1
|
Glick VJ, Webber CA, Simmons LE, Martin MC, Ahmad M, Kim CH, Adams AND, Bang S, Chao MC, Howard NC, Fortune SM, Verma M, Jost M, Beura LK, James MJ, Lee SY, Mitchell CM, Clardy J, Kim KH, Gopinath S. Vaginal lactobacilli produce anti-inflammatory β-carboline compounds. Cell Host Microbe 2024:S1931-3128(24)00358-5. [PMID: 39423813 DOI: 10.1016/j.chom.2024.09.014] [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: 02/27/2024] [Revised: 08/12/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
The optimal vaginal microbiome is a Lactobacillus-dominant community. Apart from Lactobacillus iners, the presence of Lactobacillus species is associated with reduced vaginal inflammation and reduced levels of pro-inflammatory cytokines. Loss of Lactobacillus-dominance is associated with inflammatory conditions, such as bacterial vaginosis (BV). We have identified that Lactobacillus crispatus, a key vaginal bacterial species, produces a family of β-carboline compounds with anti-inflammatory activity. These compounds suppress nuclear factor κB (NF-κB) and interferon (IFN) signaling downstream of multiple pattern recognition receptors in primary human cells and significantly dampen type I IFN receptor (IFNAR) activation in monocytes. Topical application of an anti-inflammatory β-carboline compound, perlolyrine, was sufficient to significantly reduce vaginal inflammation in a mouse model of genital herpes infection. These compounds are enriched in cervicovaginal lavage (CVL) of healthy people compared with people with BV. This study identifies a family of compounds by which vaginal lactobacilli mediate host immune homeostasis and highlights a potential therapeutic avenue for vaginal inflammation.
Collapse
Affiliation(s)
- Virginia J Glick
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Cecilia A Webber
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lauren E Simmons
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Morgan C Martin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Maryam Ahmad
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Cecilia H Kim
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Amanda N D Adams
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Sunghee Bang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, MA 02115, USA
| | - Michael C Chao
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Nicole C Howard
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Manasvi Verma
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Marco Jost
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Michael J James
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, MA 02115, USA
| | - Seo Yoon Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Caroline M Mitchell
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, MA 02115, USA
| | - Ki Hyun Kim
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, MA 02115, USA; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Smita Gopinath
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Pagar R, Deshkar S, Mahore J, Patole V, Deshpande H, Gandham N, Mirza S, Junnarkar M, Nawani N. The microbial revolution: Unveiling the benefits of vaginal probiotics and prebiotics. Microbiol Res 2024; 286:127787. [PMID: 38851010 DOI: 10.1016/j.micres.2024.127787] [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: 11/04/2023] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Vaginal health is essential to a woman's overall well-being, as abnormalities in vaginal health can lead to a variety of gynaecological disorders, such as urinary tract infections, yeast infections, and bacterial vaginosis. The vaginal microbiome is essential for the prevention of these infections. Disruptions in this microbial ecosystem can significantly impact vaginal health. The concept of utilizing probiotics and prebiotics to stimulate the growth of protective vaginal microbiota has gathered substantial interest in recent years. Probiotics are live micro-organisms that strengthen and restore vaginal microbial balance by lowering pH levels, production of bacteriocins, biofilm disruption, modulation of immune response, and production of hydrogen peroxide (H2O2), consequently combating the development of pathogens. Prebiotics are oligosaccharides that encourage the development of probiotics such as lactobacilli species. Probiotics and prebiotics also have some broader implications for vaginal health, including their role in minimizing the incidence of premature birth, optimizing fertility, managing menopausal symptoms, and preventing vaginal infections. Synbiotics are a combination of probiotics and prebiotics that deliver additional benefits by encouraging the development and activity of beneficial microbes. Furthermore, postbiotics are bioactive compounds derived from probiotic bacteria during fermentation that have immunomodulatory actions and provide an additional layer of protection against vaginal infections. The present study highlights the most prevalent vaginal infections and limitations of existing therapies that influence the vaginal microbiota. The profound consequences of probiotics and prebiotics in women's health, including their role in minimizing the prevalence of vaginal infections and promoting overall vaginal health, as well as advanced therapeutic strategies such as synbiotics and postbiotics, are also discussed. The literature offers significant insights into the mechanism, efficacy, and safety of probiotics and prebiotics to healthcare providers and researchers.
Collapse
Affiliation(s)
- Roshani Pagar
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | - Sanjeevani Deshkar
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India.
| | - Jayashri Mahore
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | - Vinita Patole
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | - Hemant Deshpande
- Department of Obstetrics and Gynaecology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Nageswari Gandham
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Shahzad Mirza
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Manisha Junnarkar
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| | - Neelu Nawani
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| |
Collapse
|
3
|
Li S, Wang Q, Mi J, Chen H, Yuan T, Wang Y, Zhao L, Ma Q, Huang S. Lactobacillus crispatus-Mediated Gut-Reproductive Tract Axis-Alleviated Microbial Dysbiosis and Oviductal Inflammation in a Laying Hen Model. Microorganisms 2024; 12:1559. [PMID: 39203401 PMCID: PMC11356123 DOI: 10.3390/microorganisms12081559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Oviductal inflammation (OI) significantly reduces the egg production and economic returns in poultry farming. While Lactobacillus crispatus (LAC) is effective against inflammation, its role in treating or preventing oviductal inflammation is understudied. In this study, we investigated the therapeutic mechanisms of LAC on oviductal inflammation, with a focus on reproductive tract health, microbiome, gene expression, and cytokine levels. This study involved 24 Jingfen No. 6 laying hens aged 60 weeks, divided into four groups: the CON, OI, OI + LAC, and OI + heat-killed Lactobacillus crispatus (HLAC) groups. And it included a 10-day adaptation, a 7-day period for the development of OI using inflammation-inducing drugs (the control received saline), followed by an 8-day treatment in which the CON and OI groups received 1 mL of MRS broth daily, and the OI + LAC and OI + HLAC groups were treated with live and heat-killed Lactobacillus crispatus (109 CFUs/mL), respectively, with six hens in each group. This study showed that Lactobacillus crispatus supplementation significantly reduced the oviductal inflammation and atrophy in the hens, with the affected hens showing markedly lower egg production rates (p < 0.001) compared to the control and treated groups (OI + HLAC and OI + LAC). The daily intake of fresh (OI + LAC, p = 0.076) or heat-killed (OI + HLAC, p < 0.01) Lactobacillus crispatus notably enhanced the feed conversion efficiency. The OI group suffered significant ovarian damage and vascular rupture, more so than the CON group, while Lactobacillus crispatus supplementation mitigated this damage. The IL-1β, IL-6, and IL-8 levels were significantly elevated in the OI group compared to those in the OI + LAC group (p < 0.05), with a significant reduction in the TNF-α levels in the latter (p < 0.001). The supplementation improved the microbial composition in the cecum, isthmus, and shell gland, enriching the cecum with beneficial bacteria, such as Ruminococcus_torques_group and Megamonas. This approach fostered ovarian health and follicle differentiation and preserved the epithelial cell barrier function in the shell gland, reducing inflammatory damage in the genital tract. This dual efficacy underscores the role of the probiotic in diminishing oviductal inflammation, regardless of its state.
Collapse
Affiliation(s)
- Shinuo Li
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Qingfeng Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Jinqiu Mi
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Haotian Chen
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Tianhao Yuan
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Yue Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Lihong Zhao
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Qiugang Ma
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Shimeng Huang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| |
Collapse
|
4
|
Li H, Han M, Xu J, Li N, Cui H. The vaginal microbial signatures of preterm birth woman. BMC Pregnancy Childbirth 2024; 24:428. [PMID: 38877389 PMCID: PMC11177453 DOI: 10.1186/s12884-024-06573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 05/10/2024] [Indexed: 06/16/2024] Open
Abstract
To explore the differences of vaginal microbes in women with preterm birth (PTB), and to construct prediction model. We searched for articles related to vaginal microbiology in preterm women and obtained four 16S rRNA-sequence datasets. We analyzed that for species diversity and differences, and constructed a random forest model with 20 differential genera. We introduce an independent whole genome-sequencing (WGS) data for validation. In addition, we collected vaginal and cervical swabs from 33 pregnant women who delivered spontaneously full-term and preterm infants, performed WGS in our lab to further validate the model. Compared to term birth (TB) samples, PTB women vagina were characterized by a decrease in Firmicutes, Lactobacillus, and an increase in diversity accompanied by the colonization of pathogenic bacteria such as Gardnerella, Atopobium and Prevotella. Twenty genus markers, including Lactobacillus, Prevotella, Streptococcus, and Gardnerella performed well in predicting PTB, with study-to-study transfer validation and LODO validation, different gestation validation showing good results, and in two independent cohorts (external WGS cohorts and woman samples WGS cohorts) in which the accuracy was maintained. PTB women have unique vaginal microbiota characteristics. A predictive model of PTB was constructed and its value validated from multiple perspectives.
Collapse
Affiliation(s)
- Huan Li
- Department of Obstetrics and Gynecology, Research Center, Shengjing Hospital of China Medical University, China Medical University Birth Cohort, Shenyang, 110004, China
| | - Mengzhen Han
- Department of Breast Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Junnan Xu
- Department of Breast Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Na Li
- Department of Obstetrics and Gynecology, Research Center, Shengjing Hospital of China Medical University, China Medical University Birth Cohort, Shenyang, 110004, China.
| | - Hong Cui
- Department of Obstetrics and Gynecology, Research Center, Shengjing Hospital of China Medical University, China Medical University Birth Cohort, Shenyang, 110004, China.
| |
Collapse
|
5
|
Elovitz M, Anton L, Cristancho A, Ferguson B, Joseph A, Ravel J. Vaginal microbes alter epithelial transcriptome and induce epigenomic modifications providing insight into mechanisms for susceptibility to adverse reproductive outcomes. RESEARCH SQUARE 2024:rs.3.rs-4385224. [PMID: 38854063 PMCID: PMC11160883 DOI: 10.21203/rs.3.rs-4385224/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The cervicovaginal microbiome is highly associated with women's health, with microbial communities dominated by Lactobacillus species considered optimal. Conversely, a lack of lactobacilli and a high abundance of strict and facultative anaerobes, including Gardnerella vaginalis, have been associated with adverse reproductive outcomes. However, how host-microbial interactions alter specific molecular pathways and impact cervical and vaginal epithelial function remains unclear. Using RNA-sequencing, we characterized the in vitro cervicovaginal epithelial transcriptional response to different vaginal bacteria and their culture supernatants. We showed that G. vaginalis upregulates genes associated with an activated innate immune response. Unexpectedly, G. vaginalis specifically induced inflammasome pathways through activation of NLRP3-mediated increases in caspase-1, IL-1β and cell death, while live L. crispatus had minimal transcriptomic changes on epithelial cells. L. crispatus culture supernatants resulted in a shift in the epigenomic landscape of cervical epithelial cells that was confirmed by ATAC-sequencing showing reduced chromatin accessibility. This study reveals new insights into host-microbe interactions in the lower reproductive tract and suggests potential therapeutic strategies leveraging the vaginal microbiome to improve reproductive health.
Collapse
|
6
|
McKee K, Bassis CM, Golob J, Palazzolo B, Sen A, Comstock SS, Rosas-Salazar C, Stanford JB, O'Connor T, Gern JE, Paneth N, Dunlop AL. Host factors are associated with vaginal microbiome structure in pregnancy in the ECHO Cohort Consortium. Sci Rep 2024; 14:11798. [PMID: 38782975 PMCID: PMC11116393 DOI: 10.1038/s41598-024-62537-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Using pooled vaginal microbiota data from pregnancy cohorts (N = 683 participants) in the Environmental influences on Child Health Outcomes (ECHO) Program, we analyzed 16S rRNA gene amplicon sequences to identify clinical and demographic host factors that associate with vaginal microbiota structure in pregnancy both within and across diverse cohorts. Using PERMANOVA models, we assessed factors associated with vaginal community structure in pregnancy, examined whether host factors were conserved across populations, and tested the independent and combined effects of host factors on vaginal community state types (CSTs) using multinomial logistic regression models. Demographic and social factors explained a larger amount of variation in the vaginal microbiome in pregnancy than clinical factors. After adjustment, lower education, rather than self-identified race, remained a robust predictor of L. iners dominant (CST III) and diverse (CST IV) (OR = 8.44, 95% CI = 4.06-17.6 and OR = 4.18, 95% CI = 1.88-9.26, respectively). In random forest models, we identified specific taxonomic features of host factors, particularly urogenital pathogens associated with pregnancy complications (Aerococcus christensenii and Gardnerella spp.) among other facultative anaerobes and key markers of community instability (L. iners). Sociodemographic factors were robustly associated with vaginal microbiota structure in pregnancy and should be considered as sources of variation in human microbiome studies.
Collapse
Grants
- U24OD023382 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- U2C OD023375 NIH HHS
- UH3 OD023271 NIH HHS
- UH3 OD023282 NIH HHS
- UH3OD023282 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- UH3 OD023287 NIH HHS
- U24 OD023319 NIH HHS
- UH3 OD023305 NIH HHS
- K01 AI153558 NIAID NIH HHS
- UH3 OD023288 NIH HHS
- UH3OD023249 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- U24OD023319 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- UH3 OD023349 NIH HHS
- UH3 OD023337 NIH HHS
- UH3 OD023328 NIH HHS
- U24 OD023382 NIH HHS
- UH3 OD023313 NIH HHS
- UH3 OD023289 NIH HHS
- UH3 OD023249 NIH HHS
- UH3 OD023389 NIH HHS
- UH3 OD023290 NIH HHS
- UH3OD023251 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- UH3 OD023285 NIH HHS
- UH3 OD023275 NIH HHS
- UH3 OD023318 NIH HHS
- UH3 OD023248 NIH HHS
- U2COD023375 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- UH3 OD023253 NIH HHS
- UH3 OD023272 NIH HHS
- UH3 OD023347 NIH HHS
- UH3OD023318 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- UH3 OD023251 NIH HHS
- UH3 OD023279 NIH HHS
- UH3OD023285 Environmental influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health
- UH3 OD023244 NIH HHS
- UH3 OD023320 NIH HHS
Collapse
Affiliation(s)
- Kimberly McKee
- Department of Family Medicine, University of Michigan, 1018 Fuller St, Ann Arbor, MI, 48104, USA.
| | - Christine M Bassis
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Golob
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Beatrice Palazzolo
- Department of Family Medicine, University of Michigan, 1018 Fuller St, Ann Arbor, MI, 48104, USA
| | - Ananda Sen
- Department of Family Medicine, University of Michigan, 1018 Fuller St, Ann Arbor, MI, 48104, USA
| | - Sarah S Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | | | - Joseph B Stanford
- Department of Family and Preventive Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Thomas O'Connor
- Departments of Neuroscience and Obstetrics & Gynecology, University of Rochester, Rochester, NY, USA
| | - James E Gern
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Nigel Paneth
- Departments of Epidemiology & Biostatistics and Pediatrics & Human Development, Michigan State University, East Lansing, MI, USA
| | - Anne L Dunlop
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
7
|
Moore KA, Petersen AP, Zierden HC. Microorganism-derived extracellular vesicles: emerging contributors to female reproductive health. NANOSCALE 2024; 16:8216-8235. [PMID: 38572613 DOI: 10.1039/d3nr05524h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that carry small molecules, nucleic acids, and proteins long distances in the body facilitating cell-cell communication. Microorganism-derived EVs mediate communication between parent cells and host cells, with recent evidence supporting their role in biofilm formation, horizontal gene transfer, and suppression of the host immune system. As lipid-bound bacterial byproducts, EVs demonstrate improved cellular uptake and distribution in vivo compared to cell-free nucleic acids, proteins, or small molecules, allowing these biological nanoparticles to recapitulate the effects of parent cells and contribute to a range of human health outcomes. Here, we focus on how EVs derived from vaginal microorganisms contribute to gynecologic and obstetric outcomes. As the composition of the vaginal microbiome significantly impacts women's health, we discuss bacterial EVs from both healthy and dysbiotic vaginal microbiota. We also examine recent work done to evaluate the role of EVs from common vaginal bacterial, fungal, and parasitic pathogens in pathogenesis of female reproductive tract disease. We highlight evidence for the role of EVs in women's health, gaps in current knowledge, and opportunities for future work. Finally, we discuss how leveraging the innate interactions between microorganisms and mammalian cells may establish EVs as a novel therapeutic modality for gynecologic and obstetric indications.
Collapse
Affiliation(s)
- Kaitlyn A Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA.
| | - Alyssa P Petersen
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Hannah C Zierden
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA.
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD, 20742, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
8
|
Gerson KD, Loder A, Landau Z, Anton L. Xenobiotic metabolites modify immune responses of the cervicovaginal epithelium: potential mechanisms underlying barrier disruption. BJOG 2024; 131:665-674. [PMID: 37705143 DOI: 10.1111/1471-0528.17654] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/28/2023] [Accepted: 08/14/2023] [Indexed: 09/15/2023]
Abstract
OBJECTIVE Xenobiotic metabolites are exogenous biochemicals that can adversely impact reproductive health. We previously identified xenobiotics in cervicovaginal fluid during pregnancy in association with short cervix. In other organ systems, xenobiotics can modify epithelial barrier function. We hypothesise that xenobiotics dysregulate epithelial cell and macrophage immune responses as a mechanism to disrupt the cervicovaginal barrier. DESIGN In vitro cell culture system. SETTING Laboratory within academic institution. SAMPLE Vaginal, ectocervical and endocervical epithelial cell lines and primary macrophages. METHODS Cells were treated with diethanolamine (2.5 mM), ethyl glucoside (5 mM) or tartrate (2.5 mM) for 24 h. MAIN OUTCOME MEASURES Cytokines and matrix metalloproteinases were measured in cell supernatants (n = 3 per condition). One-way analysis of variance (ANOVA) with Dunnett's test for multiple comparisons was performed. RESULTS Diethanolamine induces inflammatory cytokines, whereas ethyl glucoside and tartrate generally exert anti-inflammatory effects across all cells. Diethanolamine increases interleukin 6 (IL-6), IL-8, interferon γ-induced protein 10 kDa (IP-10), growth-regulated oncogene (GRO), fractalkine, matrix metalloproteinase 1 (MMP-1), MMP-9 and MMP-10 (p < 0.05 for all), factors involved in acute inflammation and recruitment of monocytes, neutrophils and lymphocytes. Ethyl glucoside and tartrate decrease multiple cytokines, including RANTES and MCP-1 (p < 0.05 for all), which serve as chemotactic factors. Vaginal cells exhibit heightened inflammatory tone compared with cervical cells and macrophages, with a greater number of differentially expressed analytes after xenobiotic exposure. CONCLUSIONS Xenobiotic metabolites present in the cervicovaginal space during pregnancy modify immune responses, unveiling potential pathways through which environmental exposures may contribute to the pathogenesis of cervical remodelling preceding preterm birth. Future work identifying xenobiotic sources and routes of exposure offers the potential to modify environmental risks to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Kristin D Gerson
- Center for Research on Reproduction and Women's Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Aaron Loder
- Center for Research on Reproduction and Women's Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zachary Landau
- Center for Research on Reproduction and Women's Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lauren Anton
- Center for Research on Reproduction and Women's Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Gao H, Liu Q, Wang X, Li T, Li H, Li G, Tan L, Chen Y. Deciphering the role of female reproductive tract microbiome in reproductive health: a review. Front Cell Infect Microbiol 2024; 14:1351540. [PMID: 38562966 PMCID: PMC10982509 DOI: 10.3389/fcimb.2024.1351540] [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: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
Relevant studies increasingly indicate that female reproductive health is confronted with substantial challenges. Emerging research has revealed that the microbiome interacts with the anatomy, histology, and immunity of the female reproductive tract, which are the cornerstone of maintaining female reproductive health and preventing adverse pregnancy outcomes. Currently, the precise mechanisms underlying their interaction and impact on physiological functions of the reproductive tract remain elusive, constituting a prominent area of investigation within the field of female reproductive tract microecology. From this new perspective, we explore the mechanisms of interactions between the microbiome and the anatomy, histology, and immunity of the female reproductive tract, factors that affect the composition of the microbiome in the female reproductive tract, as well as personalized medicine approaches in managing female reproductive tract health based on the microbiome. This study highlights the pivotal role of the female reproductive tract microbiome in maintaining reproductive health and influencing the occurrence of reproductive tract diseases. These findings support the exploration of innovative approaches for the prevention, monitoring and treatment of female reproductive tract diseases based on the microbiome.
Collapse
Affiliation(s)
- Hong Gao
- Nursing Department, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| | - Qiao Liu
- School of Nursing, University of South China, Hengyang, China
| | - Xiaolan Wang
- Center for a Combination of Obstetrics and Gynecology and Reproductive Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ting Li
- Department of Obstetrics, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Huanhuan Li
- Department of Gynaecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Genlin Li
- Center for a Combination of Obstetrics and Gynecology and Reproductive Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Lingling Tan
- Nursing Department, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yahui Chen
- School of Nursing, University of South China, Hengyang, China
| |
Collapse
|
10
|
Burris HH, Yang N, Riis V, Valeri L, South EC, Ravel J, Elovitz MA. The role of neighborhood deprivation in the cervicovaginal microbiota. Am J Obstet Gynecol MFM 2024; 6:101291. [PMID: 38246324 PMCID: PMC10948309 DOI: 10.1016/j.ajogmf.2024.101291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND Lactobacillus-deficient cervicovaginal microbiota is associated with spontaneous preterm birth and is more common among Black individuals. Persistent racial segregation in the United States has led to differential neighborhood exposures by race that can affect pregnancy outcomes. The extent to which neighborhood exposures may explain racial differences in the cervicovaginal microbiota is unknown. OBJECTIVE This study aimed to determine whether neighborhood deprivation, defined as material community deprivation, is associated with a Lactobacillus-deficient cervicovaginal microbiota in a prospective cohort of pregnant individuals. Our hypothesis was that racial differences in neighborhood deprivation may explain the higher prevalence of Lactobacillus-deficient cervicovaginal microbiota in Black birthing people. STUDY DESIGN This study analyzed data from Motherhood and Microbiome, a prospective pregnancy cohort enrolled from prenatal clinics in a single hospital system 2013-2016 in which a Lactobacillus-deficient cervicovaginal microbiota was previously shown to be associated with spontaneous preterm birth. This study geocoded addresses to obtain census tract neighborhood deprivation data from the Brokamp Nationwide Community Deprivation Index that uses weighted proportions of poverty, income, public assistance, lack of health insurance, and vacant housing. Generalized linear mixed models quantified associations of deprivation with the cervicovaginal microbiota accounting for geographic clustering by census tract and potential confounders. Because of different distributions of neighborhood deprivation and the cervicovaginal microbiota, race-stratified models were used. Mediation analyses quantified the extent to which deprivation may contribute to racial differences in the cervicovaginal microbiota. RESULTS Higher neighborhood deprivation was associated with a Lactobacillus-deficient cervicovaginal microbiota. Per standard deviation increment of deprivation, participants had 28% higher adjusted odds (adjusted odds ratio, 1.28; 95% confidence interval, 1.04-1.58) of a Lactobacillus-deficient microbiota. Black participants had higher odds of a Lactobacillus-deficient microbiota than White participants (adjusted odds ratio, 4.00; 95% confidence interval, 2.05-8.26), and mediation analysis revealed that deprivation accounted for 22% (P=.046) of that disparity. CONCLUSION Neighborhood deprivation was associated with Lactobacillus-deficient cervicovaginal microbiota and may partially explain Black-White disparities in the cervicovaginal microbiota. Mechanistic studies to explore how environmental exposures modify the cervicovaginal microbiota are warranted to identify novel opportunities for future interventional strategies to prevent preterm birth. As the findings demonstrate a potential biological effect from neighborhood conditions, policies that drive urban planning should be explored to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Heather H Burris
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA (Dr Burris); Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (Dr Burris); Leonard Davis Institute for Health Economics, University of Pennsylvania, Philadelphia, PA (Dr Burris).
| | - Nancy Yang
- University of California San Francisco Medical School, San Francisco, CA (Ms. Yang)
| | - Valerie Riis
- Women's Biomedical Research Institute, Icahn School of Medicine, New York, NY (Ms. Riis and Dr. Elovitz)
| | - Linda Valeri
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY (Dr. Valeri)
| | - Eugenia C South
- Penn Urban Health Lab, Department of Emergency Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (Dr. South)
| | - Jacques Ravel
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD (Dr. Ravel)
| | - Michal A Elovitz
- Women's Biomedical Research Institute, Icahn School of Medicine, New York, NY (Ms. Riis and Dr. Elovitz); Department of Obstetrics, Gynecology, and Reproductive Sciences, Icahn School of Medicine, New York, NY (Dr. Elovitz)
| |
Collapse
|
11
|
Huang X, Lin R, Mao B, Tang X, Zhao J, Zhang Q, Cui S. Lactobacillus crispatus CCFM1339 Inhibits Vaginal Epithelial Barrier Injury Induced by Gardnerella vaginalis in Mice. Biomolecules 2024; 14:240. [PMID: 38397477 PMCID: PMC10886512 DOI: 10.3390/biom14020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The vaginal epithelial barrier, which integrates mechanical, immune, chemical, and microbial defenses, is pivotal in safeguarding against external pathogens and upholding the vaginal microecological equilibrium. Although the widely used metronidazole effectively curtails Gardnerella vaginalis, a key pathogen in bacterial vaginosis, it falls short in restoring the vaginal barrier or reducing recurrence rates. Our prior research highlighted Lactobacillus crispatus CCFM1339, a vaginally derived Lactobacillus strain, for its capacity to modulate the vaginal epithelial barrier. In cellular models, L. crispatus CCFM1339 fortified the integrity of the cellular monolayer, augmented cellular migration, and facilitated repair. Remarkably, in animal models, L. crispatus CCFM1339 substantially abated the secretion of the barrier disruption biomarker E-cadherin (from 101.45 to 82.90 pg/mL) and increased the anti-inflammatory cytokine IL-10 (35.18% vs. the model), consequently mitigating vaginal inflammation in mice. Immunological assays in vaginal tissues elucidated increased secretory IgA levels (from 405.56 to 740.62 ng/mL) and curtailed IL-17 gene expression. Moreover, L. crispatus CCFM1339 enhanced Lactobacilli abundance and attenuated Enterobacterium and Enterococcus within the vaginal microbiome, underscoring its potential in probiotic applications for vaginal barrier regulation.
Collapse
Affiliation(s)
- Xiaoyan Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
| | - Rumeng Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (X.H.); (R.L.); (B.M.); (X.T.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
12
|
Ottinger S, Robertson CM, Branthoover H, Patras KA. The human vaginal microbiota: from clinical medicine to models to mechanisms. Curr Opin Microbiol 2024; 77:102422. [PMID: 38215548 PMCID: PMC11160953 DOI: 10.1016/j.mib.2023.102422] [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: 09/27/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/14/2024]
Abstract
The composition of the vaginal microbiota is linked to numerous reproductive health problems, including increased susceptibility to infection, pregnancy complications, and impaired vaginal tissue repair; however, the mechanisms contributing to these adverse outcomes are not yet fully defined. In this review, we highlight recent clinical advancements associating vaginal microbiome composition and function with health outcomes. Subsequently, we provide a summary of emerging models employed to identify microbe-microbe interactions contributing to vaginal health, including metagenomic sequencing, multi-omics approaches, and advances in vaginal microbiota cultivation. Last, we review new in vitro, ex vivo, and in vivo models, such as organoids and humanized microbiota murine models, used to define and mechanistically explore host-microbe interactions at the vaginal mucosa.
Collapse
Affiliation(s)
- Samantha Ottinger
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Clare M Robertson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Holly Branthoover
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Onyango S, Mi JD, Koech A, Okiro P, Temmerman M, von Dadelszen P, Tribe RM, Omuse G. Microbiota dynamics, metabolic and immune interactions in the cervicovaginal environment and their role in spontaneous preterm birth. Front Immunol 2023; 14:1306473. [PMID: 38196946 PMCID: PMC10774218 DOI: 10.3389/fimmu.2023.1306473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/11/2023] [Indexed: 01/11/2024] Open
Abstract
Differences in the cervicovaginal microbiota are associated with spontaneous preterm birth (sPTB), a significant cause of infant morbidity and mortality. Although establishing a direct causal link between cervicovaginal microbiota and sPTB remains challenging, recent advancements in sequencing technologies have facilitated the identification of microbial markers potentially linked to sPTB. Despite variations in findings, a recurring observation suggests that sPTB is associated with a more diverse and less stable vaginal microbiota across pregnancy trimesters. It is hypothesized that sPTB risk is likely to be modified via an intricate host-microbe interactions rather than due to the presence of a single microbial taxon or broad community state. Nonetheless, lactobacilli dominance is generally associated with term outcomes and contributes to a healthy vaginal environment through the production of lactic acid/maintenance of a low pH that excludes other pathogenic microorganisms. Additionally, the innate immunity of the host and metabolic interactions between cervicovaginal microbiota, such as the production of bacteriocins and the use of proteolytic enzymes, exerts a profound influence on microbial populations, activities, and host immune responses. These interplays collectively impact pregnancy outcomes. This review aims to summarize the complexity of cervicovaginal environment and microbiota dynamics, and associations with bacterial vaginosis and sPTB. There is also consideration on how probiotics may mitigate the risk of sPTB and bacterial vaginosis.
Collapse
Affiliation(s)
- Stanley Onyango
- Department of Pathology, Aga Khan University, Nairobi, Kenya
- Centre of Excellence Women and Child Health, Aga Khan University, Nairobi, Kenya
| | - Jia Dai Mi
- Faculty of Life Sciences and Medicine, Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, London, United Kingdom
| | - Angela Koech
- Centre of Excellence Women and Child Health, Aga Khan University, Nairobi, Kenya
| | - Patricia Okiro
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | - Marleen Temmerman
- Centre of Excellence Women and Child Health, Aga Khan University, Nairobi, Kenya
| | - Peter von Dadelszen
- Faculty of Life Sciences and Medicine, Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, London, United Kingdom
| | - Rachel M. Tribe
- Faculty of Life Sciences and Medicine, Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, London, United Kingdom
| | - Geoffrey Omuse
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | | |
Collapse
|
14
|
Galaz J, Romero R, Greenberg JM, Theis KR, Arenas-Hernandez M, Xu Y, Farias-Jofre M, Miller D, Kanninen T, Garcia-Flores V, Gomez-Lopez N. Host-microbiome interactions in distinct subsets of preterm labor and birth. iScience 2023; 26:108341. [PMID: 38047079 PMCID: PMC10692673 DOI: 10.1016/j.isci.2023.108341] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/06/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
Preterm birth, the leading cause of perinatal morbidity, often follows premature labor, a syndrome whose prevention remains a challenge. To better understand the relationship between premature labor and host-microbiome interactions, we conducted a mechanistic investigation using three preterm birth models. We report that intra-amniotic delivery of LPS triggers inflammatory responses in the amniotic cavity and cervico-vaginal microenvironment, causing vaginal microbiome changes and signs of active labor. Intra-amniotic IL-1α delivery causes a moderate inflammatory response in the amniotic cavity but increasing inflammation in the cervico-vaginal space, leading to vaginal microbiome disruption and signs of active labor. Conversely, progesterone action blockade by RU-486 triggers local immune responses accompanying signs of active labor without altering the vaginal microbiome. Preterm labor facilitates ascension of cervico-vaginal bacteria into the amniotic cavity, regardless of stimulus. This study provides compelling mechanistic insights into the dynamic host-microbiome interactions within the cervico-vaginal microenvironment that accompany premature labor and birth.
Collapse
Affiliation(s)
- Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Jonathan M. Greenberg
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kevin R. Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tomi Kanninen
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
15
|
Ardizzone CM, Taylor CM, Toh E, Lillis RA, Elnaggar JH, Lammons JW, Mott PD, Duffy EL, Shen L, Quayle AJ. Association of Chlamydia trachomatis burden with the vaginal microbiota, bacterial vaginosis, and metronidazole treatment. Front Cell Infect Microbiol 2023; 13:1289449. [PMID: 38149008 PMCID: PMC10750252 DOI: 10.3389/fcimb.2023.1289449] [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: 09/05/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023] Open
Abstract
Bacterial vaginosis (BV), a dysbiosis of the vaginal microbiota, is a common coinfection with Chlamydia trachomatis (Ct), and BV-associated bacteria (BVAB) and their products have been implicated in aiding Ct evade natural immunity. Here, we determined if a non-optimal vaginal microbiota was associated with a higher genital Ct burden and if metronidazole, a standard treatment for BV, would reduce Ct burden or aid in natural clearance of Ct infection. Cervicovaginal samples were collected from women at enrollment and, if testing positive for Ct infection, at a follow-up visit approximately one week later. Cervical Ct burden was assessed by inclusion forming units (IFU) and Ct genome copy number (GCN), and 16S rRNA gene sequencing was used to determine the composition of the vaginal microbiota. We observed a six-log spectrum of IFU and an eight-log spectrum of GCN in our study participants at their enrollment visit, but BV, as indicated by Amsel's criteria, Nugent scoring, or VALENCIA community state typing, did not predict infectious and total Ct burden, although IFU : GCN increased with Amsel and Nugent scores and in BV-like community state types. Ct burden was, however, associated with the abundance of bacterial species in the vaginal microbiota, negatively with Lactobacillus crispatus and positively with Prevotella bivia. Women diagnosed with BV were treated with metronidazole, and Ct burden was significantly reduced in those who resolved BV with treatment. A subset of women naturally cleared Ct infection in the interim, typified by low Ct burden at enrollment and resolution of BV. Abundance of many BVAB decreased, and Lactobacillus increased, in response to metronidazole treatment, but no changes in abundances of specific vaginal bacteria were unique to women who spontaneously cleared Ct infection.
Collapse
Affiliation(s)
- Caleb M. Ardizzone
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Christopher M. Taylor
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Evelyn Toh
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rebecca A. Lillis
- Department of Medicine, Section of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jacob H. Elnaggar
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - John W. Lammons
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Patricia Dehon Mott
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Emily L. Duffy
- Department of Medicine, Section of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Li Shen
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Alison J. Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
16
|
Elovitz M, Anton L, Cristancho A, Ferguson B, Joseph A, Ravel J. Vaginal microbes alter epithelial transcriptomic and epigenomic modifications providing insight into the molecular mechanisms for susceptibility to adverse reproductive outcomes. RESEARCH SQUARE 2023:rs.3.rs-3580132. [PMID: 38014044 PMCID: PMC10680926 DOI: 10.21203/rs.3.rs-3580132/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The cervicovaginal microbiome is highly associated with women's health with microbial communities dominated by Lactobacillus spp. being considered optimal. Conversely, a lack of lactobacilli and a high abundance of strict and facultative anaerobes including Gardnerella vaginalis , have been associated with adverse reproductive outcomes. However, the molecular pathways modulated by microbe interactions with the cervicovaginal epithelia remain unclear. Using RNA-sequencing, we characterize the in vitro cervicovaginal epithelial transcriptional response to different vaginal bacteria and their culture supernatants. We showed that G. vaginalis upregulated genes were associated with an activated innate immune response including anti-microbial peptides and inflammasome pathways, represented by NLRP3-mediated increases in caspase-1, IL-1β and cell death. Cervicovaginal epithelial cells exposed to L. crispatus showed limited transcriptomic changes, while exposure to L. crispatus culture supernatants resulted in a shift in the epigenomic landscape of cervical epithelial cells. ATAC-sequencing confirmed epigenetic changes with reduced chromatin accessibility. This study reveals new insight into host-microbe interactions in the lower reproductive tract and suggest potential therapeutic strategies leveraging the vaginal microbiome to improve reproductive health.
Collapse
|
17
|
Schwecht I, Nazli A, Gill B, Kaushic C. Lactic acid enhances vaginal epithelial barrier integrity and ameliorates inflammatory effects of dysbiotic short chain fatty acids and HIV-1. Sci Rep 2023; 13:20065. [PMID: 37973920 PMCID: PMC10654711 DOI: 10.1038/s41598-023-47172-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
The vaginal microenvironment is key in mediating susceptibility to sexually transmitted infections. A polymicrobial environment with reduced Lactobacilllus spp. is characteristic of vaginal dysbiosis, associated with increased production of several short chain fatty acids (SCFAs), vaginal inflammation and an increased risk of HIV-1 acquisition. In contrast, a eubiotic vaginal microbiome (VMB), dominated by Lactobacillus spp. correlates with increased production of lactic acid (LA), an acidic milieu and protection against HIV-1. Vaginal metabolites, specifically LA and SCFAs including butyric, succinic and acetic acids are associated with modulation of HIV-1 risk. We assessed the impact of combined and individual SCFAs and LA on vaginal epithelial cells (VK2) grown in air-liquid interface cultures. Treatment of VK2 cells with eubiotic SCFA + LA mixture showed increased epithelial barrier integrity, reduced FITC dextran leakage and enhanced expression of cell-cell adhesion proteins. Treatment with dysbiotic SCFA + LA mixture diminished epithelial barrier integrity, increased NFκB activation and inflammatory mediators: TNF-α, IL-6, IL-8 and RANTES. LA was found to be the primary contributor of the beneficial effects. Eubiotic SCFA + LA mixture ameliorated HIV-1 mediated barrier disruption and HIV-1 leakage, whereas dysbiotic SCFA + LA treatment exacerbated HIV-1 effects. These findings indicate a key role for LA in future prophylactic strategies.
Collapse
Affiliation(s)
- Ingrid Schwecht
- 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
| | - Biban Gill
- 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
|
18
|
Zhu B, Spaine KM, Edupuganti L, Matveyev A, Serrano MG, Buck GA. Characteristics of vaginal microbes and classification of the vaginal microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553525. [PMID: 37645743 PMCID: PMC10462076 DOI: 10.1101/2023.08.16.553525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Background The vaginal microbiome (VMB) has been classified into several discrete community state types, some of which have been associated with adverse human health conditions. However, the roles of the many vaginal bacteria in modulating the VMB and health remain unclear. Methods The associations among the vaginal taxa and other vaginal taxa, the vaginal pH, and the host gene expression responses were determined by calculating the correlation among the relative abundance of the vaginal taxa, the association between the vaginal pH and the predominant taxon in the VMB, and the correlation between the relative abundance of the vaginal taxa and human gene expression at the transcriptional level, respectively. Using these associations, an alternative more informative method, the biological vagitype (BVT), is proposed to classify community state types of the VMB. Findings Most Lactobacillus spp., with the exception of Lactobacillus iners , show significant correlations with host gene expression profiles and negative associations with dysbiosis-associated vaginal taxa. Many non- Lactobacillus spp. exhibit varied correlations with Lactobacillus spp., the vaginal pH, and host gene expression. Compared to other dysbiotic taxa, including Candidatus Lachnocurva vaginae, Gardnerella vaginalis has a stronger positive correlation with vaginal pH and a stronger negative correlation with Lactobacillus spp. Most dysbiosis-associated taxa are associated with stress responses of the host at the transcriptional level, but the genus Mycoplasma has a uniquely strong positive correlation with host immune responses. The association between BVTs of the VMBs and host characteristics, e.g., race/ethnicity, microbial infection, smoking, antibiotics, high blood pressure, economic state, diet, and others, was examined. The BVT classification method improved overall performance in associating specific vaginal microbial populations with host characteristics and phenotypes. Interpretation This study sheds light on the biological characteristics of the vaginal microbiota, including some less abundant or still unculturable taxa. Since the BVT method was established based on these biological characteristics, the classification outcome of the VMB may have more clinical relevance. Because the BVT method performs better in associating specific vaginal community types with diseases, e.g., bacterial vaginosis and gonorrhea, it could be beneficial for the predictive modeling of adverse health. Funding This work was supported by grants [UH3AI083263, U54HD080784, and R01HD092415] from the National Institutes of Health; and support from the [GAPPS BMGF PPB] grant from the Global Alliance to Prevent Prematurity and Stillbirth. We would also like to thank the Office of Research on Women's Health at NIH for their generous support. Research in context Evidence before this study: The vaginal microbiome (VMB) refers to the community of microorganisms in the female lower reproductive tract. The VMB is often a simple ecosystem dominated by a single species. The most predominant bacteria in the VMB include several Lactobacillus species and two non- Lactobacillus species, i.e., Candidatus Lachnocurva vaginae and Gardnerella vaginalis. Lactobacillus species produce lactic acid to lower the vaginal pH and inhibit the growth of disease-associated bacteria. Thus, the predominance of protective Lactobacilli, i.e., L. crispatus, L. jensenii , and L. gasseri , in the VMB is associated with overall vaginal health. However, the role of L. iners in promoting a healthy vaginal ecosystem is less clear. Actually, the biological and health relevance of many bacteria in the female lower reproductive tract is largely unknown. Some bacteria have low relative abundances, e.g., Peptostreptococcus and Coriobacteriaceae spp.; and others are not yet culturable, e.g., Candidatus Lachnocurva vaginae and BVAB TM7. When abundance of a taxon is low, its association with a host characteristic is a challenge. Previous methods to classify the VMB were based simply on their microbial compositions, and the biological characteristics of the vaginal bacteria were largely ignored. Thus, classification of these VMBs into biologically relevant community types, as described herein, should be helpful in determining their relevance to women's reproductive health. Added value of this study: This study examines three biological characteristics of bacteria in the VMB, i.e., the associations among different bacterial taxa, the vaginal pH, and the host response. Based on these three characteristics, the influence of these bacteria, particularly low abundant and unculturable bacteria, on vaginal health is evaluated. L. iners seems to be neutral in maintaining overall vaginal health. Gardnerella vaginalis is apparently more easily inhibited by Lactobacillus spp. than Candidatus Lachnocurva vaginae because of its stronger positive correlation with vaginal pH and negative correlation with Lactobacillus . The genus of Mycoplasma has a unique positive correlation with local immune responses, implying a role for Mycoplasma in promoting inflammation. Compared with previous methods to classify the VMB, a new method, considering the above three biological characteristics of bacteria in the VMB, has been established. The new method performs better in associating specific vaginal communities with host characteristics and phenotypes; e.g., bacterial vaginosis and gonorrhea. Implications of all the available evidence: Accurate biological classification of the VMB is fundamental for assessing its impact on women's health. Our classification scheme represents a step further toward that correct classification, eventually leading to new strategies for clinical assessment of the potential use of the VMB to diagnose or predict women's reproductive health.
Collapse
|
19
|
Joseph A, Lewis EL, Ferguson B, Guan Y, Anton L, Elovitz MA. Intrauterine colonization with Gardnerella vaginalis and Mobiluncus mulieris induces maternal inflammation but not preterm birth in a mouse model. Am J Reprod Immunol 2023; 90:e13749. [PMID: 37491927 PMCID: PMC11423284 DOI: 10.1111/aji.13749] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Preterm birth (PTB) remains a leading cause of childhood mortality. Recent studies demonstrate that the risk of spontaneous PTB (sPTB) is increased in individuals with Lactobacillus-deficient vaginal microbial communities. One proposed mechanism is that vaginal microbes ascend through the cervix, colonize the uterus, and activate inflammatory pathways leading to sPTB. This study assessed whether intrauterine colonization with either Gardnerella vaginalis and Mobiluncus mulieris alone is sufficient to induce maternal-fetal inflammation and induce sPTB. METHOD OF STUDY C56/B6J mice, on embryonic day 15, received intrauterine inoculation of saline or 108 colony-forming units of G. vaginalis (n = 30), M. mulieris (n = 17), or Lactobacillus crispatus (n = 16). Dams were either monitored for maternal morbidity and sPTB or sacrificed 6 h post-infusion for analysis of bacterial growth and cytokine/chemokine expression in maternal and fetal tissues. RESULTS Six hours following intrauterine inoculation with G. vaginalis, M. mulieris, or L. crispatus, live bacteria were observed in both blood and amniotic fluid, and a potent immune response was identified in the uterus and maternal serum. In contrast, only a limited immune response was identified in the amniotic fluid and the fetus after intrauterine inoculation. High bacterial load (108 CFU/animal) of G. vaginalis was associated with maternal morbidity and mortality but not sPTB. Intrauterine infusion with L. crispatus or M. mulieris at 108 CFU/animal did not induce sPTB, alter pup viability, litter size, or maternal mortality. CONCLUSIONS Despite inducing an immune response, intrauterine infusion of live G. vaginalis or M. mulieris is not sufficient to induce sPTB in our mouse model. These results suggest that ascension of common vaginal microbes into the uterine cavity alone is not causative for sPTB.
Collapse
Affiliation(s)
- Andrea Joseph
- Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn School of Medicine, New York, New York, USA
| | - Emma L Lewis
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Briana Ferguson
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yuxia Guan
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lauren Anton
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michal A Elovitz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn School of Medicine, New York, New York, USA
| |
Collapse
|
20
|
Doroftei B, Ilie OD, Armeanu T, Stoian IL, Anton N, Babici RG, Ilea C. A Narrative Review Discussing the Obstetric Repercussions Due to Alterations of Personalized Bacterial Sites Developed within the Vagina, Cervix, and Endometrium. J Clin Med 2023; 12:5069. [PMID: 37568471 PMCID: PMC10419759 DOI: 10.3390/jcm12155069] [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: 05/31/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND The reproductive tract microbiota that evolved as an integrative component has been studied intensively in the last decade. As a result, novel research, clinical opportunities, and perspectives have been derived following the close investigation of this microecological environment. This has paved the way for an update to and improvement of the management strategies and therapeutic approaches. However, obscurities, contradictions, and controversies arise regarding the ascension route from the vagina to the endometrium via the cervix, with finality in adverse obstetric outcomes. METHODS Starting from these considerations, we aimed to gather all existing data and information from four major academic databases (PubMed, ISI Web of Knowledge, Scopus, and ScienceDirect) published in the last 13 years (2010-2023) using a controlled vocabulary and dedicated terminology to enhance the coverage, identification, and sorting of potentially eligible studies. RESULTS Despite the high number of returned entries (n = 804), only a slight percentage (2.73%) of all manuscripts were deemed eligible following two rounds of evaluation. Cumulatively, a low level of Lactobacillus spp. and of other core microbiota members is mandatory, with a possible eubiosis-to-dysbiosis transition leading to an impairment of metabolic and endocrine network homeostasis. This transposes into a change in the pro-inflammatory landscape and activation of signaling pathways due to activity exerted by the bacterial lipopolysaccharides (LPSs)/endotoxins that further reflect a high risk of miscarriage in various stages. While the presence of some pathogenic entities may be suggestive of an adverse obstetric predisposition, there are still pros and cons of the role of specific strains, as only the vagina and cervix have been targeted as opposed to the endometrium, which recently started to be viewed as the key player in the vagina-cervix-endometrium route. Consequently, based on an individual's profile, diet, and regime, antibiotics and probiotics might be practical or not. CONCLUSIONS Resident bacteria have a dual facet and are beneficial for women's health, but, at the same time, relaying on the abundance, richness, and evenness that are definitory indexes standing as intermediaries of a miscarriage.
Collapse
Affiliation(s)
- Bogdan Doroftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street No. 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street No. 3C, 700032 Iasi, Romania
| | | | - Theodora Armeanu
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street No. 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street No. 3C, 700032 Iasi, Romania
| | - Irina-Liviana Stoian
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
| | - Nicoleta Anton
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
| | - Ramona-Geanina Babici
- Department of Genetics, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
| | - Ciprian Ilea
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street No. 34, 700038 Iasi, Romania
| |
Collapse
|
21
|
Santacroce L, Palmirotta R, Bottalico L, Charitos IA, Colella M, Topi S, Jirillo E. Crosstalk between the Resident Microbiota and the Immune Cells Regulates Female Genital Tract Health. Life (Basel) 2023; 13:1531. [PMID: 37511906 PMCID: PMC10381428 DOI: 10.3390/life13071531] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The female genital tract (FGT) performs several functions related to reproduction, but due to its direct exposure to the external environment, it may suffer microbial infections. Both the upper (uterus and cervix) and lower (vagina) FGT are covered by an epithelium, and contain immune cells (macrophages, dendritic cells, T and B lymphocytes) that afford a robust protection to the host. Its upper and the lower part differ in terms of Lactobacillus spp., which are dominant in the vagina. An alteration of the physiological equilibrium between the local microbiota and immune cells leads to a condition of dysbiosis which, in turn, may account for the outcome of FGT infection. Aerobic vaginitis, bacterial vaginosis, and Chlamydia trachomatis are the most frequent infections, and can lead to severe complications in reproduction and pregnancy. The use of natural products, such as probiotics, polyphenols, and lactoferrin in the course of FGT infections is an issue of current investigation. In spite of positive results, more research is needed to define the most appropriate administration, according to the type of patient.
Collapse
Affiliation(s)
- Luigi Santacroce
- Microbiology and Virology Section, Interdisciplinary Department of Medicine, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Raffaele Palmirotta
- Microbiology and Virology Section, Interdisciplinary Department of Medicine, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Lucrezia Bottalico
- Department of Clinical Disciplines, School of Technical Medical Sciences, "Alexander Xhuvani" University of Elbasan, 3001 Elbasan, Albania
| | | | - Marica Colella
- Microbiology and Virology Section, Interdisciplinary Department of Medicine, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Skender Topi
- Department of Clinical Disciplines, School of Technical Medical Sciences, "Alexander Xhuvani" University of Elbasan, 3001 Elbasan, Albania
| | - Emilio Jirillo
- Microbiology and Virology Section, Interdisciplinary Department of Medicine, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| |
Collapse
|
22
|
Costa-Fujishima M, Yazdanpanah A, Horne S, Lamont A, Lopez P, Farr Zuend C, Birse K, Taverner M, Greenslade R, Abou M, Noel-Romas L, Abrenica B, Ajibola O, Ikeogu N, Su RC, McKinnon LR, Pymar H, Poliquin V, Berard AR, Burgener AD, Murooka TT. Nonoptimal bacteria species induce neutrophil-driven inflammation and barrier disruption in the female genital tract. Mucosal Immunol 2023; 16:341-356. [PMID: 37121385 DOI: 10.1016/j.mucimm.2023.04.001] [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: 02/09/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 05/02/2023]
Abstract
Neutrophil recruitment and activation within the female genital tract are often associated with tissue inflammation, loss of vaginal epithelial barrier integrity, and increased risk for sexually transmitted infections, such as HIV-1. However, the direct role of neutrophils on vaginal epithelial barrier function during genital inflammation in vivo remains unclear. Using complementary proteome and immunological analyses, we show high neutrophil influx into the lower female genital tract in response to physiological surges in progesterone, stimulating distinct stromal, immunological, and metabolic signaling pathways. However, despite the release of extracellular matrix-modifying proteases and inflammatory mediators, neutrophils contributed little to physiological mucosal remodeling events such as epithelial shedding or re-epithelialization during transition from diestrus to estrus phase. In contrast, the presence of bacterial vaginosis-associated bacteria resulted in a rapid and sustained neutrophil recruitment, resulting in vaginal epithelial barrier leakage and decreased cell-cell junction protein expression in vivo. Thus, neutrophils are important mucosal sentinels that rapidly respond to various biological cues within the female genital tract, dictating the magnitude and duration of the ensuing inflammatory response at steady state and during disease processes.
Collapse
Affiliation(s)
- Marina Costa-Fujishima
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Atta Yazdanpanah
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Samantha Horne
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Alana Lamont
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada; National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Paul Lopez
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Christina Farr Zuend
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA
| | - Kenzie Birse
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Morgan Taverner
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada
| | - Riley Greenslade
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Max Abou
- National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Laura Noel-Romas
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Bernard Abrenica
- National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Oluwaseun Ajibola
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Nnamdi Ikeogu
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada
| | - Ruey-Chyi Su
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada; National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Lyle R McKinnon
- University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada; Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa; Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Helen Pymar
- University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Vanessa Poliquin
- University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Alicia R Berard
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada
| | - Adam D Burgener
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, USA; University of Manitoba, Department of Obstetrics, Gynecology, and Reproductive Sciences, Winnipeg, Canada; Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas T Murooka
- University of Manitoba, Rady Faculty of Health Sciences, Department of Immunology, Winnipeg, Canada; University of Manitoba, Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Canada.
| |
Collapse
|
23
|
Shvartsman E, Hill JE, Sandstrom P, MacDonald KS. Gardnerella Revisited: Species Heterogeneity, Virulence Factors, Mucosal Immune Responses, and Contributions to Bacterial Vaginosis. Infect Immun 2023; 91:e0039022. [PMID: 37071014 PMCID: PMC10187134 DOI: 10.1128/iai.00390-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
Gardnerella species are associated with bacterial vaginosis (BV) and have been investigated as etiological agents of the condition. Nonetheless, the isolation of this taxon from healthy individuals has raised important questions regarding its etiological role. Recently, using advanced molecular approaches, the Gardnerella genus was expanded to include several different species that exhibit differences in virulence potential. Understanding the significance of these different species with respect to mucosal immunity and the pathogenesis and complications of BV could be crucial to solving the BV enigma. Here, we review key findings regarding the unique genetic and phenotypic diversity within this genus, virulence factors, and effects on mucosal immunity as they stand. We also comment on the relevance of these findings to the proposed role of Gardnerella in BV pathogenesis and in reproductive health and identify key gaps in knowledge that should be explored in the future.
Collapse
Affiliation(s)
- Elinor Shvartsman
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, Manitoba, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Janet E. Hill
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Paul Sandstrom
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, Manitoba, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Kelly S. MacDonald
- Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, Manitoba, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
24
|
Barczyński B, Frąszczak K, Grywalska E, Kotarski J, Korona-Głowniak I. Vaginal and Cervical Microbiota Composition in Patients with Endometrial Cancer. Int J Mol Sci 2023; 24:ijms24098266. [PMID: 37175971 PMCID: PMC10179515 DOI: 10.3390/ijms24098266] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
According to recent data, changes in the vaginal microbiota could affect the risk of gynaecological cancers. Women suffering from endometrial cancer present significant changes in cervicovaginal microbiota composition. The objective of our study was to characterize the cervicovaginal microbiota of women undergoing hysterectomy due to benign disease, atypical hyperplasia, and endometrial cancer; The study included 96 patients, who undergone surgical treatment due to benign uterine disease, precancerous endometrial lesion, and endometrial cancer. Quantitative and qualitative real-time PCR analysis of DNA isolated from vaginal fornix and endocervical canal samples was performed to detect the 19 most commonly identified microorganisms, including different Lactobacillus spp., Atopobium, Bifidobacterium, Chlamydia, and Gardnerella; At least one of the tested microorganisms was identified in 88.5% of vaginal and 83.3% of cervical samples. Lactobacillus iners was significantly more frequent in patients with benign condition, whereas Dialister pneumosintes and Mobiluncus curtisii was more frequent in cancer patients; Mobiluncus curtisi and Dialister pneumosintes, which were identified as significantly more common in endometrial cancer vaginal samples, may be considered as potential endometrial cancer co-factors which promote/stimulate carcinogenesis. However, the exact mechanism of such activity remains unexplained and requires further investigations.
Collapse
Affiliation(s)
- Bartłomiej Barczyński
- 1st Department of Oncological Gynaecology and Gynaecology, Medical University in Lublin, 20-081 Lublin, Poland
| | - Karolina Frąszczak
- 1st Department of Oncological Gynaecology and Gynaecology, Medical University in Lublin, 20-081 Lublin, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University in Lublin, 20-093 Lublin, Poland
| | - Jan Kotarski
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University in Lublin, 20-093 Lublin, Poland
| | - Izabela Korona-Głowniak
- Department of Pharmaceutical Microbiology, Medical University in Lublin, 20-093 Lublin, Poland
| |
Collapse
|
25
|
Armstrong E, Kaul R, Cohen CR. Optimizing the vaginal microbiome as a potential strategy to reduce heterosexual HIV transmission. J Intern Med 2023; 293:433-444. [PMID: 36544257 DOI: 10.1111/joim.13600] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bacterial vaginosis (BV) is a proinflammatory genital condition characterized by high vaginal bacterial diversity and a paucity of Lactobacillus species. BV has been linked to an elevated risk of HIV acquisition among HIV-negative women and of forward HIV transmission to male sex partners among women living with HIV (adjusted hazard ratios of 1.69 and 3.17, respectively), potentially by eliciting genital inflammation in women with BV and their male sex partners. BV is also highly prevalent among women in sub-Saharan Africa, suggesting that BV treatment may have potential as an HIV prevention strategy. BV is typically treated with antibiotics but recurrence rates are high, possibly because treatment does not directly promote Lactobacillus growth. More recently, BV treatment strategies incorporating live biotherapeutic lactobacilli have led to sustained optimization of the vaginal microbiome and a decrease in inflammatory biomarkers previously associated with HIV susceptibility. Future studies are urgently needed to evaluate BV treatment strategies that can optimize the vaginal microbiome in the long term through colonization with H2 O2 -producing vaginal lactobacilli and to assess whether vaginal microbiota optimization is able to reduce the risk of HIV transmission.
Collapse
Affiliation(s)
- Eric Armstrong
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Canada.,Department of Medicine, University Health Network, Toronto, Canada
| | - Craig R Cohen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
26
|
Zierden HC, DeLong K, Zulfiqar F, Ortiz JO, Laney V, Bensouda S, Hernández N, Hoang TM, Lai SK, Hanes J, Burke AE, Ensign LM. Cervicovaginal mucus barrier properties during pregnancy are impacted by the vaginal microbiome. Front Cell Infect Microbiol 2023; 13:1015625. [PMID: 37065197 PMCID: PMC10103693 DOI: 10.3389/fcimb.2023.1015625] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/27/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction Mucus in the female reproductive tract acts as a barrier that traps and eliminates pathogens and foreign particles via steric and adhesive interactions. During pregnancy, mucus protects the uterine environment from ascension of pathogens and bacteria from the vagina into the uterus, a potential contributor to intrauterine inflammation and preterm birth. As recent work has demonstrated the benefit of vaginal drug delivery in treating women's health indications, we sought to define the barrier properties of human cervicovaginal mucus (CVM) during pregnancy to inform the design of vaginally delivered therapeutics during pregnancy. Methods CVM samples were self-collected by pregnant participants over the course of pregnancy, and barrier properties were quantified using multiple particle tracking. 16S rRNA gene sequencing was performed to analyze the composition of the vaginal microbiome. Results Participant demographics differed between term delivery and preterm delivery cohorts, with Black or African American participants being significantly more likely to delivery prematurely. We observed that vaginal microbiota is most predictive of CVM barrier properties and of timing of parturition. Lactobacillus crispatus dominated CVM samples showed increased barrier properties compared to polymicrobial CVM samples. Discussion This work informs our understanding of how infections occur during pregnancy, and directs the engineering of targeted drug treatments for indications during pregnancy.
Collapse
Affiliation(s)
- Hannah C. Zierden
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Kevin DeLong
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Fareeha Zulfiqar
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jairo Ortiz Ortiz
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Victoria Laney
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Sabrine Bensouda
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nicole Hernández
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Thuy M. Hoang
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Samuel K. Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina/North Carolina State University (UNC/NCSU) Joint Department of Biomedical Engineering, Department of Microbiology & Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, United States
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Anne E. Burke
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laura M. Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
27
|
Mirzaei R, Kavyani B, Nabizadeh E, Kadkhoda H, Asghari Ozma M, Abdi M. Microbiota metabolites in the female reproductive system: Focused on the short-chain fatty acids. Heliyon 2023; 9:e14562. [PMID: 36967966 PMCID: PMC10031489 DOI: 10.1016/j.heliyon.2023.e14562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
Several disorders have been linked to modifications in the gut microbial imbalance, intestinal epithelium, and host immune system. In this regard, microbiota derived short-chain fatty acids (SCFAs) play a key function in the regulation of histone deacetylases (HDACs), which affect modulation of immunity and regulation of inflammatory responses in the intestine and other organs. Studies examining the metabolites produced by polymicrobial bacterial vaginosis (BV) states and Lactobacillus-dominated microbiota have noted a dramatic reduction of lactic acid and a shift toward SCFA synthesis. Along with higher levels of SCFAs, acetate is typically the main metabolite in the cervicovaginal fluid of women with symptomatic bacterial vaginosis. The fact that SCFAs made by the vaginal microbiota have been shown to exhibit antibacterial and immune-modulating properties suggests that they may have promise as indicators of disease and/or disease susceptibility. In this review, we overview and summarize the current findings on the detrimental or protective roles of microbiota metabolites especially SCFAs in the health and disease of the female reproductive system.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Batoul Kavyani
- Department of Medical Microbiology (Bacteriology & Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Edris Nabizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hiva Kadkhoda
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Asghari Ozma
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Abdi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
- Corresponding author.
| |
Collapse
|
28
|
Jimenez NR, Maarsingh JD, Łaniewski P, Herbst-Kralovetz MM. Commensal Lactobacilli Metabolically Contribute to Cervical Epithelial Homeostasis in a Species-Specific Manner. mSphere 2023; 8:e0045222. [PMID: 36629413 PMCID: PMC9942568 DOI: 10.1128/msphere.00452-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/09/2022] [Indexed: 01/12/2023] Open
Abstract
In reproductive-age women, the vaginal microbiome is typically dominated by one or a few Lactobacillus species, including Lactobacillus crispatus, Lactobacillus iners, Lactobacillus paragasseri, Lactobacillus mulieris, and Lactobaccillus crispatus, has been associated with optimal cervicovaginal health; however, much is still unknown about how other lactobacilli metabolically contribute to cervicovaginal health. We hypothesized that metabolites of each Lactobacillus species differ and uniquely contribute to health and homeostasis. To address this hypothesis, we utilized a human three-dimensional (3D) cervical epithelial cell model in conjunction with genomics analyses and untargeted metabolomics to determine the metabolic contributions of less-studied vaginal lactobacilli-L. iners, L. paragasseri, and L. mulieris. Our study validated that vaginal lactobacilli exhibit a close phylogenetic relationship. Genomic findings from publicly available strains and those used in our study indicated that L. iners is metabolically distinct from other species of lactobacilli, likely due to a reduced genome size. Lactobacilli and mock controls were distinguishable based on global metabolic profiles. We identified 95 significantly altered metabolites (P < 0.05) between individual lactobacilli and mock controls. Metabolites related to amino acid metabolism were shared among the lactobacilli. N-Acetylated amino acids with potential antimicrobial properties were significantly elevated in a species-specific manner. L. paragasseri and L. iners shared aromatic, but not carbohydrate-derived, lactic acid metabolites with potential antimicrobial properties that may contribute to homeostasis of the cervicovaginal environment. Additionally, L. iners uniquely altered lipid metabolism, which may be a sign of adaptation to the cervicovaginal niche. Overall, these findings further elucidate the metabolic contributions of three key vaginal Lactobacillus species in gynecological health. IMPORTANCE Lactobacillus species contribute to cervicovaginal health by their production of lactic acid and other antimicrobial compounds. Yet, much is still unknown regarding the metabolic potential of lesser-studied but common vaginal lactobacilli. Here, we used untargeted metabolomics coupled with our 3D cervical epithelial cell model to identify metabolic differences among vaginal Lactobacillus species (Lactobacillus iners, Lactobacillus paragasseri, and Lactobacillus mulieris) and how those differences related to maintaining homeostasis of the cervical epithelium. Human 3D cell models are essential tools for studying host-bacteria interactions and reducing confounding factors inherent in clinical studies. Therefore, these unique models allowed us to decipher the putative lactobacilli mechanisms that contribute to their roles in health or disease. Metabolic analyses revealed distinct profiles of each Lactobacillus species but also shared metabolic contributions associated with antimicrobial activity: amino acid metabolism, N-acetylated amino acids, and aromatic lactic acids. These patterns provided validation of metabolites associated with health in clinical studies and provided novel targets, including immunomodulatory and antimicrobial metabolites, for postbiotic therapies.
Collapse
Affiliation(s)
- Nicole R. Jimenez
- Department of Obstetrics and Gynecology, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Jason D. Maarsingh
- Department of Obstetrics and Gynecology, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Melissa M. Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Basic Medical Sciences, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| |
Collapse
|
29
|
Dong M, Dong Y, Bai J, Li H, Ma X, Li B, Wang C, Li H, Qi W, Wang Y, Fan A, Han C, Xue F. Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front Cell Infect Microbiol 2023; 13:1124591. [PMID: 36909729 PMCID: PMC9998931 DOI: 10.3389/fcimb.2023.1124591] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/31/2023] [Indexed: 02/26/2023] Open
Abstract
The female reproductive tract harbours hundreds of bacterial species and produces numerous metabolites. The uterine cervix is located between the upper and lower parts of the female genital tract. It allows sperm and birth passage and hinders the upward movement of microorganisms into a relatively sterile uterus. It is also the predicted site for sexually transmitted infection (STI), such as Chlamydia, human papilloma virus (HPV), and human immunodeficiency virus (HIV). The healthy cervicovaginal microbiota maintains cervical epithelial barrier integrity and modulates the mucosal immune system. Perturbations of the microbiota composition accompany changes in microbial metabolites that induce local inflammation, damage the cervical epithelial and immune barrier, and increase susceptibility to STI infection and relative disease progression. This review examined the intimate interactions between the cervicovaginal microbiota, relative metabolites, and the cervical epithelial-, immune-, and mucus barrier, and the potent effect of the host-microbiota interaction on specific STI infection. An improved understanding of cervicovaginal microbiota regulation on cervical microenvironment homeostasis might promote advances in diagnostic and therapeutic approaches for various STI diseases.
Collapse
Affiliation(s)
- Mengting Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yalan Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Junyi Bai
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotong Ma
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bijun Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huiyang Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenhui Qi
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Aiping Fan
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| | - Fengxia Xue
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| |
Collapse
|
30
|
Zhu B, Tao Z, Edupuganti L, Serrano MG, Buck GA. Roles of the Microbiota of the Female Reproductive Tract in Gynecological and Reproductive Health. Microbiol Mol Biol Rev 2022; 86:e0018121. [PMID: 36222685 PMCID: PMC9769908 DOI: 10.1128/mmbr.00181-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The microbiome of the female reproductive tract defies the convention that high biodiversity is a hallmark of an optimal ecosystem. Although not universally true, a homogeneous vaginal microbiome composed of species of Lactobacillus is generally associated with health, whereas vaginal microbiomes consisting of other taxa are generally associated with dysbiosis and a higher risk of disease. The past decade has seen a rapid advancement in our understanding of these unique biosystems. Of particular interest, substantial effort has been devoted to deciphering how members of the microbiome of the female reproductive tract impact pregnancy, with a focus on adverse outcomes, including but not limited to preterm birth. Herein, we review recent research efforts that are revealing the mechanisms by which these microorganisms of the female reproductive tract influence gynecologic and reproductive health of the female reproductive tract.
Collapse
Affiliation(s)
- Bin Zhu
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Zhi Tao
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Obstetrics and Gynecology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Laahirie Edupuganti
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Myrna G. Serrano
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Gregory A. Buck
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
- Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
31
|
Vidal MS, Lintao RCV, Severino MEL, Tantengco OAG, Menon R. Spontaneous preterm birth: Involvement of multiple feto-maternal tissues and organ systems, differing mechanisms, and pathways. Front Endocrinol (Lausanne) 2022; 13:1015622. [PMID: 36313741 PMCID: PMC9606232 DOI: 10.3389/fendo.2022.1015622] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Survivors of preterm birth struggle with multitudes of disabilities due to improper in utero programming of various tissues and organ systems contributing to adult-onset diseases at a very early stage of their lives. Therefore, the persistent rates of low birth weight (birth weight < 2,500 grams), as well as rates of neonatal and maternal morbidities and mortalities, need to be addressed. Active research throughout the years has provided us with multiple theories regarding the risk factors, initiators, biomarkers, and clinical manifestations of spontaneous preterm birth. Fetal organs, like the placenta and fetal membranes, and maternal tissues and organs, like the decidua, myometrium, and cervix, have all been shown to uniquely respond to specific exogenous or endogenous risk factors. These uniquely contribute to dynamic changes at the molecular and cellular levels to effect preterm labor pathways leading to delivery. Multiple intervention targets in these different tissues and organs have been successfully tested in preclinical trials to reduce the individual impacts on promoting preterm birth. However, these preclinical trial data have not been effectively translated into developing biomarkers of high-risk individuals for an early diagnosis of the disease. This becomes more evident when examining the current global rate of preterm birth, which remains staggeringly high despite years of research. We postulate that studying each tissue and organ in silos, as how the majority of research has been conducted in the past years, is unlikely to address the network interaction between various systems leading to a synchronized activity during either term or preterm labor and delivery. To address current limitations, this review proposes an integrated approach to studying various tissues and organs involved in the maintenance of normal pregnancy, promotion of normal parturition, and more importantly, contributions towards preterm birth. We also stress the need for biological models that allows for concomitant observation and analysis of interactions, rather than focusing on these tissues and organ in silos.
Collapse
Affiliation(s)
- Manuel S. Vidal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ryan C. V. Lintao
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Mary Elise L. Severino
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ourlad Alzeus G. Tantengco
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
32
|
Morales CG, Jimenez NR, Herbst-Kralovetz MM, Lee NR. Novel Vaccine Strategies and Factors to Consider in Addressing Health Disparities of HPV Infection and Cervical Cancer Development among Native American Women. Med Sci (Basel) 2022; 10:52. [PMID: 36135837 PMCID: PMC9503187 DOI: 10.3390/medsci10030052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is the 4th most common type of cancer in women world-wide. Many factors play a role in cervical cancer development/progression that include genetics, social behaviors, social determinants of health, and even the microbiome. The prevalence of HPV infections and cervical cancer is high and often understudied among Native American communities. While effective HPV vaccines exist, less than 60% of 13- to 17-year-olds in the general population are up to date on their HPV vaccination as of 2020. Vaccination rates are higher among Native American adolescents, approximately 85% for females and 60% for males in the same age group. Unfortunately, the burden of cervical cancer remains high in many Native American populations. In this paper, we will discuss HPV infection, vaccination and the cervicovaginal microbiome with a Native American perspective. We will also provide insight into new strategies for developing novel methods and therapeutics to prevent HPV infections and limit HPV persistence and progression to cervical cancer in all populations.
Collapse
Affiliation(s)
- Crystal G. Morales
- Department of Biology, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Nicole R. Jimenez
- Department of Obstetrics and Gynecology, College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Melissa M. Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
- Department of Basic Medical Sciences, College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Naomi R. Lee
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ 86011, USA
| |
Collapse
|
33
|
Delgado-Diaz DJ, Jesaveluk B, Hayward JA, Tyssen D, Alisoltani A, Potgieter M, Bell L, Ross E, Iranzadeh A, Allali I, Dabee S, Barnabas S, Gamieldien H, Blackburn JM, Mulder N, Smith SB, Edwards VL, Burgener AD, Bekker LG, Ravel J, Passmore JAS, Masson L, Hearps AC, Tachedjian G. Lactic acid from vaginal microbiota enhances cervicovaginal epithelial barrier integrity by promoting tight junction protein expression. MICROBIOME 2022; 10:141. [PMID: 36045402 PMCID: PMC9429363 DOI: 10.1186/s40168-022-01337-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/30/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Women with a cervicovaginal microbiota dominated by Lactobacillus spp. are at reduced risk of acquiring sexually transmitted infections including HIV, but the biological mechanisms involved remain poorly defined. Here, we performed metaproteomics on vaginal swab samples from young South African women (n = 113) and transcriptomics analysis of cervicovaginal epithelial cell cultures to examine the ability of lactic acid, a metabolite produced by cervicovaginal lactobacilli, to modulate genital epithelial barrier function. RESULTS Compared to women with Lactobacillus-depleted microbiota, women dominated by vaginal lactobacilli exhibit higher abundance of bacterial lactate dehydrogenase, a key enzyme responsible for lactic acid production, which is independently associated with an increased abundance of epithelial barrier proteins. Physiological concentrations of lactic acid enhance epithelial cell culture barrier integrity and increase intercellular junctional molecule expression. CONCLUSIONS These findings reveal a novel ability of vaginal lactic acid to enhance genital epithelial barrier integrity that may help prevent invasion by sexually transmitted pathogens. Video abstract.
Collapse
Affiliation(s)
- David Jose Delgado-Diaz
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia
| | - Brianna Jesaveluk
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia
| | - Joshua A Hayward
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia
| | - David Tyssen
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Arghavan Alisoltani
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA, 92521, USA
| | - Matthys Potgieter
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Liam Bell
- Centre for Proteomic and Genomic Research, Cape Town, 7925, South Africa
| | - Elizabeth Ross
- Centre for Proteomic and Genomic Research, Cape Town, 7925, South Africa
| | - Arash Iranzadeh
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Imane Allali
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, 1014, Rabat, Morocco
| | - Smritee Dabee
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Shaun Barnabas
- Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, 7505, South Africa
| | - Hoyam Gamieldien
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
| | - Jonathan M Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for Infectious Diseases Research (CIDRI) in Africa Wellcome Trust Centre, University of Cape Town, Cape Town, 7925, South Africa
| | - Steven B Smith
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vonetta L Edwards
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Adam D Burgener
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Obstetrics and Gynecology, University of Manitoba, Winnipeg, Canada
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, 7925, South Africa
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jo-Ann S Passmore
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, 4013, South Africa
- National Health Laboratory Service, Cape Town, 7925, South Africa
| | - Lindi Masson
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, 4013, South Africa
- Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Anna C Hearps
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia
- Central Clinical School, Monash University, Melbourne, 3004, Australia
| | - Gilda Tachedjian
- Life Sciences Discipline, Burnet Institute, 85 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Microbiology, Monash University, Clayton, VIC, 3168, Australia.
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
34
|
Anton L, Ferguson B, Friedman ES, Gerson KD, Brown AG, Elovitz MA. Gardnerella vaginalis alters cervicovaginal epithelial cell function through microbe-specific immune responses. MICROBIOME 2022; 10:119. [PMID: 35922830 PMCID: PMC9351251 DOI: 10.1186/s40168-022-01317-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/26/2022] [Indexed: 05/26/2023]
Abstract
BACKGROUND The cervicovaginal (CV) microbiome is highly associated with vaginal health and disease in both pregnant and nonpregnant individuals. An overabundance of Gardnerella vaginalis (G. vaginalis) in the CV space is commonly associated with adverse reproductive outcomes including bacterial vaginosis (BV), sexually transmitted diseases, and preterm birth, while the presence of Lactobacillus spp. is often associated with reproductive health. While host-microbial interactions are hypothesized to contribute to CV health and disease, the mechanisms by which these interactions regulate CV epithelial function remain largely unknown. RESULTS Using an in vitro co-culture model, we assessed the effects of Lactobacillus crispatus (L. crispatus) and G. vaginalis on the CV epithelial barrier, the immune mediators that could be contributing to decreased barrier integrity and the immune signaling pathways regulating the immune response. G. vaginalis, but not L. crispatus, significantly increased epithelial cell death and decreased epithelial barrier integrity in an epithelial cell-specific manner. A G. vaginalis-mediated epithelial immune response including NF-κB activation and proinflammatory cytokine release was initiated partially through TLR2-dependent signaling pathways. Additionally, investigation of the cytokine immune profile in human CV fluid showed distinctive clustering of cytokines by Gardnerella spp. abundance and birth outcome. CONCLUSIONS The results of this study show microbe-specific effects on CV epithelial function. Altered epithelial barrier function through cell death and immune-mediated mechanisms by G. vaginalis, but not L. crispatus, indicates that host epithelial cells respond to bacteria-associated signals, resulting in altered epithelial function and ultimately CV disease. Additionally, distinct immune signatures associated with Gardnerella spp. or birth outcome provide further evidence that host-microbial interactions may contribute significantly to the biological mechanisms regulating reproductive outcomes. Video Abstract.
Collapse
Affiliation(s)
- Lauren Anton
- Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Briana Ferguson
- Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Elliot S Friedman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kristin D Gerson
- Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Amy G Brown
- Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michal A Elovitz
- Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
35
|
Gandhi K, Manales NJ, Garza J, David S, Sanchez A, Ventolini G. Lactobacilli and Cytokine Modifications during Menopause and Their Relation to Vulvar and Vulvovaginal Disorders. J Menopausal Med 2022; 28:52-59. [PMID: 36070870 PMCID: PMC9452597 DOI: 10.6118/jmm.22001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 07/13/2022] [Accepted: 08/08/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES Female sexual and reproductive health is heavily influenced by the levels and ratios of Lactobacilli species and vaginal cytokines. Menopause marks a profound body change as it shifts to a natural and permanent non-reproductive state. Vulvovaginal diseases encompass a broad variety of sexual health conditions. Furthermore, both menopause and vulvovaginal diseases affect vaginal Lactobacilli and cytokine levels. Thus, this study aimed to investigate the correlation between menopause, vulvovaginal diseases, and vaginal Lactobacilli and cytokine levels. METHODS Vaginal swab samples were collected as part of a prospective data bank creation to study vaginal conditions as approved by the Institutional Review Board of Texas Tech University Health Sciences Center, Lubbock, USA. This study utilized 38 samples in this database, which were assigned to the pre-menopausal with no vulvovaginal conditions (n = 20) and post-menopausal with vulvovaginal conditions (n = 18) groups. A real-time polymerase chain reaction was conducted to determine the relative concentration of Lactobacilli species, while cytokine analysis was performed using multiplex enzyme-linked immunosorbent assay immunoassay. The standardized mean difference, multivariate analysis of variance, and permutational unequal variance t test were used for the statistical analysis. RESULTS Cytokines, interleukin (IL)-6, macrophage inflammatory protein-1α, IL-8, and Lactobacillus iners expression were significantly elevated in the control group compared to the study group (P = 0.03 for the cytokines, P = 0.0194 for Lactobacilli). CONCLUSIONS The levels of vaginal cytokine and Lactobacillus profile were significantly different between the pre-menopausal and post-menopausal groups.
Collapse
Affiliation(s)
- Kushal Gandhi
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin, Odessa, TX, USA
| | - Nathan Joshua Manales
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin, Odessa, TX, USA
| | - John Garza
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin, Odessa, TX, USA.,Department of Mathematics, The University of Texas Permian Basin, Odessa, TX, USA
| | - Samuel David
- Department of Chemistry, Southern Oregon University, Ashland, OR, USA
| | - Asley Sanchez
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin, Odessa, TX, USA
| | - Gary Ventolini
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center at the Permian Basin, Odessa, TX, USA.
| |
Collapse
|
36
|
GERSON KD, YANG N, ANTON L, LEVY M, RAVEL J, ELOVITZ MA, BURRIS HH. Second trimester short cervix is associated with decreased abundance of cervicovaginal lipid metabolites. Am J Obstet Gynecol 2022; 227:273.e1-273.e18. [PMID: 35469813 PMCID: PMC9382664 DOI: 10.1016/j.ajog.2022.04.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND A short cervix is a risk factor for preterm birth. The molecular drivers of a short cervix remain elusive. Metabolites may function as mediators of pathologic processes. OBJECTIVE We sought to determine if a distinct cervicovaginal metabolomic profile is associated with a short cervix (<25 mm) to unveil the potential mechanisms by which premature cervical remodeling leads to a short cervix. STUDY DESIGN This was a secondary analysis of a completed prospective pregnancy cohort. Cervicovaginal fluid was obtained between 20 and 24 weeks' gestation. The participants selected for metabolomic profiling were frequency-matched by birth outcome and cervicovaginal microbiota profile. This analysis included 222 participants with cervical length measured. A short cervix was defined as one having length <25 mm, as measured by transvaginal ultrasound. Unpaired t-tests were performed with a Bonferroni correction for multiple comparisons. RESULTS There were 27 participants with a short cervix, and 195 with normal cervical length. Of the 637 metabolites detected, 26 differed between those with a short cervix and those with normal cervical lengths; 22 were decreased, of which 21 belonged to the lipid metabolism pathway (all P<.000079). Diethanolamine, erythritol, progesterone, and mannitol or sorbitol were increased in the cases of short cervix. Among participants with Lactobacillus-deficient microbiota, only diethanolamine and mannitol or sorbitol differed between short cervix (n=17) and normal cervical length (n=75), both increased. CONCLUSION A short cervix is associated with decreased cervicovaginal lipid metabolites, particularly sphingolipids. This class of lipids stabilizes cell membranes and protects against environmental exposures. Increased diethanolamine-an immunostimulatory xenobiotic-is associated with a short cervix. These observations begin to identify the potential mechanisms by which modifiable environmental factors may invoke cell damage in the setting of biological vulnerability, thus promoting premature cervical remodeling in spontaneous preterm birth.
Collapse
Affiliation(s)
- Kristin D. GERSON
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA,Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nancy YANG
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lauren ANTON
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Maayan LEVY
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacques RAVEL
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Michal A. ELOVITZ
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA,Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Heather H. BURRIS
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA,Children’s Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
37
|
Age-Stratified Analysis of Vaginal Microbiota Dysbiosis and the Relationship with HPV Viral Load in HPV-Positive Women. J Immunol Res 2022; 2022:1372926. [PMID: 35935589 PMCID: PMC9348945 DOI: 10.1155/2022/1372926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/04/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022] Open
Abstract
Objective This study evaluated the distribution of vaginal microbiota dysbiosis and the association with HPV viral load test in high-risk HPV-positive women before and after 50 years old. Methods For this cross-sectional study, 388 HPV-positive women prior to referral to colposcopy in Peking University Peoples' Hospital were included and classified as younger than 50 years (n = 307) and aged 50 years or older (n = 81), midvagina bacterial community composition was characterized by FlashDetect™ MAX vaginal microbe detection kit, and BMRT-HPV reported type-specific viral loads/10,000 cells. Results The community state type (CST) IV was the most common CST occurring in 148 women (38.1%). The proportion of CST IV in those aged 50 years or older was significantly higher than those younger than 50 years (women) (66.7% vs. 30.6%); the difference was statistically significant (<0.001). CST distribution has no statistical difference in different grades of cervical lesion, regardless of the age (p = 0.238 and 0.263). However, the women with high-grade cervical lesion presented a more complicated trend and the abundance of vaginal microbiota dysbiosis than low-grade lesion. HPV16/18 viral load was found to be significantly higher in CST III and CST IV than CST I/II/V (p < 0.05)in women younger than 50 years. Conclusions In women younger than 50 years, higher HPV16/18 load was more closely associated with CST IV; however, it had no significant correlation in women aged 50 years or older.
Collapse
|
38
|
Hayashida S, Takada K, Melnikov VG, Komine-Aizawa S, Tsuji NM, Hayakawa S. How were Lactobacillus species selected as single dominant species in the human vaginal microbiota? Coevolution of humans and Lactobacillus. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110858] [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
|
Li C, Zhang Z, Yang Y, Liao H. Changes in the cervicovaginal microbiota composition of HPV16-infected patients after clinical treatment. Cancer Med 2022; 11:5037-5049. [PMID: 35569127 PMCID: PMC9761074 DOI: 10.1002/cam4.4801] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND High-risk human papillomavirus (hrHPV) infection is a key factor that alters cervicovaginal microbiota patterns and causes cervical intraepithelial neoplasias (CINs) or even cervical cancer. Although local excisional treatment can clear hrHPV infection and restore the cervicovaginal microbiota, it is unclear which cervicovaginal microbiota represents recovery. Our objective was to describe the cervicovaginal microbiota before and after treatments and to assess the association between the microbiota and HPV persistence. RESULTS A cohort of 91 participants was classified into four groups (healthy control women and HPV16-infected women with CIN I, CIN II/III, and squamous cell carcinoma [SCC]). Endocervical swabs were collected 3 months prior to treatment and at 3 months post-treatment for bacterial 16S rRNA gene pyrosequencing and for HPV DNA testing. There was an increase in the number of Lactobacillus bacterial species present after the clinical treatments, and the community state type (CST) profiles were shifted from dysbiotic CSTs II and IV to Lactobacillus-dominated CSTs I and III. Specifically, the composition of Geobacter and Prevotella before treatment and Lactobacillus secaliphilus after treatment might have been related to CIN I, the composition of Burkholderia before treatment and Lactobacillus iners after treatment might have been related to CIN II/III, and the composition of Atopobium and Aerococcus before treatment and Bacilli after treatment might have been related to SCC. Further functional predictions revealed that the composition differences were linked to infectious disease- and cancer-related genes. CONCLUSION Our study provides an illustration of the changes in CSTs and the cervicovaginal microbiota before and after HPV16 clearance in each disease state.
Collapse
Affiliation(s)
- Chao Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Zhenbo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Yixia Yang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Hong Liao
- Department of Lab Medicine, Shanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
40
|
Gardnerella vaginalis induces matrix metalloproteinases in the cervicovaginal epithelium through TLR-2 activation. J Reprod Immunol 2022; 152:103648. [PMID: 35679790 PMCID: PMC9313515 DOI: 10.1016/j.jri.2022.103648] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022]
Abstract
Lactobacillus-deficient cervicovaginal microbiota, including Gardnerella vaginalis, are implicated in cervical remodeling and preterm birth. Mechanisms by which microbes drives outcomes are not fully elucidated. We hypothesize that Gardnerella vaginalis induces matrix metalloproteinases through TLR-2, leading to epithelial barrier dysfunction and premature cervical remodeling. Cervicovaginal cells were treated with live Gardnerella vaginalis or Lactobacillus crispatus or their bacteria-free supernatants for 24 h. For TLR-2 experiments, cells were pretreated with TLR-2 blocking antibody. A Luminex panel was run on cell media. For human data, we conducted a case-control study from a prospective pregnancy cohort of Black individuals with spontaneous preterm (sPTB) (n = 40) or term (n = 40) births whose vaginal microbiota had already been characterized. Cervicovaginal fluid was obtained between 20 and 24 weeks' gestation. Short cervix was defined as < 25 mm by second trimester transvaginal ultrasound. MMP-9 was quantified by ELISA. Standard analytical approaches were used to determine differences across in vitro conditions, as well as MMP-9 and associations with clinical outcomes. Gardnerella vaginalis induced MMP-1 in cervical cells (p = 0.01) and MMP-9 in cervical and vaginal (VK2) cells (p ≤ 0.001 for all). TLR-2 blockade mitigated MMP-9 induction by Gardnerella vaginalis. MMP-9 in cervicovaginal fluid is higher among pregnant individuals with preterm birth, short cervix, and Lactobacillus-deficient microbiota (p < 0.05 for all). MMP-9 is increased in the cervicovaginal fluid of pregnant individuals with subsequent sPTB. Our in vitro work ascribes a potential mechanism by which a cervicovaginal microbe, commonly associated with adverse pregnancy outcomes, may disrupt the cervicovaginal epithelial barrier and promote premature cervical remodeling in spontaneous preterm birth.
Collapse
|
41
|
Jie Z, Chen C, Hao L, Li F, Song L, Zhang X, Zhu J, Tian L, Tong X, Cai K, Zhang Z, Ju Y, Yu X, Li Y, Zhou H, Lu H, Qiu X, Li Q, Liao Y, Zhou D, Lian H, Zuo Y, Chen X, Rao W, Ren Y, Wang Y, Zi J, Wang R, Liu N, Wu J, Zhang W, Liu X, Zong Y, Liu W, Xiao L, Hou Y, Xu X, Yang H, Wang J, Kristiansen K, Jia H. Life History Recorded in the Vagino-cervical Microbiome Along with Multi-omes. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:304-321. [PMID: 34118463 PMCID: PMC9684086 DOI: 10.1016/j.gpb.2021.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/31/2021] [Indexed: 01/05/2023]
Abstract
The vagina contains at least a billion microbial cells, dominated by lactobacilli. Here we perform metagenomic shotgun sequencing on cervical and fecal samples from a cohort of 516 Chinese women of reproductive age, as well as cervical, fecal, and salivary samples from a second cohort of 632 women. Factors such as pregnancyhistory, delivery history, cesarean section, and breastfeeding were all more important than menstrual cycle in shaping the microbiome, and such information would be necessary before trying to interpret differences between vagino-cervical microbiome data. Greater proportion of Bifidobacterium breve was seen with older age at sexual debut. The relative abundance of lactobacilli especially Lactobacillus crispatus was negatively associated with pregnancy history. Potential markers for lack of menstrual regularity, heavy flow, dysmenorrhea, and contraceptives were also identified. Lactobacilli were rare during breastfeeding or post-menopause. Other features such as mood fluctuations and facial speckles could potentially be predicted from the vagino-cervical microbiome. Gut and salivary microbiomes, plasma vitamins, metals, amino acids, and hormones showed associations with the vagino-cervical microbiome. Our results offer an unprecedented glimpse into the microbiota of the female reproductive tract and call for international collaborations to better understand its long-term health impact other than in the settings of infection or pre-term birth.
Collapse
Affiliation(s)
- Zhuye Jie
- BGI-Shenzhen, Shenzhen 518083, China,Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China,Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Chen Chen
- BGI-Shenzhen, Shenzhen 518083, China,Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China,Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark,Corresponding authors.
| | - Lilan Hao
- BGI-Shenzhen, Shenzhen 518083, China
| | - Fei Li
- BGI-Shenzhen, Shenzhen 518083, China
| | - Liju Song
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Jie Zhu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Liu Tian
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xin Tong
- BGI-Shenzhen, Shenzhen 518083, China
| | - Kaiye Cai
- BGI-Shenzhen, Shenzhen 518083, China,Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen 518083, China
| | - Zhe Zhang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yanmei Ju
- BGI-Shenzhen, Shenzhen 518083, China,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China
| | - Xinlei Yu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Ying Li
- BGI-Shenzhen, Shenzhen 518083, China
| | - Hongcheng Zhou
- China National Genebank, BGI-Shenzhen, Shenzhen 518120, China
| | - Haorong Lu
- China National Genebank, BGI-Shenzhen, Shenzhen 518120, China
| | | | - Qiang Li
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | - Heng Lian
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yong Zuo
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | - Yan Ren
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yuan Wang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Jin Zi
- BGI-Shenzhen, Shenzhen 518083, China
| | - Rong Wang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Na Liu
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Wei Zhang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xiao Liu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yang Zong
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Liang Xiao
- BGI-Shenzhen, Shenzhen 518083, China,Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen 518083, China,BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
| | - Yong Hou
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518083, China,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518083, China,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen 518083, China,Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark,BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen 518083, China,Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China,Corresponding authors.
| |
Collapse
|
42
|
Armstrong E, Hemmerling A, Miller S, Burke KE, Newmann SJ, Morris SR, Reno H, Huibner S, Kulikova M, Nagelkerke N, Coburn B, Cohen CR, Kaul R. Sustained effect of LACTIN-V (Lactobacillus crispatus CTV-05) on genital immunology following standard bacterial vaginosis treatment: results from a randomised, placebo-controlled trial. THE LANCET MICROBE 2022; 3:e435-e442. [PMID: 35659905 PMCID: PMC9188188 DOI: 10.1016/s2666-5247(22)00043-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022] Open
Abstract
Background Bacterial vaginosis might increase HIV risk by eliciting genital inflammation and epithelial barrier disruption, whereas vaginal Lactobacillus crispatus is associated with immune quiescence and HIV protection. We investigated the effect of a live biotherapeutic containing L crispatus CTV-05 (LACTIN-V) on genital immunology and key vaginal bacteria. Methods This substudy included women aged 18–45 years who participated in the randomised, placebo-controlled, phase 2b trial of LACTIN-V to reduce bacterial vaginosis recurrence, conducted at four universities and hospitals in the USA. Women with negative results for sexually transmitted infection, pregnancy, and urinary tract infection were provided a 5-day course of vaginal metronidazole 0·75% gel. Those who met at least three of four clinical Amsel criteria for bacterial vaginosis and had a Nugent score of 4–10 from Gram staining were eligible. Participants in the LACTIN-V trial were randomly assigned (2:1) to receive either LACTIN-V or placebo, applied vaginally once per day for 5 days during the first week and then twice per week for 10 more weeks. Follow-up visits occurred 4, 8, 12, and 24 weeks after enrolment. Soluble immune factors and the absolute abundance of bacterial taxa were assayed by mutliplex ELISA and quantitative PCR. The primary outcomes were vaginal levels of IL-1α and soluble E-cadherin at 24 weeks (ie, 13 weeks after treatment cessation). Findings Between Feb 21, 2020 and March 18, 2021, we characterised genital immune parameters and the vaginal microbiota in a subset of 66 highly adherent participants who were randomly selected, with no exclusion criteria, from those who had attended all study follow-up visits (n=166) in the larger LACTIN-V clinical trial (n=288). 32 (48%) participants received LACTIN-V and 34 (52%) received placebo. LACTIN-V treatment was significantly associated with lower concentrations of the proinflammatory cytokine IL-1α (β coefficient 0·310, SE 0·149; p=0·042) and soluble E-cadherin (0·429, 0·199; p=0·035), a biomarker of epithelial barrier disruption. Interpretation Vaginal administration of LACTIN-V following standard bacterial vaginosis therapy resulted in a sustained reduction in genital inflammation and a biomarker of epithelial integrity. The potential of LACTIN-V to reduce HIV susceptibility merits further investigation. Funding Canadian Institutes of Health Research and the National Institutes of Health National Institute of Allergy and Infectious Diseases.
Collapse
|
43
|
Holubekova V, Kolkova Z, Kasubova I, Samec M, Mazurakova A, Koklesova L, Kubatka P, Rokos T, Kozubik E, Biringer K, Kudela E. Interaction of cervical microbiome with epigenome of epithelial cells: Significance of inflammation to primary healthcare. Biomol Concepts 2022; 13:61-80. [PMID: 35245973 DOI: 10.1515/bmc-2022-0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
One pillar of the predictive, preventive, and personalized medicine framework strategies is the female health. The evaluation of women's lifestyle and dietary habits in context with genetic and modifiable risk factors may reflect the prevention of cervical cancer before the occurrence of clinical symptoms and prediction of cervical lesion behavior. The main aim of this review is to analyze publications in the field of precision medicine that allow the use of research knowledge of cervical microbiome, epigenetic modifications, and inflammation in potential application in clinical practice. Personalized approach in evaluating patient's risk of future development of cervical abnormality should consider the biomarkers of the local microenvironment characterized by the microbial composition, epigenetic pattern of cervical epithelium, and presence of chronic inflammation. Novel sequencing techniques enable a more detailed characterization of actual state in cervical epithelium. Better understanding of all changes in multiomics level enables a better assessment of disease prognosis and selects the eligible targeted therapy in personalized medicine. Restoring of healthy vaginal microflora and reversing the outbreak of cervical abnormality can be also achieved by dietary habits as well as uptake of prebiotics, probiotics, synbiotics, microbial transplantation, and others.
Collapse
Affiliation(s)
- Veronika Holubekova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Zuzana Kolkova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Ivana Kasubova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Marek Samec
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Alena Mazurakova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Tomas Rokos
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Erik Kozubik
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Erik Kudela
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| |
Collapse
|
44
|
Rosen EM, Martin CL, Siega-Riz AM, Dole N, Basta PV, Serrano M, Fettweis J, Wu M, Sun S, Thorp JM, Buck G, Fodor AA, Engel SM. Is prenatal diet associated with the composition of the vaginal microbiome? Paediatr Perinat Epidemiol 2022; 36:243-253. [PMID: 34841560 PMCID: PMC8881389 DOI: 10.1111/ppe.12830] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND The vaginal microbiome has been associated with adverse pregnancy outcomes, but information on the impact of diet on microbiome composition is largely unexamined. OBJECTIVE To estimate the association between prenatal diet and vaginal microbiota composition overall and by race. METHODS We leveraged a racially diverse prenatal cohort of North Carolina women enrolled between 1995 and 2001 to conduct this analysis using cross-sectional data. Women completed food frequency questionnaires about diet in the previous 3 months and foods were categorised into subgroups: fruits, vegetables, nuts/seeds, whole grains, low-fat dairy, sweetened beverages and red meat. We additionally assessed dietary vitamin D, fibre and yogurt consumption. Stored vaginal swabs collected in mid-pregnancy were sequenced using 16S taxonomic profiling. Women were categorised into three groups based on predominance of species: Lactobacillus iners, Lactobacillus miscellaneous and Bacterial Vaginosis (BV)-associated bacteria. Adjusted Poisson models with robust variance estimators were run to assess the risk of being in a specific vagitype compared to the referent. Race-stratified models (Black/White) were also run. RESULTS In this study of 634 women, higher consumption of dairy was associated with increased likelihood of membership in the L. crispatus group compared to the L. iners group in a dose-dependent manner (risk ratio quartile 4 vs. 1: 2.01, 95% confidence interval 1.36, 2.95). Increased intake of fruit, vitamin D, fibre and yogurt was also associated with increased likelihood of membership in L. crispatus compared to L. iners, but only among black women. Statistical heterogeneity was only detected for fibre intake. There were no detected associations between any other food groups or risk of membership in the BV group. CONCLUSIONS Higher consumption of low-fat dairy was associated with increased likelihood of membership in a beneficial vagitype, potentially driven by probiotics.
Collapse
Affiliation(s)
- Emma M. Rosen
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Chantel L. Martin
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Anna Maria Siega-Riz
- Departments of Nutrition and Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst MA 01003
| | - Nancy Dole
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516
| | - Patricia V. Basta
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Myrna Serrano
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Jennifer Fettweis
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Michael Wu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Shan Sun
- Department of Bioinformatics, University of North Carolina at Charlotte, Charlotte, NC
| | - John M. Thorp
- Department of Obstetrics and Gynecology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Gregory Buck
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284
| | - Anthony A. Fodor
- Department of Bioinformatics, University of North Carolina at Charlotte, Charlotte, NC
| | - Stephanie M. Engel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
45
|
Armstrong E, Hemmerling A, Miller S, Burke KE, Newmann SJ, Morris SR, Reno H, Huibner S, Kulikova M, Liu R, Crawford ED, Castañeda GR, Nagelkerke N, Coburn B, Cohen CR, Kaul R. Metronidazole treatment rapidly reduces genital inflammation through effects on bacterial vaginosis-associated bacteria rather than lactobacilli. J Clin Invest 2022; 132:152930. [PMID: 35113809 PMCID: PMC8920324 DOI: 10.1172/jci152930] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 02/02/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Bacterial vaginosis (BV) causes genital inflammation and increases HIV risk, while a vaginal microbiota dominated by Lactobacillus species is associated with immune quiescence and relative HIV protection. BV treatment reduces genital inflammation, but it is unclear whether this is driven by a decrease in BV-associated bacteria or an increase in Lactobacillus. METHODS To evaluate the short-term impact of standard BV treatment on genital immunology and the vaginal microbiota, vaginal swabs were collected immediately before and after metronidazole treatment for BV and analyzed with multiplex ELISA, metagenomic sequencing, and quantitative polymerase chain reaction. RESULTS Topical metronidazole treatment rapidly reduced vaginal levels of proinflammatory cytokines, chemokines, and soluble immune markers of epithelial barrier disruption. Although the vaginal microbiota shifted to dominance by L. iners or L. jensenii, this proportional shift was primarily driven by a 2-4 log10 fold reduction in BV-associated bacteria absolute abundance; BV treatment induced no change in the absolute abundance of L. crispatus or L. iners, and only minor (<1 log10 fold) increases in L. gasseri and L. jensenii that were not independently associated with reduced inflammation in multivariable models. CONCLUSION The genital immune benefits that are associated with Lactobacillus dominance following BV treatment were not directly attributable to an absolute increase in lactobacilli, but rather to the loss of BV-associated bacteria.TRAIL REGISTRATION. Participants were recruited as part of a randomized controlled trial (NCT02766023) from 2016 to 2020. FUNDING Canadian Institutes of Health Research (PJT-156123) and the National Institute of Allergy and Infectious Diseases (HHSN2722013000141 and HHSN27200007).
Collapse
Affiliation(s)
| | - Anke Hemmerling
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, United States of America
| | - Steve Miller
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, United States of America
| | - Kerianne E Burke
- Ruth M. Rothstein CORE Centre, Stroger Hospital of Cook County Health, Chicago, United States of America
| | - Sara J Newmann
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, United States of America
| | - Sheldon R Morris
- Department of Family Medicine and Public Health, University of California, San Diego, San Diego, United States of America
| | - Hilary Reno
- Department of Medicine, Washington University, St. Louis, United States of America
| | - Sanja Huibner
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Maria Kulikova
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Rachel Liu
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Emily D Crawford
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States of America
| | - Gloria R Castañeda
- Infectious Diseases, Chan Zuckerberg Biohub, San Francisco, United States of America
| | - Nico Nagelkerke
- Centre for Global Health Research, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Bryan Coburn
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Craig R Cohen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, United States of America
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
46
|
Tantengco OAG, Menon R. Breaking Down the Barrier: The Role of Cervical Infection and Inflammation in Preterm Birth. Front Glob Womens Health 2022; 2:777643. [PMID: 35118439 PMCID: PMC8803751 DOI: 10.3389/fgwh.2021.777643] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/27/2021] [Indexed: 01/06/2023] Open
Abstract
Approximately 40% of cases of spontaneous preterm birth (sPTB) are associated with ascending intrauterine infections. The cervix serves as a physical and immunological gatekeeper, preventing the ascent of microorganisms from the vagina to the amniotic cavity. The cervix undergoes remodeling during pregnancy. It remains firm and closed from the start until the late third trimester of pregnancy and then dilates and effaces to accommodate the passage of the fetus during delivery. Remodeling proceeds appropriately and timely to maintain the pregnancy until term delivery. However, risk factors, such as acute and chronic infection and local inflammation in the cervix, may compromise cervical integrity and result in premature remodeling, predisposing to sPTB. Previous clinical studies have established bacterial (i.e., chlamydia, gonorrhea, mycoplasma, etc.) and viral infections (i.e., herpesviruses and human papillomaviruses) as risk factors of PTB. However, the exact mechanism leading to PTB is still unknown. This review focuses on: (1) the epidemiology of cervical infections in pregnant patients; (2) cellular mechanisms that may explain the association of cervical infections to premature cervical ripening and PTB; (3) endogenous defense mechanisms of the cervix that protect the uterine cavity from infection and inflammation; and (4) potential inflammatory biomarkers associated with cervical infection that can serve as prognostic markers for premature cervical ripening and PTB. This review will provide mechanistic insights on cervical functions to assist in managing cervical infections during pregnancy.
Collapse
Affiliation(s)
- Ourlad Alzeus G. Tantengco
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- *Correspondence: Ramkumar Menon
| |
Collapse
|
47
|
Galiwango RM, Park DE, Huibner S, Onos A, Aziz M, Roach K, Anok A, Nnamutete J, Isabirye Y, Wasswa JB, Male D, Kigozi G, Tobian AAR, Prodger JL, Liu CM, Kaul R. Immune milieu and microbiome of the distal urethra in Ugandan men: impact of penile circumcision and implications for HIV susceptibility. MICROBIOME 2022; 10:7. [PMID: 35042542 PMCID: PMC8764836 DOI: 10.1186/s40168-021-01185-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 11/01/2021] [Indexed: 05/24/2023]
Abstract
BACKGROUND Coronal sulcus (CS) anaerobe abundance and IL-8 levels are linked to HIV acquisition, and are dramatically reduced after penile circumcision (PC). The distal urethra may be the site of some HIV acquisition before PC, and presumably most acquisition post PC. We describe the immune milieu and microbiome of the distal urethra in uncircumcised Ugandan men, and define the impact of PC. Participants consisted of HIV-negative, genital symptom-free adult Ugandan men undergoing PC (n = 51). Urethral and coronal sulcus swabs were collected at baseline and at 6- and 12-months post-PC. Soluble immune factors were quantified by multiplex ELISA, and bacterial abundance assessed by 16S rRNA qPCR and sequencing. RESULTS At baseline, the urethra was enriched compared to the CS for most cytokines (including IL-8 and MIP-1β) and soluble E-cadherin (sE-cadherin, an epithelial disruption marker), although CS levels of IL-1α and IL-1β were higher. Baseline total bacterial abundance was ≥ 20-fold higher in the CS than the urethra (median 27,100 vs. 1200 gene copies/swab, p = 0.001), and anaerobes comprised 58% of CS bacteria vs. 42% of urethral bacteria. PC did not alter urethral IL-8 (median 806 at baseline vs. 1130 pg/ml at 12 months; p = 0.062) and urethral sE-cadherin increased (113,223 vs. 158,385 pg/ml, p = 0.009), despite five- and sevenfold drops in total bacterial and anaerobe abundance after PC, respectively. However, PC dramatically reduced CS levels of sE-cadherin (15,843 vs. 837 pg/ml, p < 0.001) and most cytokines (IL-8; 34 vs. 3 pg/ml, p < 0.001), while reducing total bacterial and anaerobe abundance by 13-fold and 60-fold, respectively (both P ≤ 0.004). CONCLUSIONS The urethra is immunologically rich with characteristics of an HIV-susceptible tissue site. However, PC had no impact on urethral immunology and may have reduced epithelial integrity, despite modest reductions in total bacteria and anaerobes, suggesting that HIV protection from PC is not mediated via immune or microbiome alterations in the urethra. Video abstract.
Collapse
Affiliation(s)
- Ronald M Galiwango
- Departments of Immunology and Medicine, University of Toronto, St. George Campus, Medical Sciences Building, 1 King's College Circle, Room 6356, Toronto, Ontario, M5S1A8, Canada
| | - Daniel E Park
- George Washington Milken Institute School of Public Health, Washington, DC, USA
| | - Sanja Huibner
- Departments of Immunology and Medicine, University of Toronto, St. George Campus, Medical Sciences Building, 1 King's College Circle, Room 6356, Toronto, Ontario, M5S1A8, Canada
| | - Abigail Onos
- George Washington Milken Institute School of Public Health, Washington, DC, USA
| | - Maliha Aziz
- Departments of Immunology and Medicine, University of Toronto, St. George Campus, Medical Sciences Building, 1 King's College Circle, Room 6356, Toronto, Ontario, M5S1A8, Canada
| | - Kelsey Roach
- George Washington Milken Institute School of Public Health, Washington, DC, USA
| | - Aggrey Anok
- Rakai Health Sciences Program, Kalisizo, Uganda
| | | | | | | | - Deo Male
- Rakai Health Sciences Program, Kalisizo, Uganda
| | | | | | | | - Cindy M Liu
- George Washington Milken Institute School of Public Health, Washington, DC, USA
| | - Rupert Kaul
- Departments of Immunology and Medicine, University of Toronto, St. George Campus, Medical Sciences Building, 1 King's College Circle, Room 6356, Toronto, Ontario, M5S1A8, Canada.
| |
Collapse
|
48
|
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: 23] [Impact Index Per Article: 11.5] [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
|
49
|
LAWLOR ML, GOLDKAMP JM, BOERRIGTER A, JAKES C, PYON R, VRICELLA LK, GROSS GA, AURORA R. Cervicovaginal microbiome in twin versus singleton gestations. Am J Obstet Gynecol MFM 2022; 4:100579. [DOI: 10.1016/j.ajogmf.2022.100579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/11/2022] [Accepted: 01/27/2022] [Indexed: 11/16/2022]
|
50
|
Pawar K, Aranha C. Lactobacilli metabolites restore E-cadherin and suppress MMP9 in cervical cancer cells. Curr Res Toxicol 2022; 3:100088. [PMID: 36176311 PMCID: PMC9513734 DOI: 10.1016/j.crtox.2022.100088] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 08/18/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
| | - Clara Aranha
- Corresponding author at: Department of Molecular Immunology and Microbiology, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai 400102, India
| |
Collapse
|