1
|
Srinivasan S, Richardson BA, Wallis JM, Fiedler TL, Strenk SM, Hoffman NG, Proll S, Chirenje ZM, Livant EW, Fredricks DN, Hillier SL, Marrazzo JM. Vaginal Bacteria and Proinflammatory Host Immune Mediators as Biomarkers of Human Immunodeficiency Virus Acquisition Risk Among African Women. J Infect Dis 2024; 230:1444-1455. [PMID: 39248500 DOI: 10.1093/infdis/jiae406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Few investigations have assessed contributions of both vaginal bacteria and proinflammatory immune mediators to human immunodeficiency virus (HIV) acquisition risk in a prospective cohort. METHODS We conducted a nested case-control study of African women who participated in a randomized placebo-controlled trial of daily oral versus vaginal tenofovir-based preexposure prophylaxis for HIV infection. Vaginal concentrations of 23 bacterial taxa and 16 immune mediators were measured. Relationships between individual bacterial concentrations or immune mediators and HIV risk were analyzed using generalized estimating equations in a multivariable model. Factor analysis assessed relationships between combinations of bacterial taxa, immune mediators, and HIV acquisition risk. RESULTS We identified 177 HIV pre-seroconversion visits from 150 women who acquired HIV and 531 visits from 436 women who remained HIV uninfected. Fourteen bacterial taxa and 6 proinflammatory cytokines and chemokines were individually associated with greater HIV risk after adjusting for confounders. Women with all 14 taxa versus <14 taxa (adjusted odds ratio [aOR], 4.45 [95% confidence interval {CI}, 2.20-8.98]; P < .001) or all 6 immune mediators versus <6 mediators (aOR, 1.77 [95% CI, 1.24-2.52]; P < .001) had greater risk for HIV acquisition. Factor analysis demonstrated that a bacterial factor comprised of 14 high-risk bacterial taxa (aOR, 1.57 [95% CI, 1.27-1.93]; P < 0.001) and the interferon gamma-induced protein 10 (highest quartile: aOR, 3.19 [95% CI, 1.32-7.72]; P = 0.002) contributed to the highest HIV risk. CONCLUSIONS Bacterial and host biomarkers for predicting HIV acquisition risk identify women at greatest risk for HIV infection and can focus prevention efforts.
Collapse
Affiliation(s)
- Sujatha Srinivasan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Barbra A Richardson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Biostatistics
- Department of Global Health
| | - Jacqueline M Wallis
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Tina L Fiedler
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Susan M Strenk
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Noah G Hoffman
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Sean Proll
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Z Mike Chirenje
- Clinical Trial Research Center, University of Zimbabwe, Harare
| | | | - David N Fredricks
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle
| | - Sharon L Hillier
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pennsylvania
| | - Jeanne M Marrazzo
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
2
|
Joseph A, Anton L, Guan Y, Ferguson B, Mirro I, Meng N, France M, Ravel J, Elovitz MA. Extracellular vesicles from vaginal Gardnerella vaginalis and Mobiluncus mulieris contain distinct proteomic cargo and induce inflammatory pathways. NPJ Biofilms Microbiomes 2024; 10:28. [PMID: 38514622 PMCID: PMC10957959 DOI: 10.1038/s41522-024-00502-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
Colonization of the vaginal space with bacteria such as Gardnerella vaginalis and Mobiluncus mulieris is associated with increased risk for STIs, bacterial vaginosis, and preterm birth, while Lactobacillus crispatus is associated with optimal reproductive health. Although host-microbe interactions are hypothesized to contribute to reproductive health and disease, the bacterial mediators that are critical to this response remain unclear. Bacterial extracellular vesicles (bEVs) are proposed to participate in host-microbe communication by providing protection of bacterial cargo, delivery to intracellular targets, and ultimately induction of immune responses from the host. We evaluated the proteome of bEVs produced in vitro from G. vaginalis, M. mulieris, and L. crispatus, identifying specific proteins of immunologic interest. We found that bEVs from each bacterial species internalize within cervical and vaginal epithelial cells, and that epithelial and immune cells express a multi-cytokine response when exposed to bEVs from G. vaginalis and M. mulieris but not L. crispatus. Further, we demonstrate that the inflammatory response induced by G. vaginalis and M. mulieris bEVs is TLR2-specific. Our results provide evidence that vaginal bacteria communicate with host cells through secreted bEVs, revealing a mechanism by which bacteria lead to adverse reproductive outcomes associated with inflammation. Elucidating host-microbe interactions in the cervicovaginal space will provide further insight into the mechanisms contributing to microbiome-mediated adverse outcomes and may reveal new therapeutic targets.
Collapse
Affiliation(s)
- Andrea Joseph
- Women's Biomedical Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.
- Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.
| | - Lauren Anton
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yuxia Guan
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Briana Ferguson
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Isabella Mirro
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nova Meng
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael France
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Michal A Elovitz
- Women's Biomedical Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| |
Collapse
|
3
|
Qulu WP, Mzobe G, Mtshali A, Letsoalo MP, Osman F, San JE, Kama AO, Garrett N, Mindel A, Rompalo A, Liebenberg LJP, Archary D, Sivro A, Ngcapu S. Metronidazole Treatment Failure and Persistent BV Lead to Increased Frequencies of Activated T- and Dendritic-Cell Subsets. Microorganisms 2023; 11:2643. [PMID: 38004655 PMCID: PMC10673474 DOI: 10.3390/microorganisms11112643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Metronidazole (MDZ) treatment failure and bacterial vaginosis (BV) recurrence rates are high among African women. This cohort study identified genital immune parameters associated with treatment response by comparing vaginal microbiota and immune cell frequencies in endocervical cytobrushes obtained from 32 South African women with symptomatic BV pre- and post-metronidazole treatment. Cervical T- and dendritic-cell subsets were phenotyped using multiparameter flow cytometry and the composition of vaginal microbial communities was characterized using 16S rRNA gene sequencing. MDZ treatment led to a modest decrease in the relative abundance of BV-associated bacteria, but colonization with Lactobacillus species (other than L. iners) was rare. At 6 and 12 weeks, MDZ-treated women had a significant increase in the frequencies of CCR5+ CD4+ T cells and plasmacytoid dendritic cells compared to the pre-treatment timepoint. In addition, MDZ non-responders had significantly higher frequencies of activated CD4 T cells and monocytes compared to MDZ responders. We conclude that MDZ treatment failure was characterized by an increased expression of activated T- and dendritic-cell subsets that may enhance HIV susceptibility. These data suggest the need to further assess the long-term impact of MDZ treatment on mucosal immune response and the vaginal microbiota.
Collapse
Affiliation(s)
- Wenkosi Perez Qulu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Gugulethu Mzobe
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Andile Mtshali
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Marothi Peter Letsoalo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
| | - Farzana Osman
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
| | - James Emmanuel San
- KwaZulu-Natal Research Innovation and Sequencing Platform, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Asavela Olona Kama
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Discipline of Public Health Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Adrian Mindel
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
| | - Anne Rompalo
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Lenine J. P. Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Centre for Epidemic Response and Innovation (CERI), Stellenbosch 7600, South Africa
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 3L5, Canada
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa; (W.P.Q.); (G.M.); (A.M.); (M.P.L.); (F.O.); (A.O.K.); (N.G.); (A.M.); (L.J.P.L.); (D.A.); (A.S.)
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
| |
Collapse
|
4
|
Lee CY, Diegel J, France MT, Ravel J, Arnold KB. Evaluation of vaginal microbiome equilibrium states identifies microbial parameters linked to resilience after menses and antibiotic therapy. PLoS Comput Biol 2023; 19:e1011295. [PMID: 37566641 PMCID: PMC10446192 DOI: 10.1371/journal.pcbi.1011295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/23/2023] [Accepted: 06/23/2023] [Indexed: 08/13/2023] Open
Abstract
The vaginal microbiome (VMB) is a complex microbial community that is closely tied to reproductive health. Optimal VMB communities have compositions that are commonly defined by the dominance of certain Lactobacillus spp. and can remain stable over time or transition to non-optimal states dominated by anaerobic bacteria and associated with bacterial vaginosis (BV). The ability to remain stable or undergo transitions suggests a system with either single (mono-stable) or multiple (multi-stable) equilibrium states, though factors that contribute to stability have been difficult to determine due to heterogeneity in microbial growth characteristics and inter-species interactions. Here, we use a computational model to determine whether differences in microbial growth and interaction parameters could alter equilibrium state accessibility and account for variability in community composition after menses and antibiotic therapies. Using a global uncertainty and sensitivity analysis that captures parameter sets sampled from a physiologically relevant range, model simulations predicted that 79.7% of microbial communities were mono-stable (gravitate to one composition type) and 20.3% were predicted to be multi-stable (can gravitate to more than one composition type, given external perturbations), which was not significantly different from observations in two clinical cohorts (HMP cohort, 75.2% and 24.8%; Gajer cohort, 78.1% and 21.9%, respectively). The model identified key microbial parameters that governed equilibrium state accessibility, such as the importance of non-optimal anaerobic bacteria interactions with Lactobacillus spp., which is largely understudied. Model predictions for composition changes after menses and antibiotics were not significantly different from those observed in clinical cohorts. Lastly, simulations were performed to illustrate how this quantitative framework can be used to gain insight into the development of new combinatorial therapies involving altered prebiotic and antibiotic dosing strategies. Altogether, dynamical models could guide development of more precise therapeutic strategies to manage BV.
Collapse
Affiliation(s)
- Christina Y. Lee
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jenna Diegel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michael T. France
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jacques Ravel
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
5
|
Thurman AR, Moench TR, Hoke M, Politch JA, Cabral H, Mausser E, Nador E, Morton J, Hamorsky K, Swope K, Bratcher B, Anderson DJ, Whaley KJ. ZB-06, a vaginal film containing an engineered human contraceptive antibody (HC4-N), demonstrates safety and efficacy in a phase 1 postcoital test and safety study. Am J Obstet Gynecol 2023; 228:716.e1-716.e12. [PMID: 36870409 PMCID: PMC10247457 DOI: 10.1016/j.ajog.2023.02.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/31/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND With an unplanned pregnancy rate of 50% or more in many countries, there is an urgent need for contraceptives that are more accessible and acceptable. To meet the growing demand for new contraceptives, ZabBio developed ZB-06, a vaginal film containing HC4-N, a human contraceptive antibody that inactivates sperm. OBJECTIVE This study aimed to assess the potential contraceptive activity of the ZB-06 film using a surrogate assessment for contraceptive efficacy, the postcoital test. We also assessed clinical safety of film use among healthy heterosexual couples. Serum, cervical mucus, and vaginal fluid HC4-N antibody concentrations and sperm agglutination potency were determined after single film use. Changes in the concentration of soluble proinflammatory cytokines and vaginal Nugent score after film use were measured as subclinical safety endpoints. STUDY DESIGN This was a phase 1, first-in-woman, open-label, proof-of-concept, postcoital test and safety study. RESULTS A total of 20 healthy women were enrolled in the study, and 8 heterosexual couples completed all study visits. The product was safe for both female participants and their male sexual partners. The postcoital test performed on ovulatory cervical mucus at baseline (no product use) revealed a mean of 25.9 (±30.6) progressively motile sperm per high-power field. After use of a single ZB-06 film before intercourse, this number dropped to 0.04 (±0.06) progressively motile sperm per high-power field (P<.0001). At the follow-up postcoital test visit approximately 1 month later (no product use), a mean of 47.4 (±37.4) progressively motile sperm per high-power field was observed, indicating contraceptive reversibility. CONCLUSION A single dose of the ZB-06 film applied before intercourse was safe and met efficacy surrogate benchmarks of excluding progressively motile sperm from ovulatory cervical mucus. These data indicate that ZB-06 is a viable contraceptive candidate warranting further development and testing.
Collapse
Affiliation(s)
- Andrea R Thurman
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA.
| | | | | | - Joseph A Politch
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Howard Cabral
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Emilie Mausser
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Ellena Nador
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | | | | | | | | | - Deborah J Anderson
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Kevin J Whaley
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| |
Collapse
|
6
|
Wang S, Wu J, Ran D, Ou G, Chen Y, Xu H, Deng L, Chen X. Study of the Relationship between Mucosal Immunity and Commensal Microbiota: A Bibliometric Analysis. Nutrients 2023; 15:nu15102398. [PMID: 37242281 DOI: 10.3390/nu15102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
This study presents the first bibliometric evaluation and systematic analysis of publications related to mucosal immunity and commensal microbiota over the last two decades and summarizes the contribution of countries, institutions, and scholars in the study of this field. A total of 1423 articles related to mucosal immunity and commensal microbiota in vivo published in 532 journals by 7774 authors from 1771 institutions in 74 countries/regions were analyzed. The interaction between commensal microbiota in vivo and mucosal immunity is essential in regulating the immune response of the body, maintaining communication between different kinds of commensal microbiota and the host, and so on. Several hot spots in this field have been found to have received extensive attention in recent years, especially the effects of metabolites of key strains on mucosal immunity, the physiopathological phenomena of commensal microbiota in various sites including the intestine, and the relationship between COVID-19, mucosal immunity and microbiota. We hope that the full picture of the last 20 years in this research area provided in this study will serve to deliver necessary cutting-edge information to relevant researchers.
Collapse
Affiliation(s)
- Shiqi Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jialin Wu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Duo Ran
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Guosen Ou
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yaokang Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Li Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
7
|
Shvartsman E, Hill JE, Sandstrom P, MacDonald KS. Gardnerella Revisited: Species Heterogeneity, Virulence Factors, Mucosal Immune Responses, and Contributions to Bacterial Vaginosis. Infect Immun 2023; 91:e0039022. [PMID: 37071014 PMCID: PMC10187134 DOI: 10.1128/iai.00390-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
Gardnerella species are associated with bacterial vaginosis (BV) and have been investigated as etiological agents of the condition. Nonetheless, the isolation of this taxon from healthy individuals has raised important questions regarding its etiological role. Recently, using advanced molecular approaches, the Gardnerella genus was expanded to include several different species that exhibit differences in virulence potential. Understanding the significance of these different species with respect to mucosal immunity and the pathogenesis and complications of BV could be crucial to solving the BV enigma. Here, we review key findings regarding the unique genetic and phenotypic diversity within this genus, virulence factors, and effects on mucosal immunity as they stand. We also comment on the relevance of these findings to the proposed role of Gardnerella in BV pathogenesis and in reproductive health and identify key gaps in knowledge that should be explored in the future.
Collapse
Affiliation(s)
- Elinor Shvartsman
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, Manitoba, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Janet E. Hill
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Paul Sandstrom
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, Manitoba, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Kelly S. MacDonald
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, Manitoba, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
8
|
Ziogou A, Ziogos E, Giannakodimos I, Giannakodimos A, Sifakis S, Ioannou P, Tsiodras S. Bacterial Vaginosis and Post-Operative Pelvic Infections. Healthcare (Basel) 2023; 11:healthcare11091218. [PMID: 37174760 PMCID: PMC10178576 DOI: 10.3390/healthcare11091218] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Bacterial vaginosis (BV) represents a condition in which the normal protective Lactobacilli, especially those that produce H2O2, are replaced by high quantities of facultative anaerobes, leading to gynecologic and obstetric post-operative complications. BV is an important cause of obstetric and gynecological adverse sequelae and it could lead to an increased risk of contracting sexually transmitted infections such as gonorrhea, genital herpes, Chlamydia, Trichomonas, and human immunodeficiency virus. Herein, we reviewed bacterial vaginosis and its association with post-operative pelvic infections. In Obstetrics, BV has been associated with increased risk of preterm delivery, first-trimester miscarriage in women undergoing in vitro fertilization, preterm premature rupture of membranes, chorioamnionitis, amniotic fluid infections, postpartum and postabortal endomyometritis as well as postabortal pelvic inflammatory disease (PID). In gynecology, BV increases the risk of post-hysterectomy infections such as vaginal cuff cellulitis, pelvic cellulitis, pelvic abscess, and PID. BV is often asymptomatic, can resolve spontaneously, and often relapses with or without treatment. The American College of Obstetricians and Gynecologists recommends testing for BV in women having an increased risk for preterm delivery. Women with symptoms should be evaluated and treated. Women with BV undergoing gynecological surgeries must be treated to reduce the frequency of post-operative pelvic infections. Metronidazole and clindamycin are the mainstays of therapy. Currently, there is no consensus on pre-surgery screening for BV; decisions are made on a case-by-case basis.
Collapse
Affiliation(s)
- Afroditi Ziogou
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eleftherios Ziogos
- Department of Gynecology and Obstetrics, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Ilias Giannakodimos
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Alexios Giannakodimos
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Petros Ioannou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Sotirios Tsiodras
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Fourth Department of Internal Medicine, Attikon General Hospital, 12462 Athens, Greece
| |
Collapse
|
9
|
Influence of Hormonal Contraceptive Use and HIV on Cervicovaginal Cytokines and Microbiota in Malawi. mSphere 2023; 8:e0058522. [PMID: 36622252 PMCID: PMC9942570 DOI: 10.1128/msphere.00585-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Important questions remain on how hormonal contraceptives alter the local immune environment and the microbiota in the female genital tract and how such effects may impact susceptibility to HIV infection. We leveraged samples from a previously conducted clinical trial of Malawian women with (n = 73) and without (n = 24) HIV infection randomized to depot medroxyprogesterone acetate (DMPA) or the levonogestrel implant in equal numbers within each group and determined the effects of these hormonal contraceptives (HCs) on the vaginal immune milieu and the composition of the vaginal microbiota. Longitudinal data for soluble immune mediators, measured by multiplex bead arrays and enzyme-linked immunosorbent assays (ELISAs), and vaginal microbiota, assessed by 16S rRNA gene amplicon, were collected prior to and over a period of 180 days post-HC initiation. DMPA and levonogestrel had only minimal effects on the vaginal immune milieu and microbiota. In women with HIV, with the caveat of a small sample size, there was an association between the median log10 change in the interleukin-12 (IL-12)/IL-10 ratio in vaginal fluid at day 180 post-HC compared to baseline when these women were classified as having a community state type (CST) IV vaginal microbiota and were randomized to DMPA. Long-lasting alterations in soluble immune markers or shifts in microbiota composition were not observed. Furthermore, women with HIV did not exhibit increased viral shedding in the genital tract after HC initiation. Consistent with the results of the ECHO (Evidence for Contraceptive Options and HIV Outcomes) trial, our data imply that the progestin-based HC DMPA and levonorgestrel are associated with minimal risk for women with HIV. (This study has been registered at ClinicalTrials.gov under registration no. NCT02103660). IMPORTANCE The results of the Evidence for Contraceptive Options and HIV Outcomes (ECHO) trial, the first large randomized controlled clinical trial comparing the HIV acquisition risk of women receiving DMPA, the levonorgestrel (LNG) implant, or the copper intrauterine device (IUD), did not reveal an increased risk of HIV acquisition for women on any of these three contraceptives. Our study results confirm that the two different progestin-based hormonal contraceptives DMPA and levonogestrel will not increase the risk for HIV infection. Furthermore, DMPA and levonogestrel have only minimal effects on the immune milieu and the microbiota in the vaginal tract, attesting to the safety of these hormonal contraceptives.
Collapse
|
10
|
Shvartsman E, Perciani CT, Richmond MEI, Russell JNH, Tough RH, Vancuren SJ, Hill JE, KAVI-ICR, Jaoko W, McKinnon LR, Sandstrom PA, MacDonald KS. Gardnerella subgroup dominant microbiomes are associated with divergent cervicovaginal immune responses in a longitudinal cohort of Kenyan women. Front Immunol 2023; 13:974195. [PMID: 36726972 PMCID: PMC9886495 DOI: 10.3389/fimmu.2022.974195] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/09/2022] [Indexed: 01/19/2023] Open
Abstract
Most cervicovaginal microbiome-immunology studies to date have relied on 16S rDNA microbial profiling which does not resolve the molecular subgroups of Gardnerella, believed to be central to the pathogenesis of bacterial vaginosis (BV) and subsequent risk of HIV acquisition. Here we used the cpn60 universal target which in addition to other microbial taxa, resolves four Gardnerella subgroups, for cervicovaginal microbial profiling in a longitudinal cohort of Kenyan women to examine associations with cellular and soluble markers of inflammation and HIV susceptibility. Participants (N = 41) were sampled, contributing 362 samples for microbiome analysis. All non-Lactobacillus dominant microbial communities were associated with high pro-inflammatory cytokine levels. Divergent associations were observed among different Gardnerella subgroup dominated communities with respect to the chemokine IP-10. Specifically, Gardnerella subgroup A dominant and polymicrobial communities were associated with reduced concentrations of IP-10 in adjusted linear mixed models (p<0.0001), compared to microbial communities dominated by Lactobacillus (non-iners) species. However, these associations did not translate to significant differences in the proportion or absolute number of CCR5, HLA-DR and CD38 expressed on cervical CD4+ T- cells. These findings suggest that some associations between Gardnerella subgroup dominant microbiomes and mucosal immunity differ and are relevant for the study of BV-pathogenesis and understanding the mechanisms of BV-associated HIV risk.
Collapse
Affiliation(s)
- Elinor Shvartsman
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB, Canada,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada,Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Catia T. Perciani
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Meika E. I. Richmond
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB, Canada,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada
| | - Justen N. H. Russell
- JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Riley H. Tough
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB, Canada,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada
| | - Sarah J. Vancuren
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Janet E. Hill
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - KAVI-ICR
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Walter Jaoko
- Kenyan AIDS Vaccine Initiative-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Lyle R. McKinnon
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB, Canada,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada,Centre for the AIDS Program of Research in South Africa (CAPRISA), Durban, South Africa
| | - Paul A. Sandstrom
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB, Canada,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada
| | - Kelly S. MacDonald
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB, Canada,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada,Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada,Department of Immunology, University of Toronto, Toronto, ON, Canada,*Correspondence: Kelly S. MacDonald,
| |
Collapse
|
11
|
Dong M, Dong Y, Bai J, Li H, Ma X, Li B, Wang C, Li H, Qi W, Wang Y, Fan A, Han C, Xue F. Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front Cell Infect Microbiol 2023; 13:1124591. [PMID: 36909729 PMCID: PMC9998931 DOI: 10.3389/fcimb.2023.1124591] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/31/2023] [Indexed: 02/26/2023] Open
Abstract
The female reproductive tract harbours hundreds of bacterial species and produces numerous metabolites. The uterine cervix is located between the upper and lower parts of the female genital tract. It allows sperm and birth passage and hinders the upward movement of microorganisms into a relatively sterile uterus. It is also the predicted site for sexually transmitted infection (STI), such as Chlamydia, human papilloma virus (HPV), and human immunodeficiency virus (HIV). The healthy cervicovaginal microbiota maintains cervical epithelial barrier integrity and modulates the mucosal immune system. Perturbations of the microbiota composition accompany changes in microbial metabolites that induce local inflammation, damage the cervical epithelial and immune barrier, and increase susceptibility to STI infection and relative disease progression. This review examined the intimate interactions between the cervicovaginal microbiota, relative metabolites, and the cervical epithelial-, immune-, and mucus barrier, and the potent effect of the host-microbiota interaction on specific STI infection. An improved understanding of cervicovaginal microbiota regulation on cervical microenvironment homeostasis might promote advances in diagnostic and therapeutic approaches for various STI diseases.
Collapse
Affiliation(s)
- Mengting Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yalan Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Junyi Bai
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotong Ma
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bijun Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huiyang Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenhui Qi
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Aiping Fan
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| | - Fengxia Xue
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| |
Collapse
|
12
|
Adapen C, Réot L, Menu E. Role of the human vaginal microbiota in the regulation of inflammation and sexually transmitted infection acquisition: Contribution of the non-human primate model to a better understanding? FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:992176. [PMID: 36560972 PMCID: PMC9763629 DOI: 10.3389/frph.2022.992176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
The human vaginal microbiota has a central role in the regulation of the female reproductive tract (FRT) inflammation. Indeed, on one hand an optimal environment leading to a protection against sexually transmitted infections (STI) is associated with a high proportion of Lactobacillus spp. (eubiosis). On the other hand, a more diverse microbiota with a high amount of non-Lactobacillus spp. (dysbiosis) is linked to a higher local inflammation and an increased STI susceptibility. The composition of the vaginal microbiota is influenced by numerous factors that may lead to a dysbiotic environment. In this review, we first discuss how the vaginal microbiota composition affects the local inflammation with a focus on the cytokine profiles, the immune cell recruitment/phenotype and a large part devoted on the interactions between the vaginal microbiota and the neutrophils. Secondly, we analyze the interplay between STI and the vaginal microbiota and describe several mechanisms of action of the vaginal microbiota. Finally, the input of the NHP model in research focusing on the FRT health including vaginal microbiota or STI acquisition/control and treatment is discussed.
Collapse
Affiliation(s)
- Cindy Adapen
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Louis Réot
- Université Paris-Saclay, Inserm, Commissariat à l'énergie Atomique et aux énergies Alternatives (CEA), Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB)/Department of Infectious Disease Models and Innovative Therapies (IDMIT), Fontenay-aux-Roses, France
| | - Elisabeth Menu
- Université Paris-Saclay, Inserm, Commissariat à l'énergie Atomique et aux énergies Alternatives (CEA), Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB)/Department of Infectious Disease Models and Innovative Therapies (IDMIT), Fontenay-aux-Roses, France
- Mucosal Immunity and Sexually Transmitted Infection Control (MISTIC) Group, Department of Virology, Institut Pasteur, Paris, France
| |
Collapse
|
13
|
Haddad LB, Herring GB, Mehta CC, Staple T, Young MR, Govindaraj S, Velu V, Smith AK. Evaluating the impact of three progestin-based hormonal contraceptive methods on immunologic changes in the female genital tract and systemically (CHIME Study): a prospective cohort study protocol. BMC Womens Health 2022; 22:456. [PMID: 36401326 PMCID: PMC9673204 DOI: 10.1186/s12905-022-02053-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/05/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Gonadal hormones can modify immune function, which may impact susceptibility to infectious diseases, including Human Immunodeficiency Virus (HIV). There is limited knowledge about how hormonal contraceptives (HC) influence the immune response during the course of use. The CHIME study aims to evaluate the effect of long-acting progestin-based hormonal contraceptives (depot medroxyprogesterone acetate, etonogestrel implant, and levonorgestrel intrauterine device) on immunologic changes in the female genital tract (FGT) and systemic compartment. METHODS CHIME is an observational cohort study where participants attend 2 visits prior to initiating the HC method of their choice, and then attend 6 visits over 12 months with biological sampling (vaginal swabs, cervicovaginal lavage, cytobrush and blood) for immunological, bacteriological, and virological analyses at each visit. Immune profiling will be evaluated by multi-color flow cytometry to determine how different T-cell subsets, in particular the CD4 T-cell subsets, change during the course of contraceptive use and whether they have different profiles in the FGT compared to the systemic compartment. The study aims are (1) to characterize the alterations in FGT and systemic immune profiles associated with three long-acting progestin-only HC and (2) to evaluate the vaginal microenvironment, determined by 16 s rRNA sequencing, as an individual-level risk factor and moderator of genital and systemic immune profile changes following exposure to three commonly used HC. Data collection started in March 2019 and is scheduled to be completed in October 2024. DISCUSSION The CHIME study aims to contribute to the body of research designed to evaluate the comparative impact of three long-acting progestin-only HC on innate and adaptive immune functions to understand how immunologic effects alter STI and HIV susceptibility.
Collapse
Affiliation(s)
- Lisa B Haddad
- Center for Biomedical Research, Population Council, New York, NY, USA
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, 101 Woodruff Circle NE, GA, 30322, Atlanta, USA
| | - Gina Bailey Herring
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Grady Infectious Disease Program, Grady Health System, Atlanta, GA, USA
| | - C Christina Mehta
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 101 Woodruff Circle NE, Atlanta, GA, 30322, USA
| | - Tyree Staple
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, 101 Woodruff Circle NE, GA, 30322, Atlanta, USA
| | - Marisa R Young
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, 101 Woodruff Circle NE, GA, 30322, Atlanta, USA
| | - Sakthivel Govindaraj
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Vijayakumar Velu
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, 101 Woodruff Circle NE, GA, 30322, Atlanta, USA.
| |
Collapse
|
14
|
Hughes SM, Levy CN, Katz R, Lokken EM, Anahtar MN, Hall MB, Bradley F, Castle PE, Cortez V, Doncel GF, Fichorova R, Fidel PL, Fowke KR, Francis SC, Ghosh M, Hwang LY, Jais M, Jespers V, Joag V, Kaul R, Kyongo J, Lahey T, Li H, Makinde J, McKinnon LR, Moscicki AB, Novak RM, Patel MV, Sriprasert I, Thurman AR, Yegorov S, Mugo NR, Roxby AC, Micks E, Hladik F. Changes in concentrations of cervicovaginal immune mediators across the menstrual cycle: a systematic review and meta-analysis of individual patient data. BMC Med 2022; 20:353. [PMID: 36195867 PMCID: PMC9533580 DOI: 10.1186/s12916-022-02532-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 08/16/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hormonal changes during the menstrual cycle play a key role in shaping immunity in the cervicovaginal tract. Cervicovaginal fluid contains cytokines, chemokines, immunoglobulins, and other immune mediators. Many studies have shown that the concentrations of these immune mediators change throughout the menstrual cycle, but the studies have often shown inconsistent results. Our understanding of immunological correlates of the menstrual cycle remains limited and could be improved by meta-analysis of the available evidence. METHODS We performed a systematic review and meta-analysis of cervicovaginal immune mediator concentrations throughout the menstrual cycle using individual participant data. Study eligibility included strict definitions of the cycle phase (by progesterone or days since the last menstrual period) and no use of hormonal contraception or intrauterine devices. We performed random-effects meta-analyses using inverse-variance pooling to estimate concentration differences between the follicular and luteal phases. In addition, we performed a new laboratory study, measuring select immune mediators in cervicovaginal lavage samples. RESULTS We screened 1570 abstracts and identified 71 eligible studies. We analyzed data from 31 studies, encompassing 39,589 concentration measurements of 77 immune mediators made on 2112 samples from 871 participants. Meta-analyses were performed on 53 immune mediators. Antibodies, CC-type chemokines, MMPs, IL-6, IL-16, IL-1RA, G-CSF, GNLY, and ICAM1 were lower in the luteal phase than the follicular phase. Only IL-1α, HBD-2, and HBD-3 were elevated in the luteal phase. There was minimal change between the phases for CXCL8, 9, and 10, interferons, TNF, SLPI, elafin, lysozyme, lactoferrin, and interleukins 1β, 2, 10, 12, 13, and 17A. The GRADE strength of evidence was moderate to high for all immune mediators listed here. CONCLUSIONS Despite the variability of cervicovaginal immune mediator measurements, our meta-analyses show clear and consistent changes during the menstrual cycle. Many immune mediators were lower in the luteal phase, including chemokines, antibodies, matrix metalloproteinases, and several interleukins. Only interleukin-1α and beta-defensins were higher in the luteal phase. These cyclical differences may have consequences for immunity, susceptibility to infection, and fertility. Our study emphasizes the need to control for the effect of the menstrual cycle on immune mediators in future studies.
Collapse
Affiliation(s)
- Sean M Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Claire N Levy
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Erica M Lokken
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Melis N Anahtar
- Ragon Institute of MIT and Harvard, Massachusetts General Hospital, Boston, MA, USA
| | | | - Frideborg Bradley
- Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Philip E Castle
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Valerie Cortez
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - Raina Fichorova
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Paul L Fidel
- Louisiana State University Health, New Orleans, LA, USA
| | - Keith R Fowke
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Suzanna C Francis
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Mimi Ghosh
- Department of Epidemiology, The George Washington University, Washington, DC, USA
| | - Loris Y Hwang
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mariel Jais
- Office of Laboratory Safety, The George Washington University, Washington, DC, USA
| | | | - Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jordan Kyongo
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Timothy Lahey
- University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Huiying Li
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Julia Makinde
- IAVI Human Immunology Laboratory, Imperial College, London, England, UK
- IAVI, New York, NY, USA
| | - Lyle R McKinnon
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Anna-Barbara Moscicki
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Mickey V Patel
- Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Intira Sriprasert
- Department of OB/GYN, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Sergey Yegorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Nelly Rwamba Mugo
- Department of Global Health, University of Washington, Seattle, WA, USA
- Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Alison C Roxby
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | - Elizabeth Micks
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA.
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA.
| |
Collapse
|
15
|
Tyssen D, Hearps AC, Guntur K, Masson L, Cook S, Moulton SE, Ravel J, Bradshaw CS, Ayehunie S, Tachedjian G. The Impact of Over-The-Counter Lactic Acid Containing Vaginal Gels on the Integrity and Inflammatory State of the Vaginal Epithelium in vitro. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:915948. [PMID: 36303665 PMCID: PMC9580709 DOI: 10.3389/frph.2022.915948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/24/2022] [Indexed: 12/19/2024] Open
Abstract
The vaginal microbiome influences a wide range of health outcomes in women, where a microbiome dominated by Lactobacillus spp. is considered optimal and associated with reduced risk of pre-term birth and acquisition of sexually transmitted infections including HIV. Conversely, replacement of lactobacilli by non-optimal bacteria leads to the development of bacterial vaginosis, which is associated with increased risk of these outcomes. Lactobacilli produce the metabolite lactic acid (LA) which is a potent antibacterial and antiviral agent. The potential therapeutic benefits of LA have prompted the development of numerous over-the-counter LA-containing gels for use in the vagina, although a comprehensive analysis of the impact of these formulations on the cervicovaginal epithelium and pro-inflammatory cytokine/chemokine responses, has not been assessed. Here, we evaluated the properties of 11 over-the-counter gels, including 9 containing LA, marketed for use in the vagina. Ten of the 11 gels had an osmolality greater than vaginal fluid from women with Lactobacillus-dominated microbiota (370 ± 40 mOsmol/kg in women with Nugent score 0-3), with six gels that were hyperosmolal >2,000 mOsmol/kg. Using a reconstructed primary cell model of the vaginal epithelium, we found hyperosmolal gels had a detrimental impact on epithelial barrier integrity, resulting in substantial cellular toxicity (<10% viability as compared to untreated cells) and reduced epithelial barrier integrity [≈30% of untreated cells, assessed by transepithelial electrical resistance (TEER)]. Treatment of vaginal tissues with most of the gels elicited the production of pro-inflammatory factors including IL-1α (8 of 11) and IL-1β (10 of 11) which are associated with heightened risk of HIV acquisition in vivo. The majority of the OTC gels elicited moderate tissue damage as determined by histology. The detrimental effects of these gels on the human vaginal epithelium in vitro may predict compromised epithelial barrier integrity and genital inflammation in vivo, which has implications for sexual and reproductive health. This study highlights the importance of evaluating the impact of intravaginal products on the integrity and inflammatory status of the mucosal epithelium to avoid unfavorable off target effects.
Collapse
Affiliation(s)
- David Tyssen
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC, Australia
| | - Anna C. Hearps
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Lindi Masson
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Simon Cook
- Australian Research Council (ARC) Centre of Excellence for Electromaterials Science, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Simon E. Moulton
- Australian Research Council (ARC) Centre of Excellence for Electromaterials Science, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent's Hospital Melbourne, Melbourne, VIC, Australia
- Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, United States
| | - Catriona S. Bradshaw
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC, Australia
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Gilda Tachedjian
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Gustin AT, Thurman AR, Chandra N, Schifanella L, Alcaide M, Fichorova R, Doncel GF, Gale M, Klatt NR. Recurrent bacterial vaginosis following metronidazole treatment is associated with microbiota richness at diagnosis. Am J Obstet Gynecol 2022; 226:225.e1-225.e15. [PMID: 34560047 PMCID: PMC8887553 DOI: 10.1016/j.ajog.2021.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/13/2021] [Accepted: 09/12/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Bacterial vaginosis-a condition defined by a shift from Lactobacillus dominance to a polymicrobial, anaerobic bacterial community-increases the risk of acquiring sexually transmitted infections and other complications of the female reproductive tract. Antibiotic treatment frequently fails to return the microbiome to an optimal Lactobacillus-dominated state. No criteria currently exist to identify the patients likely to experience treatment failure. OBJECTIVE We sought to identify the pretreatment community signatures associated with treatment failure through 16S ribosomal RNA gene analysis. STUDY DESIGN Twenty-eight women who were enrolled in an oral metronidazole treatment trial of bacterial vaginosis were studied. Cervicovaginal lavage samples were collected before metronidazole treatment and at 7 and 30 days posttreatment. Cervicovaginal lavage DNA was amplified and sequenced using a paired-end, V4 region 2×150 MiSeq run. RESULTS Of the 28 women, 25% failed to clear bacterial vaginosis; 35.7% demonstrated a transient clearance, shifting to community-type 2 (Lactobacillus iners dominant) at visit 2 only; 7.1% demonstrated a delayed clearance, reaching community-type 2 at the final visit only; and 32.1% of patients experienced sustained bacterial vaginosis clearance. Examination of the community composition and structure demonstrated that both the richness and the evenness were significantly lower for the women who experienced sustained clearance, whereas the women who failed to clear bacterial vaginosis possessed the highest median levels of richness, evenness, and diversity pretreatment. Soluble immune factors in the lower reproductive tract improved significantly following a shift from community-type 4 to a Lactobacillus-dominant microbiome, with the samples categorized as community-type 2 possessing significantly higher levels of secretory leukocyte protease inhibitor, growth-regulated alpha protein, and macrophage inflammatory protein-3 and significantly lower levels of intercellular adhesion molecule-1. Although the shifts to Lactobacillus dominance improved the markers of mucosal tissue health, these gains were only temporary among the women who experienced recurrence. CONCLUSION Assemblies of highly diverse microbiota are associated with the enhanced resilience of bacterial vaginosis to standard metronidazole treatment. These communities may be foundational to treatment resistance or simply an indication of a well-established community made possible by canonical biofilm-forming taxa. Future studies must target the transcriptional activity of these communities under the pressure of antibiotic treatment to resolve the mechanisms of their resistance.
Collapse
Affiliation(s)
- Andrew T Gustin
- Department of Global Health, University of Washington, Seattle, WA; Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN; Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | | | | | - Luca Schifanella
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN
| | | | - Raina Fichorova
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA
| | | | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Nichole R Klatt
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN; Department of Pediatrics, University of Miami, Miami, FL.
| |
Collapse
|
17
|
Teleshova N, Keller MJ, Fernández Romero JA, Friedland BA, Creasy GW, Plagianos MG, Ray L, Barnable P, Kizima L, Rodriguez A, Cornejal N, Melo C, Cruz Rodriguez G, Mukhopadhyay S, Calenda G, Sinkar SU, Bonnaire T, Wesenberg A, Zhang S, Kleinbeck K, Palmer K, Alami M, O’Keefe BR, Gillevet P, Hur H, Liang Y, Santone G, Fichorova RN, Kalir T, Zydowsky TM. Results of a phase 1, randomized, placebo-controlled first-in-human trial of griffithsin formulated in a carrageenan vaginal gel. PLoS One 2022; 17:e0261775. [PMID: 35051209 PMCID: PMC8775213 DOI: 10.1371/journal.pone.0261775] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
HIV pre-exposure prophylaxis (PrEP) is dominated by clinical therapeutic antiretroviral (ARV) drugs. Griffithsin (GRFT) is a non-ARV lectin with potent anti-HIV activity. GRFT’s preclinical safety, lack of systemic absorption after vaginal administration in animal studies, and lack of cross-resistance with existing ARV drugs prompted its development for topical HIV PrEP. We investigated safety, pharmacokinetics (PK), pharmacodynamics (PD), and immunogenicity of PC-6500 (0.1% GRFT in a carrageenan (CG) gel) in healthy women after vaginal administration. This randomized, placebo-controlled, parallel group, double-blind first-in-human phase 1 study enrolled healthy, HIV-negative, non-pregnant women aged 24–45 years. In the open label period, all participants (n = 7) received single dose of PC-6500. In the randomized period, participants (n = 13) were instructed to self-administer 14 doses of PC-6500 or its matching CG placebo (PC-535) once daily for 14 days. The primary outcomes were safety and PK after single dose, and then after 14 days of dosing. Exploratory outcomes were GRFT concentrations in cervicovaginal fluids, PD, inflammatory mediators and gene expression in ectocervical biopsies. This trial is registered with ClinicalTrials.gov, number NCT02875119. No significant adverse events were recorded in clinical or laboratory results or histopathological evaluations in cervicovaginal mucosa, and no anti-drug (GRFT) antibodies were detected in serum. No cervicovaginal proinflammatory responses and no changes in the ectocervical transcriptome were evident. Decreased levels of proinflammatory chemokines (CXCL8, CCL5 and CCL20) were observed. GRFT was not detected in plasma. GRFT and GRFT/CG in cervicovaginal lavage samples inhibited HIV and HPV, respectively, in vitro in a dose-dependent fashion. These data suggest GRFT formulated in a CG gel is a safe and promising on-demand multipurpose prevention technology product that warrants further investigation.
Collapse
Affiliation(s)
- Natalia Teleshova
- Center for Biomedical Research, Population Council, New York, New York, United States of America
- * E-mail:
| | - Marla J. Keller
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - José A. Fernández Romero
- Center for Biomedical Research, Population Council, New York, New York, United States of America
- Science Department, Borough of Manhattan Community College, New York, New York, United States of America
| | - Barbara A. Friedland
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - George W. Creasy
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Marlena G. Plagianos
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Laurie Ray
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Patrick Barnable
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Larisa Kizima
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Aixa Rodriguez
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Nadjet Cornejal
- Science Department, Borough of Manhattan Community College, New York, New York, United States of America
| | - Claudia Melo
- Science Department, Borough of Manhattan Community College, New York, New York, United States of America
| | - Gearoff Cruz Rodriguez
- Science Department, Borough of Manhattan Community College, New York, New York, United States of America
| | - Sampurna Mukhopadhyay
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Giulia Calenda
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Shweta U. Sinkar
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Thierry Bonnaire
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Asa Wesenberg
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Shimin Zhang
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Kyle Kleinbeck
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Kenneth Palmer
- University of Louisville, Louisville, Kentucky, United States of America
| | - Mohcine Alami
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Barry R. O’Keefe
- Division of Cancer Treatment and Diagnosis, Molecular Targets Program, Center for Cancer Research and Natural Products Branch, Developmental Therapeutics Program, National Cancer Institute, Frederick, Maryland, United States of America
| | - Patrick Gillevet
- George Mason University, Manassas, Virginia, United States of America
| | - Hong Hur
- Rockefeller University, New York, New York, United States of America
| | - Yupu Liang
- Rockefeller University, New York, New York, United States of America
| | - Gabriela Santone
- Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Tamara Kalir
- Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Thomas M. Zydowsky
- Center for Biomedical Research, Population Council, New York, New York, United States of America
| |
Collapse
|
18
|
Hu M, Valicherla GR, Zhou T, Hillier SL, Rohan LC. Expression, Activity, and Regulation of Phosphorylating Enzymes in Tissues and Cells Relevant to HIV-1 Sexual Transmission. AIDS Res Hum Retroviruses 2022; 38:22-32. [PMID: 33567990 PMCID: PMC8785762 DOI: 10.1089/aid.2020.0250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Phosphorylating enzymes (PEs) are responsible for activating nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) such as tenofovir (TFV) and are critical for their conversion to obtain intracellular antiviral activity. However, there are limited data available regarding the expression of PEs and their activity in the female genital tract. This work compared the messenger RNA (mRNA) expression levels of PEs in human female genital tissue, immune cells, and animal models that are commonly used in human immunodeficiency virus (HIV) research. Furthermore, the effect of contraceptive hormones and proinflammatory cytokines on tenofovir diphosphate (TFV-DP) formation and efficacy in human vaginal, epithelial, and immune cells was also evaluated. We found that human vaginal and ectocervical tissues had similar mRNA expression for seven PEs tested. Polymerase chain reaction results revealed that creatine kinase brain (CKB), mitochondrial creatine kinase 1 (CKMT1), mitochondrial creatine kinase 2 (CKMT2), adenylate kinase AK3L1 (AK4), and nucleoside diphosphate kinase 1 (NME1) exhibited a 10- to 10,000-fold higher expression level in a vaginal epithelial cell line, VK2, compared with CD4+ T cells (p < .05). Medroxyprogesterone acetate (MPA)/progesterone (P4) and IL-1β/IL-8 treatment resulted in altered TFV-DP levels in VK2 and PM1 cells. MPA and P4 at concentrations above 0.1 μM, as well as IL-1β and IL-8 at concentrations above 10 ng/mL, significantly decreased HIV-1BaL inhibition in PM1 cells when 1 μM TFV was added. However, this observed effect of hormones and cytokines was abrogated when TFV concentration was raised to 1 mM. These in vitro results elucidate the role of PEs in TFV metabolism and provide information regarding differences in PE tissue expression for animal models commonly used in HIV testing. This information can be applied to better understand and interpret data obtained using these models.
Collapse
Affiliation(s)
- Minlu Hu
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Guru R. Valicherla
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tian Zhou
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sharon L. Hillier
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lisa C. Rohan
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Address correspondence to: Lisa C. Rohan, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
19
|
Liu CW, Su BC, Chen JY. Tilapia Piscidin 4 (TP4) Reprograms M1 Macrophages to M2 Phenotypes in Cell Models of Gardnerella vaginalis-Induced Vaginosis. Front Immunol 2021; 12:773013. [PMID: 34925343 PMCID: PMC8674419 DOI: 10.3389/fimmu.2021.773013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/15/2021] [Indexed: 01/24/2023] Open
Abstract
Gardnerella vaginalis is associated with bacterial vaginosis (BV). The virulence factors produced by G. vaginalis are known to stimulate vaginal mucosal immune response, which is largely driven by activated macrophages. While Tilapia piscidin 4 (TP4), an antimicrobial peptide isolated from Nile tilapia, is known to display a broad range of antibacterial functions, it is unclear whether TP4 can affect macrophage polarization in the context of BV. In this study, we used the culture supernatants from G. vaginalis to stimulate differentiation of THP-1 and RAW264.7 cells to an M1 phenotype. The treatment activated the NF-κB/STAT1 signaling pathway, induced reactive nitrogen and oxygen species, and upregulated inflammatory mediators. We then treated the induced M1 macrophages directly with a non-toxic dose of TP4 or co-cultured the M1 macrophages with TP4-treated vaginal epithelial VK2 cells. The results showed that TP4 could not only decrease pro-inflammatory mediators in the M1 macrophages, but it also enriched markers of M2 macrophages. Further, we found that direct treatment with TP4 switched M1 macrophages toward a resolving M2c phenotype via the MAPK/ERK pathway and IL-10-STAT3 signaling. Conversely, tissue repair M2a macrophages were induced by TP4-treated VK2 cells; TP4 upregulated TSG-6 in VK2 cells, which subsequently activated STAT6 and M2a-related gene expression in the macrophages. In conclusion, our results imply that TP4 may be able to attenuate the virulence of G. vaginalis by inducing resolving M2c and tissue repair M2a macrophage polarizations, suggesting a novel strategy for BV therapy.
Collapse
Affiliation(s)
- Chia-Wen Liu
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Taiwan
| | - Bor-Chyuan Su
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Taiwan.,The iEGG and Animal Biotechnology Center, The Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
20
|
Gustin A, Cromarty R, Schifanella L, Klatt NR. Microbial mismanagement: how inadequate treatments for vaginal dysbiosis drive the HIV epidemic in women. Semin Immunol 2021; 51:101482. [PMID: 34120819 DOI: 10.1016/j.smim.2021.101482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022]
Abstract
Women and girls represent a key population driving new HIV infections and persistence of the HIV pandemic. A key determinant of HIV susceptibility is the composition of the vaginal microbiome, which can influence the local immune cell population, inflammation status, and HIV prevention drug levels. While a low-diversity composition dominated by Lactobacillus crispatus is associated with a decreased risk of HIV acquisition, high diversity environments associated with bacterial vaginosis increase risk of HIV. Given the important role of the vaginal microbiome in determining HIV susceptibility, altering the microbiome towards a Lactobacillus-dominated state is an attractive complementary strategy to reduce HIV incidence rates. Here, we provide an overview of the mechanisms by which the vaginal microbiome may contribute to HIV acquisition risk. Furthermore, we address the advantages and limitations of historical treatments and emerging technologies under investigation to modify the vaginal microbiome, including: antibiotics, bacteriophages, probiotics, topicals, and engineered bacteria. By addressing the current state of vaginal microbiome knowledge and strategies for manipulation, we hope to amplify the growing calls for increased resources and research into vaginal microbial health, which will be essential to accelerating preventative efforts amongst the world's most vulnerable populations.
Collapse
Affiliation(s)
- Andrew Gustin
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Ross Cromarty
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, University of Minnesota, Minneapolis, MN, USA; Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Luca Schifanella
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, University of Minnesota, Minneapolis, MN, USA
| | - Nichole R Klatt
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
21
|
Raglan O, MacIntyre DA, Mitra A, Lee YS, Smith A, Assi N, Nautiyal J, Purkayastha S, Gunter MJ, Gabra H, Marchesi JR, Bennett PR, Kyrgiou M. The association between obesity and weight loss after bariatric surgery on the vaginal microbiota. MICROBIOME 2021; 9:124. [PMID: 34049596 PMCID: PMC8164250 DOI: 10.1186/s40168-021-01011-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 02/02/2021] [Indexed: 05/08/2023]
Abstract
BACKGROUND Obesity and vaginal microbiome (VMB) dysbiosis are each risk factors for adverse reproductive and oncological health outcomes in women. Here, we investigated the relationship between obesity, vaginal bacterial composition, local inflammation and bariatric surgery. METHODS Vaginal bacterial composition assessed by high-throughput sequencing of bacterial 16S rRNA genes and local cytokine levels measured using a multiplexed Magnetic Luminex Screening Assay were compared between 67 obese and 42 non-obese women. We further assessed temporal changes in the microbiota and cytokines in a subset of 27 women who underwent bariatric surgery. RESULTS The bacterial component of the vaginal microbiota in obese women was characterised by a lower prevalence of a Lactobacillus-dominant VMB and higher prevalence of a high diversity (Lactobacillus spp., and Gardnerella- spp. depleted) VMB, compared with non-obese subjects (p<0.001). Obese women had higher relative abundance of Dialister species (p<0.001), Anaerococcus vaginalis (p=0.021), and Prevotella timonensis (p=0.020) and decreased relative abundance of Lactobacillus crispatus (p=0.014). Local vaginal IL-1β, IL-4, IL-6, IL-8, IFNγ, MIP-1α and TNFα levels were all higher among obese women, however, only IL-1β and IL-8 correlated with VMB species diversity. In a subset of obese women undergoing bariatric surgery, there were no significant overall differences in VMB following surgery; however, 75% of these women remained obese at 6 months. Prior to surgery, there was no relationship between body mass index (BMI) and VMB structure; however, post-surgery women with a Lactobacillus-dominant VMB had a significantly lower BMI than those with a high diversity VMB. CONCLUSIONS Obese women have a significantly different vaginal microbiota composition with increased levels of local inflammation compared to non-obese women. Bariatric surgery does not change the VMB; however, those with the greatest weight loss 6-month post-surgery are most likely to have a Lactobacillus-dominant VMB. Video abstract.
Collapse
Affiliation(s)
- Olivia Raglan
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
- Queen Charlotte’s and Chelsea-Hammersmith Hospital, Imperial College NHS Trust, W12 OHS London, UK
| | - David A. MacIntyre
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
- March of Dimes European Prematurity Research Centre, Imperial College London, London, W12 0NN UK
| | - Anita Mitra
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
- Queen Charlotte’s and Chelsea-Hammersmith Hospital, Imperial College NHS Trust, W12 OHS London, UK
| | - Yun S. Lee
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
- March of Dimes European Prematurity Research Centre, Imperial College London, London, W12 0NN UK
| | - Ann Smith
- Division of Population Medicine, Cardiff University, Heath Park, Cardiff, CF14 4YS UK
| | - Nada Assi
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Jaya Nautiyal
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
| | - Sanjay Purkayastha
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
- St Mary’s Hospital, Imperial College NHS Trust, W2 1NY London, UK
| | - Marc J. Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Hani Gabra
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
| | - Julian R. Marchesi
- March of Dimes European Prematurity Research Centre, Imperial College London, London, W12 0NN UK
- Division of Integrative Systems Medicine and Digestive Disease, St. Mary’s Hospital, Imperial College London, South Wharf Road, London, W2 1NY UK
| | - Phillip R. Bennett
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
- Queen Charlotte’s and Chelsea-Hammersmith Hospital, Imperial College NHS Trust, W12 OHS London, UK
- March of Dimes European Prematurity Research Centre, Imperial College London, London, W12 0NN UK
| | - Maria Kyrgiou
- IRDB, Department of Metabolism, Digestion and Reproduction - Surgery and Cancer, Hammersmith Campus, Imperial College London, W12 0NN London, UK
- Queen Charlotte’s and Chelsea-Hammersmith Hospital, Imperial College NHS Trust, W12 OHS London, UK
| |
Collapse
|
22
|
Sultan JS, Wang T, Hunte R, Srinivasan S, McWalters J, Tharp GK, Bosinger SE, Fiedler TL, Atrio JM, Murphy K, Barnett R, Ray LR, Krows ML, Fredricks DN, Irungu E, Ngure K, Mugo N, Marrazzo J, Keller MJ, Herold BC. Differences in vaginal microbiota, host transcriptome and proteins in women with bacterial vaginosis are associated with metronidazole treatment response. J Infect Dis 2021; 224:2094-2104. [PMID: 34003290 DOI: 10.1093/infdis/jiab266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/17/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Bacterial vaginosis (BV) treatment failures and recurrences are common. To identify features associated with treatment response, we compared vaginal microbiota and host ectocervical transcriptome before and after oral metronidazole therapy. METHODS Women with BV (Bronx, NY and Thika, Kenya) received 7 days of oral metronidazole at enrollment (Day 0) and underwent genital tract sampling of microbiome (16S rRNA gene sequencing), transcriptome (RNAseq), and immune mediator concentrations on Day 0, 15 and 35. RESULTS Bronx participants were more likely than Thika participants to clinically respond to metronidazole (19/20 vs 10/18, respectively, p=0.0067) and by changes in microbiota composition and diversity. After dichotomizing the cohort into responders and non-responders by change in alpha diversity between Day 35 and Day 0, we identified transcription differences associated with chemokine signaling (q=0.002) and immune system process (q=2.5e-8) that differentiated responders from non-responders were present at enrollment. Responders had significantly lower levels of CXCL9 in cervicovaginal lavage on Day 0 (p< 0.007) and concentrations of CXCL9, CXCL10 and MCP-1 increased significantly between Day 0 and Day 35 in responders versus non-responders. CONCLUSIONS Response to metronidazole is characterized by significant changes in chemokines and related transcripts suggesting that treatments that promote these pathways may prove beneficial.
Collapse
Affiliation(s)
- Joyce Serebrenik Sultan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Richard Hunte
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sujatha Srinivasan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jessica McWalters
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gregory K Tharp
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Steven E Bosinger
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, USA.,Emory University School of Medicine, Department of Pathology & Laboratory Medicine, Atlanta, GA, USA.,Emory Vaccine Center, Emory University, Atlanta,GA, USA
| | - Tina L Fiedler
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jessica M Atrio
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kerry Murphy
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rebecca Barnett
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laurie R Ray
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Meighan L Krows
- Department of Global Health, University of Washington, Seattle WA, USA
| | - David N Fredricks
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Elizabeth Irungu
- Department of Medicine, Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kenneth Ngure
- Department of Medicine, Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Nelly Mugo
- Department of Medicine, Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Jeanne Marrazzo
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Marla J Keller
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Betsy C Herold
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
23
|
Farr Zuend C, Noël-Romas L, Hoger S, McCorriser S, Westmacott G, Marrazzo J, Hillier SL, Dezzutti C, Squires K, Bunge KE, Burgener A. Influence of dapivirine vaginal ring use on cervicovaginal immunity and functional microbiome in adolescent girls. AIDS 2021; 35:369-380. [PMID: 33181534 PMCID: PMC7924934 DOI: 10.1097/qad.0000000000002751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The antiretroviral-based dapivirine vaginal ring reduced HIV risk among women in phase III clinical trials. However, limited data exists on the impact of dapivirine on the vaginal microenvironment in adolescents. DESIGN A comprehensive metaproteomics approach was used to assess host proteome and microbiome changes in cervicovaginal mucus with dapivirine ring use in adolescents enrolled in the MTN-023/IPM 030 (MTN-023) trial. METHODS Participants were randomized 3 : 1 to use dapivirine or placebo vaginal rings monthly for 6 months. Cervicovaginal samples from a subset of 35 participants (8 placebo, 27 dapivirine) were analyzed. RESULTS Mass spectrometry analysis identified 405 human and 2467 bacterial proteins belonging to 15 unique genera. The host proteome belonged to many functional pathways primarily related to inflammation. When stratified by study treatment arm, 18 (4.4%) and 28 (6.9%) human proteins were differentially abundant (adjusted P < 0.05) between baseline and follow-up in the placebo and dapivirine arms, respectively. The vaginal microbiome was predominantly composed of Lactobacillus, Gardnerella, and Prevotella. Although bacterial taxa did not differ by arm or change significantly, Lactobacillus crispatus increased (P < 0.001) and Lactobacillus iners decreased (P < 0.001) during the 6-month follow-up. There were no significant differences in bacterial functions by arm or time in the trial. Protected vaginal sex significantly associated with decreased neutrophil inflammatory biomarkers and may be associated with changes in bacterial taxa and metabolism. CONCLUSION Condom use may associate with differences to inflammation and bacterial function but dapivirine ring use does not, thereby supporting the mucosal safety profile of this vaginal ring for adolescents.
Collapse
Affiliation(s)
- Christina Farr Zuend
- Center for Global health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - Laura Noël-Romas
- Center for Global health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
- Departments of Obstetrics and Gynecology and Medical Microbiology, University of Manitoba
| | - Sarah Hoger
- Departments of Obstetrics and Gynecology and Medical Microbiology, University of Manitoba
| | - Stuart McCorriser
- Mass Spectrometry and Proteomics Core Facility, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Garrett Westmacott
- Mass Spectrometry and Proteomics Core Facility, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Jeanne Marrazzo
- University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Sharon L. Hillier
- University of Pittsburgh School of Medicine
- Magee-Women's Research Institute, Pittsburgh
| | - Charlene Dezzutti
- University of Pittsburgh School of Medicine
- Magee-Women's Research Institute, Pittsburgh
| | - Kathleen Squires
- Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Katherine E. Bunge
- University of Pittsburgh School of Medicine
- Magee-Women's Research Institute, Pittsburgh
| | - Adam Burgener
- Center for Global health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
- Departments of Obstetrics and Gynecology and Medical Microbiology, University of Manitoba
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
24
|
Rodríguez-Nava C, Cortés-Sarabia K, Avila-Huerta MD, Ortiz-Riaño EJ, Estrada-Moreno AK, Alarcón-Romero LDC, Mata-Ruíz O, Medina-Flores Y, Vences-Velázquez A, Morales-Narváez E. Nanophotonic Sialidase Immunoassay for Bacterial Vaginosis Diagnosis. ACS Pharmacol Transl Sci 2021; 4:365-371. [PMID: 33615186 DOI: 10.1021/acsptsci.0c00211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 01/10/2023]
Abstract
Bacterial vaginosis (BV) affects reproductive-age women and can lead to pelvic inflammatory disease, postpartum endometritis, and preterm labor/delivery and predisposes the infection of sexually transmitted diseases. Typically, BV diagnosis involves the analysis of vaginal swab samples via microscopy operated by highly skilled personnel. Hence, novel approaches for BV diagnosis are an existing need. In response, the first immunosensing platform targeting sialidase, a BV biomarker, is reported. The nanophotonic operational principle of this biosensing platform allows for a cheaper, faster, and simpler analysis when compared with an indirect enzyme-linked immunosorbent assay (ELISA). The clinical evaluation of such a nanotechnology is highlighted, where 162 vaginal swab samples were analyzed with high sensitivity and specificity (96.29%, respectively). The resulting nanoimmunosensing platform offers a resourceful approach to perform a timely BV diagnosis.
Collapse
Affiliation(s)
- Cynthia Rodríguez-Nava
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica A. C., León 37150, Guanajuato, Mexico.,Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Karen Cortés-Sarabia
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Mariana D Avila-Huerta
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica A. C., León 37150, Guanajuato, Mexico
| | - Edwin J Ortiz-Riaño
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica A. C., León 37150, Guanajuato, Mexico
| | - Ana K Estrada-Moreno
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Luz Del C Alarcón-Romero
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Olga Mata-Ruíz
- Laboratorio de Anticuerpos Monoclonales, Instituto de Diagnóstico y Referencia Epidemiológicos "Dr. Manuel Martínez Báez", Francisco de P. Miranda 177, Lomas de Plateros 01480, Ciudad de México 02210, Mexico
| | - Yolanda Medina-Flores
- Laboratorio de Anticuerpos Monoclonales, Instituto de Diagnóstico y Referencia Epidemiológicos "Dr. Manuel Martínez Báez", Francisco de P. Miranda 177, Lomas de Plateros 01480, Ciudad de México 02210, Mexico
| | - Amalia Vences-Velázquez
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Guerrero, Mexico
| | - Eden Morales-Narváez
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica A. C., León 37150, Guanajuato, Mexico
| |
Collapse
|
25
|
Lee CY, Cheu RK, Lemke MM, Gustin AT, France MT, Hampel B, Thurman AR, Doncel GF, Ravel J, Klatt NR, Arnold KB. Quantitative modeling predicts mechanistic links between pre-treatment microbiome composition and metronidazole efficacy in bacterial vaginosis. Nat Commun 2020; 11:6147. [PMID: 33262350 PMCID: PMC7708644 DOI: 10.1038/s41467-020-19880-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Bacterial vaginosis is a condition associated with adverse reproductive outcomes and characterized by a shift from a Lactobacillus-dominant vaginal microbiota to a polymicrobial microbiota, consistently colonized by strains of Gardnerella vaginalis. Metronidazole is the first-line treatment; however, treatment failure and recurrence rates remain high. To understand complex interactions between Gardnerella vaginalis and Lactobacillus involved in efficacy, here we develop an ordinary differential equation model that predicts bacterial growth as a function of metronidazole uptake, sensitivity, and metabolism. The model shows that a critical factor in efficacy is Lactobacillus sequestration of metronidazole, and efficacy decreases when the relative abundance of Lactobacillus is higher pre-treatment. We validate results in Gardnerella and Lactobacillus co-cultures, and in two clinical cohorts, finding women with recurrence have significantly higher pre-treatment levels of Lactobacillus relative to bacterial vaginosis-associated bacteria. Overall results provide mechanistic insight into how personalized differences in microbial communities influence vaginal antibiotic efficacy.
Collapse
Affiliation(s)
- Christina Y Lee
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Ryan K Cheu
- University of Miami Department of Pediatrics, University of Miami, Miami, FL, USA
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Melissa M Lemke
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Andrew T Gustin
- University of Miami Department of Pediatrics, University of Miami, Miami, FL, USA
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Michael T France
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Benjamin Hampel
- Division of Infectious Diseases and Hospital Epidemiology, University of Zurich, Zürich, Switzerland
| | | | | | - Jacques Ravel
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nichole R Klatt
- University of Miami Department of Pediatrics, University of Miami, Miami, FL, USA.
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | - Kelly B Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
26
|
Castanheira CP, Sallas ML, Nunes RAL, Lorenzi NPC, Termini L. Microbiome and Cervical Cancer. Pathobiology 2020; 88:187-197. [PMID: 33227782 DOI: 10.1159/000511477] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/07/2020] [Indexed: 11/19/2022] Open
Abstract
Persistent infection with some types of mucosal human papillomavirus (HPV) is the etiological factor for the development of cervical cancer and its precursor lesions. Besides, several cofactors are known to play a role in cervical disease onset and progression either by favoring or by preventing HPV infection and persistence. The microbiome of a healthy female genital tract is characterized by the presence of 1 or few varieties of lactobacilli. However, high-throughput studies addressing the bacterial diversity and abundance in the female genital tract have shown that several factors, including hormonal levels, hygiene habits, and sexually transmitted diseases may disrupt the natural balance, favoring the outgrowth of some groups of bacteria, which in turn may favor some pathological states. Recently, the vaginal microbiome has emerged as a new variable that could greatly influence the natural history of HPV infections and their clinical impact. In this context, changes in the vaginal microbiome have been detected in women infected with HPV and women with HPV-associated lesions and cancer. However, the role of specific bacteria groups in the development/progression or prevention/regression of HPV-associated pathologies is not well understood. In this review we summarize the current knowledge concerning changes in vaginal microbiome and cervical disease. We discuss the potential functional interplay between specific bacterial groups and HPV infection outcomes.
Collapse
Affiliation(s)
| | - Mayara Luciana Sallas
- Innovation in Cancer Laboratory, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo Octavio Frias de Oliveira, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Rafaella Almeida Lima Nunes
- Innovation in Cancer Laboratory, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo Octavio Frias de Oliveira, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Lara Termini
- Innovation in Cancer Laboratory, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo Octavio Frias de Oliveira, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil,
| |
Collapse
|
27
|
Inflammation, HIV, and Immune Quiescence: Leveraging on Immunomodulatory Products to Reduce HIV Susceptibility. AIDS Res Treat 2020; 2020:8672850. [PMID: 33178456 PMCID: PMC7609152 DOI: 10.1155/2020/8672850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/23/2020] [Accepted: 10/15/2020] [Indexed: 12/29/2022] Open
Abstract
The relationship between inflammation and HIV has been a focus of research over the last decade. In HIV-infected individuals, increased HIV-associated immune activation significantly correlated to disease progression. While genital inflammation (GI) has been shown to significantly increase the risk of HIV acquisition and transmission, immune correlates for reduced risk remain limited. In certain HIV-exposed seronegative individuals, an immune quiescent phenotype characterized reduced risk. Immune quiescence is defined by specific, targeted, highly regulated immune responses that hinder overt inflammation or immune activation. Targeted management of inflammation, therefore, is a plausible strategy to mitigate HIV risk and slow disease progression. Nonsteroidal anti-inflammatory drugs (NSAIDs) such as hydroxychloroquine and aspirin have shown encouraging preliminary results in low-risk women by reducing systemic and genital immune activation. A topical NSAID, containing ibuprofen, is effective in treating vulvovaginal inflammation. Additionally, the glucocorticoids (GCs), prednisolone, and dexamethasone are used to treat HIV-associated immune activation. Collectively, these data inform on immune-modulating drugs to reduce HIV risk. However, the prolonged use of these pharmaceutical drugs is associated with adverse effects, both systemically and to a lesser extent topically. Natural products with their reduced side effects coupled with anti-inflammatory properties render them viable options. Lactic acid (LA) has immunomodulatory properties. LA regulates the genital microbiome by facilitating the growth of Lactobacillus species, while simultaneously limiting bacterial species that cause microbial dysbiosis and GI. Glycerol monolaurate, besides being anti-inflammatory, also inhibited SIV infections in rhesus macaques. The proposed pharmaceutical and natural products could be used in combination with either antiretrovirals for treatment or preexposure prophylaxis for HIV prevention. This review provides a summary on the associations between inflammation, HIV risk, and disease progression. Furthermore, we use the knowledge from immune quiescence to exploit the use of pharmaceutical and natural products as strategic interventions to manage inflammation, toward mitigating HIV infections.
Collapse
|
28
|
Lacroix G, Gouyer V, Gottrand F, Desseyn JL. The Cervicovaginal Mucus Barrier. Int J Mol Sci 2020; 21:ijms21218266. [PMID: 33158227 PMCID: PMC7663572 DOI: 10.3390/ijms21218266] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/19/2022] Open
Abstract
Preterm births are a global health priority that affects 15 million babies every year worldwide. There are no effective prognostic and therapeutic strategies relating to preterm delivery, but uterine infections appear to be a major cause. The vaginal epithelium is covered by the cervicovaginal mucus, which is essential to health because of its direct involvement in reproduction and functions as a selective barrier by sheltering the beneficial lactobacilli while helping to clear pathogens. During pregnancy, the cervical canal is sealed with a cervical mucus plug that prevents the vaginal flora from ascending toward the uterine compartment, which protects the fetus from pathogens. Abnormalities of the cervical mucus plug and bacterial vaginosis are associated with a higher risk of preterm delivery. This review addresses the current understanding of the cervicovaginal mucus and the cervical mucus plug and their interactions with the microbial communities in both the physiological state and bacterial vaginosis, with a focus on gel-forming mucins. We also review the current state of knowledge of gel-forming mucins contained in mouse cervicovaginal mucus and the mouse models used to study bacterial vaginosis.
Collapse
|
29
|
Chen S, Gu Z, Zhang W, Jia S, Wu Y, Zheng P, Dai Y, Leng J. Microbiome of the lower genital tract in Chinese women with endometriosis by 16s-rRNA sequencing technique: a pilot study. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1440. [PMID: 33313185 PMCID: PMC7723586 DOI: 10.21037/atm-20-1309] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Endometriosis is a benign, chronic, gynecological disease which affect the women in reproductive age. The dysfunction of immune system is associated with endometriosis and the diversity of microbiota in genital tract. According to previous studies, microbiota significantly contributes to multi-systemic function, but the evidence of relationship between microbiota and endometriosis remains insufficient. Methods There are 68 participants were included in this study and 134 samples obtained from the cervical canal, posterior fornix and uterine cavity were analyzed by 16s-rRNA sequencing. The raw data was filtered, analyzed, and visualized, and bio-information methods were used to identify the characteristics of microbiota. Results Two different locations near the cervix, cervical canal, and posterior fornix, exhibited no differences in alpha diversity. The microbiota profile of adenomyosis with endometriosis patients is different from control group through PCoA. Among the different disease groups, five microbiotas were distinctive in the genus level, and Atopobium presented with the greatest significance in adenomyoisis-endometriosis patients. The LeFSe analysis failed to identify the special biomarkers, while several characteristic functions were identified through PICRUSt. Conclusions Lactobacillus is the predominant genus in the female lower genital tract, and Atopobium is higher in patients with endometriosis combined with adenomyosis. Several different functions of microbiota were explored, some of them are found to be associated with endometriosis or adenomyosis, other functions are needed to be further verified. These findings may provide a new concept of microbiota/immune system/endometriosis system.
Collapse
Affiliation(s)
- Sikai Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China.,Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Zhiyue Gu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China.,Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Shuangzheng Jia
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China.,Department of Gynecologic Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yushi Wu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Ping Zheng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China.,Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yi Dai
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Jinhua Leng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Cromarty R, Sigal A, Liebenberg LJ, Mckinnon LR, Abdool Karim SS, Passmore JAS, Archary D. Betamethasone induces potent immunosuppression and reduces HIV infection in a PBMC in vitro model. J Investig Med 2020; 69:28-40. [PMID: 33004468 PMCID: PMC7803916 DOI: 10.1136/jim-2020-001424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 01/15/2023]
Abstract
Genital inflammation is an established risk factor for increased HIV acquisition risk. Certain HIV-exposed seronegative populations, who are naturally resistant to HIV infection, have an immune quiescent phenotype defined by reduced immune activation and inflammatory cytokines at the genital tract. Therefore, the aim of this study was to create an immune quiescent environment using immunomodulatory drugs to mitigate HIV infection. Using an in vitro peripheral blood mononuclear cell (PBMC) model, we found that inflammation was induced using phytohemagglutinin and Toll-like receptor (TLR) agonists Pam3CSK4 (TLR1/2), lipopolysaccharide (LPS) (TLR4) and R848 (TLR7/8). After treatment with anti-inflammatory drugs, ibuprofen (IBF) and betamethasone (BMS), PBMCs were exposed to HIV NL4-3 AD8. Multiplexed ELISA was used to measure 28 cytokines to assess inflammation. Flow cytometry was used to measure immune activation (CD38, HLA-DR and CCR5) and HIV infection (p24 production) of CD4+ T cells. BMS potently suppressed inflammation (soluble cytokines, p<0.05) and immune activation (CD4+ T cells, p<0.05). BMS significantly reduced HIV infection of CD4+ T cells only in the LPS (0.98%) and unstimulated (1.7%) conditions (p<0.02). In contrast, IBF had minimal anti-inflammatory and immunosuppressive but no anti-HIV effects. BMS demonstrated potent anti-inflammatory effects, regardless of stimulation condition. Despite uniform immunosuppression, BMS differentially affected HIV infection according to the stimulation conditions, highlighting the complex nature of these interactions. Together, these data underscore the importance of interrogating inflammatory signaling pathways to identify novel drug targets to mitigate HIV infection.
Collapse
Affiliation(s)
- Ross Cromarty
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
| | - Alexander Sigal
- Africa Health Research Institute (AHRI), Durban, KwaZulu-Natal, South Africa
- Max-Planck-Institute for Infection Biology, Berlin, Germany
| | - Lenine Julie Liebenberg
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Lyle Robert Mckinnon
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Salim Safurdeen Abdool Karim
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Jo-Ann Shelly Passmore
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town Faculty of Health Sciences, Cape Town, Western Cape, South Africa
| | - Derseree Archary
- Mucosal Immunology Laboratory, CAPRISA, Durban, KwaZulu-Natal, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
31
|
Redelinghuys MJ, Geldenhuys J, Jung H, Kock MM. Bacterial Vaginosis: Current Diagnostic Avenues and Future Opportunities. Front Cell Infect Microbiol 2020; 10:354. [PMID: 32850469 PMCID: PMC7431474 DOI: 10.3389/fcimb.2020.00354] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/09/2020] [Indexed: 12/16/2022] Open
Abstract
A healthy female genital tract harbors a microbiome dominated by lactic acid and hydrogen peroxide producing bacteria, which provide protection against infections by maintaining a low pH. Changes in the bacterial compositions of the vaginal microbiome can lead to bacterial vaginosis (BV), which is often associated with vaginal inflammation. Bacterial vaginosis increases the risk of acquiring sexually transmitted infections (STIs) like human immunodeficiency virus (HIV) and affects women's reproductive health negatively. In pregnant women, BV can lead to chorioamnionitis and adverse pregnancy outcomes, including preterm premature rupture of the membranes and preterm birth. In order to manage BV effectively, good diagnostic procedures are required. Traditionally clinical and microscopic methods have been used to diagnose BV; however, these methods require skilled staff and time and suffer from reduced sensitivity and specificity. New diagnostics, including highly sensitive and specific point-of-care (POC) tests, treatment modalities and vaccines can be developed based on the identification of biomarkers from the growing pool of vaginal microbiome and vaginal metabolome data. In this review the current and future diagnostic avenues will be discussed.
Collapse
Affiliation(s)
- Mathys J. Redelinghuys
- School of Clinical Medicine, Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Johannesburg, South Africa
| | - Janri Geldenhuys
- UP-Ampath Translational Genomics Initiative, Department of Biochemistry, Genetics and Microbiology, Faculty of Health Sciences and Faculty of Natural and Agricultural Sciences, Division of Genetics, University of Pretoria, Pretoria, South Africa
| | - Hyunsul Jung
- Department of Medical Microbiology, University of Pretoria, Pretoria, South Africa
| | - Marleen M. Kock
- Department of Medical Microbiology, University of Pretoria, Pretoria, South Africa
- Department of Medical Microbiology, Tshwane Academic Division, National Health Laboratory Service, Pretoria, South Africa
| |
Collapse
|
32
|
Cortés-Sarabia K, Rodríguez-Nava C, Medina-Flores Y, Mata-Ruíz O, López-Meza JE, Gómez-Cervantes MD, Parra-Rojas I, Illades-Aguiar B, Flores-Alfaro E, Vences-Velázquez A. Production and characterization of a monoclonal antibody against the sialidase of Gardnerella vaginalis using a synthetic peptide in a MAP8 format. Appl Microbiol Biotechnol 2020; 104:6173-6183. [PMID: 32462244 PMCID: PMC7253150 DOI: 10.1007/s00253-020-10691-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/07/2020] [Accepted: 05/17/2020] [Indexed: 11/27/2022]
Abstract
Abstract Bacterial vaginosis is one of the most frequent vaginal infections. Its main etiological agent is Gardnerella vaginalis, which produces several virulence factors involved in vaginal infection and colonization, in particular, sialidase (SLD), a potential clinical biomarker that participates in immune response modulation and mucus degradation. The main objective of this work was the production and evaluation of a monoclonal antibody against G. vaginalis sialidase and its validation in immunoassays. For immunization of mice, a synthetic multiantigenic peptide was used, and hybridomas were generated. After fusion, hybridomas were evaluated for antibody production and cloned by limited dilution. One clone producing IgG1 was selected and characterized by indirect ELISA, dot blot, and Western blot, and we also tested clinical isolates and HeLa cells infected with G. vaginalis. The results showed that the anti-SLD antibody recognized a single protein of ~90 kDa that correlated with the estimated molecular weight of SLD. In addition, anti-SLD antibody recognized SLD from complete bacteria and from culture supernatants of infected Hela cells. In conclusion, our results showed that the anti-SLD antibody recognized SLD from different sources and could be considered a new tool for the diagnosis of bacterial vaginosis. Key Points • Anti-sialidase mAb was generated using a synthetic peptide • The mAb recognizes synthetic peptide and intact protein from multiple sources • The antibody was characterized by several immunological methods
Collapse
Affiliation(s)
- Karen Cortés-Sarabia
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Cynthia Rodríguez-Nava
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Yolanda Medina-Flores
- Laboratorio de Anticuerpos Monoclonales, Instituto de Diagnóstico y Referencia Epidemiológicos "Dr. Manuel Martínez Báez", Francisco de P. Miranda 177, Lomas de Plateros, 01480, Ciudad de México, Mexico
| | - Olga Mata-Ruíz
- Laboratorio de Anticuerpos Monoclonales, Instituto de Diagnóstico y Referencia Epidemiológicos "Dr. Manuel Martínez Báez", Francisco de P. Miranda 177, Lomas de Plateros, 01480, Ciudad de México, Mexico
| | - Joel E López-Meza
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | | | - Isela Parra-Rojas
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Berenice Illades-Aguiar
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Eugenia Flores-Alfaro
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Amalia Vences-Velázquez
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
33
|
O'Hanlon DE, Gajer P, Brotman RM, Ravel J. Asymptomatic Bacterial Vaginosis Is Associated With Depletion of Mature Superficial Cells Shed From the Vaginal Epithelium. Front Cell Infect Microbiol 2020; 10:106. [PMID: 32211347 PMCID: PMC7076050 DOI: 10.3389/fcimb.2020.00106] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/27/2020] [Indexed: 12/26/2022] Open
Abstract
Previous studies have described bacterial vaginosis (BV) as associated with increased cell-shedding from the cervicovaginal epithelium. Cell-shedding in excess of cell-proliferation is thought to decrease epithelial barrier function and increase susceptibility to infection. This study evaluated the number of shed cells in mid-vaginal smears from women with a diagnosis of symptomatic BV (sBV, n = 17), asymptomatic BV (aBV, n = 71), or no BV (n = 104) by Amsel criteria. The sBV smears contained significantly more shed cells (median 158/100X field) than no BV smears (median 91/100X field), p = 7.2e−9. However, we observed that aBV smears contained significantly fewer shed cells (median 35/100X field) than no BV smears, p = 22.0e−16. The sizes of cell-aggregates (cells shed in sometimes multilayered sections with intact cell-cell attachments) followed the same pattern. Cell-aggregates in sBV smears were significantly larger (median ~220,000 μm2) than those in no BV smears (median ~50,000 μm2), p = 1.8e−6, but cell-aggregates in aBV smears were significantly smaller (median ~7,000 μm2) than those in no BV smears, p = 0.0028. We also compared the superficial cell index (SCI), a measure of cervicovaginal epithelial cell maturity, in no BV and aBV smears with relatively low numbers of shed cells (≤50/100X field). The SCI of no BV smears was significantly higher (median 0.86) than that of aBV smears (median 0.35), p = 4.3e−98, suggesting a depletion of mature cells with exposure and shedding of underlying immature cells in aBV with low number of shed cells. These results indicate that aBV may contribute disproportionately to the increased susceptibility to reproductive tract infections associated with BV. Our findings remained true when considering only those smears in which the microbiota comprised a diverse set of strict and facultative anaerobic bacteria [Community State Type IV (n = 162)], thus excluding those dominated by Lactobacillus spp. This is consistent with our developing hypothesis that high-shedding sBV and low-shedding aBV could be temporally separated phases of the same condition, rather than two separate forms of BV. These findings might inform future work on clinical management of symptomatic and asymptomatic bacterial vaginosis.
Collapse
Affiliation(s)
- D Elizabeth O'Hanlon
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Pawel Gajer
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rebecca M Brotman
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
34
|
Wessels JM, Lajoie J, Cooper MIJH, Omollo K, Felker AM, Vitali D, Dupont HA, Nguyen PV, Mueller K, Vahedi F, Kimani J, Oyugi J, Cheruiyot J, Mungai JN, Deshiere A, Tremblay MJ, Mazzulli T, Stearns JC, Ashkar AA, Fowke KR, Surette MG, Kaushic C. Medroxyprogesterone acetate alters the vaginal microbiota and microenvironment in women and increases susceptibility to HIV-1 in humanized mice. Dis Model Mech 2019; 12:dmm.039669. [PMID: 31537512 PMCID: PMC6826019 DOI: 10.1242/dmm.039669] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
The hormonal contraceptive medroxyprogesterone acetate (MPA) is associated with increased risk of human immunodeficiency virus (HIV), via incompletely understood mechanisms. Increased diversity in the vaginal microbiota modulates genital inflammation and is associated with increased HIV-1 acquisition. However, the effect of MPA on diversity of the vaginal microbiota is relatively unknown. In a cohort of female Kenyan sex workers, negative for sexually transmitted infections (STIs), with Nugent scores <7 (N=58 of 370 screened), MPA correlated with significantly increased diversity of the vaginal microbiota as assessed by 16S rRNA gene sequencing. MPA was also significantly associated with decreased levels of estrogen in the plasma, and low vaginal glycogen and α-amylase, factors implicated in vaginal colonization by lactobacilli, bacteria that are believed to protect against STIs. In a humanized mouse model, MPA treatment was associated with low serum estrogen, low glycogen and enhanced HIV-1 susceptibility. The mechanism by which the MPA-mediated changes in the vaginal microbiota may contribute to HIV-1 susceptibility in humans appears to be independent of inflammatory cytokines and/or activated T cells. Altogether, these results suggest MPA-induced hypo-estrogenism may alter key metabolic components that are necessary for vaginal colonization by certain bacterial species including lactobacilli, and allow for greater bacterial diversity in the vaginal microbiota. This article has an associated First Person interview with the first author of the paper. Summary: MPA may increase susceptibility to HIV-1 in sex workers through the suppression of endogenous estrogen, reducing vaginal glycogen and α-amylase levels, which increases bacterial diversity, potentially reducing protective bacterial species such as lactobacilli.
Collapse
Affiliation(s)
- Jocelyn M Wessels
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Julie Lajoie
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.,Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | - Maeve I J Hay Cooper
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Kenneth Omollo
- Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | - Allison M Felker
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Danielle Vitali
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Haley A Dupont
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Philip V Nguyen
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Kristen Mueller
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Fatemeh Vahedi
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Joshua Kimani
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.,Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya.,Kenyan AIDS Control Program, P.O. Box 19361 - 00202, Nairobi, Kenya
| | - Julius Oyugi
- Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | | | - John N Mungai
- Kenyan AIDS Control Program, P.O. Box 19361 - 00202, Nairobi, Kenya
| | - Alexandre Deshiere
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec City, Québec G1V 4G2, Canada.,Department of Microbiology and Immunology Medical Biology, Université Laval, Québec City, Québec G1V 0A6, Canada
| | - Michel J Tremblay
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec City, Québec G1V 4G2, Canada.,Department of Microbiology and Immunology Medical Biology, Université Laval, Québec City, Québec G1V 0A6, Canada
| | - Tony Mazzulli
- Public Health Laboratories, Public Health Ontario, Toronto, Ontario M5G 1V2, Canada.,Mount Sinai Hospital/University Health Network, Department of Microbiology, Toronto, Ontario M5G 1X5, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jennifer C Stearns
- Department of Medicine, Farncombe Family Digestive Health Institute, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Keith R Fowke
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.,Department of Medical Microbiology, University of Nairobi, P.O. BOX 30197-00100, Nairobi, Kenya
| | - Michael G Surette
- Department of Medicine, Farncombe Family Digestive Health Institute, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,McMaster Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4K1, Canada .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4K1, Canada.,McMaster Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
35
|
Thurman A, Chandra N, Schwartz JL, Brache V, Chen BA, Asin S, Rollenhagen C, Herold BC, Fichorova RN, Hillier SL, Weiner DH, Mauck C, Doncel GF. The Effect of Hormonal Contraception on Cervicovaginal Mucosal End Points Associated with HIV Acquisition. AIDS Res Hum Retroviruses 2019; 35:853-864. [PMID: 30997816 DOI: 10.1089/aid.2018.0298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reproductive age women may choose to concurrently use topical antiretrovirals and hormonal contraceptives (HCs) to simultaneously prevent HIV-1 infection and unintended/mistimed pregnancy. There are conflicting data on the effect of HCs on mucosal susceptibility to HIV-1. The objective of this study was to evaluate cervicovaginal (CV) mucosal data from healthy women before and after initiation of either oral contraceptive pills (OCPs) or depot medroxyprogesterone acetate (DMPA) injection. CONRAD A10-114 was a prospective, open-label, parallel cohort study. We enrolled 74 women and 62 completed the visits (32 and 30 who selected OCPs and DMPA, respectively). Participants provided CV lavage, vaginal biopsies, and CV swabs at baseline in the luteal phase and then ∼6 weeks after initiating HCs. After contraceptive initiation, there were significant increases in vaginal immune cell density among both DMPA and OCP users. Changes for OCP users were concentrated in the subepithelial lamina propria, whereas for DMPA users, they were distributed throughout the vaginal tissue, including the epithelium (CD45+, CD3+, CD4+, and CD1a+). Contraceptive use altered concentrations of soluble CV inflammatory and immune mediators, with significant reductions in some proinflammatory cytokines and secretory leukoprotease inhibitor. Compared with baseline, p24 antigen production after ex vivo HIV-1 infection of vaginal biopsies doubled after DMPA use, but all p-values were >.05. HIV-1 replication was significantly higher in DMPA-exposed tissues compared with those from the OCP group at the end of the tissue culture (p = .01). Although not statistically significant, median in vitro inhibition of HIV-1 by CV fluid (innate antiviral activity), was reduced by ∼50% with HCs (p > .21). Exposure to exogenous contraceptive hormones significantly increased vaginal immune cells and reduced CV proinflammatory cytokines and antimicrobial peptides. DMPA users showed higher susceptibility to HIV-1 ex vivo infection.
Collapse
Affiliation(s)
- Andrea Thurman
- Eastern Virginia Medical School, CONRAD, Norfolk, Virginia
| | | | | | | | - Beatrice A. Chen
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Susana Asin
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine, Dartmouth, New Hampshire
| | - Christiane Rollenhagen
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine, Dartmouth, New Hampshire
| | - Betsy C. Herold
- Department of Pediatric Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York
| | - Raina N. Fichorova
- Laboratory of Genital Tract Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sharon L. Hillier
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|
36
|
Kalia N, Singh J, Kaur M. Immunopathology of Recurrent Vulvovaginal Infections: New Aspects and Research Directions. Front Immunol 2019; 10:2034. [PMID: 31555269 PMCID: PMC6722227 DOI: 10.3389/fimmu.2019.02034] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/12/2019] [Indexed: 12/25/2022] Open
Abstract
Recurrent vulvovaginal infections (RVVI), a devastating group of mucosal infection, are severely affecting women's quality of life. Our understanding of the vaginal defense mechanisms have broadened recently with studies uncovering the inflammatory nature of bacterial vaginosis, inflammatory responses against novel virulence factors, innate Type 17 cells/IL-17 axis, neutrophils mediated killing of pathogens by a novel mechanism, and oxidative stress during vaginal infections. However, the pathogens have fine mechanisms to subvert or manipulate the host immune responses, hijack them and use them for their own advantage. The odds of hijacking increases, due to impaired immune responses, the net magnitude of which is the result of numerous genetic variations, present in multiple host genes, detailed in this review. Thus, by underlining the role of the host immune responses in disease etiology, modern research has clarified a major hypothesis shift in the pathophilosophy of RVVI. This knowledge can further be used to develop efficient immune-based diagnosis and treatment strategies for this enigmatic disease conditions. As for instance, plasma-derived MBL replacement, adoptive T-cell, and antibody-based therapies have been reported to be safe and efficacious in infectious diseases. Therefore, these emerging immune-therapies could possibly be the future therapeutic options for RVVI.
Collapse
Affiliation(s)
- Namarta Kalia
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, India
| | - Jatinder Singh
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, India
| | - Manpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
37
|
Thurman A, Cunningham T, Fichorova R, Herold BC, Hillier SL, Chandra N, Doncel GF. A phase I randomized safety study of a single-size silicone rubber diaphragm used with or without a lactic-acid-containing diaphragm gel. Contraception 2019; 100:430-437. [PMID: 31442441 DOI: 10.1016/j.contraception.2019.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/24/2019] [Accepted: 06/02/2019] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To evaluate a lactic-acid-containing diaphragm gel (Contragel®) approved outside the United States for use with a silicone rubber diaphragm (Caya®). The study gel is being evaluated as a safer alternative to nonoxynol-9 (N-9) gel, which has been associated with risk of increasing susceptibility to human immunodeficiency virus (HIV). STUDY DESIGN This was a Phase I randomized, parallel study evaluating the safety of the novel diaphragm gel versus hydroxyethylcellulose (HEC) universal placebo gel delivered by the study diaphragm for two 7-day test cycles of daily use, without and with intercourse. The primary clinical safety endpoint was treatment emergent adverse events. Mucosal safety endpoints included colposcopic findings, anti-Escherichia coli activity of endocervical and vaginal fluid, immune mediators, Nugent score and ectocervical immune cell density. Endpoints were assessed prior to each test cycle and at day 7 of each test cycle. We compared the two independent groups and also evaluated paired changes from baseline in each gel cohort. RESULTS Twenty-three participants used the study diaphragm with the novel gel (n=11) or with HEC (n=12). Use of either gel resulted in few genital AEs and no colposcopic findings. There were no differences in ectocervical histology and lymphocyte density or phenotype between the two cohorts at baseline or after each test cycle. We found no clinically important differences in the anti-microbial (anti Escherichia coli) activity of endocervical or vaginal fluid or concentrations of genital immune mediators (e.g. anti-inflammatory secretory leukocyte protease inhibitor (SLPI) or pro-inflammatory mediator RANTES) between the two gel cohorts at any visit. There were no important paired changes from baseline among participants using either gel in Nugent score, ectocervical histology or anti-microbial activity of genital secretions. CONCLUSIONS We found no clinically significant differences in clinical and mucosal safety endpoints between the two cohorts. The mucosal safety profiles of the study gel and HEC placebo gel were similar. IMPLICATIONS Our data demonstrate no clinically important differences between the safety profiles of the lactic-acid-containing diaphragm gel versus HEC placebo gel when used with the study diaphragm. N-9 can no longer be used with contraceptive diaphragms in high HIV prevalence regions. Although larger studies are needed, the novel gel appears safe for use with the study diaphragm, which is the first over-the-counter, non-hormonal, diaphragm.
Collapse
Affiliation(s)
- Andrea Thurman
- CONRAD, Eastern Virginia Medical School, 601 Colley Ave, Norfolk, VA, USA 23507.
| | - Tina Cunningham
- Healthcare Delivery Science Program, Healthcare Analytics Program, Eastern Virginia Medical School, 651 Colley Ave, Norfolk, VA, USA 23507
| | - Raina Fichorova
- Laboratory of Genital Tract Biology, Brigham and Women's Hospital, Harvard University, 221 Longwood Ave., Boston, MA, 02115
| | - Betsy C Herold
- Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Sharon L Hillier
- Magee-Womens Research Institute and the University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA 15213
| | - Neelima Chandra
- CONRAD, Eastern Virginia Medical School, 601 Colley Ave, Norfolk, VA, USA 23507
| | - Gustavo F Doncel
- CONRAD, Eastern Virginia Medical School, 601 Colley Ave, Norfolk, VA, USA 23507; CONRAD, Eastern Virginia Medical School, 1911 North Fort Myer Drive, Arlington, Virginia, USA 22209
| |
Collapse
|
38
|
Keller MJ, Huber A, Espinoza L, Serrano MG, Parikh HI, Buck GA, Gold JA, Wu Y, Wang T, Herold BC. Impact of Herpes Simplex Virus Type 2 and Human Immunodeficiency Virus Dual Infection on Female Genital Tract Mucosal Immunity and the Vaginal Microbiome. J Infect Dis 2019; 220:852-861. [PMID: 31111902 PMCID: PMC6667798 DOI: 10.1093/infdis/jiz203] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/22/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Mechanisms linking herpes simplex virus type 2 (HSV-2) with human immunodeficiency virus (HIV) are not fully defined. We tested the hypothesis that HSV-2 and HIV dual infection is associated with cervicovaginal inflammation and/or vaginal dysbiosis. METHODS Genital tract samples were obtained weekly over a 12-week period from 30 women seropositive (+) for HIV and HSV-2 and 15 women each who were seropositive for one or seronegative (-) for both viruses. Immune mediators, antimicrobial activity, and microbial composition and diversity were compared. RESULTS Significant differences in the concentrations of interferon-γ (P = .002), tumor necrosis factor-α (P = .03), human beta defensin 1 (P = .001), secretory leukocyte protease inhibitor (P = .01), and lysozyme (P = .03) were observed across the 4 groups (Kruskal-Wallis). There were also significant differences in vaginal microbial alpha diversity (Simpson index) (P = .0046). Specifically, when comparing HIV-1+/HSV-2+ to HIV-1-/HSV-2- women, a decrease in Lactobacillus crispatus and increase in diverse anaerobes was observed. The number of genital HSV outbreaks was greater in HIV+ versus HIV- women (39 versus 12) (P = .04), but there were no significant differences when comparing outbreak to non-outbreak visits. CONCLUSIONS Increased microbial diversity and cervicovaginal inflammation in HIV and HSV-2 dually infected women may adversely impact genital health and, in the absence of antiretroviral therapy, facilitate HIV shedding.
Collapse
Affiliation(s)
- Marla J Keller
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Ashley Huber
- Departments of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
- Present Affiliation: Department of Obstetrics, Gynecology and Reproductive Medicine, Stony Brook University School of Medicine, Stony Brook, New York
| | - Lilia Espinoza
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Myrna G Serrano
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond
| | - Hardik I Parikh
- School of Medicine Research Computing, University of Virginia, Charlottesville
| | - Gregory A Buck
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond
- Computer Science Department, Virginia Commonwealth University, Richmond
| | - Jeremy A Gold
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Present Affiliation: Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Yiqun Wu
- Departments of Epidemiology and Population Health, Bronx, New York
- Present Affiliation: Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tao Wang
- Departments of Epidemiology and Population Health, Bronx, New York
| | - Betsy C Herold
- Departments of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
- Department of Pediatrics, Albert Einstein College of Medicine and Children’s Hospital at Montefiore, Bronx, New York
| |
Collapse
|
39
|
Cromarty R, Sigal A, Liebenberg LJP, McKinnon LR, Abdool Karim SS, Passmore JAS, Archary D. Diminished HIV Infection of Target CD4+ T Cells in a Toll-Like Receptor 4 Stimulated in vitro Model. Front Immunol 2019; 10:1705. [PMID: 31396221 PMCID: PMC6664077 DOI: 10.3389/fimmu.2019.01705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Genital inflammation is associated with increased HIV acquisition risk. Induction of an inflammatory response can occur through the recognition of pathogenic or commensal microbes by Toll-like receptors (TLRs) on various immune cells. We used a in vitro peripheral blood mononuclear cell (PBMC) system to understand the contribution of TLR stimulation in inducing inflammation and the activation of target T cells, and its effect on HIV susceptibility. PBMCs were stimulated with TLR agonists LPS (TLR4), R848 (TLR7/8), and Pam3CSK4 (TLR1/2), and then infected with HIV NL4-3 AD8. Multiplexed ELISA was used to measure 28 cytokines in cell culture supernatants. Flow cytometry was used to measure the activation state (CD38 and HLA-DR), and CCR5 expression on CD4+ and CD8+ T cells. Although TLR agonists induced higher cytokine and chemokine secretion, they did not significantly activate CD4+ and CD8+ T cells and showed decreased CCR5 expression relative to the unstimulated control. Despite several classes of inflammatory cytokines and chemokines being upregulated by TLR agonists, CD4+ T cells were significantly less infectable by HIV after TLR4-stimulation than the unstimulated control. These data demonstrate that the inflammatory effects that occur in the presence TLR agonist stimulations do not necessarily translate to the activation of T cells. Most importantly, the finding that TLR4-stimulation reduces rather than increases susceptibility of CD4+ T cells to HIV infection in this in vitro system strongly suggests that the increased chemokine and possible antiviral factor expression induced by these TLR agonists play a powerful although complex role in determining HIV infection risk.
Collapse
Affiliation(s)
- Ross Cromarty
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Alex Sigal
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Lenine J P Liebenberg
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Lyle R McKinnon
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Jo-Ann S Passmore
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Medical School, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
40
|
Amerson-Brown MH, Miller AL, Maxwell CA, White MM, Vincent KL, Bourne N, Pyles RB. Cultivated Human Vaginal Microbiome Communities Impact Zika and Herpes Simplex Virus Replication in ex vivo Vaginal Mucosal Cultures. Front Microbiol 2019; 9:3340. [PMID: 30692980 PMCID: PMC6340164 DOI: 10.3389/fmicb.2018.03340] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/24/2018] [Indexed: 01/07/2023] Open
Abstract
The human vaginal microbiome (VMB) is a complex bacterial community that interacts closely with vaginal epithelial cells (VECs) impacting the mucosal phenotype and its responses to pathogenic insults. The VMB and VEC relationship includes nutrient exchange and regulation of signaling molecules that controls numerous host functions and defends against invading pathogens. To better understand infection and replication of sexually transmitted viral pathogens in the human vaginal mucosa we used our ex vivo VEC multilayer culture system. We tested the hypothesis that selected VMB communities could be identified that alter the replication of sexually transmitted viruses consistent with reported clinical associations. Sterile VEC multilayer cultures or those colonized with VMB dominated by specific Lactobacillus spp., or VMB lacking lactobacilli, were infected with Zika virus, (ZIKV) a single stranded RNA virus, or Herpes Simplex Virus type 2 (HSV-2), a double stranded DNA virus. The virus was added to the apical surface of the cultured VEC multilayer to model transmission during vaginal intercourse. Viral replication was measured 48 h later by qPCR. The results indicated that VEC cultures colonized by VMB containing Staphylococcus spp., previously reported as inflammatory, significantly reduced the quantity of viral genomes produced by ZIKV. HSV-2 titers were decreased by nearly every VMB tested relative to the sterile control, although Lactobacillus spp.-dominated VMBs caused the greatest reduction in HSV-2 titer consistent with clinical observations. To explore the mechanism for reduced ZIKV titers, we investigated inflammation created by ZIKV infection, VMB colonization or pre-exposure to selected TLR agonists. Finally, expression levels of human beta defensins 1–3 were quantified in cultures infected by ZIKV and those colonized by VMBs that impacted ZIKV titers. Human beta defensins 1–3 produced by the VEC showed no association with ZIKV titers. The data presented expands the utility of this ex vivo model system providing controlled and reproducible methods to study the VMB impact on STIs and indicated an association between viral replication and specific bacterial species within the VMB.
Collapse
Affiliation(s)
- Megan H Amerson-Brown
- Graduate School of Biomedical Sciences, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Aaron L Miller
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Carrie A Maxwell
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Mellodee M White
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Kathleen L Vincent
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Nigel Bourne
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Richard B Pyles
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
41
|
|
42
|
Thurman AR, Schwartz JL, Brache V, Clark MR, McCormick T, Chandra N, Marzinke MA, Stanczyk FZ, Dezzutti CS, Hillier SL, Herold BC, Fichorova R, Asin SN, Rollenhagen C, Weiner D, Kiser P, Doncel GF. Randomized, placebo controlled phase I trial of safety, pharmacokinetics, pharmacodynamics and acceptability of tenofovir and tenofovir plus levonorgestrel vaginal rings in women. PLoS One 2018; 13:e0199778. [PMID: 29953547 PMCID: PMC6023238 DOI: 10.1371/journal.pone.0199778] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 06/07/2018] [Indexed: 01/28/2023] Open
Abstract
To prevent the global health burdens of human immunodeficiency virus [HIV] and unintended/mistimed pregnancies, we developed an intravaginal ring [IVR] that delivers tenofovir [TFV] at ~10mg/day alone or with levonorgestrel [LNG] at ~20μg/day for 90 days. We present safety, pharmacokinetics, pharmacodynamics, acceptability and drug release data in healthy women. CONRAD A13-128 was a randomized, placebo controlled phase I study. We screened 86 women; 51 were randomized to TFV, TFV/LNG or placebo IVR [2:2:1] and 50 completed all visits, using the IVR for approximately 15 days. We assessed safety by adverse events, colposcopy, vaginal microbiota, epithelial integrity, mucosal histology and immune cell numbers and phenotype, cervicovaginal [CV] cytokines and antimicrobial proteins and changes in systemic laboratory measurements, and LNG and TFV pharmacokinetics in multiple compartments. TFV pharmacodynamic activity was measured by evaluating CV fluid [CVF] and tissue for antiviral activity using in vitro models. LNG pharmacodynamic assessments were timed based on peak urinary luteinizing hormone levels. All IVRs were safe with no significant colposcopic, mucosal, immune and microbiota changes and were acceptable. Among TFV containing IVR users, median and mean CV aspirate TFV concentrations remained above 100,000 ng/mL 4 hours post IVR insertion and mean TFV-diphosphate [DP] concentrations in vaginal tissue remained above 1,000 fmol/mg even 3 days post IVR removal. CVF of women using TFV-containing IVRs completely inhibited [94-100%] HIV infection in vitro. TFV/LNG IVR users had mean serum LNG concentrations exceeding 300 pg/mL within 1 hour, remaining high throughout IVR use. All LNG IVR users had a cervical mucus Insler score <10 and the majority [95%] were anovulatory or had abnormal cervical mucus sperm penetration. Estimated in vivo TFV and LNG release rates were within expected ranges. All IVRs were safe with the active ones delivering sustained high concentrations of TFV locally. LNG caused changes in cervical mucus, sperm penetration, and ovulation compatible with contraceptive efficacy. The TFV and TFV/LNG rings are ready for expanded 90 day clinical testing. Trial registration ClinicalTrials.gov #NCT02235662.
Collapse
Affiliation(s)
- Andrea Ries Thurman
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| | - Jill L. Schwartz
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | | | - Meredith R. Clark
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| | - Timothy McCormick
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| | - Neelima Chandra
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Mark A. Marzinke
- Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Frank Z. Stanczyk
- University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Charlene S. Dezzutti
- University of Pittsburgh, Department of Obstetrics, Gynecology & Reproductive Sciences, Department of Infectious Diseases & Microbiology, Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Sharon L. Hillier
- University of Pittsburgh School of Medicine, Departments of Obstetrics, Gynecology and Reproductive Sciences and Microbiology and Molecular Genetics, Pittsburgh, Pennsylvania, United States of America
| | - Betsy C. Herold
- Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Raina Fichorova
- Laboratory of Genital Tract Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Susana N. Asin
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine at Dartmouth, New Hampshire
| | - Christiane Rollenhagen
- V.A. Medical Center, White River Junction, VT and Geisel School of Medicine at Dartmouth, New Hampshire
| | - Debra Weiner
- FHI360, Durham, North Carolina, United States of America
| | - Patrick Kiser
- Northwestern University, Evanston, Illinois, United States of America
| | - Gustavo F. Doncel
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, United States of America
| |
Collapse
|
43
|
Abstract
: Bacterial vaginosis, characterized by the replacement of the Lactobacillus-dominant microbiota with anaerobic bacteria and facultative Gram-negative rods, has been associated with adverse reproductive health outcomes including HIV acquisition. With the advent of newer molecular techniques, the vaginal microbiota can be investigated in more detail and the association with HIV examined more thoroughly. This review examines recent evidence suggesting that vaginal dysbiosis with increased microbial diversity, specific vaginal bacterial communities, and the presence and concentrations of some individual bacterial species, may increase HIV susceptibility. Potential mechanisms through which vaginal microbiota could impact HIV susceptibility are discussed. On the basis of the available data, this review finds that there is a modest, but growing, body of evidence linking vaginal microbiota to HIV susceptibility in women. The evidence could be strengthened through two main pathways. First, laboratory studies such as ex-vivo or animal experiments are needed to move from plausible mechanisms towards proven mechanisms that explain an effect of the vaginal microbiota on HIV susceptibility. Second, experimental evidence could directly test the hypothesis that sustaining optimal microbiota reduces HIV risk, though there are important obstacles to conducting such studies. Finally, this review examines strong evidence from a recent publication suggesting that deviations from an optimal vaginal microbiome, and particularly the presence of some bacterial communities with high relative abundance of Gardnerella vaginalis, reduces the efficacy of vaginal tenofovir-based microbicides.
Collapse
|
44
|
Park YJ, Lee HK. The Role of Skin and Orogenital Microbiota in Protective Immunity and Chronic Immune-Mediated Inflammatory Disease. Front Immunol 2018; 8:1955. [PMID: 29375574 PMCID: PMC5767596 DOI: 10.3389/fimmu.2017.01955] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/19/2017] [Indexed: 12/11/2022] Open
Abstract
The skin and orogenital mucosae, which constitute complex protective barriers against infection and injuries, are not only the first to come into contact with pathogens but are also colonized by a set of microorganisms that are essential to maintain a healthy physiological environment. Using 16S ribosomal RNA metagenomic sequencing, scientists recognized that the microorganism colonization has greater diversity and variability than previously assumed. These microorganisms, such as commensal bacteria, affect the host’s immune response against pathogens and modulate chronic inflammatory responses. Previously, a single pathogen was thought to cause a single disease, but current evidence suggests that dysbiosis of the tissue microbiota may underlie the disease status. Dysbiosis results in aberrant immune responses at the surface and furthermore, affects the systemic immune response. Hence, understanding the initial interaction between the barrier surface immune system and local microorganisms is important for understanding the overall systemic effects of the immune response. In this review, we describe current evidence for the basis of the interactions between pathogens, microbiota, and immune cells on surface barriers and offer explanations for how these interactions may lead to chronic inflammatory disorders.
Collapse
Affiliation(s)
- Young Joon Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| |
Collapse
|
45
|
Lennard K, Dabee S, Barnabas SL, Havyarimana E, Blakney A, Jaumdally SZ, Botha G, Mkhize NN, Bekker LG, Lewis DA, Gray G, Mulder N, Passmore JAS, Jaspan HB. Microbial Composition Predicts Genital Tract Inflammation and Persistent Bacterial Vaginosis in South African Adolescent Females. Infect Immun 2018; 86:e00410-17. [PMID: 29038128 PMCID: PMC5736802 DOI: 10.1128/iai.00410-17] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/10/2017] [Indexed: 11/20/2022] Open
Abstract
Young African females are at an increased risk of HIV acquisition, and genital inflammation or the vaginal microbiome may contribute to this risk. We studied these factors in 168 HIV-negative South African adolescent females aged 16 to 22 years. Unsupervised clustering of 16S rRNA gene sequences revealed three clusters (subtypes), one of which was strongly associated with genital inflammation. In a multivariate model, the microbiome compositional subtype and hormonal contraception were significantly associated with genital inflammation. We identified 40 taxa significantly associated with inflammation, including those reported previously (Prevotella, Sneathia, Aerococcus, Fusobacterium, and Gemella) as well as several novel taxa (including increased frequencies of bacterial vaginosis-associated bacterium 1 [BVAB1], BVAB2, BVAB3, Prevotella amnii, Prevotella pallens, Parvimonas micra, Megasphaera, Gardnerella vaginalis, and Atopobium vaginae and decreased frequencies of Lactobacillus reuteri, Lactobacillus crispatus, Lactobacillus jensenii, and Lactobacillus iners). Women with inflammation-associated microbiomes had significantly higher body mass indices and lower levels of endogenous estradiol and luteinizing hormone. Community functional profiling revealed three distinct vaginal microbiome subtypes, one of which was characterized by extreme genital inflammation and persistent bacterial vaginosis (BV); this subtype could be predicted with high specificity and sensitivity based on the Nugent score (≥9) or BVAB1 abundance. We propose that women with this BVAB1-dominated subtype may have chronic genital inflammation due to persistent BV, which may place them at a particularly high risk for HIV infection.
Collapse
Affiliation(s)
- Katie Lennard
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Smritee Dabee
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Shaun L Barnabas
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Enock Havyarimana
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Anna Blakney
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Shameem Z Jaumdally
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Gerrit Botha
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla N Mkhize
- National Institute for Communicable Diseases, Sandringham, Johannesburg, South Africa
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - David A Lewis
- Western Sydney Sexual Health Centre, Parramatta, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
- Sydney Medical School-Westmead, University of Sydney, Sydney, Australia
- National Institute for Communicable Diseases, Sandringham, Johannesburg, South Africa
| | - Glenda Gray
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Nicola Mulder
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Jo-Ann S Passmore
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Johannesburg, South Africa
| | - Heather B Jaspan
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
46
|
Thurman AR, Yousefieh N, Chandra N, Kimble T, Asin S, Rollenhagen C, Anderson SM, Herold BC, Freiermuth JL, Starkman BS, Mesquita PM, Richardson-Harman N, Cunningham T, Hillier S, Rabe L, Schwartz JL, Doncel GF. Comparison of Mucosal Markers of Human Immunodeficiency Virus Susceptibility in Healthy Premenopausal Versus Postmenopausal Women. AIDS Res Hum Retroviruses 2017; 33:807-819. [PMID: 28398069 DOI: 10.1089/aid.2016.0320] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The objective of this study was to characterize cervicovaginal (CV) mucosal factors modulating susceptibility to human immunodeficiency virus (HIV) acquisition in healthy premenopausal (PRE) and postmenopausal (POST) women before and after treatment with estradiol (E2). We compared CV mucosal epithelial histology and immune cells, vaginal microbiota, antimicrobial activity of and soluble mucosal protein concentrations in the CV fluid lavage (CVL), and p24 antigen production after ex vivo infection of ectocervical tissues with HIV-1BaL among PRE women (n = 20) in the follicular and luteal phases of the menstrual cycle and POST women (n = 17) at baseline and after ∼1 month of treatment with 0.01% vaginal E2 cream. Compared to PRE women, we measured higher levels of p24 antigen after ex vivo infection in tissues from POST women. POST women had a significantly thinner vaginal epithelium with decreased tight junction proteins and a higher density of mucosal immune T cells and lower levels of CD1a antigen-presenting cells, antimicrobial peptides, and inflammatory cytokines in the CVL (p values <.05). POST women had higher vaginal pH and lower vaginal Lactobacilli (p values <.05) than PRE women. After vaginal E2 therapy, CV endpoints and ex vivo HIV replication in POST tissues were similar to those observed in PRE tissues. The CV mucosa in POST women is thinned and compromised, with increased HIV-target immune cells and decreased antimicrobial factors, being more susceptible to HIV infection. After POST women receive topical E2 treatment, mucosal endpoints are similar to PRE levels.
Collapse
Affiliation(s)
- Andrea Ries Thurman
- CONRAD Clinical Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Nazita Yousefieh
- CONRAD Clinical Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Neelima Chandra
- CONRAD Clinical Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Thomas Kimble
- CONRAD Clinical Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Susana Asin
- V.A. Medical Center, White River Junction, Vermont
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Christiane Rollenhagen
- V.A. Medical Center, White River Junction, Vermont
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Sharon M. Anderson
- CONRAD Clinical Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | | | | | | | | | | | - Tina Cunningham
- Center for Health Analytics and Discovery, Eastern Virginia Medical School, Norfolk, Virginia
| | - Sharon Hillier
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lorna Rabe
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Jill L. Schwartz
- CONRAD Clinical Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Gustavo F. Doncel
- CONRAD Clinical Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
47
|
Klatt NR, Cheu R, Birse K, Zevin AS, Perner M, Noël-Romas L, Grobler A, Westmacott G, Xie IY, Butler J, Mansoor L, McKinnon LR, Passmore JAS, Abdool Karim Q, Abdool Karim SS, Burgener AD. Vaginal bacteria modify HIV tenofovir microbicide efficacy in African women. Science 2017; 356:938-945. [DOI: 10.1126/science.aai9383] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/25/2017] [Accepted: 04/13/2017] [Indexed: 12/15/2022]
|
48
|
Sanders AP, Gennings C, Svensson K, Motta V, Mercado-Garcia A, Solano M, Baccarelli AA, Tellez-Rojo MM, Wright RO, Burris HH. Bacterial and cytokine mixtures predict the length of gestation and are associated with miRNA expression in the cervix. Epigenomics 2016; 9:33-45. [PMID: 27936911 DOI: 10.2217/epi-2016-0095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Bacterial vaginosis may lead to preterm birth through epigenetic programming of the inflammatory response, specifically via miRNA expression. METHODS We quantified bacterial 16S rRNA, cytokine mRNA and 800 miRNA from cervical swabs obtained from 80 women at 16-19 weeks' gestation. We generated bacterial and cytokine indices using weighted quantile sum regression and examined associations with miRNA and gestational age at delivery. RESULTS & DISCUSSION Each decile of the bacterial and cytokine indices was associated with shorter gestations (p < 0.005). The bacterial index was associated with miR-494, 371a, 4286, 185, 320e, 888 and 23a (p < 0.05). miR-494 remained significant after false discovery rate correction (q < 0.1). The cytokine index was associated with 27 miRNAs (p < 0.05; q < 0.01). CONCLUSION Future investigation into the role of bacterial vaginosis- and inflammation-associated miRNA and preterm birth is warranted.
Collapse
Affiliation(s)
- Alison P Sanders
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Chris Gennings
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Katherine Svensson
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Valeria Motta
- Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Department of Clinical Sciences & Community Health University of Milan - Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Adriana Mercado-Garcia
- Center for Evaluation Research & Surveys, National Institute of Public Health, Cuernavaca, Morelos 62100, Mexico
| | - Maritsa Solano
- Center for Evaluation Research & Surveys, National Institute of Public Health, Cuernavaca, Morelos 62100, Mexico
| | - Andrea A Baccarelli
- Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Martha M Tellez-Rojo
- Center for Evaluation Research & Surveys, National Institute of Public Health, Cuernavaca, Morelos 62100, Mexico
| | - Robert O Wright
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Heather H Burris
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Boston Children's Hospital & Harvard Medical School, Boston, MA, USA.,Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
49
|
Abstract
Bacterial vaginosis (BV) is the most commonly reported microbiological syndrome among women of childbearing age. BV is characterized by a shift in the vaginal flora from the dominant Lactobacillus to a polymicrobial flora. BV has been associated with a wide array of health issues, including preterm births, pelvic inflammatory disease, increased susceptibility to HIV infection, and other chronic health problems. A number of potential microbial pathogens, singly and in combinations, have been implicated in the disease process. The list of possible agents continues to expand and includes members of a number of genera, including Gardnerella, Atopobium, Prevotella, Peptostreptococcus, Mobiluncus, Sneathia, Leptotrichia, Mycoplasma, and BV-associated bacterium 1 (BVAB1) to BVAB3. Efforts to characterize BV using epidemiological, microscopic, microbiological culture, and sequenced-based methods have all failed to reveal an etiology that can be consistently documented in all women with BV. A careful analysis of the available data suggests that what we term BV is, in fact, a set of common clinical signs and symptoms that can be provoked by a plethora of bacterial species with proinflammatory characteristics, coupled to an immune response driven by variability in host immune function.
Collapse
|
50
|
Jais M, Younes N, Chapman S, Cu-Uvin S, Ghosh M. Reduced levels of genital tract immune biomarkers in postmenopausal women: implications for HIV acquisition. Am J Obstet Gynecol 2016; 215:324.e1-324.e10. [PMID: 27026477 DOI: 10.1016/j.ajog.2016.03.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Rates of HIV infections are increasing in older adults. Although it is known that the HIV/AIDS epidemics affects women disproportionately, little is known regarding immune functions in the genital tract of postmenopausal women, as relevant to HIV susceptibility. OBJECTIVE The objective of the study was to compare levels of female reproductive tract immune mediators that are important for HIV-associated immune responses as well as intrinsic anti-HIV activity in the cervical vaginal lavages collected from HIV-negative pre- and postmenopausal women. STUDY DESIGN Cervical vaginal lavage from 20 premenopausal and 20 postmenopausal women were assayed for interleukin-6, interleukin-8, tumor necrosis factor-α, secretory leukocyte protease inhibitor, elafin, human β-defensin-2, and macrophage inflammatory protein-3α using standard enzyme-linked immunosorbent assays. Anti-HIV activity of cervical-vaginal lavage was measured using TZM-bl indicator cells against HIV-1 IIIB and BaL. Whereas each postmenopausal woman provided only 1 sample, each premenopausal woman provided 3 samples, during proliferative, ovulatory, and secretory stages, based on menstrual dates. RESULTS We observed significantly lower levels of tumor necrosis factor-α, MIP-3α, secretory leukocyte protease inhibitor, elafin, and human β-defensin-2 in cervical vaginal lavage from postmenopausal women compared with premenopausal women. Inhibition of HIV-1 infection was observed for both pre- and postmenopausal women, but cervical vaginal lavage from postmenopausal women showed significantly higher inhibition against HIV-1 BaL after adjusting for total protein concentration, genital pH, and reproductive tract infections. No change in mediators or HIV inhibition was observed through the stages of menstrual cycle. In addition, we observed that postmenopausal women with reproductive tract infections had significantly higher levels of tumor necrosis factor-α and significantly lower levels of interleukin-8, which were not observed in premenopausal women. CONCLUSION Our findings suggest that female reproductive tract immune microenvironment is distinct in HIV-negative postmenopausal women. Further studies are needed to assess the risk of HIV acquisition/transmission in this population.
Collapse
Affiliation(s)
- Mariel Jais
- Department of Epidemiology and Biostatistics, The George Washington University, Washington, DC
| | - Naji Younes
- Department of Epidemiology and Biostatistics, The George Washington University, Washington, DC
| | - Stacey Chapman
- Department of Medicine, Alpert School of Medicine, Brown University, Providence, RI
| | - Susan Cu-Uvin
- Department of Obstetrics and Gynecology, Alpert School of Medicine, Brown University, Providence, RI
| | - Mimi Ghosh
- Department of Epidemiology and Biostatistics, The George Washington University, Washington, DC.
| |
Collapse
|