1
|
Hemmerling A, Mitchell CM, Demby S, Gebremichael M, Elsherbini J, Xu J, Xulu N, Shih J, Dong K, Govender V, Pillay V, Ismail N, Casillas G, Moodley J, Bergerat A, Brunner T, Liebenberg L, Ngcapu S, Mbano I, Lagenaur L, Parks TP, Ndung'u T, Kwon DS, Cohen CR. Effect of the vaginal live biotherapeutic LACTIN-V (Lactobacillus crispatus CTV-05) on vaginal microbiota and genital tract inflammation among women at high risk of HIV acquisition in South Africa: a phase 2, randomised, placebo-controlled trial. THE LANCET. MICROBE 2025:101037. [PMID: 40194532 DOI: 10.1016/j.lanmic.2024.101037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/13/2024] [Accepted: 10/30/2024] [Indexed: 04/09/2025]
Abstract
BACKGROUND Absence of vaginal lactobacilli and accompanying genital inflammation is associated with HIV acquisition. We aimed to assess how a vaginal live biotherapeutic containing Lactobacillus crispatus affects cervicovaginal microbiota and markers of HIV susceptibility in South African women. METHODS This randomised, placebo-controlled, phase 2 trial evaluated LACTIN-V (L crispatus CTV-05), a vaginal live biotherapeutic, compared with placebo in cisgender women in South Africa, aged 18-23 years, recruited at a community-based research clinic. Eligible participants with a Nugent score of 4-10 (indicating intermediate vaginal microbiota or bacterial vaginosis) completed 7 days of oral metronidazole and were randomly assigned (2:1) to LACTIN-V (2 × 109 colony forming units per dose) or placebo (the substrate alone) via an independently generated randomisation sequence. Pharmacists, participants, and investigators were masked to treatment assignment. The study product (or placebo) was dosed daily for 5 days in week 1, then twice per week for an additional 3 weeks. Adverse events were evaluated 4 weeks and 8 weeks after starting the study product. Vaginal swabs (for 16S rRNA sequencing of the vaginal microbiome) and cervicovaginal lavage (for Luminex analysis of immune markers) were collected before metronidazole treatment, before study product (or placebo) administration, and at the week 4 and week 8 follow-up visits. An endocervical cytobrush for flow cytometry analysis of immune cell populations (including CD3+CD4+ T cells, and presence of CCR5 and the activation markers CD38 or HLA-DR) was collected before study product use and at 4 weeks and 8 weeks after study product use. The coprimary outcomes for the trial were (1) safety and acceptability of LACTIN-V, as measured by number of adverse events and a validated questionnaire; (2) presence of a Lactobacillus-dominant vaginal microbial community by 16S rRNA gene sequencing at week 4 and week 8; and (3) comparison of change in genital tract inflammatory markers from before metronidazole treatment to week 4 and week 8 between groups. Safety analyses were done in the intention-to-treat population and efficacy analyses in a modified intent-to-treat population (ie, excluding one person assigned placebo who erroneously received LACTIN-V). This trial is completed and registered on ClinicalTrials.gov (NCT05022212). FINDINGS 45 Black South African women were randomly assigned to receive LACTIN-V (n=32) or placebo (n=13). One woman in each group discontinued the trial during the intervention and two women discontinued during the follow-up. No severe or serious adverse events were observed. Solicited adverse events occurred in 35 (78%) of 45 participants with no significant difference by group (risk ratio 1·17, 95% CI 0·79-1·75; p=0·44). All local solicited adverse events were mild. 32 (71%) of 45 participants strongly agreed or agreed they would use the product again. L crispatus dominant microbiomes were identified in 13 (41%) of 32 participants in the LACTIN-V group at week 4 and eight (26%) of 31 at week 8, compared with none at week 4 and one (9%) of 11 in week 8 in the placebo group (week 4 p=0·0088; week 8 p=0·40). The proportion of activated endocervical HIV target cells out of total T cells increased from after metronidazole treatment to week 4 in the placebo group (median log2 fold change 1·891, IQR 1·731-4·018) but not in the LACTIN-V group (1·062, 0·449-1·424; p=0·016). Changes in the concentrations of 13 immune markers from before metronidazole treatment to week 4 or week 8 were not significantly different by group. INTERPRETATION The use of LACTIN-V after metronidazole significantly increased vaginal L crispatus colonisation during 4 weeks of use, although this increase was transient, and women in the placebo group had an increase in endocervical CD4+ HIV target cells during recovery compared with the LACTIN-V group. These results show that vaginal colonisation with an L crispatus live biotherapeutic is possible in an African context, and that optimisation of this strategy might be a way to decrease risk for HIV. FUNDING US National Institute of Child Health and Human Development and US National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
- Anke Hemmerling
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Caroline M Mitchell
- Department of Obstetrics & Gynecology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Suuba Demby
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | | | - Joseph Elsherbini
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Jiawu Xu
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Nondumiso Xulu
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Johnathan Shih
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Krista Dong
- Harvard Medical School, Boston, MA, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA; FRESH Clinic South Africa, Umlazi, South Africa
| | | | | | - Nasreen Ismail
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Gardenia Casillas
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | | | - Agnes Bergerat
- Department of Obstetrics & Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Tess Brunner
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Lenine Liebenberg
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa; Center for Epidemic Response and Innovation, Stellenbosch University, Stellenbosch, South Africa; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Ian Mbano
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | | | | | - Thumbi Ndung'u
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Douglas S Kwon
- Harvard Medical School, Boston, MA, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA.
| | - Craig R Cohen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Murphy K, Shi Q, Hoover DR, Adimora AA, Alcaide ML, Brockmann S, Daubert E, Duggal P, Merenstein D, Dionne JA, Sheth AN, Keller MJ, Herold BC, Anastos K, Aouizerat B. Genetic predictors for bacterial vaginosis in women living with and at risk for HIV infection. Am J Reprod Immunol 2024; 91:e13845. [PMID: 38720636 PMCID: PMC11410097 DOI: 10.1111/aji.13845] [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: 08/31/2023] [Revised: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 08/29/2024] Open
Abstract
PROBLEM Bacterial vaginosis (BV) disproportionally impacts Black and Hispanic women, placing them at risk for HIV, sexually transmitted infections and preterm birth. It is unknown whether there are differences by genetic ancestry in BV risk or whether polymorphisms associated with BV risk differ by ancestry. METHODS Women's Interagency HIV Study (WIHS) participants with longitudinal Nugent scores were dichotomized as having (n = 319, Nugent 7-10) or not having BV (n = 367, Nugent 0-3). Genetic ancestry was defined by clustering of principal components from ancestry informative markers and further stratified by BV status. 627 single nucleotide polymorphisms (SNPs) across 41 genes important in mucosal defense were identified in the WIHS GWAS. A logistic regression analysis was adjusted for nongenetic predictors of BV and self-reported race/ethnicity to assess associations between genetic ancestry and genotype. RESULTS Self-reported race and genetic ancestry were associated with BV risk after adjustment for behavioral factors. Polymorphisms in mucosal defense genes including syndecans, cytokines and toll-like receptors (TLRs) were associated with BV in all ancestral groups. CONCLUSIONS The common association of syndecan, cytokine and TLR genes and the importance of immune function and inflammatory pathways in BV, suggests these should be targeted for further research on BV pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Kerry Murphy
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Quihu Shi
- School of Health Sciences and Practice, New York Medical College, Valhalla, New York, United States of America
| | - Donald R. Hoover
- Department of Statistics and Institute for Health, Health Care Policy and Aging Research Rutgers the State University of New Jersey, Piscataway, NJ, United States of America
| | - Adaora A. Adimora
- Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC. United States of America
| | - Maria L. Alcaide
- Department of Medicine, Obstetrics & Gynecology and Public Health, University of Miami Miller School of Medicine
| | - Susan Brockmann
- State University of New York, Health Sciences Center, Brooklyn, NY
| | | | - Priya Duggal
- Department of Epidemiology and International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | | | - Jodie A. Dionne
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anandi N. Sheth
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Marla J. Keller
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Betsy C. Herold
- Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Pediatrics, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Kathryn Anastos
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | | |
Collapse
|
3
|
Matassoli F, Cagigi A, Shen CH, Henry AR, Johnston TS, Schramm CA, Cottrell CA, Kalyuzhniy O, Spangler A, Eller L, Robb M, Eller M, Naluyima P, Kwong PD, Douek DC, Schief WR, Andrews SF, McDermott AB. High frequency of HIV precursor-target-specific B cells in sub-Saharan populations. Cell Rep 2023; 42:113450. [PMID: 38019653 PMCID: PMC10886445 DOI: 10.1016/j.celrep.2023.113450] [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: 06/05/2023] [Revised: 10/11/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
HIV gp120 engineered outer domain germline-targeting version 8 (eOD-GT8) was designed specifically to engage naive B cell precursors of VRC01-class antibodies. However, the frequency and affinity of naive B cell precursors able to recognize eOD-GT8 have been evaluated only in U.S. populations. HIV infection is disproportionally concentrated in sub-Saharan Africa, so we seek to characterize naive B cells able to recognize eOD-GT8 in sub-Saharan cohorts. We demonstrate that people from sub-Saharan Africa have a higher or equivalent frequency of naive B cells able to engage eOD-GT8 compared with people from the U.S. Genetically, the higher frequency of eOD-GT8-positive cells is accompanied by a higher level of naive B cells with gene signatures characteristic of the VRC01 class, as well as other CD4bs-directed antibodies. Our study demonstrates that vaccination with eOD-GT8 in sub-Saharan Africa could be successful at expanding and establishing a pool of CD4bs-directed memory B cells from naive precursors.
Collapse
Affiliation(s)
- Flavio Matassoli
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Alberto Cagigi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chen-Hsiang Shen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Timothy S Johnston
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chaim A Schramm
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher A Cottrell
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Oleksandr Kalyuzhniy
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Abby Spangler
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leigh Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Merlin Robb
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michael Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - William R Schief
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Sarah F Andrews
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Waltmann A, Thomas C, Duncan JA. The role of the genital microbiota in the acquisition and pathogenesis of sexually transmitted infections. Curr Opin Infect Dis 2023; 36:35-48. [PMID: 36729748 PMCID: PMC10500551 DOI: 10.1097/qco.0000000000000893] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW There are an estimated 374 million new sexually transmitted infections (STIs) worldwide every year. Our review article examines the current evidence of how STI acquisition, transmission, and pathogenesis is impacted upon by the genital microbiota, with a focus on epidemiological, biochemical, and immunological features. RECENT FINDINGS At least in women, a genital microbiota dominated by lactobacilli has long been considered optimal for reproductive health, while depletion of lactobacilli may lead to a genital microenvironment dominated by anaerobic pathogens, which can manifest clinically as bacterial vaginosis. Recent research efforts have characterized genital microbiota composition in greater resolution, sometimes at species-level, using proteomics, metabolomics, and deep sequencing. This has enhanced our understanding of how specific microbiota members influence acquisition or clinical manifestation of STI pathogen infection. Other advances include a steady, though still slow, increase in the number of studies that sought to determine the genital (penile or urethral) microbiota of males and how it may impact that of their female partners' genital microbiota and risk of STI acquisition. Altogether, these data enabled us to explore the concept that genital microbiota may be sexually transmitted and influence pathogenesis and clinical presentation of other STI. SUMMARY With STI infection rates increasing worldwide, it is important now more than ever to find novel STI prevention strategies. Understanding if and how the genital microbiota is a modifiable risk factor for STI transmission, acquisition, and clinical manifestation may prove to be an important strategy in our efforts to curb morbidity in at risk populations.
Collapse
Affiliation(s)
- Andreea Waltmann
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Institute for Global Health & Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cynthia Thomas
- Graduate Program, Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Joseph A Duncan
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Institute for Global Health & Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
5
|
Kamau E, Chaudhury S, Bolton JS, Slike BM, Jian N, Eller MA, Eller LA, Ake J, Robb ML, Krebs SJ, Bergmann-Leitner ES. Susceptibility to HIV-1 Acquisition linked to Malaria Exposure: A Case-control Study. Clin Infect Dis 2022; 75:1834-1837. [PMID: 35594548 DOI: 10.1093/cid/ciac384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
HIV and malaria infection rates overlap across sub-Saharan Africa, but factors influencing their co-occurrence are unclear. In a case-control study, we investigated whether malaria exposure increases risk of HIV-1 acquisition. Prior to seroconverting, HIV-positive cases had significantly higher malaria-associated antibodies compared to HIV-negative controls, linking malaria exposure to HIV-1 acquisition.
Collapse
Affiliation(s)
- Edwin Kamau
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Sidhartha Chaudhury
- Center for Enabling Capabilities, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jessica S Bolton
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,Biologics Research and Development, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Bonnie M Slike
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Ningbo Jian
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Michael A Eller
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Leigh Anne Eller
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Julie Ake
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Merlin L Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Shelly J Krebs
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Elke S Bergmann-Leitner
- Biologics Research and Development, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
6
|
Happel AU, Sivro A, Liebenberg L, Passmore JA, Mitchell CM. Considerations for Choosing Soluble Immune Markers to Determine Safety of Novel Vaginal Products. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:899277. [PMID: 36303630 PMCID: PMC9580790 DOI: 10.3389/frph.2022.899277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/11/2022] [Indexed: 11/29/2022] Open
Abstract
Several soluble cytokines have been associated with microbicide-induced cervicovaginal inflammation, non-optimal vaginal microbiota, and risk of HIV acquisition. Many of these biomarkers are used in preclinical assays to estimate the safety of vaginally applied products. However, there are currently no validated biomarkers to evaluate the safety of novel vaginal products in clinical trials. This hinders the rapid and rational selection of novel products being tested in first-in-human trials. We reviewed available literature to assess how best to select and measure soluble immune markers to determine product safety in first in human clinical trials of novel vaginal products.
Collapse
Affiliation(s)
- Anna-Ursula Happel
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Lenine Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Jo Ann Passmore
- 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 (CAPRISA), Durban, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Caroline M. Mitchell
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, United States
- *Correspondence: Caroline M. Mitchell
| |
Collapse
|
7
|
Lovett A, Seña AC, Macintyre AN, Sempowski GD, Duncan JA, Waltmann A. Cervicovaginal Microbiota Predicts Neisseria gonorrhoeae Clinical Presentation. Front Microbiol 2022; 12:790531. [PMID: 35222300 PMCID: PMC8867028 DOI: 10.3389/fmicb.2021.790531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/13/2021] [Indexed: 01/11/2023] Open
Abstract
Neisseria gonorrhoeae infection of the female lower genital tract can present with a spectrum of phenotypes ranging from asymptomatic carriage to symptomatic cervical inflammation, or cervicitis. The factors that contribute to the development of asymptomatic or symptomatic infections are largely uncharacterized. We conducted a pilot study to assess differences in the cervicovaginal microbial community of patients presenting with symptomatic vs. asymptomatic N. gonorrhoeae infections to a sexually transmitted infections (STI) clinic. DNA was isolated from cervicovaginal swab specimens from women who tested positive for N. gonorrhoeae infection using a clinical diagnostic nucleic acid amplification test. We performed deep sequencing of 16S ribosomal RNA gene amplicons, followed by microbiome analyses with QIIME, and species-specific real-time PCR to assess the composition of microbial communities cohabitating the lower genital tract with the infecting N. gonorrhoeae. Specimens collected from asymptomatic individuals with N. gonorrhoeae infection and no co-infection with Chlamydia trachomatis and/or Trichomonas vaginalis carried Lactobacillus-dominant microbial communities more frequently than symptomatic patients without co-infection. When compared to asymptomatic individuals, symptomatic women had microbial communities characterized by more diverse and heterogenous bacterial taxa, typically associated with bacterial vaginosis (BV) [Prevotella, Sneathia, Mycoplasma hominis, and Bacterial Vaginosis-Associated Bacterium-1 (BVAB1)/"Candidatus Lachnocurva vaginae"]. Both symptomatic and asymptomatic N. gonorrhoeae patients with additional STI co-infection displayed a BV-like microbial community. These findings suggest that Lactobacillus-dominant vaginal microbial community may protect individuals from developing symptoms during lower genital tract infection with N. gonorrhoeae.
Collapse
Affiliation(s)
- Angela Lovett
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Arlene C. Seña
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Andrew N. Macintyre
- School of Medicine and Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| | - Gregory D. Sempowski
- School of Medicine and Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| | - Joseph A. Duncan
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Andreea Waltmann
- Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,*Correspondence: Andreea Waltmann,
| |
Collapse
|
8
|
Wang R, Trent ME, Bream JH, Nilles TL, Gaydos CA, Carson KA, Coleman JS. Mycoplasma genitalium Infection Is Not Associated With Genital Tract Inflammation Among Adolescent and Young Adult Women in Baltimore, Maryland. Sex Transm Dis 2022; 49:139-144. [PMID: 34321450 PMCID: PMC8755577 DOI: 10.1097/olq.0000000000001524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Mycoplasma genitalium (MG) is a prevalent sexually transmitted infection, but little is known about the associated inflammatory signatures in the genital tract of adolescents and young adult women. METHODS Adolescents and young adult women aged 13 to 24 years were recruited. Demographic information, sexual behavior history, and medical history were collected. Vaginal swab samples were tested for MG, Chlamydia trachomatis, Neisseria gonorrhoeae, Trichomonas vaginalis, bacterial vaginosis, and measurement of 13 cytokines, chemokines, and antimicrobial proteins. Vaginal cytokine concentrations were compared by MG infection status. The strength of associations between multiple factors and MG infection was evaluated. RESULTS Of 215 participants, 16.7% (95% confidence interval [CI], 12.0%-22.4%) had MG infection. Inflammation was not associated with MG infection (P > 0.05). M. genitalium infection was associated with C. trachomatis infection (adjusted prevalence ratio [aPrR], 3.02; 95% CI, 1.69-5.39), bisexual behavior in the past 3 months (aPrR, 2.07; 95% CI, 1.18-3.64), genitourinary symptoms (aPrR, 2.06; 95% CI, 1.22-3.49), and self-reported Black race (aPrR, 3.53; 95% CI, 1.11-11.18). CONCLUSIONS Higher levels of genital tract cytokines were not associated with MG infection. C. trachomatis infection, bisexual behavior, self-reported Black race, and genitourinary symptoms were associated with an increased likelihood of MG infection.
Collapse
Affiliation(s)
- Runzhi Wang
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Maria E. Trent
- Department of Pediatrics, Adolescent /Young Adult Medicine Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jay H. Bream
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Immunology Training Program, Johns Hopkins School of Medicine
| | - Tricia L. Nilles
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Charlotte A. Gaydos
- Department of Medicine, Infectious Disease Division, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kathryn A. Carson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jenell S. Coleman
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
The Potential Role of FREM1 and Its Isoform TILRR in HIV-1 Acquisition through Mediating Inflammation. Int J Mol Sci 2021; 22:ijms22157825. [PMID: 34360591 PMCID: PMC8346017 DOI: 10.3390/ijms22157825] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
FREM1 (Fras-related extracellular matrix 1) and its splice variant TILRR (Toll-like interleukin-1 receptor regulator) have been identified as integral components of innate immune systems. The potential involvement of FREM1 in HIV-1 (human immunodeficiency virus 1) acquisition was suggested by a genome-wide SNP (single nucleotide polymorphism) analysis of HIV-1 resistant and susceptible sex workers enrolled in the Pumwani sex worker cohort (PSWC) in Nairobi, Kenya. The studies showed that the minor allele of a FREM1 SNP rs1552896 is highly enriched in the HIV-1 resistant female sex workers. Subsequent studies showed that FREM1 mRNA is highly expressed in tissues relevant to mucosal HIV-1 infection, including cervical epithelial tissues, and TILRR is a major modulator of many genes in the NF-κB signal transduction pathway. In this article, we review the role of FREM1 and TILRR in modulating inflammatory responses and inflammation, and how their influence on inflammatory responses of cervicovaginal tissue could enhance the risk of vaginal HIV-1 acquisition.
Collapse
|
10
|
Achilles SL, Meyn LA, Mhlanga FG, Matubu AT, Stoner KA, Beamer MA, Chirenje ZM, Hillier SL. Zim CHIC: A cohort study of immune changes in the female genital tract associated with initiation and use of contraceptives. Am J Reprod Immunol 2020; 84:e13287. [PMID: 32533883 PMCID: PMC7507197 DOI: 10.1111/aji.13287] [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: 02/20/2020] [Revised: 05/13/2020] [Accepted: 06/05/2020] [Indexed: 12/27/2022] Open
Abstract
Problem Contraceptive hormones are systemically active, potent, and likely to invoke biological responses other than known fertility regulation impacts. We hypothesized that initiation of depot medroxyprogesterone acetate (DMPA) would increase genital HIV‐target‐cells and soluble immune mediators compared with baseline and initiation of other contraceptive methods. Method of Study We collected cervical cytobrushes and cervicovaginal fluid from healthy Zimbabwean women aged 18‐34 to assess immune cell populations, cytokines, and innate anti‐HIV activity at baseline and after 30, 90, and 180 days use of DMPA (n = 38), norethisterone enanthate (n = 41), medroxyprogesterone acetate/estradiol cypionate (n = 36), levonorgestrel implant (n = 43), etonogestrel implant (n = 47), or copper intrauterine device (Cu‐IUD) (n = 45). Cells were quantified by flow cytometry, cytokines were detected by multiplex assays, and innate anti‐HIV activity was assessed by in vitro HIV challenge. Results Compared to baseline, the number of cervical HIV target cells (#CD4 cells P < .04 and #CD11c cells P < .04), the concentration of the inflammatory cytokine IL‐1β (P < .01), and the innate in vitro anti‐HIV activity (P < .001) significantly decreased following DMPA initiation. In Cu‐IUD users, genital HIV target cells increased (#CD4 cells P < .001, #CD4CCR5 cells P = .02, #CD4CD69 cells P < .001, #CD8CD69 P = .01, and #CD11c cells P = .003) at day 30 and resolved by day 180. IFN‐γ (P < .001), IL‐1β (P < .001), IL‐6 (P < .001), IL‐8 (P < .001), IL‐10 (P < .01), and RANTES (P < .001) were also significantly increased at day 30. Minimal alterations were observed following initiation of subdermal implantable contraceptives. Conclusions This head‐to‐head study compared six contraceptives and found increased HIV target cells and cervical inflammation temporally associated with Cu‐IUD initiation. Use of hormonal contraception, including DMPA, did not increase cervical HIV target cells or inflammation. Clinical Trial Number: NCT02038335
Collapse
Affiliation(s)
- Sharon L Achilles
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Family Planning Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Leslie A Meyn
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Family Planning Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Felix G Mhlanga
- Department of Obstetrics and Gynaecology, University of Zimbabwe College of Health Science, Harare, Zimbabwe
| | - Allen T Matubu
- Department of Obstetrics and Gynaecology, University of Zimbabwe College of Health Science, Harare, Zimbabwe
| | | | - May A Beamer
- Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Zvavahera M Chirenje
- Department of Obstetrics and Gynaecology, University of Zimbabwe College of Health Science, Harare, Zimbabwe
| | - Sharon L Hillier
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Family Planning Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Magee-Womens Research Institute, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Schiffer JT, Gottlieb SL. Biologic interactions between HSV-2 and HIV-1 and possible implications for HSV vaccine development. Vaccine 2019; 37:7363-7371. [PMID: 28958807 PMCID: PMC5867191 DOI: 10.1016/j.vaccine.2017.09.044] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022]
Abstract
Development of a safe and effective vaccine against herpes simplex virus type 2 (HSV-2) has the potential to limit the global burden of HSV-2 infection and disease, including genital ulcer disease and neonatal herpes, and is a global sexual and reproductive health priority. Another important potential benefit of an HSV-2 vaccine would be to decrease HIV infections, as HSV-2 increases the risk of HIV-1 acquisition several-fold. Acute and chronic HSV-2 infection creates ulcerations and draws dendritic cells and activated CD4+ T cells into genital mucosa. These cells are targets for HIV entry and replication. Prophylactic HSV-2 vaccines (to prevent infection) and therapeutic vaccines (to modify or treat existing infections) are currently under development. By preventing or modifying infection, an effective HSV-2 vaccine could limit HSV-associated genital mucosal inflammation and thus HIV risk. However, a vaccine might have competing effects on HIV risk depending on its mechanism of action and cell populations generated in the genital mucosa. In this article, we review biologic interactions between HSV-2 and HIV-1, consider HSV-2 vaccine development in the context of HIV risk, and discuss implications and research needs for future HSV vaccine development.
Collapse
Affiliation(s)
- Joshua T Schiffer
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, United States; Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, United States; University of Washington, Department of Medicine, Seattle, WA, United States.
| | - Sami L Gottlieb
- World Health Organization, Department of Reproductive Health and Research, Geneva, Switzerland
| |
Collapse
|
12
|
Defining characteristics of genital health in South African adolescent girls and young women at high risk for HIV infection. PLoS One 2019; 14:e0213975. [PMID: 30947260 PMCID: PMC6448899 DOI: 10.1371/journal.pone.0213975] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 03/05/2019] [Indexed: 01/24/2023] Open
Abstract
The genital tract of African women has been shown to differ from what is currently accepted as ‘normal’, defined by a pH≤4.5 and lactobacilli-dominated microbiota. Adolescent girls and young women (AGYW) from sub-Saharan Africa are at high risk for HIV, and we hypothesized that specific biological factors are likely to be influential. This study aimed to compare characteristics of vaginal health in HIV-negative AGYW (16-22-years-old), from two South African communities, to international norms. We measured plasma hormones, vaginal pH, presence of BV (Nugent scoring), sexually transmitted infections (multiplex PCR for Chlamydia trachomatis, Neisseria gonorrhoea, Trichomonas vaginalis, Mycoplasma genitalium) and candidiasis (Gram stain) in AGYW (n = 298) from Cape Town and Soweto. Cervicovaginal microbiota was determined by 16S pyrosequencing; 44 genital cytokines were measured by Luminex; and cervical T-cell activation/proliferation (CCR5, HLA-DR, CD38, Ki67) was measured by multiparametric flow cytometry. 90/298 (30.2%) AGYW were negative for BV, candidiasis and bacterial STIs. L. crispatus and L. iners were the dominant bacteria in cervicovaginal swabs, and the median vaginal pH was 4.7. AGYW with L. crispatus-dominant microbiota (42.4%) generally had the lowest cytokine concentrations compared to women with more diverse microbiota (34/44 significantly upregulated cytokines). Frequencies of CCR5+CD4+ T-cells co-expressing CD38 and HLA-DR correlated positively with interleukin (IL)-6, TNF-α, GRO-α, macrophage inflammatory protein (MIP)-1α, and IL-9. While endogenous oestrogen had an immune-dampening effect on IL-6, TNF-related apoptosis-inducing ligand (TRAIL) and IL-16, injectable hormone contraceptives (DMPA and Net-EN) were associated with significantly lower endogenous hormone concentrations (p<0.0001 for oestrogen and progesterone) and upregulation of 34/44 cytokines. Since genital inflammation and the presence of activated CD4+ T cells in the genital tract have been implicated in increased HIV risk in South African women, the observed high levels of genital cellular activation and cytokines from AGYW may point towards biological factors increasing HIV risk in this region.
Collapse
|
13
|
Deese J, Pradhan S, Goetz H, Morrison C. Contraceptive use and the risk of sexually transmitted infection: systematic review and current perspectives. Open Access J Contracept 2018; 9:91-112. [PMID: 30519127 PMCID: PMC6239113 DOI: 10.2147/oajc.s135439] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Purpose Evidence on the association between contraceptive use and risk of sexually transmitted infections (STIs) and bacterial vaginosis (BV) is lacking, with few prospective studies. We systematically reviewed the last 10 years' evidence on the association between contraception and STI/BV, building on the most recent systematic reviews published in 2006 and 2009. Methods We searched the MEDLINE and POPLINE databases for peer-reviewed articles p ublished between January 1, 2008 and January 31, 2018 reporting prospective studies that assessed the association between contraceptive use and incident STI and/or incident or recurrent BV. Results We identified 33 articles that evaluated combined oral contraceptives (COC), depot medroxyprogesterone acetate (DMPA), the copper intrauterine device (Cu-IUD), the levonorgestrel intrauterine system (LNG-IUS) and other methods. The strength of the evidence for many specific contraceptive method/STI associations is limited by few prospective studies with comparably defined exposures and outcomes. Available data suggest no association of COCs and Neisseria gonorrhoeae, Trichomonas vaginalis, HSV-2 or syphilis, and mixed evidence on the association with HPV, Chlamydia trachomatis, and BV. For DMPA, none of the studies identified found an association with N. gonorrhoeae or syphilis, and data on C. trachomatis, T. vaginalis, HPV and BV were mixed. Two large studies showed a highly clinically significant increased risk of HSV-2 infection with DMPA use. Data on the effect of Cu-IUD and the LNG-IUS on the acquisition of C. trachomatis, N. gonorrhoeae and T. vaginalis are sparse, and data on HPV and BV are mixed. Conclusion Few data are available from prospective studies, including randomized trials, to draw strong conclusions about the relationships between contraceptive methods and specific STIs. The overall evidence on the association between contraceptive use and STI/BV risk is limited by the lack of any randomized trials, few published prospective studies designed to analyze these associations, wide variability in exposure definitions and comparator groups, potential for confounding due to inaccurate sexual behavior data, differential confounder adjustment and differences in study populations and sizes. Despite these limitations, new evidence is supportive of a significantly increased risk of HSV-2 infection among DMPA users which warrants additional research to better understand this association.
Collapse
Affiliation(s)
- Jennifer Deese
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA,
| | - Subarna Pradhan
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA,
| | - Hannah Goetz
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA,
| | - Charles Morrison
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA,
| |
Collapse
|
14
|
Morrison CS, Fichorova R, Chen PL, Kwok C, Deese J, Yamamoto H, Anderson S, Chipato T, Salata R, Doncel GF. A Longitudinal Assessment of Cervical Inflammation and Immunity Associated with HIV-1 Infection, Hormonal Contraception, and Pregnancy. AIDS Res Hum Retroviruses 2018; 34:889-899. [PMID: 30047279 DOI: 10.1089/aid.2018.0022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hormonal contraception (HC), particularly injectable depot-medroxyprogesterone acetate (DMPA), has been associated with increased HIV acquisition and higher levels of cervical regulated upon activation, normal T-cell expressed, and secreted (RANTES), also associated with HIV seroconversion. Longitudinal changes in cervical immunity associated with DMPA and combined oral contraceptives (COCs) have not been studied. Cervical samples from 216 HIV seroconverters in Uganda and Zimbabwe with matched samples from 727 HIV-uninfected controls were collected at two quarterly visits before (t - 2, t - 1), at (t0), and two visits following (t + 1, t + 2) HIV seroconversion and corresponding visits for HIV-negative controls. We measured 10 biomarkers of inflammation and immunity and used generalized linear models to estimate and compare biomarker levels across HIV status, contraceptive, and pregnancy groups. Biomarkers remained relatively stable across visits for controls, while in HIV-infected women cervical immunity started to change before seroconversion with RANTES and BD-2 increased and secretory leukocyte protease inhibitor (SLPI) decreased at t - 1 and continued to change at t0 with ICAM-1 up and IL-8 down and with more biomarkers after seroconversion (IL-1β, IL-6, MIP-3α, VEGF, and IL-1RA down and IL-1RA:IL-1β ratio up). In multivariable analyses, seroconverters had higher BD-2 at t - 1, higher RANTES and lower SLPI from t - 1 through t + 2, and lower IL-8 and IL-1RA at and/or after seroconversion compared to nonseroconverters. Compared to non-HC users, DMPA users had higher RANTES at all visits and lower BD-2 at t - 2 through t0, while COC users and pregnant women had higher IL-8 and SLPI at all visits; COC users also had lower BD-2 preseroconversion; pregnant women had lower RANTES at t0 - t + 2. Longitudinal patterns of cervical immunity differ between HIV seroconverters and HIV-negative women; seroconverters demonstrate increased RANTES and decreased SLPI starting before and continuing postseroconversion. Furthermore, these patterns are differentially regulated by DMPA, COC, and pregnancy.
Collapse
Affiliation(s)
| | - Raina Fichorova
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pai-Lien Chen
- Global Health, Population and Nutrition, FHI 360, Durham, North Carolina
| | - Cynthia Kwok
- Global Health, Population and Nutrition, FHI 360, Durham, North Carolina
| | - Jennifer Deese
- Global Health, Population and Nutrition, FHI 360, Durham, North Carolina
| | - Hidemi Yamamoto
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sharon Anderson
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Arlington, Virginia
| | - Tsungai Chipato
- Department of Obstetrics and Gynaecology, University of Zimbabwe, Harare, Zimbabwe
| | - Robert Salata
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Gustavo F. Doncel
- CONRAD, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
15
|
Zalenskaya IA, Chandra N, Yousefieh N, Fang X, Adedipe OE, Jackson SS, Anderson SM, Mauck CK, Schwartz JL, Thurman AR, Doncel GF. Use of contraceptive depot medroxyprogesterone acetate is associated with impaired cervicovaginal mucosal integrity. J Clin Invest 2018; 128:4622-4638. [PMID: 30222141 DOI: 10.1172/jci120583] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Injectable depot medroxyprogesterone acetate (DMPA) is one of the most popular contraception methods in areas of high HIV seroprevalence. Evidence is accumulating that use of DMPA might be associated with an increased risk of HIV-1 acquisition by women; however, mechanisms of this association are not completely understood. The goal of this study was to gain insight into mechanisms underlying the possible link between use of DMPA and risk of HIV-1 acquisition, exploring transcription profiling of ectocervical tissues. METHODS Healthy women received either DMPA (n = 31) or combined oral contraceptive (COC), which has not been linked to an increased risk of HIV acquisition (n = 32). We conducted a comparative microarray-based whole-genome transcriptome profiling of human ectocervical tissues before and after 6 weeks of hormonal contraception use. RESULTS The analysis identified that expression of 235 and 76 genes was significantly altered after DMPA and COC use, respectively. The most striking effect of DMPA, but not COC, was significantly altered expression (mostly downregulation) of many genes strategically involved in the maintenance of mucosal barrier function; the alterations, as indicated by Ingenuity Pathway Analysis (IPA), were most likely due to the DMPA-induced estrogen deficiency. Furthermore, IPA predicted that transcriptome alterations related to ectocervical immune responses were in general compatible with an immunosuppressive effect of DMPA, but, in some women, also with an inflammatory-like response. CONCLUSION Our results suggest that impairment of cervicovaginal mucosal integrity in response to DMPA administration is an important mechanism contributing to the potential increased risk of HIV-1 acquisition in DMPA users. TRIAL REGISTRATION ClinicalTrials.gov NCT01421368. FUNDING This study was supported by the United States Agency for International Development (USAID) under Cooperative Agreement GPO-A-00-08-00005-00.
Collapse
Affiliation(s)
| | - Neelima Chandra
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | - Xi Fang
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | | | | | | | - Jill L Schwartz
- CONRAD, Eastern Virginia Medical School, Arlington, Virginia, USA
| | | | - Gustavo F Doncel
- CONRAD, Eastern Virginia Medical School, Norfolk, Virginia, USA.,CONRAD, Eastern Virginia Medical School, Arlington, Virginia, USA
| |
Collapse
|
16
|
Abstract
Cervicovaginal microbiota play a critical role in women's health and reproductive outcomes. Despite being one of the simplest commensal bacterial communities in the human body, we are only beginning to appreciate its complex dynamic nature and important role in host immune modulation. In this review, we discuss the "optimal" cervicovaginal bacterial community composition, the impact of microbiota on gynecologic and obstetric outcomes, and the hurdles to developing a deeper mechanistic understanding of the function of the cervicovaginal microbiome. We then describe efforts to durably alter microbial composition in this compartment by promotion of Lactobacillus colonization with probiotics, modulation of vaginal pH, hormonal administration, and the eradication of pathogenic bacteria with antibiotics. Finally, we draw on lessons learned from the deeply investigated gut microbiome to suggest future avenues of research into host-pathogen interactions in the female genital tract.
Collapse
|
17
|
Jaumdally SZ, Picton A, Tiemessen CT, Paximadis M, Jaspan HB, Gamieldien H, Masson L, Coetzee D, Williamson AL, Little F, Gumbi PP, Passmore JAS. CCR5 expression, haplotype and immune activation in protection from infection in HIV-exposed uninfected individuals in HIV-serodiscordant relationships. Immunology 2017; 151:464-473. [PMID: 28398593 DOI: 10.1111/imm.12743] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/16/2017] [Accepted: 03/24/2017] [Indexed: 12/01/2022] Open
Abstract
Several host factors have been implicated in resistance to HIV infection in individuals who remain HIV-seronegative despite exposure. In a cohort of HIV-serodiscordant heterosexual couples, we investigated interactions between systemic inflammation and T-cell activation in resistance to HIV infection. Males and females in stable long-term relationships with either HIV-infected or uninfected partners were recruited, blood T-cell activation (CD38, HLA-DR, CCR5 and Ki67) and plasma cytokine concentrations were evaluated. The HIV-negative exposed individuals had significantly lower frequencies of CCR5+ CD4+ and CD8+ T cells than unexposed individuals. Mean fluorescence intensity of CCR5 expression on CD4+ T cells was significantly lower in HIV-negative exposed than unexposed individuals. Protective CCR5 haplotypes (HHA/HHF*2, HHF*2/HHF*2, HHC/HHF*2, HHA/HHA, HHA/HHC and HHA/HHD) tended to be over-represented in exposed compared with unexposed individuals (38% versus 28%, P = 0·58) whereas deleterious genotypes (HHC/HHD, HHC/HHE, HHD/HHE, HHD/HHD and HHE/HHE) were under-represented (26% versus 44%; P = 0·16). Plasma concentrations of interleukin-2 (P = 0·02), interferon-γ (P = 0·05) and granulocyte-macrophage colony-stimulating factor (P = 0·006) were lower in exposed compared with unexposed individuals. Activation marker expression and systemic cytokine concentrations were not influenced by gender. We conclude that the dominant signature of resistance to HIV infection in this cohort of exposed but uninfected individuals was lower T-cell CCR5 expression and plasma cytokine concentrations.
Collapse
Affiliation(s)
- Shameem Z Jaumdally
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,NRF-DST Centre of Excellence in HIV Prevention, CAPRISA, Durban, South Africa
| | - Anabela Picton
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline T Tiemessen
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maria Paximadis
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Heather B Jaspan
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hoyam Gamieldien
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Lindi Masson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,NRF-DST Centre of Excellence in HIV Prevention, CAPRISA, Durban, South Africa
| | - David Coetzee
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,National Health Laboratory Service, Cape Town, South Africa
| | - Francesca Little
- Department of Statistical Sciences, University of Cape Town, Cape Town, South Africa
| | - Pamela P Gumbi
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,NRF-DST Centre of Excellence in HIV Prevention, CAPRISA, Durban, South Africa
| | - Jo-Ann S Passmore
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,NRF-DST Centre of Excellence in HIV Prevention, CAPRISA, Durban, South Africa.,National Health Laboratory Service, Cape Town, South Africa
| |
Collapse
|
18
|
Houlihan CF, Baisley K, Bravo IG, Kapiga S, de Sanjosé S, Changalucha J, Ross DA, Hayes RJ, Watson-Jones D. Rapid acquisition of HPV around the time of sexual debut in adolescent girls in Tanzania. Int J Epidemiol 2016; 45:762-773. [PMID: 26944311 PMCID: PMC5005945 DOI: 10.1093/ije/dyv367] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND No reports exist on genotype-specific human papillomavirus (HPV) acquisition in girls after first sex in sub-Saharan Africa, despite high HPV prevalence and cervical cancer incidence. METHODS We followed 503 HP-unvaccinated girls aged 15-16 years in Mwanza, Tanzania, 3-monthly for 18 months with interviews and self-administered vaginal swabs. Swabs were tested for 13 higHRisk and 24 low-risk HPV genotypes. Incidence, clearance and duration of overall HPV and genotype-specific infections were calculated and associated factors evaluated. RESULTS A total of 106 participants reported first sex prior to enrolment (N = 29) or during follow-up (N = 77). One was HIV-positive at the final visit. The remaining 105 girls contributed 323 adequate specimens. Incidence of any new HPV genotype was 225/100 person-years (pys), and incidence of vaccine types HPV-6, -11, -16 and -18 were 12, 2, 2 and 7/100 pys, respectively. Reporting sex in the past 3 months and knowing the most recent sexual partner for a longer period before sex were associated with HPV acquisition. Median time from reported sexual debut to first HPVinfection was 5 months, and infection duration was 6 months. CONCLUSION This is the first description of HPV acquisition after first sex in sub-Saharan Africa where the incidence of cervical cancer is amongst the highest in the world. HPV incidence was very high after first sex, including some vaccine genotypes, and infection duration was short. This very high HPV incidence may help explain high cervical cancer rates, and supports recommendations that the HPV vaccine should be given to girls before first sex.
Collapse
Affiliation(s)
- Catherine F Houlihan
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK Mwanza Intervention Trials Unit, Mwanza, Tanzania
| | - Kathy Baisley
- MRC Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Ignacio G Bravo
- Unit of Infections and Cancer, Institut Català d'Oncologia, Barcelona, Spain
| | - Saidi Kapiga
- Mwanza Intervention Trials Unit, Mwanza, Tanzania MRC Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Silvia de Sanjosé
- Unit of Infections and Cancer, Institut Català d'Oncologia, Barcelona, Spain CIBER ESP, Barcelona, Spain
| | | | - David A Ross
- MRC Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Richard J Hayes
- MRC Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Deborah Watson-Jones
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK Mwanza Intervention Trials Unit, Mwanza, Tanzania
| |
Collapse
|
19
|
Hughes SM, Shu Z, Levy CN, Ferre AL, Hartig H, Fang C, Lentz G, Fialkow M, Kirby AC, Adams Waldorf KM, Veazey RS, Germann A, von Briesen H, McElrath MJ, Dezzutti CS, Sinclair E, Baker CAR, Shacklett BL, Gao D, Hladik F. Cryopreservation of Human Mucosal Leukocytes. PLoS One 2016; 11:e0156293. [PMID: 27232996 PMCID: PMC4883784 DOI: 10.1371/journal.pone.0156293] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/11/2016] [Indexed: 01/09/2023] Open
Abstract
Background Understanding how leukocytes in the cervicovaginal and colorectal mucosae respond to pathogens, and how medical interventions affect these responses, is important for developing better tools to prevent HIV and other sexually transmitted infections. An effective cryopreservation protocol for these cells following their isolation will make studying them more feasible. Methods and Findings To find an optimal cryopreservation protocol for mucosal mononuclear leukocytes, we compared cryopreservation media and procedures using human vaginal leukocytes and confirmed our results with endocervical and colorectal leukocytes. Specifically, we measured the recovery of viable vaginal T cells and macrophages after cryopreservation with different cryopreservation media and handling procedures. We found several cryopreservation media that led to recoveries above 75%. Limiting the number and volume of washes increased the fraction of cells recovered by 10–15%, possibly due to the small cell numbers in mucosal samples. We confirmed that our cryopreservation protocol also works well for both endocervical and colorectal leukocytes. Cryopreserved leukocytes had slightly increased cytokine responses to antigenic stimulation relative to the same cells tested fresh. Additionally, we tested whether it is better to cryopreserve endocervical cells on the cytobrush or in suspension. Conclusions Leukocytes from cervicovaginal and colorectal tissues can be cryopreserved with good recovery of functional, viable cells using several different cryopreservation media. The number and volume of washes has an experimentally meaningful effect on the percentage of cells recovered. We provide a detailed, step-by-step protocol with best practices for cryopreservation of mucosal leukocytes.
Collapse
Affiliation(s)
- Sean M. Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Zhiquan Shu
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- School of Mechanical and Materials Engineering, Washington State University, Everett, Washington, United States of America
| | - Claire N. Levy
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - April L. Ferre
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Heather Hartig
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Cifeng Fang
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
| | - Gretchen Lentz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Michael Fialkow
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Anna C. Kirby
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Anja Germann
- Division of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach/Saar, Germany
| | - Hagen von Briesen
- Division of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach/Saar, Germany
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Charlene S. Dezzutti
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth Sinclair
- Division of Experimental Medicine, Department of Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | - Chris A. R. Baker
- Division of Experimental Medicine, Department of Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
- Core Immunology Lab, University of California San Francisco, San Francisco, California, United States of America
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
- Department of Medicine, Division of Infectious Diseases, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DG); (FH)
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DG); (FH)
| |
Collapse
|
20
|
Biomarkers and biometric measures of adherence to use of ARV-based vaginal rings. J Int AIDS Soc 2016; 19:20746. [PMID: 27142091 PMCID: PMC4854848 DOI: 10.7448/ias.19.1.20746] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 03/21/2016] [Accepted: 03/31/2016] [Indexed: 11/13/2022] Open
Abstract
Introduction Poor adherence to product use has been observed in recent trials of antiretroviral (ARV)-based oral and vaginal gel HIV prevention products, resulting in an inability to determine product efficacy. The delivery of microbicides through vaginal rings is widely perceived as a way to achieve better adherence but vaginal rings do not eliminate the adherence challenges exhibited in clinical trials. Improved objective measures of adherence are needed as new ARV-based vaginal ring products enter the clinical trial stage. Methods To identify technologies that have potential future application for vaginal ring adherence measurement, a comprehensive literature search was conducted that covered a number of biomedical and public health databases, including PubMed, Embase, POPLINE and the Web of Science. Published patents and patent applications were also searched. Technical experts were also consulted to gather more information and help evaluate identified technologies. Approaches were evaluated as to feasibility of development and clinical trial implementation, cost and technical strength. Results Numerous approaches were identified through our landscape analysis and classified as either point measures or cumulative measures of vaginal ring adherence. Point measurements are those that give a measure of adherence at a particular point in time. Cumulative measures attempt to measure ring adherence over a period of time. Discussion Approaches that require modifications to an existing ring product are at a significant disadvantage, as this will likely introduce additional regulatory barriers to the development process and increase manufacturing costs. From the point of view of clinical trial implementation, desirable attributes would be high acceptance by trial participants, and little or no additional time or training requirements on the part of participants or clinic staff. We have identified four promising approaches as being high priority for further development based on the following measurements: intracellular drug levels, drug levels in hair, the accumulation of a vaginal analyte that diffuses into the ring, and the depletion of an intrinsic ring constituent. Conclusions While some approaches show significant promise over others, it is recommended that a strategy of using complementary biometric and behavioural approaches be adopted to best understand participants’ adherence to ARV-based ring products in clinical trials.
Collapse
|
21
|
Shu Z, Hughes SM, Fang C, Huang J, Fu B, Zhao G, Fialkow M, Lentz G, Hladik F, Gao D. A study of the osmotic characteristics, water permeability, and cryoprotectant permeability of human vaginal immune cells. Cryobiology 2016; 72:93-9. [PMID: 26976225 PMCID: PMC4829375 DOI: 10.1016/j.cryobiol.2016.03.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 12/03/2022]
Abstract
Cryopreservation of specimens taken from the genital tract of women is important for studying mucosal immunity during HIV prevention trials. However, it is unclear whether the current, empirically developed cryopreservation procedures for peripheral blood cells are also ideal for genital specimens. The optimal cryopreservation protocol depends on the cryobiological features of the cells. Thus, we obtained tissue specimens from vaginal repair surgeries, isolated and flow cytometry-purified immune cells, and determined fundamental cryobiological characteristics of vaginal CD3+ T cells and CD14+ macrophages using a microfluidic device. The osmotically inactive volumes of the two cell types (Vb) were determined relative to the initial cell volume (V0) by exposing the cells to hypotonic and hypertonic saline solutions, evaluating the equilibrium volume, and applying the Boyle van't Hoff relationship. The cell membrane permeability to water (Lp) and to four different cryoprotective agent (CPA) solutions (Ps) at room temperature were also measured. Results indicated Vb values of 0.516 V0 and 0.457 V0 for mucosal T cells and macrophages, respectively. Lp values at room temperature were 0.196 and 0.295 μm/min/atm for T cells and macrophages, respectively. Both cell types had high Ps values for the three CPAs, dimethyl sulfoxide (DMSO), propylene glycol (PG) and ethylene glycol (EG) (minimum of 0.418 × 10−3 cm/min), but transport of the fourth CPA, glycerol, occurred 50–150 times more slowly. Thus, DMSO, PG, and EG are better options than glycerol in avoiding severe cell volume excursion and osmotic injury during CPA addition and removal for cryopreservation of human vaginal immune cells.
Collapse
Affiliation(s)
- Zhiquan Shu
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA; School of Mechanical and Materials Engineering, Washington State University, Everett, WA 98201, USA
| | - Sean M Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Cifeng Fang
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Jinghua Huang
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA; College of Information Technology, Beijing Union University, Beijing 100101, China
| | - Baiwen Fu
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA; College of Information Technology, Beijing Union University, Beijing 100101, China
| | - Gang Zhao
- Department of Electronic Science & Technology, University of Science and Technology of China, Hefei 230027, China
| | - Michael Fialkow
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Gretchen Lentz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
22
|
Kaul R, Prodger J, Joag V, Shannon B, Yegorov S, Galiwango R, McKinnon L. Inflammation and HIV Transmission in Sub-Saharan Africa. Curr HIV/AIDS Rep 2016; 12:216-22. [PMID: 25877253 DOI: 10.1007/s11904-015-0269-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
While the per-contact risk of sexual HIV transmission is relatively low, it is fourfold higher in sub-Saharan Africa, and this may partly explain the major global disparities that exist in HIV prevalence. Genital immune parameters are key determinants of HIV transmission risk, including epithelial integrity and the presence of highly HIV-susceptible intraepithelial or submucosal CD4+ T cell target cells. Biological parameters that may enhance mucosal HIV susceptibility in highly HIV-affected regions of sub-Saharan Africa include increased levels of mucosal inflammation, which can affect both epithelial integrity and target cell availability, as well as the increased mucosal surface area that is afforded by an intact foreskin, contraceptive choices, and intravaginal practices. There are multifactorial causes for increased mucosal inflammation, with the prevalence and nature of common co-infections being particularly relevant.
Collapse
Affiliation(s)
- Rupert Kaul
- Departments of Medicine and Immunology, University of Toronto, Medical Sciences Building Rm. 6356, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada,
| | | | | | | | | | | | | |
Collapse
|
23
|
Ait-Lounis A, Laraba-Djebari F. TNF-alpha modulates adipose macrophage polarization to M1 phenotype in response to scorpion venom. Inflamm Res 2015; 64:929-36. [PMID: 26403661 DOI: 10.1007/s00011-015-0876-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/07/2015] [Accepted: 09/09/2015] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE We previously reported that Androctonus australis hector (Aah) venom and its toxic fraction affect adipose tissue metabolism. However, the contribution of immune system and the role of adipose tissue macrophages (ATMs) in the progression of inflammation induced by scorpion venom remain largely unknown. METHODS Here we evaluate the capacity of the toxic fraction of Aah venom (FTox-G50) to induce the expression of M1 and M2 markers genes on adipose tissue and isolated stromal vascular cells (SVC). Quantitative real-time PCR was performed on the SVC 24 h after FTox-G50 venom injection to assess the gene expressions of IL12p40, IL23, and other macrophages-associated markers. RESULTS We found that ATM from FTox-G50-venom-injected mice markedly increased the expressions of IL-12p40 and IL-23. Furthermore, the expression of nitric oxide synthase 2 (an M1 marker) was up-regulated, but the expression of Arginase1 (an M2 marker) was not. Systemic injection of a chemical inhibitor directed against TNF-α binding reduced the expression of inflammatory M1 macrophage markers and the MAPKpk2 gene, a key mediator of inflammatory signaling. CONCLUSION These results indicate that TNF-α is a physiological regulator of inflammation and macrophage activation induced by scorpion venom.
Collapse
Affiliation(s)
- Aouatef Ait-Lounis
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, BP 32 El-Alia, Bab Ezzouar, Algiers, Algeria
| | - Fatima Laraba-Djebari
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, BP 32 El-Alia, Bab Ezzouar, Algiers, Algeria.
| |
Collapse
|
24
|
Moodie Z, Metch B, Bekker LG, Churchyard G, Nchabeleng M, Mlisana K, Laher F, Roux S, Mngadi K, Innes C, Mathebula M, Allen M, Bentley C, Gilbert PB, Robertson M, Kublin J, Corey L, Gray GE. Continued Follow-Up of Phambili Phase 2b Randomized HIV-1 Vaccine Trial Participants Supports Increased HIV-1 Acquisition among Vaccinated Men. PLoS One 2015; 10:e0137666. [PMID: 26368824 PMCID: PMC4569275 DOI: 10.1371/journal.pone.0137666] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/19/2015] [Indexed: 12/14/2022] Open
Abstract
Background The Phase 2b double-blinded, randomized Phambili/HVTN 503 trial evaluated safety and efficacy of the MRK Ad5 gag/pol/nef subtype B HIV-1 preventive vaccine vs placebo in sexually active HIV-1 seronegative participants in South Africa. Enrollment and vaccinations stopped and participants were unblinded but continued follow-up when the Step study evaluating the same vaccine in the Americas, Caribbean, and Australia was unblinded for non-efficacy. Final Phambili analyses found more HIV-1 infections amongst vaccine than placebo recipients, impelling the HVTN 503-S recall study. Methods HVTN 503-S sought to enroll all 695 HIV-1 uninfected Phambili participants, provide HIV testing, risk reduction counseling, physical examination, risk behavior assessment and treatment assignment recall. After adding HVTN 503-S data, HIV-1 infection hazard ratios (HR vaccine vs. placebo) were estimated by Cox models. Results Of the 695 eligible, 465 (67%) enrolled with 230 from the vaccine group and 235 from the placebo group. 38% of the 184 Phambili dropouts were enrolled. Enrollment did not differ by treatment group, gender, or baseline HSV-2. With the additional 1286 person years of 503-S follow-up, the estimated HR over Phambili and HVTN 503-S follow-up was 1.52 (95% CI 1.08–2.15, p = 0.02, 82 vaccine/54 placebo infections). The HR was significant for men (HR = 2.75, 95% CI 1.49, 5.06, p = 0.001) but not for women (HR = 1.12, 95% CI 0.73, 1.72, p = 0.62). Conclusion The additional follow-up from HVTN 503-S supported the Phambili finding of increased HIV-1 acquisition among vaccinated men and strengthened the evidence of lack of vaccine effect among women. Trial Registration clinicaltrials.gov NCT00413725 SA National Health Research Database DOH-27-0207-1539
Collapse
Affiliation(s)
- Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail:
| | - Barbara Metch
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Linda-Gail Bekker
- Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | - Gavin Churchyard
- Aurum Institute for Health Research, Johannesburg, South Africa
- School of Public Health, University of Witwatersrand, Johannesburg, South Africa
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Maphoshane Nchabeleng
- Mecru Clinical Research Unit, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - Koleka Mlisana
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal and National Health Laboratory Service, Durban, South Africa
| | - Fatima Laher
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Surita Roux
- Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | - Kathryn Mngadi
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal and National Health Laboratory Service, Durban, South Africa
| | - Craig Innes
- Aurum Institute Clinical Research Site, Klerksdorp, South Africa
| | - Matsontso Mathebula
- Mecru Clinical Research Unit, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - Mary Allen
- Vaccine Research Program, Division of AIDS, NIAID, NIH, Rockville, United States of America
| | - Carter Bentley
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Michael Robertson
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - James Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Glenda E. Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
25
|
Schistosoma mansoni Infection in Ugandan Men Is Associated with Increased Abundance and Function of HIV Target Cells in Blood, but Not the Foreskin: A Cross-sectional Study. PLoS Negl Trop Dis 2015; 9:e0004067. [PMID: 26335139 PMCID: PMC4559468 DOI: 10.1371/journal.pntd.0004067] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/18/2015] [Indexed: 01/01/2023] Open
Abstract
Background Schistosoma mansoni infection has been associated with an increased HIV prevalence in humans and SHIV incidence in primate models. We hypothesized that immune activation from this gastrointestinal mucosa infection would increase highly HIV-susceptible CD4 T cell subsets in the blood and the foreskin through common mucosal homing. Methodology/Principal Findings Foreskin tissue and blood were obtained from 34 HIV- and malaria-uninfected Ugandan men who volunteered for elective circumcision, 12 of whom were definitively positive for S. mansoni eggs in stool and 12 definitively negative for both S. mansoni eggs and worm antigen. Tissue and blood T cell subsets were characterized by flow cytometry and immunohistochemistry (IHC). Th17 and Th1 cells from both the blood and foreskin expressed higher levels of CCR5 and were more activated than other CD4 T cell subsets. S. mansoni-infected men had a higher frequency of systemic Th1 cells (22.9 vs. 16.5% of blood CD4 T cells, p<0.05), Th17 cells (2.3 vs. 1.5%, p<0.05), and Th22 cells (0.5 vs. 0.3%, p<0.01) than uninfected men. Additionally, Th17 cells in the blood of S. mansoni-infected men demonstrated enhanced function (28.1 vs. 16.3% producing multiple cytokines, p = 0.046). However, these immune alterations were not observed in foreskin tissue. Conclusions/Significance S. mansoni infection was associated with an increased frequency of highly HIV-susceptible Th1, Th17 and Th22 cell subsets in the blood, but these T cell immune differences did not extend to the foreskin. S. mansoni induced changes in T cell immunology mediated through the common mucosal immune system are not likely to increase HIV susceptibility in the foreskin. Fishing communities in East Africa have a very high prevalence of HIV, and also high rates of other endemic infections such as malaria and the fluke Schistosoma mansoni. Genital infections are known to increase HIV susceptibility through the recruitment and activation of mucosal CD4 T cells to the site of HIV sexual exposure. These activated CD4 T cells are necessary for an effective host immune response but are also preferentially infected by HIV. We hypothesized that S. mansoni infection in the gut mucosa might increase recruitment and activation of HIV target cells at other mucosal sites, and thereby contribute to high HIV rates in fishing communities. We enrolled men from a fishing community in Uganda and examined the frequency of highly HIV-susceptible cell types in their blood and foreskin tissue (a main site of HIV acquisition in heterosexual men). We found that men with S. mansoni infection had a greater frequency of HIV target cells in their blood, but not their foreskin tissue, perhaps because foreskin cells did not express mucosal homing markers. It is possible that HIV target cells observed in the blood of S. mansoni-infected individuals may traffic to other mucosae, such as the vagina or gut, and so the possibility that S. mansoni infection increases risk at these sites should be explored.
Collapse
|
26
|
Cervicovaginal bacteria are a major modulator of host inflammatory responses in the female genital tract. Immunity 2015; 42:965-76. [PMID: 25992865 DOI: 10.1016/j.immuni.2015.04.019] [Citation(s) in RCA: 509] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/26/2015] [Accepted: 03/10/2015] [Indexed: 12/18/2022]
Abstract
Colonization by Lactobacillus in the female genital tract is thought to be critical for maintaining genital health. However, little is known about how genital microbiota influence host immune function and modulate disease susceptibility. We studied a cohort of asymptomatic young South African women and found that the majority of participants had genital communities with low Lactobacillus abundance and high ecological diversity. High-diversity communities strongly correlated with genital pro-inflammatory cytokine concentrations in both cross-sectional and longitudinal analyses. Transcriptional profiling suggested that genital antigen-presenting cells sense gram-negative bacterial products in situ via Toll-like receptor 4 signaling, contributing to genital inflammation through activation of the NF-κB signaling pathway and recruitment of lymphocytes by chemokine production. Our study proposes a mechanism by which cervicovaginal microbiota impact genital inflammation and thereby might affect a woman's reproductive health, including her risk of acquiring HIV.
Collapse
|
27
|
Deese J, Masson L, Miller W, Cohen M, Morrison C, Wang M, Ahmed K, Agot K, Crucitti T, Abdellati S, Van Damme L. Injectable Progestin-Only Contraception is Associated With Increased Levels of Pro-Inflammatory Cytokines in the Female Genital Tract. Am J Reprod Immunol 2015. [PMID: 26202107 DOI: 10.1111/aji.12415] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PROBLEM Genital inflammatory changes may be a mechanism of increased HIV risk among injectable progestin-only contraception (IPC) users. METHOD OF STUDY We conducted a cross-sectional analysis of 376 Kenyan and South African women. Genital cytokines and secretory leukocyte peptidase inhibitor concentrations in a reference population were compared to IPC users and women with reproductive tract infections. RESULTS No significant variability in marker concentrations was observed by age or site. Depot medroxyprogesterone acetate (DMPA) users had significantly higher MIP-1α, MIP-1β, IL-6, IL-8, IP-10, and RANTES concentrations. Norethisterone oenanthate users had significantly higher IL-6, IL-8, and RANTES concentrations. Women with sexually transmitted infections had variable inflammation, and women with bacterial vaginosis exhibited a mixed profile of up and downregulation. CONCLUSION The finding of substantial mucosal inflammation among DMPA users provides evidence which, combined with the results of prior studies, suggests that DMPA may create an immune environment conducive to HIV target cell recruitment and inhibitory for antiviral activity.
Collapse
Affiliation(s)
- Jennifer Deese
- FHI 360, Durham, NC, USA.,Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lindi Masson
- Division of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - William Miller
- Division of Infectious Diseases, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Myron Cohen
- Division of Infectious Diseases, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Khatija Ahmed
- Setshaba Research Centre, Soshanguve, Pretoria, South Africa
| | - Kawango Agot
- Impact Research and Development Organization, Kisumu, Kenya
| | | | | | - Lut Van Damme
- The Bill & Melinda Gates Foundation, Seattle, WA, USA
| |
Collapse
|
28
|
Murphy K, Richardson BA, Dezzutti CS, Marrazzo J, Hillier SL, Hendrix CW, Herold BC. Levels of Genital Tract Defensins and Cytokines Differ between HIV-Uninfected US and African Women. Am J Reprod Immunol 2015; 74:313-22. [PMID: 26094732 DOI: 10.1111/aji.12411] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022] Open
Abstract
PROBLEM To explore the impact of race and geographic region on biomarkers of HIV risk and vaginal health, differences in soluble immune mediators were measured in US versus African and US white versus US black women at enrollment into a phase 2 microbicide trial. METHODS Levels of soluble mucosal immune mediators and inhibitory activity against E. coli, which may serve as biomarkers of risk for HIV and other genital tract infections, were quantified in cervicovaginal lavage (CVL) collected from HIV-uninfected women in the United States (n = 73) and Africa (n = 73). Differences between groups were analyzed with multivariable logistic regression models for dichotomous variables and linear regression models for continuous variables. RESULTS Secretory leukocyte protease inhibitor, lactoferrin, human beta defensins, interleukin (IL)-8, and interferon-gamma-induced protein-10 were significantly higher in US compared to African women in multivariable analysis, but only IL-1β was significantly different between US white and black women. E. coli inhibitory activity did not differ among groups in adjusted analyses. CONCLUSION Differences in soluble mucosal immunity between US and African women may play an important role in women's risk for HIV and other genital tract infections and response to prevention strategies including vaginal microbicides and should be considered in future studies.
Collapse
Affiliation(s)
- Kerry Murphy
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Charlene S Dezzutti
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeanne Marrazzo
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Sharon L Hillier
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Craig W Hendrix
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Betsy C Herold
- Department of Pediatrics, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
29
|
Reduced immune activation during tenofovir-emtricitabine therapy in HIV-negative individuals. J Acquir Immune Defic Syndr 2015; 68:495-501. [PMID: 25763783 DOI: 10.1097/qai.0000000000000529] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Elevated immune activation is associated with an increased risk of HIV acquisition. Tenofovir (TFV) has immunomodulatory properties in vitro, but how this extends in vivo remains unknown. METHODS HIV-negative adults received daily coformulated TFV disoproxil fumarate 300 mg/emtricitabine (FTC) 200 mg for 30 days followed by a 30-day washout. Markers of T-cell activation, inflammation, and cytokines were measured before drug and on days 30 (on drug) and 60 (30-day washout). Data were analyzed using one-way analysis of variance/pairwise comparisons. Intracellular disposition of TFV-diphosphate and FTC-triphosphate in CD4 and CD8 T-cells and monocytes was characterized, and the relationship with immune activation was evaluated using Pearson's correlation coefficient. RESULTS T-cell activation was available in 19 participants. CD38/HLA-DR coexpression on CD8 T-cells decreased from baseline to day 30 (3.97% vs. 2.71%; P = 0.03) and day 60 (3.97% vs. 2.41%; P = 0.008). Soluble CD27 decreased from baseline to day 60 (184.1 vs. 168.4 pg/mL; P = 0.001). Cytokines and inflammation markers were not significantly different. TFV-diphosphate and FTC-triphosphate were approximately 4-fold higher in monocytes vs. CD4 and CD8 T-cells but neither correlated with activation markers. CONCLUSIONS TFV disoproxil fumarate/FTC therapy was associated with decreased T-cell activation in HIV-negative adults, which could contribute to the antiviral effect of pre-exposure prophylaxis (NCT01040091; www.clinicaltrials.gov).
Collapse
|
30
|
Ramjee G, Abbai NS, Naidoo S. Women and Sexually Transmitted Infections in Africa. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/ojog.2015.57056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Kahle EM, Bolton M, Hughes JP, Donnell D, Celum C, Lingappa JR, Ronald A, Cohen CR, de Bruyn G, Fong Y, Katabira E, McElrath MJ, Baeten JM. Plasma cytokine levels and risk of HIV type 1 (HIV-1) transmission and acquisition: a nested case-control study among HIV-1-serodiscordant couples. J Infect Dis 2014; 211:1451-60. [PMID: 25389306 DOI: 10.1093/infdis/jiu621] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/31/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND A heightened proinflammatory state has been hypothesized to enhance human immunodeficiency virus type 1 (HIV-1) transmission - both susceptibility of HIV-1-exposed persons and infectiousness of HIV-1-infected persons. METHODS Using prospective data from heterosexual African couples with HIV-1 serodiscordance, we conducted a nested case-control analysis to assess the relationship between cytokine concentrations and the risk of HIV-1 acquisition. Case couples (n = 120) were initially serodiscordant couples in which HIV-1 was transmitted to the seronegative partner during the study; control couples (n = 321) were serodiscordant couples in which HIV-1 was not transmitted to the seronegative partner. Differences in a panel of 30 cytokines were measured using plasma specimens from both HIV-1-susceptible and HIV-1-infected partners. Plasma was collected before seroconversion for cases. RESULTS For both HIV-1-infected and HIV-1-susceptible partners, cases and controls had significantly different mean responses in cytokine panels (P < .001, by the Hotelling T(2) test), suggesting a broadly different pattern of immune activation for couples in which HIV-1 was transmitted, compared with couples without transmission. Individually, log10 mean concentrations of interleukin 10 (IL-10) and CXCL10 were significantly higher for both HIV-1-susceptible and HIV-1-infected case partners, compared with HIV-1-susceptible and HIV-1-infected control partners (P < .01 for all comparisons). In multivariate analysis, HIV-1 transmission was significantly associated with elevated CXCL10 concentrations in HIV-1-susceptible partners (P = .001) and with elevated IL-10 concentrations in HIV-1-infected partners (P = .02). CONCLUSIONS Immune activation, as measured by levels of cytokine markers, particularly elevated levels of IL-10 and CXCL1, are associated with increased HIV-1 susceptibility and infectiousness.
Collapse
Affiliation(s)
| | - Michael Bolton
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle
| | | | - Deborah Donnell
- Department of Epidemiology Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle
| | - Connie Celum
- Department of Epidemiology Department of Global Health Department of Medicine
| | - Jairam R Lingappa
- Department of Global Health Department of Medicine Department of Pediatrics, University of Washington
| | - Allan Ronald
- Department of Medicine, University of Manitoba, Winnepeg, Canada
| | - Craig R Cohen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco
| | - Guy de Bruyn
- Perinatal HIV Research Unit, University of Witwatersrand, Johannesburg, South Africa
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle
| | - Elly Katabira
- Infectious Disease Institute, Makerere University, Kampala, Uganda
| | - M Juliana McElrath
- Department of Global Health Department of Medicine Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle
| | - Jared M Baeten
- Department of Epidemiology Department of Global Health Department of Medicine
| | | |
Collapse
|
32
|
Cervical inflammation and immunity associated with hormonal contraception, pregnancy, and HIV-1 seroconversion. J Acquir Immune Defic Syndr 2014; 66:109-17. [PMID: 24413042 DOI: 10.1097/qai.0000000000000103] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Hormonal contraception (HC), younger age, and pregnancy have been associated with increased HIV risk in some studies. We sought to elucidate the biological mechanisms for these associations. DESIGN Case-control selection of specimens from a large, prospective, clinical study. METHODS We enrolled and followed 4531 HIV-negative women from Uganda and Zimbabwe using either the injectable depo-medroxyprogesterone acetate (DMPA), combined oral contraception, or no HC (NH). Innate immunity mediators were measured in cervical samples collected from women at their visit before HIV seroconversion (n = 199) and matched visits from women remaining HIV uninfected (n = 633). Generalized linear models were applied after Box-Cox power transformation. RESULTS Higher RANTES and lower secretory leukocyte protease inhibitor (SLPI) levels were associated with HIV seroconversion. DMPA users had higher RANTES and lower BD-2 levels. Most inflammation-promoting and/or inflammation-inducible mediators were higher [interleukin (IL)-1β, IL-6, IL-8, MIP-3α, vascular endothelial growth factor, and SLPI], and the protective BD-2 and IL-1RA:IL-1β ratio were lower among combined oral contraception users. Pregnant women showed a similar cervical immunity status (higher IL-1β, IL-6, IL-8, vascular endothelial growth factor, SLPI, and IL-1RA; lower IL-1RA:IL-1β). Age <25 years was associated with lower SLPI, IL-8, MIP-3α but higher IL-1RA:IL-1β. Zimbabwean women (with higher HIV seroconversion rates) had overall higher pro-inflammatory and lower anti-inflammatory protein levels than Ugandan women. CONCLUSIONS HC use, pregnancy, and young age alter cervical immunity in different ways known to increase risk of HIV, for example, through increased levels of pro-inflammatory cytokines or decreased levels of SLPI. Higher levels of RANTES may be one factor underlying a possible association between DMPA use and risk of HIV acquisition.
Collapse
|
33
|
Murine Plasmodium chabaudi malaria increases mucosal immune activation and the expression of putative HIV susceptibility markers in the gut and genital mucosae. J Acquir Immune Defic Syndr 2014; 65:517-25. [PMID: 24256632 DOI: 10.1097/qai.0000000000000056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To evaluate if systemic murine malarial infection enhances HIV susceptibility through parasite-induced mucosal immune alterations at sites of HIV sexual exposure. BACKGROUND Malaria and HIV have a high degree of geographical overlap and interact substantially within coinfected individuals. We used a murine model to test the hypothesis that malaria might also enhance HIV susceptibility at mucosal sites of HIV sexual exposure. METHODS Female C57/BL6 mice were infected with Plasmodium chabaudi malaria using a standardized protocol. Blood, gastrointestinal tissues, upper and lower genital tract tissues, and iliac lymph nodes were sampled 10 days postinfection, and the expression of putative HIV susceptibility and immune activation markers on T cells was assessed by flow cytometry. RESULTS P. chabaudi malaria increased expression of mucosal homing integrin α4β7 on blood CD4 and CD8 T cells, and these α4β7 T cells had significantly increased co-expression of both CCR5 and CD38. In addition, malaria increased expression of the HIV co-receptor CCR5 on CD4 T cells from the genital tract and gut mucosa as well as mucosal T-cell expression of the immune activation markers CD38, Major Histocompatibility Complex -II (MHC-II) and CD69. CONCLUSIONS Systemic murine malarial infection induced substantial upregulation of the mucosal homing integrin α4β7 in blood as well as gut and genital mucosal T-cell immune activation and HIV co-receptor expression. Human studies are required to confirm these murine findings and to examine whether malarial infection enhances the sexual acquisition of HIV.
Collapse
|
34
|
Kleppa E, Ramsuran V, Zulu S, Karlsen GH, Bere A, Passmore JAS, Ndhlovu P, Lillebø K, Holmen SD, Onsrud M, Gundersen SG, Taylor M, Kjetland EF, Ndung’u T. Effect of female genital schistosomiasis and anti-schistosomal treatment on monocytes, CD4+ T-cells and CCR5 expression in the female genital tract. PLoS One 2014; 9:e98593. [PMID: 24896815 PMCID: PMC4045760 DOI: 10.1371/journal.pone.0098593] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 05/06/2014] [Indexed: 11/19/2022] Open
Abstract
Background Schistosoma haematobium is a waterborne parasite that may cause female genital schistosomiasis (FGS), characterized by genital mucosal lesions. There is clinical and epidemiological evidence for a relationship between FGS and HIV. We investigated the impact of FGS on HIV target cell density and expression of the HIV co-receptor CCR5 in blood and cervical cytobrush samples. Furthermore we evaluated the effect of anti-schistosomal treatment on these cell populations. Design The study followed a case-control design with post treatment follow-up, nested in an on-going field study on FGS. Methods Blood and cervical cytobrush samples were collected from FGS negative and positive women for flow cytometry analyses. Urine samples were investigated for schistosome ova by microscopy and polymerase chain reaction (PCR). Results FGS was associated with a higher frequency of CD14+ cells (monocytes) in blood (11.5% in FGS+ vs. 2.2% in FGS-, p = 0.042). Frequencies of CD4+ cells expressing CCR5 were higher in blood samples from FGS+ than from FGS- women (4.7% vs. 1.5%, p = 0.018). The CD14+ cell population decreased significantly in both compartments after anti-schistosomal treatment (p = 0.043). Although the frequency of CD4+ cells did not change after treatment, frequencies of CCR5 expression by CD4+ cells decreased significantly in both compartments (from 3.4% to 0.5% in blood, p = 0.036; and from 42.4% to 5.6% in genital samples, p = 0.025). Conclusions The results support the hypothesis that FGS may increase the risk of HIV acquisition, not only through damage of the mucosal epithelial barrier, but also by affecting HIV target cell populations, and that anti-schistosomal treatment can modify this.
Collapse
Affiliation(s)
- Elisabeth Kleppa
- Norwegian Centre for Imported and Tropical Diseases, Department of Infectious Diseases, Oslo University Hospital (OUH), Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- * E-mail:
| | - Veron Ramsuran
- HIV Pathogenesis Programme, Nelson R Mandela School of Medicine, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Siphosenkosi Zulu
- School of Public Health Medicine, Nelson R Mandela School of Medicine, UKZN, Durban, South Africa
| | | | - Alfred Bere
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
| | - Jo-Ann S. Passmore
- Division of Medical Virology, IDM, University of Cape Town, Cape Town, South Africa
| | | | - Kristine Lillebø
- Norwegian Centre for Imported and Tropical Diseases, Department of Infectious Diseases, Oslo University Hospital (OUH), Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sigve D. Holmen
- Norwegian Centre for Imported and Tropical Diseases, Department of Infectious Diseases, Oslo University Hospital (OUH), Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Svein Gunnar Gundersen
- Research Unit, Sorlandet Hospital, Kristiansand, Norway
- Centre for Development Studies, University of Agder, Kristiansand, Norway
| | - Myra Taylor
- School of Public Health Medicine, Nelson R Mandela School of Medicine, UKZN, Durban, South Africa
| | - Eyrun F. Kjetland
- Norwegian Centre for Imported and Tropical Diseases, Department of Infectious Diseases, Oslo University Hospital (OUH), Oslo, Norway
- School of Public Health Medicine, Nelson R Mandela School of Medicine, UKZN, Durban, South Africa
| | - Thumbi Ndung’u
- HIV Pathogenesis Programme, Nelson R Mandela School of Medicine, University of KwaZulu-Natal (UKZN), Durban, South Africa
| |
Collapse
|
35
|
Gray GE, Moodie Z, Metch B, Gilbert PB, Bekker LG, Churchyard G, Nchabeleng M, Mlisana K, Laher F, Roux S, Mngadi K, Innes C, Mathebula M, Allen M, McElrath MJ, Robertson M, Kublin J, Corey L. Recombinant adenovirus type 5 HIV gag/pol/nef vaccine in South Africa: unblinded, long-term follow-up of the phase 2b HVTN 503/Phambili study. THE LANCET. INFECTIOUS DISEASES 2014; 14:388-96. [PMID: 24560541 DOI: 10.1016/s1473-3099(14)70020-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The HVTN 503/Phambili study, which assessed the efficacy of the Merck Ad5 gag/pol/nef subtype B HIV-1 preventive vaccine in South Africa, was stopped when futility criteria in the Step study (assessing the same vaccine in the Americas, Caribbean, and Australia) were met. Here we report long-term follow-up data. METHODS HVTN 503/Phambili was a double-blind, placebo-controlled, randomised trial that recruited HIV-1 uninfected, sexually active adults aged 18-35 years from five sites in South Africa. Eligible participants were randomly assigned (1:1) by computer-generated random numbers to either vaccine or placebo, stratified by site and sex. Cox proportional hazards models were used to estimate HIV-1 infection in the modified intention-to-treat cohort, all of whom were unmasked early in follow-up. The trial is registered with ClinicalTrials.gov, number NCT00413725 and the South African National Health Research Database, number DOH-27-0207-1539. FINDINGS Between Jan 24, 2007, and Sept 19, 2007, 801 participants (26·7%) of a planned 3000 were randomly assigned (400 to vaccine, 401 to placebo); 216 (27%) received only one injection, 529 (66%) received only two injections, and 56 (7%) received three injections. At a median follow-up of 42 months (IQR 31-42), 63 vaccine recipients (16%) had HIV-1 infection compared with 37 placebo recipients (9%; adjusted HR 1·70, 95% CI 1·13-2·55; p=0·01). Risk for HIV-1 infection did not differ according to the number of vaccinations received, sex, circumcision, or adenovirus type 5 (Ad5) serostatus. Differences in risk behaviour at baseline or during the study, or annualised dropout rate (7·7% [95% CI 6·2-9·5] for vaccine recipients vs 8·8% [7·1-10·7] for placebo recipients; p=0·40) are unlikely explanations for the increased rate of HIV-1 infections seen in vaccine recipients. INTERPRETATION The increased risk of HIV-1 acquisition in vaccine recipients, irrespective of number of doses received, warrants further investigation to understand the biological mechanism. We caution against further use of the Ad5 vector for HIV vaccines. FUNDING National Institute of Allergy and Infectious Diseases, Merck, and South African Medical Research Council.
Collapse
Affiliation(s)
- Glenda E Gray
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa; South African Medical Research Council, Cape Town, South Africa.
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Barbara Metch
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Linda-Gail Bekker
- Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | | | | | - Koleka Mlisana
- Centre for AIDS Programme for Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - Fatima Laher
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Surita Roux
- Desmond Tutu HIV Foundation, University of Cape Town, Cape Town, South Africa
| | - Kathryn Mngadi
- Centre for AIDS Programme for Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - Craig Innes
- Aurum Institute for Health Research, Johannesburg, South Africa
| | - Matsontso Mathebula
- MEDUNSA HIV Clinical Research Unit, University of Limpopo, Mankweng-E, South Africa
| | - Mary Allen
- Vaccine Research Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M Julie McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michael Robertson
- Infectious Diseases and Vaccines Clinical Research, Merck and Company, North Wales, PA, USA
| | - James Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | |
Collapse
|
36
|
Chamoun-Emanuelli AM, Bobardt M, Moncla B, Mankowski MK, Ptak RG, Gallay P, Chen Z. Evaluation of PD 404,182 as an anti-HIV and anti-herpes simplex virus microbicide. Antimicrob Agents Chemother 2013; 58:687-97. [PMID: 24217696 PMCID: PMC3910842 DOI: 10.1128/aac.02000-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 11/05/2013] [Indexed: 12/17/2022] Open
Abstract
PD 404,182 (PD) is a synthetic compound that was found to compromise HIV integrity via interaction with a nonenvelope protein viral structural component (A. M. Chamoun et al., Antimicrob. Agents Chemother. 56:672-681, 2012). The present study evaluates the potential of PD as an anti-HIV microbicide and establishes PD's virucidal activity toward another pathogen, herpes simplex virus (HSV). We show that the anti-HIV-1 50% inhibitory concentration (IC50) of PD, when diluted in seminal plasma, is ∼1 μM, similar to the IC50 determined in cell culture growth medium, and that PD retains full anti-HIV-1 activity after incubation in cervical fluid at 37°C for at least 24 h. In addition, PD is nontoxic toward vaginal commensal Lactobacillus species (50% cytotoxic concentration [CC50], >300 μM), freshly activated human peripheral blood mononuclear cells (CC50, ∼200 μM), and primary CD4(+) T cells, macrophages, and dendritic cells (CC50, >300 μM). PD also exhibited high stability in pH-adjusted Dulbecco's phosphate-buffered saline with little to no activity loss after 8 weeks at pH 4 and 42°C, indicating suitability for formulation for transportation and storage in developing countries. Finally, for the first time, we show that PD inactivates herpes simplex virus 1 (HSV-1) and HSV-2 at submicromolar concentrations. Due to the prevalence of HSV infection, the ability of PD to inactivate HSV may provide an additional incentive for use as a microbicide. The ability of PD to inactivate both HIV-1 and HSV, combined with its low toxicity and high stability, warrants additional studies for the evaluation of PD's microbicidal candidacy in animals and humans.
Collapse
Affiliation(s)
- Ana M. Chamoun-Emanuelli
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Michael Bobardt
- Department of Immunology and Microbial Sciences, IMM-9, The Scripps Research Institute, La Jolla, California, USA
| | - Bernard Moncla
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA, and Magee-Womens Research Institute Foundation, Pittsburgh, Pennsylvania, USA
| | - Marie K. Mankowski
- Southern Research Institute, Infectious Disease Research Department, Frederick, Maryland, USA
| | - Roger G. Ptak
- Southern Research Institute, Infectious Disease Research Department, Frederick, Maryland, USA
| | - Philippe Gallay
- Department of Immunology and Microbial Sciences, IMM-9, The Scripps Research Institute, La Jolla, California, USA
| | - Zhilei Chen
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
- Department of Microbial and Molecular Pathogenesis, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
37
|
The complexity of contraceptives: understanding their impact on genital immune cells and vaginal microbiota. AIDS 2013; 27 Suppl 1:S5-15. [PMID: 24088684 DOI: 10.1097/qad.0000000000000058] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Abstract
OBJECTIVE The HIV epidemic has carved contrasting trajectories around the world with sub-Saharan Africa (SSA) being most affected. We hypothesized that mean HIV-1 plasma RNA viral loads are higher in SSA than other areas, and that these elevated levels may contribute to the scale of epidemics in this region. DESIGN AND METHODS To evaluate this hypothesis, we constructed a database of means of 71,668 viral load measurements from 44 cohorts in seven regions of the world. We used linear regression statistical models to estimate differences in viral load between regions. We also constructed and analyzed a mathematical model to describe the impact of the regional viral load differences on HIV epidemic trajectory. RESULTS We found substantial regional viral load heterogeneity. The mean viral load in SSA was 0.58 log(10) copies/ml higher than in North America (95% confidence interval 0.45-0.71); this represents about a four-fold increase. The highest mean viral loads were found in Southern and East Africa, whereas in Asia, Europe, North America, and South America, mean viral loads were comparable. Mathematical modeling indicated that conservatively 14% of HIV infections in a representative population in Kenya could be attributed to the enhanced infectiousness of patients with heightened viral load. CONCLUSION We conclude that community viral load appears to be higher in SSA than in other regions and this may be a central driver of the massive HIV epidemics in this region. The elevated viral loads in SSA may reflect, among other factors, the high burden of co-infections or the preponderance of HIV-1 subtype C infection.
Collapse
|
39
|
Mitchell C, Balkus JE, McKernan-Mullin J, Cohn SE, Luque AE, Mwachari C, Cohen CR, Coombs R, Frenkel LM, Hitti J. Associations between genital tract infections, genital tract inflammation, and cervical cytobrush HIV-1 DNA in US versus Kenyan women. J Acquir Immune Defic Syndr 2013; 62:143-8. [PMID: 23018377 DOI: 10.1097/qai.0b013e318274577d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Cervical shedding of HIV-1 DNA may influence HIV-1 sexual transmission. HIV-1 DNA was detected in 250 (80%) of 316 and 207 (79%) of 259 cervical cytobrush specimens from 56 US and 80 Kenyan women, respectively. Plasma HIV-1 RNA concentration was associated with increased HIV-1 DNA shedding among US and Kenyan women. Kenyan women had higher cervicovaginal concentrations of proinflammatory interleukins (IL)-1β, IL-6, IL-8, and anti-inflammatory secretory leukocyte protease inhibitor compared with US women (all P < 0.01). HIV-1 DNA shedding was associated with increased concentrations of IL-1β and IL-6 and lower secretory leukocyte protease inhibitor among US women but not Kenyan women.
Collapse
Affiliation(s)
- Caroline Mitchell
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98104, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mitchell C, Balkus JE, Fredricks D, Liu C, McKernan-Mullin J, Frenkel LM, Mwachari C, Luque A, Cohn SE, Cohen CR, Coombs R, Hitti J. Interaction between lactobacilli, bacterial vaginosis-associated bacteria, and HIV Type 1 RNA and DNA Genital shedding in U.S. and Kenyan women. AIDS Res Hum Retroviruses 2013; 29:13-9. [PMID: 23020644 DOI: 10.1089/aid.2012.0187] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Bacterial vaginosis has been associated with genital HIV-1 shedding; however, the effect of specific vaginal bacterial species has not been assessed. We tested cervicovaginal lavage from HIV-1-seropositive women for common Lactobacillus species: L. crispatus, L. jensenii, and seven BV-associated species: BVAB1, BVAB2, BVAB3, Leptotrichia, Sneathia, Megasphaera, and Atopobium spp. using quantitative PCR. We used linear and Poisson regression to evaluate associations between vaginal bacteria and genital HIV-1 RNA and DNA. Specimens from 54 U.S. (310 visits) and 50 Kenyan women (137 visits) were evaluated. Controlling for plasma viral load, U.S. and Kenyan women had similar rates of HIV-1 RNA (19% of visits vs. 24%; IRR=0.95; 95% CI 0.61, 1.49) and DNA shedding (79% vs. 76%; IRR=0.90; 0.78, 1.05). At visits during antiretroviral therapy (ART), the likelihood of detection of HIV-1 RNA shedding was greater with BVAB3 (IRR=3.16; 95% CI 1.36, 7.32), Leptotrichia, or Sneathia (IRR=2.13; 1.02, 4.72), and less with L. jensenii (IRR=0.39; 0.18, 0.84). At visits without ART, only L. crispatus was associated with a lower likelihood of HIV-1 RNA detection (IRR=0.6; 0.40, 0.91). Vaginal Lactobacillus species were associated with lower risk of genital HIV-1 shedding, while the presence of certain BV-associated species may increase that risk.
Collapse
Affiliation(s)
- Caroline Mitchell
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Jennifer E. Balkus
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - David Fredricks
- Department of Medicine, University of Washington, Seattle, Washington
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Congzhou Liu
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | | | - Amneris Luque
- Department of Medicine, University of Rochester, Rochester, New York
| | - Susan E. Cohn
- Department of Medicine, Northwestern University, Chicago, Illinois
| | - Craig R. Cohen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California
| | - Robert Coombs
- Department of Medicine, University of Washington, Seattle, Washington
| | - Jane Hitti
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| |
Collapse
|
41
|
Picton ACP, Shalekoff S, Paximadis M, Tiemessen CT. Marked differences in CCR5 expression and activation levels in two South African populations. Immunology 2012; 136:397-407. [PMID: 22509959 DOI: 10.1111/j.1365-2567.2012.03592.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The chemokine receptor CCR5 is pivotal in determining an individual's susceptibility to HIV-1 infection and rate of disease progression. To establish whether population-based differences exist in cell surface expression of CCR5 we evaluated the extent of CCR5 expression across all peripheral blood cell types in individuals from two populations, South African Africans (SAA) and South African Caucasians (SAC). Significant differences in CCR5 expression, both in number of CCR5 molecules per cell (density) and the percentage of CCR5-expressing cells, were observed between the two study groups, within all cell subsets. Most notably, the percentage of all CCR5(+) cell subsets was significantly lower in SAC compared with SAA individuals (P < 0·01) among natural killer (NK) -cell subsets (CD56(+) , CD16(+) CD56(+) and CD56(dim) ) whereas CCR5 density was significantly higher in SAC compared with SAA individuals in CCR5(+) CD8(+) T-cell subsets and CCR5(+) NK-cell subsets (CD56(+) , CD16(+) CD56(+) and CD56(dim) ) (all P < 0·05). These relationships were maintained after exclusion of CCR5Δ32 heterozygous individuals (n = 7) from the SAC dataset. The SAA individuals exhibited significantly higher cell activation levels, as measured by HLA-DR expression, than SAC individuals in CD4(+) T-cell subsets (P = 0·002) and CD56(+) NK-cell subsets (P < 0·001). This study serves to demonstrate that ethnically divergent populations show marked differences in both cell activation and CCR5 expression, which are likely to impact on both susceptibility to HIV-1 infection and the rate of HIV-1 disease progression.
Collapse
Affiliation(s)
- Anabela C P Picton
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| | | | | | | |
Collapse
|
42
|
Cooper MD, Roberts MH, Barauskas OL, Jarvis GA. Secretory leukocyte protease inhibitor binds to Neisseria gonorrhoeae outer membrane opacity protein and is bactericidal. Am J Reprod Immunol 2012; 68:116-27. [PMID: 22537232 PMCID: PMC3395761 DOI: 10.1111/j.1600-0897.2012.01149.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 03/29/2012] [Indexed: 01/10/2023] Open
Abstract
PROBLEM Secretory leukocyte protease inhibitor (SLPI) is an innate immune peptide present on the genitourinary tract mucosa that has antimicrobial activity. In this study, we investigated the interaction of SLPI with Neisseria gonorrhoeae. METHOD OF STUDY ELISA and far-Western blots were used to analyze binding of SLPI to gonococci. The binding site for SLPI was identified by tryptic digests and mass spectrometry. Antimicrobial activity of SLPI for gonococci was determined using bactericidal assays. SLPI protein levels in cell supernatants were measured by ELISA, and SLPI mRNA levels were assessed by quantitative RT-PCR. RESULTS SLPI bound directly to the gonococcal Opa protein and was bactericidal. Epithelial cells from the reproductive tract constitutively expressed SLPI at different levels. Gonococcal infection of cells did not affect SLPI expression. CONCLUSION We conclude that SLPI is bactericidal for gonococci and is expressed by reproductive tract epithelial cells and thus is likely to play a role in the pathogenesis of gonococcal infection.
Collapse
Affiliation(s)
- Morris D Cooper
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University, Springfield, IL, USA
| | | | | | | |
Collapse
|
43
|
Thayil SM, Ho YC, Bollinger RC, Blankson JN, Siliciano RF, Karakousis PC, Page KR. Mycobacterium tuberculosis complex enhances susceptibility of CD4 T cells to HIV through a TLR2-mediated pathway. PLoS One 2012; 7:e41093. [PMID: 22844428 PMCID: PMC3402510 DOI: 10.1371/journal.pone.0041093] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/18/2012] [Indexed: 01/19/2023] Open
Abstract
Among HIV-infected individuals, co-infection with Mycobacterium tuberculosis is associated with faster progression to AIDS. We investigated the hypothesis that M. bovis BCG and M. tuberculosis (Mtb complex) could enhance susceptibility of CD4+ cells to HIV infection. Peripheral blood mononuclear cells (PBMCs) collected from healthy donors were stimulated with M. bovis BCG, M. tuberculosis CDC1551 and M. smegmatis MC2155, and stimulated CD4+ cells were infected with R5-and X4-tropic single replication-competent pseudovirus. CD4+ cells stimulated with Mtb complex showed enhanced infection with R5- and X4-tropic HIV, compared to unstimulated cells or cells stimulated with M. smegmatis (p<0.01). Treatment with TLR2 siRNA reversed the increased susceptibility of CD4+ cells with R5- and X4-tropic virus induced by Mtb complex. These findings suggest that TB infection and/or BCG vaccination may be a risk factor for HIV acquisition.
Collapse
Affiliation(s)
- Seema M. Thayil
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ya-Chi Ho
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Robert C. Bollinger
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Joel N. Blankson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Robert F. Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Petros C. Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Kathleen R. Page
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
44
|
Measurement of mucosal biomarkers in a phase 1 trial of intravaginal 3% StarPharma LTD 7013 gel (VivaGel) to assess expanded safety. J Acquir Immune Defic Syndr 2012; 59:134-40. [PMID: 22067666 DOI: 10.1097/qai.0b013e31823f2aeb] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The aim of this study was to examine the effect of the 3% StarPharma LTD 7013 gel (VivaGel) on mucosal immune markers hypothesized to be associated with HIV-1 acquisition. DESIGN Phase 1, placebo-controlled, randomized, double-blind clinical trial was performed in 54 young women in the United States and Kenya. Participants used carbopol gel with and without (placebo) StarPharma LTD 7013 twice daily over 14 days. Cervical specimens were collected for cytokines, chemokines, T cells, and dendritic cells at days 0, 7, 14, and 21. A negative binomial regression model was used to assess differences between study arms. RESULTS Several mucosal immune parameters were increased in the VivaGel arm compared with placebo. For cytokines D7, IL-6 (P = 0.05); D 14, interferon gamma (P = 0.03), IL-2 (P = 0.04), IL-5 (P = 0.003), and IL-10 (P = 0.001) were increased. On D7, CD8+/CD69+ T cells tended to be increased (P < 0.08); limiting analysis to visits without blood or bacterial vaginosis, these findings were stronger as follows: at D7, CD8+/CD69+ T cells were increased in the VivaGel arm (P < 0.005), as were CD4+/CD69+ cells (P = 0.001) and CD4+/CCR5+ T cells (P = 0.01). The changes described for D7 and 14 were no longer seen at D21. CONCLUSIONS Markers associated with inflammation and epithelial damage were reversibly elevated in the VivaGel arm compared with the placebo arm after 7-14 days of twice daily product use.
Collapse
|
45
|
|
46
|
Scott ME, Wilson SS, Cosentino LA, Richardson BA, Moscicki AB, Hillier SL, Herold BC. Interlaboratory reproducibility of female genital tract cytokine measurements by Luminex: implications for microbicide safety studies. Cytokine 2011; 56:430-4. [PMID: 21764598 PMCID: PMC3185204 DOI: 10.1016/j.cyto.2011.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 06/14/2011] [Accepted: 06/17/2011] [Indexed: 10/18/2022]
Abstract
The interlaboratory reproducibility of cytokine measurements from cervicovaginal samples by Luminex has not been reported. Using cervicovaginal lavage specimens collected on three study days from 12 women participating in a Phase I microbicide study, we measured a panel of eight cytokines in three independent laboratories. Four (IFN-γ, IL-10, IL-17, and TNF) were below the limit of detection in the majority (85%) of samples in either two or all three laboratories, an observation that may guide analyte selection for future studies. Good interlaboratory agreement (intraclass correlation coefficient, r>0.7) in absolute levels was observed for IL-1β, IL-6, and IL-8, while poor agreement was seen for IFN-α2 (r=0.47). When considering within-subject change from baseline (pre-product, at study-day 0) to either post-product visit (study-days 7 and 14), IL-1β and IL-6 exhibited good interlaboratory agreement (r>0.7), while IFN-α2 and IL-8 did not. Future studies addressing the clinical utility of specific biomarkers of inflammation for microbicide trials should consider reproducibility in the context of defining biologically meaningful thresholds of change for candidate biomarkers, ensuring that such change can be reliably distinguished from background variability.
Collapse
Affiliation(s)
- Mark E. Scott
- Division of Adolescent Medicine, Department of Pediatrics, School of Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA. (MES) and (ABM)
| | - Sarah S. Wilson
- Departments of Pediatrics and Microbiology-Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA. (SW) and (BCH)
| | - Lisa A. Cosentino
- Department of Obstetrics, Gynecology, and Infectious Disease Research, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA 15213, USA. (LAC) and (SLL)
| | - Barbra A. Richardson
- Department of Biostatistics, University of Washington, Box 359909, 325 Ninth Avenue, Seattle, WA 98104, USA.
| | - Anna-Barbara Moscicki
- Division of Adolescent Medicine, Department of Pediatrics, School of Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA. (MES) and (ABM)
| | - Sharon L. Hillier
- Department of Obstetrics, Gynecology, and Infectious Disease Research, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA 15213, USA. (LAC) and (SLL)
- Department of Obstetrics, Gynecology, and Reproductive Services, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA 15213, USA
| | - Betsy C. Herold
- Departments of Pediatrics and Microbiology-Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA. (SW) and (BCH)
| |
Collapse
|
47
|
Jaspan HB, Liebenberg L, Hanekom W, Burgers W, Coetzee D, Williamson AL, Little F, Myer L, Coombs RW, Sodora D, Passmore JA. Immune activation in the female genital tract during HIV infection predicts mucosal CD4 depletion and HIV shedding. J Infect Dis 2011; 204:1550-6. [PMID: 21940422 DOI: 10.1093/infdis/jir591] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Plasma viral load predicts genital tract human immunodeficiency virus (HIV) shedding in HIV-infected women. We investigated whether local mucosal T-cell activation (HLA-DR, CD38, CCR5, and Ki67) contributed to HIV shedding in the genital tracts of HIV-infected women. We showed that cervical cytobrush-derived T cells expressed higher frequencies of T-cell activation markers (CD38+ and HLA-DR+) than blood-derived T cells. Expression was significantly higher in HIV-infected women than in uninfected women. We found that the frequency of activated proliferating cervical T cells (Ki67+; Ki67+CCR5+) broadly predicted HIV shedding in the genital tract in HIV-infected women, independently of plasma viral loads. Furthermore, activated cervical T cells (HLA-DR+CD38+ and HLA-DR+CCR5+) and local HIV shedding were independently associated with CD4 depletion in the genital tract. These data suggest that the presence of high frequencies of activated T cells in the female genital mucosa during HIV infection facilitates both local HIV shedding and CD4 T-cell depletion.
Collapse
Affiliation(s)
- Heather B Jaspan
- Institute of Infectious Disease and Molecular Medicine, Cape Town, South Africa
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Jaspan H. The wrong place at the wrong time: geographic disparities in young people's HIV Risk. J Adolesc Health 2011; 49:227-9. [PMID: 21856512 PMCID: PMC3878907 DOI: 10.1016/j.jadohealth.2011.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 07/07/2011] [Indexed: 10/17/2022]
Affiliation(s)
- Heather Jaspan
- Seattle Biomedical Research Institute Seattle, Washington Division of Immunology Institute of Infectious Diseases and Molecular Medicine University of Cape Town Cape Town, South Africa
| |
Collapse
|
49
|
Horbul JE, Schmechel SC, Miller BRL, Rice SA, Southern PJ. Herpes simplex virus-induced epithelial damage and susceptibility to human immunodeficiency virus type 1 infection in human cervical organ culture. PLoS One 2011; 6:e22638. [PMID: 21818356 PMCID: PMC3144918 DOI: 10.1371/journal.pone.0022638] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 07/03/2011] [Indexed: 11/20/2022] Open
Abstract
Normal human premenopausal cervical tissue has been used to derive primary cell populations and to establish ex vivo organ culture systems to study infections with herpes simplex virus (HSV-1 or HSV-2) and human immunodeficiency virus type 1 (HIV-1). Infection with either HSV-1 or HSV-2 rapidly induced multinuclear giant cell formation and widespread damage in mucosal epithelial cells. Subsequent exposure of the damaged mucosal surfaces to HIV-1 revealed frequent co-localization of HSV and HIV-1 antigens. The short-term organ culture system provides direct experimental support for the epidemiological findings that pre-existing sexually transmitted infections, including primary and recurrent herpes virus infections at mucosal surfaces, represent major risk factors for acquisition of primary HIV-1 infection. Epithelial damage in combination with pre-existing inflammation, as described here for overtly normal human premenopausal cervix, creates a highly susceptible environment for the initiation and establishment of primary HIV-1 infection in the sub-mucosa of the cervical transformation zone.
Collapse
Affiliation(s)
- Julie E. Horbul
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Stephen C. Schmechel
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Barrie R. L. Miller
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Stephen A. Rice
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Peter J. Southern
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
50
|
Coming of age: reconstruction of heterosexual HIV-1 transmission in human ex vivo organ culture systems. Mucosal Immunol 2011; 4:383-96. [PMID: 21430654 DOI: 10.1038/mi.2011.12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Heterosexual transmission of human immunodeficiency virus-1 (HIV-1), from men to women, involves exposure to infectious HIV-1 in semen. Therefore, the cellular and molecular processes that underlie HIV-1 transmission are closely interconnected with fundamental principles of human reproductive biology. Human ex vivo organ culture systems allow experimental reconstruction of HIV-1 transmission, using human semen and premenopausal cervicovaginal mucosal tissue, with specific emphasis on the progression from exposure to development of primary HIV-1 infection. Clearly, an isolated piece of human tissue cannot duplicate the full complexity of events in natural infections, but with correct observation of conventional medical and ethical standards, there is no opportunity to study HIV-1 exposure and primary infection in young women. Human mucosal organ cultures allow direct study of HIV-1 infection in a reproducible format while retaining major elements of complexity and variability that typify community-based HIV-1 transmission. Experimental manipulation of human mucosal tissue both allows and requires acquisition of new insights into basic processes of human mucosal immunology. Expanding from the current foundations, we believe that human organ cultures will become increasingly prominent in experimental studies of HIV-1 transmission and continuing efforts to prevent HIV-1 infection at human mucosal surfaces.
Collapse
|