1
|
Arab Bafrani M, Rios V, Kim MJ, Balan A, Bove R. Gynecological health: A missing link in comprehensive treatment monitoring for multiple sclerosis. Mult Scler 2025:13524585251346371. [PMID: 40528461 DOI: 10.1177/13524585251346371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2025]
Abstract
Safety monitoring of disease-modifying therapies (DMTs) used to treat multiple sclerosis (MS) has largely overlooked the domain of gynecological health. This topical review aims to provide MS clinicians with an overview of the three categories of complications described to date, as well as risk mitigation strategies. These are increased risk of human papilloma virus (HPV) positivity and related cervical dysplasia/cancers; inflammatory and infectious vaginitis and susceptibility to bacterial vaginosis (BV); and herpesvirus infections, including genital Herpes Simplex Virus (HSV). Current knowledge may be biased due to limited studies and lack of gynecological focus in neurological encounters. Risk mitigation strategies include promoting HPV vaccination, following guidance for immune compromised individuals relating to cervical cancer screening and antiviral suppression, and proactive communication with patients about gynecological health when starting DMTs. Together, these might improve gynecological health and thereby quality of life in females with neuroinflammatory diseases.
Collapse
Affiliation(s)
- Melika Arab Bafrani
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Viviana Rios
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Min Ji Kim
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Ayushi Balan
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Riley Bove
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
Jung W, Son YM. Nanoparticle-Driven Modulation of Mucosal Immunity and Interplay with the Microbiome. J Microbiol Biotechnol 2025; 35:e2404033. [PMID: 40537897 PMCID: PMC12197821 DOI: 10.4014/jmb.2504.04033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 06/29/2025]
Abstract
Mucosal surfaces are dynamic immunological interfaces that play a critical role in maintaining host defense and microbial homeostasis. Disruptions in the interaction between the mucosal immune system and its commensal microbiota have been associated with the onset of several diseases, including inflammatory bowel disease, asthma, and bacterial vaginosis. This review examines recent advances in nanoparticle (NP)-based strategies aimed at modulating mucosal immunity and restructuring microbial communities. It highlights how organic and inorganic NPs such as polysaccharide-based carriers, lipid NPs, and metallic nanomaterials enhance the delivery and stability of probiotics, prebiotics, and synbiotics, and facilitate targeted immunomodulation across gastrointestinal, respiratory, and female reproductive mucosal tissues. NP-based strategies are particularly emphasized for their ability to penetrate mucus barriers, facilitate microbial colonization, modulate cytokine activity, and enhance the restoration of epithelial barrier function. Disease-specific applications, including NP-based therapies for colitis, respiratory inflammation, and vaginal dysbiosis, are also discussed. In addition, this review outlines current challenges related to biosafety, targeting specificity, and clinical translation, and suggests future directions for research. Altogether, NP platforms offer a promising avenue for the precise modulation of mucosal immunity and microbiota, with significant potential in the prevention and treatment of mucosal-associated diseases.
Collapse
Affiliation(s)
- Won Jung
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Republic of Korea
| | - Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Republic of Korea
| |
Collapse
|
3
|
Adelfio M, Callen GE, He X, Paster BJ, Hasturk H, Ghezzi CE. Engineered Tissue Models to Decode Host-Microbiota Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417687. [PMID: 40364768 PMCID: PMC12199434 DOI: 10.1002/advs.202417687] [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] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 04/13/2025] [Indexed: 05/15/2025]
Abstract
A mutualistic co-evolution exists between the host and its associated microbiota in the human body. Bacteria establish ecological niches in various tissues of the body, locally influencing their physiology and functions, but also contributing to the well-being of the whole organism through systemic communication with other distant niches (axis). Emerging evidence indicates that when the composition of the microbiota inhabiting the niche changes toward a pathogenic state (dysbiosis) and interactions with the host become unbalanced, diseases may present. In addition, imbalances within a single niche can cause dysbiosis in distant organs. Current research efforts are focused on elucidating the mechanisms leading to dysbiosis, with the goal of restoring tissue homeostasis. In vitro models can provide critical experimental platforms to address this need, by reproducing the niche cyto-architecture and physiology with high fidelity. This review surveys current in in vitro host-microbiota research strategies and provides a roadmap that can guide the field in further developing physiologically relevant in vitro models of ecological niches, thus enabling investigation of the role of the microbiota in human health and diseases. Lastly, given the Food and Drug Administration Modernization Act 2.0, this review highlights emerging in vitro strategies to support the development and validation of new therapies on the market.
Collapse
Affiliation(s)
- Miryam Adelfio
- Department of Biomedical EngineeringUniversity of Massachusetts‐LowellLowellMA01854USA
| | - Grace E. Callen
- Department of Biomedical EngineeringUniversity of Massachusetts‐LowellLowellMA01854USA
| | - Xuesong He
- ADA Forsyth Institute245 First StCambridgeMA02142USA
| | | | | | - Chiara E. Ghezzi
- Department of Biomedical EngineeringUniversity of Massachusetts‐LowellLowellMA01854USA
| |
Collapse
|
4
|
Dorjee K, Sadoff RC, Mansour FR, Dorjee S, Binder EM, Stetson M, Yuen R, Kim H. Menstrual disturbance associated with COVID-19 vaccines: A comprehensive systematic review and meta-analysis. PLoS One 2025; 20:e0320162. [PMID: 40378132 PMCID: PMC12083795 DOI: 10.1371/journal.pone.0320162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/12/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND The relationship between COVID-19 vaccines and menstrual disturbance is unclear, in part because researchers have measured different outcomes (e.g., delays vs. changes to cycle length) with various study designs. Menstrual disruption could be a decisive factor in people's willingness to accept the COVID-19 vaccine. METHODS We searched Medline, Embase, and Web of Science for studies investigating menstrual cycle length, flow volume, post-menopausal bleeding, and unexpected or intermenstrual bleeding. Data were analyzed using fixed-effects meta-analysis with Shore's adjusted confidence intervals for heterogeneity. FINDINGS Seventeen studies with >1·9 million participants were analyzed. We found a 19% greater risk of increase in menstrual cycle length as compared to unvaccinated people or pre-vaccination time-periods (summary relative risk (sRR): 1·19; 95% CI: 1·11-1·26; n = 23,718 participants). The increase in risk was the same for Pfizer-BioNTech (sRR: 1·15; 1·05-1·27; n = 16,595) and Moderna vaccines (sRR: 1·15; 1·05-1·25; n = 7,523), similar for AstraZeneca (sRR: 1·27; 1·02-1·59; n = 532), and higher for the Janssen (sRR: 1·69; 1·14-2·52; n = 751) vaccine. In the first cycle after vaccination, length increased by INTERPRETATION We observed a mild increase in the risk of menstrual disturbance associated with COVID-19 vaccines. Such risks are likely clinically unmeaningful. Vaccine recipients should be appropriately counseled.
Collapse
Affiliation(s)
- Kunchok Dorjee
- Center for Tuberculosis and AIDS Research, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - R. C. Sadoff
- Center for Tuberculosis and AIDS Research, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Farima Rahimi Mansour
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sangyal Dorjee
- Center for Tuberculosis and AIDS Research, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Eli M. Binder
- Adams County Health Department, Brighton, Colorado, United States of America
| | - Maria Stetson
- Center for Tuberculosis and AIDS Research, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Center for Humanitarian Health, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Regina Yuen
- Center for Tuberculosis and AIDS Research, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
5
|
MacLean F, Tsegaye AT, Graham JB, Swarts JL, Vick SC, Potchen NB, Cruz Talavera I, Warrier L, Dubrulle J, Schroeder LK, Saito A, Mar C, Thomas KK, Mack M, Sabo MC, Chohan BH, Ngure K, Mugo NR, Lingappa JR, Lund JM, for the Kinga Study Team. Bacterial vaginosis associates with dysfunctional T cells and altered soluble immune factors in the cervicovaginal tract. J Clin Invest 2025; 135:e184609. [PMID: 40131862 PMCID: PMC12077898 DOI: 10.1172/jci184609] [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: 07/08/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUNDBacterial vaginosis (BV) is a dysbiosis of the vaginal microbiome that is prevalent among reproductive-age females worldwide. Adverse health outcomes associated with BV include an increased risk of sexually acquired HIV, yet the immunological mechanisms underlying this association are not well understood.METHODSTo investigate BV-driven changes to cervicovaginal tract (CVT) and circulating T cell phenotypes, Kinga Study participants with or without BV provided vaginal tract (VT) and ectocervical (CX) tissue biopsies and PBMC samples.RESULTSHigh-parameter flow cytometry revealed an increased frequency of cervical CD4+ conventional T (Tconv) cells expressing CCR5 in BR+ versus BR- women. However, we found no difference in the number of CD3+CD4+CCR5+ cells in the CX or VT of BV+ versus BV- individuals, suggesting that BV-driven increased HIV susceptibility may not be solely attributed to increased CVT HIV target cell abundance. Flow cytometry also revealed that individuals with BV had an increased frequency of dysfunctional CX and VT CD39+ Tconv and CX tissue-resident CD69+CD103+ Tconv cells, reported to be implicated in HIV acquisition risk and replication. Many soluble immune factor differences in the CVT further support that BV elicits diverse and complex CVT immune alterations.CONCLUSIONOur comprehensive analysis expands on potential immunological mechanisms that may underlie the adverse health outcomes associated with BV, including increased HIV susceptibility.TRIAL REGISTRATIONClinicalTrials.gov NCT03701802.FUNDINGThis work was supported by National Institutes of Health grants R01AI131914, R01AI141435, and R01AI129715.
Collapse
Affiliation(s)
- Finn MacLean
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Jessica B. Graham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jessica L. Swarts
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sarah C. Vick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Nicole B. Potchen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Irene Cruz Talavera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Lakshmi Warrier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Julien Dubrulle
- Cellular Imaging Shared Resource, Fred Hutchinson Cancer Center. Seattle, Washington, USA
| | - Lena K. Schroeder
- Cellular Imaging Shared Resource, Fred Hutchinson Cancer Center. Seattle, Washington, USA
| | - Ayumi Saito
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Corinne Mar
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Katherine K. Thomas
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Matthias Mack
- Department of Internal Medicine–Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Michelle C. Sabo
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Bhavna H. Chohan
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Center for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kenneth Ngure
- Department of Global Health, University of Washington, Seattle, Washington, USA
- School of Public Health, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Nelly Rwamba Mugo
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Jairam R. Lingappa
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jennifer M. Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
6
|
Taylor BD, Haggerty CL, Amabebe E, Richardson LS. Current Evidence of Maternal Infection With Chlamydia trachomatis and Preeclampsia Risk. Am J Reprod Immunol 2025; 93:e70080. [PMID: 40298141 DOI: 10.1111/aji.70080] [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: 07/19/2024] [Revised: 01/13/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Chlamydia trachomatis is the most common bacterial sexually transmitted infection (STI) in the United States. Ascending C. trachomatis can cause pelvic inflammatory disease (PID), potentially leading to subsequent infertility, ectopic pregnancy, and adverse pregnancy outcomes. There is growing evidence implicating infections (e.g., COVID-19, cytomegalovirus) in preeclampsia etiology, a maternal hypertensive disorder and leading cause of maternal morbidity and mortality. However, few studies have investigated the impact of STIs on preeclampsia risk. In this review, we provide an overview of the potential association between C. trachomatis and preeclampsia and identify future research needs through a critical evaluation of epidemiologic, in vitro, and in vivo studies. Unfortunately, current methodological limitations such as lower-quality study designs, selection bias, confounding bias, and variations in chlamydia diagnostic methods inhibit our understanding of the impact of C. trachomatis on preeclampsia. In addition, bench-side approaches such as animal models and in vitro studies have not elucidated the mechanisms linking C. trachomatis to preeclampsia. Understanding the biological pathways that could be disrupted by chlamydia is important as it may ultimately guide the development and use of novel therapeutics to augment standard antibiotic therapy to reduce pathology.
Collapse
Affiliation(s)
- Brandie DePaoli Taylor
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
- Academic Research, Advocate Aurora Research Institute, Milwaukee, Wisconsin, USA
| | - Catherine L Haggerty
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emmanuel Amabebe
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lauren S Richardson
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
7
|
Pérez-Ibave DC, Burciaga-Flores CH, García-Mejía X, Alcorta-Nuñez F, Solis-Coronado O, Escamilla MG, Vidal-Gutiérrez O, Garza-Rodríguez ML. Hallmarks of Bacterial Vaginosis. Diagnostics (Basel) 2025; 15:1090. [PMID: 40361908 PMCID: PMC12071977 DOI: 10.3390/diagnostics15091090] [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: 03/28/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Bacterial vaginosis (BV) is considered the most common cause of vaginal discharge, which is related to several public health issues, such as an increased risk for sexually transmitted infections, pelvic inflammatory disease, pregnancy-related problems such as abortion, stillbirth or premature birth, and tubal factor infertility. BV is not considered an infection but an imbalance in the vaginal microbiota, characterized by a substitution of the normal Lactobacilli flora by anaerobe. Reducing resistance against infections by several mechanisms, including bacterial homeostasis, stabilization of acid pH, inhibition of pathogens adhesion by polyamine degradation, production of anti-inflammatory molecules, surfactants, and antimicrobial substances like hydrogen peroxide, acids, and bacteriocins. Approximately half of women with BV can experience symptoms, which mainly include vaginal malodor, fishy discharge, stinging sensation, and increased vaginal pH. The treatment of BV is based primarily on promoting Lactobacilli restoration and eliminating dangerous microbiota with antibiotic therapy. However, there is a high rate of recurrence and relapse. Objective: Based on the current literature, this review aims to propose a list of ten BV hallmarks: dysbiosis, inflammation, apoptosis, pH basification, mucosal barrier integrity, pathway activation, epithelial damage, genomic instability, oxidative stress (OS), and metabolic reconfiguration. Conclusions: Understanding the causes of BV and the pathogenicity mechanisms is critical for preventing and improving the current therapeutic management of patients.
Collapse
Affiliation(s)
- Diana Cristina Pérez-Ibave
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico; (D.C.P.-I.); (C.H.B.-F.); (F.A.-N.); (O.S.-C.); (M.G.E.)
| | - Carlos Horacio Burciaga-Flores
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico; (D.C.P.-I.); (C.H.B.-F.); (F.A.-N.); (O.S.-C.); (M.G.E.)
| | - Ximena García-Mejía
- Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico;
| | - Fernando Alcorta-Nuñez
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico; (D.C.P.-I.); (C.H.B.-F.); (F.A.-N.); (O.S.-C.); (M.G.E.)
| | - Orlando Solis-Coronado
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico; (D.C.P.-I.); (C.H.B.-F.); (F.A.-N.); (O.S.-C.); (M.G.E.)
| | - Moisés González Escamilla
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico; (D.C.P.-I.); (C.H.B.-F.); (F.A.-N.); (O.S.-C.); (M.G.E.)
| | - Oscar Vidal-Gutiérrez
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico; (D.C.P.-I.); (C.H.B.-F.); (F.A.-N.); (O.S.-C.); (M.G.E.)
| | - María Lourdes Garza-Rodríguez
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 66451, Mexico; (D.C.P.-I.); (C.H.B.-F.); (F.A.-N.); (O.S.-C.); (M.G.E.)
| |
Collapse
|
8
|
Lyon LM, Marroquin SM, Thorstenson JC, Joyce LR, Fuentes EJ, Doran KS, Horswill AR. Genome-wide mutagenesis identifies factors involved in MRSA vaginal colonization. Cell Rep 2025; 44:115421. [PMID: 40085646 DOI: 10.1016/j.celrep.2025.115421] [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: 07/29/2024] [Revised: 01/17/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an opportunistic pathogen that colonizes various body sites, including the nares, skin, and vagina. During pregnancy,colonization can lead to dysbiosis, adverse pregnancy outcomes, and invasive disease. To identify genes contributing to MRSA vaginal fitness, we performed transposon sequencing (Tn-seq) using a murine model of vaginal colonization, identifying over 250 conditionally essential genes. Five genes were validated in our murine model, including those encoding the aerobic respiration protein QoxB, bacillithiol biosynthesis component BshB2, sialic acid catabolism enzyme NanE, and staphylococcal regulator of respiration SrrAB. RNA sequencing and comparative analysis identified over 30 SrrAB-regulated genes potentially important for fitness in vaginal-like conditions, particularly under oxygen stress. These findings highlight pathways such as aerobic respiration, bacillithiol biosynthesis, sialic acid catabolism, and transcriptional regulation that support MRSA's competitive fitness in the vaginal tract.
Collapse
Affiliation(s)
- Laurie M Lyon
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, CO, USA
| | - Stephanie M Marroquin
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, CO, USA
| | - John C Thorstenson
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, CO, USA
| | - Luke R Joyce
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, CO, USA
| | - Ernesto J Fuentes
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kelly S Doran
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, CO, USA
| | - Alexander R Horswill
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, CO, USA; Department of Veterans Affairs Eastern, Colorado Healthcare System, Aurora, CO, USA.
| |
Collapse
|
9
|
Castellanos-Ruiz D, Ojeda-Borbolla JG, Ruiz-García OV, Peña-Corona SI, Martínez-Peña AA, Ibarra-Rubio ME, Gavilanes-Ruiz M, Mendoza-Rodríguez CA. Uterine Microbiota and Bisphenols: Novel Influencers in Reproductive Health. J Xenobiot 2025; 15:26. [PMID: 39997369 PMCID: PMC11856463 DOI: 10.3390/jox15010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Infertility affects 8-12% of couples worldwide, and 30-75% of preclinical pregnancy losses are due to a failure during the implantation process. Exposure to endocrine disruptors, like bisphenols, among others, has been associated with the increase in infertility observed in the past decades. An increase in infertility has correlated with exposure to endocrine disruptors like bisphenols. The uterus harbors its own microbiota, and changes in this microbiota have been linked to several gynecological conditions, including reproductive failure. There are no studies on the effects of bisphenols on the uterine-microbiota composition, but some inferences can be gleaned by looking at the gut. Bisphenols can alter the gut microbiota, and the molecular mechanism by which gut microbiota regulates intestinal permeability involves Toll-like receptors (TLRs) and tight junction (TJ) proteins. TJs participate in embryo implantation in the uterus, but bisphenol exposure disrupts the expression and localization of TJ proteins. The aim of this review is to summarize the current knowledge on the microbiota of the female reproductive tract (FRT), its association with different reproductive diseases-particularly reproductive failure-the effects of bisphenols on microbiota composition and reproductive health, and the molecular mechanisms regulating uterine-microbiota interactions crucial for embryo implantation. This review also highlights existing knowledge gaps and outlines research needs for future risk assessments regarding the effects of bisphenols on reproduction.
Collapse
Affiliation(s)
- Dafne Castellanos-Ruiz
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - J. Gerardo Ojeda-Borbolla
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - Olga V. Ruiz-García
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - Sheila I. Peña-Corona
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Annia A. Martínez-Peña
- División de Ciencias de la Salud, Universidad Intercontinental, A. C., Ciudad de México 14420, Mexico
| | - María Elena Ibarra-Rubio
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - Marina Gavilanes-Ruiz
- Facultad de Química, Departamento de Bioquímica, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - C. Adriana Mendoza-Rodríguez
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| |
Collapse
|
10
|
Sun Y, Cai J, Zhang Y, Bao S. A high concentration of neutrophil extracellular traps is observed in humans and mice suffering from endometriosis. J Reprod Immunol 2025; 167:104414. [PMID: 39657366 DOI: 10.1016/j.jri.2024.104414] [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: 08/02/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024]
Abstract
We wished to ascertain if there is an association between neutrophil extracellular traps and endometriosis (EMS). We collected the lesional tissues and normal endometrium of 30 patients suffering from endometriosis. Samples were also taken from healthy controls. Blood from the peripheral circulation was collected to isolate serum and neutrophils. A mouse model of endometriosis was also created. Expression of citrullinated histone and the myeloperoxidase level in tissue were measured by immunofluorescence staining and western blotting. The myeloperoxidase level in peripheral blood serum was measured by enzyme-linked immunosorbent assay. Staining (Trypan Blue) and flow cytometry were used to measure the apoptosis of neutrophils in peripheral blood. BALB/C mice were modeled by allotransplantation, and the experimental parameters noted above quantified. The myeloperoxidase content in the peripheral blood of patients with endometriosis was increased compared with that in healthy controls. Flow cytometry showed that the percent apoptosis of neutrophils in patients with endometriosis was lower than that in healthy controls. Expression of citrullinated histone was higher in the endometriosis group in humans and mice compared with respective controls according to immunofluorescence staining and western blotting. Our data suggest that a high concentration of neutrophil extracellular traps was observed in humans and mice suffering from endometriosis.
Collapse
Affiliation(s)
- Yuting Sun
- Department of Gynecology and Obstetrics Mudanjiang Medical University Affliated Honggi Hospital, No.5 Tongxiang Road, Aimin District, Mudanjiang,Heilongjiang 157011, China
| | - Junhong Cai
- Medical Laboratory Center, Hainan General Hospital, Haikou 570102, China
| | - Yanan Zhang
- Department of Gynecology and Obstetrics Mudanjiang Medical University Affliated Honggi Hospital, No.5 Tongxiang Road, Aimin District, Mudanjiang,Heilongjiang 157011, China
| | - Shan Bao
- Department of Gynaecology and Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China.
| |
Collapse
|
11
|
MacLean F, Tsegaye AT, Graham JB, Swarts JL, Vick SC, Potchen N, Talavera IC, Warrier L, Dubrulle J, Schroeder LK, Saito A, Thomas KK, Mack M, Sabo MC, Chohan BH, Ngure K, Mugo N, Lingappa JR, Lund JM. Bacterial vaginosis-driven changes in cervicovaginal immunity that expand the immunological hypothesis for increased HIV susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.03.601916. [PMID: 39005354 PMCID: PMC11245000 DOI: 10.1101/2024.07.03.601916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Bacterial vaginosis (BV) is a dysbiosis of the vaginal microbiome that is prevalent among reproductive-age females worldwide. Adverse health outcomes associated with BV include an increased risk of sexually-acquired HIV, yet the immunological mechanisms underlying this association are not well understood. To investigate BV-driven changes to cervicovaginal tract (CVT) and circulating T cell phenotypes, participants with or without BV provided vaginal tract (VT) and ectocervical (CX) tissue biopsies and PBMC samples. High-parameter flow cytometry revealed an increased frequency of cervical conventional CD4+ T cells (Tconv) expressing CCR5. However, we found no difference in number of CD3+CD4+CCR5+ cells in the CX or VT of BV+ vs BV- individuals, suggesting that BV-driven increased HIV susceptibility may not be solely attributed to increased CVT HIV target cell abundance. Flow cytometry also revealed that individuals with BV have an increased frequency of dysfunctional CX and VT CD39+ Tconv and CX tissue-resident CD69+CD103+ Tconv, reported to be implicated in HIV acquisition risk and replication. Many soluble immune factor differences in the CVT further support that BV elicits diverse and complex CVT immune alterations. Our comprehensive analysis expands on potential immunological mechanisms that may underlie the adverse health outcomes associated with BV including increased HIV susceptibility.
Collapse
Affiliation(s)
- Finn MacLean
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | | | - Jessica B. Graham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Jessica L. Swarts
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Sarah C. Vick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Nicole Potchen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Irene Cruz Talavera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Lakshmi Warrier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Julien Dubrulle
- Cellular Imaging Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Lena K. Schroeder
- Cellular Imaging Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Ayumi Saito
- Department of Global Health, University of Washington, Seattle, USA
| | | | - Matthias Mack
- Department of Internal Medicine-Nephrology, University Hospital Regensburg, Regensburg, Germany
| | | | - Bhavna H. Chohan
- Department of Global Health, University of Washington, Seattle, USA
- Center for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kenneth Ngure
- Department of Global Health, University of Washington, Seattle, USA
- School of Public Health, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Nelly Mugo
- Department of Global Health, University of Washington, Seattle, USA
- Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Jairam R. Lingappa
- Department of Global Health, University of Washington, Seattle, USA
- Department of Medicine, University of Washington, Seattle, USA
- Department of Pediatrics, University of Washington, Seattle, USA
| | - Jennifer M. Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
- Department of Global Health, University of Washington, Seattle, USA
| |
Collapse
|
12
|
Zhang YL, Zhou YY, Ke LJ, Sheng J, Zou DY, Tang TT, Yang ZY, Chen L, Hou XC, Zhu J, Xu JB, Zhu YX, Zhou WL. Lipopolysaccharide Triggers Luminal Acidification to Promote Defense Against Bacterial Infection in Vaginal Epithelium. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2290-2301. [PMID: 39222908 DOI: 10.1016/j.ajpath.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
The vaginal epithelium plays pivotal roles in host defense against pathogen invasion, contributing to the maintenance of an acidic microenvironment within the vaginal lumen through the activity of acid-base transport proteins. However, the precise defense mechanisms of the vaginal epithelium after a bacterial infection remain incompletely understood. This study showed that bacterial lipopolysaccharide (LPS) potentiated net proton efflux by up-regulating the expression of Na+-H+ exchanger 1 (NHE1) in vaginal epithelial cells. Pharmacologic inhibition or genetic knockdown of Toll-like receptor-4 and the extracellular signal-regulated protein kinase signaling pathway effectively counteracted the up-regulation of NHE1 and the enhanced proton efflux triggered by LPS in vaginal epithelial cells. In vivo studies revealed that LPS administration led to luminal acidification through the up-regulation of NHE1 expression in the rat vagina. Moreover, inhibition of NHE exhibited an impaired defense against acute bacterial infection in the rat vagina. These findings collectively indicate the active involvement of vaginal epithelial cells in facilitating luminal acidification during acute bacterial infection, offering potential insights into the treatment of bacterial vaginosis.
Collapse
Affiliation(s)
- Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, Sun Yat-sen University, Guangzhou, China.
| | - Yu-Yun Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Li-Jiao Ke
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jie Sheng
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Dan-Yang Zou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ting-Ting Tang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zi-Ying Yang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Chun Hou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jie Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jian-Bang Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
13
|
Zhao X, Shi W, Li Z, Zhang W. Linking reproductive tract microbiota to premature ovarian insufficiency: Pathophysiological mechanisms and therapies. J Reprod Immunol 2024; 166:104325. [PMID: 39265315 DOI: 10.1016/j.jri.2024.104325] [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: 06/20/2024] [Revised: 08/06/2024] [Accepted: 09/01/2024] [Indexed: 09/14/2024]
Abstract
Over the past decade, research on the human microbiota has become a hot topic. Among them, the female reproductive tract (FRT) also has a specific microbiota that maintains the body's health and dynamic balance, especially in the reproductive aspect. When the FRT ecosystem is dysregulated, changes in immune and metabolic signals can lead to pathological and physiological changes such as chronic inflammation, epithelial barrier disruption, changes in cell proliferation and apoptosis, and dysregulation of angiogenesis and metabolism, thereby causing disruption of the female endocrine system. Premature ovarian insufficiency (POI), a clinical syndrome of ovarian dysfunction, is primarily influenced by immune, genetic, and environmental factors. New evidence suggests that dysbiosis of the FRT microbiota and/or the presence of specific bacteria may contribute to the occurrence and progression of POI. This influence occurs through both direct and indirect mechanisms, including the regulation of estrogen metabolism. The use of probiotics or microbiota transplantation to regulate the microbiome has also been proven to be beneficial in improving ovarian function and the quality of life in women with premature aging. This article provides an overview of the interrelationships and roles between the FRT microbiome and POI in recent years, to fully understand the risk factors affecting female reproductive health, and to offer insights for the future diagnosis, treatment, and application of the FRT microbiome in POI patients.
Collapse
Affiliation(s)
- Xi Zhao
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| | - Wenying Shi
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| | - Zhengyu Li
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| | - Wei Zhang
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| |
Collapse
|
14
|
Obuobi S, Škalko-Basnet N. Understanding vaginal biofilms: The first step in harnessing antimicrobial nanomedicine. J Control Release 2024; 376:1190-1208. [PMID: 39510257 DOI: 10.1016/j.jconrel.2024.10.064] [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: 06/17/2024] [Revised: 10/02/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
In spite of multipurpose technologies offering broad-spectrum prevention for sexually transmitted infections (STIs) and contraception, the STIs incidences rise worldwide. The situation is even more alarming considering continuous rise in antimicrobial resistance (AMR) that limits therapy options. In this review we address the specific challenges of efficiently treating vaginal infections locally, at the infection site, by understanding the underlying barriers to efficient treatment such as vaginal biofilms. Knowledge on vaginal biofilms remains, up to now, rather scarce and requires more attention. We therefore propose a 'back to basics' insight that seeks to probe the complexity and role of the vaginal microbiota, its relationship with vaginal biofilms and implications to future therapeutic modalities utilizing advanced nano delivery systems. Our key objective is to highlight the interplay between biofilm, (nano)formulation and therapy outcome rather than provide an overview of all nanoformulations that were challenged against biofilms. We focused on the anatomy of the female reproductive organ and its physiological changes from birth, the unique vaginal microenvironment in premenopausal and postmenopausal women, vaginal biofilm infections and current nanomedicine-based approaches to treat infections in the vaginal site. Finally, we offer our perspectives on the current challenges associated with vaginal delivery and key considerations that can aid in the design and development of safer and potent products against persisting vaginal infections.
Collapse
Affiliation(s)
- Sybil Obuobi
- Drug Transport and Delivery Research Group, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Tromsø, Norway.
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
15
|
Sadat Larijani M, Eybpoosh S, Doroud D, Bavand A, Moradi L, Ashrafian F, Tajmehrabi Namini P, Zali M, Ramezani A. Assessment of COVID-19 Vaccine Impact on Women's Menstrual Health within an 18-Month Follow-Up. Obstet Gynecol Int 2024; 2024:7344506. [PMID: 39364209 PMCID: PMC11449560 DOI: 10.1155/2024/7344506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Considering menstruation as a crucial factor in females' health and fertility, any factor that could change its cycle is important. This study was conducted from April 2021 to October 2022 on females who got 3 doses of vaccines against SARS-CoV-2 through different platforms. The participants were requested to provide the trained experts with any changes regarding menstrual cycles after each dose of the vaccine up to 6 months after the booster shots. The disturbances related to the vaccines were identified by the adverse events committee to find possible associations with the applied vaccines. Of 308 women who participated until the end of the study, 22 (7.1%) complained about at least one abnormality in their menstrual patterns. The most common disturbance was metrorrhagia as 10 (48%) incidences followed by menorrhagia as 6 events (24.2%). Notably, the identified complaints were persistent in 59% of the patients. In addition, 14 studied cases developed COVID-19 infection after menstrual disorders. In these cases, COVID-19 could also play a role in the persistence of postvaccine menstrual disturbances. COVID-19 vaccination could affect menstrual cycle in women with no remarkable previous medical history. More longitudinal studies are required regarding this issue.
Collapse
Affiliation(s)
| | - Sana Eybpoosh
- Department of Epidemiology and BiostatisticsResearch Centre for Emerging and Reemerging Infectious DiseasesPasteur Institute of Iran, Tehran, Iran
| | - Delaram Doroud
- Quality Control DepartmentProduction and Research ComplexPasteur Institute of Iran, Tehran, Iran
| | - Anahita Bavand
- Clinical Research DepartmentPasteur Institute of Iran, Tehran, Iran
| | - Ladan Moradi
- Clinical Research DepartmentPasteur Institute of Iran, Tehran, Iran
| | | | | | - Mahsan Zali
- Clinical Research DepartmentPasteur Institute of Iran, Tehran, Iran
| | - Amitis Ramezani
- Clinical Research DepartmentPasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
16
|
Al Shahrani A, Alhumaidan N, Alzelfawi L, AlDosari L, Alhindawi Z, Alotaibi N, Aljohani R. Prevalence of menstrual alterations following COVID-19 vaccination: systematic review & meta-analysis. BMC Womens Health 2024; 24:523. [PMID: 39300461 PMCID: PMC11412023 DOI: 10.1186/s12905-024-03349-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND COVID-19 vaccines can lead to diverse local and systemic side effects, but there is limited evidence concerning their association with menstrual cycle changes. This study aimed to assess the prevalence of menstrual cycle alterations after COVID-19 vaccination among adult women. METHODS We systematically searched the PubMed, Web of Science and Science Direct databases for observational studies that included adult women and investigated the range of menstrual alterations. The quality of the studies was evaluated via the Newcastle-Ottawa scale. All the data were analyzed via Comprehensive Meta-Analysis Software Version 4.0. Forest plots were created to calculate the individual and pooled prevalence rates of different types of menstrual changes and 95% confidence intervals (CI) via fixed-effects and random-effects models, as appropriate. Heterogeneity was assessed with Q statistics and the I2 test. RESULTS Eleven studies, encompassing 26,283 adult women, met our eligibility criteria. Among the selected studies, five were cohort studies, five were cross-sectional studies, and one employed a case‒control design. The menstrual changes included abnormal cycle duration, dysmenorrhea, irregular cycles, and abnormal cycle flow (heavy and light flow), with pooled percentages of 27.3% (CI: 7.2-64.6%), 22% (CI: 5.2-59.4%), 16% (CI: 5.8-37.2%), 11.7% (CI: 5.8-22%), and 5.5% (CI: 2.3-12.5%), respectively. CONCLUSIONS This review highlights the prevalence of menstrual changes after COVID-19 vaccination and emphasizes the importance of considering menstrual health as an integral part of postvaccination monitoring and health care interventions. However, longitudinal studies are essential for establishing a definitive causal relationship between COVID-19 vaccination and menstrual alterations.
Collapse
Affiliation(s)
- Abeer Al Shahrani
- Department of Family and Community Medicine, College of Medicine, Princess Nourah bint Abdulrahman University, P.O.Box 84428, Riyadh, 11671, Saudi Arabia.
| | - Norah Alhumaidan
- College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Lama Alzelfawi
- College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Lena AlDosari
- College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Zeena Alhindawi
- College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Nahlah Alotaibi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Renad Aljohani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
van Rosmalen L, Zhu J, Maier G, Gacasan EG, Lin T, Zhemchuzhnikova E, Rothenberg V, Razu S, Deota S, Ramasamy RK, Sah RL, McCulloch AD, Hut RA, Panda S. Multi-organ transcriptome atlas of a mouse model of relative energy deficiency in sport. Cell Metab 2024; 36:2015-2037.e6. [PMID: 39232281 PMCID: PMC11378950 DOI: 10.1016/j.cmet.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/23/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
Insufficient energy intake to meet energy expenditure demands of physical activity can result in systemic neuroendocrine and metabolic abnormalities in activity-dependent anorexia and relative energy deficiency in sport (REDs). REDs affects >40% of athletes, yet the lack of underlying molecular changes has been a hurdle to have a better understanding of REDs and its treatment. To assess the molecular changes in response to energy deficiency, we implemented the "exercise-for-food" paradigm, in which food reward size is determined by wheel-running activity. By using this paradigm, we replicated several aspects of REDs in female and male mice with high physical activity and gradually reduced food intake, which results in weight loss, compromised bone health, organ-specific mass changes, and altered rest-activity patterns. By integrating transcriptomics of 19 different organs, we provide a comprehensive dataset that will guide future understanding of REDs and may provide important implications for metabolic health and (athletic) performance.
Collapse
Affiliation(s)
- Laura van Rosmalen
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jiaoyue Zhu
- Chronobiology unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, the Netherlands
| | - Geraldine Maier
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Erica G Gacasan
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Terry Lin
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Elena Zhemchuzhnikova
- Chronobiology unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, the Netherlands
| | - Vince Rothenberg
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Swithin Razu
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shaunak Deota
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ramesh K Ramasamy
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Robert L Sah
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrew D McCulloch
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Roelof A Hut
- Chronobiology unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, the Netherlands
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
18
|
Mertelsmann AM, Bowers SF, Wright D, Maganga JK, Mazigo HD, Ndhlovu LC, Changalucha JM, Downs JA. Effects of Schistosoma haematobium infection and treatment on the systemic and mucosal immune phenotype, gene expression and microbiome: A systematic review. PLoS Negl Trop Dis 2024; 18:e0012456. [PMID: 39250522 PMCID: PMC11412685 DOI: 10.1371/journal.pntd.0012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/19/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Urogenital schistosomiasis caused by Schistosoma haematobium affects approximately 110 million people globally, with the majority of cases in low- and middle-income countries. Schistosome infections have been shown to impact the host immune system, gene expression, and microbiome composition. Studies have demonstrated variations in pathology between schistosome subspecies. In the case of S. haematobium, infection has been associated with HIV acquisition and bladder cancer. However, the underlying pathophysiology has been understudied compared to other schistosome species. This systematic review comprehensively investigates and assimilates the effects of S. haematobium infection on systemic and local host mucosal immunity, cellular gene expression and microbiome. METHODS We conducted a systematic review assessing the reported effects of S. haematobium infections and anthelmintic treatment on the immune system, gene expression and microbiome in humans and animal models. This review followed PRISMA guidelines and was registered prospectively in PROSPERO (CRD42022372607). Randomized clinical trials, cohort, cross-sectional, case-control, experimental ex vivo, and animal studies were included. Two reviewers performed screening independently. RESULTS We screened 3,177 studies and included 94. S. haematobium was reported to lead to: (i) a mixed immune response with a predominant type 2 immune phenotype, increased T and B regulatory cells, and select pro-inflammatory cytokines; (ii) distinct molecular alterations that would compromise epithelial integrity, such as increased metalloproteinase expression, and promote immunological changes and cellular transformation, specifically upregulation of genes p53 and Bcl-2; and (iii) microbiome dysbiosis in the urinary, intestinal, and genital tracts. CONCLUSION S. haematobium induces distinct alterations in the host's immune system, molecular profile, and microbiome. This leads to a diverse range of inflammatory and anti-inflammatory responses and impaired integrity of the local mucosal epithelial barrier, elevating the risks of secondary infections. Further, S. haematobium promotes cellular transformation with oncogenic potential and disrupts the microbiome, further influencing the immune system and genetic makeup. Understanding the pathophysiology of these interactions can improve outcomes for the sequelae of this devastating parasitic infection.
Collapse
Affiliation(s)
- Anna M Mertelsmann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Sheridan F Bowers
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Drew Wright
- Samuel J. Wood Library & C.V. Starr Biomedical Information Center, Weill Cornell Medical College, New York, New York, United States of America
| | - Jane K Maganga
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Humphrey D Mazigo
- Department of Parasitology and Entomology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - John M Changalucha
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Jennifer A Downs
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
- Weill Bugando School of Medicine, Mwanza, Tanzania
| |
Collapse
|
19
|
Marečková M, Garcia-Alonso L, Moullet M, Lorenzi V, Petryszak R, Sancho-Serra C, Oszlanczi A, Icoresi Mazzeo C, Wong FCK, Kelava I, Hoffman S, Krassowski M, Garbutt K, Gaitskell K, Yancheva S, Woon EV, Male V, Granne I, Hellner K, Mahbubani KT, Saeb-Parsy K, Lotfollahi M, Prigmore E, Southcombe J, Dragovic RA, Becker CM, Zondervan KT, Vento-Tormo R. An integrated single-cell reference atlas of the human endometrium. Nat Genet 2024; 56:1925-1937. [PMID: 39198675 PMCID: PMC11387200 DOI: 10.1038/s41588-024-01873-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/17/2024] [Indexed: 09/01/2024]
Abstract
The complex and dynamic cellular composition of the human endometrium remains poorly understood. Previous endometrial single-cell atlases profiled few donors and lacked consensus in defining cell types. We introduce the Human Endometrial Cell Atlas (HECA), a high-resolution single-cell reference atlas (313,527 cells) combining published and new endometrial single-cell transcriptomics datasets of 63 women with and without endometriosis. HECA assigns consensus and identifies previously unreported cell types, mapped in situ using spatial transcriptomics and validated using a new independent single-nuclei dataset (312,246 nuclei, 63 donors). In the functionalis, we identify intricate stromal-epithelial cell coordination via transforming growth factor beta (TGFβ) signaling. In the basalis, we define signaling between fibroblasts and an epithelial population expressing progenitor markers. Integration of HECA with large-scale endometriosis genome-wide association study data pinpoints decidualized stromal cells and macrophages as most likely dysregulated in endometriosis. The HECA is a valuable resource for studying endometrial physiology and disorders, and for guiding microphysiological in vitro systems development.
Collapse
Affiliation(s)
- Magda Marečková
- Wellcome Sanger Institute, Cambridge, UK
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | | | | | - Valentina Lorenzi
- Wellcome Sanger Institute, Cambridge, UK
- European Bioinformatics Institute-European Molecular Biology Laboratory, Cambridge, UK
| | | | | | | | | | | | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | | | - Michał Krassowski
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kurtis Garbutt
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Kezia Gaitskell
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford, UK
| | - Slaveya Yancheva
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford, UK
| | - Ee Von Woon
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
- The Fertility Centre, Chelsea and Westminster Hospital, London, UK
| | - Victoria Male
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Ingrid Granne
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Karin Hellner
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Krishnaa T Mahbubani
- Department of Haematology, University of Cambridge, Cambridge, UK
- Cambridge Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Cambridge Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Mohammad Lotfollahi
- Wellcome Sanger Institute, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | | - Jennifer Southcombe
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Rebecca A Dragovic
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Christian M Becker
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Krina T Zondervan
- Oxford Endometriosis Care Centre, Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK.
- Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | |
Collapse
|
20
|
Li Y, Zhai Y, Fu B, He Y, Feng Y, Ma F, Lu H. A comprehensive N-glycome map of porcine sperm membrane before and after capacitation. Carbohydr Polym 2024; 335:122084. [PMID: 38616102 DOI: 10.1016/j.carbpol.2024.122084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/05/2024] [Accepted: 03/21/2024] [Indexed: 04/16/2024]
Abstract
Mapping the N-glycome of porcine sperm before and after sperm capacitation is important for understanding the rearrangement of glycoconjugates during capacitation. In this work, we characterized the N-glycome on the membranes of 18 pairs of fresh porcine sperm before capacitation and porcine sperm after capacitation by MALDI-MS (Matrix-assisted laser desorption/ionization-mass spectrometry). A total of 377 N-glycans were detected and a comprehensive N-glycome map of porcine sperm membranes before and after capacitation was generated, which presents the largest N-glycome dataset of porcine sperm cell membranes. Statistical analysis revealed a significantly higher level of high mannose glycosylation and a significantly lower level of fucosylation, galactosylation, and α-2,6-NeuAc after capacitation, which is further verified by flow cytometry and lectin blotting. This research reveals new insights into the relationship between N-glycosylation variations and sperm capacitation, including the underlying mechanisms of the capacitation process.
Collapse
Affiliation(s)
- Yueyue Li
- Liver Cancer Institute, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yujia Zhai
- Department of Medical Genetics/Prenatal Diagnostic Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China
| | - Bing Fu
- Department of Chemistry, NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China
| | - Yuanlin He
- College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ying Feng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Haojie Lu
- Liver Cancer Institute, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Chemistry, NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China.
| |
Collapse
|
21
|
Shaffer Z, Romero R, Tarca AL, Galaz J, Arenas-Hernandez M, Gudicha DW, Chaiworapongsa T, Jung E, Suksai M, Theis KR, Gomez-Lopez N. The vaginal immunoproteome for the prediction of spontaneous preterm birth: A retrospective longitudinal study. eLife 2024; 13:e90943. [PMID: 38913421 PMCID: PMC11196114 DOI: 10.7554/elife.90943] [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: 07/12/2023] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Background Preterm birth is the leading cause of neonatal morbidity and mortality worldwide. Most cases of preterm birth occur spontaneously and result from preterm labor with intact (spontaneous preterm labor [sPTL]) or ruptured (preterm prelabor rupture of membranes [PPROM]) membranes. The prediction of spontaneous preterm birth (sPTB) remains underpowered due to its syndromic nature and the dearth of independent analyses of the vaginal host immune response. Thus, we conducted the largest longitudinal investigation targeting vaginal immune mediators, referred to herein as the immunoproteome, in a population at high risk for sPTB. Methods Vaginal swabs were collected across gestation from pregnant women who ultimately underwent term birth, sPTL, or PPROM. Cytokines, chemokines, growth factors, and antimicrobial peptides in the samples were quantified via specific and sensitive immunoassays. Predictive models were constructed from immune mediator concentrations. Results Throughout uncomplicated gestation, the vaginal immunoproteome harbors a cytokine network with a homeostatic profile. Yet, the vaginal immunoproteome is skewed toward a pro-inflammatory state in pregnant women who ultimately experience sPTL and PPROM. Such an inflammatory profile includes increased monocyte chemoattractants, cytokines indicative of macrophage and T-cell activation, and reduced antimicrobial proteins/peptides. The vaginal immunoproteome has improved predictive value over maternal characteristics alone for identifying women at risk for early (<34 weeks) sPTB. Conclusions The vaginal immunoproteome undergoes homeostatic changes throughout gestation and deviations from this shift are associated with sPTB. Furthermore, the vaginal immunoproteome can be leveraged as a potential biomarker for early sPTB, a subset of sPTB associated with extremely adverse neonatal outcomes. Funding This research was conducted by the Perinatology 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) under contract HHSN275201300006C. ALT, KRT, and NGL were supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health.
Collapse
Affiliation(s)
- Zachary Shaffer
- 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, University of MichiganAnn ArborUnited States
- Department of Epidemiology and Biostatistics, Michigan State UniversityEast LansingUnited States
| | - Adi L Tarca
- 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Computer Science, Wayne State University College of EngineeringDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de ChileSantiagoChile
| | - 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Dereje W Gudicha
- 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Tinnakorn Chaiworapongsa
- 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Eunjung Jung
- 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Manaphat Suksai
- 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of MedicineDetroitUnited States
| | - 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, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of MedicineDetroitUnited States
| |
Collapse
|
22
|
Zhang Y, He Z. Inflammatory mediators in bacterial vaginosis: The role of cytokines. APMIS 2024; 132:245-255. [PMID: 38345182 DOI: 10.1111/apm.13380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/03/2024] [Indexed: 03/14/2024]
Abstract
BV is a significant concern in women's health with a varying prevalence rate in different cities of China. The condition has been linked to the acquisition of STIs, including HIV and HPV, and can lead to infertility, adverse obstetric outcomes. We conducted a comprehensive literature search in the PubMed. The search was performed from 01/01/2018 to 01/09/2023. The following search terms were used: bacterial vaginosis and cytokine. We also manually searched the reference lists of included studies and relevant reviews to identify additional articles. The presence of Gardnerella spp. can lead to changes in cytokine levels. The immune system of the female reproductive tract consists of various immune cells and molecules that play a vital role in defending against infections. Cytokines, signaling molecules involved in immune cell recruitment and activation, have been identified as potential biomarkers for diagnosing BV and predicting STIs. Current treatments for BV primarily involve antibiotics, but there is a high recurrence rate posttreatment. BV is a complex condition that affects a significant number of women worldwide. The role of cytokines in the onset, progression, and treatment of BV offers promising avenues for future research and potential diagnostic and therapeutic advancements.
Collapse
Affiliation(s)
- Yuexin Zhang
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Zhi He
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
23
|
Das S, Konwar BK. Influence of connatural factors in shaping vaginal microflora and ensuring its health. Arch Gynecol Obstet 2024; 309:871-886. [PMID: 37676318 DOI: 10.1007/s00404-023-07200-8] [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: 06/12/2022] [Accepted: 08/21/2023] [Indexed: 09/08/2023]
Abstract
Vaginal canal (VC) is exposed to the external environment affected by habitual factors like hygiene and sexual behaviour as well as physiological factors like puberty, menstrual cycle, pregnancy, child birth and menopause. Healthy VC harbours beneficial microflora supported by vaginal epithelium and cervical fluid. Connatural antimicrobial peptide (AMPs) of female reproductive tract (FRT) conjunctly with these beneficial microbes provide protection from a large number of infectious diseases. Such infections may either be caused by native microbes of the VC or transitory microbes like bacteria or virus which are not a part of VC microflora. This review highlight's the role of hormones, enzymes, innate immunological factors, epithelial cells and vaginal mucus that support beneficial microbes over infectious ones thus, helping to maintain homeostasis in VC and further protect the FRT. We also discuss the prospective use of vaginal probiotics and AMPs against pathogens which can serve as a potential cure for vaginal infections.
Collapse
Affiliation(s)
- Shreaya Das
- Department of MBBT, Tezpur University, Napaam, Assam, 784028, India.
| | - Bolin K Konwar
- Department of MBBT, Tezpur University, Napaam, Assam, 784028, India
| |
Collapse
|
24
|
Panneer Selvam S, Ramadoss R, Shanmugam R, Sundar S, Ta L, Ramani P. Assessment of Female Hormonal Influence on COVID-19 Vaccine Response: A Prospective Cohort Study. Cureus 2024; 16:e54417. [PMID: 38510901 PMCID: PMC10950846 DOI: 10.7759/cureus.54417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
INTRODUCTION The diversity of oral epithelial cells offers potential viral infection sites. The lower level of ACE2 inhibitors in women's blood renders them more resistant to coronavirus disease 2019 (COVID-19). In order to determine the effect of COVID-19 vaccination on female hormones, salivary levels of total antibody, immunoglobulin G (IgG), and cortisol were measured in young and elderly women. METHODS Saliva samples from 88 participants were collected and subjected to ELISA for detecting total antibody, IgG, and cortisol. RESULTS Women who were infected with COVID-19 and who completed two doses of vaccination had more IgG antibodies when compared to the uninfected individuals/single-dose/non-vaccinated individuals. The cortisol levels in post-menopausal women were higher than those in women with normal menstrual cycles, and the difference was statistically significant (P-value 0.00). The increased cortisol levels were well correlated with increased levels of IgG antibodies which was statistically significant (Spearman rho P value 0.00) Conclusions: COVID variants will continue to mutate and evolve as long as the epidemic persists. The higher cortisol and IgG antibodies produced by female hormones protect them from COVID-19 infection.
Collapse
Affiliation(s)
- Suganya Panneer Selvam
- Oral Pathology and Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Ramya Ramadoss
- Oral Pathology and Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - RajeshKumar Shanmugam
- Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Sandhya Sundar
- Oral Pathology and Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Lakshmi Ta
- Oral Pathology and Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Pratibha Ramani
- Oral Pathology, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| |
Collapse
|
25
|
Murphy K, Gromisch M, Srinivasan S, Wang T, Wood L, Proll S, Liu C, Fiedler T, Valint DJ, Fredricks DN, Keller MJ, Herold BC. IgA coating of vaginal bacteria is reduced in the setting of bacterial vaginosis (BV) and preferentially targets BV-associated species. Infect Immun 2024; 92:e0037323. [PMID: 38099624 PMCID: PMC10790818 DOI: 10.1128/iai.00373-23] [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: 09/11/2023] [Accepted: 11/29/2023] [Indexed: 01/17/2024] Open
Abstract
Immunoglobulin (Ig) bacterial coating has been described in the gastrointestinal tract and linked to inflammatory bowel disease; however, little is known about Ig coating of vaginal bacteria and whether it plays a role in vaginal health including bacterial vaginosis (BV). We examined Ig coating in 18 women with symptomatic BV followed longitudinally before, 1 week, and 1 month after oral metronidazole treatment. Immunoglobulin A (IgA) and/or immunoglobulin G (IgG) coating of vaginal bacteria was assessed by flow cytometry, and Ig coated and uncoated bacteria were sorted and characterized using 16S rRNA sequencing. Despite higher levels of IgG compared to IgA in cervicovaginal fluid, the predominant Ig coating the bacteria was IgA. The majority of bacteria were uncoated at all visits, but IgA coating significantly increased after treatment for BV. Despite similar amounts of uncoated and IgA coated majority taxa ( >1% total) across all visits, there was preferential IgA coating of minority taxa (0.2%-1% total) associated with BV including Sneathia, several Prevotella species, and others. At the time of BV, we identified a principal component (PC) driven by proinflammatory mediators that correlated positively with an uncoated BV-associated bacterial community and negatively with an IgA coated protective Lactobacillus bacterial community. The preferential coating of BV-associated species, increase in coating following metronidazole treatment, and positive correlation between uncoated BV-associated species and inflammation suggest that coating may represent a host mechanism designed to limit bacterial diversity and reduce inflammatory responses. Elucidating the role of Ig coating in vaginal mucosal immunity may promote new strategies to prevent recurrent BV.
Collapse
Affiliation(s)
- Kerry Murphy
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Matthew Gromisch
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sujatha Srinivasan
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Lianna Wood
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sean Proll
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Congzhou Liu
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tina Fiedler
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - D. J. Valint
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - David N. Fredricks
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Marla J. Keller
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Betsy C. Herold
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
26
|
Kumar N, Gangane N, Mohapatra I, Rukadikar C, Sharmila V, Pushpalatha K, Eerike M, Santhoshi G, Samantaray SR, Seth S, Trigunait P, Reddy NJ, Patel S, Rani S, Mishra R, Negi K. Effect of COVID-19 Vaccination on Menstrual Cycle Patterns of Reproductive-age Women: A Multi-centric Observational Study. Curr Drug Res Rev 2024; 16:237-248. [PMID: 37291775 DOI: 10.2174/2589977515666230608140606] [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: 04/07/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/10/2023]
Abstract
AIM The study was conducted to know the impact of COVID-19 vaccination on menstrual cycle patterns and pre- and post-menstrual symptoms in women aged 18-45 years. BACKGROUND COVID-19 vaccination was introduced to combat the dreadful impacts of human coronavirus infection. The two indigenously developed COVID-19 vaccines approved for use in India are COVISHIELD and COVAXIN. OBJECTIVES To investigate the effects of COVID-19 vaccination on the menstrual cycle, pre- and post-menstrual symptoms and to establish the correlation with the type of vaccine received. METHODS Multi-centric observational study conducted in six institutes of national importance in different states of India over one year. A total of 5709 female participants fulfilling inclusion criteria were enrolled. Data about the impact of vaccines (COVISHIELD and COVAXIN) and prior COVID-19 infection on the menstrual cycle and its associated symptoms were obtained using all participants' online and offline interviews. RESULTS Of 5709 participants, 78.2% received COVISHIELD and 21.8% COVAXIN. Of the total 5709 participants, 333 (5.8%) developed post-vaccination menstrual disturbances, with 32.7% having frequent cycles, 63.7% prolonged cycles, and 3.6% inter-menstrual bleeding. A total of 301 participants noticed changes in the amount of bleeding, with 50.2% excessive, 48.8% scanty, and 0.99% amenorrhea followed by heavy bleeding. Furthermore, the irregularities of the menstrual cycle (p = 0.011) and length (0.001) were significantly higher in the COVAXIN group (7.2%) as compared to the COVISHIELD (5.3%) group. A total of 721 participants complained of newly developed/worsening pre- and post-menstrual symptoms. These symptoms were significantly higher in the COVISHIELD group (p = 0.031), with generalized weakness and body pains as the main complaints (p = 0.001). No significant difference was observed in the incidence of COVID-19 infection with these vaccines. No significant associations were observed when comparing menstrual abnormalities among those with COVID-19 infection (p > 0.05). CONCLUSION COVISHIELD and COVAXIN vaccines were associated with menstrual cycle disturbances and pre-and post-menstrual symptoms in a small proportion of participants, with 94.7% having no change in the amount of bleeding during menstruation post-vaccination. The menstrual irregularities observed were significantly higher with the COVAXIN vaccine. Others: Further, long-term studies are required to confirm that the impact of COVID-19 vaccination on the menstrual cycle may be short-lasting, with no severe effects on women's menstrual health.
Collapse
Affiliation(s)
- Naina Kumar
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Neha Gangane
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Nagpur, Maharashtra, India
| | - Ipsita Mohapatra
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Kalyani, West Bengal, India
| | - Charushila Rukadikar
- Department of Physiology, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | - Vijayan Sharmila
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - K Pushpalatha
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Madhavi Eerike
- Department of Pharmacology, All India Institute of Medical Sciences, Hyderabad, Telangana, India
| | - G Santhoshi
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Subha Ranjan Samantaray
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Kalyani, West Bengal, India
| | - Shikha Seth
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | - Pragati Trigunait
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Nanditha Jangam Reddy
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Shweta Patel
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Sandhya Rani
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Roopanshi Mishra
- Department of Physiology, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | - Kamlesh Negi
- Department of Physiology, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| |
Collapse
|
27
|
Assiri AM, Alamaa T, Elenezi F, Alsagheir A, Alzubaidi L, TIeyjeh I, Alhomod AS, Gaffas EM, Amer SA. Unveiling the Clinical Spectrum of Post-COVID-19 Conditions: Assessment and Recommended Strategies. Cureus 2024; 16:e52827. [PMID: 38406111 PMCID: PMC10884364 DOI: 10.7759/cureus.52827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
SARS-CoV-2 caused the pandemic of the rapidly evolving COVID-19. As of December 6, 2023, there were 765,152,854 COVID-19-recovering cases. Long-term consequences known as "long COVID" and "post-COVID-19 conditions" (PCCs) or "post-acute COVID-19 syndrome" are being reported more frequently in a subset of recovering patients. Systemic, neuropsychiatric, cardio-respiratory, and gastrointestinal symptoms are the most prevalent. The management of PCCs poses unique challenges due to the lack of official guidelines and the complex nature of the illness. This abstract highlights key principles derived from recent reviews and expert recommendations to provide healthcare professionals with a comprehensive approach to manage post-COVID-19 patients. Preventive medicine plays a crucial role in managing PCCs. While no specific medications are available for treatment, preventive measures such as COVID-19 vaccination, adherence to precautionary measures, regular consultations with medical professionals, monitoring symptoms and progress, and seeking information on symptom management are essential to assist patients in their recovery and improve their quality of life. Medical management requires transparent goal-setting and collaborative decision-making based on the patient's symptoms, comorbidities, and treatment objectives. Treatment plans for post-COVID-19 patients should focus on patient education, using registries and calendars to track symptoms and triggers, providing support and reassurance, and offering holistic support through peer networks and supportive psychotherapy techniques. Symptomatic and rehabilitative care, including well-established symptom management techniques, physical rehabilitation programs, and addressing mental health and well-being, are vital components of post-COVID-19 management. Lifestyle factors such as stress reduction, nutrition, and sleep should be incorporated into managing underlying medical conditions in post-COVID-19 patients. Regular follow-up visits and referrals to specialists are recommended to monitor the patient's progress and address specific organ system involvement or additional care needs. In summary, for the effective management of PCCs, a holistic approach should include preventive measures, patient education, supportive psychotherapy, symptomatic and rehabilitative care, medical management, counseling on lifestyle elements, and appropriate follow-up plans. However, it is crucial to stay updated with evolving guidelines and recommendations from healthcare authorities to provide the most effective and evidence-based care to post-COVID-19 patients.
Collapse
Affiliation(s)
| | - Tareef Alamaa
- Therapeutic Services, Saudi Ministry of Health, Riyadh, SAU
| | - Faisal Elenezi
- Assistant Agency for Hospital Affairs, Saudi Ministry of Health, Riyadh, SAU
| | - Aeshah Alsagheir
- Health Quality Index Measuring, Saudi Ministry of Health, Riyadh, SAU
| | - Lamya Alzubaidi
- Assistant Agency for Hospital Affairs, Saudi Ministry of Health, Riyadh, SAU
| | - Imad TIeyjeh
- Infectious Diseases, Mayo Clinic, Rochester, USA
- Infectious Diseases, King Fahad Medical City, Riyadh, SAU
| | | | - Eisha M Gaffas
- Mental Health and Social Services, Saudi Ministry of Health, Riyadh, SAU
| | - Samar A Amer
- Public Health and Community Medicine, Zagazig University, Zagazig, EGY
- General Administration of Health Programs and Non-communicable Diseases, Saudi Ministry of Health, Riyadh, SAU
| |
Collapse
|
28
|
Dai M, Xu Y, Gong G, Zhang Y. Roles of immune microenvironment in the female reproductive maintenance and regulation: novel insights into the crosstalk of immune cells. Front Immunol 2023; 14:1109122. [PMID: 38223507 PMCID: PMC10786641 DOI: 10.3389/fimmu.2023.1109122] [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: 11/27/2022] [Accepted: 09/25/2023] [Indexed: 01/16/2024] Open
Abstract
Female fertility decline is an accumulative consequence caused by complex factors, among them, the disruption of the immune profile in female reproduction stands out as a crucial contributor. Presently, the effects of immune microenvironment (IME) on the female reproductive process have attracted increasing attentions for their dynamic but precisive roles. Immunocytes including macrophages, dendritic cells, T cells, B cells and neutrophils, with diverse subpopulations as well as high plasticity functioned dynamically in the process of female reproduction through indirect intercellular communication via specific cytokine release transduced by molecular signal networks or direct cell-cell contact to maintain the stability of the reproductive process have been unveiled. The immune profile of female reproduction in each stage has also been meticulously unveiled. Especially, the application of single-cell sequencing (scRNA-seq) technology in this process reveals the distribution map of immune cells, which gives a novel insight for the homeostasis of IME and provides a research direction for better exploring the role of immune cells in female reproduction. Here, we provide an all-encompassing overview of the latest advancements in immune modulation within the context of the female reproductive process. Our approach involves structuring our summary in accordance with the physiological sequence encompassing gonadogenesis, folliculogenesis within the ovaries, ovulation through the fallopian tubes, and the subsequent stages of embryo implantation and development within the uterus. Our overarching objective is to construct a comprehensive portrayal of the immune microenvironment (IME), thereby accentuating the pivotal role played by immune cells in governing the intricate female reproductive journey. Additionally, we emphasize the pressing need for heightened attention directed towards strategies that focus on immune interventions within the female reproductive process, with the ultimate aim of enhancing female fertility.
Collapse
Affiliation(s)
- Mengyuan Dai
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, China
| | - Ying Xu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, China
| | - Guidong Gong
- National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, China
| | - Yaoyao Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, China
| |
Collapse
|
29
|
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: 4] [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
|
30
|
Li S, Liu H, Li D, Chen F. Female reproductive health during the COVID-19 pandemic: latest evidence and understanding. Arch Gynecol Obstet 2023; 308:1691-1696. [PMID: 36856820 PMCID: PMC9976669 DOI: 10.1007/s00404-023-06976-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE The ongoing coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has imposed a heavy burden on medical systems. In addition to the respiratory system, the virus also causes injuries to other organs and systems such as the gastroenteric system, kidneys, and reproductive system. Female reproductive health requires more attention in this context. METHODS We have performed a thorough review of the relevant literature that addresses the impacts of SARS-CoV-2 infection and COVID-19 vaccination on the female reproductive system. RESULTS Most evidence shows that SARS-CoV-2 does not infect the female reproductive system. However, the virus may indirectly influence sex hormone concentrations through inflammation associated with cytokine storms and nervous system damage. Menstrual disorders in women infected with SARS-CoV-2 may be caused by down-regulation of angiotensin-converting enzyme 2, abnormal hormone levels, medications, and stress. There is no significant difference in ovarian follicle quality and in vitro fertilization parameters between the pre- and post-COVID-19 vaccination groups. In addition, most symptoms due to side effects of vaccination could recover within a short period of time. CONCLUSION SARS-CoV-2 infection affects female reproductive system function through multiple mechanisms. It is recommended that women of childbearing age be vaccinated with COVID-19 vaccine.
Collapse
Affiliation(s)
- Shuhui Li
- Department of Physiology, Jining Medical University, 133 Hehua Road, Jining, 272067, China
| | - Hongxin Liu
- Department of Physiology, Jining Medical University, 133 Hehua Road, Jining, 272067, China
| | - Di Li
- Department of Physiology, Jining Medical University, 133 Hehua Road, Jining, 272067, China
| | - Fei Chen
- Department of Physiology, Jining Medical University, 133 Hehua Road, Jining, 272067, China.
| |
Collapse
|
31
|
Pathare ADS, Loid M, Saare M, Gidlöf SB, Zamani Esteki M, Acharya G, Peters M, Salumets A. Endometrial receptivity in women of advanced age: an underrated factor in infertility. Hum Reprod Update 2023; 29:773-793. [PMID: 37468438 PMCID: PMC10628506 DOI: 10.1093/humupd/dmad019] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/24/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Modern lifestyle has led to an increase in the age at conception. Advanced age is one of the critical risk factors for female-related infertility. It is well known that maternal age positively correlates with the deterioration of oocyte quality and chromosomal abnormalities in oocytes and embryos. The effect of age on endometrial function may be an equally important factor influencing implantation rate, pregnancy rate, and overall female fertility. However, there are only a few published studies on this topic, suggesting that this area has been under-explored. Improving our knowledge of endometrial aging from the biological (cellular, molecular, histological) and clinical perspectives would broaden our understanding of the risks of age-related female infertility. OBJECTIVE AND RATIONALE The objective of this narrative review is to critically evaluate the existing literature on endometrial aging with a focus on synthesizing the evidence for the impact of endometrial aging on conception and pregnancy success. This would provide insights into existing gaps in the clinical application of research findings and promote the development of treatment options in this field. SEARCH METHODS The review was prepared using PubMed (Medline) until February 2023 with the keywords such as 'endometrial aging', 'receptivity', 'decidualization', 'hormone', 'senescence', 'cellular', 'molecular', 'methylation', 'biological age', 'epigenetic', 'oocyte recipient', 'oocyte donation', 'embryo transfer', and 'pregnancy rate'. Articles in a language other than English were excluded. OUTCOMES In the aging endometrium, alterations occur at the molecular, cellular, and histological levels suggesting that aging has a negative effect on endometrial biology and may impair endometrial receptivity. Additionally, advanced age influences cellular senescence, which plays an important role during the initial phase of implantation and is a major obstacle in the development of suitable senolytic agents for endometrial aging. Aging is also accountable for chronic conditions associated with inflammaging, which eventually can lead to increased pro-inflammation and tissue fibrosis. Furthermore, advanced age influences epigenetic regulation in the endometrium, thus altering the relation between its epigenetic and chronological age. The studies in oocyte donation cycles to determine the effect of age on endometrial receptivity with respect to the rates of implantation, clinical pregnancy, miscarriage, and live birth have revealed contradictory inferences indicating the need for future research on the mechanisms and corresponding causal effects of women's age on endometrial receptivity. WIDER IMPLICATIONS Increasing age can be accountable for female infertility and IVF failures. Based on the complied observations and synthesized conclusions in this review, advanced age has been shown to have a negative impact on endometrial functioning. This information can provide recommendations for future research focusing on molecular mechanisms of age-related cellular senescence, cellular composition, and transcriptomic changes in relation to endometrial aging. Additionally, further prospective research is needed to explore newly emerging therapeutic options, such as the senolytic agents that can target endometrial aging without affecting decidualization. Moreover, clinical trial protocols, focusing on oocyte donation cycles, would be beneficial in understanding the direct clinical implications of endometrial aging on pregnancy outcomes.
Collapse
Affiliation(s)
- Amruta D S Pathare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Marina Loid
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Sebastian Brusell Gidlöf
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Masoud Zamani Esteki
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Ganesh Acharya
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Medicine, Women’s Health and Perinatology Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Maire Peters
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Santos LC, Silva JF. Molecular Factors Involved in the Reproductive Morphophysiology of Female Domestic Cat ( Felis catus). Animals (Basel) 2023; 13:3153. [PMID: 37835759 PMCID: PMC10571923 DOI: 10.3390/ani13193153] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 10/15/2023] Open
Abstract
The domestic cat (Felis catus) is considered an important model for the study of feline reproductive morphophysiology. However, although the morphological changes and clinical signs that occur during the estrous cycle and pregnancy are well known, little is known about the molecular mechanisms involved in the reproductive physiology of this animal species. Thus, this paper reviews the current knowledge about the modulation and expression profile of hormonal, immunological, redox, and growth mediators involved in the uterine, ovarian, and placental morphophysiology of domestic cats.
Collapse
Affiliation(s)
| | - Juneo Freitas Silva
- Nucleo de Pesquisas em Reproducao e Endocrinologia, Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus 45662-900, Brazil;
| |
Collapse
|
33
|
Muyayalo KP, Gong GS, Kiyonga Aimeé K, Liao AH. Impaired immune response against SARS-CoV-2 infection is the major factor indirectly altering reproductive function in COVID-19 patients: a narrative review. HUM FERTIL 2023; 26:778-796. [PMID: 37811836 DOI: 10.1080/14647273.2023.2262757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/25/2023] [Indexed: 10/10/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease affecting multiple systems and organs, including the reproductive system. SARS-CoV-2, the virus that causes COVID-19, can damage reproductive organs through direct (angiotensin converting enzyme-2, ACE-2) and indirect mechanisms. The immune system plays an essential role in the homeostasis and function of the male and female reproductive systems. Therefore, an altered immune response related to infectious and inflammatory diseases can affect reproductive function and fertility in both males and females. This narrative review discussed the dysregulation of innate and adaptive systems induced by SARS-CoV-2 infection. We reviewed the evidence showing that this altered immune response in COVID-19 patients is the major indirect mechanism leading to adverse reproduction outcomes in these patients. We summarized studies reporting the long-term effect of SARS-CoV-2 infection on women's reproductive function and proposed the chronic inflammation and chronic autoimmunity characterizing long COVID as potential underlying mechanisms. Further studies are needed to clarify the role of autoimmunity and chronic inflammation (long COVID) in altered female reproduction function in COVID-19.
Collapse
Affiliation(s)
- Kahindo P Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Department of Obstetrics and Gynecology, University of Kinshasa, Kinshasa, D. R. Congo
| | - Guang-Shun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Kahindo Kiyonga Aimeé
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, People's Republic of China
- Department of Tropical Medicine Infectious and Parasitic Diseases, University of Kinshasa, Kinshasa, D. R. Congo
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| |
Collapse
|
34
|
Wang J, Li W, Li N, Wang B. Immunization with Multiple Virulence Factors Provides Maternal and Neonatal Protection against Group B Streptococcus Serotypes. Vaccines (Basel) 2023; 11:1459. [PMID: 37766135 PMCID: PMC10535937 DOI: 10.3390/vaccines11091459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Group B streptococcus (GBS) commonly colonizes the vaginal tract and is a leading cause of life-threatening neonatal infections and adverse pregnancy outcomes. No effective vaccine is clinically available. Conserved bacterial virulence factors, including those of GBS, have been employed as vaccine components. We investigated serotype-independent protection against GBS by intranasal immunization with six conserved GBS virulence factors (GBSV6). GBSV6 induced systemic and vaginal antibodies and T cell responses in mice. The immunity reduced mouse mortality and vaginal colonization by various GBS serotypes and protected newborn mice of immunized dams against GBS challenge. Intranasal GBSV6 immunization also provided long-lasting protective immunity and had advantages over intramuscular GBSV6 immunization regarding restricting vaginal GBS colonization. Our findings indicate that intranasal immunization targeting multiple conserved GBS virulence factors induces serotype-independent immunity, which protects against GBS infection systemically and vaginally in dams and prevents newborn death. The study presents valuable strategies for GBS vaccine development.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Varnotech Biopharm Ltd., Beijing 100176, China
| | - Wenbo Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Varnotech Biopharm Ltd., Beijing 100176, China
| | - Ning Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Beinan Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
35
|
Lingasamy P, Modhukur V, Mändar R, Salumets A. Exploring Immunome and Microbiome Interplay in Reproductive Health: Current Knowledge, Challenges, and Novel Diagnostic Tools. Semin Reprod Med 2023; 41:172-189. [PMID: 38262441 PMCID: PMC10846929 DOI: 10.1055/s-0043-1778017] [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] [Indexed: 01/25/2024]
Abstract
The dynamic interplay between the immunome and microbiome in reproductive health is a complex and rapidly advancing research field, holding tremendously vast possibilities for the development of reproductive medicine. This immunome-microbiome relationship influences the innate and adaptive immune responses, thereby affecting the onset and progression of reproductive disorders. However, the mechanisms governing these interactions remain elusive and require innovative approaches to gather more understanding. This comprehensive review examines the current knowledge on reproductive microbiomes across various parts of female reproductive tract, with special consideration of bidirectional interactions between microbiomes and the immune system. Additionally, it explores innate and adaptive immunity, focusing on immunoglobulin (Ig) A and IgM antibodies, their regulation, self-antigen tolerance mechanisms, and their roles in immune homeostasis. This review also highlights ongoing technological innovations in microbiota research, emphasizing the need for standardized detection and analysis methods. For instance, we evaluate the clinical utility of innovative technologies such as Phage ImmunoPrecipitation Sequencing (PhIP-Seq) and Microbial Flow Cytometry coupled to Next-Generation Sequencing (mFLOW-Seq). Despite ongoing advancements, we emphasize the need for further exploration in this field, as a deeper understanding of immunome-microbiome interactions holds promise for innovative diagnostic and therapeutic strategies for reproductive health, like infertility treatment and management of pregnancy.
Collapse
Affiliation(s)
| | - Vijayachitra Modhukur
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Reet Mändar
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
36
|
Kaya YA, de Zoete MR, Steba GS. Advanced Technologies for Studying Microbiome-Female Reproductive Tract Interactions: Organoids, Organoids-on-a-Chip, and Beyond. Semin Reprod Med 2023; 41:160-171. [PMID: 38262440 PMCID: PMC11444813 DOI: 10.1055/s-0043-1778067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
The female reproductive tract (FRT) is home to diverse microbial communities that play a pivotal role in reproductive health and disorders such as infertility, endometriosis, and cervical cancer. To understand the complex host-microbiota interactions within the FRT, models that authentically replicate the FRT's environment, including the interplay between the microbiota, mucus layer, immune system, and hormonal cycle, are key. Recent strides in organoid and microfluidic technologies are propelling research in this domain, offering insights into FRT-microbiota interactions and potential therapeutic avenues. This review delves into the current state of FRT organoid models and microbe integration techniques, evaluating their merits and challenges for specific research objectives. Emphasis is placed on innovative approaches and applications, including integrating organoids with microfluidics, and using patient-derived biobanks, as this offers potential for deeper mechanistic insights and personalized therapeutic strategies. Modeling various FRT properties in organoids is explored, from encompassing age-related epithelial features, oxygen levels, and hormonal effects to mucus layers, immune responses, and microbial interactions, highlighting their potential to transform reproductive health research and predict possible outcomes.
Collapse
Affiliation(s)
| | - Marcel R de Zoete
- Department of Medical Microbiology, University Medical Centre, Utrecht, The Netherlands
| | - Gaby S Steba
- Division of Female and Baby, Department of Reproductive Medicine and Gynaecology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
37
|
Dabee S, Balle C, Onono M, Innes S, Nair G, Palanee-Phillips T, Burgener AD, Bosinger SE, Passmore JAS, Heffron R, Jaspan H, Happel AU. Update on the Impact of Depot Medroxyprogesterone Acetate on Vaginal Mucosal Endpoints and Relevance to Sexually Transmitted Infections. Curr HIV/AIDS Rep 2023; 20:251-260. [PMID: 37341916 PMCID: PMC10403392 DOI: 10.1007/s11904-023-00662-0] [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] [Accepted: 05/29/2023] [Indexed: 06/22/2023]
Abstract
PURPOSE OF REVIEW The long-acting reversible intramuscularly-injected contraceptive depot medroxyprogesterone acetate (DMPA-IM) is widely used by cisgender women in Africa. Although DMPA-IM provides reliable contraception, potential effects on the female genital tract (FGT) mucosa have raised concern, including risk of HIV infection. This review summarises and compares evidence from observational cohort studies and the randomised Evidence for Contraceptive Options in HIV Outcomes (ECHO) Trial. RECENT FINDINGS Although previous observational studies found women using DMPA-IM had higher abundance of bacterial vaginosis (BV)-associated bacteria, increased inflammation, increased cervicovaginal HIV target cell density, and epithelial barrier damage, sub-studies of the ECHO Trial found no adverse changes in vaginal microbiome, inflammation, proteome, transcriptome, and risk of viral and bacterial STIs, other than an increase in Th17-like cells. Randomised data suggest that DMPA-IM use does not adversely change mucosal endpoints associated with acquisition of infections. These findings support the safe use of DMPA-IM in women at high risk of acquiring STIs, including HIV.
Collapse
Affiliation(s)
- Smritee Dabee
- Center for Global Infectious Disease, Seattle Children’s Research Institute, 307 Westlake Ave. N, Seattle, WA 98109 USA
| | - Christina Balle
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925 South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925 South Africa
| | | | - Steve Innes
- Desmond Tutu Health Foundation, 3 Woodlands Rd, Woodstock, Cape Town, 7915 South Africa
| | - Gonasagrie Nair
- Desmond Tutu Health Foundation, 3 Woodlands Rd, Woodstock, Cape Town, 7915 South Africa
| | - Thesla Palanee-Phillips
- Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Klein St & Esselen St, Hillbrow, Johannesburg 2001 South Africa
| | - Adam D. Burgener
- Center for Global Health and Diseases, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Obstetrics and Gynecology, University of Manitoba, 66 Chancellors Cir, Winnipeg, MB R3T 2N2 Canada
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Visionsgatan 18, L8, 171 76 Solna, Sweden
| | - Steven E. Bosinger
- ENPRC Genomics Core Laboratory, Emory National Primate Research Center, 954 Gatewood Rd NE, Atlanta, GA 30329 USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA 30307 USA
- Emory Vaccine Center, Emory University, 7 1st Ave, Atlanta, GA 30317 USA
| | - Jo-Ann S. Passmore
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925 South Africa
- National Health Laboratory Service, Anzio Road, Observatory, Cape Town, 7925 South Africa
| | - Renee Heffron
- Department of Medicine, University of Alabama at Birmingham, 845 19th Street South, AL 35294-2170 Birmingham, USA
- Department of Global Health, University of Washington, 1510 San Juan Road NE, Seattle, WA 98195 USA
| | - Heather Jaspan
- Center for Global Infectious Disease, Seattle Children’s Research Institute, 307 Westlake Ave. N, Seattle, WA 98109 USA
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925 South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925 South Africa
- Department of Pediatrics, University of Washington, 1959 NE Pacific St., Seattle, WA 98195 USA
- Department of Global Health, University of Washington, 1510 San Juan Road NE, Seattle, WA 98195 USA
| | - Anna-Ursula Happel
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925 South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925 South Africa
| |
Collapse
|
38
|
Rahimi Mansour F, Keyvanfar A, Najafiarab H, Rajaei Firouzabadi S, Sefidgar S, Hooshmand Chayijan S, Tarom M, Fadaei M, Farzaneh F, Karimzadeh Bardeei L, Tehrani S. Menstrual disturbances following COVID-19 vaccination: A probable puzzle about the role of endocrine and immune pathways. J Reprod Immunol 2023; 158:103952. [PMID: 37201456 PMCID: PMC10174728 DOI: 10.1016/j.jri.2023.103952] [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: 11/18/2022] [Revised: 04/15/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
Menstruation is a monthly shedding of the uterine wall, presented by menstrual bleeding in women of reproductive age. Menstruation is regulated by fluctuation of estrogen and progesterone, as well as other endocrine and immune pathways. Many women experienced menstrual disturbances after vaccination against the novel coronavirus in the last two years. Vaccine-induced menstrual disturbances have led to discomfort and concern among reproductive-age women, such that some decided not to receive the subsequent doses of the vaccine. Although many vaccinated women report these menstrual disturbances, the mechanism is still poorly understood. This review article discusses the endocrine and immune changes following COVID-19 vaccination and the possible mechanisms of vaccine-related menstrual disturbances.
Collapse
Affiliation(s)
- Farima Rahimi Mansour
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Amirreza Keyvanfar
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hanieh Najafiarab
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sahar Sefidgar
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammadreza Tarom
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahta Fadaei
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farah Farzaneh
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shabnam Tehrani
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Kumar V, Bauer C, Stewart JH. TIME Is Ticking for Cervical Cancer. BIOLOGY 2023; 12:941. [PMID: 37508372 PMCID: PMC10376148 DOI: 10.3390/biology12070941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023]
Abstract
Cervical cancer (CC) is a major health problem among reproductive-age females and comprises a leading cause of cancer-related deaths. Human papillomavirus (HPV) is the major risk factor associated with CC incidence. However, lifestyle is also a critical factor in CC pathogenesis. Despite HPV vaccination introduction, the incidence of CC is increasing worldwide. Therefore, it becomes critical to understand the CC tumor immune microenvironment (TIME) to develop immune cell-based vaccination and immunotherapeutic approaches. The current article discusses the immune environment in the normal cervix of adult females and its role in HPV infection. The subsequent sections discuss the alteration of different immune cells comprising CC TIME and their targeting as future therapeutic approaches.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| | - Caitlin Bauer
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| | - John H Stewart
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
- Louisiana Children's Medical Center Cancer Center, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| |
Collapse
|
40
|
Wesselink AK, Lovett SM, Weinberg J, Geller RJ, Wang TR, Regan AK, Willis MD, Perkins RB, Yland JJ, Koenig MR, Rothman KJ, Hatch EE, Wise LA. COVID-19 vaccination and menstrual cycle characteristics: A prospective cohort study. Vaccine 2023; 41:4327-4334. [PMID: 37301706 PMCID: PMC10239900 DOI: 10.1016/j.vaccine.2023.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
We prospectively examined the association between COVID-19 vaccination and menstrual cycle characteristics in an internet-based prospective cohort study. We included a sample of 1,137 participants who enrolled in Pregnancy Study Online (PRESTO), a preconception cohort study of couples trying to conceive, during January 2021-August 2022. Eligible participants were aged 21-45 years, United States or Canadian residents, and trying to conceive without fertility treatment. At baseline and every 8 weeks for up to 12 months, participants completed questionnaires on which they provided information on COVID-19 vaccination and menstrual cycle characteristics, including cycle regularity, cycle length, bleed length, heaviness of bleed, and menstrual pain. We fit generalized estimating equation (GEE) models with a log link function and Poisson distribution to estimate the adjusted risk ratio (RR) for irregular cycles associated with COVID-19 vaccination. We used linear regression with GEE to estimate adjusted mean differences in menstrual cycle length associated with COVID-19 vaccination. We adjusted for sociodemographic, lifestyle, medical and reproductive factors. Participants had 1.1 day longer menstrual cycles after receiving the first dose of COVID-19 vaccine (95 % CI: 0.4, 1.9) and 1.3 day longer cycles after receiving the second dose (95 % CI: 0.2, 2.5). Associations were attenuated at the second cycle post-vaccination. We did not observe strong associations between COVID-19 vaccination and cycle regularity, bleed length, heaviness of bleed, or menstrual pain. In conclusion, COVID-19 vaccination was associated with a ∼1 day temporary increase in menstrual cycle length, but was not appreciably associated with other menstrual cycle characteristics.
Collapse
Affiliation(s)
- Amelia K Wesselink
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States.
| | - Sharonda M Lovett
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Janice Weinberg
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Ruth J Geller
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Tanran R Wang
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | | | - Mary D Willis
- Department of Obstetrics & Gynecology, Boston University Chobanian and Avedesian School of Medicine, Boston, MA, United States
| | - Rebecca B Perkins
- Department of Obstetrics & Gynecology, Boston University Chobanian and Avedesian School of Medicine, Boston, MA, United States
| | - Jennifer J Yland
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Martha R Koenig
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Kenneth J Rothman
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Elizabeth E Hatch
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Lauren A Wise
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| |
Collapse
|
41
|
Rastegar T, Feryduni L, Fakhraei M. COVID-19 vaccine side effects on menstrual disturbances among Iranian women. New Microbes New Infect 2023; 53:101114. [PMID: 37065964 PMCID: PMC10085868 DOI: 10.1016/j.nmni.2023.101114] [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: 04/25/2022] [Revised: 03/15/2023] [Accepted: 03/23/2023] [Indexed: 04/18/2023] Open
Abstract
Background Many studies reported of menstrual disturbances as possible side effects of COVID-19 vaccination. Our objective was to evaluate the association between vaccination and the occurrence of menstrual disturbances among Iranian women. Methods We used to google form questionnaires to collect reports of menstrual disturbances from 455 women aged 15-55 years in Iran. We estimated the relative risk of menstrual disturbances according to vaccination in a self-controlled case-series design after vaccination. We examined the occurrence of such disorders after the first, second and third doses of vaccine. Results Findings The prevalence of menstrual disturbance was more in latency and heavy bleeding than others disorder after vaccination, although ֮ %50 of women doesn't have any disturbance. We observed increased risks after vaccination also for other menstrual disturbances, in menopausal women too (>10%). Discussion Menstrual disturbances were generally common regardless of vaccination. We found a significant increase in menstrual disturbances after vaccination, particularly for latency and heavier bleeding than usual, longer duration and for short interval between menstruations. Mechanisms underlying these findings may involve bleeding disturbances in general, as well as endocrine alterations of immune system stimulation and relation to hormonal secretion.
Collapse
Affiliation(s)
- Tayebeh Rastegar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Burns and Regenerative Medicine Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Lila Feryduni
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Fakhraei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
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: 19] [Impact Index Per Article: 9.5] [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
|
43
|
Human Leukocyte Antigen Alleles Compatibility and Immunophenotypic Profile Associations in Infertile Couples. Cureus 2023; 15:e36584. [PMID: 36968684 PMCID: PMC10035384 DOI: 10.7759/cureus.36584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2023] [Indexed: 03/25/2023] Open
Abstract
Introduction: The maternal immune system has a major role in the successful embryo implantation and maintenance of the pregnancy. This study aimed to investigate the maternal immunophenotyping profile (percentage of Natural Killer [NK] cells and the CD4/CD8 [cluster designation] ratio in peripheral blood lymphocytes) and the HLA (Human Leukocyte Antigen)-DQA1 alleles sharing in infertile couples. Methods: This cross-sectional study included 78 women who had experienced at least two spontaneous miscarriages and 110 women with a history of recurrent implantation failures after in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) and embryo transfer (ET) (IVF-ET failures). The NK cell percentage and the CD4/CD8 ratio were determined by flow cytometry. Genotyping of the HLA-DQA1 alleles was carried out for all women and their partners, and couple HLA-DQA1 compatibility was expressed as the percentage of common HLA-DQA1 alleles (totaling 35 alleles) shared between spouses to the sum of the unique alleles observed. Results: In women with recurrent miscarriages, high values (%) of the NK population with a median (interquartile range [IQR]) of 10.3% (7.7% to 12.5%) and CD4/CD8 ratio (1.7) (1.5 to 2.1) were found. In women with IVF-ET failures, the (%) NK population (10.5%) (8.6% to 12.5%) and CD4/CD8 ratio (1.8) (1.5 to 2.1) were similarly increased (p=0.390, and p=0.490, respectively). The proportion of women with >10% NK cells was 53.8% and 58.2% in women with miscarriages and IVF-ET failures, respectively (p=0.554). The prevalence of HLA-DQA1*5 allele carriage was elevated in women with miscarriages as well as those with IVF-ET failures (52.6% and 61.8%, respectively; p=0.206). The proportion of couples with high (>50%) HLA-DQA1 sharing was 65.4% in the group with miscarriages and 73.6% in the group with IVF-ET failures, respectively (p=0.222). The CD4/CD8 ratio was statistically significantly positively correlated with the (%) NK population in women with IVF-ET failures (rho = 0.297, p=0.002) and with the (%) HLA-DQA1 sharing in the group with miscarriages (rho = 0.266, p=0.019). The couples in which both spouses were carriers of the HLA-DQA1*5 allele had an increased probability of high (>50%) HLA-DQA1 compatibility compared with the couples in which neither of the spouses carried the allele in the miscarriage group (OR = 24.3, 95% CI: 3.0 to 198.9, p<0.001), and the IVF-ET failure group (OR = 10.5, 95% CI: 2.2 to 49.8, p<0.001). Conclusion: The peripheral NK (%) population and CD4/CD8 ratio, as well as the prevalence of the HLA-DQA1*5 allele, were elevated in women with recurrent miscarriages and IVF-ET failures. Furthermore, these couples with negative reproductive outcomes had a high percentage of HLA-DQA1 allele similarity. The presence of the HLA-DQA1*5 allele in spouses was strongly associated with overall couple HLA-DQA1 compatibility, implying that it could be used as a surrogate marker for assessing overall immunological compatibility in infertile couples.
Collapse
|
44
|
Control of human cytomegalovirus replication by liver resident natural killer cells. Nat Commun 2023; 14:1409. [PMID: 36918610 PMCID: PMC10014884 DOI: 10.1038/s41467-023-37181-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 03/06/2023] [Indexed: 03/15/2023] Open
Abstract
Natural killer cells are considered to be important for control of human cytomegalovirus- a major pathogen in immune suppressed transplant patients. Viral infection promotes the development of an adaptive phenotype in circulating natural killer cells that changes their anti-viral function. In contrast, less is understood how natural killer cells that reside in tissue respond to viral infection. Here we show natural killer cells resident in the liver have an altered phenotype in cytomegalovirus infected individuals and display increased anti-viral activity against multiple viruses in vitro and identify and characterise a subset of natural killer cells responsible for control. Crucially, livers containing natural killer cells with better capacity to control cytomegalovirus replication in vitro are less likely to experience viraemia post-transplant. Taken together, these data suggest that virally induced expansion of tissue resident natural killer cells in the donor organ can reduce the chance of viraemia post-transplant.
Collapse
|
45
|
Al Kadri HM, Al Sudairy AA, Alangari AS, Al Khateeb BF, El-Metwally AA. COVID-19 vaccination and menstrual disorders among women: Findings from a meta-analysis study. J Infect Public Health 2023; 16:697-704. [PMID: 36934644 PMCID: PMC9979695 DOI: 10.1016/j.jiph.2023.02.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/31/2022] [Accepted: 02/19/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND COVID - 19 vaccine can lead to various local and systemic side effects, including menstrual irregularities in women. There is no robust quantitative evidence of the association between the COVID - 19 vaccine and menstrual irregularities. A meta-analysis was performed to estimate the pooled prevalence of a range of menstrual disorders that may occur in women following COVID - 19 vaccination. METHODS After searching for epidemiological studies, we systematically performed a meta-analysis on PubMed/Medline, EMBASE, and Science Direct. Sixteen studies were finally included in the study. We estimated the pooled prevalence and corresponding 95 % confidence intervals (CIs) for a group of menstrual disorders, including menorrhagia, polymenorrhea, abnormal cycle length, and oligomenorrhea. Heterogeneity was assessed using the I2 statistic and the Q test. RESULTS Overall, the pooled prevalence of menorrhagia was 24.24 % (pooled prevalence 24.24 %; 95 % CI: 12.8-35.6 %). The pooled prevalence of polymenorrhea was 16.2 % (pooled prevalence: 16.2 %; 95 % CI: 10.7-21.6 %). The pooled prevalence of abnormal cycle length was relatively lower than that of the other disorders (pooled prevalence: 6.6 %; 95 % CI: 5.0-8.2 %). The pooled prevalence of oligomenorrhea was 22.7 % (95 % CI: 13.5-32.0 %). CONCLUSION The findings indicate that menorrhagia, oligomenorrhea, and polymenorrhea were the most common menstrual irregularities after vaccination. The findings also suggest that a relatively high proportion of women suffer from menstrual irregularities. Further longitudinal studies are needed to confirm the causal relationship between COVID-19 vaccination and menstrual irregularities.
Collapse
Affiliation(s)
- Hanan M Al Kadri
- Department of Obstetrics and Gynecology, King Abdulaziz Medical City, Ministry of the National Guard-Health affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | - Atika A Al Sudairy
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abdulaziz S Alangari
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Badr F Al Khateeb
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; Department of Family Medicine, King Abdulaziz Medical City, Ministry of the National Guard-Health affairs, Riyadh, Saudi Arabia
| | - Ashraf A El-Metwally
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
46
|
Korchagina AA, Koroleva E, Tumanov AV. Innate Lymphoid Cell Plasticity in Mucosal Infections. Microorganisms 2023; 11:461. [PMID: 36838426 PMCID: PMC9967737 DOI: 10.3390/microorganisms11020461] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Mucosal tissue homeostasis is a dynamic process that involves multiple mechanisms including regulation of innate lymphoid cells (ILCs). ILCs are mostly tissue-resident cells which are critical for tissue homeostasis and immune response against pathogens. ILCs can sense environmental changes and rapidly respond by producing effector cytokines to limit pathogen spread and initiate tissue recovery. However, dysregulation of ILCs can also lead to immunopathology. Accumulating evidence suggests that ILCs are dynamic population that can change their phenotype and functions under rapidly changing tissue microenvironment. However, the significance of ILC plasticity in response to pathogens remains poorly understood. Therefore, in this review, we discuss recent advances in understanding the mechanisms regulating ILC plasticity in response to intestinal, respiratory and genital tract pathogens. Key transcription factors and lineage-guiding cytokines regulate this plasticity. Additionally, we discuss the emerging data on the role of tissue microenvironment, gut microbiota, and hypoxia in ILC plasticity in response to mucosal pathogens. The identification of new pathways and molecular mechanisms that control functions and plasticity of ILCs could uncover more specific and effective therapeutic targets for infectious and autoimmune diseases where ILCs become dysregulated.
Collapse
Affiliation(s)
| | | | - Alexei V. Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA
| |
Collapse
|
47
|
Gilan A, Laster-Haim S, Rottenstreich A, Porat S, Lessans N, Saar TD, Dior UP. The effect of SARS-CoV-2 BNT162b2 vaccine on the symptoms of women with endometriosis. Arch Gynecol Obstet 2023; 307:121-127. [PMID: 36098831 PMCID: PMC9468530 DOI: 10.1007/s00404-022-06765-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/22/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE As the use of the messenger RNA (mRNA) BNT162b2 (Pfizer-BioNTech) Coronavirus disease 2019 vaccine has grown, reports on menstrual changes have arisen. We aimed to examine menstrual bleeding patterns and endometriosis-associated symptoms after receiving the mRNA BNT162b2 SARS-CoV-2 vaccine in women with endometriosis, as compared to the control group. METHODS This is a questionnaire-based cross-sectional study including a total of 174 women. The study group included 86 women with a confirmed diagnosis of endometriosis and the control group included 88 women with no diagnosis or suspected diagnosis of endometriosis. Each woman completed a questionnaire on menstrual bleeding patterns and endometriosis-associated symptoms before and after receiving two doses of the BNT162b2 vaccine. Primary outcomes were changes in amount or length of menstrual bleeding, rates of intermenstrual bleeding and worsening in dysmenorrhea in the endometriosis patient group, as compared to the control group. Secondary outcomes included changes in all endometriosis-associated symptoms. RESULTS In our cohort, women with endometriosis were more likely to experience changes in bleeding patterns (women with endometriosis: 39.5%, control group: 31.0%, p = 0.02), and a significant worsening in endometriosis-associated symptoms with an almost 4.3-fold worsening in dysmenorrhea [95% CI 1.9-9.9, p < 0.01] and 5.5-fold odds for any worsening in symptoms in endometriosis patients, as compared to the control group [95% CI 2.7-11.1, p < 0.01]. CONCLUSION In our cohort, endometriosis was shown to be a significant risk factor for worsening of menstrual symptoms, after receiving the SARS-CoV-2 BNT162b2 mRNA vaccine. Further research is needed to confirm these findings.
Collapse
Affiliation(s)
- Adi Gilan
- Endometriosis Centre, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel.
| | - Sarit Laster-Haim
- Endometriosis Centre, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Amihai Rottenstreich
- Endometriosis Centre, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Shay Porat
- Endometriosis Centre, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Naama Lessans
- Endometriosis Centre, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Tal D Saar
- Endometriosis Centre, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Uri P Dior
- Endometriosis Centre, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| |
Collapse
|
48
|
Menzies FM. Immunology of Pregnancy and Systemic Consequences. Curr Top Microbiol Immunol 2023; 441:253-280. [PMID: 37695432 DOI: 10.1007/978-3-031-35139-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Pregnancy is an immunological paradox, with renowned Nobel Prize winning transplantation biologist Sir Peter Brian Medawar being the first to introduce this concept back in 1953. This concept considers how the maternal immune system can tolerate the developing fetus, which is 50% antigenically foreign to the uterus. There have been significant advances in our understanding of the immune system in regulating fertility, pregnancy and in complications of these, and what was once considered a paradox can be seen as a highly evolved system. Indeed, the complexity of the maternal-fetal interface along with our ever-advancing knowledge of immune cells and mediators means that we have a better understanding of these interactions, with gaps still present. This chapter will summarise the key aspects of the role of the immune system at each stage of pregnancy and highlight the recent advances in our knowledge.
Collapse
Affiliation(s)
- Fiona M Menzies
- School of Health and Life Sciences, University of the West of Scotland, Lanarkshire, UK.
| |
Collapse
|
49
|
Qashqari FSI, Dahlawi M, Assaggaf HM, Alsafi R, Gari A, Abudawood A, Al-Doboke A, Alsulami S, Bukhari R, Majeed SA, Salih EA, Alfelali M, Makhdoom H, Jalal NA. Effect of the COVID-19 Vaccine on the Menstrual Cycle among Females in Saudi Arabia. Ethiop J Health Sci 2022; 32:1083-1092. [PMID: 36475264 PMCID: PMC9692149 DOI: 10.4314/ejhs.v32i6.4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/28/2022] [Indexed: 12/13/2022] Open
Abstract
Background The number of reports of menstrual changes after COVID-19 vaccination in the Saudi population is still unknown. Therefore, this study aimed to assess the effect of the COVID-19 vaccine(Pfizer, AstraZeneca, and Moderna) on the menstrual cycle among females in Saudi Arabia. Methods This descriptive cross-sectional study was conducted in Saudi Arabia at Umm Al-Qura University (UQU) from August 2021 to February 2022. Data was collected through a previously validated online questionnaire. Results A total of 2338 participants who received the first dose of the COVID-19 vaccine participated in this study; 1606 (68.7%) of them received the second dose in addition to the first. The mean age of the study participants was 35.4±9.5 years. No significant associations were found between the type of COVID-19 vaccine and the impact on the menstrual cycle, either for the first or second dose (P-values > 0.05). A significant association was found only between the first dose vaccination day and the impact on the menstrual cycle in the second question of "After receiving the COVID-19 vaccine, your next period was" (P-value ≤ 0.05). Significant associations were found between the second dose vaccination day and the impact on the menstrual cycle in the first and second questions of "After receiving the COVID-19 vaccine, your next period was", and "After receiving the first dose, your next period was," respectively (P-values ≤ 0.05). Conclusion The study found a potential association between the COVID-19 vaccine and menstrual cycle irregularities, which could impact females' quality of life.
Collapse
Affiliation(s)
- Fadi S I Qashqari
- Department of Microbiology, College of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Maryam Dahlawi
- College of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Hamza M Assaggaf
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Radi Alsafi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Abdulrahim Gari
- Department of Obstetrics and Gynecology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Abdulrahman Abudawood
- Department of Family and Community Medicine, Rabigh Medical College, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amal Al-Doboke
- College of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Seham Alsulami
- College of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Rahaf Bukhari
- College of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | | | - Elaf Abdullah Salih
- Department of Obstetrics and Gynecology, International Medical Center, Jeddah, Saudi Arabi
| | - Mohammad Alfelali
- Department of Family and Community Medicine, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hatim Makhdoom
- Applied Medical Sciences College, Laboratory Technology Department, Taibah University, Almadinah Almunwarah, Jeddah, Saudi Arabia
| | - Naif A Jalal
- Department of Microbiology, College of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| |
Collapse
|
50
|
Wang S, Mortazavi J, Hart JE, Hankins JA, Katuska LM, Farland LV, Gaskins AJ, Wang YX, Tamimi RM, Terry KL, Rich-Edwards JW, Missmer SA, Chavarro JE. A prospective study of the association between SARS-CoV-2 infection and COVID-19 vaccination with changes in usual menstrual cycle characteristics. Am J Obstet Gynecol 2022; 227:739.e1-739.e11. [PMID: 35841938 PMCID: PMC9277995 DOI: 10.1016/j.ajog.2022.07.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/23/2022] [Accepted: 07/06/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Despite anecdotal reports, the impacts of SARS-CoV-2 infection or COVID-19 vaccination on menstrual health have not been systemically investigated. OBJECTIVE This study aimed to examine the associations of SARS-CoV-2 infection and COVID-19 vaccination with menstrual cycle characteristics. STUDY DESIGN This study prospectively observed 3858 premenopausal women in the Nurses' Health Study 3 living in the United States or Canada who received biannual follow-up questionnaires between January 2011 and December 2021 and completed additional monthly and quarterly surveys related to the COVID-19 pandemic between April 2020 and November 2021. History of positive SARS-CoV-2 test, COVID-19 vaccination status, and vaccine type were self-reported in surveys conducted in 2020 and 2021. Current menstrual cycle length and regularity "before COVID-19" were reported at baseline between 2011 and 2016, and current menstrual cycle length and regularity "after COVID-19" were reported in late 2021. Pre- to post-COVID change in menstrual cycle length and regularity was calculated between reports. Logistic or multinomial logistic regression models were used to assess the associations between SARS-CoV-2 infection and COVID-19 vaccination and change in menstrual cycle characteristics. RESULTS The median age at baseline and the median age at end of follow-up were 33 years (range, 21-51) and 42 years (range, 27-56), respectively, with a median follow-up time of 9.2 years. This study documented 421 SARS-CoV-2 infections (10.9%) and 3527 vaccinations (91.4%) during follow-up. Vaccinated women had a higher risk of increased cycle length than unvaccinated women (odds ratio, 1.48; 95% confidence interval, 1.00-2.19), after adjusting for sociodemographic and behavioral factors. These associations were similar after in addition accounting for pandemic-related stress. COVID-19 vaccination was only associated with change to longer cycles in the first 6 months after vaccination (0-6 months: odds ratio, 1.67 [95% confidence interval, 1.05-2.64]; 7-9 months: odds ratio, 1.43 [95% confidence interval, 0.96-2.14]; >9 months: odds ratio, 1.41 [95% confidence interval, 0.91-2.18]) and among women whose cycles were short, long, or irregular before vaccination (odds ratio, 2.82 [95% confidence interval, 1.51-5.27]; odds ratio, 1.10 [95% confidence interval, 0.68-1.77] for women with normal length, regular cycles before vaccination). Messenger RNA and adenovirus-vectored vaccines were both associated with this change. SARS-CoV-2 infection was not associated with changes in usual menstrual cycle characteristics. CONCLUSION COVID-19 vaccination may be associated with short-term changes in usual menstrual cycle length, particularly among women whose cycles were short, long, or irregular before vaccination. The results underscored the importance of monitoring menstrual health in vaccine clinical trials. Future work should examine the potential biological mechanisms.
Collapse
Affiliation(s)
- Siwen Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA; Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jasmine Mortazavi
- Department of Obstetrics, Gynecology, and Reproductive Biologym, Michigan State University College of Human Medicine, Grand Rapids, MI
| | - Jaime E Hart
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jennifer A Hankins
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Laura M Katuska
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Leslie V Farland
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, The University of Arizona, Tucson, AZ
| | - Audrey J Gaskins
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Yi-Xin Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Rulla M Tamimi
- Population Health Sciences, Weill Cornell Medicine, New York, NY
| | - Kathryn L Terry
- Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Janet W Rich-Edwards
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Stacey A Missmer
- Department of Obstetrics, Gynecology, and Reproductive Biologym, Michigan State University College of Human Medicine, Grand Rapids, MI; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.
| | - Jorge E Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|