Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 1.
Published in final edited form as: Clin Lab Med. 2014 Sep 18;34(4):733–745. doi: 10.1016/j.cll.2014.08.005

Microbiome in HIV infection

January T Salas a, Theresa L Chang b,*
PMCID: PMC4254659  NIHMSID: NIHMS625720  PMID: 25439273

Abstract

HIV primary infection occurs at mucosa tissues, suggesting an intricate interplay between microbiome and HIV infection. Recent advanced technologies of high-throughput sequencing and bioinformatics allow researchers to explore nonculturable microbes including bacteria, virus and fungi and their association with diseases. HIV/SIV infection is associated with microbiome shifts and immune activation that may affect the outcome of disease progression. Similarly, altered microbiome and inflammation are associated with increased risks of HIV acquisition, suggesting the role of microbiome in HIV transmission. In this review, we will focus on microbiome in HIV infection at various mucosal compartments. Understanding the relationship between microbiome and HIV may offer insights into development of better strategies for HIV prevention and treatment.

Keywords: Microbiome, HIV transmission, HIV pathogenesis, immune activation, microbial translocation

Introduction

Recent studies have demonstrated the important role of microbiota/microbiome in human health and diseases at cellular and molecular levels 1-6. Interactions among microbes, nutrition, and immune response affect our health. For example, commensal bacteria protect the body from colonizing pathogenic bacteria by competing for space and nutrients 7. Bacterial metabolites (i.e. indole-3-aldehyde, butyric acid, hydrogen peroxide) could indirectly shape the host immune repertoire 8, 9. The advancement of techniques in sequencing and bioinformatics has made possible the characterization of microbial communities in health and diseased states. With this came an explosion of information shedding light on the important role microbiota in the human body, from nutrition and autoimmunity to its role in brain diseases 10.

An estimated 35.3 million people are living with human immunodeficiency virus (HIV) worldwide in addition to more than 2 million new cases since 2012 (UNAIDS 2013). Increasing evidence indicates microbiome plays a crucial role in HIV transmission and pathogenesis 11-13. Alteration of vaginal and rectal microbiome may influence HIV acquisition 11, 14-16 and mother-to-child transmission (MTCT) 17. Microbiome and immune response co-evolve in response to HIV infection during HIV pathogenesis that may determine the disease progression. Understanding the interplay between microbiome and HIV is vital for developing effective strategies for HIV prevention and treatment. This review will summarize the recent progress on microbiome in HIV transmission and pathogenesis. Although microbes are composed of bacteria, fungi, protozoa and virus, we primarily focus on bacteria due to the availability of published data.

Chronic immune activation in HIV pathogenesis

Persistent immune activation is a key feature of HIV and markers of inflammation are a better predictor of clinical outcome than viral load 18, 19. Heightened immune activation and inflammation in HIV patients are associated with increased age-related diseases such as cardiovascular, kidney, diabetes mellitus and bone fracture in HIV patients 20, 21. Immune activation and inflammatory markers decline with anti-retroviral therapy (ART), but always remain high compared to healthy controls 22, 23. Natural hosts of simian immunodeficiency virus (SIV), despite high viral loads, are able to avoid chronic infection through rapid controlled immune response hence do not develop acquired immune deficiency syndrome (AIDS) 24-26.

Depletion of CD4+ T cells occurs within weeks after HIV infection, notably in the gut compartment 27-30. Gut-associated interleukin-17 (IL-17) secreting CD4+ cells (Th17), important for mucosal defense against invading pathogens, are preferentially depleted 31-33. Microbial translocation, translocation of microbes or microbial products without overt bacteremia, occurs after disruption of gut mucosal membrane integrity and mucosal immune homeostasis which then could cause systemic immune activation 18. Plasma lipopolysaccharide (LPS) was elevated in HIV-infected patients and associated with an increase in plasma IFNα and frequency of activated CD8+ T cells (CD38+HLA-DR+) 18. Despite their activation status, only small portions of these CD8+ T cells are specific against HIV 30. Additionally, expression of program death-1 (PD-1) on HIV-specific CD8+ T cells (marker also elevated in Cytomegalovirus (CMV) and Epstein-Barr (EBV) infections) increases cell apoptosis and decreases their proliferative capacity 34. Thus, in response to HIV infection, microbial translocation may cause chronic activation, leading to inflammatory associated pathology and immune exhaustion. Note that the cause-and-effect relationship between microbial translocation and immune activation during chronic infection remains debatable despite several studies using animal models supporting the contribution of microbial translocation to immune activation (review in 21). While a recent study demonstrates early blockade of microbial translocation reduces inflammation and viral replication in SIV models35, the reciprocal interaction between microbial translocation and immune activation may contribute to SIV/HIV pathogenesis.

Microbiome shapes immune response

Microbiota has major effects on immune cells and epithelial cells at the mucosa36. Mice raised in a clean facility, a condition which prevents natural colonized by microbiota, have an increased mortality, bacteria burden upon challenge, and susceptibility to infection compared to counterparts raised in conventional environment7, 37, indicating the beneficial effects of early bacterial exposure on shaping host immunity. For instance, Lactobacillus casei induces IgA and IL-6 producing cells in mouse gut lamina propia 38, whereas segmented filamentous bacterium (SBF) promotes the differentiation of Th17 cells in the gut 39. In addition to the direct impact on immune response, microbes facilitate the processing and absorption of nutrients essential for immune functions such as short fatty acids (butyrate and acetate) and amino acids (tryptophan) 40. Since different bacterial species induce different immune responses, the type of bacterial composition in the compartment could influence the balance between inflammation and homeostasis 7.

Bacterial communities are diverse and their compositions fluctuate with hormones, diet, and immune responses. They can be classified into 3 classes: 1) symbionts, bacteria known to promote health, 2) commensals, permanent residents with no known beneficial or detrimental effect to the host, and 3) pathobionts, permanent residents with possibility to become pathogenic 7. Patients with HIV infection or other diseases have alterations of microbial compositions 41. Opportunistic pathogens such as Pseudomonas aeruginosa and Candida albicans are frequently found in HIV-infected patients who often have low to barely detectable levels of Bifidobacteria and Lactobacilli species in the gut 42. Bifidobacteria and Lactobacilli are known to help improve gut health and immune function 43, 44. Prebiotics/probiotics supplements in HIV patients on ART enhanced reconstitution of CD4+ T cells in the gastrointestinal (GI) tract, improved Th17 functionality, increased functionality and frequency of antigen presenting cells (APC), and decreased markers of immune activation. Likewise, an increase in Prevotella with decreased Bacteroides was associated with increased immune activation and microbial translocation 12. Colonization of commensal Lactobacillus crispatus, L. jensenii, and L. rhamnosus on vaginal epithelial cell in vitro dampened inflammatory cytokine induction via toll-like receptor (TLR) activation 36, 45. Taken together, symbiotic/commensal bacteria modulate mucosal immune cells and maintain immune homeostasis. When symbiotic/commensal bacteria are compromised by overgrowth of indigenous pathobionts, leading to dysbiosis, immune cells will be activated to control pathogens. Immune activation and inflammation will result in collateral damage to surrounding tissues 7, 37.

Bacterial metabolic products can modulate immune responses. For instance the lack of butyric acid, a fermentation product from butyrate producing bacteria in the gut, may lead to a decrease in regulatory T cells (Tregs) in inflammatory bowel disease 46. In HIV patients, enrichment of gut bacteria that catabolizes tryptophan, such as Pseudomonas fluorescens, inhibits Th17 cell differentiation and correlates with mucosal disruption 47. Likewise, intestinal Lactobacilli such as L. reuteri metabolize tryptophan to produce indole-3-aldehyde, which promotes IL-22 transcription 8. Attachment of commensal L. crispatus and L. jensinii to the vaginal epithelium down-regulates inflammatory cytokines such as IL-6, tumor necrosis factor-α (TNF-α), and IL-8 upon TLR-3 agonist polyinosinic:polycytidylic acid (polyIC) exposure, suggesting the immune-modulatory effect of colonization of commensal bacteria on epithelial cells 36. This indicates the presence of commensal bacteria regulates immune response of epithelial cells.

Changes in microbial communities in response HIV/SIV infection and their association with immune activation have been recently documented 14, 18, 26, 42, 47, 48. HIV-infected patients given prebiotic/probiotic supplements exhibited reduced inflammation, enhanced CD4 reconstitution and a subsequent improvement of prognosis, all highlighting the role of microbiota in HIV pathogenesis 5. In the following sections, we summarize microbiome in various compartments in context of HIV infection.

Oral and periodontal microbiome

Oral lesions, frequently observed in HIV-positive patients without ART, are considered as indicators of disease progression 49. Oral lesions are often the first manifestation of HIV in places where the access to regular health care or ART is limited. Periodontal pathogens are more prevalent in HIV-infected individuals 50. Oral microbial diversity with increased levels of total Lactobacillus species and Candida species was found to be greater in HIV-infected patients than uninfected controls 41. Conversely, HIV seropositive children have lower saliva bacterial species than uninfected children 51. Macaques with infection by SIVmac251 intravenously had fewer oral bacterial species than uninfected animals followed by an outgrowth of Gemella morbillorum 52. Certain microbial species are frequently found in HIV-infected patients but not in healthy individuals 53. Preferential overgrowth of bacterial taxa such Candida spp. Gemella, Streptococcus, Veillonella, and Porphyromonas gingivalis in HIV-infected patients has been described 28, 48, 49. Bacterial culture supernatant of P. gingivalis reactivates HIV infection in cells lines with latent HIV proviruses through butyric acid-mediated histone acetylation and chromatin remodeling 54. Elevated inflammatory cytokines such as IL-6, IL-8, and granulocyte macrophage colony stimulating factor (GM-CSF) in periodontal pockets is observed in HIV-infected patients which could be immune response to opportunistic bacteria 49.

ART decreases oral lesions such as candidiasis in HIV-infected patients 49, 55, although the mechanism is not well defined. Reduction in viral load may have a direct impact on microbiota composition and oral epithelial cells. In monkeys, SIV down-regulates genes involved in oral epithelial regeneration, leading to slow healing of lesions 56. A positive correlation between viral copies and bacterial growth has also been described 48. In healthy rhesus macaques, Streptococcus, Gemella, and Granulicatella are three major genera in the lingual epithelium of the tongue dorsum, whereas Streptococcus and Lachnospiraceae are the core bacterial taxa in the dental plaque 52. In response to intravenous SIV infection, there is a reduction of bacterial community diversity. Additionally, Gemella species became predominant and Streptococcus species were significantly reduced in the tongue dorsum. This dysbiosis is accompanied by induction of IFNγ signaling pathways in tongue tissues. In vitro stimulation of oral epithelial cells with IFNγ results in the reduction of antimicrobial peptides, suggesting that SIV infection changes the microbial community through IFNγ-mediated down-regulation of antimicrobial peptides 52. The findings in SIV-infected macaques are similar to those in humans. Streptococcus is one of the predominant commensal genera in the human lingual epithelium 57. The outgrown of Gemella species is also found in HIV-infected patients 48. Specifically, Gemella morbillorum is a common opportunistic pathogen in HIV-infected patients with periodontitis 28.

Gut microbiome

Enteropathy is a common disorder in HIV-infected patients with AIDS 58. Increasing evidence strongly indicates that HIV/SIV infection alters gut microbiome. Microbial analysis of colon biopsies reveals HIV-infected patients have more Proteobacteria and less Firmicutes than uninfected individuals 12. Further analysis at a genus level showed an increase in Prevotella and a decrease in Bacteroides in HIV-infected patients. Alteration of the microbial community is accompanied by an increase in activation of colonic T cells and myeloid dendritic cells 12. In chimpanzees, SIV infection alters gut microbial communities which become more diverse overtime 59. The abundances of Staphylococcus, Sarcina, and Selenomonas were increased in response to infection 59. Similarly, HIV-infected patients on ART with low viral loads have reduced populations of commensal bacteria that are replaced by bacterial communities including Brachyspira, Campylobacter, Catenibacterium, Escherichia, Enterobacteriaceae, Fusobacteriaceae, Mogibacterium, Prevotella and Ralstonia, 60. Total bacterial loads are similar among HIV-positive patients, HIV-positive patients on ART, and uninfected individuals. However there is a difference in microbial compositions between HIV-positive patients and uninfected group 47. HIV-positive group exhibited a profile of enriched pathobionts (i.e. Prevotella, Salmonella, Escherichia, Staphylococcus, and Campylobacter species) accompanied reduced symbionts (i.e. Clostridia and Bacteroides), a microbial profile referred as disease-associated microbial communities (DMC) 47, 60. HIV-positive patients on ART have intermediate DMC profile between untreated HIV-positive patients and uninfected individuals. ART results in a shift in the DMC that resembles microbial communities in uninfected individuals. Studies of HIV-infected patients on ART, comparing partial immunological responders (>200/ l CD4 count) and immunological non-responders (<200/ l), indicate that immunological nonresponders have increased microbial translocation with elevated circulating 16S rDNA levels of more pathogenic Enterobacteriaceae and less of immunomodulatory Lactobacillus species 61. Enrichment of genus Prevotella in HIV-positive patients was associated with increased immune activation and microbial translocation 12. Prevotella malaninogenica enhanced HIV-1 expression in THP-1/NL4-3luc cells by induction TLR-2 in vitro 62. Gut mucosal dysbiosis characterized by pathobionts, a common feature in HIV infected individuals, is strongly associated with microbial translocation, immune activation, and viral persistence. The role of microbiome on the immune response inHIV-infected patients was demonstrated by administration of probiotics/prebiotics in conjunction with ART leading to improved reconstitution of CD4 T cells, enhanced IL-23 secretion and number of APC, and increased the percentage of multifunctional Th17 cells in the gut 5, 44, 63.

Altered microbiome in HIV-positive patients is also associated with high T cell activation, plasma inflammation markers (IL-6, TNF), diminished mucosal IL-17 and IL-22 secreting cells, and increased tryptophan catabolism 47, 60. Diminished gut Th17 CD4+ T cells in HIV are believed to be the catalyst in the disruption of mucosal barrier 42. Indeed, Th17 cells are essential to mucosal homeostasis by orchestrating immune responses against invading pathogens and promoting mucosal barrier integrity through supporting the production of tight junction proteins and claudins 64. Th17 cells are preferentially infected by the HIV leading to diminished numbers 31-33. Recent evidence suggests gut microbiota may determine the fate of Th17 cells 7, 31. Firmicutes, a segmented filamentous bacterium, is essential for the development of Th17 cells 39. By contrast, the gut bacteria Pseudomonas fluorescens is enriched in HIV-infected patients and has the capacity to catabolize tryptophan, generating catabolites that inhibit Th17 differentiation 47. Indolamine-2,3-dioxygenase (IDO1) generated by myeloid dendritic cells catabolizes tryptophan to skew the differentiation of Tregs over Th17 cells 9.

Gut microbiota provides the host the necessary nutrients and is critical in the development of a proper immune response 65. A recent study by Shulzhenko et al reveals a bidirectional interaction between gut microbiota, mucosal epithelium and gut-associated B cells, whereby microbiota influences the balance between epithelial immune function and metabolic function. In the absence of IgA-secreting B cells, gut microbiota up-regulates epithelial immune functions (up-regulation IFN-induced genes and the complement system) at the expense of metabolic functions (down-regulation of lipid, carbohydrate, and micronutrients related genes) 2. Similarly, probiotic Lactobacillus casei administration to mice increased IgA producing cells that were not specific to L. casei 38. Gene expression analysis of duodenal biopsies from HIV-infected patients reveals there is an up-regulation of epithelial immune functions and down-regulation of epithelial metabolism, which could explain malnutrition and malabsorption found in HIV-infected patients 2. Taken together, microbiota/microbiome not only regulates immune response but also nutrition and health in gut epithelium. It also modulates epithelial turnover, shedding, tight-junctions, apoptosis, and autophagy 66.

Rectal microbiome in HIV

Rectal transmission is the main route of HIV transmission in men who have sex with men (MSM). In 2010, 78% of new infections were among MSM (CDC). HIV infection is associated with a significant reduction in the diversity of microbial species in rectal mucosa however combined ART reverses this reduction to levels similar to healthy controls 14. HIV-infected individuals have enrichment of Fusobacteria, Anaerococcus, Peptostreptococcus and Porphyromonas with a depletion of Roseburia, Ruminococcus, Eubacterium, Coprococcus and Lachnospira 14. Analysis of metagenomic pathways demonstrated a down-regulation of genes related to amino acid, fructose/mannose metabolism and CoA biosynthesis 14. Similarly, as infection progressed in HIV-positive MSMs, there was reduced alpha diversity and enrichment of Fusobacteria 67.

Fusobacteria, enriched in HIV-infected individuals 14, 67, are abundant in the colorectal adenoma and associated with an increase in the local cytokine milieu suggestive of mucosal inflammation 68. Fusobacterium isolated from ulcerative colitis secrete n-butyric acid 69, a known HDAC inhibitor that can reactivate latent HIV 70, 71. It remains to be determined whether enrichment of butyric acid producing Fusobacteria in rectal mucosa could increase HIV transmission.

Vaginal and cervical microbiome

Most studies that associate HIV with alterations of vaginal microbiota are performed in women diagnosed with bacterial vaginosis (BV) 72-74, which is a clinical symptom caused by an imbalance of commensal bacteria in the female genital tract. The predominant Lactobacillus species in the vagina are replaced by overgrowth of anaerobic bacteria such as Gardnerella, Atopobium, Prevotella and several other taxa 72, 73. A meta-analysis study comprising of 23 publications with a total of 30,739 women clearly indicates an association between BV and increased risk of HIV acquisition 1. BV not only increases the risk HIV acquisition but also transmission. HIV positive women with BV are greater than 3-fold more likely to transmit HIV to their HIV negative male partners than HIV positive women without BV 11. HIV MTCT was increased when there was alteration of vaginal microbiota to a Gardnerella vaginalis dominant species 17. This clearly shows that the predominant community in a dysbiotic female reproductive tract alters environment facilitating HIV entry and transmission 75.

The vaginal microbiome comprises a dynamic ecosystem with important host defense capabilities that promote reproductive health. It is a kinetic ecosystem where the proportion of bacterial communities varies with hormonal changes, sexual activity, age, and race 10, 76. Nevertheless, the vaginal microbiome can be classified into 7 community types. The majority of vaginal communities, types I, II, III, and V, are dominated by one or more species of Lactobacillus that constitute the majority of all sequences obtained, and are associated with low vaginal pH (pH 4.0-5.0) 77. Lactobacillus dominance is lost in the type IV community, while Gardnerella vaginalis dominates the type VI community. Both types IV and VI are at higher risk for BV 72, 76. The type VII community has high, approximately even proportions of G. vaginalis and Lactobacillus spp. Lactobacilli ferment glucose and produce lactic acid to maintain vaginal pH at an acidic state. The acidic pH environment is thought to prevent non-resident pathobionts from inhabiting and protects hosts against viral infection. H2O2-producing Lactobacillus acidophillus has an anti-HIV effect 78. Lactobacilllus species do not trigger proinflammatory cytokine production in vaginal epithelial cells but non-resident skin bacteria Staphylococcus epidermis does 36.

It is not clear how BV increases HIV acquisition and transmission but it has been shown that BV increases viral shedding, increases the abundance of target cells, promotes the production of pro-inflammatory cytokines, and disrupts membrane integrity 3, 15, 79-81. BV results in an increase in inflammatory cytokines 15 which remains present even during immunosuppressive states such as pregnancy 80. Also, high levels of inflammatory cytokines such as IL-1β, IL-6 and IL-8 in cervicovaginal lavages (CVL) correlates with reduction of CD4 T cells 79. Furthermore, high concentrations of proinflammatory cytokines in CVL increased HIV-1 RNA levels and viral shedding 81. Atopobium vaginae, not L. inners nor L. crispatus, induce proinflammatory cytokines, antimicrobial peptides, and disrupt the mucosal barrier 13. To summarize, vaginal dysbiosis in the setting of an inflammatory condition such as BV could increase HIV transmission by increasing target cells at the site, disrupting the mucosal barrier, and increasing viral shedding.

Penile microbiome in HIV

Male circumcision reduces the incidences of urinary tract infection 6 and HIV acquisition 82. Although the underlying mechanism remains to be defined, it is thought that penile foreskin, which is removed during circumcision, can trap HIV virions during sexual intercourse, increasing the opportunity for infection 6. Additionally, the foreskin is lined with moist mucosal epithelium that provides an ideal environment for pro-inflammatory anaerobic bacteria and immune cells such as Langerhans cells (LC) and CD4 T cells, target cells for HIV infection 83. Foreskin inflammation manifested by massive infiltrates of CD4 and CD8 T cells increases HIV infection 84. Male circumcision significantly reduces the bacterial load, prevalence of some but not all anaerobic bacteria, and reduces microbiota diversity and composition 85, 86. Studies have clearly shown that circumcision reduces the risk of HIV acquisition and bacterial and viral sexually transmitted infections (STIs) 87, 88; however, the underlying mechanism remains to be defined.

Lung microbiome in HIV

Bacterial pneumonia is frequently observed in HIV-positive patients 89. Opportunistic infections in HIV-infected patients have been attributed to their state of immunosuppression. Bacterial rDNAs are detectable in lungs of HIV-infected individuals with no respiratory symptoms 89. Broncheoalveolar lavage of HIV-infected patients with pneumonia (from the San Francisco area) has greater a number of taxa including Actinobacteria, Firmicutes, Cyanobacteria, Bacteroidetes, and Chloroflexi when compared to non-HIV patients with pneumonia 90. On the other hand, HIV-infected patients (from Uganda cohort) showed a enrichment in members of Lachnospiraceae and sulfur reducing bacteria (Desulfovibrionaceae and Desulfuromonadaceae) and exhibited even more diverse bacterial communities than the western cohort 91. Pseudomonas aeruginosa pathogen was associated with pneumonia in the Uganda HIV cohort 91. A metabolomics study of BAL from HIV-positive patients demonstrated increased levels of pyochelin metabolite, a by-product of P.aeroginosa, which was not observed in healthy individuals 92. HIV-infected patients have a high abundance of Tropheryma whipplei, the causative agent of Whipple’s disease, in the lung, which is significantly reduced by ART 4.

Diversity in lung microbiome between different cohorts of HIV-infected patients can be attributed to factors such as antimicrobial therapy, clinical status, ethnicity (genetics), diet and environmental exposure 91, 93. Germ-free mice upon challenge with ovalbumin exhibited exaggerated airway eosinophilia with increased production of IgE and Th2 cytokines compared to pathogen-free mice 93. The contribution of environmental microbial exposures to HIV pathogenesis remains to be explored.

Conclusion

HIV infection alters microbial communities in various mucosal compartments that may contribute to microbial translocation and immune activation during HIV disease progression. Conversely, microbiomes in the genital or rectal mucosa can impact the local immune response and HIV transmission. The bidirectional cross-talk between microbiome and the immune response may influence HIV transmission and pathogenesis. Further studies delineating the interplay between microbiome and immunity will offer insight into the development of a better strategy for HIV prevention and treatment.

Key points.

  • HIV infected individuals have altered microbiome associated with immune activation that impact the consequence of disease progression.

  • Genital and rectal microbiome may modulate immune response and affect HIV transmission.

Acknowledgments

This work was supported by NIH R01AI110372.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

The authors have nothing to disclose.

References

  • 1.Atashili J, Poole C, Ndumbe PM, et al. Bacterial vaginosis and HIV acquisition: a meta-analysis of published studies. Aids. 2008;22(12):1493–1501. doi: 10.1097/QAD.0b013e3283021a37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Shulzhenko N, Morgun A, Hsiao W, et al. Crosstalk between B lymphocytes, microbiota and the intestinal epithelium governs immunity versus metabolism in the gut. Nature medicine. 2011;17(12):1585–1593. doi: 10.1038/nm.2505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Petrova MI, van den Broek M, Balzarini J, et al. Vaginal microbiota and its role in HIV transmission and infection. FEMS microbiology reviews. 2013;37(5):762–792. doi: 10.1111/1574-6976.12029. [DOI] [PubMed] [Google Scholar]
  • 4.Lozupone C, Cota-Gomez A, Palmer BE, et al. Widespread colonization of the lung by Tropheryma whipplei in HIV infection. American journal of respiratory and critical care medicine. 2013;187(10):1110–1117. doi: 10.1164/rccm.201211-2145OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Klatt NR, Canary LA, Sun X, et al. Probiotic/prebiotic supplementation of antiretrovirals improves gastrointestinal immunity in SIV-infected macaques. The Journal of clinical investigation. 2013;123(2):903–907. doi: 10.1172/JCI66227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Anderson D, Politch JA, Pudney J. HIV infection and immune defense of the penis. American journal of reproductive immunology. 2011;65(3):220–229. doi: 10.1111/j.1600-0897.2010.00941.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nature reviews Immunology. 2009;9(5):313–323. doi: 10.1038/nri2515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zelante T, Iannitti RG, Cunha C, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39(2):372–385. doi: 10.1016/j.immuni.2013.08.003. [DOI] [PubMed] [Google Scholar]
  • 9.Favre D, Mold J, Hunt PW, et al. Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease. Science translational medicine. 2010;2(32):32ra36. doi: 10.1126/scitranslmed.3000632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Human Microbiome Project C Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–214. doi: 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cohen CR, Lingappa JR, Baeten JM, et al. Bacterial vaginosis associated with increased risk of female-to-male HIV-1 transmission: a prospective cohort analysis among African couples. PLoS medicine. 2012;9(6):e1001251. doi: 10.1371/journal.pmed.1001251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dillon SM, Lee EJ, Kotter CV, et al. An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia. Mucosal immunology. 2014;7(4):983–994. doi: 10.1038/mi.2013.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.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. The Journal of infectious diseases. 2014;209(12):1989–1999. doi: 10.1093/infdis/jiu004. [DOI] [PubMed] [Google Scholar]
  • 14.McHardy IH, Li X, Tong M, et al. HIV Infection is associated with compositional and functional shifts in the rectal mucosal microbiota. Microbiome. 2013;1(1):26. doi: 10.1186/2049-2618-1-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sturm-Ramirez K, Gaye-Diallo A, Eisen G, et al. High levels of tumor necrosis factor-alpha and interleukin-1beta in bacterial vaginosis may increase susceptibility to human immunodeficiency virus. The Journal of infectious diseases. 2000;182(2):467–473. doi: 10.1086/315713. [DOI] [PubMed] [Google Scholar]
  • 16.Martin HL, Richardson BA, Nyange PM, et al. Vaginal lactobacilli, microbial flora, and risk of human immunodeficiency virus type 1 and sexually transmitted disease acquisition. The Journal of infectious diseases. 1999;180(6):1863–1868. doi: 10.1086/315127. [DOI] [PubMed] [Google Scholar]
  • 17.Frank DN, Manigart O, Leroy V, et al. Altered vaginal microbiota are associated with perinatal mother-to-child transmission of HIV in African women from Burkina Faso. Journal of acquired immune deficiency syndromes. 2012;60(3):299–306. doi: 10.1097/QAI.0b013e31824e4bdb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nature medicine. 2006;12(12):1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 19.Hunt PW. HIV and inflammation: mechanisms and consequences. Current HIV/AIDS reports. 2012;9(2):139–147. doi: 10.1007/s11904-012-0118-8. [DOI] [PubMed] [Google Scholar]
  • 20.Guaraldi G, Orlando G, Zona S, et al. Premature age-related comorbidities among HIV-infected persons compared with the general population. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2011;53(11):1120–1126. doi: 10.1093/cid/cir627. [DOI] [PubMed] [Google Scholar]
  • 21.Marchetti G, Tincati C, Silvestri G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clinical microbiology reviews. 2013;26(1):2–18. doi: 10.1128/CMR.00050-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hazenberg MD, Stuart JW, Otto SA, et al. T-cell division in human immunodeficiency virus (HIV)-1 infection is mainly due to immune activation: a longitudinal analysis in patients before and during highly active antiretroviral therapy (HAART) Blood. 2000;95(1):249–255. [PubMed] [Google Scholar]
  • 23.Neuhaus J, Jacobs DR, Jr., Baker JV, et al. Markers of inflammation, coagulation, and renal function are elevated in adults with HIV infection. The Journal of infectious diseases. 2010;201(12):1788–1795. doi: 10.1086/652749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Brenchley JM, Silvestri G, Douek DC. Nonprogressive and progressive primate immunodeficiency lentivirus infections. Immunity. 2010;32(6):737–742. doi: 10.1016/j.immuni.2010.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Jacquelin B, Mayau V, Targat B, et al. Nonpathogenic SIV infection of African green monkeys induces a strong but rapidly controlled type I IFN response. The Journal of clinical investigation. 2009;119(12):3544–3555. doi: 10.1172/JCI40093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Silvestri G, Sodora DL, Koup RA, et al. Nonpathogenic SIV infection of sooty mangabeys is characterized by limited bystander immunopathology despite chronic high-level viremia. Immunity. 2003;18(3):441–452. doi: 10.1016/s1074-7613(03)00060-8. [DOI] [PubMed] [Google Scholar]
  • 27.Lim SG, Condez A, Lee CA, et al. Loss of mucosal CD4 lymphocytes is an early feature of HIV infection. Clinical and experimental immunology. 1993;92(3):448–454. doi: 10.1111/j.1365-2249.1993.tb03419.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Aas JA, Barbuto SM, Alpagot T, et al. Subgingival plaque microbiota in HIV positive patients. Journal of clinical periodontology. 2007;34(3):189–195. doi: 10.1111/j.1600-051X.2006.01034.x. [DOI] [PubMed] [Google Scholar]
  • 29.Veazey RS, DeMaria M, Chalifoux LV, et al. Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection. Science. 1998;280(5362):427–431. doi: 10.1126/science.280.5362.427. [DOI] [PubMed] [Google Scholar]
  • 30.Brenchley JM, Schacker TW, Ruff LE, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. The Journal of experimental medicine. 2004;200(6):749–759. doi: 10.1084/jem.20040874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Blaschitz C, Raffatellu M. Th17 cytokines and the gut mucosal barrier. Journal of clinical immunology. 2010;30(2):196–203. doi: 10.1007/s10875-010-9368-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Raffatellu M, Santos RL, Verhoeven DE, et al. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut. Nature medicine. 2008;14(4):421–428. doi: 10.1038/nm1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Brenchley JM, Paiardini M, Knox KS, et al. Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood. 2008;112(7):2826–2835. doi: 10.1182/blood-2008-05-159301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Petrovas C, Casazza JP, Brenchley JM, et al. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. The Journal of experimental medicine. 2006;203(10):2281–2292. doi: 10.1084/jem.20061496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kristoff J, Haret-Richter G, Ma D, et al. Early microbial translocation blockade reduces SIV-mediated inflammation and viral replication. The Journal of clinical investigation. 2014;124(6):2802–2806. doi: 10.1172/JCI75090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rose WA, 2nd, McGowin CL, Spagnuolo RA, et al. Commensal bacteria modulate innate immune responses of vaginal epithelial cell multilayer cultures. PloS one. 2012;7(3):e32728. doi: 10.1371/journal.pone.0032728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cerf-Bensussan N, Gaboriau-Routhiau V. The immune system and the gut microbiota: friends or foes? Nature reviews Immunology. 2010;10(10):735–744. doi: 10.1038/nri2850. [DOI] [PubMed] [Google Scholar]
  • 38.Galdeano CM, Perdigon G. The probiotic bacterium Lactobacillus casei induces activation of the gut mucosal immune system through innate immunity. Clinical and vaccine immunology : CVI. 2006;13(2):219–226. doi: 10.1128/CVI.13.2.219-226.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–498. doi: 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kau AL, Ahern PP, Griffin NW, et al. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474(7351):327–336. doi: 10.1038/nature10213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Saxena D, Li Y, Yang L, et al. Human microbiome and HIV/AIDS. Current HIV/AIDS reports. 2012;9(1):44–51. doi: 10.1007/s11904-011-0103-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Gori A, Tincati C, Rizzardini G, et al. Early impairment of gut function and gut flora supporting a role for alteration of gastrointestinal mucosa in human immunodeficiency virus pathogenesis. Journal of clinical microbiology. 2008;46(2):757–758. doi: 10.1128/JCM.01729-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Saavedra JM, Bauman NA, Oung I, et al. Feeding of Bifidobacterium bifidum and Streptococcus thermophilus to infants in hospital for prevention of diarrhoea and shedding of rotavirus. Lancet. 1994;344(8929):1046–1049. doi: 10.1016/s0140-6736(94)91708-6. [DOI] [PubMed] [Google Scholar]
  • 44.Hummelen R, Changalucha J, Butamanya NL, et al. Effect of 25 weeks probiotic supplementation on immune function of HIV patients. Gut microbes. 2011;2(2):80–85. doi: 10.4161/gmic.2.2.15787. [DOI] [PubMed] [Google Scholar]
  • 45.Pyles RB, Vincent KL, Baum MM, et al. Cultivated vaginal microbiomes alter HIV-1 infection and antiretroviral efficacy in colonized epithelial multilayer cultures. PloS one. 2014;9(3):e93419. doi: 10.1371/journal.pone.0093419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–450. doi: 10.1038/nature12721. [DOI] [PubMed] [Google Scholar]
  • 47.Vujkovic-Cvijin I, Dunham RM, Iwai S, et al. Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism. Science translational medicine. 2013;5(193):193ra191. doi: 10.1126/scitranslmed.3006438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Dang AT, Cotton S, Sankaran-Walters S, et al. Evidence of an increased pathogenic footprint in the lingual microbiome of untreated HIV infected patients. BMC microbiology. 2012;12:153. doi: 10.1186/1471-2180-12-153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ryder MI. Periodontal management of HIV-infected patients. Periodontology. 2000;23:85–93. doi: 10.1034/j.1600-0757.2000.2230108.x. 2000. [DOI] [PubMed] [Google Scholar]
  • 50.Pereira VT, Pavan P, Souza RC, et al. The association between detectable plasmatic human immunodeficiency virus (HIV) viral load and different subgingival microorganisms in Brazilian adults with HIV: a multilevel analysis. Journal of periodontology. 2014;85(5):697–705. doi: 10.1902/jop.2013.130273. [DOI] [PubMed] [Google Scholar]
  • 51.Silva-Boghossian C, Castro GF, Teles RP, et al. Salivary microbiota of HIV-positive children and its correlation with HIV status, oral diseases, and total secretory IgA. International journal of paediatric dentistry / the British Paedodontic Society [and] the International Association of Dentistry for Children. 2008;18(3):205–216. doi: 10.1111/j.1365-263X.2007.00864.x. [DOI] [PubMed] [Google Scholar]
  • 52.Ocon S, Murphy C, Dang AT, et al. Transcription profiling reveals potential mechanisms of dysbiosis in the oral microbiome of rhesus macaques with chronic untreated SIV infection. PloS one. 2013;8(11):e80863. doi: 10.1371/journal.pone.0080863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mataftsi M, Skoura L, Sakellari D. HIV infection and periodontal diseases: an overview of the post-HAART era. Oral diseases. 2011;17(1):13–25. doi: 10.1111/j.1601-0825.2010.01727.x. [DOI] [PubMed] [Google Scholar]
  • 54.Imai K, Ochiai K, Okamoto T. Reactivation of latent HIV-1 infection by the periodontopathic bacterium Porphyromonas gingivalis involves histone modification. Journal of immunology. 2009;182(6):3688–3695. doi: 10.4049/jimmunol.0802906. [DOI] [PubMed] [Google Scholar]
  • 55.Greenspan D, Gange SJ, Phelan JA, et al. Incidence of oral lesions in HIV-1-infected women: reduction with HAART. Journal of dental research. 2004;83(2):145–150. doi: 10.1177/154405910408300212. [DOI] [PubMed] [Google Scholar]
  • 56.George MD, Verhoeven D, Sankaran S, et al. Heightened cytotoxic responses and impaired biogenesis contribute to early pathogenesis in the oral mucosa of simian immunodeficiency virus-infected rhesus macaques. Clinical and vaccine immunology : CVI. 2009;16(2):277–281. doi: 10.1128/CVI.00265-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Aas JA, Paster BJ, Stokes LN, et al. Defining the normal bacterial flora of the oral cavity. Journal of clinical microbiology. 2005;43(11):5721–5732. doi: 10.1128/JCM.43.11.5721-5732.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kotler DP, Gaetz HP, Lange M, et al. Enteropathy associated with the acquired immunodeficiency syndrome. Annals of internal medicine. 1984;101(4):421–428. doi: 10.7326/0003-4819-101-4-421. [DOI] [PubMed] [Google Scholar]
  • 59.Moeller AH, Shilts M, Li Y, et al. SIV-induced instability of the chimpanzee gut microbiome. Cell host & microbe. 2013;14(3):340–345. doi: 10.1016/j.chom.2013.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mutlu EA, Keshavarzian A, Losurdo J, et al. A compositional look at the human gastrointestinal microbiome and immune activation parameters in HIV infected subjects. PLoS pathogens. 2014;10(2):e1003829. doi: 10.1371/journal.ppat.1003829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Merlini E, Bai F, Bellistri GM, et al. Evidence for polymicrobic flora translocating in peripheral blood of HIV-infected patients with poor immune response to antiretroviral therapy. PloS one. 2011;6(4):e18580. doi: 10.1371/journal.pone.0018580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ahmed N, Hayashi T, Hasegawa A, et al. Suppression of human immunodeficiency virus type 1 replication in macrophages by commensal bacteria preferentially stimulating Toll-like receptor 4. The Journal of general virology. 2010;91(Pt 11):2804–2813. doi: 10.1099/vir.0.022442-0. [DOI] [PubMed] [Google Scholar]
  • 63.Perez-Santiago J, Gianella S, Massanella M, et al. Gut Lactobacillales are associated with higher CD4 and less microbial translocation during HIV infection. Aids. 2013;27(12):1921–1931. doi: 10.1097/qad.0b013e3283611816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Dandekar S, George MD, Baumler AJ. Th17 cells, HIV and the gut mucosal barrier. Current opinion in HIV and AIDS. 2010;5(2):173–178. doi: 10.1097/COH.0b013e328335eda3. [DOI] [PubMed] [Google Scholar]
  • 65.Smith PM, Garrett WS. The gut microbiota and mucosal T cells. Frontiers in microbiology. 2011;2:111. doi: 10.3389/fmicb.2011.00111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kim M, Ashida H, Ogawa M, et al. Bacterial interactions with the host epithelium. Cell host & microbe. 2010;8(1):20–35. doi: 10.1016/j.chom.2010.06.006. [DOI] [PubMed] [Google Scholar]
  • 67.Yu G, Fadrosh D, Ma B, et al. Anal microbiota profiles in HIV-positive and HIV-negative MSM. Aids. 2013 doi: 10.1097/QAD.0000000000000154. [DOI] [PubMed] [Google Scholar]
  • 68.McCoy AN, Araujo-Perez F, Azcarate-Peril A, et al. Fusobacterium is associated with colorectal adenomas. PloS one. 2013;8(1):e53653. doi: 10.1371/journal.pone.0053653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ohkusa T, Okayasu I, Ogihara T, et al. Induction of experimental ulcerative colitis by Fusobacterium varium isolated from colonic mucosa of patients with ulcerative colitis. Gut. 2003;52(1):79–83. doi: 10.1136/gut.52.1.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shirakawa K, Chavez L, Hakre S, et al. Reactivation of latent HIV by histone deacetylase inhibitors. Trends in microbiology. 2013;21(6):277–285. doi: 10.1016/j.tim.2013.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Victoriano AF, Imai K, Okamoto T. Interaction between endogenous bacterial flora and latent HIV infection. Clinical and vaccine immunology : CVI. 2013;20(6):773–779. doi: 10.1128/CVI.00766-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hummelen R, Fernandes AD, Macklaim JM, et al. Deep sequencing of the vaginal microbiota of women with HIV. PloS one. 2010;5(8):e12078. doi: 10.1371/journal.pone.0012078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Spear GT, Gilbert D, Sikaroodi M, et al. Identification of rhesus macaque genital microbiota by 16S pyrosequencing shows similarities to human bacterial vaginosis: implications for use as an animal model for HIV vaginal infection. AIDS research and human retroviruses. 2010;26(2):193–200. doi: 10.1089/aid.2009.0166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Spear GT, Sikaroodi M, Zariffard MR, et al. Comparison of the diversity of the vaginal microbiota in HIV-infected and HIV-uninfected women with or without bacterial vaginosis. The Journal of infectious diseases. 2008;198(8):1131–1140. doi: 10.1086/591942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Schellenberg JJ, Plummer FA. The Microbiological Context of HIV Resistance: Vaginal Microbiota and Mucosal Inflammation at the Viral Point of Entry. International journal of inflammation. 2012;2012:131243. doi: 10.1155/2012/131243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gajer P, Brotman RM, Bai G, et al. Temporal dynamics of the human vaginal microbiota. Science translational medicine. 2012;4(132):132ra152. doi: 10.1126/scitranslmed.3003605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ravel J, Gajer P, Abdo Z, et al. Vaginal microbiome of reproductive-age women. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(Suppl 1):4680–4687. doi: 10.1073/pnas.1002611107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Klebanoff SJ, Coombs RW. Viricidal effect of Lactobacillus acidophilus on human immunodeficiency virus type 1: possible role in heterosexual transmission. The Journal of experimental medicine. 1991;174(1):289–292. doi: 10.1084/jem.174.1.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bebell LM, Passmore JA, Williamson C, et al. Relationship between levels of inflammatory cytokines in the genital tract and CD4+ cell counts in women with acute HIV-1 infection. The Journal of infectious diseases. 2008;198(5):710–714. doi: 10.1086/590503. [DOI] [PubMed] [Google Scholar]
  • 80.Beigi RH, Yudin MH, Cosentino L, et al. Cytokines, pregnancy, and bacterial vaginosis: comparison of levels of cervical cytokines in pregnant and nonpregnant women with bacterial vaginosis. The Journal of infectious diseases. 2007;196(9):1355–1360. doi: 10.1086/521628. [DOI] [PubMed] [Google Scholar]
  • 81.Mitchell C, Hitti J, Paul K, et al. Cervicovaginal shedding of HIV type 1 is related to genital tract inflammation independent of changes in vaginal microbiota. AIDS research and human retroviruses. 2011;27(1):35–39. doi: 10.1089/aid.2010.0129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Auvert B, Taljaard D, Lagarde E, et al. Randomized, controlled intervention trial of male circumcision for reduction of HIV infection risk: the ANRS 1265 Trial. PLoS medicine. 2005;2(11):e298. doi: 10.1371/journal.pmed.0020298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Price LB, Liu CM, Johnson KE, et al. The effects of circumcision on the penis microbiome. PloS one. 2010;5(1):e8422. doi: 10.1371/journal.pone.0008422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Johnson KE, Sherman ME, Ssempiija V, et al. Foreskin inflammation is associated with HIV and herpes simplex virus type-2 infections in Rakai, Uganda. Aids. 2009;23(14):1807–1815. doi: 10.1097/QAD.0b013e32832efdf1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Liu CM, Hungate BA, Tobian AA, et al. Male circumcision significantly reduces prevalence and load of genital anaerobic bacteria. mBio. 2013;4(2):e00076. doi: 10.1128/mBio.00076-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Schneider JA, Vadivelu S, Liao C, et al. Increased Likelihood of Bacterial Pathogens in the Coronal Sulcus and Urethra of Uncircumcised Men in a Diverse Group of HIV Infected and Uninfected Patients in India. Journal of global infectious diseases. 2012;4(1):6–9. doi: 10.4103/0974-777X.93750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Corey L, Wald A, Celum CL, et al. The effects of herpes simplex virus-2 on HIV-1 acquisition and transmission: a review of two overlapping epidemics. Journal of acquired immune deficiency syndromes. 2004;35(5):435–445. doi: 10.1097/00126334-200404150-00001. [DOI] [PubMed] [Google Scholar]
  • 88.Weiss HA, Thomas SL, Munabi SK, et al. Male circumcision and risk of syphilis, chancroid, and genital herpes: a systematic review and meta-analysis. Sexually transmitted infections. 2006;82(2):101–109. doi: 10.1136/sti.2005.017442. discussion 110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Segal LN, Methe BA, Nolan A, et al. HIV-1 and bacterial pneumonia in the era of antiretroviral therapy. Proceedings of the American Thoracic Society. 2011;8(3):282–287. doi: 10.1513/pats.201006-044WR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Iwai S, Fei M, Huang D, et al. Oral and airway microbiota in HIV-infected pneumonia patients. Journal of clinical microbiology. 2012;50(9):2995–3002. doi: 10.1128/JCM.00278-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Iwai S, Huang D, Fong S, et al. The lung microbiome of Ugandan HIV-infected pneumonia patients is compositionally and functionally distinct from that of San Franciscan patients. PloS one. 2014;9(4):e95726. doi: 10.1371/journal.pone.0095726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cribbs SK, Park Y, Guidot DM, et al. Metabolomics of Bronchoalveolar Lavage Differentiate Healthy HIV-1-Infected Subjects from Controls. AIDS research and human retroviruses. 2014;30(6):579–585. doi: 10.1089/aid.2013.0198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Beck JM, Young VB, Huffnagle GB. The microbiome of the lung. Translational research : the journal of laboratory and clinical medicine. 2012;160(4):258–266. doi: 10.1016/j.trsl.2012.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES