1
|
Incognito GG, Ronsini C, Palmara V, Romeo P, Vizzielli G, Restaino S, La Verde M, De Tommasi O, Palumbo M, Cianci S. The Interplay Between Cervicovaginal Microbiota Diversity, Lactobacillus Profiles and Human Papillomavirus in Cervical Cancer: A Systematic Review. Healthcare (Basel) 2025; 13:599. [PMID: 40150449 PMCID: PMC11942255 DOI: 10.3390/healthcare13060599] [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: 09/12/2024] [Revised: 02/05/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Background and Objectives: Interest in defining the characteristics of the cervicovaginal microbiota (CVM) in invasive cervical cancer (ICC) is growing, particularly concerning Lactobacillus species, as evidence suggests that these may differ in affected women and potentially interact with Human Papillomavirus (HPV) infection. Understanding these features could have important implications for disease management. Thus, this study aims to systematically review the main characteristics of available literature exploring the relationship between CVM diversity, Lactobacillus profiles, and HPV in ICC; Methods: A comprehensive bibliographic search was conducted across databases, including Medline, Embase, Scopus, the Cochrane Database of Systematic Reviews, and ClinicalTrials.gov, in accordance with the to the PRISMA guidelines. The review included studies that met the following inclusion criteria: studies comparing CVM in women with ICC to controls, focusing on Community State Types (CSTs), Lactobacillus profiles, and microbial diversity. Exclusion criteria included commentaries, letters, reviews, and studies without control groups. Variables were analyzed using the Kruskal-Wallis and Fisher's exact tests, with statistical significance level set at 0.05. Data analysis was conducted and reviewed in a blinded manner. Results: A total of 28 studies published between 2015 and 2024 met the inclusion criteria. A total of 2082 patients were included, with 323 (41.9%) of the 770 cases testing positive for HPV and 327 (24.9%) of the 1312 controls testing positive for HPV. A total of 18 studies specifically examined HPV genotypes. Cervical swabs were employed in 19 out of 28 studies (67.9%), while vaginal swabs were used in 17 studies (60.7%). Additionally, two studies included samples collected via cervical biopsy (7.1%), four studies utilized cervicovaginal lavage (14.3%), and one study used a cervical brush for sample collection (3.6%). Regarding microbiota profiling, 26 studies (92.9%) employed 16S rRNA analysis, while one study (3.6%) utilized whole-genome sequencing (WGS), and another (3.6%) used 16s rDNA. A total of 10 studies (35.7%) examined the distribution of CSTs. Five studies (17.9%) reported on Lactobacillus profiles. Different levels of Lactobacillus crispatus and Lactobacillus iners were observed, along with variations between Lactobacillus-dominant and Lactobacillus-depleted communities. A total of 22 studies (78.6%) assessed α-diversity, and 17 studies (60.7%) examined β-diversity; Conclusions: This study emphasizes the heterogeneous features of the studies exploring the association between alterations in the CVM, HPV, and the development of ICC, suggesting the need for further research to better understand this relationship. These findings could inform new strategies for prevention and treatment.
Collapse
Affiliation(s)
- Giosuè Giordano Incognito
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy;
| | - Carlo Ronsini
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.R.); (M.L.V.)
| | - Vittorio Palmara
- Unit of Gynecology and Obstetrics, Policlinico “G. Martino”, Department of Human Pathology of Adult and Childhood “G. Barresi”, University of Messina, 98122 Messina, Italy; (V.P.); (P.R.); (S.C.)
| | - Paola Romeo
- Unit of Gynecology and Obstetrics, Policlinico “G. Martino”, Department of Human Pathology of Adult and Childhood “G. Barresi”, University of Messina, 98122 Messina, Italy; (V.P.); (P.R.); (S.C.)
| | - Giuseppe Vizzielli
- Department of Maternal and Child Health, Obstetrics and Gynecology Clinic, University Hospital of Udine, 33100 Udine, Italy; (G.V.); (S.R.)
- Division of Gynecologic Oncology, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Stefano Restaino
- Department of Maternal and Child Health, Obstetrics and Gynecology Clinic, University Hospital of Udine, 33100 Udine, Italy; (G.V.); (S.R.)
- PhD School in Biomedical Sciences, Gender Medicine, Child and Women Health, University of Sassari, 07100 Sassari, Italy
| | - Marco La Verde
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.R.); (M.L.V.)
| | - Orazio De Tommasi
- Department of Women and Children’s Health, Clinic of Gynecology and Obstetrics, University of Padua, 35100 Padua, Italy;
| | - Marco Palumbo
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy;
| | - Stefano Cianci
- Unit of Gynecology and Obstetrics, Policlinico “G. Martino”, Department of Human Pathology of Adult and Childhood “G. Barresi”, University of Messina, 98122 Messina, Italy; (V.P.); (P.R.); (S.C.)
| |
Collapse
|
2
|
Kim J, Ślęczkowska M, Nobre B, Wieringa P. Study Models for Chlamydia trachomatis Infection of the Female Reproductive Tract. Microorganisms 2025; 13:553. [PMID: 40142446 PMCID: PMC11945960 DOI: 10.3390/microorganisms13030553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Chlamydia trachomatis (Ct) is a leading cause of sexually transmitted infections globally, often resulting in inflammatory disorders, ectopic pregnancies, and infertility. Studying Ct's pathogenesis remains challenging due to its unique life cycle and host-specific interactions, which require diverse experimental models. Animal studies using mouse, guinea pig, pig, and non-human primate models provide valuable insights into immune responses, hormonal influences, and disease progression. However, they face limitations in terms of translational relevance due to physiological differences, as well as ethical concerns. Complementing these, in vitro systems, ranging from simple monolayer to advanced three-dimensional models, exhibit improved physiological relevance by replicating the human tissue architecture. This includes the detailed investigation of epithelial barrier disruptions, epithelium-stroma interactions, and immune responses at a cellular level. Nonetheless, in vitro models fall short in mimicking the intricate tissue structures found in vivo and, therefore, cannot faithfully replicate the host-pathogen interactions or infection dynamics observed in living organisms. This review presents a comprehensive overview of the in vivo and in vitro models employed over the past few decades to investigate Ct and its pathogenesis, addressing their strengths and limitations. Furthermore, we explore emerging technologies, including organ-on-chip and in silico models, as promising tools to overcome the existing challenges and refine our understanding of Ct infections.
Collapse
Affiliation(s)
| | | | | | - Paul Wieringa
- Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.K.); (M.Ś.); (B.N.)
| |
Collapse
|
3
|
Alizhan D, Ukybassova T, Bapayeva G, Aimagambetova G, Kongrtay K, Kamzayeva N, Terzic M. Cervicovaginal Microbiome: Physiology, Age-Related Changes, and Protective Role Against Human Papillomavirus Infection. J Clin Med 2025; 14:1521. [PMID: 40094958 PMCID: PMC11900180 DOI: 10.3390/jcm14051521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/17/2025] [Accepted: 02/23/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: Persistent high-risk human papillomavirus (HPV) infections are the leading cause of cervical cancer. Developing evidence suggests that the cervicovaginal microbiome plays a significant role in modulating HPV persistence and progression to cervical neoplasia. This review synthesizes the current knowledge on the interplay between the cervicovaginal microbiome and local immunity in HPV infections, emphasizing microbial diversity, immune responses, and potential therapeutic implications. Methods: A thorough review of the literature was performed using Embase, PubMed, Scopus, and Google Scholar, encompassing studies published between 2000 and 2024. Studies examining the composition of the microbiome, immune responses, and HPV-related outcomes were evaluated and synthesized into a comprehensive review. Results: A Lactobacillus-dominant microbiome, particularly with L. crispatus, creates a protective environment through lactic acid production, maintenance of low pH, and anti-inflammatory immune modulation, facilitating HPV clearance. Dysbiosis, often characterized by a dominance of L. iners and overgrowth of anaerobic bacteria, fosters chronic inflammation, cytokine imbalance, and a microenvironment conducive to HPV persistence and progression. Hormonal changes and menopause exacerbate these microbial shifts, increasing the risk of cervical lesions. Studies suggest that cytokine profiles and antimicrobial peptides significantly influence local immune responses, further modulating infection outcomes. Conclusions: The cervicovaginal microbiome is a critical determinant in HPV infection outcomes, with therapeutic potential for modulating the microbiome to enhance immune responses and prevent cervical cancer. Personalized microbiome-targeted therapies may offer a novel avenue for managing HPV and reducing cervical cancer incidence.
Collapse
Affiliation(s)
- Diana Alizhan
- School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
| | - Talshyn Ukybassova
- Clinical Academic Department of Women’s Health, CF “University Medical Center”, Astana 010000, Kazakhstan (K.K.); (N.K.); (M.T.)
| | - Gauri Bapayeva
- Clinical Academic Department of Women’s Health, CF “University Medical Center”, Astana 010000, Kazakhstan (K.K.); (N.K.); (M.T.)
| | - Gulzhanat Aimagambetova
- Clinical Academic Department of Women’s Health, CF “University Medical Center”, Astana 010000, Kazakhstan (K.K.); (N.K.); (M.T.)
- Department of Surgery, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | - Kuralay Kongrtay
- Clinical Academic Department of Women’s Health, CF “University Medical Center”, Astana 010000, Kazakhstan (K.K.); (N.K.); (M.T.)
- Department of Surgery, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | - Nazira Kamzayeva
- Clinical Academic Department of Women’s Health, CF “University Medical Center”, Astana 010000, Kazakhstan (K.K.); (N.K.); (M.T.)
| | - Milan Terzic
- Clinical Academic Department of Women’s Health, CF “University Medical Center”, Astana 010000, Kazakhstan (K.K.); (N.K.); (M.T.)
- Department of Surgery, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
4
|
Gill B, Wessels JM, Hayes CL, Ratcliffe J, Wokuri J, Ball E, Reid G, Kaul R, Rana J, Alkhaifi M, Tharao W, Smaill F, Kaushic C. Feasibility, safety and tolerability of estrogen and/or probiotics for improving vaginal health in Canadian African, Caribbean, and Black women: A pilot phase 1 clinical trial. PLoS One 2025; 20:e0315576. [PMID: 39836666 PMCID: PMC11750099 DOI: 10.1371/journal.pone.0315576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 11/24/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND A dysbiotic vaginal microbiome (VMB) is associated with clinical conditions such as bacterial vaginosis (BV) and an increased risk of human immunodeficiency virus (HIV-1) infection. Considering the high prevalence of BV among African, Caribbean and Black (ACB) women, we conducted a prospective, randomized, open-label phase 1 clinical trial to determine the feasibility, safety and tolerability of administering low-dose estrogen, probiotics or both in combination to improve vaginal health and decrease HIV-1 susceptibility. METHODS ACB women aged 18-49 from the Greater Toronto Area (GTA) were randomized to one of four study arms: intravaginal estradiol (Estring©; 7.5mg/day); a vaginal probiotic (RepHresh™ Pro-B™) administered twice daily; a combination of Estring© and vaginal RepHresh™ Pro-B™ (twice daily); or the Estring© and oral RepHresh™ Pro-B™ (twice daily), for a duration of 30 days. Feasibility was evaluated through enrolment, retention, and adherence rates, while safety and tolerability were determined by a pre- and post-treatment blood panel and reported adverse events (AEs). RESULTS Overall, 63 ACB women were screened, 50 were enrolled and received the intervention while 41 completed the study, resulting in 80% enrollment and 82% retention rates. Overall adherence to the study protocol was high at 93%, with an adherence of 92% for RepHresh™ Pro-B™ and 97% for Estring©. A total of 88 AEs were reported by 29 participants which were mild (66/88; 75%) and largely resolved (82/88;93%) by the end of the study, with no serious AEs (SAEs) noted. In addition, a panel of safety blood markers measured pre- and post-intervention confirmed no clinically significant changes in blood chemistry or blood cell count. CONCLUSION Overall, the administration of intravaginal estrogen and/or probiotics in pre-menopausal ACB women is feasible, safe, and well tolerated. TRIAL REGISTRATION The trial was registered with Clinicaltrials.gov (NCT03837015) and CIHR HIV Clinical Trials (CTN308).
Collapse
Affiliation(s)
- Biban Gill
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jocelyn M. Wessels
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON, Canada
- Afynia Laboratories, Hamilton, ON, Canada
| | - Christina L. Hayes
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jenna Ratcliffe
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Junic Wokuri
- Women’s Health in Women’s Hands Community Health Centre, Toronto, ON, Canada
| | - Elizabeth Ball
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Gregor Reid
- Departments of Microbiology & Immunology and Surgery, Western University, and Canadian Research and Development Centre for Human Microbiome and Probiotics, The Lawson Health Research Institute, London, ON, Canada
| | - Rupert Kaul
- Departments of Immunology and Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Jesleen Rana
- Women’s Health in Women’s Hands Community Health Centre, Toronto, ON, Canada
| | - Muna Alkhaifi
- Women’s Health in Women’s Hands Community Health Centre, Toronto, ON, Canada
| | - Wangari Tharao
- Women’s Health in Women’s Hands Community Health Centre, Toronto, ON, Canada
| | - Fiona Smaill
- Department of Pathology and Molecular Medicine and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
5
|
Velmurugan S, Ganesan K, Rajasundaram A, Thangam C, Cyril R, Subbaraj GK. Nanoparticles and the Vaginal Microbiota: Diagnostic and Therapeutic Innovations in Human Papilloma Virus-associated Cervical Cancer - A Systematic Review. Niger Postgrad Med J 2025; 32:1-13. [PMID: 40091465 DOI: 10.4103/npmj.npmj_265_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 03/19/2025]
Abstract
Cervical cancer (CC) remains a significant global health challenge, with the interplay between microbiome and cancer progression becoming increasingly evident. A comprehensive literature search was conducted across multiple databases, including Embase, NCBI, Google Scholar, Elsevier, Science Direct and PubMed, utilising the specific keywords such as 'cervical cancer', 'cervical microbiome', 'vaginal microbiome', 'Human papillomavirus', 'nanoparticles', 'drug delivery' and 'dysbiosis'. Studies published over the past 15 years were reviewed. A total of 400 articles were identified and 65 research articles were finally included. This systematic approach identified relevant studies, enabling an in-depth analysis of microbial species, such as Parabacteroides, Escherichia, Shigella, Gardnerella and Fusobacterium which are recognised as the potential biomarkers for CC diagnosis and progression. Dysbiosis is characterised by a reduction in helpful bacteria and a proliferation of harmful ones. It is linked to chronic inflammation and human papillomavirus infection, both of which facilitate the CC progression. Advanced nanotechnology presents innovative therapeutic options for CC treatment, enhancing drug delivery systems and targeting tumour cells more effectively. Moreover, incorporating nanocarriers into treatment regimens aims to improve the bioavailability and efficacy of existing therapies, potentially transforming the clinical approaches to CC management. This review highlights the dual role of the microbiome as both a diagnostic and prognostic biomarker and elucidates the potential of nanotechnology in optimising treatment strategies, advocating for further research on microbial interactions in cancer pathology.
Collapse
Affiliation(s)
- Saranya Velmurugan
- Medical Genetics Division, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Karthikeyan Ganesan
- Department of Pharmacology, Vinayaka Mission's Kirupananda Variyar Medical College and Hospitals, Vinayaka Mission's Research Foundation (DU), Salem, Tamil Nadu, India
| | - Archana Rajasundaram
- Department of Anatomy, Sree Balaji Medical College and Hospital, Chennai, Tamil Nadu, India
| | - C Thangam
- Department of Pharmacology, KSR Institute of Dental Science and Research, Tiruchengode, Tamil Nadu, India
| | - Rozario Cyril
- Department of Pharmacology, JKK Nattraja Dental College and Hospital, Namakkal, Tamil Nadu, India
| | - Gowtham Kumar Subbaraj
- Medical Genetics Division, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
6
|
Govindaraj S, Tyree S, Herring GB, Rahman SJ, Babu H, Ibegbu C, Young MR, Mehta CC, Haddad LB, Smith AK, Velu V. Differential expression of HIV target cells CCR5 and α4β7 in tissue resident memory CD4 T cells in endocervix during the menstrual cycle of HIV seronegative women. Front Immunol 2024; 15:1456652. [PMID: 39386203 PMCID: PMC11461385 DOI: 10.3389/fimmu.2024.1456652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Background Ovarian hormones are known to modulate the immune system in the female genital tract (FGT). We sought to define the impact of the menstrual cycle on the mucosal HIV target cell levels, and tissue-resident CD4 T cells. Materials and methods Here, we characterized the distribution, phenotype, and function of CD4 T cells with special emphasis on HIV target cells (CCR5+ and α4β7+) as well as tissue-resident memory (TRM; CD69+ and CD103+) CD4 T cells in FGT of cycling women. Peripheral blood and Endocervical cells (EC-collected from cytobrush) were collected from 105 healthy women and performed multicolor flow cytometry to characterize the various subsets of CD4 T cells. Cervicovaginal lavage (CVL) were collected for cytokine analysis and plasma were collected for hormonal analysis. All parameters were compared between follicular and luteal phase of menstrual cycle. Results Our findings revealed no significant difference in the blood CD4 T cell subsets between the follicular and luteal phase. However, in EC, the proportion of several cell types was higher in the follicular phase compared to the luteal phase of menstrual cycle, including CCR5+α4β7-cells (p=0.01), CD69+CD103+ TRM (p=0.02), CCR5+CD69+CD103+ TRM (p=0.001) and FoxP3+ CD4 T cells (p=0.0005). In contrast, α4β7+ CCR5- cells were higher in the luteal phase (p=0.0004) compared to the follicular phase. In addition, we also found that hormonal levels (P4/E2 ratio) and cytokines (IL-5 and IL-6) were correlated with CCR5+ CD4 T cells subsets during the follicular phase of the menstrual cycle. Conclusion Overall, these findings suggest the difference in the expression of CCR5 and α4β7 in TRM CD4 T cell subsets in endocervix of HIV seronegative women between the follicular and luteal phase. Increase in the CCR5+ expression on TRM subsets could increase susceptibility to HIV infection during follicular phase of the menstrual cycle.
Collapse
Affiliation(s)
- Sakthivel Govindaraj
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Staple Tyree
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Gina Bailey Herring
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
- Grady Ponce de Leon Center, Grady Health System, Atlanta, GA, United States
| | - Sadia J. Rahman
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Hemalatha Babu
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Chris Ibegbu
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Marisa R. Young
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - C. Christina Mehta
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Lisa B. Haddad
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Biomedical Research, Population Council, New York, NY, United States
| | - Alicia K. Smith
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Vijayakumar Velu
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| |
Collapse
|
7
|
Busonero F, Lenarduzzi S, Crobu F, Gentile RM, Carta A, Cracco F, Maschio A, Camarda S, Marongiu M, Zanetti D, Conversano C, Di Lorenzo G, Mazzà D, De Seta F, Girotto G, Sanna S. The Women4Health cohort: a unique cohort to study women-specific mechanisms of cardio-metabolic regulation. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae012. [PMID: 38532851 PMCID: PMC10964981 DOI: 10.1093/ehjopen/oeae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Aims Epidemiological research has shown relevant differences between sexes in clinical manifestations, severity, and progression of cardiovascular and metabolic disorders. To date, the mechanisms underlying these differences remain unknown. Given the rising incidence of such diseases, gender-specific research on established and emerging risk factors, such as dysfunction of glycaemic and/or lipid metabolism, of sex hormones and of gut microbiome, is of paramount importance. The relationships between sex hormones, gut microbiome, and host glycaemic and/or lipid metabolism are largely unknown even in the homoeostasis status. Yet this knowledge gap would be pivotal to pinpoint to key mechanisms that are likely to be disrupted in disease context. Methods and results Here we present the Women4Health (W4H) cohort, a unique cohort comprising up to 300 healthy women followed up during a natural menstrual cycle, set up with the primary goal to investigate the combined role of sex hormones and gut microbiota variations in regulating host lipid and glucose metabolism during homoeostasis, using a multi-omics strategy. Additionally, the W4H cohort will take into consideration another ecosystem that is unique to women, the vaginal microbiome, investigating its interaction with gut microbiome and exploring-for the first time-its role in cardiometabolic disorders. Conclusion The W4H cohort study lays a foundation for improving current knowledge of women-specific mechanisms in cardiometabolic regulation. It aspires to transform insights on host-microbiota interactions into prevention and therapeutic approaches for personalized health care.
Collapse
Affiliation(s)
- Fabio Busonero
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), c/o Cittadella Universitaria di Monserrato, SS554 Km 4500, Monserrato, 09042, CA, Italy
| | - Stefania Lenarduzzi
- Institute for Maternal and Child Health—IRCCS ‘Burlo Garofolo’, Via dell'Istria 65/1, Trieste, 34137, TS, Italy
| | - Francesca Crobu
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), c/o Cittadella Universitaria di Monserrato, SS554 Km 4500, Monserrato, 09042, CA, Italy
| | - Roberta Marie Gentile
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste, 34137, TS, Italy
| | - Andrea Carta
- Department of Business and Economics, University of Cagliari, via Università 40, 09124, Cagliari, CA, Italy
| | - Francesco Cracco
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste, 34137, TS, Italy
| | - Andrea Maschio
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), c/o Cittadella Universitaria di Monserrato, SS554 Km 4500, Monserrato, 09042, CA, Italy
| | - Silvia Camarda
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste, 34137, TS, Italy
| | - Michele Marongiu
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), c/o Cittadella Universitaria di Monserrato, SS554 Km 4500, Monserrato, 09042, CA, Italy
| | - Daniela Zanetti
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), c/o Cittadella Universitaria di Monserrato, SS554 Km 4500, Monserrato, 09042, CA, Italy
| | - Claudio Conversano
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), c/o Cittadella Universitaria di Monserrato, SS554 Km 4500, Monserrato, 09042, CA, Italy
- Department of Business and Economics, University of Cagliari, via Università 40, 09124, Cagliari, CA, Italy
| | - Giovanni Di Lorenzo
- Institute for Maternal and Child Health—IRCCS ‘Burlo Garofolo’, Via dell'Istria 65/1, Trieste, 34137, TS, Italy
| | - Daniela Mazzà
- Institute for Maternal and Child Health—IRCCS ‘Burlo Garofolo’, Via dell'Istria 65/1, Trieste, 34137, TS, Italy
| | - Francesco De Seta
- Institute for Maternal and Child Health—IRCCS ‘Burlo Garofolo’, Via dell'Istria 65/1, Trieste, 34137, TS, Italy
| | - Giorgia Girotto
- Institute for Maternal and Child Health—IRCCS ‘Burlo Garofolo’, Via dell'Istria 65/1, Trieste, 34137, TS, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste, 34137, TS, Italy
| | - Serena Sanna
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), c/o Cittadella Universitaria di Monserrato, SS554 Km 4500, Monserrato, 09042, CA, Italy
- Department of Genetics, University Medical Center Groningen, Hanzeplein 1, 97123 GZ, Groningen, The Netherlands
| |
Collapse
|
8
|
Rahman N, Mian MF, Nazli A, Kaushic C. Human vaginal microbiota colonization is regulated by female sex hormones in a mouse model. Front Cell Infect Microbiol 2023; 13:1307451. [PMID: 38156321 PMCID: PMC10753781 DOI: 10.3389/fcimb.2023.1307451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/22/2023] [Indexed: 12/30/2023] Open
Abstract
Introduction Clinically, a Lactobacillus rich vaginal microbiota (VMB) is considered optimal for reproductive outcomes, while a VMB populated by anaerobes is associated with dysbiosis and the clinical condition bacterial vaginosis (BV), which is linked to increased susceptibility to sexually transmitted infections and adverse reproductive outcomes. Mouse models that mimic eubiotic and dysbiotic VMB are currently lacking but could play a critical role in improving protective interventions. Methods In this study, probiotic, eubiotic, and dysbiotic models were developed in C57BL/6 mice, using probiotic strains Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14, eubiotic Lactobacillus crispatus, or dysbiotic Gardnerella vaginalis strains. Endogenous sex hormones were manipulated by either ovariectomizing (OVX) mice or administering 17β-estradiol or progesterone pellets in OVX mice. Hormone-altered mice were inoculated with probiotic Lactobacillus species, L. crispatus, or G. vaginalis, and colonization was tracked using quantitative plating assays. Glycogen and MUC-1 levels in hormone-treated mice were determined with ELISA and MUC-1 staining. Results Following a single administration, L. rhamnosus and L. reuteri persisted in the mouse vaginal tract for up to eight days, L. crispatus persisted for up to three days, and G. vaginalis persisted for up to two days, as measured by quantitative plating assays and qPCR. Colonization of G. vaginalis was facilitated by the presence of mucin. The lack of endogenous hormones in OVX mice dramatically decreased VMB bacterial load compared to normal mice. None of the exogenous bacteria including Lactobacilli could colonize OVX mice for more than 24 hours. Treatment with 17β-estradiol but not progesterone restored the endogenous VMB and colonization with Lactobacilli and G. vaginalis. Interestingly, 17β-estradiol treated mice had significantly increased levels of glycogen compared to OVX and progesterone-treated mice. Discussion Based on the results, we have shown that estrogen played a significant role in the ability for human VMB species to colonize in our mouse models, potentially through a glycogen mediated mechanism. These results suggest there is a dynamic interaction between sex hormones and the VMB, which can affect bacterial diversity and the ability for a VMB to colonize.
Collapse
Affiliation(s)
- Nuzhat Rahman
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Center, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON, Canada
| | - M. Firoz Mian
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Center, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON, Canada
| | - Aisha Nazli
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Center, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON, Canada
| | - Charu Kaushic
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Center, Michael G. DeGroote Center for Learning and Discovery, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
9
|
Chu B, Liu Z, Liu Y, Jiang H. The Role of Advanced Parental Age in Reproductive Genetics. Reprod Sci 2023; 30:2907-2919. [PMID: 37171772 PMCID: PMC10556127 DOI: 10.1007/s43032-023-01256-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/29/2023] [Indexed: 05/13/2023]
Abstract
The increase of parental reproductive age is a worldwide trend in modern society in recent decades. In general, older parents have a significant impact on reproductive genetics and the health of offspring. In particular, advanced parental age contributes to the increase in the risk of adverse neurodevelopmental outcomes in offspring. However, it is currently under debate how and to what extent the health of future generations was affected by the parental age. In this review, we aimed to (i) provide an overview of the effects of age on the fertility and biology of the reproductive organs of the parents, (ii) highlight the candidate biological mechanisms underlying reproductive genetic alterations, and (iii) discuss the relevance of the effect of parental age on offspring between animal experiment and clinical observation. In addition, we think that the impact of environmental factors on cognitive and emotional development of older offspring will be an interesting direction.
Collapse
Affiliation(s)
- Boling Chu
- Department of Biobank, Suining Central Hospital, Suining, 629000, China
| | - Zhi Liu
- Department of Pathology, Suining Central Hospital, Suining, 629000, China
| | - Yihong Liu
- College of Humanities And Management, Guizhou University of Traditional Chinese Medicine, Guizhou, 550025, China
| | - Hui Jiang
- Department of Biobank, Suining Central Hospital, Suining, 629000, China.
| |
Collapse
|
10
|
Pérez-López FR, Fernández-Alonso AM, Mezones-Holguín E, Vieira-Baptista P. Low genitourinary tract risks in women living with the human immunodeficiency virus. Climacteric 2023:1-7. [PMID: 37054721 DOI: 10.1080/13697137.2023.2194528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
This review analyzes the clinical associations between specific low genitourinary tract clinical circumstances in perimenopausal and postmenopausal women living with human immunodeficiency virus (WLHIV). Modern antiretroviral therapy (ART) improves survival and reduces opportunistic infections and HIV transmission. Despite appropriate ART, WLHIV may display menstrual dysfunction, risk of early menopause, vaginal microbiome alterations, vaginal dryness, dyspareunia, vasomotor symptoms and low sexual function as compared to women without the infection. They have increased risks of intraepithelial and invasive cervical, vaginal and vulvar cancers. The reduced immunity capacity may also increase the risk of urinary tract infections, side-effects or toxicity of ARTs, and opportunistic infections. Menstrual dysfunction and early menopause may contribute to the early onset of vascular atherosclerosis and plaque formation, and increased osteoporosis risks requiring specific early interventions. On the other hand, the association between being postmenopausal and having a low sexual function is significant and related to low adherence to ART. WLHIV deserve a specific approach to manage different low genitourinary risks and complications related to hormone dysfunction and early menopause.
Collapse
Affiliation(s)
- F R Pérez-López
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain
- Obstetrics and Reproduction, Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain
| | | | - E Mezones-Holguín
- Centro de Estudios Económicos y Sociales en Salud, Universidad San Ignacio de Loyola, Lima, Perú
| | - P Vieira-Baptista
- Department of Gynecology-Obstetrics and Pediatrics, Hospital Lusíadas Porto, Porto, Portugal
- Lower Genital Tract Unit, Centro Hospitalar de São João, Porto, Portugal
- Department of Gynecology-Obstetrics and Pediatrics, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| |
Collapse
|
11
|
Luthuli B, Gounder K, Deymier MJ, Dong KL, Balazs AB, Mann JK, Ndung'u T. Generation and characterization of infectious molecular clones of transmitted/founder HIV-1 subtype C viruses. Virology 2023; 583:14-26. [PMID: 37084644 DOI: 10.1016/j.virol.2023.04.001] [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: 07/05/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023]
Abstract
The genetic diversity of HIV impedes vaccine development. Identifying the viral properties of transmitted/founder (T/F) variants may provide a common vaccine target. To study the biological nature of T/F viruses, we constructed full-length clones from women detected during Fiebig stage I acute HIV-1 infection (AHI) from heterosexual male-to-female (MTF) transmission; and clones after one year of infection using In-Fusion-based cloning. Eighteen full-length T/F clones were generated from 9 women and six chronic infection clones were from 2 individuals. All clones but one were non-recombinant subtype C. Three of the 5 T/F clones and 3 chronic clones tested replicated efficiently in PBMCs and utilised CCR5 coreceptor for cell entry. Transmitted/founder and chronic infection clones displayed heterogenous in vitro replicative capacity and resistance to type I interferon. T/F viruses had shorter Env glycoproteins and fewer N-linked glycosylation sites in Env. Our findings suggest MTF transmission may select viruses with compact envelopes.
Collapse
Affiliation(s)
| | - Kamini Gounder
- Africa Health Research Institute, Durban, South Africa; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Martin J Deymier
- The Ragon Institute of MGH, MIT and Harvard University, Cambridge, MA, USA
| | - Krista L Dong
- The Ragon Institute of MGH, MIT and Harvard University, Cambridge, MA, USA
| | - Alejandro B Balazs
- The Ragon Institute of MGH, MIT and Harvard University, Cambridge, MA, USA
| | - Jaclyn K Mann
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Thumbi Ndung'u
- Africa Health Research Institute, Durban, South Africa; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa; The Ragon Institute of MGH, MIT and Harvard University, Cambridge, MA, USA; Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
12
|
Chopra C, Bhushan I, Mehta M, Koushal T, Gupta A, Sharma S, Kumar M, Khodor SA, Sharma S. Vaginal microbiome: considerations for reproductive health. Future Microbiol 2022; 17:1501-1513. [DOI: 10.2217/fmb-2022-0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The microbial communities are an indispensable part of the human defense system and coexist with humans as symbionts, contributing to the metabolic functions and immune defense against pathogens. An ecologically stable vaginal microbiota is dominated by Lactobacillus species, which plays an important role in the prevention of genital infections by controlling the vaginal pH, reducing glycogen to lactic acid, and stimulating bacteriocins and hydrogen peroxide. In contrast, an abnormal vaginal microbial composition is associated with an increased risk of bacterial vaginosis, trichomoniasis, sexually transmitted diseases, preterm labor and other birth defects. This microbial diversity is affected by race, ethnicity, pregnancy, hormonal changes, sexual activities, hygiene practices and other conditions. In the present review, we discuss the changes in the microbial community of the vaginal region at different stages of a female's life cycle and its influence on her reproductive health and pathological conditions.
Collapse
Affiliation(s)
- Chitrakshi Chopra
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, 182320, India
| | - Indu Bhushan
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, 182320, India
| | - Malvika Mehta
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, 182320, India
| | - Tanvi Koushal
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, 182320, India
| | - Amita Gupta
- Department of Gynecology, Government Medical College, Jammu, (J&K), 180001, India
| | - Sarika Sharma
- Department of Sponsored Research, Division of Research & Development, Lovely Professional University, Phagwara, 144411, India
| | - Manoj Kumar
- Research Department, Sidra Medicine, Doha, 26999, Qatar
| | | | - Sandeep Sharma
- Department of Medical Laboratory Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| |
Collapse
|
13
|
Gholiof M, Adamson-De Luca E, Wessels JM. The female reproductive tract microbiotas, inflammation, and gynecological conditions. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:963752. [PMID: 36303679 PMCID: PMC9580710 DOI: 10.3389/frph.2022.963752] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
The intricate interactions between the host cells, bacteria, and immune components that reside in the female reproductive tract (FRT) are essential in maintaining reproductive tract homeostasis. Much of our current knowledge surrounding the FRT microbiota relates to the vaginal microbiota, where ‘health’ has long been associated with low bacterial diversity and Lactobacillus dominance. This concept has recently been challenged as women can have a diverse vaginal microbial composition in the absence of symptomatic disease. The structures of the upper FRT (the endocervix, uterus, Fallopian tubes, and ovaries) have distinct, lower biomass microbiotas than the vagina; however, the existence of permanent microbiotas at these sites is disputed. During homeostasis, a balance exists between the FRT bacteria and the immune system that maintains immune quiescence. Alterations in the bacteria, immune system, or local environment may result in perturbances to the FRT microbiota, defined as dysbiosis. The inflammatory signature of a perturbed or “dysbiotic” FRT microbiota is characterized by elevated concentrations of pro-inflammatory cytokines in cervical and vaginal fluid. It appears that vaginal homeostasis can be disrupted by two different mechanisms: first, a shift toward increased bacterial diversity can trigger vaginal inflammation, and second, local immunity is altered in some manner, which disrupts the microbiota in response to an environmental change. FRT dysbiosis can have negative effects on reproductive health. This review will examine the increasing evidence for the involvement of the FRT microbiotas and inflammation in gynecologic conditions such as endometriosis, infertility, and endometrial and ovarian cancer; however, the precise mechanisms by which bacteria are involved in these conditions remains speculative at present. While only in their infancy, the use of antibiotics and probiotics to therapeutically alter the FRT microbiota is being studied and is discussed herein. Our current understanding of the intimate relationship between immunity and the FRT microbiota is in its early days, and more research is needed to deepen our mechanistic understanding of this relationship and to assess how our present knowledge can be harnessed to assist in diagnosis and treatment of gynecologic conditions.
Collapse
Affiliation(s)
- Mahsa Gholiof
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON, Canada
| | - Emma Adamson-De Luca
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON, Canada
- AIMA Laboratories Inc., Hamilton, ON, Canada
| | - Jocelyn M. Wessels
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON, Canada
- AIMA Laboratories Inc., Hamilton, ON, Canada
- *Correspondence: Jocelyn M. Wessels
| |
Collapse
|
14
|
Radzey N, Harryparsad R, Meyer B, Chen PL, Gao X, Morrison C, Taku O, Williamson A, Mehou‐Loko C, Lefebvre d'Hellencourt F, Buck G, Smit J, Strauss J, Nanda K, Ahmed K, Beksinska M, Serrano M, Bailey V, Masson L, Deese J. Genital inflammatory status and the innate immune response to contraceptive initiation. Am J Reprod Immunol 2022; 88:e13542. [PMID: 35394678 PMCID: PMC10909525 DOI: 10.1111/aji.13542] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
PROBLEM Data on the effects of contraceptives on female genital tract (FGT) immune mediators are inconsistent, possibly in part due to pre-existing conditions that influence immune mediator changes in response to contraceptive initiation. METHODS This study included 161 South African women randomised to injectable depot medroxyprogesterone acetate (DMPA-IM), copper intrauterine device (IUD), or levonorgestrel (LNG) implant in the Evidence for Contraceptive Options and HIV Outcomes (ECHO) trial. We measured thirteen cytokines and antimicrobial peptides previously associated with HIV acquisition in vaginal swabs using Luminex and ELISA, before, and at 1 and 3 months after contraceptive initiation. Women were grouped according to an overall baseline inflammatory profile. We evaluated modification of the relationships between contraceptives and immune mediators by baseline inflammation, demographic, and clinical factors. RESULTS Overall, LNG implant and copper IUD initiation were associated with increases in inflammatory cytokines, while no changes were observed following DMPA-IM initiation. However, when stratifying by baseline inflammatory profile, women with low baseline inflammation in all groups experienced significant increases in inflammatory cytokines, while those with a high baseline inflammatory profile experienced no change or decreases in inflammatory cytokines. CONCLUSION We conclude that pre-contraceptive initiation immune profile modifies the effect of contraceptives on the FGT innate immune response.
Collapse
Affiliation(s)
- Nina Radzey
- Institute of Infectious Disease and Molecular Medicine (IDM)University of Cape TownCape TownSouth Africa
| | - Rushil Harryparsad
- Institute of Infectious Disease and Molecular Medicine (IDM)University of Cape TownCape TownSouth Africa
| | - Bahiah Meyer
- Institute of Infectious Disease and Molecular Medicine (IDM)University of Cape TownCape TownSouth Africa
| | | | | | | | - Ongeziwe Taku
- Institute of Infectious Disease and Molecular Medicine (IDM)University of Cape TownCape TownSouth Africa
| | - Anna‐Lise Williamson
- Institute of Infectious Disease and Molecular Medicine (IDM)University of Cape TownCape TownSouth Africa
| | - Celia Mehou‐Loko
- Institute of Infectious Disease and Molecular Medicine (IDM)University of Cape TownCape TownSouth Africa
| | | | - Gregory Buck
- Virginia Commonwealth UniversityRichmondVirginiaUSA
| | - Jennifer Smit
- MatCH Research Unit (MRU), Department of Obstetrics and GynaecologyUniversity of the WitwatersrandDurbanSouth Africa
| | | | | | - Khatija Ahmed
- Setshaba Research CentreTshwaneSouth Africa
- Department of Medical MicrobiologyUniversity of PretoriaPretoriaSouth Africa
| | - Mags Beksinska
- MatCH Research Unit (MRU), Department of Obstetrics and GynaecologyUniversity of the WitwatersrandDurbanSouth Africa
| | | | | | - Lindi Masson
- Institute of Infectious Disease and Molecular Medicine (IDM)University of Cape TownCape TownSouth Africa
- Disease Elimination Program, Life Sciences DisciplineBurnet InstituteMelbourneAustralia
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
- Central Clinical SchoolMonash UniversityMelbourneAustralia
| | - Jennifer Deese
- RTI InternationalResearch Triangle ParkNorth CarolinaUSA
| |
Collapse
|
15
|
Lendamba RW, Mbeang Nguema PP, Onanga R, Landry-Erik M. Determination of prevalence of Mycoplasma hominis and Ureaplasma species in bacterial vaginosis in association with antibiotic resistance in Franceville, Gabon. Microb Pathog 2022; 166:105528. [DOI: 10.1016/j.micpath.2022.105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/09/2022] [Accepted: 04/06/2022] [Indexed: 11/30/2022]
|
16
|
Plesniarski A, Siddik AB, Su RC. The Microbiome as a Key Regulator of Female Genital Tract Barrier Function. Front Cell Infect Microbiol 2022; 11:790627. [PMID: 34976864 PMCID: PMC8719631 DOI: 10.3389/fcimb.2021.790627] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The microbiome, the collection of microbial species at a site or compartment, has been an underappreciated realm of human health up until the last decade. Mounting evidence suggests the microbiome has a critical role in regulating the female genital tract (FGT) mucosa's function as a barrier against sexually transmitted infections (STIs) and pathogens. In this review, we provide the most recent experimental systems and studies for analyzing the interplay between the microbiome and host cells and soluble factors with an influence on barrier function. Key components, such as microbial diversity, soluble factors secreted by host and microbe, as well as host immune system, all contribute to both the physical and immunologic aspects of the FGT mucosal barrier. Current gaps in what is known about the effects of the microbiome on FGT mucosal barrier function are compared and contrasted with the literature of the gut and respiratory mucosa. This review article presents evidence supporting that the vaginal microbiome, directly and indirectly, contributes to how well the FGT protects against infection.
Collapse
Affiliation(s)
- Andrew Plesniarski
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Abu Bakar Siddik
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Ruey-Chyi Su
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
17
|
Wu J, Ning Y, Tan L, Chen Y, Huang X, Zhuo Y. Characteristics of the vaginal microbiome in women with premature ovarian insufficiency. J Ovarian Res 2021; 14:172. [PMID: 34879874 PMCID: PMC8655991 DOI: 10.1186/s13048-021-00923-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/18/2021] [Indexed: 11/30/2022] Open
Abstract
PURPOSE To investigate the relationship between vaginal microbial community structure and premature ovarian insufficiency (POI). METHODS Twenty-eight women with POI and 12 healthy women were recruited at Shenzhen Maternity and Child Healthcare Hospital between August and September 2020. Blood samples were collected for glucose tests and detection of sex hormone levels and vaginal secretions were collected for microbial group determination. Vaginal microbial community profiles were analysed by 16S rRNA gene sequencing using the Illumina MiSeq system (Illumina Inc., San Diego, CA, USA). RESULTS Compared to the controls, the serum levels of follicle-stimulating hormone, luteinizing hormone, testosterone, and the follicle-stimulating hormone/luteinizing hormone ratio, significantly increased, and oestradiol and anti-Müllerian hormone levels significantly decreased in women with POI. Higher weighted UniFrac values were observed in women with POI than in healthy women. Bacteria in the genera Lactobacillus, Brevundimonas, and Odoribacter were more abundant in the microbiomes of healthy women, while the quantity of bacteria in the genus Streptococcus was significantly increased in the microbiomes of women with POI. Moreover, these differences in microbes in women with POI were closely related to follicle-stimulating hormone, luteinizing hormone, oestradiol, and anti-Müllerian hormone levels and to the follicle-stimulating hormone/luteinizing hormone ratio. CONCLUSIONS Women with POI had altered vaginal microbial profiles compared to healthy controls. The alterations in their microbiomes were associated with serum hormone levels. These results will improve our understanding of the vaginal microbial community structure in women with POI. TRIAL REGISTRATION CHICTR, ChiCTR2000029576 . Registered 3 August 2020 - Retrospectively registered, https://www.chictr.org.cn/showproj.aspx?proj=48844 .
Collapse
Affiliation(s)
- Jiaman Wu
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Yan Ning
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Liya Tan
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yan Chen
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Xingxian Huang
- Department of Acupuncture and Moxibustion, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Yuanyuan Zhuo
- Department of Acupuncture and Moxibustion, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| |
Collapse
|
18
|
Intimate Partner Violence and Sexually Transmitted Infections Among Women in Sub-Saharan Africa. J Immigr Minor Health 2021; 23:191-198. [PMID: 32767184 DOI: 10.1007/s10903-020-01064-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Intimate partner violence (IPV) is a serious public health issue that increases risk for sexually transmitted infections (STIs). Data was obtained from women (n = 32,409) who completed the Demographic Health Survey's (DHS) domestic violence module in 7 countries in sub-Saharan Africa between 2011 and 2015. DHS questions assessed lifetime physical, emotional and sexual IPV, cumulative exposure to IPV as well as the presence of a STI in the past 12 months. Multivariate logistic regression examined the association between IPV and STIs adjusting for potentially influential covariates. Data were weighted and analyzed using STATA Software (version 14.0). Women who had experienced physical, emotional sexual and cumulative IPV were significantly more likely to have had a STIs in the past 12 months. In order to reduce the burden of STIs, initiatives may need to address underlying mechanisms such as gender norms and power inequalities which perpetuate IPV.
Collapse
|
19
|
Bastianelli C, Farris M, Bianchi P, Benagiano G. The effect of different contraceptive methods on the vaginal microbiome. Expert Rev Clin Pharmacol 2021; 14:821-836. [PMID: 33863265 DOI: 10.1080/17512433.2021.1917373] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Following a historical overview, the effect of different contraceptive methods on vaginal microbiome has been reviewed and summarized.Areas covered: Effects of combined hormonal contraceptives (oral or vaginal) and of progestin only (injectable and implantable), intrauterine devices/systems (copper- or levonorgestrel-releasing), on vaginal microbiome. In addition, mention is made of vaginal rings releasing antiviral drugs and lactic acid.Expert opinion: The vaginal microbiota (VM) is unique in that it is normally dominated by Lactobacillus species providing a degree of protection against infections; this however may vary, depending on the species and strains of Lactobacillus. Bacterial Vaginosis represents the most common dysbiosis of the VM and its prevalence can be influenced by use of contraception. Available evidence indicates that, under the influence of oral or systemically administered female sex hormones, there is apromotion of vaginal eubiosis, with aprevalence of ahealthy VM in which Lactobacilli predominate.
Collapse
Affiliation(s)
- Carlo Bastianelli
- Department of Maternal & Child Health, Gynecology and Urology, Sapienza, University of Rome, Rome, Italy
| | - Manuela Farris
- Department of Maternal & Child Health, Gynecology and Urology, Sapienza, University of Rome, Rome, Italy.,Italian Association for Demographic Education, AIED, Rome, Italy
| | - Paola Bianchi
- Department of Medico-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Giuseppe Benagiano
- Department of Maternal & Child Health, Gynecology and Urology, Sapienza, University of Rome, Rome, Italy
| |
Collapse
|
20
|
Tuddenham S, Ravel J, Marrazzo JM. Protection and Risk: Male and Female Genital Microbiota and Sexually Transmitted Infections. J Infect Dis 2021; 223:S222-S235. [PMID: 33576776 DOI: 10.1093/infdis/jiaa762] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Unique compositional and functional features of the cervicovaginal microbiota have been associated with protection against and risk for sexually transmitted infections (STI). In men, our knowledge of the interaction between the penile microbiota and STI is less developed. The current state of our understanding of these microbiota and their role in select STIs is briefly reviewed, along with strategies that leverage existing findings to manipulate genital microbiota and optimize protection against STIs. Finally, we focus on major research gaps and present a framework for future studies.
Collapse
Affiliation(s)
- Susan Tuddenham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeanne M Marrazzo
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
21
|
Predominance of Atopobium vaginae at Midtrimester: a Potential Indicator of Preterm Birth Risk in a Nigerian Cohort. mSphere 2021; 6:6/1/e01261-20. [PMID: 33504666 PMCID: PMC7885325 DOI: 10.1128/msphere.01261-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Giving birth too soon accounts for half of all newborn deaths worldwide. Clinical symptoms alone are not sufficient to identify women at risk of giving birth too early, as such a pragmatic approach to reducing the incidence of preterm birth entails developing early strategies for intervention before it materializes. Preterm birth (PTB) is the largest contributor to infant death in sub-Saharan Africa and globally. With a global estimate of 773,600, Nigeria has the third highest rate of PTB worldwide. There have been a number of microbiome profiling studies to identify vaginal microbiomes suggestive of preterm and healthy birth outcome. However, studies on the pregnancy vaginal microbiome in Africa are sparse with none performed in Nigeria. Moreover, few studies have considered the concurrent impact of steroid hormones and the vaginal microbiome on pregnancy outcome. We assessed two key determinants of pregnancy progression to gain a deeper understanding of the interactions between vaginal microbiome composition, steroid hormone concentrations, and pregnancy outcome. Vaginal swabs and blood samples were prospectively collected from healthy midtrimester pregnant women. Vaginal microbiome compositions were assessed by analysis of the V3-V5 region of 16S rRNA genes, and potential functional metabolic traits of identified vaginal microbiomes were imputed by PICRUSt (phylogenetic investigation of communities by reconstruction of unobserved states) analysis, while plasma estradiol (E2) and progesterone (P1) levels were quantified by the competitive enzyme-linked immunosorbent assay (ELISA). PTB vaginal samples were characterized by increased microbial richness, high diversity, and depletion of lactobacilli compared to term delivery samples. Women who delivered preterm were characterized by an Atopobium vaginae-dominated vagitype. High relative abundance of Atopobium vaginae at the midtrimester was highly predictive of PTB (area under the receiving operator characteristics [AUROC] of 0.983). There was a marked overlap in the range of plasma E2 and P1 values between term and PTB groups. IMPORTANCE Giving birth too soon accounts for half of all newborn deaths worldwide. Clinical symptoms alone are not sufficient to identify women at risk of giving birth too early, as such a pragmatic approach to reducing the incidence of preterm birth entails developing early strategies for intervention before it materializes. In view of the role played by the vaginal microbiome and maternal steroid hormones in determining obstetric outcome, we assessed the vaginal microbiome composition and steroid hormone during pregnancy and examined their relationship in predicting preterm birth risk in Nigerian women. This study highlights a potential early-driver microbial marker for prediction of preterm birth risk and supports the notion that vaginal microbiome composition varies across populations. A knowledge of relevant preterm birth microbial markers specific to populations would enhance the development of personalized therapeutic interventions toward restoring a microbiome that optimizes reproductive health fitness, therefore reducing the incidence of preterm birth.
Collapse
|
22
|
Transcriptional response of vaginal epithelial cells to medroxyprogesterone acetate treatment results in decreased barrier integrity. J Reprod Immunol 2020; 143:103253. [PMID: 33285485 DOI: 10.1016/j.jri.2020.103253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/12/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022]
Abstract
Medroxyprogesterone acetate (MPA) is a frequently used hormonal contraceptive that has been shown to significantly increase HIV-1 susceptibility by approximately 40 %. However, the underlying mechanism by which this occurs remains unknown. Here, we examined the biological response to MPA by vaginal epithelial cells, the first cells to encounter HIV-1 during sexual transmission, in order to understand the potential mechanism(s) of MPA-mediated increase of HIV-1 infection. Using microarray analysis and in vitro assays, we characterized the response of vaginal epithelial cells, grown in biologically relevant air-liquid interface (ALI) cultures, to physiological levels of female sex hormones, estradiol (E2), progesterone (P4), or MPA. Transcriptional profiling of E2, P4 or MPA-treated vaginal epithelial cells indicated unique transcriptional profiles associated with each hormone. MPA treatment increased transcripts of genes related to cholesterol/sterol synthesis and decreased transcripts related to cell division and cell-cell adhesion, results not seen with E2 or P4 treatments. MPA treatment also resulted in unique gene expression indicative of decreased barrier integrity. Functional assays confirmed that MPA, but not E2 or P4 treatments, resulted in increased epithelial barrier permeability and inhibited cell cycle progression. The effects of MPA on vaginal epithelial cells seen in this study may help explain the increase of HIV-1 infection in women who use MPA as a hormonal contraceptive.
Collapse
|
23
|
Deese J, Philip N, Lind M, Ahmed K, Batting J, Beksinska M, Edward VA, Louw CE, Onono M, Palanee-Phillips T, Smit JA, Baeten JM, Donnell D, Mastro TD, Mugo NR, Nanda K, Rees H, Morrison C. Sexually transmitted infections among women randomised to depot medroxyprogesterone acetate, a copper intrauterine device or a levonorgestrel implant. Sex Transm Infect 2020; 97:249-255. [PMID: 33208512 PMCID: PMC8165154 DOI: 10.1136/sextrans-2020-054590] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 11/27/2022] Open
Abstract
Objectives Reproductive aged women are at risk of pregnancy and sexually transmitted infections (STI). Understanding drivers of STI acquisition, including any association with widely used contraceptives, could help us to reduce STI prevalence and comorbidities. We compared the risk of STI among women randomised to three contraceptive methods. Methods We conducted a secondary analysis to assess the risk of chlamydia and gonorrhoea in a clinical trial evaluating HIV risk among 7829 women aged 16–35 randomised to intramuscular depot medroxyprogesterone acetate (DMPA-IM), a copper intrauterine device (IUD) or a levonorgestrel (LNG) implant. We estimated chlamydia and gonorrhoea prevalences by contraceptive group and prevalence ratios (PR) using log-binomial regression. Results At baseline, chlamydia and gonorrhoea prevalences were 18% and 5%, respectively. Final visit chlamydia prevalence did not differ significantly between DMPA-IM and copper IUD groups or between copper IUD and LNG implant groups. The DMPA-IM group had significantly lower risk of chlamydia compared with the LNG implant group (PR 0.83, 95% CI 0.72 to 0.95). Final visit gonorrhoea prevalence differed significantly only between the DMPA-IM and the copper IUD groups (PR 0.67, 95% CI 0.52 to 0.87). Conclusions The findings suggest that chlamydia and gonorrhoea risk may vary with contraceptive method use. Further investigation is warranted to better understand the mechanisms of chlamydia and gonorrhoea susceptibility in the context of contraceptive use.
Collapse
Affiliation(s)
- Jennifer Deese
- Global Public Health Impact Center, RTI International, Research Triangle Park, North Carolina, USA (formerly with FHI 360)
| | - Neena Philip
- Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Margaret Lind
- Department of Global Health, International Clinical Research Center, University of Washington, Seattle, Washington, USA
| | | | - Joanne Batting
- Effective Care Research Unit, University of the Witwatersrand, East London, South Africa
| | - Mags Beksinska
- MatCH Research Unit, Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| | - Vinodh A Edward
- The Aurum Institute, Johannesburg, South Africa.,School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cheryl E Louw
- Madibeng Centre for Research, Brits, South Africa.,Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Maricianah Onono
- Center for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Thesla Palanee-Phillips
- Faculty of Health Sciences, Wits Reproductive Health and HIV Institute, Johannesburg, South Africa
| | - Jennifer A Smit
- MatCH Research Unit, Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Durban, South Africa
| | - Jared M Baeten
- Global Health, Medicine, and Epidemiology, University of Washington, Seattle, Washington, USA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Deborah Donnell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Nelly R Mugo
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kavita Nanda
- Product Development and Introduction, FHI 360, Durham, North Carolina, USA
| | - Helen Rees
- Wits Reproductive Health & HIV Institute, University of the Witwatersrand, Johannesburg, South Africa
| | - Charles Morrison
- Behavioral, Epidemiological and Clinical Sciences, FHI 360, Durham, North Carolina, USA
| |
Collapse
|
24
|
Carosso A, Revelli A, Gennarelli G, Canosa S, Cosma S, Borella F, Tancredi A, Paschero C, Boatti L, Zanotto E, Sidoti F, Bottino P, Costa C, Cavallo R, Benedetto C. Controlled ovarian stimulation and progesterone supplementation affect vaginal and endometrial microbiota in IVF cycles: a pilot study. J Assist Reprod Genet 2020; 37:2315-2326. [PMID: 32671734 PMCID: PMC7492325 DOI: 10.1007/s10815-020-01878-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/30/2020] [Indexed: 12/23/2022] Open
Abstract
Purpose Does controlled ovarian stimulation (COS) and progesterone (P) luteal supplementation modify the vaginal and endometrial microbiota of women undergoing in vitro fertilization? Methods Fifteen women underwent microbiota analysis at two time points: during a mock transfer performed in the luteal phase of the cycle preceding COS, and at the time of fresh embryo transfer (ET). A vaginal swab and the distal extremity of the ET catheter tip were analyzed using next-generation 16SrRNA gene sequencing. Heterogeneity of the bacterial microbiota was assessed according to both the Bray-Curtis similarity index and the Shannon diversity index. Results Lactobacillus was the most prevalent genus in the vaginal samples, although its relative proportion was reduced by COS plus P supplementation (71.5 ± 40.6% vs. 61.1 ± 44.2%). In the vagina, an increase in pathogenic species was observed, involving Prevotella (3.5 ± 8.9% vs. 12.0 ± 19.4%), and Escherichia coli-Shigella spp. (1.4 ± 5.6% vs. 2.0 ± 7.8%). In the endometrium, the proportion of Lactobacilli slightly decreased (27.4 ± 34.5% vs. 25.0 ± 29.9%); differently, both Prevotella and Atopobium increased (3.4 ± 9.5% vs. 4.7 ± 7.4% and 0.7 ± 1.5% vs. 5.8 ± 12.0%). In both sites, biodiversity was greater after COS (p < 0.05), particularly in the endometrial microbiota, as confirmed by Bray-Curtis analysis of the phylogenetic distance among bacteria genera. Bray-Curtis analysis confirmed significant differences also for the paired endometrium-vagina samples at each time point. Conclusions Our findings suggest that COS and P supplementation significantly change the composition of vaginal and endometrial microbiota. The greater instability could affect both endometrial receptivity and placentation. If our findings are confirmed, they may provide a further reason to encourage the freeze-all strategy.
Collapse
Affiliation(s)
- Andrea Carosso
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy.
| | - Alberto Revelli
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| | - Gianluca Gennarelli
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| | - Stefano Canosa
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| | - Stefano Cosma
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| | - Fulvio Borella
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| | - Annalisa Tancredi
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| | - Carlotta Paschero
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| | - Lara Boatti
- Arrow Diagnostics S.r.l., Via Francesco Rolla 26, 16152, Genoa, Italy
| | - Elisa Zanotto
- Virology, Public Health and Pediatrics Department, University of Torino, Turin, Italy
| | - Francesca Sidoti
- Virology, Public Health and Pediatrics Department, University of Torino, Turin, Italy
| | - Paolo Bottino
- Virology, Public Health and Pediatrics Department, University of Torino, Turin, Italy
| | - Cristina Costa
- Virology, Public Health and Pediatrics Department, University of Torino, Turin, Italy
| | - Rossana Cavallo
- Virology, Public Health and Pediatrics Department, University of Torino, Turin, Italy
| | - Chiara Benedetto
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant Anna Hospital, University of Torino, Via Ventimiglia 1, 10126, Turin, Italy
| |
Collapse
|
25
|
Characterization of the Vaginal Microbiome in Women with Infertility and Its Potential Correlation with Hormone Stimulation during In Vitro Fertilization Surgery. mSystems 2020; 5:5/4/e00450-20. [PMID: 32665329 PMCID: PMC7363005 DOI: 10.1128/msystems.00450-20] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The microbiome had been hypothesized to be involved in the physiology and pathophysiology of assisted reproduction before the first success in IVF, while the data supporting or refuting this hypothesis were less than conclusive. Thanks to sequencing data from the 16S rRNA subunit, we characterized the microbiome in the reproductive tract of infertile women, and we found that changes in the vaginal microbiome are related to female infertility. We also found that the characteristic microbiome bacteria are mainly members of several genera and that the vaginal microbiome of infertile women is not sensitive to hormonal changes during IVF. In conclusion, our report provides data that can be used for discovering the role of the vaginal microbiome in patients suffering from secondary infertility. Perturbation of vaginal microbiome of reproductive-age women influences all the phases of a woman's reproductive life. Although studies have shown that dynamic changes in vaginal microbiome can affect pregnancy, its role in secondary infertility (i.e., inability to become pregnant or to carry a pregnancy successfully after previous success in delivering a child) and in vitrofertilization (IVF) remains to be unraveled. To determine the vaginal microbiome in women undergoing in vitrofertilization and embryo transfer (IVF-ET) and investigate its potential correlations with hormone stimulation, we recruited 30 patients with secondary infertility and receiving IVF and 92 matched healthy women and analyzed their vaginal microbiome composition using 16S rRNA gene sequencing. Our results show that women suffering from infertility (infertile women) exhibit a significant decrease in microbiome diversity and richness compared with healthy women during the nonovulation period (follicular phase) (P < 0.01), whereas vaginal microbiome of healthy women reveals dramatic fluctuations during ovulation (P < 0.05). Interestingly, infertility patients show no change of the vaginal microbiome under conditions of gonadotropin-releasing hormone (GnRH) agonist and recombinant human chorionic gonadotropin (r-hCG) induction (P > 0.05). Moreover, our results indicate that infertile women show characteristic variations in vaginal microbiome, such as increased abundance of Atopobium, Aerococcus, and Bifidobacterium and decreased abundance of Lactobacillus and Leuconostoc. IMPORTANCE The microbiome had been hypothesized to be involved in the physiology and pathophysiology of assisted reproduction before the first success in IVF, while the data supporting or refuting this hypothesis were less than conclusive. Thanks to sequencing data from the 16S rRNA subunit, we characterized the microbiome in the reproductive tract of infertile women, and we found that changes in the vaginal microbiome are related to female infertility. We also found that the characteristic microbiome bacteria are mainly members of several genera and that the vaginal microbiome of infertile women is not sensitive to hormonal changes during IVF. In conclusion, our report provides data that can be used for discovering the role of the vaginal microbiome in patients suffering from secondary infertility.
Collapse
|
26
|
Kaur H, Merchant M, Haque MM, Mande SS. Crosstalk Between Female Gonadal Hormones and Vaginal Microbiota Across Various Phases of Women's Gynecological Lifecycle. Front Microbiol 2020; 11:551. [PMID: 32296412 PMCID: PMC7136476 DOI: 10.3389/fmicb.2020.00551] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/13/2020] [Indexed: 01/02/2023] Open
Abstract
Functional equilibrium between vaginal microbiota and the host is important for maintaining gynecological and reproductive health. Apart from host genetics, infections, changes in diet, life-style and hygiene status are known to affect this delicate state of equilibrium. More importantly, the gonadal hormones strongly influence the overall structure and function of vaginal microbiota. Several studies have attempted to understand (a) the composition of vaginal microbiota in specific stages of women's reproductive cycle as well as in menopause (b) their association with gonadal hormones, and their potential role in manifestation of specific health conditions (from the perspective of cause/consequence). However, a single study that places, in context, the structural variations of the vaginal microbiome across the entire life-span of women's reproductive cycle and during various stages of menopause is currently lacking. With the objective to obtain a holistic overview of the community dynamics of vaginal micro-environment 'across' various stages of women's reproductive and post-reproductive life-cycle, we have performed a meta-analysis of approximately 1,000 vaginal microbiome samples representing various stages of the reproductive cycle and menopausal states. Objectives of this analysis included (a) understanding temporal changes in vaginal community taxonomic structure and composition as women pass through various reproductive and menopausal stages (b) exploring correlations between the levels of female sex hormones with vaginal microbiome diversity (c) analyzing changes in the pattern of community diversity in cases of dysbiotic conditions such as bacterial vaginosis, and viewing the analyzed changes in the context of a healthy state. Results reveal interesting temporal trends with respect to vaginal microbial community diversity and its pattern of correlation with host physiology. Results indicate significant differences in alpha-diversity and overall vaginal microbial community members in various reproductive and post-reproductive phases. In addition to reinforcing the known influence/role of gonadal hormones in maintaining gynecological health, results indicate how hormonal level perturbations cause/contribute to imbalances in vaginal microbiota. The nature of resulting dysbiotic state and its influence on vaginal health is also analyzed and discussed. Results also suggest that elevated vaginal microbial diversity in pregnancy does not necessarily indicate a state of bacterial infection. The study puts forward a hormone-level driven microbiome diversity hypothesis for explaining temporal patterns in vaginal microbial diversity during various stages of women's reproductive cycle and at menopause.
Collapse
Affiliation(s)
| | | | | | - Sharmila S. Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services, Pune, India
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW This review highlights current knowledge on the dichotomous role played by T helper 17 cells (Th17)-polarized CD4 T cells in maintaining mucosal immunity homeostasis versus fueling HIV/simian immunodeficiency virus (SIV) replication/persistence during antiretroviral therapy (ART), with a focus on molecular mechanisms underlying these processes. RECENT FINDING Th17 cells bridge innate and adaptive immunity against pathogens at mucosal barrier surfaces. Th17 cells are located at portal sites of HIV/SIV entry, express a unique transcriptional/metabolic status compatible with viral replication, and represent the first targets of infection. The paucity of Th17 cells during HIV/SIV infection is caused by infection itself, but also by an altered Th17 differentiation, survival, and trafficking into mucosal sites. This causes major alterations of mucosal barrier integrity, microbial translocation, and disease progression. Unless initiated during the early acute infection phases, ART fails to restore the frequency/functionality of mucosal Th17 cells. A fraction of Th17 cells is long-lived and carry HIV reservoir during ART. Recent studies identified Th17-specific host factors controlling HIV transcription, a step untargeted by current ART. SUMMARY The identification of molecular mechanisms contributing to HIV replication/persistence in mucosal Th17 cells paves the way toward the design of new Th17-specific therapeutic strategies aimed at improving mucosal immunity in HIV-infected individuals.
Collapse
|
28
|
Abstract
Rheumatoid Arthritis (RA) is a severe, chronic autoimmune disease that affects 1% of the world's population. Familial risk contributes 50% of the risk of seropositive RA, with strongest risks seen in first-degree relatives. Smoking increases the risk of developing anti-citrullinated peptide antibody (ACPA)+ RA, particularly in individuals with high-risk RA-susceptibility alleles. Other contributory environmental risks including particulate exposure, periodontal disease, bronchiectasis, diet, obesity and the oral contraceptive impact respiratory, oral, intestinal and genital tract mucosal sites. Furthermore, the first signs of autoimmunity may appear at mucosal sites e.g. sputum ACPA-IgA and IgG. While oral and faecal dysbiosis are well described, there is no consistent single bacterial species that appears to drive RA. Animal and human data suggest a model in which multiple environmental influences impact mucosal immune function through the host genetics through enhanced mucosal permeability and the traffic of pro-inflammatory PAMPs and the amplification of autoimmune responses. In some cases, autoimmunity may be driven by cross-reactivity, or mimicry, to pathogen-specific antigens, particularly where the host immune system fails to support their rapid control and elimination.
Collapse
Affiliation(s)
- Anne-Sophie Bergot
- The University of Queensland Diamantina Institute, University of Queensland, Princess Alexandra Hospital, Brisbane, 4102, QLD, Australia
| | - Rabina Giri
- Mater Research Institute-UQ, Faculty of Medicine, University of Queensland, Brisbane, 4102, QLD, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, University of Queensland, Princess Alexandra Hospital, Brisbane, 4102, QLD, Australia.
| |
Collapse
|
29
|
Al-Nasiry S, Ambrosino E, Schlaepfer M, Morré SA, Wieten L, Voncken JW, Spinelli M, Mueller M, Kramer BW. The Interplay Between Reproductive Tract Microbiota and Immunological System in Human Reproduction. Front Immunol 2020; 11:378. [PMID: 32231664 PMCID: PMC7087453 DOI: 10.3389/fimmu.2020.00378] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
In the last decade, the microbiota, i.e., combined populations of microorganisms living inside and on the surface of the human body, has increasingly attracted attention of researchers in the medical field. Indeed, since the completion of the Human Microbiome Project, insight and interest in the role of microbiota in health and disease, also through study of its combined genomes, the microbiome, has been steadily expanding. One less explored field of microbiome research has been the female reproductive tract. Research mainly from the past decade suggests that microbial communities residing in the reproductive tract represent a large proportion of the female microbial network and appear to be involved in reproductive failure and pregnancy complications. Microbiome research is facing technological and methodological challenges, as detection techniques and analysis methods are far from being standardized. A further hurdle is understanding the complex host-microbiota interaction and the confounding effect of a multitude of constitutional and environmental factors. A key regulator of this interaction is the maternal immune system that, during the peri-conceptional stage and even more so during pregnancy, undergoes considerable modulation. This review aims to summarize the current literature on reproductive tract microbiota describing the composition of microbiota in different anatomical locations (vagina, cervix, endometrium, and placenta). We also discuss putative mechanisms of interaction between such microbial communities and various aspects of the immune system, with a focus on the characteristic immunological changes during normal pregnancy. Furthermore, we discuss how abnormal microbiota composition, “dysbiosis,” is linked to a spectrum of clinical disorders related to the female reproductive system and how the maternal immune system is involved. Finally, based on the data presented in this review, the future perspectives in diagnostic approaches, research directions and therapeutic opportunities are explored.
Collapse
Affiliation(s)
- Salwan Al-Nasiry
- Department of Obstetrics and Gynecology, GROW School of Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, Netherlands
| | - Elena Ambrosino
- Department of Genetics and Cell Biology, Faculty of Health, Medicine and Life Sciences, Research School GROW (School for Oncology & Developmental Biology), Institute for Public Health Genomics, Maastricht University, Maastricht, Netherlands
| | - Melissa Schlaepfer
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Servaas A Morré
- Department of Genetics and Cell Biology, Faculty of Health, Medicine and Life Sciences, Research School GROW (School for Oncology & Developmental Biology), Institute for Public Health Genomics, Maastricht University, Maastricht, Netherlands.,Laboratory of Immunogenetics, Department Medical Microbiology and Infection Control, VU University Medical Center, Amsterdam UMC, Amsterdam, Netherlands
| | - Lotte Wieten
- Tissue Typing Laboratory, Department of Transplantation Immunology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jan Willem Voncken
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Marialuigia Spinelli
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Martin Mueller
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland.,Department of Pediatrics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
30
|
Xu J, Bian G, Zheng M, Lu G, Chan WY, Li W, Yang K, Chen ZJ, Du Y. Fertility factors affect the vaginal microbiome in women of reproductive age. Am J Reprod Immunol 2020; 83:e13220. [PMID: 31925865 PMCID: PMC7078941 DOI: 10.1111/aji.13220] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 12/17/2019] [Accepted: 01/04/2020] [Indexed: 12/29/2022] Open
Abstract
Problem For women of reproductive age, achieving a successful pregnancy requires both the normal functioning of reproductive endocrine and the health of the reproductive tract environment. We aimed to study how these fertility factors, such as female age, baseline sexual hormone levels, tubal patency, and vaginal pH, affect the composition of vaginal microbiome. Method of study The 16S rRNA sequencing was carried on vaginal microbiome samples from 85 women of reproductive age without vaginal infections or reproductive endocrine diseases. The detailed correlations between fertility factors and vaginal microbiome were quantified by Spearman's rank tests. A linear discriminant analysis was carried out to explore the effects of fertility factors on the relative abundances of vaginal bacterial species. Results The vaginal pH, levels of basal E2, LH, and FSH all had significant effects on the distribution of vaginal microbiome. The relative abundances of vaginal bacterial species, including Escherichia coli, Streptococcus agalactiae, and Prevotella intermedia, were significantly different due to the host's state of reproductive endocrine and tubal patency. It was worth noting that women with tubal obstruction, or prolonged menstrual cycle, or antral follicle count >15, or vaginal pH > 4.5 all had a higher abundance of Escherichia coli in vagina. Conclusion The fertility factors associated with the reproductive endocrine and the genital tract environment affected vaginal microbiome in women of reproductive age. The species Escherichia coli, Streptococcus agalactiae, Prevotella intermedia, etc could be used as biomarkers to reflect the pathological state of reproductive endocrine and genital tract.
Collapse
Affiliation(s)
- Jieying Xu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Gaorui Bian
- Department of NGS Sequencing, Tianyi Health Sciences Institute, Zhenjiang, China
| | - Min Zheng
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Gang Lu
- The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Wai-Yee Chan
- The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Weiping Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Kaiping Yang
- Department of NGS Sequencing, Tianyi Health Sciences Institute, Zhenjiang, China.,Departments of Obstetrics & Gynaecology and Physiology & Pharmacology, Children's Health Research Institute & Lawson Health Research Institute, Western University, London, ON, Canada
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
31
|
Berry A, Hall JV. The complexity of interactions between female sex hormones and Chlamydia trachomatis infections. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019; 6:67-75. [PMID: 31890462 PMCID: PMC6936955 DOI: 10.1007/s40588-019-00116-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW This review focuses specifically on the mechanisms by which female sex hormones, estrogen and progesterone, affect Chlamydia trachomatis infections in vivo and in vitro. RECENT FINDINGS Recent data support previous work indicating that estrogen enhances chlamydial development via multiple mechanisms. Progesterone negatively impacts Chlamydia infections also through multiple mechanisms, particularly by altering the immune response. Conflicting data exist regarding the effect of synthetic hormones, such as those found in hormonal contraceptives, on chlamydial infections. SUMMARY Numerous studies over the years have indicated that female sex hormones affect C. trachomatis infection. However, we still do not have a clear understanding of how these hormones alter Chlamydia disease transmission and progression. The studies reviewed here indicate that there are many variables that determine the outcome of Chlamydia/hormone interactions, including: 1) the specific hormone, 2) hormone concentration, 3) cell type or area of the genital tract, 4) hormone responsiveness of cell lines, and 5) animal models.
Collapse
Affiliation(s)
- Amy Berry
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
- Center for Infectious Disease, Inflammation and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Jennifer V. Hall
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
- Center for Infectious Disease, Inflammation and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| |
Collapse
|
32
|
Boodhram R, Moodley D, Abbai N, Ramjee G. Association of endogenous progesterone levels in young women using hormonal contraception with recent HIV-1 infection. BMC WOMENS HEALTH 2019; 19:63. [PMID: 31068152 PMCID: PMC6505278 DOI: 10.1186/s12905-019-0761-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 04/26/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND A high endogenous progesterone luteal state in the menstrual cycle has been independently associated with Human Immunodeficiency Virus (HIV) incidence in epidemiological studies. Hormonal contraception particularly high dose Depot Medroxyprogesterone Acetate (DMPA) is also thought to increase the risk of HIV acquisition. Inconsistent reports of this association have led us to hypothesize that unsuppressed endogenous progesterone level in women who reported hormonal contraception (HC) use may be an explanation for increased vulnerability to HIV. METHODS This pilot study was a secondary cross-sectional analysis of data and laboratory testing of stored specimens collected from women who participated in the SAMRC HIV prevention MDP 301 trial during 2005-2009 in South Africa. Serum progesterone levels were measured in 39 women at the point of first positive HIV diagnosis during study follow-up and 36 women who remained HIV uninfected at the 52-week study exit visit. RESULTS Overall, the median (IQR) progesterone level in 49 women using hormonal contraception was 0.39 ng/ml (IQR 0.13-0.45) and 48 (97.9%) women had a progesterone level < 3.0 ng/ml suggestive of adequate progesterone suppression for contraceptive efficacy. After excluding the one woman with a progesterone level of > 3.0 ng/ml, the median progesterone level in women using DMPA remained marginally higher at 0.42 ng/ml (IQR 0.27-0.45) than women using Norethisterone Enanthate (NET-EN) (0.31 ng/ml; IQR 0.13-0.41, p = 0.061). For women using hormonal contraception, the median progesterone level did not differ between women with recent HIV infection or women who remained HIV negative (0.39 vs 0.38 ng/ml, p = 0.959). Similarly, the median progesterone level in women using DMPA or NET-EN did not differ by HIV status (0.43 vs 0.41 ng/ml, p = 0.905; 0.24 vs 0.31 ng/ml, p = 0.889). CONCLUSION Among women using hormonal contraception, DMPA or NET-EN we did not observe a significant difference in progesterone levels between women with recently acquired HIV infection and women who remained HIV negative. Our findings suggest that endogenous progesterone levels remain suppressed in the presence of hormonal contraception and are not likely to be associated with HIV acquisition.
Collapse
Affiliation(s)
- Resha Boodhram
- HIV Prevention Research Unit, South African Medical Research Council, Durban, South Africa
| | - Dhayendre Moodley
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu Natal, 719 Umbilo Road, Durban, 4051, South Africa.
| | - Nathlee Abbai
- Department of Clinical Medicine Laboratory, School of Clinical Medicine, College of Health Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Gita Ramjee
- HIV Prevention Research Unit, South African Medical Research Council, Durban, South Africa.,Department of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK.,Department of Global Health, University of Washington, Seattle, USA
| |
Collapse
|
33
|
Pandit H, Kale K, Yamamoto H, Thakur G, Rokade S, Chakraborty P, Vasudevan M, Kishore U, Madan T, Fichorova RN. Surfactant Protein D Reverses the Gene Signature of Transepithelial HIV-1 Passage and Restricts the Viral Transfer Across the Vaginal Barrier. Front Immunol 2019; 10:264. [PMID: 30984160 PMCID: PMC6447669 DOI: 10.3389/fimmu.2019.00264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/31/2019] [Indexed: 01/02/2023] Open
Abstract
Effective prophylactic strategy against the current epidemic of sexually transmitted HIV-1 infection requires understanding of the innate gatekeeping mechanisms at the genital mucosa. Surfactant protein D (SP-D), a member of the collectin family of proteins naturally present in the vaginal tract, is a potential HIV-1 entry inhibitor at the cellular level. Human EpiVaginal tissues compartmentalized in culture inserts were apically exposed to HIV-1 and/or a recombinant fragment of human SP-D (rfhSP-D) and viral passage was assessed in the basal chamber containing mononuclear leukocytes. To map the gene signature facilitating or resisting the transepithelial viral transfer, microarray analysis of the HIV-1 challenged EpiVaginal tissues was performed in the absence or presence of rfhSP-D. Mucosal biocompatibility of rfhSP-D was assessed ex vivo and in the standard rabbit vaginal irritation model. The passage of virus through the EpiVaginal tissues toward the underlying target cells was associated with a global epithelial gene signature including differential regulation of genes primarily involved in inflammation, tight junctions and cytoskeletal framework. RfhSP-D significantly inhibited HIV-1 transfer across the vaginal tissues and was associated with a significant reversal of virus induced epithelial gene signature. Pro-inflammatory NF-κB and mTOR transcripts were significantly downregulated, while expression of the tight junctions and cytoskeletal genes was upheld. In the absence of virus, rfhSP-D directly interacted with the EpiVaginal tissues and upregulated expression of genes related to structural stability of the cell and epithelial integrity. There was no increment in the viral acquisition by the PBMCs present in basal chambers wherein, the EpiVaginal tissues in apical chambers were treated with rfhSP-D. The effective concentrations of rfhSP-D had no effect on lactobacilli, epithelial barrier integrity and were safe on repeated applications onto the rabbit vaginal mucosa. This pre-clinical safety data, coupled with its efficacy of restricting viral passage via reversal of virus-induced gene expression of the vaginal barrier, make a strong argument for clinical trials of rfhSP-D as a topical anti-HIV microbicide.
Collapse
Affiliation(s)
- Hrishikesh Pandit
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India.,Laboratory of Genital Tract Biology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Kavita Kale
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Hidemi Yamamoto
- Laboratory of Genital Tract Biology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Gargi Thakur
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Sushama Rokade
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Payal Chakraborty
- Genome Informatics Research Group, Bionivid Technology Pvt. Ltd., Bengaluru, India
| | - Madavan Vasudevan
- Genome Informatics Research Group, Bionivid Technology Pvt. Ltd., Bengaluru, India
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Taruna Madan
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Raina Nakova Fichorova
- Laboratory of Genital Tract Biology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
34
|
Gonzalez SM, Aguilar-Jimenez W, Su RC, Rugeles MT. Mucosa: Key Interactions Determining Sexual Transmission of the HIV Infection. Front Immunol 2019; 10:144. [PMID: 30787929 PMCID: PMC6373783 DOI: 10.3389/fimmu.2019.00144] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 01/17/2019] [Indexed: 12/26/2022] Open
Abstract
In the context of HIV sexual transmission at the genital mucosa, initial interactions between the virus and the mucosal immunity determine the outcome of the exposure. Hence, these interactions have been deeply explored in attempts to undercover potential targets for developing preventative strategies. The knowledge gained has led to propose a hypothetical model for mucosal HIV transmission. Subsequent research studies on this topic further revealed new mechanisms and identified new host-HIV interactions. This review aims at integrating these findings to inform better and update the current model of HIV transmission. At the earliest stage of virus exposure, the epithelial integrity and the presence of antiviral factors are critical in preventing viral entry to the submucosa. However, the virus has been shown to enter to the submucosa in the presence of physical abrasion or via epithelial transmigration using paracellular passage or transcytosis mechanisms. The efficiency of these processes is greater with cell-associated viral inoculums and can be influenced by the presence of viral and immune factors, and by the structure of the exposed epithelium. Once the virus reaches the submucosa, dendritic cells and fibroblasts, as recently described, have been shown in vitro of being capable of facilitating the transfer of viral particles to susceptible cells, leading to viral dissemination, most likely in a trans-infection manner. The presence of activated CD4+ T cells in submucosa increases the probability of infection, where the predominant microbiota could be implicated through the modulation of an inflammatory microenvironment. Other factors such as genital fluids and hormones could also play an essential role in HIV transmission. Here, we review the most recent evidence described for mucosal HIV-transmission contributing with the understanding of this phenomenon.
Collapse
Affiliation(s)
- Sandra M Gonzalez
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.,National HIV and Retrovirology Laboratory, JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada
| | | | - Ruey-Chyi Su
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Maria T Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
35
|
Wessels JM, Felker AM, Dupont HA, Kaushic C. The relationship between sex hormones, the vaginal microbiome and immunity in HIV-1 susceptibility in women. Dis Model Mech 2018; 11:dmm035147. [PMID: 30154116 PMCID: PMC6177003 DOI: 10.1242/dmm.035147] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The role of sex hormones in regulating immune responses in the female genital tract has been recognized for decades. More recently, it has become increasingly clear that sex hormones regulate susceptibility to sexually transmitted infections through direct and indirect mechanisms involving inflammation and immune responses. The reproductive cycle can influence simian/human immunodeficiency virus (SHIV) infections in primates and HIV-1 infection in ex vivo cervical tissues from women. Exogenous hormones, such as those found in hormonal contraceptives, have come under intense scrutiny because of the increased susceptibility to sexually transmitted infections seen in women using medroxyprogesterone acetate, a synthetic progestin-based contraceptive. Recent meta-analyses concluded that medroxyprogesterone acetate enhanced HIV-1 susceptibility in women by 40%. In contrast, estradiol-containing hormonal contraceptives were not associated with increased susceptibility and some studies reported a protective effect of estrogen on HIV/SIV infection, although the underlying mechanisms remain incompletely understood. Recent studies describe a key role for the vaginal microbiota in determining susceptibility to sexually transmitted infections, including HIV-1. While Lactobacillus spp.-dominated vaginal microbiota is associated with decreased susceptibility, complex microbiota, such as those seen in bacterial vaginosis, correlates with increased susceptibility to HIV-1. Interestingly, sex hormones are inherently linked to microbiota regulation in the vaginal tract. Estrogen has been postulated to play a key role in establishing a Lactobacillus-dominated microenvironment, whereas medroxyprogesterone acetate is linked to hypo-estrogenic effects. The aim of this Review is to contribute to a better understanding of the sex-hormone-microbiome-immunity axis, which can provide key information on the determinants of HIV-1 susceptibility in the female genital tract and, consequently, inform HIV-1 prevention strategies.
Collapse
Affiliation(s)
- Jocelyn M Wessels
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Allison M Felker
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Haley A Dupont
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
36
|
Dupont HA, Lam J, Woods MW, Zahoor MA, Kaushic C. Hormonal influence on HIV-1 transmission in the female genital tract: New insights from systems biology. Am J Reprod Immunol 2018; 80:e13019. [PMID: 30014538 DOI: 10.1111/aji.13019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Although anti-retroviral treatments have significantly slowed down the spread of the HIV-1 pandemic, approximately 2 million new infections occur every year. The majority of new infections are in sub-Saharan Africa where rates of infection are much higher in women than men. Young women are disproportionately affected and have higher susceptibility to HIV-1. The complex interactions between HIV-1 and the female genital tract (FGT) and the mechanisms regulating susceptibility in women remain incompletely understood. In this review, we focus on the current understanding of the acute events that occur in the FGT following HIV-1 exposure with a particular focus on the effect of endogenous and exogenous sex hormones on HIV-1 susceptibility. We highlight the contribution of the recent transcriptomic and proteomic studies in providing new insights.
Collapse
Affiliation(s)
- Haley A Dupont
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jeff Lam
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew W Woods
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Mohammed A Zahoor
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Michael G. DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
37
|
Hofmeyr GJ, Morrison CS, Baeten JM, Chipato T, Donnell D, Gichangi P, Mugo N, Nanda K, Rees H, Steyn P, Taylor D. Rationale and design of a multi-center, open-label, randomised clinical trial comparing HIV incidence and contraceptive benefits in women using three commonly-used contraceptive methods (the ECHO study). Gates Open Res 2018; 1:17. [PMID: 29355224 PMCID: PMC5771152 DOI: 10.12688/gatesopenres.12775.2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2018] [Indexed: 11/20/2022] Open
Abstract
Background:In vitro, animal, biological and observational clinical studies suggest that some hormonal methods, particularly depot medroxyprogesterone acetate - DMPA, may increase women's risk of HIV acquisition. DMPA is the most common contraceptive used in many countries worst affected by the HIV epidemic. To provide robust evidence for contraceptive decision-making among women, clinicians and planners, we are conducting the Evidence for Contraceptive Options and HIV Outcomes (ECHO) study in four countries with high HIV incidence and DMPA use: Kenya, South Africa, Swaziland, and Zambia (Clinical Trials.gov identifier NCT02550067). Study design: We randomized HIV negative, sexually active women 16-35 years old requesting effective contraception and agreeing to participate to either DMPA, the copper T 380A intrauterine device or levonorgestrel implant. Participants attend a contraception support visit after 1 month and quarterly visits thereafter for up to 18 months. Participants receive a standard HIV prevention package and contraceptive side-effect management at each visit. The primary outcome is HIV seroconversion. Secondary outcomes include pregnancy, serious adverse events and method discontinuation. The sample size of 7800 women provides 80% power to detect a 50% relative increase in HIV risk between any of the three method pairs, assuming 250 incident infections per comparison. Ethical considerations: Several WHO consultations have concluded that current evidence on HIV risk associated with DMPA is inconclusive and that a randomized trial is needed to guide policy, counselling and choice. Previous studies suggest that women without a specific contraceptive preference are willing to accept randomization to different contraceptive methods. Stringent performance standards are monitored by an independent data and safety monitoring board approximately every 6 months. The study has been conducted with extensive stakeholder engagement. Conclusions: The ECHO study is designed to provide robust evidence on the relative risks (HIV acquisition) and benefits (pregnancy prevention) between three effective contraceptive methods.
Collapse
Affiliation(s)
- G. Justus Hofmeyr
- Effective Care Research Unit, Universities of Witwatersrand and Fort Hare, Eastern Cape Department of Health, East London, South Africa
| | | | - Jared M. Baeten
- Departments of Global Health, Medicine, and Epidemiology, University of Washington, Seattle, WA, USA
| | - Tsungai Chipato
- Department of Obstetrics and Gynaecology, University of Zimbabwe, Harare, Zimbabwe
| | - Deborah Donnell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter Gichangi
- University of Nairobi, Nairobi, Kenya
- Ghent University, Ghent, Belgium
- International Centre for Reproductive Health (ICRH), Mombasa, Kenya
| | - Nelly Mugo
- Kenya Medical Research Institute, Nairobi, Kenya
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Kavita Nanda
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA
| | - Helen Rees
- Wits RHI, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Douglas Taylor
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA
| | - ECHO Trial Team
- Effective Care Research Unit, Universities of Witwatersrand and Fort Hare, Eastern Cape Department of Health, East London, South Africa
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA
- Departments of Global Health, Medicine, and Epidemiology, University of Washington, Seattle, WA, USA
- Department of Obstetrics and Gynaecology, University of Zimbabwe, Harare, Zimbabwe
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Nairobi, Nairobi, Kenya
- Ghent University, Ghent, Belgium
- International Centre for Reproductive Health (ICRH), Mombasa, Kenya
- Kenya Medical Research Institute, Nairobi, Kenya
- Department of Global Health, University of Washington, Seattle, WA, USA
- Wits RHI, University of the Witwatersrand, Johannesburg, South Africa
- World Health Organization, Geneva, Switzerland
| |
Collapse
|
38
|
Popovici R. Das Mikrobiom von Vagina bis Tuben und seine Relation zu uterinen Erkrankungen. GYNAKOLOGISCHE ENDOKRINOLOGIE 2018. [DOI: 10.1007/s10304-017-0171-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
39
|
Hofmeyr GJ, Morrison CS, Baeten JM, Chipato T, Donnell D, Gichangi P, Mugo N, Nanda K, Rees H, Steyn P, Taylor D. Rationale and design of a multi-center, open-label, randomised clinical trial comparing HIV incidence and contraceptive benefits in women using three commonly-used contraceptive methods (the ECHO study). Gates Open Res 2017; 1:17. [PMID: 29355224 PMCID: PMC5771152 DOI: 10.12688/gatesopenres.12775.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND In vitro, animal, biological and observational clinical studies suggest that some hormonal methods, particularly depot medroxyprogesterone acetate - DMPA, may increase women's risk of HIV acquisition. DMPA is the most common contraceptive used in many countries worst affected by the HIV epidemic. To provide robust evidence for contraceptive decision-making among women, clinicians and planners, we are conducting the Evidence for Contraceptive Options and HIV Outcomes (ECHO) study in four countries with high HIV incidence and DMPA use: Kenya, South Africa, Swaziland, and Zambia (Clinical Trials.gov identifier NCT02550067). STUDY DESIGN We randomized HIV negative, sexually active women 16-35 years old requesting effective contraception and agreeing to participate to either DMPA, the copper T 380A intrauterine device or levonorgestrel implant. Participants attend a contraception support visit after 1 month and quarterly visits thereafter for 12 to 18 months. Participants receive a standard HIV prevention package and contraceptive side-effect management at each visit. The primary outcome is HIV seroconversion. Secondary outcomes include pregnancy, serious adverse events and method discontinuation. The sample size of 7800 women provides 80% power to detect a 50% difference in HIV risk between any of the three method pairs, assuming 250 incident infections per comparison. Ethical considerations: Several WHO consultations have concluded that current evidence on HIV risk associated with DMPA is inconclusive and that a randomized trial is needed to guide policy, counselling and choice. Previous studies suggest that women without a specific contraceptive preference are willing to accept randomization to different contraceptive methods. Stringent performance standards are monitored by an independent data and safety monitoring board approximately every 6 months. The study has been conducted with extensive stakeholder engagement. CONCLUSIONS The ECHO study is designed to provide robust evidence on the relative risks (HIV acquisition) and benefits (pregnancy prevention) between three effective contraceptive methods.
Collapse
Affiliation(s)
- G. Justus Hofmeyr
- Effective Care Research Unit, Universities of Witwatersrand and Fort Hare, Eastern Cape Department of Health, East London, South Africa
| | | | - Jared M. Baeten
- Departments of Global Health, Medicine, and Epidemiology, University of Washington, Seattle, WA, USA
| | - Tsungai Chipato
- Department of Obstetrics and Gynaecology, University of Zimbabwe, Harare, Zimbabwe
| | - Deborah Donnell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter Gichangi
- University of Nairobi, Nairobi, Kenya
- Ghent University, Ghent, Belgium
- International Centre for Reproductive Health (ICRH), Mombasa, Kenya
| | - Nelly Mugo
- Kenya Medical Research Institute, Nairobi, Kenya
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Kavita Nanda
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA
| | - Helen Rees
- Wits RHI, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Douglas Taylor
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA
| | - ECHO Trial Team
- Effective Care Research Unit, Universities of Witwatersrand and Fort Hare, Eastern Cape Department of Health, East London, South Africa
- Global Health, Population and Nutrition, FHI 360, Durham, NC, USA
- Departments of Global Health, Medicine, and Epidemiology, University of Washington, Seattle, WA, USA
- Department of Obstetrics and Gynaecology, University of Zimbabwe, Harare, Zimbabwe
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Nairobi, Nairobi, Kenya
- Ghent University, Ghent, Belgium
- International Centre for Reproductive Health (ICRH), Mombasa, Kenya
- Kenya Medical Research Institute, Nairobi, Kenya
- Department of Global Health, University of Washington, Seattle, WA, USA
- Wits RHI, University of the Witwatersrand, Johannesburg, South Africa
- World Health Organization, Geneva, Switzerland
| |
Collapse
|
40
|
Shattock R. HIV vaccine research in Canada. AIDS Res Ther 2017; 14:54. [PMID: 28893293 PMCID: PMC5594529 DOI: 10.1186/s12981-017-0181-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 11/24/2022] Open
|