulates NFκB. We also observed strong upregulation of OLR1, a gene for oxidized low density lipoprotein (lectin-like) receptor 1—cell surface protein belonging to the C-type lectin family. OLR1 can be rapidly activated by a wide range of stimuli including pro-inflammatory and tissue damaging ones. Its activation triggers several signaling pathways including NFκB [115,116].

NFκB signaling pathway is the central regulator of inflammation and immune activation [60]. We demonstrate here that NFκB is activated in vaginal cells in response to all PIC treatments used in this study (Fig 4). The network of PIC discriminatory gene interactions illustrates the central role of NFκB complex in their regulation (Fig 3). Importantly, NFkB binding elements are present in enhancer located in HIV-1 LTR and provide signal-specific activation of HIV expression in response to NFkB stimuli [117,118]. It might be possible that PIC treatments activate NFkB pathway in the TLR-expressing HIV-1-target cells present in the vaginal epithelium. Multiple genes activated by PICs (INHBA, FGF2, PRDM1, MafB, and SERPINB2) are involved in signal transduction or regulation of immune response [54,55,119121], which can also promote HIV-1 transcription and replication [122].

Conclusion

Robust comparative gene expression profiling revealed 20 genes (called here PIC-DGs) that were significantly altered by diverse pro-inflammatory/immunomodulatory compounds in human vaginal epithelial cells. Although the pattern of expression of these genes in vivo has not been defined yet (a study with this goal is underway), the observed consistent deregulation of these genes by PICs in the vaginal epithelial cell line suggests that PIC-DGs can be employed as in vitro molecular markers of potential inflammatory/ immunomodulatory untoward mucosal effects of candidate microbicides and other vaginal products. We believe that in vitro evaluation of the immunoinflammatory potential of candidate microbicides using the PIC-DGs defined in this study could help in the initial screening of microbicide candidates prior to entering clinical trials. Further analysis of these genes can provide better insight into the cervicovaginal immunoinflammatory and mucosal altering processes that facilitate or limit HIV transmission having important implications for the design of novel prevention strategies.

Supporting Information

S2 Table. Probesets significantly deregulated in Vk2 cells by TNF-α.

https://doi.org/10.1371/journal.pone.0128557.s002

(XLSX)

S3 Table. Probesets significantly deregulated in Vk2 cells by Pam3CSK4.

https://doi.org/10.1371/journal.pone.0128557.s003

(XLSX)

S4 Table. Probesets significantly deregulated in Vk2 cells by MALP2.

https://doi.org/10.1371/journal.pone.0128557.s004

(XLSX)

S5 Table. Probesets significantly deregulated in Vk2 cells by imiquimod.

https://doi.org/10.1371/journal.pone.0128557.s005

(XLSX)

S6 Table. Probesets significantly deregulated in Vk2 cells by N-9.

https://doi.org/10.1371/journal.pone.0128557.s006

(XLSX)

S7 Table. Probesets significantly deregulated in Vk2 cells by HEC.

https://doi.org/10.1371/journal.pone.0128557.s007

(XLSX)

S8 Table. Probesets significantly deregulated in Vk2 cells by CMC

https://doi.org/10.1371/journal.pone.0128557.s008

(XLSX)

S9 Table. Significantly deregulated probesets common to all TLR ligands.

https://doi.org/10.1371/journal.pone.0128557.s009

(XLSX)

S10 Table. Probesets significantly deregulated in Vk2 cells that are common to all treatments with proinflammatory-immunomodulatory compounds.

https://doi.org/10.1371/journal.pone.0128557.s010

(DOCX)

Author Contributions

Conceived and designed the experiments: IAZ GFD. Performed the experiments: TJ NY IAZ SSJ TF HSY RNF. Analyzed the data: IAZ RS JB. Wrote the paper: IAZ GFD. Editorial comments/critiques: RS RNF.

References

  1. 1. Shattock RJ, Moore JP (2003) Inhibiting sexual transmission of HIV-1 infection. Nat Rev Microbiol 1: 25–34. pmid:15040177
  2. 2. Romano JW, Robbiani M, Doncel GF, Moench T (2012) Non-specific microbicide product development: then and now. Curr HIV Res 10: 9–18. pmid:22264041
  3. 3. Abdool Karim SS, Baxter C (2012) Overview of microbicides for the prevention of human immunodeficiency virus. Best Pract Res Clin Obstet Gynaecol 26: 427–439. pmid:22386823
  4. 4. Obiero J, Mwethera PG, Hussey GD, Wiysonge CS (2012) Vaginal microbicides for reducing the risk of sexual acquisition of HIV infection in women: systematic review and meta-analysis. BMC Infect Dis 12: 289. pmid:23130761
  5. 5. Friend DR, Kiser PF (2013) Assessment of topical microbicides to prevent HIV-1 transmission: concepts, testing, lessons learned. Antiviral Res 99: 391–400. pmid:23845918
  6. 6. Abdool Karim Q, Abdool Karim SS, Frohlich JA, Grobler AC, Baxter C, et al. (2010) Effectiveness and safety of tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in women. Science 329: 1168–1174. pmid:20643915
  7. 7. Van Damme L, Govinden R, Mirembe FM, Guedou F, Solomon S, et al. (2008) Lack of effectiveness of cellulose sulfate gel for the prevention of vaginal HIV transmission. N Engl J Med 359: 463–472. pmid:18669425
  8. 8. Feldblum PJ, Adeiga A, Bakare R, Wevill S, Lendvay A, et al. (2008) SAVVY vaginal gel (C31G) for prevention of HIV infection: a randomized controlled trial in Nigeria. PLoS ONE 3: e1474. pmid:18213382
  9. 9. Mesquita PM, Cheshenko N, Wilson SS, Mhatre M, Guzman E, et al. (2009) Disruption of tight junctions by cellulose sulfate facilitates HIV infection: model of microbicide safety. J Infect Dis 200: 599–608. pmid:19586414
  10. 10. Stone AB, Harrison PF, Lusti-Narasimhan M (2013) Microbicides from a regulatory perspective. AIDS 27: 2261–2269. pmid:23612007
  11. 11. Gray RH, Wawer MJ (2012) Probability of heterosexual HIV-1 transmission per coital act in sub-Saharan Africa. J Infect Dis 205: 351–352. pmid:22241799
  12. 12. Hughes JP, Baeten JM, Lingappa JR, Magaret AS, Wald A, et al. (2012) Determinants of per-coital-act HIV-1 infectivity among African HIV-1-serodiscordant couples. J Infect Dis 205: 358–365. pmid:22241800
  13. 13. Li Q, Estes JD, Schlievert PM, Duan L, Brosnahan AJ, et al. (2009) Glycerol monolaurate prevents mucosal SIV transmission. Nature 458: 1034–1038. pmid:19262509
  14. 14. Royce RA, Sena A, Cates W Jr, Cohen MS (1997) Sexual transmission of HIV. N Engl J Med 336: 1072–1078. pmid:9091805
  15. 15. Appay V, Sauce D (2008) Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol 214: 231–241. pmid:18161758
  16. 16. Borrow P, Shattock RJ, Vyakarnam A (2010) Innate immunity against HIV: a priority target for HIV prevention research. Retrovirology 7: 84. pmid:20937128
  17. 17. Shacklett BL (2010) Immune responses to HIV and SIV in mucosal tissues: 'location, location, location'. Curr Opin HIV AIDS 5: 128–134. pmid:20543589
  18. 18. Haase AT (2010) Targeting early infection to prevent HIV-1 mucosal transmission. Nature 464: 217–223. pmid:20220840
  19. 19. Galvin SR, Cohen MS (2004) The role of sexually transmitted diseases in HIV transmission. Nat Rev Microbiol 2: 33–42. pmid:15035007
  20. 20. Thurman AR, Doncel GF (2011) Innate immunity and inflammatory response to Trichomonas vaginalis and bacterial vaginosis: relationship to HIV acquisition. Am J Reprod Immunol 65: 89–98. pmid:20678168
  21. 21. Ward H, Ronn M (2010) Contribution of sexually transmitted infections to the sexual transmission of HIV. Curr Opin HIV AIDS 5: 305–310. pmid:20543605
  22. 22. Jespers V, Millwood IY, Poynten IM, Van Damme L, Kaldor JM (2013) The evolving design and methods for trials evaluating the safety of candidate vaginal microbicides: a systematic review. Sex Transm Dis 40: 729–736. pmid:23945427
  23. 23. Hladik F, Doncel GF (2010) Preventing mucosal HIV transmission with topical microbicides: challenges and opportunities. Antiviral Res 88 Suppl 1: S3–9. pmid:21109065
  24. 24. Pudney J, Quayle AJ, Anderson DJ (2005) Immunological microenvironments in the human vagina and cervix: mediators of cellular immunity are concentrated in the cervical transformation zone. Biol Reprod 73: 1253–1263. pmid:16093359
  25. 25. Doncel GF, Chandra N, Fichorova RN (2004) Preclinical assessment of the proinflammatory potential of microbicide candidates. J Acquir Immune Defic Syndr 37 Suppl 3: S174–180. pmid:16419269
  26. 26. Fichorova RN (2004) Guiding the vaginal microbicide trials with biomarkers of inflammation. J Acquir Immune Defic Syndr 37 Suppl 3: S184–193. pmid:16419271
  27. 27. Zhong M, He B, Yang J, Bao R, Zhang Y, et al. (2012) L-selectin and P-selectin are novel biomarkers of cervicovaginal inflammation for preclinical mucosal safety assessment of anti-HIV-1 microbicide. Antimicrob Agents Chemother 56: 3121–3132. pmid:22391529
  28. 28. Catalone BJ, Kish-Catalone TM, Budgeon LR, Neely EB, Ferguson M, et al. (2004) Mouse model of cervicovaginal toxicity and inflammation for preclinical evaluation of topical vaginal microbicides. Antimicrob Agents Chemother 48: 1837–1847. pmid:15105142
  29. 29. Fichorova RN, Bajpai M, Chandra N, Hsiu JG, Spangler M, et al. (2004) Interleukin (IL)-1, IL-6, and IL-8 predict mucosal toxicity of vaginal microbicidal contraceptives. Biol Reprod 71: 761–769. pmid:15128598
  30. 30. Cummins JE Jr, Doncel GF (2009) Biomarkers of cervicovaginal inflammation for the assessment of microbicide safety. Sex Transm Dis 36: S84–91. pmid:19218890
  31. 31. Doncel GF, Clark MR (2010) Preclinical evaluation of anti-HIV microbicide products: New models and biomarkers. Antiviral Res 88 Suppl 1: S10–18. pmid:21109063
  32. 32. Fields S, Song B, Rasoul B, Fong J, Works MG, et al. (2014) New candidate biomarkers in the female genital tract to evaluate microbicide toxicity. PLoS One 9: e110980. pmid:25333937
  33. 33. Joseph T, Zalenskaya IA, Yousefieh N, Schriver SD, Cote LC, et al. (2012) Induction of Cyclooxygenase (COX)-2 in Human Vaginal Epithelial Cells in Response to TLR ligands and TNF-alpha. Am J Reprod Immunol 67: 482–490 pmid:22235849
  34. 34. Zalenskaya IA, Cerocchi OG, Joseph T, Donaghay MA, Schriver SD, et al. (2011) Increased COX-2 expression in human vaginal epithelial cells exposed to nonoxynol-9, a vaginal contraceptive microbicide that failed to protect women from HIV-1 infection. Am J Reprod Immunol 65: 569–577. pmid:21241401
  35. 35. Fichorova RN, Rheinwald JG, Anderson DJ (1997) Generation of papillomavirus-immortalized cell lines from normal human ectocervical, endocervical, and vaginal epithelium that maintain expression of tissue-specific differentiation proteins. Biol Reprod 57: 847–855. pmid:9314589
  36. 36. Fichorova RN, Desai PJ, Gibson FC III, Genco CA (2001) Distinct proinflammatory host responses to Neisseria gonorrhoeae infection in immortalized human cervical and vaginal epithelial cells. Infect Immun 69: 5840–5848. pmid:11500462
  37. 37. Fichorova RN, Cronin AO, Lien E, Anderson DJ, Ingalls RR (2002) Response to Neisseria gonorrhoeae by cervicovaginal epithelial cells occurs in the absence of toll-like receptor 4-mediated signaling. J Immunol 168: 2424–2432. pmid:11859134
  38. 38. Fichorova RN, Lee Y, Yamamoto HS, Takagi Y, Hayes GR, et al. (2012) Endobiont viruses sensed by the human host—beyond conventional antiparasitic therapy. PLoS One 7: e48418. pmid:23144878
  39. 39. Fichorova RN, Anderson DJ (1999) Differential expression of immunobiological mediators by immortalized human cervical and vaginal epithelial cells. Biol Reprod 60: 508–514. pmid:9916021
  40. 40. Fichorova RN, Tucker LD, Anderson DJ (2001) The molecular basis of nonoxynol-9-induced vaginal inflammation and its possible relevance to human immunodeficiency virus type 1 transmission. J Infect Dis 184: 418–428. pmid:11471099
  41. 41. Fichorova RN, Yamamoto HS, Delaney ML, Onderdonk AB, Doncel GF (2011) Novel vaginal microflora colonization model providing new insight into microbicide mechanism of action. MBio 2: e00168–00111. pmid:22027006
  42. 42. Onderdonk AB, Lee ML, Lieberman E, Delaney ML, Tuomala RE (2003) Quantitative microbiologic models for preterm delivery. J Clin Microbiol 41: 1073–1079. pmid:12624032
  43. 43. Antonio MA, Hawes SE, Hillier SL (1999) The identification of vaginal Lactobacillus species and the demographic and microbiologic characteristics of women colonized by these species. J Infect Dis 180: 1950–1956. pmid:10558952
  44. 44. Pavlova SI, Kilic AO, Kilic SS, So JS, Nader-Macias ME, et al. (2002) Genetic diversity of vaginal lactobacilli from women in different countries based on 16S rRNA gene sequences. J Appl Microbiol 92: 451–459. pmid:11872120
  45. 45. Redondo-Lopez V, Cook RL, Sobel JD (1990) Emerging role of lactobacilli in the control and maintenance of the vaginal bacterial microflora. Rev Infect Dis 12: 856–872. pmid:2237129
  46. 46. Pendharkar S, Magopane T, Larsson PG, de Bruyn G, Gray GE, et al. (2013) Identification and characterisation of vaginal lactobacilli from South African women. BMC Infect Dis 13: 43. pmid:23351177
  47. 47. Hill GB (1993) The microbiology of bacterial vaginosis. Am J Obstet Gynecol 169: 450–454. pmid:8357043
  48. 48. Ling Z, Kong J, Liu F, Zhu H, Chen X, et al. (2010) Molecular analysis of the diversity of vaginal microbiota associated with bacterial vaginosis. BMC Genomics 11: 488. pmid:20819230
  49. 49. Shipitsyna E, Roos A, Datcu R, Hallen A, Fredlund H, et al. (2013) Composition of the vaginal microbiota in women of reproductive age—sensitive and specific molecular diagnosis of bacterial vaginosis is possible? PLoS One 8: e60670. pmid:23585843
  50. 50. Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, et al. (2003) Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 31: e15. pmid:12582260
  51. 51. Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: A practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B 57: 289–300.
  52. 52. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408. pmid:11846609
  53. 53. Zalenskaya IA, Bradbury EM, Zalensky AO (2000) Chromatin structure of telomere domain in human sperm. Biochem Biophys Res Commun 279: 213–218. pmid:11112441
  54. 54. Kim H, Seed B (2010) The transcription factor MafB antagonizes antiviral responses by blocking recruitment of coactivators to the transcription factor IRF3. Nat Immunol 11: 743–750. pmid:20581830
  55. 55. Schroder WA, Major L, Suhrbier A (2011) The role of SerpinB2 in immunity. Crit Rev Immunol 31: 15–30. pmid:21395508
  56. 56. Turini ME, DuBois RN (2002) Cyclooxygenase-2: a therapeutic target. Annu Rev Med 53: 35–57. pmid:11818462
  57. 57. Williams CS, Mann M, DuBois RN (1999) The role of cyclooxygenases in inflammation, cancer, and development. Oncogene 18: 7908–7916. pmid:10630643
  58. 58. Doyle SL, O'Neill LA (2006) Toll-like receptors: from the discovery of NFkappaB to new insights into transcriptional regulations in innate immunity. Biochem Pharmacol 72: 1102–1113. pmid:16930560
  59. 59. Drexler SK, Foxwell BM (2010) The role of toll-like receptors in chronic inflammation. Int J Biochem Cell Biol 42: 506–518. pmid:19837184
  60. 60. Hoffmann A, Baltimore D (2006) Circuitry of nuclear factor kappaB signaling. Immunol Rev 210: 171–186. pmid:16623771
  61. 61. Medzhitov R, Janeway C Jr (2000) The Toll receptor family and microbial recognition. Trends Microbiol 8: 452–456. pmid:11044679
  62. 62. Sabroe I, Parker LC, Dower SK, Whyte MK (2008) The role of TLR activation in inflammation. J Pathol 214: 126–135. pmid:18161748
  63. 63. Szameit S, Vierlinger K, Farmer L, Tuschl H, Noehammer C (2008) Microarray-based in vitro test system for the discrimination of contact allergens and irritants: identification of potential marker genes. Clin Chem 54: 525–533. pmid:18202158
  64. 64. Tureci O, Bian H, Nestle FO, Raddrizzani L, Rosinski JA, et al. (2003) Cascades of transcriptional induction during dendritic cell maturation revealed by genome-wide expression analysis. FASEB J 17: 836–847. pmid:12724343
  65. 65. Hladik F, Sakchalathorn P, Ballweber L, Lentz G, Fialkow M, et al. (2007) Initial events in establishing vaginal entry and infection by human immunodeficiency virus type-1. Immunity 26: 257–270. pmid:17306567
  66. 66. Coombs RW, Reichelderfer PS, Landay AL (2003) Recent observations on HIV type-1 infection in the genital tract of men and women. AIDS 17: 455–480. pmid:12598766
  67. 67. Weiler AM, Li Q, Duan L, Kaizu M, Weisgrau KL, et al. (2008) Genital ulcers facilitate rapid viral entry and dissemination following intravaginal inoculation with cell-associated simian immunodeficiency virus SIVmac239. J Virol 82: 4154–4158. pmid:18272571
  68. 68. Beutler B, Cerami A (1989) The biology of cachectin/TNF—a primary mediator of the host response. Annu Rev Immunol 7: 625–655. pmid:2540776
  69. 69. Warren JS (1990) Interleukins and tumor necrosis factor in inflammation. Crit Rev Clin Lab Sci 28: 37–59. pmid:2121159
  70. 70. Niruthisard S, Roddy RE, Chutivongse S (1991) The effects of frequent nonoxynol-9 use on the vaginal and cervical mucosa. Sex Transm Dis 18: 176–179. pmid:1658953
  71. 71. Van Damme L, Ramjee G, Alary M, Vuylsteke B, Chandeying V, et al. (2002) Effectiveness of COL-1492, a nonoxynol-9 vaginal gel, on HIV-1 transmission in female sex workers: a randomised controlled trial. Lancet 360: 971–977. pmid:12383665
  72. 72. Fazeli A, Bruce C, Anumba DO (2005) Characterization of Toll-like receptors in the female reproductive tract in humans. Hum Reprod 20: 1372–1378. pmid:15695310
  73. 73. Herbst-Kralovetz MM, Quayle AJ, Ficarra M, Greene S, Rose WA Jr, et al. (2008) Quantification and comparison of toll-like receptor expression and responsiveness in primary and immortalized human female lower genital tract epithelia. Am J Reprod Immunol 59: 212–224. pmid:18201283
  74. 74. Nasu K, Narahara H (2010) Pattern recognition via the toll-like receptor system in the human female genital tract. Mediators Inflamm 2010: 976024. pmid:20396665
  75. 75. Yu L, Wang L, Chen S (2009) Toll-like receptors, inflammation and tumor in the human female reproductive tract. Am J Reprod Immunol 62: 1–8. pmid:19527227
  76. 76. Peterson L, Nanda K, Opoku BK, Ampofo WK, Owusu-Amoako M, et al. (2007) SAVVY (C31G) gel for prevention of HIV infection in women: a Phase 3, double-blind, randomized, placebo-controlled trial in Ghana. PLoS One 2: e1312. pmid:18091987
  77. 77. Adams JL, Kashuba AD (2012) Formulation, pharmacokinetics and pharmacodynamics of topical microbicides. Best Pract Res Clin Obstet Gynaecol.
  78. 78. Cone RA, Hoen T, Wong X, Abusuwwa R, Anderson DJ, et al. (2006) Vaginal microbicides: detecting toxicities in vivo that paradoxically increase pathogen transmission. BMC Infect Dis 6: 90. pmid:16740164
  79. 79. Van Damme L (2004) Clinical microbicide research: an overview. Trop Med Int Health 9: 1290–1296. pmid:15598260
  80. 80. Ballagh SA, Baker JM, Henry DM, Archer DF (2002) Safety of single daily use for one week of C31G HEC gel in women. Contraception 66: 369–375. pmid:12443969
  81. 81. Sokal DC, Karim QA, Sibeko S, Yende-Zuma N, Mansoor LE, et al. (2013) Safety of tenofovir gel, a vaginal microbicide, in South African women: results of the CAPRISA 004 Trial. Antivir Ther 18: 301–310. pmid:22914267
  82. 82. Gengiah TN, Baxter C, Mansoor LE, Kharsany AB, Abdool Karim SS (2012) A drug evaluation of 1% tenofovir gel and tenofovir disoproxil fumarate tablets for the prevention of HIV infection. Expert Opin Investig Drugs 21: 695–715. pmid:22394224
  83. 83. Johnson TJ, Clark MR, Albright TH, Nebeker JS, Tuitupou AL, et al. (2012) A 90-day tenofovir reservoir intravaginal ring for mucosal HIV prophylaxis. Antimicrob Agents Chemother 56: 6272–6283. pmid:23006751
  84. 84. Patton DL, Sweeney YT, Balkus JE, Rohan LC, Moncla BJ, et al. (2007) Preclinical safety assessments of UC781 anti-human immunodeficiency virus topical microbicide formulations. Antimicrob Agents Chemother 51: 1608–1615. pmid:17353240
  85. 85. Anderson A, Sanunu M, Schneider C, Clad A, Karygianni L, et al. (2014) Rapid species-level identification of vaginal and oral lactobacilli using MALDI-TOF MS analysis and 16S rDNA sequencing. BMC Microbiol 14: 312. pmid:25495549
  86. 86. Atashili J, Poole C, Ndumbe PM, Adimora AA, Smith JS (2008) Bacterial vaginosis and HIV acquisition: a meta-analysis of published studies. AIDS 22: 1493–1501. pmid:18614873
  87. 87. Buve A, Jespers V, Crucitti T, Fichorova RN (2014) The vaginal microbiota and susceptibility to HIV. AIDS 28: 2333–2344. pmid:25389548
  88. 88. Ravel J, Gajer P, Fu L, Mauck CK, Koenig SS, et al. (2012) Twice-daily application of HIV microbicides alter the vaginal microbiota. MBio 3.
  89. 89. Harada A, Sekido N, Akahoshi T, Wada T, Mukaida N, et al. (1994) Essential involvement of interleukin-8 (IL-8) in acute inflammation. J Leukoc Biol 56: 559–564. pmid:7964163
  90. 90. Hoffmann E, Dittrich-Breiholz O, Holtmann H, Kracht M (2002) Multiple control of interleukin-8 gene expression. J Leukoc Biol 72: 847–855. pmid:12429706
  91. 91. Baggiolini M, Moser B (1997) Blocking chemokine receptors. J Exp Med 186: 1189–1191. pmid:9379143
  92. 92. Spandorfer SD, Neuer A, Giraldo PC, Rosenwaks Z, Witkin SS (2001) Relationship of abnormal vaginal flora, proinflammatory cytokines and idiopathic infertility in women undergoing IVF. J Reprod Med 46: 806–810. pmid:11584481
  93. 93. Lane BR, Lore K, Bock PJ, Andersson J, Coffey MJ, et al. (2001) Interleukin-8 stimulates human immunodeficiency virus type 1 replication and is a potential new target for antiretroviral therapy. J Virol 75: 8195–8202. pmid:11483765
  94. 94. Narimatsu R, Wolday D, Patterson BK (2005) IL-8 increases transmission of HIV type 1 in cervical explant tissue. AIDS Res Hum Retroviruses 21: 228–233. pmid:15795529
  95. 95. Vandercappellen J, Van Damme J, Struyf S (2008) The role of CXC chemokines and their receptors in cancer. Cancer Lett 267: 226–244. pmid:18579287
  96. 96. Schutyser E, Struyf S, Van Damme J (2003) The CC chemokine CCL20 and its receptor CCR6. Cytokine Growth Factor Rev 14: 409–426. pmid:12948524
  97. 97. El Hed A, Khaitan A, Kozhaya L, Manel N, Daskalakis D, et al. (2011) Susceptibility of human Th17 cells to human immunodeficiency virus and their perturbation during infection. J Infect Dis 201: 843–854.
  98. 98. Kader M, Wang X, Piatak M, Lifson J, Roederer M, et al. (2009) Alpha4(+)beta7(hi)CD4(+) memory T cells harbor most Th-17 cells and are preferentially infected during acute SIV infection. Mucosal Immunol 2: 439–449. pmid:19571800
  99. 99. McKinnon LR, Nyanga B, Chege D, Izulla P, Kimani M, et al. (2011) Characterization of a human cervical CD4+ T cell subset coexpressing multiple markers of HIV susceptibility. J Immunol 187: 6032–6042. pmid:22048765
  100. 100. Monteiro P, Gosselin A, Wacleche VS, El-Far M, Said EA, et al. (2011) Memory CCR6+CD4+ T cells are preferential targets for productive HIV type 1 infection regardless of their expression of integrin beta7. J Immunol 186: 4618–4630. pmid:21398606
  101. 101. Prendergast A, Prado JG, Kang YH, Chen F, Riddell LA, et al. (2010) HIV-1 infection is characterized by profound depletion of CD161+ Th17 cells and gradual decline in regulatory T cells. AIDS 24: 491–502. pmid:20071976
  102. 102. Hirata T, Osuga Y, Takamura M, Kodama A, Hirota Y, et al. (2010) Recruitment of CCR6-expressing Th17 cells by CCL 20 secreted from IL-1 beta-, TNF-alpha-, and IL-17A-stimulated endometriotic stromal cells. Endocrinology 151: 5468–5476. pmid:20881253
  103. 103. Hirota K, Yoshitomi H, Hashimoto M, Maeda S, Teradaira S, et al. (2007) Preferential recruitment of CCR6-expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model. J Exp Med 204: 2803–2812. pmid:18025126
  104. 104. Miossec P (2009) IL-17 and Th17 cells in human inflammatory diseases. Microbes Infect 11: 625–630. pmid:19371791
  105. 105. Cremel M, Berlier W, Hamzeh H, Cognasse F, Lawrence P, et al. (2005) Characterization of CCL20 secretion by human epithelial vaginal cells: involvement in Langerhans cell precursor attraction. J Leukoc Biol 78: 158–166. pmid:15831560
  106. 106. Blauvelt A, Glushakova S, Margolis LB (2000) HIV-infected human Langerhans cells transmit infection to human lymphoid tissue ex vivo. AIDS 14: 647–651. pmid:10807187
  107. 107. de Jong MA, de Witte L, Bolmstedt A, van Kooyk Y, Geijtenbeek TB (2008) Dendritic cells mediate herpes simplex virus infection and transmission through the C-type lectin DC-SIGN. J Gen Virol 89: 2398–2409. pmid:18796707
  108. 108. Fahrbach KM, Barry SM, Ayehunie S, Lamore S, Klausner M, et al. (2007) Activated CD34-derived Langerhans cells mediate transinfection with human immunodeficiency virus. J Virol 81: 6858–6868. pmid:17442711
  109. 109. Sugaya M, Lore K, Koup RA, Douek DC, Blauvelt A (2004) HIV-infected Langerhans cells preferentially transmit virus to proliferating autologous CD4+ memory T cells located within Langerhans cell-T cell clusters. J Immunol 172: 2219–2224. pmid:14764689
  110. 110. Ballweber L, Robinson B, Kreger A, Fialkow M, Lentz G, et al. (2011) Vaginal langerhans cells nonproductively transporting HIV-1 mediate infection of T cells. J Virol 85: 13443–13447. pmid:21976645
  111. 111. Hata AN, Breyer RM (2004) Pharmacology and signaling of prostaglandin receptors: multiple roles in inflammation and immune modulation. Pharmacol Ther 103: 147–166. pmid:15369681
  112. 112. Harizi H, Grosset C, Gualde N (2003) Prostaglandin E2 modulates dendritic cell function via EP2 and EP4 receptor subtypes. J Leukoc Biol 73: 756–763. pmid:12773508
  113. 113. Kabashima K, Sakata D, Nagamachi M, Miyachi Y, Inaba K, et al. (2003) Prostaglandin E2-EP4 signaling initiates skin immune responses by promoting migration and maturation of Langerhans cells. Nat Med 9: 744–749. pmid:12740571
  114. 114. Dumais N, Barbeau B, Olivier M, Tremblay MJ (1998) Prostaglandin E2 Up-regulates HIV-1 long terminal repeat-driven gene activity in T cells via NF-kappaB-dependent and-independent signaling pathways. J Biol Chem 273: 27306–27314. pmid:9765256
  115. 115. Khaidakov M, Mitra S, Kang BY, Wang X, Kadlubar S, et al. (2011) Oxidized LDL receptor 1 (OLR1) as a possible link between obesity, dyslipidemia and cancer. PLoS One 6: e20277. pmid:21637860
  116. 116. Li D, Mehta JL (2000) Antisense to LOX-1 inhibits oxidized LDL-mediated upregulation of monocyte chemoattractant protein-1 and monocyte adhesion to human coronary artery endothelial cells. Circulation 101: 2889–2895. pmid:10869259
  117. 117. Jones KA, Peterlin BM (1994) Control of RNA initiation and elongation at the HIV-1 promoter. Annu Rev Biochem 63: 717–743. pmid:7979253
  118. 118. Nolan GP, Baltimore D (1992) The inhibitory ankyrin and activator Rel proteins. Curr Opin Genet Dev 2: 211–220. pmid:1386268
  119. 119. Aleman-Muench GR, Soldevila G (2012) When versatility matters: activins/inhibins as key regulators of immunity. Immunol Cell Biol 90: 137–148. pmid:21537340
  120. 120. Keller AD, Maniatis T (1991) Identification and characterization of a novel repressor of beta-interferon gene expression. Genes Dev 5: 868–879. pmid:1851123
  121. 121. Verstrepen L, Verhelst K, van Loo G, Carpentier I, Ley SC, et al. (2010) Expression, biological activities and mechanisms of action of A20 (TNFAIP3). Biochem Pharmacol 80: 2009–2020. pmid:20599425
  122. 122. Darnell GA, Schroder WA, Gardner J, Harrich D, Yu H, et al. (2006) SerpinB2 is an inducible host factor involved in enhancing HIV-1 transcription and replication. J Biol Chem 281: 31348–31358. pmid:16923810