Abstract
Purpose:
Women who have genital inflammation are at increased risk of sexual HIV infection. The purpose of this review is to evaluate the mechanisms for this relationship, causes of genital inflammation and strategies to manage this condition.
Recent Findings:
We have recently shown in a cohort of South African women that HIV seroconversion was associated with persistently raised genital inflammatory cytokines (including MIP-1α, MIP-1β and IP-10). Elevated inflammatory cytokine concentrations may facilitate HIV infection by recruiting and activating HIV target cells and disrupting the mucosal epithelial barrier. BV and STIs, which are predominantly asymptomatic in women, cause lower genital tract inflammation and increased HIV acquisition risk. In Africa, where syndromic management of STIs and BV is standard-of-care, the substantial burden of asymptomatic infections has likely contributed to high HIV incidence rates.
Summary:
A genital inflammatory profile contributes to the high risk of HIV acquisition in African women. STIs and BV are poorly managed in Africa and other developing nations and as such remain major drivers of persistent genital inflammation and HIV acquisition among these women.
Keywords: genital tract, inflammation, cytokines, T cell activation, HIV risk
Introduction
Young women in sub-Saharan Africa are disproportionately affected by HIV, with prevalences of up to 8-fold higher than in males of the same age [1]. Despite new HIV infections dropping from 3.4 million in 2001 to 2.0 million globally in 2014 [2], continued transmission in young women is one of the greatest challenges preventing an AIDS-free generation [3]. Susceptibility to HIV infection varies considerably from person-to-person, with some women remaining uninfected despite repeated exposure [4]. Bacterial vaginosis (BV) and sexually transmitted infections (STIs) [5–8], as well as other biological factors, have been shown to impact the risk of young women acquiring HIV. Genital inflammation underlies many of these risk factors, providing a unifying mechanism driving risk [8–10].
HIV risk and genital inflammation
Inflammation in the female genital tract, regardless of the cause, creates an environment that favours HIV replication and establishment of a productive infection. Women with elevated concentrations of pro-inflammatory cytokines, including MIP-1α, MIP-1β and IP-10, in their genital tracts were found to be at increased risk of HIV acquisition [9]. IP-10, MIP-1α, and MIP-1β are chemotactic for HIV target cells, including T cells, macrophages and dendritic cells [11–14]. MIP-1α and MIP-1β are also ligands for the HIV co-receptor CCR5 and specifically recruit CCR5+ target cells into tissues [15]. Pro-inflammatory cytokine signatures in the lower reproductive tract has been associated with increased frequencies of neutrophils, T and B cells, as well as higher levels of cellular activation [16,17]. Pro-inflammatory cytokines and chemokines that are involved in activation, differentiation and recruitment of immune cells to the genital tract, which may increase HIV transmission as HIV replication depends on the presence of immune cell targets, the level of immune cell activation and monocyte differentiation to macrophages or dendritic cells [13,16]. In rhesus macaques, pro-inflammatory cytokine production following vaginal SIV exposure resulted in recruitment of CD4+ T cells needed for establishment of SIV infection [11,18]. The essential role of inflammation in SIV infection was clearly demonstrated when topical application of an anti-inflammatory, glycerol-monolaurate, down-regulated chemokine concentrations, inhibited inflammatory cell influx to the genital tract, and prevented SIV infection in macaques [11].
Studies in exposed seronegative women (ESN), who remained HIV-uninfected despite high-risk sexual activity, have improved our understanding of risk factors for HIV acquisition. In ESN women, concentrations of the CCR5-binding chemokine RANTES were found to be elevated compared to low-risk controls, while MIP-1α and MIP-1β have been shown to competitively inhibit HIV binding to CCR5 in vitro, suggesting that these chemokines may protect against HIV infection [19,20]. However, ESN women may have higher genital chemokine concentrations compared to low-risk controls because they are more likely to have STIs [4], and in vitro models do not account for up-regulation of other inflammatory factors or recruitment of HIV target cells by these chemokines that may facilitate HIV replication in vivo. More recent ESN studies have shown that an immune quiescent phenotype in the female genital tract may account for reduced susceptibility to HIV infection in these women [21,22]. Although ESN women were found to have higher CD4+ T cell numbers at the cervix, fewer of these cells expressed CCR5 compared to low risk women [22].
In addition to recruiting more target cells for HIV replication, pro-inflammatory cytokines induce expression of the transcription factor, nuclear factor (NF)-ΚB, which binds to HIV-long terminal repeat (LTR) and directly up-regulates HIV replication [23]. Pro-inflammatory cytokines may also facilitate HIV infection by disrupting tight junctions between epithelial cells, reducing the integrity of this barrier [24]. In support of this, proteomic studies have shown that women with elevated genital pro-inflammatory cytokine concentrations have unique protein signatures of reduced epithelial barrier function [17]. Several proteins that regulate actin cytoskeleton organization and extracellular matrix components were found to be associated with genital inflammation, suggesting that tissue remodeling occurs in women with inflammation at the expense of effective barrier function [17].
Systemic markers of inflammation, chemokine gradients and HIV risk
Blood biomarkers have also been associated with increased risk of HIV infection [25,26]. Others from our group reported that women who later became HIV-infected had higher plasma concentrations of TNF-α, IL-2, IL-7, and IL-12p70 than women who remained uninfected [25]. We found that these and other cytokines do not correlate between blood and the genital tract, suggesting that cytokine risk factors identified in blood do not predict those in the genital tract and vice versa [9,27]. Kahle et al. found that elevated plasma IP-10 and IL-10 concentrations predicted HIV seroconversion in individuals in HIV discordant relationships [26].
ESN women had lower concentrations of HIV-target cell recruiting chemokines, including IP-10, MIP-1α and MIP-1β, in the genital tract than blood, which may result in reduced target cell influx in the absence of a chemokine gradient to the genital tract, and thereby confer a certain degree of protection against HIV infection [21,22]. This suggests that a chemokine gradient from blood to the genital mucosa may contribute to risk for HIV infection.
The level of T cell activation in blood appear to be important in HIV risk. CD4+ T cell immune quiescence has shown to be protective against HIV infection in vivo [28]. Studies in european adult ESNs showed relatively lower CD38 and HLA-DR-expressing CD4+ T cells in blood than persons who go on to become HIV-infected [28]. SIV-exposed sooty mangabey infants, with few peripheral and mucosal CD4+CCR5+ cells, are less likely to acquire SIV via low dose oral challenge than their rhesus macaque counterparts [29]. We found a strong correlation between peripheral and cervical T cell activation in HIV-uninfected women [30]. Global T cell activation may be an important contributing factor determining HIV risk.
STIs cause genital inflammation
STIs are major causes of inflammatory cytokine upregulation and immune cell recruitment to the genital mucosa [27,31–34]. Although inflammation can be important in STI clearance, it may also cause destruction of infected epithelial layers, allowing STI-associated microbes to access deeper tissues [35,36]. Relatively few women are able to clear an infection in the absence of treatment, with STIs often being recurrent or persistent [37,38]. In addition to reproductive complications, non-ulcerative STIs have been found to influence susceptibility to HIV infection [5,8]. We have shown that Chlamydia trachomatis, Neisseria gonorrhoeae and Mycoplasma genitalium infections were associated with increased risk of HIV acquisition [8]. Highly prevalent STIs, such as human papillomavirus (HPV) infections, have also been shown to increase risk of HIV infection [39]. Of the common STIs, we found chlamydia that was associated with the highest genital cytokine levels, followed by gonorrhoea, HSV-2, trichomoniasis, and BV [27].
In a cohort of African women, Masese et al. (2015) reported that the overall population risk for HIV infection was largely attributable to HSV-2, even in the absence of ulcers, with prevalent HSV-2 accounting for 48.3% and incident HSV-2 infections accounting for 4.5% of risk [40]. Although HSV-2 ulcerative lesions disrupt the mucosal barrier, higher numbers of DC-SIGN+ DCs and CCR5+ CD4+ T cells are observed in the genital tracts of women who have HSV-2, even in the absence of HSV-2 shedding or genital ulceration, and sub-clinical inflammatory responses in the mucosa are evident for months after a reactivation event [34,41]. Other infections, including yeast (6.4%), Trichomonas vaginalis (1.1%), N. gonorrhoeae (0.9%), and nonspecific cervicitis (0.7%), accounted collectively for 9% of the population attributable risk for HIV in an African cohort [40]. Masese et al. showed that prevalent HSV-2 infections continued to be the most dominant population attributable risk (40.4% to 61.8% between 1998 and 2012) over time [40]. In South Africa, over 50% of new HIV infections in women could be attributed to STIs, BV and candidiasis in 2010, with HSV-2 being the most influential infection [42].
BV and the vaginal microbiome influence genital inflammation
BV is a syndrome characterized by a displacement of healthy vaginal commensal microbiota by other Gram-positive and Gram-negative bacteria [43,44]. A recent meta-analysis found that BV was associated with 1.7-fold increased risk of HIV acquisition [7]. Masese et al. [40] reported that BV contributed substantially to HIV acquisition risk, with 15.1% of the overall population risk attributable to this condition and 7.5% attributable to intermediate microbiota [40]. Given the high prevalence and recurrence of BV, this strong association with HIV risk has important public health implications.
Several studies from North America have defined a healthy female genital tract as one harboring predominantly Lactobacillus species (particulalry L. crispatus and L. jensenii), having a pH between 3.5–4.5, having no BV, candida or other STIs [45–47], although this may not be perfectly applicable to women in Africa [47]. Recent studies from South Africa found that less than 40% of women had a vaginal microbiota dominated by Lactobacillus spp., with more than half of the women not having an easily definable predominant bacterial taxon [10].
Commensal microorganisms are recognized as an important component of vaginal mucosal defense against STIs [48], including HIV [49–52], but the mechanisms of this protection are not well elucidated and are likely multifactorial. There are several ways by which commensal bacteria could potentially affect vaginal inflammation and HIV susceptibility. These include (i) lowering vaginal pH as a result of their lactic acid and H202 metabolites; (ii) competitive antagonism of pathogens; (iii) anti-microbial factor production [53], (iv) modulation of epithelial barrier integrity, epithelial or immune cell function [17,54]; (v) generation of tolerizing cells such as Tregs [55, 56]. No single bacterial strain drives all of these effects, and it is likely that more than one of these mechanisms may be at play, and may not be mutually exclusive.
In vitro experiments have demonstrated that Lactobacillus species generally induce low or no proinflammatory cytokine production by vaginal epithelial cell lines, compared to common BV-associated organisms, such as Atopobium vaginae or Gardnerella vaginalis [57,58]. Anahtar et al. (2015) found that the presence specific combinations of non-commensal organisms (cervicotype IV defined by a high diversity of organisms, dominated by Gardnerella and Prevotella species, but also featuring Shuttleworthia, Sneathia, Megasphaera, Mobiluncus, and Atopobium) was associated with higher levels of inflammation (measured by IL-1α, IL-1β, and TNF-α concentrations) in the genital tracts of young African women [10]. Only half of the young women in this category had Nugent scores >7. Some of these non-commensal bacteria individually (Sneathia amnii, Streptococcus sanguinegens and Mobiluncus mulieris) induced inflammatory responses by vaginal epithelial cell lines [10]. These women were followed longitudinally, and changes in prevalent cervicotypes were associated with significant increases in IL-1α, IL-1β and TNF-α, implying a causal relationship [10]. Other studies have found that BV is associated with genital pro-inflammatory cytokine upregulation, but also downregulation of some cytokines [27,59,60]. This is likely due to the fact that BV is complex, and is not the same syndrome in every case.
Proteomic analysis of women with increasing levels of vaginal dysbiosis was able to identify several cytokines and cytokine receptors that increased with BV, but also found alterations in proteins associated with mucosal barrier breakdown, including mucus and cytoskeletal alterations (decreased keratins and cornified envelope proteins) [54]. Interestingly, Arnold et al. (2015) reported similar changes in women with increased genital inflammation, implying that BV may act through these same pathways to increase susceptibility to HIV [17].
Hormonal contraceptives (HCs) and genital inflammation
Over 50 studies have examined the association between HC use and HIV. Some studies have found no association [61], whereas others have found up to two-fold higher risk of HIV acquisition in seronegative women using any HC [62]. In macaques, progesterone implants increase susceptibility to vaginal inoculation with SIV [63]. This is thought to be due to epithelial thinning, which can be reversed by pretreatment with estrogen [64,65]. Studies of the effect of HC among humans on genital epithelium did not observe the thinning seen in non-human primate studies [66]. Cervical ectopy, or extension of the endocervical columnar epithelium onto the ectocervix, has been associated with HC use [67,68]. DMPA may decrease vaginal colonization by H2O2–producing Lactobacillus species [69]. On the other hand, DMPA has been shown in cohort studies to decrease the risk of BV, but to increase the risk of other STIs, including C. trachomatis and HSV-2 [70,71]. On a cellular level, HCs have been associated with cervical and vaginal inflammation [72–74], increased genital tract cellular CCR5 expression [73–77], and T cell and macrophage mucosal trafficking [78]. Conversely, DMPA may also have anti-inflammatory effects [75,79].
Other possible causes of genital inflammation that may influence HIV risk include vaginal hygiene practices [80], exposure to seminal proteins [81], lubricants [82], hormone cycling [59], and genital schistosomiasis [83], as well as host genetics [84].
Management of STIs and BV to reduce HIV incidence
In resource-limited settings, STIs and BV are managed syndromically, according to the presence of clinical signs and symptoms [85]. However, large proportions of women who have STIs or BV are asymptomatic and are thus left untreated [8,86]. In South Africa, the implementation of syndromic management in the mid-1990s, as well as increased condom use, resulted in a decline in gonorrhoea, chancroid and syphilis, although there has been little or no evidence of declining prevalence of other STIs and BV [42]. After the introduction in South Africa, the proportion of new HIV infections attributable to curable STIs decreased from 39% to 14% between 1990 and 2010, however the proportion of HIV infections attributable to HSV-2 increased and the contribution of BV remained unchanged [42].
The results of population-wide STI treatment interventions for HIV prevention have been largely dissappointing [87–91]. Two of three STI syndromic management interventions in Africa resulted in no change in HIV acquisition [92–95], suggesting that asymptomatic infections may play a significant role. We have demonstrated in South African women that asymptomatic STIs were just as inflammatory as symptomatic infections, but only 12% of women with laboratory confirmed STIs had clinical signs [8]. This suggests that women with asymptomatic infections are also at high risk of acquiring HIV. Treatment of HSV-2 has also been found to be ineffective at reducing HIV infection rates [87,96]. Although HSV-2 suppressive therapy may reduce genital ulceration, HSV-2 may induce a persistent state of susceptibility to HIV infection because of the ongoing inflammation it causes [34]. BV may also have been a significant factor contributing to the failure of these interventions, as BV has proven difficult to treat, with a recurrence rate of 50% within 6 months of antibiotic treatment [97].
Conclusion
Although we do not fully understand the causes of genital inflammation that is associated with high HIV acquisition risk in women, prevalent STIs and BV clearly play a major role. Syndromic diagnosis of these conditions are inadequate, with the vast majority of women asymptomatic. Current treatment strategies for HSV-2 and BV are ineffective, with HSV-2 suppressive therapy associated with ongoing genital inflammation and antibiotic treatment of BV having high recurrence rates. There is thus an urgent need for better strategies to manage STIs and BV in order to reduce genital inflammation in women at high risk for HIV infection.
Key points:
Genital inflammation places women at increased risk of HIV acquisition
Higher levels of cytokines in the lower genital tract result in chemotaxis of highly activated HIV target cells to the mucosa
STIs and BV are major drivers of genital inflammation
Acknowledgements
We would like to thank the CAPRISA clinical team and Prof Salim Abdool Karim for strong support with the study and conceptual input. We would also like to acknowledge the WISH clinical team, Dr Shaun Barnabas and Smritee Dabee for help with the manuscript.
Financial support and sponsorship:
This work was supported by the EDCTP Strategic Primer, Poliomyelitis Research Foundation of SA, and SA DST-NRF Centre of Excellence in HIV prevention. LM is a recipient of an NRF Research Career Advancement Award. Additionally we acknowledge K08HD 069201 to HBJ and R01 HD083040 to JAP and HBJ.
Footnotes
Conflicts of interest
All authors have no conflicts of interest to declare.
References
- 1.UNAIDS. UNAIDS Report on the global AIDS Epidemic 2010 Available from: http://www.unaids.org/globalreport/. Accessed 15 October 2015.
- 2.UNAIDS. The gap report Available from: http://www.unaids.org/en/media/unaids/contentassets/documents/unaidspublication/2014/UNAIDS_Gap_report_en.pdf. Accessed 15 October 2015.
- 3.South African Department of Health Report. The 2012 National antenatal sentinel HIV & Herpes simplex type-2 prevalence survey in South Africa. (Department of Health, Pretoria, 2014) Available from: http://www.health-e.org.za/wpcontent/uploads/2014/05/ASHIVHerp_Report2014_22May2014.pdf. Accessed 15 October 2015.
- 4.Kaul R, Ball TB, Hirbod T. Defining the genital immune correlates of protection against HIV acquisition: co-infections and other potential confounders. Sex Transm Infect 2011; 87:125e130. [DOI] [PubMed] [Google Scholar]
- 5.Laga M, Manoka A, Kivuvu M, et al. Non-ulcerative sexually transmitted diseases as risk factors for HIV-1 transmission in women: results from a cohort study. AIDS 1993; 7:95–102. [DOI] [PubMed] [Google Scholar]
- 6.Taha TE, Hoover DR, Dallabetta GA, et al. Bacterial vaginosis and disturbances of vaginal flora: association with increased acquisition of HIV. AIDS 1998;12:1699–706. [DOI] [PubMed] [Google Scholar]
- 7.Low N, Chersich MF, Schmidlin K, et al. Intravaginal practices, bacterial vaginosis, and HIV infection in women: individual participant data meta-analysis. PLoS Med 2011; 8:e1000416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Mlisana K, Naicker N, Werner L, et al. Vaginal discharge is a poor predictor of sexually transmitted infections and subclinical genital tract inflammation in women at high-risk of HIV infection. J Infect Dis 2012; 206:6–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Masson L, Passmore JAS, Liebenberg LJ, et al. Genital inflammation and the risk of HIV acquisition in women. Clin Infect Dis 2015; civ298.** This is the first study to demonstrate a relationship between elevated genital inflammatory cytokine concentrations and increased risk of HIV acquisition in vivo, suggesting that genital inflammatory responses play a crucial role in establishment of HIV infection.
- 10.Anahtar MN, Byrne EH, Doherty KE, et al. Cervicovaginal Bacteria Are a Major Modulator of Host Inflammatory Responses in the Female Genital Tract. Immunity 2015; 42:965–976.** This study comprehensively describes the relationship between high vaginal bacterial diversity and local inflammation, providing a possible cause for the genital inflammation observed in South African women that was associated with HIV acquisition risk. Additionally, this work describes the unique vaginal microbiome of South African women.
- 11.Li Q, Estes JD, Schlievert PM, et al. Glycerol monolaurate prevents mucosal SIV transmission. Nature 2009; 458:1034–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Stanford MM TB Issekutz. The relative activity of CXCR3 and CCR5 ligands in T lymphocyte migration: concordant and disparate activities in vitro and in vivo. J Leukoc Biol 2003; 74:791–799. [DOI] [PubMed] [Google Scholar]
- 13.Wira CR, Fahey JV, Sentman CL, et al. Innate and adaptive immunity in female genital tract: cellular responses and interactions. Immunol Rev 2005; 206: 306–335. [DOI] [PubMed] [Google Scholar]
- 14.Dieu-Nosjean MC, Vicari A, Lebecque S, et al. Regulation of dendritic cell trafficking: a process that involves the participation of selective chemokines. J Leukoc Biol 1999; 66:252–262. [DOI] [PubMed] [Google Scholar]
- 15.Mueller A, Strange PG. The chemokine receptor, CCR5. Int J Biochem Cell Biol 2004; 36:35–38. [DOI] [PubMed] [Google Scholar]
- 16.Nkwanyana NN, Gumbi P, Roberts L, et al. Impact of human immunodeficiency virus 1 infection and inflammation on the composition and yield of cervical mononuclear cells in the female genital tract. Immunol 2009; 128:e746–e757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Arnold KB, Burgener A, Birse K, et al. Increased levels of inflammatory cytokines in the female reproductive tract are associated with altered expression of proteases, mucosal barrier proteins, and an influx of HIV-susceptible target cells. Mucosal Immunol 2015.** This study describes the proteomic profile and activation of HIV target cells that are associated with female genital tract inflammation, as well as possible mechanisms for the relationship between elevated genital inflammatory cytokine concentrations and increased HIV acquisition risk.
- 18.Haase AT. Early events in sexual transmission of HIV and SIV and opportunities for interventions. Annu Rev Med 2011; 62:127–139. [DOI] [PubMed] [Google Scholar]
- 19.Cocchi F, DeVico AL, Garzino-Demo A, et al. Identification of RANTES, MIP-1α, and MIP-1β as the major HIV-suppressive factors produced by CD8+ T cells. Science 1995; 270:1811–1815. [DOI] [PubMed] [Google Scholar]
- 20.Hirbod T, Nilsson J, Andersson S, et al. Upregulation of interferon-α and RANTES in the Cervix of HIV-1-seronegative women with high-risk behavior. JAIDS 2006; 43:137–143. [DOI] [PubMed] [Google Scholar]
- 21.Lajoie J, Juno J, Burgener A, et al. A distinct cytokine and chemokine profile at the genital mucosa is associated with HIV-1 protection among HIV-exposed seronegative commercial sex workers. Mucosal Immunol 2012; 5:277–287. [DOI] [PubMed] [Google Scholar]
- 22.Lajoie J, Kimani M, Plummer FA, et al. Association of sex work with reduced activation of the mucosal immune system. J Infect Dis 2014; 210:319–329. [DOI] [PubMed] [Google Scholar]
- 23.Osborn L, Kunkel S, Nabel GJ. Tumor necrosis factor-alpha and interleukin-1 stimulate the human immunodeficiency virus enhancer by activation of the nuclear factor kappa B. Proc Natl Acad Sci 1989; 86:2336–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nazli A, Chan O, Dobson-Belaire WN, et al. Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLoS Path 2010; 6:e1000852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Naranbhai V, Karim SSA, Altfeld M, et al. Innate immune activation enhances HIV acquisition in women, diminishing the effectiveness of tenofovir microbicide gel. J Infect Dis 2012; 206:993–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kahle EM, Bolton M, Hughes JP, et al. Plasma Cytokine Levels and Risk of HIV Type 1 (HIV-1) Transmission and Acquisition: A Nested Case-Control Study Among HIV-1–Serodiscordant Couples. J Infect Dis 2015; 211:1451–1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Masson L, Mlisana K, Little F, et al. Defining genital tract cytokine signatures of sexually transmitted infections and bacterial vaginosis in women at high risk of HIV infection: a cross-sectional study. Sex Transm Infect 2014; 90:580–587. [DOI] [PubMed] [Google Scholar]
- 28.Koning FA, Otto SA, Hazenberg MD, et al. Low-level CD4+ T cell activation is associated with low susceptibility to HIV-1 infection. J Immunol 2005; 175:6117–6122. [DOI] [PubMed] [Google Scholar]
- 29.Paiardini M, Cervasi B, Reyes-Aviles E, et al. Low levels of SIV infection in sooty mangabey central memory CD4+ T cells are associated with limited CCR5 expression. Nature Med 2011; 17:830–836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Jaspan HB, Liebenberg LJ, Hanekom W, et al. Immune activation in the female genital tract during HIV infection predicts mucosal CD4 depletion and HIV shedding. J Infect Dis 2011; 204:1550–1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Levine WC, Pope V, Bhoomkar A, et al. Increase in endocervical CD4 lymphocytes among women with nonulcerative sexually transmitted diseases. J Infect Dis 1998; 177:167–74. [DOI] [PubMed] [Google Scholar]
- 32.Fichorova RN, Jasvantrai Desai P, Gibson FC, et al. Distinct proinflammatory host responses to Neisseria gonorrhoeae infection in immortalized human cervical and vaginal epithelial cells. Infect Immun 2001; 69:5840–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Reddy BS, Rastogi S, Das B, et al. Cytokine expression pattern in the genital tract of Chlamydia trachomatis positive infertile women–implication for T‐cell responses. Clin Exp Immunol 2004; 137:552–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rebbapragada A, Wachihi C, Pettengell C, et al. Negative mucosal synergy between Herpes simplex type 2 and HIV in the female genital tract. AIDS 2007; 21:589–98. [DOI] [PubMed] [Google Scholar]
- 35.Mcgee ZA, Jensen RL, Clemens CM, et al. Gonococcal Infection of Human Fallopian Tube Mucosa in Organ Culture: Relationship of Mucosal Tissue TNF-α Concentration to Sloughing of Ciliated Cells. Sex Transm Dis 1999; 26:160–5. [DOI] [PubMed] [Google Scholar]
- 36.Svanborg C, Godaly G, Hedlund M. Cytokine responses during mucosal infections: role in disease pathogenesis and host defence. Curr Opin Microbiol 1999; 2:99–105. [DOI] [PubMed] [Google Scholar]
- 37.Parks KS, Dixon PB, Richey CM, et al. Spontaneous Clearance of Chlamydia trachomatis Infection in Untreated Patients. Sex Transm Dis 1997; 24: 229–35. [DOI] [PubMed] [Google Scholar]
- 38.Golden MR, Schillinger JA, Markowitz L, et al. Duration of Untreated Genital Infections With Chlamydia trachomatis: A Review of the Literature. Sex Transm Dis 2000; 27:329–37. [DOI] [PubMed] [Google Scholar]
- 39.Houlihan CF, Larke NL, Watson-Jones D, et al. HPV infection and increased risk of HIV acquisition. A systematic review and meta-analysis. AIDS 2012; 26:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Masese L, Baeten JM, Richardson BA, et al. Changes in the contribution of genital tract infections to HIV acquisition among Kenyan high-risk women from 1993 to 2012. AIDS 2015; 29:1077–1085.** This analysis calculated the population attributable risk of various STIs to HIV incidence, and found HSV-2 and BV to be the biggest players through time.
- 41.Zhu J, Hladik F, Woodward A, et al. Persistence of HIV-1 receptor–positive cells after HSV-2 reactivation is a potential mechanism for increased HIV-1 acquisition. Nature Med 2009; 15:886–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Johnson LF, Dorrington RE, Bradshaw D, et al. The role of sexually transmitted infections in the evolution of the South African HIV epidemic. Trop Med Int Health 2012; 17:161–168. [DOI] [PubMed] [Google Scholar]
- 43.Eschenbach DA, Davick PR, Williams BL, et al. Prevalence of hydrogen peroxide-producing Lactobacillus species in normal women and women with bacterial vaginosis. J Clin Microbiol 1989; 27:251–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fredricks DN, Fiedler TL, Marrazzo JM. Molecular Identification of Bacteria Associated with Bacterial Vaginosis. N Engl J Med 2005; 353:1899–911. [DOI] [PubMed] [Google Scholar]
- 45.Srinivasan S, Liu C, Mitchell CM, et al. Temporal variability of human vaginal bacteria and relationship with bacterial vaginosis. PloS one 2010; 5:e10197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gajer P, Brotman RM, Bai G, et al. Temporal dynamics of the human vaginal microbiota. Sci Transl Med 2012; 4:132ra152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ravel J, Gajer P, Abdo Z, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci USA 2011; 108:4680–4687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Spurbeck RR, Arvidson CG. Lactobacilli at the front line of defense against vaginally acquired infections. Future Microbiol 2011; 6:567–582. [DOI] [PubMed] [Google Scholar]
- 49.van de Wijgert JH, Verwijs MC, Turner AN, et al. Bacterial vaginosis and vaginal yeast, but not vaginal cleansing, increase HIV-1 acquisition in African women. J AIDS 2008; 48:203–210. [DOI] [PubMed] [Google Scholar]
- 50.Mirmonsef P, Krass L, Landay A, et al. The role of bacterial vaginosis and trichomonas in HIV transmission across the female genital tract. Current HIV Res 2012; 10:202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Myer L, Denny L, Telerant R, et al. Bacterial vaginosis and susceptibility to HIV infection in South African women: a nested case-control study. J Infect Dis 2005; 192:1372–1380. [DOI] [PubMed] [Google Scholar]
- 52.Myer L, Kuhn L, Stein ZA, et al. Intravaginal practices, bacterial vaginosis, and women’s susceptibility to HIV infection: epidemiological evidence and biological mechanisms. Lancet Infect Dis 2005; 5:786–794. [DOI] [PubMed] [Google Scholar]
- 53.Madan RP, Masson L, Tugetman J, et al. Innate antibacterial activity in female genital tract secretions is associated with increased risk of HIV acquisition. AIDS Res Hum Retroviruses 2015. July 14. [DOI] [PMC free article] [PubMed]
- 54.Borgdorff H, Gautam R, Armstrong SD, et al. Cervicovaginal microbiome dysbiosis is associated with proteome changes related to alterations of the cervicovaginal mucosal barrier. Mucosal Immunol 2015.** Here the authors performed targeted and unbiased proteomic analysis of women with BV and provide an in depth description of host proteins that are altered in women with BV and are involved in disruption of mucosal barrier.
- 55.Atarashi K, Tanoue T, Shima T, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011; 331:337–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci USA 2010; 107:12204–12209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Libby EK, Pascal KE, Mordechai E, et al. Atopobium vaginae triggers an innate immune response in an in vitro model of bacterial vaginosis. Microbes Infect 2008; 10:439–446. [DOI] [PubMed] [Google Scholar]
- 58.Doerflinger SY, Throop AL, Herbst-Kralovetz MM. Bacteria in the vaginal microbiome alter the innate immune response and barrier properties of the human vaginal epithelia in a species-specific manner. J Infect Dis 2014; 209:1989–1999. [DOI] [PubMed] [Google Scholar]
- 59.Kyongo JK, Jespers V, Goovaerts O, et al. Searching for lower female genital tract soluble and cellular biomarkers: defining levels and predictors in a cohort of healthy Caucasian women. PloS one 2012; 7:e43951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Deese J, Masson L, Miller W, et al. Injectable Progestin‐Only Contraception is Associated With Increased Levels of Pro‐Inflammatory Cytokines in the Female Genital Tract. Am J Reprod Immunol 2015; 74:357–367. [DOI] [PubMed] [Google Scholar]
- 61.Myer L, Denny L, Wright TC, et al. Prospective study of hormonal contraception and women’s risk of HIV infection in South Africa. International J Epidemiol 2007; 36:166–174. [DOI] [PubMed] [Google Scholar]
- 62.Heffron R, Donnell D, Rees H, et al. Use of hormonal contraceptives and risk of HIV-1 transmission: a prospective cohort study. Lancet Infect Dis 2012; 12:19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Marx PA, Spira AI, Gettie A, et al. Progesterone implants enhance SIV vaginal transmission and early virus load. Nature Med 1996; 2:1084–1089. [DOI] [PubMed] [Google Scholar]
- 64.Sodora DL, Gettie A, Miller CJ, et al. Vaginal transmission of SIV: assessing infectivity and hormonal influences in macaques inoculated with cell-free and cell-associated viral stocks. AIDS Res Hum Retroviruses 1998; 14:S119–123. [PubMed] [Google Scholar]
- 65.Smith SM, Mefford M, Sodora D, et al. Topical estrogen protects against SIV vaginal transmission without evidence of systemic effect. AIDS 2004; 18:1637–1643. [DOI] [PubMed] [Google Scholar]
- 66.Miller L, Patton DL, Meier A, et al. Depomedroxyprogesterone-induced hypoestrogenism and changes in vaginal flora and epithelium. Obstet Gynecol 2000; 96:431–439. [DOI] [PubMed] [Google Scholar]
- 67.Critchlow CW, Wölner-Hanssen P, Eschenbach DA, et al. Determinants of cervical ectopia and of cervicitis: age, oral contraception, specific cervical infection, smoking, and douching. Am J Obstet Gynecol 1995; 173:534–543. [DOI] [PubMed] [Google Scholar]
- 68.Baeten JM, Nyange PM, Richardson BA, et al. Hormonal contraception and risk of sexually transmitted disease acquisition: results from a prospective study. Am J Obstet Gynecol 2001; 185:380–385. [DOI] [PubMed] [Google Scholar]
- 69.Miller L, Patton DL, Meier A, et al. Depomedroxyprogesterone-induced hypoestrogenism and changes in vaginal flora and epithelium. Obstet Gynecol 2000; 96:431–439. [DOI] [PubMed] [Google Scholar]
- 70.Morrison CS, Bright P, Wong EL, et al. Hormonal contraceptive use, cervical ectopy, and the acquisition of cervical infections . Sex Transm Dis 2004; 31:561–567. [DOI] [PubMed] [Google Scholar]
- 71.Pettifor A, Delaney S, Kleinschmidt I et al. Use of injectable progestin contraception and risk of STI among South African women. Contraception 2009; 80:555–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ildgruben AK, Sjoberg IM, Hammarstrom ML. Influence of hormonal contraceptives on the immune cells and thickness of human vaginal epithelium. Obstet Gynecol 2003; 102:571–582. [DOI] [PubMed] [Google Scholar]
- 73.Ghanem KG, Shah N, Klein RS, et al. Influence of sex hormones, HIV status, and concomitant sexually transmitted infection on cervicovaginal inflammation. J Infect Dis 2005; 191:358–366. [DOI] [PubMed] [Google Scholar]
- 74.Chandra N, Thurman AR, Anderson S, et al. Depot medroxyprogesterone acetate increases immune cell numbers and activation markers in human vaginal mucosal tissues. AIDS Res Hum Retroviruses 2013; 29:592–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Huijbregts RP, Helton ES, Michel KG, et al. Hormonal contraception and HIV-1 infection: medroxyprogesterone acetate suppresses innate and adaptive immune mechanisms. Endocrinol 2013; 154:1282–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Prakash M, Patterson S, Kapembwa MS. Hormonal upregulation of CCR5 expression on T lymphocytes as a possible mechanism for increased HIV-1 risk. JAIDS 2005; 38:S14–16. [DOI] [PubMed] [Google Scholar]
- 77.Prakash M, Kapembwa MS, Gotch F, et al. Oral contraceptive use induces upregulation of the CCR5 chemokine receptor on CD4(+) T cells in the cervical epithelium of healthy women. J Reprod Immunol 2002; 54:117–131. [DOI] [PubMed] [Google Scholar]
- 78.Zang YC, Halder JB, Hong J, et al. Regulatory effects of estriol on T cell migration and cytokine profile: inhibition of transcription factor NF-kappa B. J Neuroimmunol 2002; 124:106–114. [DOI] [PubMed] [Google Scholar]
- 79.Ngcapu S, Masson L, Sibeko S, et al. Lower concentrations of chemotactic cytokines and soluble innate factors in the lower female genital tract associated with the use of injectable hormonal contraceptive. J Reprod Immunol 2015; 110:14–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Scholes D, Daling JR, Stergachis A, et al. Vaginal douching as a risk factor for acute pelvic inflammatory disease. Obstet Gynecol 1993; 81:601–606. [PubMed] [Google Scholar]
- 81.Sharkey DJ, Tremellen KP, Jasper MJ, et al. Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol 2012; 188:2445–2454. [DOI] [PubMed] [Google Scholar]
- 82.Fichorova RN, Tucker LD, Anderson DJ. The molecular basis of nonoxynol-9-induced vaginal inflammation and its possible relevance to human immunodeficiency virus type 1 transmission. J Infect Dis 2001; 184:418–428. [DOI] [PubMed] [Google Scholar]
- 83.Kleppa E, Ramsuran V, Zulu S, et al. Effect of female genital schistosomiasis and anti-schistosomal treatment on monocytes, CD4+ T-cells and CCR5 expression in the female genital tract. PLoS One 2014; 9:e98593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zabaleta J, Schneider BG, Ryckman K, et al. Ethnic differences in cytokine gene polymorphisms: potential implications for cancer development. Cancer Immunol Immunother 2008; 57:107–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.World Health Organization. 2003. Guidelines for the management of sexually transmitted infections Available at: http://www.who.int/hiv/pub/sti/en/STIGuidelines2003.pdf. Accessed 15 October 2015. [PubMed]
- 86.Wilkinson D, Abdool Karim SS, Harrison A, et al. Unrecognized sexually transmitted infections in rural South African women: a hidden epidemic. Bulletin of the World health Organization 1999; 77:22–28. [PMC free article] [PubMed] [Google Scholar]
- 87.Celum C, Wald A, Hughes J, et al. Effect of aciclovir on HIV-1 acquisition in herpes simplex virus 2 seropositive women and men who have sex with men: a randomised, double-blind, placebo-controlled trial. Lancet 2008; 371:2109–2119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Gray RH, Wawer MJ, Brookmeyer R, et al. Probability of HIV-1 transmission per coital act in monogamous, heterosexual, HIV-1-discordant couples in Rakai, Uganda. Lancet 2001; 357:1149–1153. [DOI] [PubMed] [Google Scholar]
- 89.Kaul R, Kimani J, Nagelkerke NJ, et al. Monthly antibiotic chemoprophylaxis and incidence of sexually transmitted infections and HIV-1 infection in Kenyan sex workers: a randomized controlled trial. JAMA 2004; 291:2555–2562. [DOI] [PubMed] [Google Scholar]
- 90.Watson-Jones D, Weiss HA, Rusizoka M, et al. Effect of herpes simplex suppression on incidence of HIV among women in Tanzania. NEJM 2008; 358:1560–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wawer MJ, Sewankambo NK, Serwadda D, et al. Control of sexually transmitted diseases for AIDS prevention in Uganda: a randomised community trial. Lancet 1999; 353:525–535. [DOI] [PubMed] [Google Scholar]
- 92.Grosskurth H, Todd J, Mwijarubi E, et al. Impact of improved treatment of sexually transmitted diseases on HIV infection in rural Tanzania: randomised controlled trial. Lancet 1995; 346:530–536. [DOI] [PubMed] [Google Scholar]
- 93.Grosskurth H, Gray R, Hayes R, et al. Control of sexually transmitted diseases for HIV-1 prevention: understanding the implications of the Mwanza and Rakai trials. Lancet 2000; 355:1981–1987. [DOI] [PubMed] [Google Scholar]
- 94.Gregson S, Adamson S, Papaya S, et al. Impact and process evaluation of integrated community and clinic-based HIV-1 control: a cluster-randomised trial in eastern Zimbabwe. PLoS Med 2007; 4:e102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kamali A, Quigley M, Nakiyingi J, et al. Syndromic management of sexually-transmitted infections and behaviour change interventions on transmission of HIV-1 in rural Uganda: a community randomised trial. Lancet 2003; 361:645–652. [DOI] [PubMed] [Google Scholar]
- 96.Barnabas RV, Celum C. Infectious co-factors in HIV-1 transmission herpes simplex virus type-2 and HIV-1: new insights and interventions. Current HIV research 2012; 10:228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Barrons R, Tassone D. Use of Lactobacillus probiotics for bacterial genitourinary infections in women: a review. Clinical therapeutics 2008; 303:453–468. [DOI] [PubMed] [Google Scholar]