1
|
Hosseinpour F, Zeinolabedini Hezave A, Talaei-Khozani T, Rastgou-Maeini M, Hassanpour-Dehnavi A. Preparation and characterization of human decellularized ovarian scaffold based on supercritical carbon dioxide protocol. Biomed Eng Online 2025; 24:59. [PMID: 40361140 PMCID: PMC12070545 DOI: 10.1186/s12938-025-01392-7] [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: 01/10/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Primary ovarian insufficiency affecting 1-3% of women under 40, causes premature menopause and estrogen deficiency. With increasing life expectancy, a large percentage of women also face estrogen-related symptoms. A bioengineered ovary is one of the strategies to replace or enhance ovarian tissue function. A key advancement in bioengineering is the development of ovarian decellularized extracellular matrix scaffolds that mimic natural ovarian niche. Recent studies indicate that supercritical carbon dioxide (scCo2), with a density comparable to liquids and viscosity and diffusion coefficients properties similar to gases, holds substantial promise for application in engineered scaffolds. Therefore, we established a human decellularized ovarian scaffold based on a scCo2 process, as an optimized protocol. METHODS We evaluated two distinct pressure conditions (200 and 300 bar), while maintaining identical thermal (40 °C) and temporal (1.5 h) parameters, during the scCO2 decellularization process. In addition, two modifications were implemented to identify the most optimal protocol for enhancing the decellularization process: the inclusion of 70% ethanol as a co-solvent and the application of 1% sodium dodecyl sulfate (SDS) as a pretreatment for 4 h while utilizing the scCO2 system under the previously established conditions. Cell removal was confirmed by DNA quantification and H&E staining. Extracellular matrix structure evaluated by histological staining and scanning electron microscopy (SEM). Glycosaminoglycans (GAGs) content was quantified using a dimethyl methylene blue assay following extraction with HCL and MTT test was conducted to evaluate scaffold's cytocompatibility. RESULTS Application of 1% SDS, while utilizing the scCO2 system at 200 bar and 40 °C for 1.5 h, established an optimal protocol for preserving the essential characteristics of the ovarian ECM. This protocol is able to meet previously established decellularization criteria and histological staining and SEM showed that the ECM architecture was satisfactorily preserved. GAGs quantification indicated adequate preservation of GAGs content. Finally, MTT test presented the scaffolds had suitable cytocompatibility. CONCLUSIONS We propose an optimal protocol utilizing 1% SDS as a pretreatment, followed by the scCO2 system. This protocol addresses common challenges associated with traditional decellularization methods and presents a promising avenue for advancing ovarian tissue engineering applications.
Collapse
Affiliation(s)
- Fatemeh Hosseinpour
- Tissue Engineering Lab., Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Zeinolabedini Hezave
- Fanavari Atiyeh Pouyandegan Exir Company (APEX Technologies Co.), Science and Technology Park, Arak, Iran
- Fanavari Arena Exir Sabz Company (AREX Green Technologies Co.), Arak, Iran
| | - Tahereh Talaei-Khozani
- Tissue Engineering Lab., Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Rastgou-Maeini
- Tissue Engineering Lab., Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ashraf Hassanpour-Dehnavi
- Tissue Engineering Lab., Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Histomorphometry and Stereology Research Center, Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Anatomical Sciences, School of Medicine, Imam Hussain Square, Zand St., Shiraz, 7134845794, Fars, Iran.
| |
Collapse
|
2
|
Baker KM, FernandezCriado R, Eaton JL, Mensah VA. The Clinical Utility of Measures of Ovarian Reserve. Obstet Gynecol Surv 2025; 80:121-133. [PMID: 39924338 DOI: 10.1097/ogx.0000000000001362] [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: 02/11/2025]
Abstract
Importance Measures of ovarian reserve, particularly anti-Müllerian hormone, have been increasingly and inaccurately utilized as "fertility tests." It is important to understand the available measures of ovarian reserve and how to appropriately interpret and integrate their use into clinical practice. Objectives The objectives of this article are to review the process of reproductive aging, define ovarian reserve, describe the available measures of ovarian reserve, and discuss the clinical utility of these measures. Evidence Acquisition A literature search was performed using the electronic database PubMed. Relevant guidelines, systematic reviews, and original research articles investigating ovarian reserve parameters and their clinical utility were reviewed. Results The fecundity of women gradually declines with increasing reproductive age as oocyte quantity and quality decline. Ovarian reserve is defined as the quantity of oocytes remaining in the ovary. Ovarian reserve can be measured indirectly with the use of serum blood tests or ultrasound imaging. Measures of ovarian reserve are clinically useful in several circumstances, particularly for use during fertility treatment and cycles of assisted reproductive technology. However, measures of ovarian reserve are poor predictors of reproductive potential and should not be used as "fertility tests." Conclusions and Relevance Measures of ovarian reserve are poor predictors of reproductive potential and should not be used as "fertility tests." Age remains a stronger predictor of reproductive success than measures of ovarian reserve.
Collapse
Affiliation(s)
- Katherine M Baker
- Fellow, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Women and Infants Hospital and Warren Alpert Medical School of Brown University, Providence, RI
| | - Rodolfo FernandezCriado
- Resident, Department of Obstetrics and Gynecology, Women and Infants Hospital and Warren Alpert Medical School of Brown University, Providence, RI
| | - Jennifer L Eaton
- Division Director/Fellowship Director and Associate Professor, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Women and Infants Hospital and Warren Alpert Medical School of Brown University, Providence, RI
| | - Virginia A Mensah
- Assistant Professor/Clinician Educator, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Women and Infants Hospital and Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
3
|
Cipres DT, Gordon CM. Primary Ovarian Insufficiency, Bone Health, and Other Outcomes in Adolescents. Obstet Gynecol Clin North Am 2024; 51:663-678. [PMID: 39510737 PMCID: PMC11566969 DOI: 10.1016/j.ogc.2024.08.004] [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] [Indexed: 11/15/2024]
Abstract
Adolescents with primary ovarian insufficiency (POI) frequently present with arrested pubertal development or amenorrhea. Early evaluation of menstrual irregularities can avoid a delayed diagnosis. There are various genetic, autoimmune, and iatrogenic causes of POI, although most of the cases will not have an identified cause. Prompt initiation of hormone replacement therapy will restore developmentally appropriate pubertal progression, establishment of menses, and optimization of bone and cardiovascular health. The diagnosis of POI is often unexpected and life-altering for an adolescent and has broad health and psychosocial implications that are best approached with empathy, educational resources, and engagement of multidisciplinary specialists.
Collapse
Affiliation(s)
- Danielle T Cipres
- Division of Gynecology, Department of Surgery, Boston Children's Hospital, 333 Longwood Avenue, 5th Floor, Boston, MA 02115, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | - Catherine M Gordon
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, NICHD Office of the Clinical Director, 10 CRC, Room 5-2583, 10 Center Drive, MSC 1109, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Tang S, Guo T, Song C, Wang L, Zhang J, Rajkovic A, Lin X, Chen S, Liu Y, Tian W, Wu B, Wang S, Wang W, Lai Y, Wang A, Xu S, Jin L, Ke H, Zhao S, Li Y, Qin Y, Zhang F, Chen ZJ. MGA loss-of-function variants cause premature ovarian insufficiency. J Clin Invest 2024; 134:e183758. [PMID: 39545409 PMCID: PMC11563689 DOI: 10.1172/jci183758] [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: 06/11/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
Although premature ovarian insufficiency (POI), a common cause of female infertility and subfertility, has a well-established hereditary component, the genetic factors currently implicated in POI account for only a limited proportion of cases. Here, using an exome-wide, gene-based case-control analysis in a discovery cohort comprising 1,027 POI cases and 2,733 ethnically matched women controls from China, we found that heterozygous loss-of-function (LoF) variants of MAX dimerization protein (MGA) were significantly enriched in the discovery cohort, accounting for 2.6% of POI cases, while no MGA LoF variants were found in the matched control females. Further exome screening was conducted in 4 additional POI cohorts (2 from China and 2 from the United States) for replication studies, and we identified heterozygous MGA LoF variants in 1.0%, 1.4%, 1.0%, and 1.0% of POI cases, respectively. Overall, a total of 37 distinct heterozygous MGA LoF variants were discovered in 38 POI cases, accounting for approximately 2.0% of the total 1,910 POI cases analyzed in this study. Accordingly, Mga+/- female mice were subfertile, exhibiting shorter reproductive lifespan and decreased follicle number compared with WT, mimicking the observed phenotype in humans. Our findings highlight the essential role of MGA deficiency for impaired female reproductive ability.
Collapse
Affiliation(s)
- Shuyan Tang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Ting Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Chengcheng Song
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Lingbo Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aleksandar Rajkovic
- Department of Pathology, Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Xiaoqi Lin
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Shiling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yujun Liu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
| | - Weidong Tian
- School of Life Sciences, Fudan University, Shanghai, China
| | - Bangguo Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunhui Lai
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ao Wang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuhua Xu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- School of Life Sciences, Fudan University, Shanghai, China
| | - Hanni Ke
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Shidou Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- Shanghai Key Laboratory of Embryo Original Diseases, Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (no. 2021RU001), Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
5
|
Grosu-Bularda A, Lita FF, Hodea FV, Bordeanu-Diaconescu EM, Cretu A, Dumitru CS, Cacior S, Marinescu BM, Lascar I, Hariga CS. Navigating the Complexities of Radiation Injuries: Therapeutic Principles and Reconstructive Strategies. J Pers Med 2024; 14:1100. [PMID: 39590592 PMCID: PMC11595796 DOI: 10.3390/jpm14111100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Radiation injuries, particularly those resulting from therapeutic or accidental exposure, present complex challenges for medical management. These injuries can manifest localized skin damage or extend to deeper tissues, presenting as various clinical entities that require treatment strategies, ranging from conservative management to complex surgical interventions. Radiation treatment constitutes a fundamental component of neoplastic management, with nearly two out of three oncological instances undergoing it as an element of their therapeutic strategy. The therapeutic approach to radiation injury consists of expanding prophylactic measures while maintaining the efficacy of treatment, such as conservative treatment or local debridement followed by reconstruction. The armamentarium of reconstructive methods available for plastic surgeons, from secondary healing to free tissue transfer, can be successfully applied to radiation injuries. However, the unique pathophysiological changes induced by radiation necessitate a careful and specialized approach for their application, considering the altered tissue characteristics and healing dynamics. The therapeutic strategy is guided by both the severity and progression of the injury, with the primary aim of restoring functionality and aesthetic aspects while simultaneously minimizing the risk of complications. This paper explores the various conditions encompassed by the term "radiation injury," reviews both non-surgical and surgical therapeutic strategies for managing these injuries, and highlights the unique challenges associated with treating irradiated tissues within specific oncological contexts.
Collapse
Affiliation(s)
- Andreea Grosu-Bularda
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Flavia-Francesca Lita
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
- Clinical Department Plastic Surgery and Reconstructive Microsurgery, Central Military Emergency University Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Florin-Vlad Hodea
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Eliza-Maria Bordeanu-Diaconescu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Andrei Cretu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Catalina-Stefania Dumitru
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Stefan Cacior
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Bogdan-Mihai Marinescu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinical Department Plastic Surgery and Reconstructive Microsurgery, Central Military Emergency University Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Ioan Lascar
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Cristian-Sorin Hariga
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
6
|
Yang Z, Huang N, Zhuang Z, Jin M, Zhang Z, Song Y, Cui H, Zhang S, Huang T, Liu X, Li N. Earlier Age at Menopause, Plasma Metabolome, and Risk of Premature Mortality. Metabolites 2024; 14:571. [PMID: 39590807 PMCID: PMC11596455 DOI: 10.3390/metabo14110571] [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: 09/17/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Menopause and related metabolites are associated with mortality. However, the relationship between earlier menopause, premature mortality, and the role of metabolomic signatures remains underexplored. This study investigated the association between earlier menopause and premature mortality, and the mediating effect of metabolomic signatures. METHODS This prospective cohort study used data from the UK Biobank, including 33,687 post-menopausal women aged 40-69 years. Age at menopause was obtained from a baseline self-reported questionnaire and analyzed both as a continuous variable and in categories (<40, 40-49, and ≥50 years). Premature mortality was defined as deaths before 75 years. Cox regression was used to estimate hazard ratios (HRs), and elastic net regression identified metabolomic signatures related to menopause age. Mediation analysis was conducted to assess the proportion of the association explained by the metabolomic signature. RESULTS During a median follow-up of 13.7 years, 1612 cases of premature mortality occurred. Compared to menopause at ≥50 years, earlier menopause (HR 1.17, 95% CI 1.04-1.30) and premature menopause (HR 1.60, 95% CI 1.28-2.00) were associated with higher risks of premature mortality. A metabolomic signature inversely associated with premature mortality (HR per SD increment, 0.79; 95% CI, 0.75-0.83) mediated 13.6% (95% CI, 1.9%-28.3%) of the association between earlier menopause and premature mortality. CONCLUSIONS Earlier menopause is associated with an increased risk of premature mortality, partially mediated by a metabolomic signature related to age at menopause. These findings highlight the importance of metabolomic profiling in understanding menopause and mortality risks.
Collapse
Affiliation(s)
- Zeping Yang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| | - Ninghao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| | - Zhenhuang Zhuang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| | - Ming Jin
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| | - Ziyi Zhang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| | - Yimin Song
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| | - Haoliang Cui
- Department of Global Health, School of Public Health, Peking University, Beijing 100191, China;
| | - Shan Zhang
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing 100191, China;
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
- Key Laboratory of Epidemiology of Major Diseases, Peking University, Ministry of Education, Beijing 100191, China
- Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing 100191, China
| | - Xiaojing Liu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| | - Nan Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China; (Z.Y.); (N.H.); (Z.Z.); (M.J.); (Z.Z.); (Y.S.); (T.H.)
| |
Collapse
|
7
|
Le DC, Ngo MHT, Kuo YC, Chen SH, Lin CY, Ling TY, Pham QTT, Au HK, Myung J, Huang YH. Secretome from estrogen-responding human placenta-derived mesenchymal stem cells rescues ovarian function and circadian rhythm in mice with cyclophosphamide-induced primary ovarian insufficiency. J Biomed Sci 2024; 31:95. [PMID: 39390588 PMCID: PMC11468397 DOI: 10.1186/s12929-024-01085-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is an early decline in ovarian function that leads to ovarian failure. Conventional treatments for POI are inadequate, and treatments based on mesenchymal stem cells (MSCs) have emerged as an option. However, the lack of consideration of the estrogen niche in ovarian tissue significantly reduces the therapeutic efficacy, with an unclear mechanism in the MSCs in POI treatment. Furthermore, the disruption of circadian rhythm associated with POI has not been previously addressed. METHODS Conditioned medium (CM) and estradiol-conditioned medium (E2-CM) were generated from estrogen receptor positive MSCs (ER+pcMSCs). Chemotherapy-induced POI models were established using C57BL/6 mice (in vivo) and KGN cells (in vitro) treated with cyclophosphamide (CTX) or 4-hydroperoxycyclophosphamide (4-OOH-CP). Gene/protein expressions were detected using RT-qPCR, Western blotting, and immunohistochemistry assays. Locomotor activity was monitored for behavioral circadian rhythmicity. Cytokine arrays and miRNA analysis were conducted to analyze potential factors within CM/E2-CM. RESULTS The secretome of ER+pcMSCs (CM and E2-CM) significantly reduced the CTX-induced defects in ovarian folliculogenesis and circadian rhythm. CM/E2-CM also reduced granulosa cell apoptosis and rescued angiogenesis in POI ovarian tissues. E2-CM had a more favorable effect than the CM. Notably, ER+pcMSC secretome restored CTX-induced circadian rhythm defects, including the gene expressions associated with the ovarian circadian clock (e.g., Rora, E4bp4, Rev-erbα, Per2 and Dbp) and locomotor activity. Additionally, the cytokine array analysis revealed a significant increase in cytokines and growth factors associated with immunomodulation and angiogenesis, including angiogenin. Neutralizing the angiogenin in CM/E2-CM significantly reduced its ability to promote HUVEC tube formation in vitro. Exosomal miRNA analysis revealed the miRNAs involved in targeting the genes associated with POI rescue (PTEN and PDCD4), apoptosis (caspase-3, BIM), estrogen synthesis (CYP19A1), ovarian clock regulation (E4BP4, REV-ERBα) and fibrosis (COL1A1). CONCLUSION This study is the first to demonstrate that, in considering the estrogen niche in ovarian tissue, an estrogen-priming ER+pcMSC secretome achieved ovarian regeneration and restored the circadian rhythm in a CTX-induced POI mouse model. The potential factors involved include angiogenin and exosomal miRNAs in the ER+pcMSC secretome. These findings offer insights into potential stem cell therapies for chemotherapy-induced POI and circadian rhythm disruption.
Collapse
Affiliation(s)
- Duy-Cuong Le
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Laboratory, Vinmec International Hospital, Minh Khai Street, Hai Ba Trung, Hanoi, Vietnam
| | - Mai-Huong T Ngo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yung-Che Kuo
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Fishery Sciences, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Quoc Thao Trang Pham
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Heng-Kien Au
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, 11042, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
| | - Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City, 23561, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| | - Yen-Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| |
Collapse
|
8
|
Witham M, Hengel S. The role of RAD51 regulators and variants in primary ovarian insufficiency, endometriosis, and polycystic ovary syndrome. NAR MOLECULAR MEDICINE 2024; 1:ugae010. [PMID: 39359934 PMCID: PMC11443433 DOI: 10.1093/narmme/ugae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/09/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
The study of RAD51 regulators in female reproductive diseases has novel biomarker potential and implications for therapeutic advancement. Regulators of RAD51 play important roles in maintaining genome integrity and variations in these genes have been identified in female reproductive diseases including primary ovarian insufficiency (POI), endometriosis, and polycystic ovary syndrome (PCOS). RAD51 modulators change RAD51 activity in homologous recombination, replication stress, and template switching pathways. However, molecular implications of these proteins in primary ovarian insufficiency, endometriosis, and polycystic ovary syndrome have been understudied. For each reproductive disease, we provide its definition, current diagnostic and therapeutic treatment strategies, and associated genetic variations. Variants were discovered in RAD51, and regulators including DMC1, RAD51B, SWS1, SPIDR, XRCC2 and BRCA2 linked with POI. Endometriosis is associated with variants in XRCC3, BRCA1 and CSB genes. Variants in BRCA1 were associated with PCOS. Our analysis identified novel biomarkers for POI (DMC1 and RAD51B) and PCOS (BRCA1). Further biochemical and cellular analyses of RAD51 regulator functions in reproductive disorders will advance our understanding of the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Maggie Witham
- Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Sarah R Hengel
- Department of Biology, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
9
|
Dilbaz B. Editorial: Fertility preservation. Front Glob Womens Health 2024; 5:1452588. [PMID: 39188538 PMCID: PMC11345206 DOI: 10.3389/fgwh.2024.1452588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/15/2024] [Indexed: 08/28/2024] Open
Affiliation(s)
- Berna Dilbaz
- Etlik Zubeyde Hanim Women's Health Training and Research Hospital, University of Health Sciences, Istanbul, Türkiye
| |
Collapse
|
10
|
Memi E, Pavli P, Papagianni M, Vrachnis N, Mastorakos G. Diagnostic and therapeutic use of oral micronized progesterone in endocrinology. Rev Endocr Metab Disord 2024; 25:751-772. [PMID: 38652231 PMCID: PMC11294403 DOI: 10.1007/s11154-024-09882-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
Progesterone is a natural steroid hormone, while progestins are synthetic molecules. In the female reproductive system, progesterone contributes to the control of luteinizing hormone and follicle-stimulating hormone secretion and their pulsatility, via its receptors on the kisspeptin, neurokinin B, and dynorphin neurons in the hypothalamus. Progesterone together with estradiol controls the cyclic changes of proliferation and decidualization of the endometrium; exerts anti-mitogenic actions on endometrial epithelial cells; regulates normal menstrual bleeding; contributes to fertilization and pregnancy maintenance; participates in the onset of labor. In addition, it exerts numerous effects on other endocrine systems. Micronized progesterone (MP) is natural progesterone with increased bioavailability, due to its pharmacotechnical micronized structure, which makes it an attractive diagnostic and therapeutic tool. This critical literature review aims to summarize and put forward the potential diagnostic and therapeutic uses of MP in the field of endocrinology. During reproductive life, MP is used for diagnostic purposes in the evaluation of primary or secondary amenorrhea as a challenge test. Moreover, it can be prescribed to women presenting with amenorrhea or oligomenorrhea for induction of withdrawal bleeding, in order to time blood-sampling for diagnostic purposes in early follicular phase. Therapeutically, MP, alone or combined with estrogens, is a useful tool in various endocrine disorders including primary amenorrhea, abnormal uterine bleeding due to disordered ovulation, luteal phase deficiency, premenstrual syndrome, polycystic ovary syndrome, secondary amenorrhea [functional hypothalamic amenorrhea, premature ovarian insufficiency], perimenopause and menopause. When administrated per os, acting as a neurosteroid directly or through its metabolites, it exerts beneficial effects on brain function such as alleviation of symptoms of anxiety and depression, asw well as of sleep problems, while it improves working memory in peri- and menopausal women. Micronized progesterone preserves full potential of progesterone activity, without presenting many of the side-effects of progestins. Although it has been associated with more frequent drowsiness and dizziness, it can be well tolerated with nocturnal administration. Because of its better safety profile, especially with regard to metabolic ailments, breast cancer risk and veno-thromboembolism risk, MP is the preferred option for individuals with an increased risk of cardiovascular and metabolic diseases and of all-cause mortality.
Collapse
Affiliation(s)
- Eleni Memi
- Unit of Endocrinology, Diabetes mellitus, and Metabolism, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sophias Av. 76, 11528, Athens, Greece
| | - Polina Pavli
- Unit of Endocrinology, Diabetes mellitus, and Metabolism, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sophias Av. 76, 11528, Athens, Greece
| | - Maria Papagianni
- Department of Nutrition and Dietetics, School of Physical Education, Sport Science and Dietetics, University of Thessaly, 42100, Trikala, Greece
- Endocrine Unit, 3rd Department of Pediatrics, Hippokration Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54642, Thessaloniki, Greece
| | - Nikolaos Vrachnis
- Third Department of Obstetrics and Gynecology, Attikon General Hospital, School of Medicine, National and Kapodistrian University of Athens, Rimini Str. 1, 12462, Chaidari, Athens, Greece
- St George's NHS Foundation Trust Teaching Hospitals, St George's University of London, London, UK
| | - George Mastorakos
- Unit of Endocrinology, Diabetes mellitus, and Metabolism, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sophias Av. 76, 11528, Athens, Greece.
| |
Collapse
|
11
|
Serota A, D’Erasmo G. Estrogen exposure and skeletal health: Special populations and considerations. JOURNAL OF THE PEDIATRIC ORTHOPAEDIC SOCIETY OF NORTH AMERICA 2024; 7:100061. [PMID: 40433279 PMCID: PMC12088159 DOI: 10.1016/j.jposna.2024.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 05/29/2025]
Abstract
Estrogen is critical for bone health from puberty onwards. Various clinical scenarios in adolescence can impact skeletal exposure to estrogen during this vulnerable time. Primary ovarian insufficiency, premature menopause, and anorexia nervosa necessitate prompt evaluation, treatment, and replacement of estrogen in order to optimize accrual of peak bone mass. We have much still to learn about the skeletal impact of delaying puberty and gender affirming hormones in gender diverse individuals. While the choice of hormonal contraception in adolescence is often driven by patient preference and concerns about adherence, providers and patients much take the long-term impact on bone health into consideration. Key Concepts (1)Delayed, diminished or absent estrogen during adolescence has a negative impact on peak bone mass accrual.(2)Hormone replacement therapy is essential for patients with primary ovarian insufficiency and premature menopause.(3)Recovery from anorexia nervosa does not lead to a complete catch-up of bone density lost/not gained.(4)Not all hormonal contraception methods are created equal for the adolescent skeleton.(5)Skeletal health of trans youth is an emerging focus.
Collapse
Affiliation(s)
- Alana Serota
- Hospital for Special Surgery Department of Medicine, New York, NY, USA
| | - Giavanna D’Erasmo
- Hospital for Special Surgery Department of Medicine, New York, NY, USA
| |
Collapse
|
12
|
Benor A, Decherney A. Gonadotropin-Releasing Hormone (GnRH) Agonists Do Not Protect Ovarian Function in Patients Undergoing Stem Cell Transplants. Cureus 2024; 16:e58387. [PMID: 38756303 PMCID: PMC11097921 DOI: 10.7759/cureus.58387] [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: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Medical indications for fertility preservation include any malignancy, chronic illness, or disease that would require gonadotoxic chemotherapy or radiation (conditioning regimens), which would impede a woman's ability to conceive in the future. Thus, any patient who plans to undergo a gonadotoxic regimen is advised to cryopreserve oocytes or embryos, which can be used in the future at the patient's convenience. Attempts have been made to suppress ovarian function with gonadotropin-releasing hormone agonists (GnRH-a) to induce ovarian quiescence and, thereby, theoretically limit the gonadotoxic impact on the follicular pool. We explored the use of leuprolide (a type of GnRH-a) in preventing primary ovarian insufficiency (POI) in a cohort study of patients who underwent hematopoietic stem cell transplants (HSCT) at the National Institutes of Health (NIH); since the conditioning regimens for HSCT include cyclophosphamide and other gonadotoxic therapies, we hypothesized that GnRH-a would be ineffective in preventing POI. Methods We assessed patients who underwent fertility preservation prior to their stem cell transplant, as their follicular-stimulating hormone (FSH) levels were evaluated prior to and post-chemotherapy. Twenty-nine patients who underwent hormonal evaluation prior to and post-chemotherapy were included. The control group did not receive GnRH-a prior to chemotherapy, while the treatment group did receive GnRH-a pre-chemotherapy. Results Our data revealed that 80% of the control group had menopausal levels post-chemotherapy, while 91% of the treatment group still had menopausal levels post-chemotherapy (p=0.33). Conclusions Thus, our hypothesis that GnRH-a is ineffective in reducing the risk of POI in a cohort of patients who receive conditioning regimens for HSCT was confirmed.
Collapse
Affiliation(s)
- Ariel Benor
- Reproductive Endocrinology and Infertility, National Institutes of Health, Bethesda, USA
| | - Alan Decherney
- Reproductive Endocrinology and Infertility, National Institutes of Health, Bethesda, USA
| |
Collapse
|
13
|
Persico T, Tranquillo ML, Seracchioli R, Zuccarello D, Sorrentino U. PGT-M for Premature Ovarian Failure Related to CGG Repeat Expansion of the FMR1 Gene. Genes (Basel) 2023; 15:6. [PMID: 38275588 PMCID: PMC10815814 DOI: 10.3390/genes15010006] [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: 10/25/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Primary ovarian failure (POF) is caused by follicle exhaustion and is associated with menstrual irregularities and elevated gonadotropin levels, which lead to infertility before the age of 40 years. The etiology of POI is mostly unknown, but a heterogeneous genetic and familial background can be identified in a subset of cases. Abnormalities in the fragile X mental retardation 1 gene (FMR1) are among the most prevalent monogenic causes of POI. These abnormalities are caused by the expansion of an unstable CGG repeat in the 5' untranslated region of FMR1. Expansions over 200 repeats cause fragile X syndrome (FXS), whereas expansions between 55 and 200 CGG repeats, which are defined as a fragile X premutation, have been associated with premature ovarian failure type 1 (POF1) in heterozygous females. Preimplantation genetic testing for monogenic diseases (PGT-M) can be proposed when the female carries a premutation or a full mutation. In this narrative review, we aim to recapitulate the clinical and molecular features of POF1 and their implications in the context of PGT-M.
Collapse
Affiliation(s)
- Tiziana Persico
- Medically Assisted Procreation Center, Maternal and Child Department, Beauregard Hospital, Valle D’Aosta Local Public Health, 11100 Aoste, Italy
| | - Maria Lucrezia Tranquillo
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (M.L.T.); (R.S.)
| | - Renato Seracchioli
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (M.L.T.); (R.S.)
- Division of Gynaecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero, University of Bologna, 40138 Bologna, Italy
| | - Daniela Zuccarello
- Clinical Genetics and Epidemiology Unit, University of Padova, 35128 Padova, Italy; (D.Z.); (U.S.)
| | - Ugo Sorrentino
- Clinical Genetics and Epidemiology Unit, University of Padova, 35128 Padova, Italy; (D.Z.); (U.S.)
| |
Collapse
|
14
|
Kim SM, Yoo JY, Hong YH, Lee J, Kim JH, Lee JR. The effect of growth hormone on ovarian function recovery in a mouse model of ovarian insufficiency. Front Endocrinol (Lausanne) 2023; 14:1184977. [PMID: 37854196 PMCID: PMC10579899 DOI: 10.3389/fendo.2023.1184977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/08/2023] [Indexed: 10/20/2023] Open
Abstract
Objectives To evaluate the effects and mechanisms of action of growth hormone (GH) in the recovery of ovarian function in ovarian insufficiency induced by cyclophosphamide (CP) in a mouse model. Materials and methods After inducing ovarian insufficiency by administering 400 mg/kg of CP intraperitoneally to 6-week-old ICR mice, the mice were divided into four groups (control, CP, 1 mg/kg GH, and 2 mg/kg GH) with 10 mice in each group. GH was administered a week later for 7 days. Five mice from each group were sacrificed the next day, and their ovaries were collected for histological examination. The remaining mice were superovulated for in vitro fertilization (IVF). The terminal deoxynucleotidyl transferase dUTP-nick end labeling assay was performed to detect apoptosis. Masson's trichrome staining was used to analyze the degree of fibrosis. To quantify angiogenesis, CD31 immunohistochemistry was performed. Angiogenesis-related gene expression profiles were assessed using quantitative reverse transcription polymerase chain reaction. Results CP induced the loss of non-growing (primordial and primary) follicles while GH significantly protected primordial follicles and increased follicular quality. The CP group showed a decrease in fertilization and blastocyst formation rates in IVF. In contrast, the GH treatment group showed dose-dependent enhanced IVF outcomes. Furthermore, GH treatment decreased apoptosis and stromal fibrosis and increased angiogenesis. Many genes involved in angiogenesis, especially Leptin (Lep), platelet endothelial cell adhesion molecule 1 (Pecam-1), and angiogenin (Ang) were up-regulated in the GH treatment groups. Conclusion GH treatment may promote the recovery of ovarian function in ovarian insufficiency induced by the administration of CP via decreasing apoptosis and stromal fibrosis and upregulating Lep, Pecam-1, and Ang genes.
Collapse
Affiliation(s)
- Su Mi Kim
- Department of Obstetrics and Gynecology, Chungbuk National University Hospital, Cheongju, Republic of Korea
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jung Young Yoo
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Biomedical Laboratory Science, Eulji University, Seongnam, Republic of Korea
| | - Yeon Hee Hong
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jaewang Lee
- Department of Biomedical Laboratory Science, Eulji University, Seongnam, Republic of Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Bundang Medical Center, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Jung Ryeol Lee
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
15
|
Sütcüoğlu BM, Güler M. Appropriateness of premature ovarian insufficiency recommendations provided by ChatGPT. Menopause 2023; 30:1033-1037. [PMID: 37671567 DOI: 10.1097/gme.0000000000002246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
OBJECTIVE This study aimed to evaluate the appropriateness of ChatGPT's responses to frequently asked queries regarding the prevention and treatment of premature ovarian insufficiency (POI). METHODS A set of 25 questions covering topics related to disease risk factors, screening, symptoms, treatment, and treatment adverse effects were posed to ChatGPT, and the responses were evaluated by two experienced obstetrician/gynecologists. RESULTS Of the responses given by ChatGPT, 19 of 25 (76%) were deemed appropriate, five (20%) were rated as inappropriate, and the remaining one (4%) was considered unreliable. There was no question that ChatGPT answered completely incorrectly; answers that were considered inappropriate included partially correct information. CONCLUSIONS The study's findings suggest that ChatGPT provides patients with mostly accurate information about the POI but might present significantly inaccurate information. However, it is important to note that further research is needed to validate the use of artificial intelligence chatbots in the context of reproductive health, especially for individuals who may be highly impacted by the emotional and psychological effects of POI. In addition, given the complexity and individuality of medical care, AI chatbots should not be considered a replacement for medical professionals but rather a supplementary tool that can help patients access reliable information and support. Overall, this study contributes to the growing body of research on AI chatbots and their potential use in reproductive health, highlighting both the benefits and limitations of these technologies.
Collapse
Affiliation(s)
- Bengü Mutlu Sütcüoğlu
- From the Department of Obstetrics and Gynaecology, Lokman Hekim University, Ankara, Turkey
| | | |
Collapse
|
16
|
Nelson LM, Spencer H, Hijane K, Thinuan P, Nelson CW, Vincent AJ, Gordon CM, Plant TM, Fazeli PK. My 28 Days - a global digital women's health initiative for evaluation and management of secondary amenorrhea: case report and literature review. Front Endocrinol (Lausanne) 2023; 14:1227253. [PMID: 37772077 PMCID: PMC10523024 DOI: 10.3389/fendo.2023.1227253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/18/2023] [Indexed: 09/30/2023] Open
Abstract
There is a need to close the gap between knowledge and action in health care. Effective care requires a convenient and reliable distribution process. As global internet and mobile communication increase capacity, innovative approaches to digital health education platforms and care delivery are feasible. We report the case of a young African woman who developed acute secondary amenorrhea at age 18. Subsequently, she experienced a 10-year delay in the diagnosis of the underlying cause. A global digital medical hub focused on women's health and secondary amenorrhea could reduce the chance of such mismanagement. Such a hub would establish more efficient information integration and exchange processes to better serve patients, family caregivers, health care providers, and investigators. Here, we show proof of concept for a global digital medical hub for women's health. First, we describe the physiological control systems that govern the normal menstrual cycle, and review the pathophysiology and management of secondary amenorrhea. The symptom may lead to broad and profound health implications for the patient and extended family members. In specific situations, there may be significant morbidity related to estradiol deficiency: (1) reduced bone mineral density, 2) cardiovascular disease, and 3) cognitive decline. Using primary ovarian insufficiency (POI) as the paradigm condition, the Mary Elizabeth Conover Foundation has been able to address the specific global educational needs of these women. The Foundation did this by creating a professionally managed Facebook group specifically for these women. POI most commonly presents with secondary amenorrhea. Here we demonstrate the feasibility of conducting a natural history study on secondary amenorrhea with international reach to be coordinated by a global digital medical hub. Such an approach takes full advantage of internet and mobile device communication systems. We refer to this global digital women's health initiative as My 28 Days®.
Collapse
Affiliation(s)
- Lawrence M. Nelson
- Digital Women's Health Initiative, Mary Elizabeth Conover Foundation, Tysons, VA, United States
| | - Hillary Spencer
- Digital Women's Health Initiative, Mary Elizabeth Conover Foundation, Tysons, VA, United States
| | - Karima Hijane
- Digital Women's Health Initiative, Mary Elizabeth Conover Foundation, Tysons, VA, United States
| | - Payom Thinuan
- Faculty of Nursing, Boromarajonani College of Nursing Nakhon, Lampang, Thailand
| | - Chaninan W. Nelson
- Digital Women's Health Initiative, Mary Elizabeth Conover Foundation, Tysons, VA, United States
| | - Amanda J. Vincent
- Monash Centre for Health Research and Implementation (MCHRI), Monash University, Clayton, VIC, Australia
| | - Catherine M. Gordon
- Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, United States
| | - Tony M. Plant
- Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Pouneh K. Fazeli
- Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
17
|
Hoefgen HR, Benoit J, Chan S, Jayasinghe Y, Lustberg M, Pohl V, Saraf A, Schmidt D, Appiah LC. Female reproductive health in pediatric, adolescent, and young adult cancer survivors. Pediatr Blood Cancer 2023; 70 Suppl 5:e29170. [PMID: 37381166 DOI: 10.1002/pbc.29170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/02/2021] [Accepted: 05/11/2021] [Indexed: 06/30/2023]
Abstract
An estimated 500,000 cancer survivors of reproductive age in the United States will live to experience the long-term consequences of cancer treatment. Therefore, a focused aspect of cancer care has appropriately shifted to include quality of life in survivorship. Infertility is a late effect of therapy that affects 12% of female survivors of childhood cancer receiving any cancer treatment in large cohort studies and results in a 40% decreased likelihood of pregnancy in young adults of ages 18-39 years. Nonfertility gynecologic late effects such as hypoestrogenism, radiation-induced uterine and vaginal injury, genital graft-versus-host disease after hematopoietic stem cell transplant, and sexual dysfunction also significantly affect quality of life in survivorship but are underdiagnosed and require consideration. Several articles in the special edition "Reproductive Health in Adolescent and Young Adult Cancer Survivorship" address infertility, genital graft-versus-host disease, and psychosexual functioning in survivorship. This review article focuses on other adverse gynecologic outcomes of cancer therapies including hypogonadism and hormone replacement therapy, radiation-induced uterovaginal injury, vaccination and contraception, breast and cervical cancer screening, and pregnancy considerations in survivorship.
Collapse
Affiliation(s)
- Holly R Hoefgen
- Division of Pediatric and Adolescent Gynecology, Department of Obstet Gynecol, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Janie Benoit
- Gynecology & Reproductive Sciences, CHU Ste-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Serena Chan
- Division of Pediatric and Adolescent Gynecology, Department of Obstetrics, Gynecology & Reproductive Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yasmin Jayasinghe
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Maryam Lustberg
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Victoria Pohl
- Center for Cancer and Blood Disorders, Connecticut Children's Medical Center, Hartford, Connecticut, USA
| | - Amanda Saraf
- Division Pediatric Hematology Oncology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, USA
| | - Deb Schmidt
- MACC Fund Center for Cancer and Blood Disorders, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Leslie Coker Appiah
- Division of Pediatric and Adolescent Gynecology, Department of Obstet Gynecol, Children's Hospital Colorado, University of Colorado Denver, Denver, Colorado, USA
| |
Collapse
|
18
|
Kakinuma K, Kakinuma T. Analysis of oxidative stress and antioxidative potential in premature ovarian insufficiency. World J Clin Cases 2023; 11:2684-2693. [PMID: 37214574 PMCID: PMC10198121 DOI: 10.12998/wjcc.v11.i12.2684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/20/2023] [Accepted: 03/23/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is characterized by an early decline in ovarian function, inducing secondary amenorrhea. While the cause of POI has not yet been identified, the function of mitochondria in the ovaries and the cytotoxicity associated with reactive oxygen species (ROS) have been implicated in follicle pool depletion and a decline in follicle quality. Recently developed tests have enabled easy measurement of diacron-reactive oxygen metabolites (d-ROMs) and biological antioxidant potential (BAP). The combination of these two tests is used to comprehensively assess oxidative stress in the blood.
AIM To comprehensively assess the oxidative stress of d-ROMs and BAP in POI.
METHODS Participants were classified into two groups: A POI group of 11 women aged < 40 years examined between January 2021 and June 2022 with a history of secondary amenorrhea for at least 4 mo in our hospital and an FSH value of ≥ 40 mIU/mL; and a control group of healthy women of the same age with normal ovarian function in our hospital. Plasma d-ROMs and BAP were measured in both these groups underwent. Differences between groups were assessed using the t-test.
RESULTS The mean age and mean body mass index (BMI) were 35.8 ± 3.0 years and 20.1 ± 1.9 kg/m2 in the control group and 35.8 ± 2.7 years and 19.4 ± 2.5 kg/m2 in the POI group, respectively. The mean gravidity and parity in control and POI groups were 0.6 ± 0.7 and 0.4 ± 0.5 and 0.6 ± 0.9 and 0.3 ± 0.5, respectively. The two groups did not differ significantly in terms of mean age, BMI, gravidity, or parity. The d-ROMs level was significantly higher in the POI group than in the control group (478.2 ± 58.7 vs 341.1 ± 35.1 U.CARR; P < 0.001); however, the BAP level did not significantly differ between the two groups (2078.5 ± 157.4 vs 2029.0 ± 186.4 μmol/L). The oxidase stress index (d-ROMs/BAP × 100) was significantly higher in the POI group than in the control group (23.7 ± 3.3 vs 16.5 ± 2.1; P < 0.001).
CONCLUSION Oxidative stress was significantly greater in the POI group than in the control group, suggesting oxidative stress as a factor that can serve as a POI biomarker.
Collapse
Affiliation(s)
- Kaoru Kakinuma
- Department of Obstetrics and Gynecology, International Health and Welfare Hospital, Nasushiobara 327-2763, Japan
| | - Toshiyuki Kakinuma
- Department of Obstetrics and Gynecology, International Health and Welfare Hospital, Nasushiobara 327-2763, Japan
| |
Collapse
|
19
|
Reynolds AC, McKenzie LJ. Cancer Treatment-Related Ovarian Dysfunction in Women of Childbearing Potential: Management and Fertility Preservation Options. J Clin Oncol 2023; 41:2281-2292. [PMID: 36888938 PMCID: PMC10115556 DOI: 10.1200/jco.22.01885] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/22/2022] [Accepted: 01/24/2023] [Indexed: 03/10/2023] Open
Abstract
PURPOSE To review the complex concerns of oncofertility created through increased cancer survivorship and the long-term effects of cancer treatment in young adults. DESIGN Review chemotherapy-induced ovarian dysfunction, outline how fertility may be addressed before treatment initiation, and discuss barriers to oncofertility treatment and guidelines for oncologists to provide this care to their patients. CONCLUSION In women of childbearing potential, ovarian dysfunction resulting from cancer therapy has profound short- and long-term implications. Ovarian dysfunction can manifest as menstrual abnormalities, hot flashes, night sweats, impaired fertility, and in the long term, increased cardiovascular risk, bone mineral density loss, and cognitive deficits. The risk of ovarian dysfunction varies between drug classes, number of received lines of therapy, chemotherapy dosage, patient age, and baseline fertility status. Currently, there is no standard clinical practice to evaluate patients for their risk of developing ovarian dysfunction with systemic therapy or means to address hormonal fluctuations during treatment. This review provides a clinical guide to obtain a baseline fertility assessment and facilitate fertility preservation discussions.
Collapse
Affiliation(s)
| | - Laurie J. McKenzie
- Baylor College of Medicine Houston, Houston, TX
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
20
|
Patricia Wodi A, Marquez P, Mba-Jonas A, Barash F, Nguon K, Moro PL. Spontaneous reports of primary ovarian insufficiency after vaccination: A review of the vaccine adverse event reporting system (VAERS). Vaccine 2023; 41:1616-1622. [PMID: 36732165 DOI: 10.1016/j.vaccine.2022.12.038] [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: 11/26/2021] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Since 2012, reports of primary ovarian insufficiency (POI) temporally associated with receipt of human papillomavirus (HPV) vaccine have been published leading to questions about a potential causal association. A Vaccine Safety Datalink study did not find an increased risk for POI after vaccination. We reviewed the Vaccine Adverse Event Reporting System (VAERS) to describe POI reports. METHODS We searched VAERS, a U.S. passive surveillance system, for domestic POI reports received from 01/01/1990 to 12/31/2017 after any vaccination. The search used both Medical Dictionary for Regulatory Activity Preferred Terms and a text-based search for POI and its symptoms. All reports were reviewed, and the American College of Obstetricians and Gynecologists (ACOG) guidelines for POI diagnosis were applied. Data mining for disproportionate reporting was conducted. RESULTS Six hundred fifty-two reports met the search criteria and clinical review identified 19 POI reports. Most reports (n = 16) were received between 2013 and 2017. The median age at vaccination was 14.5 years (range 10-25 years) and the median interval between first dose of vaccination and reporting the event to VAERS was 43 months (range 4-132 months; mean 59.6 months). Four reports met ACOG diagnostic criteria; one with an underlying cause (47XXX chromosomal abnormality) reported. Eleven reports documented menstrual irregularity ≥ 3 months; 5 had ≥ 1 laboratory test result used to diagnose POI. Eighteen of 19 reports described receipt of HPV vaccine with or without other vaccines. Other vaccines reported were meningococcal conjugate vaccine, hepatitis A, varicella and tetanus toxoid, reduced diphtheria toxoid and acellular pertussis. Disproportionate reporting was found for three relevant coding terms after HPV vaccination. CONCLUSIONS POI is rarely reported to VAERS. Most reports contained limited diagnostic information and were submitted after published cases of POI following HPV vaccination. Results of our review do not suggest a safety concern.
Collapse
Affiliation(s)
- A Patricia Wodi
- Immunization Safety Office, Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, GA, United States.
| | - Paige Marquez
- Immunization Safety Office, Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Adamma Mba-Jonas
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Faith Barash
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Kosal Nguon
- Commonwealth Informatics, Inc., Waltham, MA, United States
| | - Pedro L Moro
- Immunization Safety Office, Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, GA, United States
| |
Collapse
|
21
|
Barrera EP, Grimstad FW, Boskey ER. Young Adult Patients with Testosterone Management Concerns after Gender-Affirming Hysterectomy and Bilateral Oophorectomy: A Case Series. J Pediatr Adolesc Gynecol 2023; 36:89-91. [PMID: 35850361 DOI: 10.1016/j.jpag.2022.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023]
Abstract
Many transgender and gender diverse adolescents and young adults will pursue hysterectomy for the purpose of gender affirmation. This procedure often includes bilateral salpingo-oophorectomy (BSO), which has potential implications for long-term health should individuals choose to stop, or lose access to, exogenous testosterone. Although most of these individuals intend to remain on testosterone indefinitely, not all do, and little information exists on such cases following bilateral oophorectomy to guide counseling and practice. This case series documents 3 individuals who had interruptions in their testosterone use after hysterectomy with BSO for reasons including external barriers, internal barriers, and concerns about side effects. Patients should be appropriately counseled on hysterectomy options as bilateral oophorectomy is not required in the absence of specific indications.
Collapse
Affiliation(s)
- Ellis P Barrera
- Division of Gynecology, Department of Surgery, Boston Children's Hospital, 333 Longwood Ave. 5th floor Room LO-545, Boston, MA 02115, United States
| | - Frances W Grimstad
- Division of Gynecology, Department of Surgery, Boston Children's Hospital, 333 Longwood Ave. 5th floor Room LO-545, Boston, MA 02115, United States; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, United States.
| | - Elizabeth R Boskey
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, United States; Center for Gender Surgery, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
22
|
Erol Koc EM, Ceyhan M, Yaman S, Neselioglu S, Erel O, Ozaksit MG. Prolidase as a marker of fibrogenesis in idiopathic primary ovarian insufficiency. Eur J Obstet Gynecol Reprod Biol 2023; 281:7-11. [PMID: 36521400 DOI: 10.1016/j.ejogrb.2022.12.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/30/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To evaluate the serumlevel of prolidase,which isa marker of fibrogenic activity, in women with idiopathic primary ovarian insufficiency (POI). STUDY DESIGN This is a prospective case-control study. Serum prolidase level was compared between the study group including 68 women with POI and control group including 65 normally menstruating women. Serum proline and hydroxyproline levels were also compared. Correlation analyses were performed between the prolidase level and POI related parameters including estradiol (E), follicle stimulating hormone (FSH), anti-mullerian hormone (AMH) levels, and presence of POI family history. RESULTS Serum prolidase and proline level were significantly increased in women with the diagnosis of POI compared to the control group (1082.57 (147.53) vs 981.13 (223.26) U/L, 233.30 (83.16) vs 218.94 (82.59) µmol/L, respectively). Prolidase level found to have significant correlations with AMH, E, FSH levels, and presence of POI family history (r = -0.49, p = 0.001; r = -0.39, p = 0.001; r = 0.42, p = 0.001; r = 0.22, p = 0.01; respectively). In receiver operating characteristics analysis, prolidase was shown to be a discriminative factor for POI at 1031.14 U/L cut-off value with 75 % sensitivity and 65 % specificity. Thearea under curve was 0.71 [(95 % CI: 0.62-0.79), p = 0.001]. CONCLUSION The current study revealed increased prolidase level in women withPOI. Serum prolidase level was also negatively correlated with the serum AMH level. Considering the present findings,prolidase may be a candidate molecule in assessment of POI cases.
Collapse
Affiliation(s)
- Esin Merve Erol Koc
- Department of Obstetrics and Gynecology, Ankara City Hospital, Ankara, Turkey.
| | - Meryem Ceyhan
- Department of Obstetrics and Gynecology, Ankara City Hospital, Ankara, Turkey
| | - Selen Yaman
- Department of Obstetrics and Gynecology, Ankara City Hospital, Ankara, Turkey
| | - Salim Neselioglu
- Department of Biochemistry, Ankara Yıldırım Beyazıt University, Faculty of Medicine, Ankara, Turkey
| | - Ozcan Erel
- Department of Biochemistry, Ankara Yıldırım Beyazıt University, Faculty of Medicine, Ankara, Turkey
| | | |
Collapse
|
23
|
Affiliation(s)
- Cynthia A Stuenkel
- From the Department of Medicine, University of California, San Diego, School of Medicine, La Jolla (C.A.S.); Unite de Gynecologie Medicale, Port Royal-Cochin, Universite de Paris Cité, Paris (A.G.)
| | - Anne Gompel
- From the Department of Medicine, University of California, San Diego, School of Medicine, La Jolla (C.A.S.); Unite de Gynecologie Medicale, Port Royal-Cochin, Universite de Paris Cité, Paris (A.G.)
| |
Collapse
|
24
|
Jang S, Kim MS, Kim PW, Kim S. Korean medicine treatment for premature ovarian failure: Three case reports. Explore (NY) 2023; 19:121-126. [PMID: 36085273 DOI: 10.1016/j.explore.2022.08.018] [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: 06/07/2021] [Revised: 08/22/2022] [Accepted: 08/28/2022] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Premature ovarian failure (POF) is defined as amenorrhea lasting for more than 4 months before 40 years of age, which is accompanied by a serum follicle-stimulating hormone (FSH) concentration above 40 mlU/mL. POF can cause a series of symptoms associated with low estrogen levels, such as hot flushes, excessive sweating, and infertility. This study aimed to report three cases of POF that were treated successfully with Korean medicine. CASE REPRESENTATION Three patients with POF were selected for inclusion in this study. The treatment regimen consisted of herbal medicines, electroacupuncture, moxibustion, and Hominis placental pharmacopuncture. The basic treatment period was 3 months, and follow-up was performed after menstrual recovery. Following treatment, all three patients resumed menstruation without any adverse events. One patient also conceived successfully. CONCLUSIONS These case reports suggest that Korean medicine could be effective for treating POF. Further preclinical and clinical studies are needed to investigate the mechanism of action of herbal medicines and acupuncture in improving menstruation and FSH levels.
Collapse
Affiliation(s)
- Soobin Jang
- Department of Preventive Medicine, College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbukdo, Republic of Korea
| | - Mi-Sun Kim
- Sojunghan Korean Medical Clinic, Seongnam, Republic of Korea
| | - Pyung-Wha Kim
- R&D Strategy Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Sungha Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
25
|
Dural O, Ulusoy HE, Tikiz MA, Gurbanova T, Yasa C, Ugurlucan FG, Akhan SE. Effects of Hormone Replacement Therapy on Low Bone Mineral Density in Adolescents and Young Women with Hypogonadism: Comparison of Oral and Transdermal 17 Beta-Estradiol Administration. J Pediatr Adolesc Gynecol 2022; 35:634-637. [PMID: 35644512 DOI: 10.1016/j.jpag.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022]
Abstract
STUDY OBJECTIVE To evaluate the effects of physiological dose 17 beta-estradiol (E2) replacement on low bone mineral density (BMD) and compare the results of oral and transdermal (TD) E2 administration in adolescents and young women with hypogonadism DESIGN, SETTING, AND PARTICIPANTS: We retrospectively reviewed the medical records of patients aged 15 to 24 years who were diagnosed with hypogonadism, who had begun receiving oral or TD E2 replacement, and whose initial dual-energy X-ray absorptiometry scan detected a lumbar spine BMD Z-score of -1 or lower between 2014 and 2018. The patients were divided into 2 groups according to the E2 route of administration as those who received 2 mg orally (Group 1) and 0.1 mg TD (Group 2). INTERVENTIONS None MAIN OUTCOME MEASURE: BMD scans of the patients at baseline and repeated within 2 years after E2 replacement RESULTS: In total, 43 patients who met the inclusion criteria were included in the study. Two groups did not differ for BMD scores at baseline. A significant improvement in BMD was observed with physiological dose E2 replacement in both groups. Mean BMD Z-score increased by +0.7 (95% CI, 0.47-0.93) in response to TD E2 administration, compared with +0.41 (95% CI, 0.25-0.58) during oral E2 replacement (P = .037). CONCLUSION We conclude that physiological dose E2 replacement, even within a short period of 2 years, has a significant beneficial effect on bone mass acquisition on the lumbar spine. Our study also demonstrates the possible superiority of TD E2 replacement over the oral route in increasing lumbar spine BMD.
Collapse
Affiliation(s)
- Ozlem Dural
- Istanbul University, Faculty of Medicine, Department of Obstetrics and Gynecology, Fatih, Istanbul, Turkey.
| | - Hevra Ekin Ulusoy
- Istanbul University, Faculty of Medicine, Department of Obstetrics and Gynecology, Fatih, Istanbul, Turkey
| | - Muge Ates Tikiz
- Istanbul University, Faculty of Medicine, Department of Obstetrics and Gynecology, Fatih, Istanbul, Turkey
| | - Turkane Gurbanova
- Medical Park Gebze Hospital, Section of Obstetrics and Gynecology, Gebze, Kocaeli, Turkey
| | - Cenk Yasa
- Istanbul University, Faculty of Medicine, Department of Obstetrics and Gynecology, Fatih, Istanbul, Turkey
| | - Funda Gungor Ugurlucan
- Istanbul University, Faculty of Medicine, Department of Obstetrics and Gynecology, Fatih, Istanbul, Turkey
| | - Suleyman Engin Akhan
- Istanbul University, Faculty of Medicine, Department of Obstetrics and Gynecology, Fatih, Istanbul, Turkey
| |
Collapse
|
26
|
Mokhtar R, Hans P, Sinha A. Comparing Non-invasive Prenatal Testing With Invasive Testing for the Detection of Trisomy 21. Cureus 2022; 14:e31252. [PMID: 36514620 PMCID: PMC9733793 DOI: 10.7759/cureus.31252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 11/09/2022] Open
Abstract
Background Non-invasive prenatal test (NIPT) is an intermediate step between serum screening and invasive diagnostic testing. It involves analysis of the cell-free fetal DNA (cffDNA) present in the maternal blood sample for determining the likelihood of fetal aneuploidy. Owing to its high sensitivity and specificity, NIPT has quickly gained popularity across the globe since its introduction to clinical practice, making it an attractive alternative to the available screening and diagnostic tests in use. Amniocentesis is currently the gold standard test for obtaining fetal DNA and diagnosing fetal trisomy prenatally, but it is invasive and has procedure-related adverse effects. This study aims to compare NIPT and amniocentesis in pregnancies screened positive for fetal trisomy. Material and methods This is an analytic cross-sectional prospective study conducted in the Department of Obstetrics & Gynecology, Patna Medical College and Hospital, for two and half years from December 2018 to June 2021. A total of 34 pregnant women screened positive for trisomy 21, attending the antenatal care outpatient department, in their second trimester, with their written consent, were enrolled in the study. Results Out of 34 pregnant patients, three refused NIPT and directly opted for amniocentesis. A total of 31 pregnant women have undergone NIPT. A total of 28 cases were positive for trisomy 21 on both NIPT and amniocentesis. The sensitivity of NIPT was 100% with the confidence interval being 87.66% to 100.00%. The specificity of NIPT was 100% with the confidence interval being 29.24% to 100.00%. Conclusion The high performance and effectiveness of NIPT are undeniable. Though the process by which this test has to be integrated into the clinical practice needs more study and should be determined with meticulous assessment.
Collapse
Affiliation(s)
- Rifat Mokhtar
- Obstetrics and Gynecology, Bhagwan Mahavir Institute of Medical Sciences, Pawapuri, IND
| | - Punit Hans
- Obstetrics and Gynecology, Patna Medical College, Patna, IND
| | - Anjana Sinha
- Obstetrics and Gynecology, Patna Medical College, Patna, IND
| |
Collapse
|
27
|
Shylasree TS, Singh P, Kazi M, Gupta S, Engineer R, Patil PS, DeSouza A, Saklani A. Laparoscopic ovarian transposition in teenage and young women with locally advanced rectal cancer: Respite amidst cancer chaos. Colorectal Dis 2022; 24:697-705. [PMID: 35133696 DOI: 10.1111/codi.16084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/02/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
AIM Ovarian transposition is an established surgical technique to salvage ovaries in premenopausal women requiring pelvic radiation. The success rate of ovarian transposition varies widely depending on the route, technique of surgery, type of cancer and treatment. Here, we aimed to analyse the effectiveness of laparoscopic ovarian transposition (LOT) in teenage and young women prior to pelvic radiation in locally advanced rectal cancers (LARC). METHODS Patients who underwent LOT for rectal cancers were retrieved from a prospectively maintained database from June 2013-September 2019. Disease characteristics, return of menstrual function and oncological outcomes were analysed. RESULTS A total of 46 women with a mean age of 25.2 years who underwent LOT at the cancer centre were included in the study. Seventy percent were nulliparous. All patients were fit for discharge by 24 h. Mean time to start radiation was 19.6 days (range 3-47 days). Median follow-up of patients was 42 months. A total of 41 patients were assessable for ovarian function, 65.5% had resumption of menses. Median ovarian survival was 79 months and 5-year ovarian survival was 54%. Median overall survival from rectal cancer was 51 months. CONCLUSION Laparoscopic ovarian transposition is a safe and effective technique of ovarian protection from the gonadotoxic effects of pelvic radiation in LARC. It does not delay primary treatment and does not compromise oncological outcomes. Long-term follow-up is required to evaluate fertility and quality of life.
Collapse
Affiliation(s)
- Thumkur S Shylasree
- Department of Gynaecology Oncology, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| | - Pooja Singh
- Department of Gynaecology Oncology, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| | - Mufaddal Kazi
- Department of Surgical Oncology, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| | - Stuti Gupta
- Department of Gynaecology Oncology, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| | - Reena Engineer
- Department of Radiation Oncology, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| | - Prachi S Patil
- Department of Digestive Diseases and Clinical Nutrition, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| | - Ashwin DeSouza
- Department of Surgical Oncology, Robotic and Colorectal Surgery, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| | - Avanish Saklani
- Department of Surgical Oncology, Robotic and Colorectal Surgery, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
28
|
Nickel RS, Maher JY, Hsieh MH, Davis MF, Hsieh MM, Pecker LH. Fertility after Curative Therapy for Sickle Cell Disease: A Comprehensive Review to Guide Care. J Clin Med 2022; 11:2318. [PMID: 35566443 PMCID: PMC9105328 DOI: 10.3390/jcm11092318] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/30/2022] Open
Abstract
Curative therapy for sickle cell disease (SCD) currently requires gonadotoxic conditioning that can impair future fertility. Fertility outcomes after curative therapy are likely affected by pre-transplant ovarian reserve or semen analysis parameters that may already be abnormal from SCD-related damage or hydroxyurea treatment. Outcomes are also likely affected by the conditioning regimen. Conditioning with myeloablative busulfan and cyclophosphamide causes serious gonadotoxicity particularly among post-pubertal females. Reduced-intensity and non-myeloablative conditioning may be acutely less gonadotoxic, but more short and long-term fertility outcome data after these approaches is needed. Fertility preservation including oocyte/embryo, ovarian tissue, sperm, and experimental testicular tissue cryopreservation should be offered to patients with SCD pursing curative therapy. Regardless of HSCT outcome, longitudinal post-HSCT fertility care is required.
Collapse
Affiliation(s)
- Robert Sheppard Nickel
- Children’s National Hospital, Division of Hematology, Washington, DC 20001, USA;
- Children’s National Hospital, Division of Blood and Marrow Transplantation, Washington, DC 20001, USA
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20001, USA;
| | - Jacqueline Y. Maher
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Pediatric and Adolescent Gynecology, National Institutes of Health, Bethesda, MD 20810, USA;
- Children’s National Hospital, Pediatric and Adolescent Gynecology Program, Washington, DC 20001, USA
| | - Michael H. Hsieh
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20001, USA;
- Children’s National Hospital, Division of Urology, Washington, DC 20001, USA
| | - Meghan F. Davis
- Department of Urology, MedStar Georgetown University Hospital, Washington, DC 20001, USA;
| | - Matthew M. Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20810, USA;
| | - Lydia H. Pecker
- Division of Hematology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 20810, USA
| |
Collapse
|
29
|
Gómez-Rojas S, Aristizábal-Duque JE, Muñoz-Fernández LF, Sarmiento-Ramón MP, Pereira-Gómez MDP. Nueva variante del gen STAG3 causante de insuficiencia ovárica prematura. REVISTA COLOMBIANA DE OBSTETRICIA Y GINECOLOGÍA 2022; 73:142-148. [PMID: 35503298 PMCID: PMC9097685 DOI: 10.18597/rcog.3806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/05/2022] [Indexed: 11/23/2022]
Abstract
Objetivos: describir un caso de falla ovárica secundaria a una variante patogénica homocigota en el gen STAG3 no reportada previamente. Materiales y métodos: paciente de 16 años con amenorrea primaria y ausencia de características sexuales secundarias, en quien se documentó hipotiroidismo autoinmune, pobre desarrollo genital y cintilla gonadal, por lo cual se realizó secuenciación de exorna clínico. Se identificó una variante homocigota patogénica previamente no reportada en el gen STAG3, el cual ha sido relacionado con insuficiencia ovárica prematura (IOP). Conclusiones: en este caso, la realización de exorna clínico fue determinante para identificar una alteración del gen STAG, probablemente asociada a la IOP y el pronóstico a largo plazo de la paciente. Se establece una nueva variante patogénica c.2773delT; p.Ser925Profs*6 del gen STAG3 asociada a la IOP.
Collapse
|
30
|
Steinman B, Kilduff S, Del Rio M, Hayde N. Amenorrhea in a pediatric kidney transplant recipient: Answers. Pediatr Nephrol 2022; 37:565-567. [PMID: 34731311 DOI: 10.1007/s00467-021-05320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/01/2022]
Affiliation(s)
- Benjamin Steinman
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, NY, USA
| | - Stella Kilduff
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, NY, USA
| | - Marcela Del Rio
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, NY, USA
| | - Nicole Hayde
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, NY, USA.
| |
Collapse
|
31
|
Turkyilmaz A, Alavanda C, Ates EA, Geckinli BB, Polat H, Gokcu M, Karakaya T, Cebi AH, Soylemez MA, Guney Aİ, Ata P, Arman A. Whole-exome sequencing reveals new potential genes and variants in patients with premature ovarian insufficiency. J Assist Reprod Genet 2022; 39:695-710. [PMID: 35066699 PMCID: PMC8995228 DOI: 10.1007/s10815-022-02408-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/19/2022] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Premature ovarian insufficiency (POI) is a heterogeneous disorder characterized by the cessation of menstrual cycles before the age of 40 years due to the depletion or dysfunction of the ovarian follicles. POI is a highly heterogeneous disease in terms of etiology. The aim of this study is to reveal the genetic etiology in POI patients. METHODS A total of 35 patients (mean age: 27.2 years) from 28 different families diagnosed with POI were included in the study. Karyotype, FMR1 premutation analysis, single nucleotide polymorphism (SNP) array, and whole-exome sequencing (WES) were conducted to determine the genetic etiology of patients. RESULTS A total of 35 patients with POI were first evaluated by karyotype analysis, and chromosomal anomaly was detected in three (8.5%) and FMR1 premutation was detected in six patients (17%) from two different families. A total of 29 patients without FMR1 premutation were included in the SNP array analysis, and one patient had a 337-kb deletion in the chromosome 6q26 region including PARK2 gene, which was thought to be associated with POI. Twenty-nine cases included in SNP array analysis were evaluated simultaneously with WES analysis, and genetic variant was detected in 55.1% (16/29). CONCLUSION In the present study, rare novel variants were identified in genes known to be associated with POI, which contribute to the mutation spectrum. The effects of detected novel genes and variations on different pathways such as gonadal development, meiosis and DNA repair, or metabolism need to be investigated by experimental studies. Molecular etiology allows accurate genetic counseling to the patient and family as well as fertility planning.
Collapse
Affiliation(s)
- Ayberk Turkyilmaz
- Department of Medical Genetics, School of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| | - Ceren Alavanda
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Esra Arslan Ates
- grid.414850.c0000 0004 0642 8921Department of Medical Genetics, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Bilgen Bilge Geckinli
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Hamza Polat
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Mehmet Gokcu
- grid.31564.350000 0001 2186 0630Department of Medical Genetics, School of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Taner Karakaya
- Department of Medical Genetics, Isparta City Hospital, Isparta, Turkey
| | - Alper Han Cebi
- grid.31564.350000 0001 2186 0630Department of Medical Genetics, School of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mehmet Ali Soylemez
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ahmet İlter Guney
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Pinar Ata
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ahmet Arman
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
32
|
Kuang X, Tang Y, Xu H, Ji M, Lai D. The Evaluation of Ovarian Function Recovery Following Treatment of Primary Ovarian Insufficiency: A Systematic Review. Front Endocrinol (Lausanne) 2022; 13:855992. [PMID: 35573993 PMCID: PMC9095968 DOI: 10.3389/fendo.2022.855992] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is gaining awareness as its prevalence increases and its effect on patients is extremely negative. To date, several therapies have been designed to treat POI, but the conclusions are conflicting, in part, due to inconsistent evaluation methods. Thus, we explore a multi-index of ovarian function assessment methods to evaluate the recovery of ovarian function after various therapies in order to evaluate effectiveness in a more comprehensive manner. AIM The purpose of this review is to assess the effectiveness of various therapies to recover ovarian function in patients with POI. The primary outcome measures were anti-Müllerian hormone (AMH) levels, follicle stimulating hormone (FSH) levels, and antral follicle count (AFC). The secondary outcomes included the change of mean ovarian volume, menstruation recovery, and pregnancy rate. METHODS Our systematic searching including PubMed, Web of Science, Cochrane, and Embase databases was conducted to find all human clinical trial articles published from January 2000 to April 2021 and related to POI treatment, including the keywords: POI, AFC, and hormones. All prospective and retrospective studies exploring ovarian function recovery that include AFC, AMH levels, and FSH levels evolution throughout treatment were included. All patients included in the studies met the POI criteria described by the European Society for Human Reproductive Embryology (ESHRE) guideline. RESULTS Six studies were selected based on the criteria: one randomized controlled trial and five observational studies. Among them, two studies focused on the intraovarian platelet-rich plasma (PRP) infusion treatment, two studies focused on dehydroepiandrosterone (DHEA) supplements, one study focused on hormone replacement therapy (HRT), and one study focused on autologous adipose-derived stromal cells (ADSCs) treatment. There was insufficient scientific evidence that any approach could help ovarian function recovery in patients with POI because the ovarian function markers in each study had inconsistent changes with 26 patients (6.2%) reporting spontaneous pregnancy. CONCLUSION Serum AMH levels, FSH levels, and AFC are sensitive indicators and reflect the evolution of ovarian function. Large randomized controlled trials are necessary, and the data on ovarian function should be collected comprehensively to evaluate the effectiveness of a variety of treatments.
Collapse
Affiliation(s)
- Xiaojun Kuang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yongzhe Tang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hong Xu
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Min Ji
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- *Correspondence: Min Ji, ; Dongmei Lai,
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- *Correspondence: Min Ji, ; Dongmei Lai,
| |
Collapse
|
33
|
Ovarian Telomerase and Female Fertility. Biomedicines 2021; 9:biomedicines9070842. [PMID: 34356906 PMCID: PMC8301802 DOI: 10.3390/biomedicines9070842] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 11/16/2022] Open
Abstract
Women's fertility is characterized both quantitatively and qualitatively mainly by the pool of ovarian follicles. Monthly, gonadotropins cause an intense multiplication of granulosa cells surrounding the oocyte. This step of follicular development requires a high proliferation ability for these cells. Telomere length plays a crucial role in the mitotic index of human cells. Hence, disrupting telomere homeostasis could directly affect women's fertility. Strongly expressed in ovaries, telomerase is the most effective factor to limit telomeric attrition and preserve ovarian reserve. Considering these facts, two situations of infertility could be correlated with the length of telomeres and ovarian telomerase activity: PolyCystic Ovary Syndrome (PCOS), which is associated with a high density of small antral follicles, and Premature Ovarian Failure (POF), which is associated with a premature decrease in ovarian reserve. Several authors have studied this topic, expecting to find long telomeres and strong telomerase activity in PCOS and short telomeres and low telomerase activity in POF patients. Although the results of these studies are contradictory, telomere length and the ovarian telomerase impact in women's fertility disorders appear obvious. In this context, our research perspectives aimed to explore the stimulation of ovarian telomerase to limit the decrease in the follicular pool while avoiding an increase in cancer risk.
Collapse
|
34
|
Verrilli L, Johnstone E, Allen-Brady K, Welt C. Shared genetics between nonobstructive azoospermia and primary ovarian insufficiency. F&S REVIEWS 2021; 2:204-213. [PMID: 36177363 PMCID: PMC9518791 DOI: 10.1016/j.xfnr.2021.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
OBJECTIVE Primary ovarian insufficiency (POI) and Non-obstructive azoospermia (NOA) both represent disease states of early, and often complete, failure of gametogenesis. Because oogenesis and spermatogenesis share the same conserved steps in meiosis I, it is possible that inherited defects in meiosis I could lead to shared causes of both POI and NOA. Currently, known genes that contribute to both POI and NOA are limited. In this review article, we provide a systematic review of genetic mutations in which both POI and NOA phenotypes exist. EVIDENCE REVIEW A PubMed literature review was conducted from January 1, 2000 through October 2020. We included all studies that demonstrated human cases of POI or NOA due to a specific genetic mutation either within the same family or in separate families. RESULTS We identified 33 papers that encompassed 10 genes of interest with mutations implicated in both NOA and POI. The genes were all involved in processes of meiosis I. CONCLUSION Mutations in genes involved in processes of meiosis I may cause both NOA and POI. Identifying these unique phenotypes among shared genotypes leads to biologic plausibility that the key error occurs early in gametogenesis with an etiology shared among both male and female offspring. From a clinical standpoint, this shared relationship may help us better understand and identify individuals at high risk for gonadal failure within families and suggests that clinicians obtain history for opposite sex family members when approaching a new diagnosis of POI or NOA.
Collapse
Affiliation(s)
- Lauren Verrilli
- University of Utah School of Medicine, Department of Obstetrics and Gynecology, 30 N 1900 E #2B200, Salt Lake City, UT 84132
| | - Erica Johnstone
- University of Utah School of Medicine, Department of Obstetrics and Gynecology, 30 N 1900 E #2B200, Salt Lake City, UT 84132
| | - Kristina Allen-Brady
- University of Utah School of Medicine, Division of Epidemiology, Department of Internal Medicine, 296 Chipeta Way, Salt Lake City, UT 84108
| | - Corrine Welt
- University of Utah School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Salt Lake City, UT 84132
| |
Collapse
|
35
|
Patel S, Vargo JA, Olson A, Mahajan A. Supportive care for toxicities in children undergoing radiation therapy. Pediatr Blood Cancer 2021; 68 Suppl 2:e28597. [PMID: 33818886 DOI: 10.1002/pbc.28597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 06/16/2020] [Accepted: 07/01/2020] [Indexed: 11/08/2022]
Abstract
Radiation therapy (RT) is an integral part of the management of many pediatric tumors; however, it is associated with both acute and permanent adverse events that can significantly impact a child's quality of life, lead to treatment delays, and potentially affect outcomes of cancer therapy. Prevention, early detection, and optimal management of these adverse effects will help reduce their impact on the patients' quality of life and overall well-being. Unfortunately, there has not been a coordinated effort to study the etiology, evaluate risk factors, and explore novel treatments for these conditions. Studies of supportive care for children undergoing RT are often small and uncontrolled. This review will focus on the impact of irradiation on the different organ systems and their current management. Further studies are required to improve our understanding of the contributing factors and explore novel treatment options for these adverse effects and to enable children and their families to better cope with some of the unavoidable toxicities following multimodality therapy.
Collapse
Affiliation(s)
- Samir Patel
- Divisions of Radiation Oncology and Pediatric Hematology, Oncology and Palliative Care, University of Alberta, Stollery Children's Hospital, Edmonton, Canada
| | - John Austin Vargo
- Department of Radiation Oncology, UPMC Magee-Womens Hospital, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Adam Olson
- Department of Radiation Oncology, UPMC Children's Hospital of Pittsburg, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Anita Mahajan
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
36
|
Debiec KE, Todd N. Gynecologic Care for Pediatric and Adolescent Patients Undergoing Hematopoietic Stem Cell Transplantation. J Pediatr Adolesc Gynecol 2021; 34:112-116. [PMID: 33338627 DOI: 10.1016/j.jpag.2020.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/01/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cell transplantation is used to treat many chronic and acute malignant and nonmalignant conditions. We review hematopoietic stem cell transplantation and its effect on the gynecologic health of pediatric and adolescent patients, including pretransplantation evaluation, contraception, menstrual suppression, sexual health, fertility, primary ovarian insufficiency, and graft vs host disease. Comprehensive and team-based care provides optimal anticipatory counseling, evaluation, and management of acute and ongoing gynecologic issues.
Collapse
Affiliation(s)
- Katherine E Debiec
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle, Washington.
| | - Nicole Todd
- Division of General Gynecology and Obstetrics, Division of Gynecologic Specialties, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
37
|
Premature ovarian insufficiency: pathogenesis and therapeutic potential of mesenchymal stem cell. J Mol Med (Berl) 2021; 99:637-650. [PMID: 33641066 DOI: 10.1007/s00109-021-02055-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 01/29/2021] [Accepted: 02/21/2021] [Indexed: 12/14/2022]
Abstract
Primary ovarian insufficiency (POI) is defined as a reduction in ovarian function before the expected age of menopause. POI is known to increase the risk of cardiovascular disorders, osteoporosis, cognitive decline, and mood disorders, resulting in a reduced quality of life. Appropriate hormone replacement for premenopausal women decreases these adverse health risks and improves quality of life for women with POI, but does not prolong life expectancy. The potential etiologies of POI include chromosomal abnormalities and genetic mutations, autoimmune factors, and iatrogenic causes, including surgery, chemotherapy, and radiation therapy. A major association is suggested to exist between reproductive longevity and the DNA damage pathway response genes. DNA damage and repair in ovarian granulosa cells is strongly associated with POI. Depletion of oocytes with damaged DNA occurs through different cell death mechanisms, such as apoptosis, autophagy, and necroptosis, mediated by the phosphatase and tensin homolog (PTEN)/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/forkhead transcription factors 3 (FOXO3) pathway. Mesenchymal stem cells (MSCs) are characterized by the ability of self-renewal and differentiation and play an important role in the regeneration of injured tissues. Transplantation of MSCs has been shown to functionally restore ovarian reserve in a POI mouse model. Recent advances in stem cell therapy are likely to be translated to new therapeutic options bringing new hope to patients with POI. The aim of this review is to summarize the pathogenic mechanisms that involve cell death and DNA damage and repair pathways and to discuss the stem cell-based therapies as potential therapeutic options for this gynecologic pathology.
Collapse
|
38
|
Biswas L, Tyc K, Yakoubi WE, Morgan K, Xing J, Schindler K. Meiosis interrupted: the genetics of female infertility via meiotic failure. Reproduction 2021; 161:R13-R35. [PMID: 33170803 PMCID: PMC7855740 DOI: 10.1530/rep-20-0422] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022]
Abstract
Idiopathic or 'unexplained' infertility represents as many as 30% of infertility cases worldwide. Conception, implantation, and term delivery of developmentally healthy infants require chromosomally normal (euploid) eggs and sperm. The crux of euploid egg production is error-free meiosis. Pathologic genetic variants dysregulate meiotic processes that occur during prophase I, meiotic resumption, chromosome segregation, and in cell cycle regulation. This dysregulation can result in chromosomally abnormal (aneuploid) eggs. In turn, egg aneuploidy leads to a broad range of clinical infertility phenotypes, including primary ovarian insufficiency and early menopause, egg fertilization failure and embryonic developmental arrest, or recurrent pregnancy loss. Therefore, maternal genetic variants are emerging as infertility biomarkers, which could allow informed reproductive decision-making. Here, we select and deeply examine human genetic variants that likely cause dysregulation of critical meiotic processes in 14 female infertility-associated genes: SYCP3, SYCE1, TRIP13, PSMC3IP, DMC1, MCM8, MCM9, STAG3, PATL2, TUBB8, CEP120, AURKB, AURKC, andWEE2. We discuss the function of each gene in meiosis, explore genotype-phenotype relationships, and delineate the frequencies of infertility-associated variants.
Collapse
Affiliation(s)
- Leelabati Biswas
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Katarzyna Tyc
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Warif El Yakoubi
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Katie Morgan
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
39
|
La Marca A, Mastellari E. Fertility preservation for genetic diseases leading to premature ovarian insufficiency (POI). J Assist Reprod Genet 2021; 38:759-777. [PMID: 33495935 DOI: 10.1007/s10815-021-02067-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The current review aims to summarize the data available concerning the applicability of fertility preservation techniques to genetic conditions at risk of premature ovarian insufficiency (POI). METHODS A literature review through the PubMed Database was carried out. RESULTS About 10% of cases of POI is related to genetic diseases. The most frequent conditions associated with POI are Turner syndrome and fragile X pre-mutation; mutation of BRCA 1-2 genes and several other mutations and genetic syndromes have recently been highlighted, although they rarely occur. If a diagnosis is issued before POI onset, counseling on currently available fertility preservation techniques is advisable. In case of spontaneous menarche (this can occur variably depending on the mutation) established techniques like embryo or oocyte cryopreservation can be proposed, even if, in some cases, their effectiveness may be reduced by ovarian alterations connected to the mutation. Ovarian tissue cryopreservation has recently been defined as an established medical procedure for fertility preservation in young cancer patients and may be an option for prepubertal patients. However, it is still experimental in special populations with genetic diseases causing POI. New innovative experimental techniques, like in vitro maturation of immature oocytes (IVM) and vitro activation (IVA) of immature follicles on ovarian tissue, have shown limited but encouraging data and they will be probably available in the near future. For a correct risk-benefit evaluation, the following aspects should be considered: actual knowledge about the pathology-specific efficacy of the various techniques, the average age of onset of POI, the possible risks associated with the procedure in relation to the underlying pathology, the probability of spontaneous conception, as well as the health implications of a possible future pregnancy.. CONCLUSIONS Fertility preservation techniques represent a crucial opportunity for patients with genetic risk of POI. Early diagnosis increases the chances to apply these techniques. No specific recommendations concerning fertility preservation for each genetic pathology are available, and clinicians should first counsel the patient and her relatives about known risks and benefits of the available techniques, both those established and those considered as experimental.
Collapse
Affiliation(s)
- Antonio La Marca
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy. .,Clinica Eugin Modena, Modena, Italy.
| | - Elisa Mastellari
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
40
|
Abstract
Fertility represents a biological and psychological requirement for women. Some genetic diseases represent a rare cause of infertility, being responsible for 10% of cases of premature ovarian insufficiency. Among these, the most frequent and also those most studied by researchers are Turner Syndrome - due to a karyotype abnormality of the X chromosome pair - and the presence of fragile X premutation (FMR1). To exclude these conditions the diagnostic workup for non-iatrogenic premature ovarian insufficiency (POI) involves the performance of a karyotype analysis and the search for the FMR1 gene mutation, as well as the search for the presence of Y-chromosomal material. However, several other mutations and genetic syndromes associated with POI development have recently been highlighted, although they occur rarely, such as the GALT gene mutation in galactosemia or the FOXL2 gene mutation in BPES and many others, and further autosomal genetic testing are indicated if clinical suspicion is present. Mutations of BRCA 1 and 2 genes, make patients at genetically determined high risk of developing early ovarian or breast cancer and of getting POIs for the treatments they must undergo to prevent it (prophylactic bilateral oophorectomy) or treat it (chemotherapy). The management of impaired fertility is not less important than that of other syndromic manifestations for the quality of life of patients. Few data are available regarding the efficiency of cryopreservation of reproductive material (oocytes, embryos or ovarian tissue) in order to preserve fertility in this particular subgroup of patients, but certainly it represents a promising chance and a hope for the future.
Collapse
Affiliation(s)
- Elisa Mastellari
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio La Marca
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy -
- Clinica Eugin Modena, Modena, Italy
| |
Collapse
|
41
|
Davis SM, Soares K, Howell S, Cree-Green M, Buyers E, Johnson J, Tartaglia NR. Diminished Ovarian Reserve in Girls and Adolescents with Trisomy X Syndrome. Reprod Sci 2020; 27:1985-1991. [PMID: 32578162 PMCID: PMC7529937 DOI: 10.1007/s43032-020-00216-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/06/2020] [Indexed: 10/24/2022]
Abstract
An extra X chromosome occurs in ~ 1 in 1000 females, resulting in a karyotype 47,XXX also known as trisomy X syndrome (TXS). Women with TXS appear to be at increased risk for premature ovarian insufficiency; however, very little research on this relationship has been conducted. The objective of this case-control study is to compare ovarian function, as measured by anti-mullerian hormone (AMH) levels, between girls with TXS and controls. Serum AMH concentrations were compared between 15 females with TXS (median age 13.4 years) and 26 controls (median age 15.1 years). Females with TXS had significantly lower serum AMH compared to controls (0.7 ng/mL (IQR 0.2-1.7) vs 2.7 (IQR 1.3-4.8), p < 0.001). Additionally, girls with TXS were much more likely to have an AMH below the 2.5th percentile for age with 67% of them meeting these criteria (OR 11, 95% CI 2.3-42). Lower AMH concentrations in females with TXS may represent an increased risk for primary ovarian insufficiency in these patients and potentially a narrow window of opportunity to pursue fertility preservation options. Additional research is needed to understand the natural history of low AMH concentrations and future risk of premature ovarian insufficiency in girls with TXS.
Collapse
Affiliation(s)
- Shanlee M Davis
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
- Department of Pediatric Endocrinology, Children's Hospital Colorado, 13123 East 16th Ave B265, Aurora, CO, 80045, USA.
| | - Katelyn Soares
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Susan Howell
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Department of Developmental Pediatrics, Children's Hospital Colorado, Aurora, CO, USA
| | - Melanie Cree-Green
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Department of Pediatric Endocrinology, Children's Hospital Colorado, 13123 East 16th Ave B265, Aurora, CO, 80045, USA
- Center for Women's Health Research, Aurora, CO, 80045, USA
| | - Eliza Buyers
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Department of Pediatric and Adolescent Gynecology, Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Joshua Johnson
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nicole R Tartaglia
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Department of Developmental Pediatrics, Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
42
|
Abstract
The diagnosis of primary ovarian insufficiency (POI) has untold effects on women and a better understanding alongside potential treatments are paramount to improve quality of life of these women. Various causes have been linked to the development of POI with genetics playing a key role. A better understanding of the genetics of POI could lead to earlier diagnosis and broaden fertility options. This chapter discusses previously known and more recently discovered genes that have been implicated in the development of POI. It explores the varying phenotypic expressions of some genes in different populations and areas for further research in the genetics of POI.
Collapse
|
43
|
Lin D, Quan H, Chen K, Lin L, Lin L, Ji Q. An adolescent girl with premature ovarian failure, Graves' disease, and chronic urticaria: a case report. J Med Case Rep 2020; 14:184. [PMID: 33038927 PMCID: PMC7548041 DOI: 10.1186/s13256-020-02491-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/07/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Premature ovarian failure is characterized by amenorrhea, hypoestrogenism, and hypergonadotropinism, and occurs in women under 40 years of age. The prevalence of premature ovarian failure in women younger than 20 years of age is only 0.01%. Immune disorders are one of the causes of premature ovarian failure. Graves' disease and chronic urticaria are also associated with immune disorders. CASE PRESENTATION We report a case of a 15-year-old Han Chinese girl with premature ovarian failure complicated by Graves' disease and chronic urticaria. She experienced menarche at 13 years of age and presented with amenorrhea after 7 months of irregular menstruation. Laboratory examinations indicated hypoestrogenism and hypergonadotropinism. Ultrasound imaging revealed that her uterus and ovaries were small in size. Gene and antibody tests related to premature ovarian failure returned negative results. Both thyroid peroxidase autoantibody and thyrotropin receptor antibody were positive. After reviewing the literature on the relationship between these three diseases and immune disorders, our patient was diagnosed as having atypical autoimmune polyglandular syndrome. After taking small doses of estrogen for 6 months, the size of her uterus increased, and her psychological anxiety was relieved. CONCLUSIONS We report a case of an unusual association of premature ovarian failure, Graves' disease, and chronic urticaria. This case presents an atypical combination of adolescent autoimmune polyglandular syndrome, which is worthy of the attention of clinicians and presents an important lesson for them. Our case highlights that premature ovarian failure in adolescents requires long-term follow-up and medical treatment as well as psychological counselling.
Collapse
Affiliation(s)
- Danhong Lin
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Xiuying District, Haikou City, Hainan Province, China
| | - Huibiao Quan
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Xiuying District, Haikou City, Hainan Province, China.
| | - Kaining Chen
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Xiuying District, Haikou City, Hainan Province, China
| | - Lu Lin
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Xiuying District, Haikou City, Hainan Province, China
| | - Leweihua Lin
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Xiuying District, Haikou City, Hainan Province, China
| | - Qun Ji
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Xiuying District, Haikou City, Hainan Province, China
| |
Collapse
|
44
|
Abstract
Primary ovarian insufficiency (POI) is an uncommon yet devastating occurrence that results from a premature depletion of the ovarian pool of primordial follicles. Our understanding of both putative and plausible mechanisms underlying POI, previously considered to be largely "idiopathic", has been furthered over the past several years, largely due to advances in the field of genetics and through expansion of translational models for experimental research. In this review, our goal is to familiarize the multidisciplinary readers of the F1000 platform with the strides made in the field of reproductive medicine that hold both preventative and therapeutic implications for those women who are at risk for or who have POI.
Collapse
Affiliation(s)
- Victoria Wesevich
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Amanada N Kellen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Lubna Pal
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
45
|
Fattet AJ, Toupance S, Thornton SN, Monnin N, Guéant JL, Benetos A, Koscinski I. Telomere length in granulosa cells and leukocytes: a potential marker of female fertility? A systematic review of the literature. J Ovarian Res 2020; 13:96. [PMID: 32825843 PMCID: PMC7442985 DOI: 10.1186/s13048-020-00702-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022] Open
Abstract
In the context of a continuously increased delay of motherhood and of an increase of the incidence of premature ovarian failure, it is of the greatest interest to dispose of a predictive marker of the duration of the fertility window. Unfortunately, current available markers of women's fertility (hormonal rates or echography count of small follicles) have a poor predictive value of premature ovarian failure. In the last ten years, some studies have suggested that telomere length may be correlated with premature ovarian failure, but the results of these studies are contradictory.In accordance with guidelines from Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA), this systematic review of the literature selected studies evaluating telomere length or telomerase activity in granulosa cells and/or in leukocytes as a premature ovarian failure marker.Five publications (252 premature ovarian failure patients) were included in this review of experimental evidence. Two of them studied telomere length and/or telomerase activity in granulosa cells and 4 in leukocytes in women with premature ovarian failure. For each study, authors determined if there was a positive or a negative correlation between telomeric parameters and premature ovarian failure.3 studies (178 premature ovarian failure patients) found shorter telomere length in granulosa cells and/or leukocytes and/or lower telomerase activity in premature ovarian failure patients. 2 studies (74 premature ovarian failure patients) presented contradictory results about the correlation of leucocyte telomere length with premature ovarian failure.Shorter telomeres and diminished telomerase activity in granulosa cells appear to be associated with ovarian insufficiency. However, the number of studies and of subjects within are low and the methodology questionable. The confirmation of these results is essential with more subjects, better defined populations and more adapted methodology, in order to consider telomere length in granulosa cells and/or in leucocytes as an early and reliable marker for the decline of ovarian function.
Collapse
Affiliation(s)
- Anne-Julie Fattet
- Laboratory of Biology of Reproduction-CECOS Lorraine, University Hospital of Nancy, 10 rue du Dr Heydenreich, 54000, Nancy, France
| | - Simon Toupance
- Université de Lorraine, Inserm, DCAC, F-54000, Nancy, France
| | | | - Nicolas Monnin
- Centre AMP Majorelle-Laboratory ATOUTBIO, 95, rue Ambroise Paré, 54000, Nancy, France
| | | | | | - Isabelle Koscinski
- Laboratory of Biology of Reproduction-CECOS Lorraine, University Hospital of Nancy, 10 rue du Dr Heydenreich, 54000, Nancy, France.
- Université de Lorraine, Inserm, NGERE, F-54000, Nancy, France.
| |
Collapse
|
46
|
Tang R, Yu Q. Novel variants in women with premature ovarian function decline identified via whole-exome sequencing. J Assist Reprod Genet 2020; 37:2487-2502. [PMID: 32789750 DOI: 10.1007/s10815-020-01919-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To investigate the potential etiologies of premature ovarian insufficiency (POI) and diminished ovarian reserve (DOR). METHODS Fourteen women with sporadic POI and 6 women with DOR were enrolled. We used whole-exome sequencing (WES) and bioinformatics analysis to identify variants in a subset of 599 selected POI candidate genes. The identified genes were subjected to gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment and protein-protein interaction (PPI) network analyses to uncover key genes and pathways. RESULTS Among the 20 patients, 79 heterozygous variants were detected in 49 genes, which were classified as "likely pathogenic" or "variants of uncertain significance" according to the guidelines of the American College of Medical Genetics and Genomics. Most patients (17/20) carried two or more variants. Monoacylglycerol O-acyltransferase 1 mutations were found in six patients, and cytochrome P450 family 26 subfamily B member 1 and Bardet-Biedl syndrome 9 mutations were each found in four patients. Some variants were shared between DOR and POI. Enrichment analyses showed that the identified genes participate in key ovarian processes, such as follicular development, gonadal development, meiosis, Fanconi anemia, homologous recombination, and transforming growth factor β signaling. A PPI network revealed interactions between these proteins. CONCLUSION Premature ovarian function decline may be polygenic, and overlap exists between the genetic backgrounds of DOR and POI. WES and in silico analyses may be a useful clinical tool for etiological diagnosis and risk prediction for high-risk women in the future.
Collapse
Affiliation(s)
- Ruiyi Tang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Qi Yu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China.
| |
Collapse
|
47
|
Benetti-Pinto CL, Soares Júnior JM, Maciel GA, Nácul AP, Yela DA, Silva ACJSRE. Premature ovarian insufficiency: A hormonal treatment approach. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2020; 42:511-518. [PMID: 32898916 PMCID: PMC10309232 DOI: 10.1055/s-0040-1716929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Cristina Laguna Benetti-Pinto
- Departamento de Tocoginecologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - José Maria Soares Júnior
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Gustavo Arantes Maciel
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Daniela Angerame Yela
- Departamento de Tocoginecologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Ana Carolina Japur Sá Rosa e Silva
- Departamento de Ginecologia e Obstetrícia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
48
|
Cattoni A, Parissone F, Porcari I, Molinari S, Masera N, Franchi M, Cesaro S, Gaudino R, Passoni P, Balduzzi A. Hormonal replacement therapy in adolescents and young women with chemo- or radio-induced premature ovarian insufficiency: Practical recommendations. Blood Rev 2020; 45:100730. [PMID: 32654893 DOI: 10.1016/j.blre.2020.100730] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/10/2020] [Accepted: 06/23/2020] [Indexed: 12/27/2022]
Abstract
In women with premature ovarian insufficiency (POI), hormonal therapy (HT) is indicated to decrease the risk of morbidity and to treat symptoms related to prolonged hypoestrogenism. While general recommendations for the management of HT in adults with POI have been published, no systematic suggestions focused on girls, adolescents and young women with POI following gonadotoxic treatments (chemotherapy, radiotherapy, stem cell transplantation) administered for pediatric cancer are available. In order to highlight the challenging issues specifically involving this cohort of patients and to provide clinicians with the proposal of practical therapeutic protocol, we revised the available literature in the light of the shared experience of a multidisciplinary team of pediatric oncologists, gynecologists and endocrinologists. We hereby present the proposals of a practical scheme to induce puberty in prepubertal girls and a decisional algorithm that should guide the clinician in approaching HT in post-pubertal adolescents and young women with iatrogenic POI.
Collapse
Affiliation(s)
- A Cattoni
- Department of Pediatrics, Università degli Studi di Milano Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma, Azienda Ospedaliera San Gerardo, Via Pergolesi 33, 20900 Monza, Italy.
| | - F Parissone
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Universitaria Integrata di Verona, Università di Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| | - I Porcari
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Universitaria Integrata di Verona, Università di Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| | - S Molinari
- Department of Pediatrics, Università degli Studi di Milano Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma, Azienda Ospedaliera San Gerardo, Via Pergolesi 33, 20900 Monza, Italy.
| | - N Masera
- Department of Pediatrics, Università degli Studi di Milano Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma, Azienda Ospedaliera San Gerardo, Via Pergolesi 33, 20900 Monza, Italy
| | - M Franchi
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Universitaria Integrata di Verona, Università di Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy.
| | - S Cesaro
- Pediatric Hematology Oncology, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata di Verona, Università di Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy.
| | - R Gaudino
- Pediatric Endocrinology, Azienda Ospedaliera Universitaria Integrata di Verona, Università di Verona, Piazzale Aristide Stefani 1, 37126 Verona, Italy.
| | - P Passoni
- Department of Obstetrics and Gynecology, Azienda Ospedaliera San Gerardo, Via Pergolesi 33, 20900 Monza, Italy
| | - A Balduzzi
- Department of Pediatrics, Università degli Studi di Milano Bicocca, Fondazione Monza e Brianza per il Bambino e la sua Mamma, Azienda Ospedaliera San Gerardo, Via Pergolesi 33, 20900 Monza, Italy.
| |
Collapse
|
49
|
McGowan MC, Doyle EA, Boulware SD. Primary Ovarian Insufficiency in the Prepubertal Adolescent: A Case Report. J Pediatr Health Care 2020; 34:256-263. [PMID: 32334747 DOI: 10.1016/j.pedhc.2019.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 11/29/2022]
Abstract
Although there are many etiologies for delayed puberty in adolescent-aged girls, the pediatric provider should consider primary ovarian insufficiency if estradiol remains undetectable despite elevated levels of gonadotropins. Adolescent girls with this diagnosis will need holistic care from their primary care provider, focusing on both their medical and psychosocial needs. The following case study describes a 14-year-old girl who was referred to pediatric endocrinology for delayed puberty, in the setting of increased gonadotropins and undetectable estradiol. The differential diagnosis, evaluation, and management of primary ovarian insufficiency are reviewed as well as potential long-term health considerations.
Collapse
|
50
|
Gruber N, Kugler S, de Vries L, Brener A, Zung A, Eyal O, Rachmiel M, Koren I, Tenenbaum-Rakover Y, Hershkovitz E, Landau Z, Oren M, Eliakim A, Zangen D, German A, Majdoub H, Mazor-Aronovitch K, Modan-Moses D, Yeshayahu Y, Naugolni L, Levy-Shraga Y, Ben-Ami M, Brill G, Zuckerman-Levin N, Levy-Khademi F, Avnon-Ziv C, Tiosano D, Harel S, Kedem E, Segev-Becker A, Shoenfeld Y, Pinhas-Hamiel O. Primary Ovarian Insufficiency Nationwide Incidence Rate and Etiology Among Israeli Adolescents. J Adolesc Health 2020; 66:603-609. [PMID: 31987720 DOI: 10.1016/j.jadohealth.2019.11.315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/13/2019] [Accepted: 11/27/2019] [Indexed: 02/08/2023]
Abstract
PURPOSE The aim of the study was to estimate the current incidence and the distribution of etiologies of primary ovarian insufficiency (POI) in a nationwide study. The prevalence of POI in young adult women has recently increased, but the data cited for adolescents are more than three decades old. METHODS Data regarding females aged <21 years diagnosed with POI during the years 2000-2016 were collected from all the pediatric endocrinology units in Israel. POI was defined by at least 4 months of amenorrhea in association with menopausal levels of follicle-stimulating hormone. Iatrogenic cases were excluded. RESULTS For the 130 females aged <21 years included in the study, the distribution of POI etiologies was Turner syndrome/mosaicism in 56 (43%), idiopathic in 35 (27%), and other (developmental, genetic, metabolic, adrenal, and autoimmune) in 39 (30%) females. During the years 2009-2016, compared with 2000-2008, the incidence rate of new POI diagnoses per 100,000 person-years doubled (4.5 vs. 2.0; p value <.0001), and incidence rates of idiopathic and other etiologies increased by 2.6 (p value = .008) and 3.0 (p value = .002), respectively. In contrast, the incidence of Turner syndrome was constant (p value = .2). In the age group of 15-21 years, the current incidence of non-Turner POI in adolescents is one per 100,000 person-years. CONCLUSIONS In this nationwide study, the incidence rate of POI in youth aged <21 years was one tenth of the rate that is commonly cited. A significant increase in the rate of POI in non-Turner females was observed over the last decade. Contributions of environmental and epigenetic factors should be studied.
Collapse
Affiliation(s)
- Noah Gruber
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| | - Shir Kugler
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Liat de Vries
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Avivit Brener
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Amnon Zung
- Pediatric Endocrinology Unit, Kaplan Medical Center, Rehovot, Israel; The School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ori Eyal
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Marianna Rachmiel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Pediatric Endocrinology Unit, Assaf Haroffeh Medical Center, Zerifin, Israel
| | - Ilana Koren
- Pediatric Clinic, Armon Child Center, Clalit Health Services, Haifa, Israel
| | - Yardena Tenenbaum-Rakover
- Pediatric Endocrine Unit, Ha'Emek Medical Center, Affula, Israel; The Rappaport Endocrinology Faculty of Medicine, Technion, Haifa, Israel
| | - Eli Hershkovitz
- Pediatric Endocrinology and Metabolic Unit, Soroka University Medical Center, Israel and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Zohar Landau
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Pediatric Endocrine and Diabetes Unit, E. Wolfson Medical Center, Holon, Israel
| | - Meirav Oren
- Paediatric Endocrine Unit, Rambam Health Care Campus, Haifa, Israel
| | - Alon Eliakim
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Pediatric Department, Meir Medical Center, Kfar Saba, Israel
| | - David Zangen
- The School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Division of Pediatric Endocrinology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Alina German
- Pediatric Department, Bnei Zion Medical Center, Haifa, Israel
| | - Hussein Majdoub
- Pediatric Clinic, Armon Child Center, Clalit Health Services, Haifa, Israel
| | - Kineret Mazor-Aronovitch
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Dalit Modan-Moses
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Yonatan Yeshayahu
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Larisa Naugolni
- Pediatric Endocrinology Unit, Assaf Haroffeh Medical Center, Zerifin, Israel
| | - Yael Levy-Shraga
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Michal Ben-Ami
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Gherta Brill
- Migdal Hamea Health Services, Clalit Health Services, Tel Aviv, Israel
| | - Nehama Zuckerman-Levin
- The Rappaport Endocrinology Faculty of Medicine, Technion, Haifa, Israel; Institute of Diabetes, Endocrinology and Metabolism, Rambam Medical Center, Haifa, Israel
| | - Floris Levy-Khademi
- The School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Division of Pediatric Endocrinology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Carmit Avnon-Ziv
- The School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Division of Pediatric Endocrinology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Dov Tiosano
- The Rappaport Endocrinology Faculty of Medicine, Technion, Haifa, Israel; Paediatric Endocrine Unit, Rambam Health Care Campus, Haifa, Israel
| | - Shira Harel
- Maccabi Health Services, Central District, Tel Aviv, Israel
| | - Einat Kedem
- Pediatric Endocrine and Diabetes Unit, E. Wolfson Medical Center, Holon, Israel
| | - Anat Segev-Becker
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yehuda Shoenfeld
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Orit Pinhas-Hamiel
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|