1
|
Jiang D, Cai X, Fang H, Li Y, Zhang Z, Chen H, Zheng Z, Wang W, Sun Y. Coexposure to ambient air pollution and temperature and its associations with birth outcomes in women undergoing assisted reproductive technology in Fujian, China: A retrospective cohort study. JOURNAL OF HAZARDOUS MATERIALS 2025; 481:136539. [PMID: 39561545 DOI: 10.1016/j.jhazmat.2024.136539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/17/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND The interactions between pollutants and temperature coexposure, the mixing effects and their potential mechanisms remain uncertain. METHODS This retrospective cohort study included 11,766 women with infertility who received treatment at Fujian Hospital between 2015 and 2024. The daily mean concentrations of the six pollutants and the relative humidity and temperature data were acquired from the Fujian region. Data on genes were obtained from the Comparative Toxicogenomics Database. RESULTS O3 (aOR=0.80, 95 % CI=0.725--0.891) and temperature (aOR=0.936, 95 % CI=0.916--0.957) were negatively correlated with live birth rates. Moreover, PM10 (aOR=1.135, 95 % CI=1.028--1.252) and PM2.5 (aOR=1.146, 95 % CI=1.03--1.274) were positively associated with preterm birth. Among the effects on live births, PM2.5, PM10, NO2, CO, and SO2 had significant synergistic effects with temperature; in addition, O3 had significant antagonistic effects with temperature. A notable trend toward declining live birth rates with elevated concentrations of mixed pollutants was observed. Different infertility patients have different sensitivities to coexposure. Gene enrichment and cell experiments are associated mainly with cellular life activities. CONCLUSIONS Individual effects, interactions, and mixed effects between temperature and air pollutants and birth outcomes persist when air pollutant levels are relatively low. AAP may trigger miscarriage through cytotoxic effects.
Collapse
Affiliation(s)
- Dongdong Jiang
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Xuefen Cai
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; Fujian Maternal-Fetal Clinical Medicine Research Center, Fuzhou, Fujian, China
| | - Hua Fang
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuehong Li
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; Fujian Maternal-Fetal Clinical Medicine Research Center, Fuzhou, Fujian, China
| | - Ziqi Zhang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Haoting Chen
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zixin Zheng
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenxiang Wang
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| | - Yan Sun
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; Fujian Maternal-Fetal Clinical Medicine Research Center, Fuzhou, Fujian, China.
| |
Collapse
|
2
|
Khateeb S. Etoricoxib-NLC Mitigates Radiation-Induced Ovarian Damage in Rats: Insights into Pro-Inflammatory Cytokines, Antioxidant Activity, and Hormonal Responses. Biomolecules 2024; 15:12. [PMID: 39858407 PMCID: PMC11761947 DOI: 10.3390/biom15010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Radiotherapy is a critical treatment for cancer but poses significant risks to ovarian tissue, particularly in young females, leading to premature ovarian failure (POF). This study examines the therapeutic potential of etoricoxib nanostructured lipid carriers (ETO-NLC) in mitigating radiation-induced ovarian damage in female Wistar rats. Twenty-four female rats were randomly assigned to four groups: a control group receiving normal saline, a group exposed to a single dose of whole-body gamma radiation (6 Gy), a group treated with etoricoxib (10 mg/kg) post-radiation, and a group treated with ETO-NLC for 14 days following radiation. Histopathological evaluations and oxidative stress biomarker assessments were conducted, including ELISAs for reactive oxygen species (ROS), pro-inflammatory cytokines (IL-1β, TNF-α), and signaling molecules (PI3K, AKT, P38MAPK, AMH). Serum levels of estrogen, FSH, and LH were measured, and gene expression analysis for TGF-β and Nrf2 was performed using qRT-PCR. The findings indicate that ETO-NLC has the potential to ameliorate the harmful effects of ovarian damage induced by γ-radiation. These therapeutic effects were achieved through the modulation of oxidative stress, inflammation, augmentation of antioxidant defenses (including Nrf2 activation), support for cell survival pathways (via PI3K/Akt signaling), regulation of MAPK, mitigation of fibrosis (TGF-β), and preservation of ovarian reserve (as evidenced by AMH, FSH/LH, and estrogen levels). ETO-NLC shows promise as an effective strategy for attenuating radiation-induced ovarian damage, highlighting the need for further research to enhance therapeutic interventions aimed at preserving ovarian function during cancer treatment.
Collapse
Affiliation(s)
- Sahar Khateeb
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Biochemistry Division, Department of Chemistry, Faculty of Science, Fayoum University, Fayoum P.O. Box 63514, Egypt;
| |
Collapse
|
3
|
Yao Y, Zhu S, Zhu X. PCOS Influences the Expression of AMHRII in the Endometrium of AEH During the Reproductive Age. Diagnostics (Basel) 2024; 14:2872. [PMID: 39767233 PMCID: PMC11675281 DOI: 10.3390/diagnostics14242872] [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] [Received: 10/26/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Endometrial proliferative lesions (EPLs) encompass endometrial hyperplasia (EH) and endometrial carcinoma (EC). Atypical endometrial hyperplasia (AEH) is associated with an elevated risk of progression to EC. Patients with polycystic ovarian syndrome (PCOS) exhibit higher serum levels of anti-Müllerian hormone (AMH) and a correspondingly increased incidence of EPLs. AMH has the capacity to inhibit the cell proliferation of EPLs derived from Müllerian duct tissue through the AMH-AMH receptor (AMHR) signaling pathway. METHODS Pairs of samples matched by preference scores were randomly selected. Immunohistochemistry was employed to assess the expression levels of AMHR type II (AMHR2) in endometrial tissue. A comparative analysis was performed between tissues from individuals with PCOS and those without, as well as between a normal endometrium and endometrial tissue from individuals with EPLs. This study aimed to elucidate differences in AMHR2 expression among these tissue types. By focusing on AMHR2 expression, the impact of the PCOS-related background on the endometrial AMH-AMHR cascade signaling pathway was initially investigated. RESULTS The AMHR2 protein was expressed in the endometrium of both the PCOS group and the non-PCOS group during the reproductive age (20-39 years). The expression of the AMHR2 protein in the AEH endometrium of PCOS patients did not differ significantly from that in the normal endometrium of PCOS patients; however, it was significantly higher than in the AEH endometrium of non-PCOS patients (p = 0.011). Conversely, the expression of the AMHR2 protein in the AEH endometrium of non-PCOS patients was significantly lower than that in the normal endometrium of non-PCOS patients (p = 0.021). Notably, there was no significant difference in AMHR2 protein expression in a normal endometrium between PCOS and non-PCOS patients. CONCLUSIONS The involvement of the endometrial AMH-AMHR cascade signaling pathway and its biological effects in the pathogenesis of AEH are evident. The pathophysiological conditions associated with PCOS, such as elevated serum AMH levels and other pathological states, may directly or indirectly influence the AMH-AMHR cascade signaling pathway in the endometrium. This influence could contribute to the progression of AEH.
Collapse
Affiliation(s)
- Yingsha Yao
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China; (Y.Y.); (S.Z.)
| | - Shulan Zhu
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China; (Y.Y.); (S.Z.)
- Hangzhou Linping District Maternal & Child Health Care Hospital, Hangzhou 311199, China
| | - Xiaoming Zhu
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China; (Y.Y.); (S.Z.)
- Women’s Reproductive Health Key Laboratory of Zhejiang Province, Hangzhou 310006, China
| |
Collapse
|
4
|
Oreja-Guevara C, Gónzalez-Suárez I, Bilbao MM, Gómez-Palomares JL, Rodríguez CH, Rabanal A, Benito YA. Multiple sclerosis: Pregnancy, fertility, and assisted reproductive technology-a review. Mult Scler Relat Disord 2024; 92:105893. [PMID: 39393162 DOI: 10.1016/j.msard.2024.105893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Pregnancy is not discouraged in multiple sclerosis (MS) patients. However, MS patients tend to delay motherhood since they must wait for a low clinical and radiological activity before considering pregnancy, which impacts their fertility and their need for assisted reproductive technology (ART). This review aimed to cover, from a multidisciplinary perspective, the most critical aspects revolving around pregnancy in MS patients. METHODS A group of seven experts (four neurologists and three gynaecologists) met for three discussion sessions to review current knowledge on ART in patients with MS. PubMed searches for journal articles published in English or Spanish between 2000 and 2024 were undertaken. 354 articles were revised at the title level. RESULTS We reviewed current evidence on fertility in women and men with MS, on the effects of pregnancy on MS, the disease's pharmacological treatment during pregnancy, MS during delivery and breastfeeding, ART (intrauterine insemination, in vitro fertilisation, intracytoplasmic sperm injection, and oocyte cryopreservation) in patients with MS. CONCLUSION Early family planning, supported by good coordination between neurology and gynaecology departments, is paramount to managing MS women with motherhood desire. Besides, although a well-planned, early pregnancy is always the most desirable outcome, ART is considered safe and valuable for MS patients. Finally, multidisciplinary units are deemed pivotal to guide MS patients with parenthood desire through pregnancy.
Collapse
Affiliation(s)
- Celia Oreja-Guevara
- Department of Neurology, Hospital Clinico San Carlos, IdISSC, Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid (UCM). Madrid, Spain.
| | | | - Mar Mendibe Bilbao
- Neuroscience Department, Biocruces Health Research Institute, Cruces University Hospital, University of the Basque Country, Bilbao, Spain
| | | | | | - Aintzane Rabanal
- Cruces University Hospital, University of the Basque Country, Obstetrics and Gynaecology Department, Human Reproduction Unit, Biocruces Health Research Institute, Bilbao, Spain
| | - Yolanda Aladro Benito
- Departament of Neurology, Research Institute, Hospital Universitario de Getafe, Madrid, Spain
| |
Collapse
|
5
|
Shaji A, Kumaresan A, Sinha MK, Nag P, Patil S, Jeyakumar S, Gowdar Veerappa V, Manimaran A, Ramesha K. Identification of potential differences in salivary proteomic profiles between estrus and diestrus stage of estrous cycle in dairy cows. Syst Biol Reprod Med 2024; 70:204-217. [PMID: 39008339 DOI: 10.1080/19396368.2024.2370328] [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: 07/20/2023] [Accepted: 06/15/2024] [Indexed: 07/16/2024]
Abstract
In the present study, a comparative global high-throughput proteomic analysis strategy was used to identify proteomic differences between estrus and diestrus stage of estrous cycle in dairy cows. Saliva was collected from cows during estrus and diestrus, and subjected to LC-MS/MS-based proteomic analysis. A total of 2842 proteins were detected in the saliva of cows, out of which, 2437 and 1428 non-redundant proteins were identified in estrous and diestrous saliva, respectively. Further, it was found that 1414 and 405 salivary proteins were specific to estrus and diestrus, respectively while 1023 proteins were common to both groups. Among the significantly dysregulated proteins, the expression of 56 proteins was down-regulated (abundance ratio <0.5) while 40 proteins were up-regulated (abundance ratio > 2) in estrous compared to diestrous saliva. The proteins, such as HSD17B12, INHBA, HSP70, ENO1, SRD5A1, MOS, AMH, ECE2, PDGFA, OPRK1, SYN1, CCNC, PLIN5, CETN1, AKR1C4, NMNAT1, CYP2E1, and CYP19A1 were detected only in the saliva samples derived from estrous cows. Considerable number of proteins detected in the saliva of estrous cows were found to be involved in metabolic pathway, PI3K-Akt signaling pathway, toll-like receptor signaling pathway, steroid biosynthesis pathway, insulin signaling pathway, calcium signaling pathway, estrogen signaling pathway, oxytocin signaling pathway, TGF-β signaling pathway and oocyte meiosis. On the other hand, proteins detected in saliva of diestrous cows were involved mainly in metabolic pathway. Collectively, these data provide preliminary evidence of a potential difference in salivary proteins at different stages of estrous cycle in dairy cows.
Collapse
Affiliation(s)
- Arsha Shaji
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Arumugam Kumaresan
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Manish Kumar Sinha
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Pradeep Nag
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Shivanagouda Patil
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Sakthivel Jeyakumar
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Vedamurthy Gowdar Veerappa
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Ayyasamy Manimaran
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Kerekoppa Ramesha
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| |
Collapse
|
6
|
Rotz SJ, Bjornard K, Hampanda K, Kumnick A, Maher JCY, Yu C, Appiah L. Limited Recommendations and Evidence for Timing and Frequency of Anti-Mullerian Hormone Screening in Female Pediatric Cancer Survivors: A Systematic Review from the Pediatric and Adolescent Committee of the Oncofertility Consortium. J Adolesc Young Adult Oncol 2024. [PMID: 39552408 DOI: 10.1089/jayao.2024.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Guidelines regarding the optimal use and timing of anti-Mullerian hormone (AMH) screening in childhood cancer survivors to evaluate for the risk of premature ovarian insufficiency or reduced fertility potential are lacking. We conducted a systematic review of the current evidence supporting AMH screening of female childhood cancer survivors with the overall objective to identify gaps in the literature needing further study, to allow for future data-driven recommendations. Search terms included "cancer, fertility, and anti-Mullerian hormone." We included original research articles that had ≥20 female childhood cancer survivors and excluded studies not including pediatric oncology survivors (≤18 years of age), did not include raw AMH values, were a mixed pediatric/young adult population which were minority pediatric, or did not separate pediatric from adult AMH data. In total, 17 studies (8 case-control, 5 cross-sectional, and 4 longitudinal prospective cohorts), encompassing 1106 total survivors met inclusion criteria and were further evaluated. Three studies evaluated the relationship of AMH to antral follicle count with generally good concordance. Four studies analyzed longitudinal changes in AMH with chemotherapy demonstrating that most patients will have an acute drop in AMH during therapy, and recovery of AMH over time is dependent on treatment intensity. No studies evaluated the optimal timing or interval of AMH testing. AMH correlates well with other markers of ovarian reserve, but there is insufficient data regarding the utility of AMH to predict the ability to conceive or timing of menopause. Optimal AMH screening initiation, duration, and intervals also require further study.
Collapse
Affiliation(s)
- Seth J Rotz
- Department of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kari Bjornard
- Division of Pediatric Hematology, Department of Pediatrics, Oncology and Stem Cell Transplant, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Karen Hampanda
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Allison Kumnick
- Department of Obstetrics and Gynecology, MedStar Washington Hospital Center, Washington, District of Columbia, USA
| | - Jacqueline C Yano Maher
- Pediatric and Adolescent Gynecology, National Institute of Child Health and Human Development, Bethesda, Maryland, USA
- Pediatric and Adolescent Gynecology Program, Children's National Hospital, Washington, District of Columbia, USA
| | - Christine Yu
- Division of Endocrinology, Department of Pediatric Medicine, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Leslie Appiah
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Obstetrics and Gynecology, Children's Hospital Colorado, Denver, Colorado, USA
| |
Collapse
|
7
|
Gu M, Wang Y, Yu Y. Ovarian fibrosis: molecular mechanisms and potential therapeutic targets. J Ovarian Res 2024; 17:139. [PMID: 38970048 PMCID: PMC11225137 DOI: 10.1186/s13048-024-01448-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 07/07/2024] Open
Abstract
Ovarian fibrosis, characterized by the excessive proliferation of ovarian fibroblasts and the accumulation of extracellular matrix (ECM), serves as one of the primary causes of ovarian dysfunction. Despite the critical role of ovarian fibrosis in maintaining the normal physiological function of the mammalian ovaries, research on this condition has been greatly underestimated, which leads to a lack of clinical treatment options for ovarian dysfunction caused by fibrosis. This review synthesizes recent research on the molecular mechanisms of ovarian fibrosis, encompassing TGF-β, extracellular matrix, inflammation, and other profibrotic factors contributing to abnormal ovarian fibrosis. Additionally, we summarize current treatment approaches for ovarian dysfunction targeting ovarian fibrosis, including antifibrotic drugs, stem cell transplantation, and exosomal therapies. The purpose of this review is to summarize the research progress on ovarian fibrosis and to propose potential therapeutic strategies targeting ovarian fibrosis for the treatment of ovarian dysfunction.
Collapse
Affiliation(s)
- Mengqing Gu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yibo Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Institute of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
8
|
Hess MK, Mersha A, Ference SS, Nafziger SR, Keane JA, Fuller AM, Kurz SG, Sutton CM, Spangler ML, Petersen JL, Cupp AS. Puberty classifications in beef heifers are moderately to highly heritable and associated with candidate genes related to cyclicity and timing of puberty. Front Genet 2024; 15:1405456. [PMID: 38939530 PMCID: PMC11208629 DOI: 10.3389/fgene.2024.1405456] [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] [Received: 03/22/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024] Open
Abstract
Introduction: Pubertal attainment is critical to reproductive longevity in heifers. Previously, four heifer pubertal classifications were identified according to attainment of blood plasma progesterone concentrations > 1 ng/ml: 1) Early; 2) Typical; 3) Start-Stop; and 4) Non-Cycling. Early and Typical heifers initiated and maintained cyclicity, Start-Stop started and then stopped cyclicity and Non-Cycling never initiated cyclicity. Start-Stop heifers segregated into Start-Stop-Discontinuous (SSD) or Start-Stop-Start (SSS), with SSD having similar phenotypes to Non-Cycling and SSS to Typical heifers. We hypothesized that these pubertal classifications are heritable, and loci associated with pubertal classifications could be identified by genome wide association studies (GWAS). Methods: Heifers (n = 532; 2017 - 2022) genotyped on the Illumina Bovine SNP50 v2 or GGP Bovine 100K SNP panels were used for variant component estimation and GWAS. Heritability was estimated using a univariate Bayesian animal model. Results: When considering pubertal classifications: Early, Typical, SSS, SSD, and Non-Cycling, pubertal class was moderately heritable (0.38 ± 0.08). However, when heifers who initiated and maintained cyclicity were compared to those that did not cycle (Early+Typical vs. SSD+Non-Cycling) heritability was greater (0.59 ± 0.19). A GWAS did not identify single nucleotide polymorphisms (SNPs) significantly associated with pubertal classifications, indicating puberty is a polygenic trait. A candidate gene approach was used, which fitted SNPs within or nearby a set of 71 candidate genes previously associated with puberty, PCOS, cyclicity, regulation of hormone secretion, signal transduction, and methylation. Eight genes/regions were associated with pubertal classifications, and twenty-two genes/regions were associated with whether puberty was attained during the trial. Additionally, whole genome sequencing (WGS) data on 33 heifers were aligned to the reference genome (ARS-UCD1.2) to identify variants in FSHR, a gene critical to pubertal attainment. Fisher's exact test determined if FSHR SNPs segregated by pubertal classification. Two FSHR SNPs that were not on the bovine SNP panel were selected for additional genotyping and analysis, and one was associated with pubertal classifications and whether they cycled during the trial. Discussion: In summary, these pubertal classifications are moderately to highly heritable and polygenic. Consequently, genomic tools to inform selection/management of replacement heifers would be useful if informed by SNPs associated with cyclicity and early pubertal attainment.
Collapse
Affiliation(s)
- Melanie K. Hess
- Department of Animal Science, University of Nebraska–Lincoln, Lincoln, NE, United States
| | | | | | | | | | | | | | | | | | | | - Andrea S. Cupp
- Department of Animal Science, University of Nebraska–Lincoln, Lincoln, NE, United States
| |
Collapse
|
9
|
Patel RH, Truong VB, Sabry R, Acosta JE, McCahill K, Favetta LA. SMAD signaling pathway is disrupted by BPA via the AMH receptor in bovine granulosa cells†. Biol Reprod 2023; 109:994-1008. [PMID: 37724935 DOI: 10.1093/biolre/ioad125] [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: 02/22/2023] [Revised: 07/18/2023] [Accepted: 09/13/2023] [Indexed: 09/21/2023] Open
Abstract
Significant events that determine oocyte competence occur during follicular growth and oocyte maturation. The anti-Mullerian hormone, a positive predictor of fertility, has been shown to be affected by exposure to endocrine disrupting compounds, such as bisphenol A and S. However, the interaction between bisphenols and SMAD proteins, mediators of the anti-Mullerian hormone pathway, has not yet been elucidated. AMH receptor (AMHRII) and downstream SMAD expression was investigated in bovine granulosa cells treated with bisphenol A, bisphenol S, and then competitively with the anti-Mullerian hormone. Here, we show that 24-h bisphenol A exposure in granulosa cells significantly increased SMAD1, SMAD4, and SMAD5 mRNA expression. No significant changes were observed in AMHRII or SMADs protein expression after 24-h treatment. Following 12-h treatments with bisphenol A (alone or with the anti-Mullerian hormone), a significant increase in SMAD1 and SMAD4 mRNA expression was observed, while a significant decrease in SMAD1 and phosphorylated SMAD1 was detected at the protein level. To establish a functional link between bisphenols and the anti-Mullerian hormone signaling pathway, antisense oligonucleotides were utilized to suppress AMHRII expression with or without bisphenol exposure. Initially, transfection conditions were optimized and validated with a 70% knockdown achieved. Our findings show that bisphenol S exerts its effects independently of the anti-Mullerian hormone receptor, while bisphenol A may act directly through the anti-Mullerian hormone signaling pathway providing a potential mechanism by which bisphenols may exert their actions to disrupt follicular development and decrease oocyte competence.
Collapse
Affiliation(s)
- Rushi H Patel
- Reproductive Health and Biotechnology Lab, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Vivien B Truong
- Reproductive Health and Biotechnology Lab, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Reem Sabry
- Reproductive Health and Biotechnology Lab, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Julianna E Acosta
- Reproductive Health and Biotechnology Lab, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Kiera McCahill
- Reproductive Health and Biotechnology Lab, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Laura A Favetta
- Reproductive Health and Biotechnology Lab, Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
10
|
Coxir SA, Costa GMJ, Santos CFD, Alvarenga RDLLS, Lacerda SMDSN. From in vivo to in vitro: exploring the key molecular and cellular aspects of human female gametogenesis. Hum Cell 2023:10.1007/s13577-023-00921-7. [PMID: 37237248 DOI: 10.1007/s13577-023-00921-7] [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: 03/20/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023]
Abstract
Human oogenesis is a highly complex and not yet fully understood process due to ethical and technological barriers that limit studies in the field. In this context, replicating female gametogenesis in vitro would not only provide a solution for some infertility problems, but also be an excellent study model to better understand the biological mechanisms that determine the formation of the female germline. In this review, we explore the main cellular and molecular aspects involved in human oogenesis and folliculogenesis in vivo, from the specification of primordial germ cells (PGCs) to the formation of the mature oocyte. We also sought to describe the important bidirectional relationship between the germ cell and the follicular somatic cells. Finally, we address the main advances and different methodologies used in the search for obtaining cells of the female germline in vitro.
Collapse
Affiliation(s)
- Sarah Abreu Coxir
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Guilherme Mattos Jardim Costa
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Camilla Fernandes Dos Santos
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | | | - Samyra Maria Dos Santos Nassif Lacerda
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
11
|
Follicular Atresia, Cell Proliferation, and Anti-Mullerian Hormone in Two Neotropical Primates (Aotus nancymae and Sapajus macrocephalus). Animals (Basel) 2023; 13:ani13061051. [PMID: 36978591 PMCID: PMC10044352 DOI: 10.3390/ani13061051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/29/2022] [Accepted: 02/07/2023] [Indexed: 03/17/2023] Open
Abstract
This study evaluated the follicular atresia, cell proliferation, and anti-Mullerian hormone action in Aotus nancymae and Sapajus macrocephalus during three sexual phases (follicular, luteal, and gestational). Follicular quantification and immunolocalization of Caspase-3 protein, B-cell lymphoma 2 (BCL-2), proliferating cell nuclear antigen (PCNA), and anti-Mullerian hormone (AMH) were performed. A significant difference in the quantification between preantral and antral follicles, with a progressive decrease in the antrals, was identified. Protein and hormonal markers varied significantly between follicle cell types (A. nancymae p = 0.001; S. macrocephalus, p = 0.002). Immunostaining in the preantral and antral follicles was present in all sexual phases; for Caspase-3, in granulosa cells, oocytes, and stroma; for BCL-2, in granulosa cells, oocytes, and theca; and for PCNA and AMH, in oocytes and granulosa cells. The immunostaining for Caspase-3 was more expressive in the preantral follicles (follicular phase, p < 0.05), while that for BCL-2 and PCNA was more expressive in the antral follicles of the follicular phase. The AMH was more expressive in the primary and antral follicles of nonpregnant females, in both the follicular and luteal phases. Our results contribute to understanding the ovarian follicular selection, recruitment, and degeneration of these species.
Collapse
|
12
|
Oreja-Guevara C, Rabanal A, Rodríguez CH, Benito YA, Bilbao MM, Gónzalez-Suarez I, Gómez-Palomares JL. Assisted Reproductive Techniques in Multiple Sclerosis: Recommendations from an Expert Panel. Neurol Ther 2023; 12:427-439. [PMID: 36746871 PMCID: PMC10043068 DOI: 10.1007/s40120-023-00439-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is mainly diagnosed in women of reproductive age. However, there is a paucity of guidelines jointly prepared by neurologists and gynaecologists on managing women with MS and the desire for motherhood. Therefore, in this review we propose recommendations for such cases, with an particular focus on those requiring assisted reproductive techniques (ART). METHODS A group of seven MS experts (4 neurologists and 3 gynaecologists) came together for three discussion sessions to achieve consensus. RESULTS The recommendations reported here focus on the importance of early preconception counselling, the management of disease-modifying therapies before and during ART procedures, important considerations for women with MS regarding ART (intrauterine insemination, in vitro fertilisation and oocyte cryopreservation) and the paramount relevance of multidisciplinary units to manage these patients. CONCLUSIONS Early preconception consultations are essential to individualising pregnancy management in women with MS, and an early, well-planned, spontaneous pregnancy should be the aim whenever possible. The management of women with MS and the desire for motherhood by multidisciplinary units is warranted to ensure appropriate guidance through the entire pregnancy.
Collapse
Affiliation(s)
- Celia Oreja-Guevara
- Department of Neurology, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), Hospital Clinico San Carlos, Madrid, Spain. .,Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.
| | - Aintzane Rabanal
- Human Reproduction Unit, Obstetrics and Gynaecology Department, Biocruces Health Research Institute, Cruces University Hospital, University of the Basque Country, Bilbao, Spain
| | | | - Yolanda Aladro Benito
- Department of Neurology, Research Institute, Hospital Universitario de Getafe, Madrid, Spain
| | - Mar Mendibe Bilbao
- Neuroscience Department, Biocruces Health Research Institute, Cruces University Hospital, University of the Basque Country, Bilbao, Spain
| | | | - José Luis Gómez-Palomares
- Wilson Fertiliy-Balearic Center for In Vitro Fertilization CEFIVBA-Wilson Fertility, Mallorca, Spain
| |
Collapse
|
13
|
Wang YC, Ma YD, Liu H, Cui ZH, Zhao D, Zhang XQ, Zhang LX, Guo WJ, Long Y, Tu SS, Yuan DZ, Zhang JH, Wang BK, Xu LZ, Shen QY, Wang Y, Nie L, Yue LM. Hyperandrogen-induced polyol pathway flux increase affects ovarian function in polycystic ovary syndrome via excessive oxidative stress. Life Sci 2023; 313:121224. [PMID: 36435224 DOI: 10.1016/j.lfs.2022.121224] [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: 08/09/2022] [Revised: 11/10/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
AIMS Polycystic ovary syndrome (PCOS) is a common endocrine disorder in the women of childbearing age. It is characterized by hyperandrogenism and abnormal follicular growth and ovulation. The polyol pathway is a glucose metabolism bypass pathway initiated by aldose reductase (ADR). Androgen induces the expression of ADR in the male reproductive tract, which has a general physiological significance for male reproductive function. Here we investigate whether hyperandrogenemia in PCOS leads to increased flux of the polyol pathway in ovarian tissue, which in turn affects follicular maturation and ovulation through oxidative stress. MAIN METHODS We used clinical epidemiological methods to collect serum and granulosa cells from clinical subjects for a clinical case-control study. At the same time, cell biology and molecular biology techniques were used to conduct animal and cell experiments to further explore the mechanism of hyperandrogen-induced ovarian polyol pathway hyperactivity and damage to ovarian function. KEY FINDINGS Here, we find that hyperandrogenism of PCOS can induce the expression of ovarian aldose reductase, which leads to the increase of the polyol pathway flux, and affects ovarian function through excessive oxidative stress. SIGNIFICANCE Our research has enriched the pathological mechanism of PCOS and may provide a new clue for the clinical treatment of PCOS.
Collapse
Affiliation(s)
- Yi-Cheng Wang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China; Department of Reproductive Health and Infertility, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, Sichuan, China
| | - Yong-Dan Ma
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Huan Liu
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhi-Hui Cui
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Dan Zhao
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xue-Qin Zhang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Li-Xue Zhang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wen-Jing Guo
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yun Long
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Sha-Sha Tu
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Dong-Zhi Yuan
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China; Reproductive Endocrinology and Regulation Joint Laboratory, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jin-Hu Zhang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China; Reproductive Endocrinology and Regulation Joint Laboratory, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Bing-Kun Wang
- Reproductive Endocrinology and Regulation Joint Laboratory, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Liang-Zhi Xu
- Reproductive Endocrinology and Regulation Joint Laboratory, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qiong-Yan Shen
- Reproductive Medicine Center, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yan Wang
- Reproductive Medicine Center, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Li Nie
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China; Reproductive Endocrinology and Regulation Joint Laboratory, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Li-Min Yue
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China; Reproductive Endocrinology and Regulation Joint Laboratory, West China Second Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
14
|
He YC, Su KZ, Cai J, Meng QX, Wu YT, Huang HF. Serum anti-Müllerian hormone levels are associated with perinatal outcomes in women undergoing IVF/ICSI: A multicenter retrospective cohort study. Front Endocrinol (Lausanne) 2023; 14:1081069. [PMID: 36896183 PMCID: PMC9990865 DOI: 10.3389/fendo.2023.1081069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
INTRODUCTION Anti-Müllerian hormone (AMH) level has long been considered as a serum biomarker of ovarian reserve clinically, while emerging data suggest that serum AMH level may also predict pregnancy outcomes. However, whether pregestational serum AMH levels are related to perinatal outcomes among women undergoing in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) cycles is unknown. OBJECTIVE To explore the association between different AMH levels and perinatal outcomes in women with live births in IVF/ICSI. METHODS This multicenter retrospective cohort study was conducted among three different provinces in China, from January 2014 to October 2019. A total of 13,763 IVF/ICSI cycles with 5657 live-delivery pregnant women and 6797 newborns were recruited. Participants were categorized into three groups according to the <25th (low), 25 to 75th (average), and >75th (high) percentile of serum AMH concentration. Perinatal outcomes were compared among groups. Subgroup analyses were conducted based on the number of live births. RESULTS Among women with singleton deliveries, low and high AMH levels increased the risk of intrahepatic cholestasis of pregnancy (ICP) (aOR1 = 6.02, 95%CI: 2.10-17.22; aOR2 = 3.65, 95%CI:1.32-10.08) and decreased the risk of macrosomia (aOR1 = 0.65, 95%CI:0.48-0.89; aOR2 = 0.72, 95%CI:0.57-0.96), while low AMH reduced the risk of large for gestational age (LGA, aOR=0.74, 95%CI:0.59-0.93) and premature rupture of membrane (PROM, aOR=0.50, 95%CI:0.31-0.79)compared with the average AMH group. In women with multiple deliveries, high AMH levels increased the risks of gestational diabetes mellitus (GDM, aOR=2.40, 95%CI:1.48-3.91) and pregnancy-induced hypertension (PIH, aOR=2.26, 95%CI:1.20-4.22) compared with the average AMH group, while low AMH levels increased the risk of ICP (aOR=14.83, 95%CI:1.92-54.30). However, there was no evidence of differences in preterm birth, congenital anomaly, and other perinatal outcomes among the three groups in both singleton and multiple deliveries. CONCLUSIONS Abnormal AMH levels increased the risk of ICP regardless of the number of live births for women undergoing IVF/ICSI, while high AMH levels increased the risks of GDM and PIH in multiple deliveries. However, serum AMH levels were not associated with adverse neonatal outcomes in IVF/ICSI. The underlying mechanism warrants further investigation.
Collapse
Affiliation(s)
- Yi-Chen He
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Kai-Zhen Su
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Cai
- Department of Reproductive Medicine, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Qing-Xia Meng
- Center of Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Yan-Ting Wu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: He-Feng Huang, ; Yan-Ting Wu,
| | - He-Feng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: He-Feng Huang, ; Yan-Ting Wu,
| |
Collapse
|
15
|
Zaami S, Melcarne R, Patrone R, Gullo G, Negro F, Napoletano G, Monti M, Aceti V, Panarese A, Borcea MC, Scorziello C, Ventrone L, Mamedov SN, Meggiorini ML, Vergine M, Giacomelli L. Oncofertility and Reproductive Counseling in Patients with Breast Cancer: A Retrospective Study. J Clin Med 2022; 11:jcm11051311. [PMID: 35268402 PMCID: PMC8911138 DOI: 10.3390/jcm11051311] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/25/2022] Open
Abstract
Introduction. Improving the prognosis of breast cancer patients is of utmost importance in terms of increasing survival rates. Modern medicine has therefore prioritized better quality of life for patients, even after the disease, through a better management of the potential long-term side effects induced by anticancer treatments. Fertility preservation and family planning are therefore crucial issues to be addressed in all cancer patients of reproductive age. Along those lines, a new branch of medicine with distinct multidisciplinary characteristics has developed over the years: oncofertility. Although both national and international guidelines value reproductive counseling as an essential aspect of the diagnostic-therapeutic pathway, part and parcel of the informed consent process, it is not included within the protocols adopted by the operating units for the care and management of neoplastic diseases. Objective. This study aimed to evaluate the activity of the Breast Unit of the Policlinico Umberto I Hospital, Rome, Italy, and the degree of compliance with guidelines. By knowing the strengths and weaknesses of such approaches, the standards of care offered to breast cancer patients can be improved. Materials and methods. A retrospective study based on a review of medical records was conducted between 2014 and 2021. Patients under 40 years of age diagnosed with non-metastatic malignancies were included who received chemotherapy treatment, namely neoadjuvant, adjuvant or adjuvant hormone therapy. Results. The data were extracted from the medical records of 51 patients who met the inclusion criteria, 41% of whom received reproductive counseling, and of these, 43% decided to undertake a path of fertility preservation. Factors such as the absence of children and young age reportedly favored both the interest in counseling proposals by the medical staff and the decision to undertake a path of fertility preservation. Conclusions. The study shows that there has been growing interest in the topic of oncofertility, especially in light of law 219/2017. Therefore, since 2018, multiple proposals for reproductive counseling have been set forth, but there was not an equally growing demand for fertility preservation practices, which can be explained by the invasive nature of such practices, the patients’ concern about their own state of health, and poor or inadequate information. Such impediments highlight the importance of standardized counseling and the need for a multidisciplinary medical team to support the patient in the decision-making process. The study also revealed a drop in the number of patients receiving counseling due to the COVID-19 pandemic, contrary to the positive trend that was recorded prior to the pandemic.
Collapse
Affiliation(s)
- Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (F.N.); (G.N.)
- Correspondence:
| | - Rossella Melcarne
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (R.M.); (M.C.B.); (C.S.); (L.V.); (S.N.M.); (M.V.); (L.G.)
| | - Renato Patrone
- ICTUS, University of Naples Federico II, 80131 Naples, Italy;
| | - Giuseppe Gullo
- In Vitro Fertilization Unit, Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy;
| | - Francesca Negro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (F.N.); (G.N.)
| | - Gabriele Napoletano
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (F.N.); (G.N.)
| | - Marco Monti
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (M.M.); (M.L.M.)
| | - Valerio Aceti
- Department of Translational and Precision Medicine, “Sapienza” University of Rome, 00161 Rome, Italy;
| | - Alessandra Panarese
- General and Transplant Surgery Department, Dipartimento di Scienze Cliniche Applicate e Biotecnologiche (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy;
| | - Maria Carola Borcea
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (R.M.); (M.C.B.); (C.S.); (L.V.); (S.N.M.); (M.V.); (L.G.)
| | - Chiara Scorziello
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (R.M.); (M.C.B.); (C.S.); (L.V.); (S.N.M.); (M.V.); (L.G.)
| | - Luca Ventrone
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (R.M.); (M.C.B.); (C.S.); (L.V.); (S.N.M.); (M.V.); (L.G.)
| | - Samira Nicole Mamedov
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (R.M.); (M.C.B.); (C.S.); (L.V.); (S.N.M.); (M.V.); (L.G.)
| | - Maria Letizia Meggiorini
- Department of Maternal and Child Health and Urological Sciences, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (M.M.); (M.L.M.)
| | - Massimo Vergine
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (R.M.); (M.C.B.); (C.S.); (L.V.); (S.N.M.); (M.V.); (L.G.)
| | - Laura Giacomelli
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (R.M.); (M.C.B.); (C.S.); (L.V.); (S.N.M.); (M.V.); (L.G.)
| |
Collapse
|
16
|
The Influence of Cesarean Delivery on Ovarian Reserve: a Prospective Cohort Study. Reprod Sci 2021; 29:639-645. [PMID: 34472035 DOI: 10.1007/s43032-021-00730-z] [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: 06/06/2021] [Accepted: 08/26/2021] [Indexed: 11/27/2022]
Abstract
To assess the association between cesarean delivery and ovarian reserve, as compared to vaginal delivery. A prospective case control study conducted at a single tertiary medical center between June 2018 and June 2019. Study population included women with singleton pregnancy that underwent first cesarean delivery that were compared to women undergoing normal vaginal delivery. Women with low ovarian reserve, endometriosis, previous pelvic surgery, chronic maternal disease, and active labor were excluded. Ovarian reserve was estimated by Anti-Mullerian hormone (AMH) levels that was determined twice for each participant: up to a week before and 3 months after delivery. Primary outcome was defined as the delta in AMH levels. Data were analyzed by non-parametric tests. During the study period, 135 women were enrolled, of them 63 (47%) underwent cesarean delivery and 72 (53%) had vaginal delivery. Women in the cesarean delivery group were older (34 (31-38) vs. 32 (29-35); p = 0.001); nevertheless, AMH levels measured before delivery were comparable between the two groups (0.92 (0.51-1.79) vs. 0.95 (0.51-1.79) pg/mL; p = 0.42). AMH levels measured after delivery were more than doubled in the study and control groups (2.15 (1.24-3.05) vs. 2.62 (1.05-5.09); p = 0.50), and delta AMH levels were also found comparable (1.25 (0.61-2.22) vs. 1.59 (0.63-3.41), respectively; p = 0.43). Linear regression analysis including age, mode of delivery, gestational age at delivery, and delta hemoglobin levels revealed that only maternal age was significantly associated with delta in AMH levels (B = - 0.09, p = 0.04). Cesarean delivery does not decrease ovarian reserve as estimated by AMH.
Collapse
|
17
|
The impact of isotretinoin on the pituitary-ovarian axis: An interpretative review of the literature. Reprod Toxicol 2021; 104:85-95. [PMID: 34224824 DOI: 10.1016/j.reprotox.2021.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022]
Abstract
Isotretinoin (13-cis-retinoic acid), a derivative of vitamin A, is used in the treatment of severe acne resulting in sebum suppression induced by sebocyte apoptosis. Isotretinoin treatment is associated with several adverse effects including teratogenicity, hepatotoxicity, and dyslipidemia. Isotretinoin's effects on endocrine systems and its potential role as an endocrine disruptor are not yet adequately investigated. This review presents clinical, endocrine, and molecular evidence showing that isotretinoin treatment adversely affects the pituitary-ovarian axis and enhances the risk of granulosa cell apoptosis reducing follicular reserve. Isotretinoin is associated with pro-apoptotic signaling in sebaceous glands through upregulated expression of p53, forkhead box O transcription factors (FOXO1, FOXO3), and tumor necrosis factor-related apoptosis inducing ligand (TRAIL). Two literature searches including clinical and experimental studies respectively support the hypothesis that isotretinoin's toxicological mode of action on the pituitary-ovarian axis might be caused by over-expressed p53/FOXO1 signaling resulting in gonadotropin suppression and granulosa cell apoptosis. The reduction of follicular reserve by isotretinoin treatment should be especially considered when this drug will be administered for the treatment of acne in post-adolescent women, in whom fertility may be adversely affected. In contrast, isotretinoin treatment may exert beneficial effects in states of hyperandrogenism, especially in patients with polycystic ovary syndrome.
Collapse
|
18
|
Miner AE, Graves JS. What telomeres teach us about MS. Mult Scler Relat Disord 2021; 54:103084. [PMID: 34371369 DOI: 10.1016/j.msard.2021.103084] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/14/2021] [Accepted: 06/09/2021] [Indexed: 02/03/2023]
Abstract
While the precise mechanisms driving progressive forms of MS are not fully understood, patient age has clear impact on disease phenotype. The very young with MS have high relapse rates and virtually no progressive disease, whereas older patients tend to experience more rapid disability accumulation with few relapses. Defining a patient's biological age may offer more precision in determining the role of aging processes in MS phenotype and pathophysiology than just working with an individual's birthdate. The most well recognized measurement of an individual's "biological clock" is telomere length (TL). While TL may differ across tissue types in an individual, most cells TL correlate well with leukocyte TL (LTL), which is the most common biomarker used for aging. LTL has been associated with risk for aging related diseases and most recently with higher levels of disability and brain atrophy in people living with MS. LTL explains 15% of the overall association of chronological age with MS disability level. While LTL may be used just as a biomarker of overall somatic aging processes, triggering of the DNA damage response by telomere attrition leads to senescence pathways that are likely highly relevant to a chronic autoimmune disease. Considering reproductive aging factors, particularly ovarian aging in women, which correlates with LTL and oocyte telomere length, may complement measurements of somatic aging in understanding MS progression. The key to stopping non-relapse related progression in MS might lie in targeting pathways related to biological aging effects on the immune and nervous systems.
Collapse
Affiliation(s)
- Annalise E Miner
- Department of Neurosciences, University of California, San Diego, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California, San Diego, USA.
| |
Collapse
|
19
|
Khaghani AJ, Farrokh P, Zavareh S. Epigenetic effects of Bisphenol A on granulosa cells of mouse follicles during in vitro culture: An experimental study. Int J Reprod Biomed 2021; 19:129-136. [PMID: 33718757 PMCID: PMC7922291 DOI: 10.18502/ijrm.v19i2.8471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 05/04/2020] [Accepted: 08/04/2020] [Indexed: 11/24/2022] Open
Abstract
Background Bisphenol A (BPA), a synthetic endocrine-disrupting chemical, is a reproductive toxicant. Granulosa cells have significant roles in follicle development, and KIT ligand (KITL) and Anti-Müllerian hormone (AMH) are essential biomolecules produced by them during folliculogenesis. Objective Due to the widespread use of BPA and its potential epigenetic effects, this study examined the impact of BPA on promoter methylation of amh and kitl genes in mouse granulosa cells. Materials and Methods Preantral follicles were isolated from ovaries of immature mice and cultured for eight days. Then, follicles were treated with 50 and 100 μM of BPA, and 0.01% (v/v) ethanol for 24 and 72 hr. Growth and degeneration of follicles and antrum formation were analyzed. The granulosa cells were isolated mechanically, and their extracted DNA was treated with sodium bisulfite. The promoter regions of the amh and kitl were analyzed with PCR and sequencing. Results BPA did not change follicle survival and antrum formation significantly (p = 0.41). However, the culture in the presence of 100 μM BPA had an inhibitory effect on growth. Before BPA treatment, the CpG of the kitl and amh promoters were unmethylated and partially methylated, respectively. While the percent of 5mC in the amh promoter reduced at 100 μM of BPA, it did not alter the kitl promoter methylation. Conclusion BPA at higher concentrations has an inhibitory effect on follicle growth. Moreover, it seems that the epigenetic impact of BPA restricts to the demethylation of CpG sites.
Collapse
Affiliation(s)
| | - Parisa Farrokh
- School of Biology, Damghan University, Damghan, Iran.,Institute of Biological Sciences, Damghan University, Damghan, Iran
| | - Saeed Zavareh
- School of Biology, Damghan University, Damghan, Iran.,Institute of Biological Sciences, Damghan University, Damghan, Iran
| |
Collapse
|
20
|
Punchoo R, Bhoora S. Variation in the Measurement of Anti-Müllerian Hormone - What Are the Laboratory Issues? Front Endocrinol (Lausanne) 2021; 12:719029. [PMID: 34539570 PMCID: PMC8446602 DOI: 10.3389/fendo.2021.719029] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022] Open
Abstract
Anti-Müllerian Hormone (AMH) is a 140 kDa homodimeric glycoprotein consisting of two identical subunits linked by disulphide bonds and is synthesised by the testes and ovaries. Its clinical applications are prediction of ovarian response and gonadotropin dose selection upon in vitro fertilization. In males, AMH is used to investigate sexual developmental disorders and gonadal function. AMH is commonly assayed by enzyme-linked immunosorbent assay or automated immunoassay formats that show variation between methods. This review applies fundamental chemical pathology concepts to explain the observed analytical variation of AMH measurement. We examine the lack of standardisation between AMH assays, the impact of antibody design on variable measurements, consider the analytical detection of AMH isoforms, review analytical interference in AMH measurement, and briefly assess systematic bias between AMH assays. The improved attempt at standardising AMH measurement by the recent approval of a WHO Reference Reagent offers promise for harmonising immunoassay results and establishing consensus medical cut-off points for AMH in disease. Standardisation, however, will need to redress the issue of poor commutability of standard reference material and further assign a standard reference procedure to quantify AMH standard reference material. The improvement of the analytical phase of AMH testing will support harmonised method development and patient care.
Collapse
Affiliation(s)
- Rivak Punchoo
- Tshwane Academic Division, National Health Laboratory Service, Pretoria, South Africa
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- *Correspondence: Rivak Punchoo,
| | - Sachin Bhoora
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
21
|
Kim J, You S. After cyclophosphamide exposure, granulosa cells recover their anti-müllerian hormone-producing ability but not their numbers. Cytometry A 2020; 99:807-813. [PMID: 33342073 PMCID: PMC8451832 DOI: 10.1002/cyto.a.24297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022]
Abstract
Anti‐müllerian hormone (AMH) produced by granulosa cells (GCs), reserves the ovarian follicle pool for future recruitment and ovulation. However, women who have undergone cyclophosphamide (Cy) treatment have decreased AMH levels due to damaged GCs. This study establishes flow cytometry protocols for identification of GCs and investigates the cause of the Cy‐induced AMH decrease by analyzing the number of GCs and their AMH production at the single cell level. Over 2 weeks, C57BL/6 mice were intraperitoneally injected 6 times with 100 mg/kg Cy and sacrificed either immediately or 4 weeks after Cy treatment. Twenty‐four hours post‐Cy exposure, a decrease in serum AMH levels was seen due to a reduction in the number of follicle‐stimulating hormone receptor (FSHR)+AMH+ GCs and their ability to produce AMH. However, 4 weeks after Cy treatment, serum AMH levels were still decreased due to the decreased number of FSHR+AMH+ GCs, however, their AMH‐producing ability was unaltered. Consistently, in vitro, Cy‐induced low AMH production in FSHR+AMH+ hGL5 cells (immortalized human GCs) was restored 24 h after Cy treatment, although their numbers remained decreased. Thus, the surviving GCs after Cy exposure had intact AMH‐producing ability. In future, an effort to minimize GC death by Cy treatment is required, while maintaining its therapeutic effects.
Collapse
Affiliation(s)
- Jihyun Kim
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Sooseong You
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| |
Collapse
|
22
|
Hashemian Z, Afsharian P, Farzaneh P, Eftekhari-Yazdi P, Vakhshiteh F, Daneshvar Amoli A, Nasimian A. Establishment and characterization of a PCOS and a normal human granulosa cell line. Cytotechnology 2020; 72:10.1007/s10616-020-00426-3. [PMID: 32989584 PMCID: PMC7695766 DOI: 10.1007/s10616-020-00426-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/16/2020] [Accepted: 09/20/2020] [Indexed: 11/29/2022] Open
Abstract
Oocyte maturation is an important phase in fertility and any disorder in this process could lead to infertility. The most common disorder during folliculogenesis is polycystic ovary syndrome (PCOS). Due to the secretive activity of granulosa cells (GCs), they play a vital role in folliculogenesis. Although scientists use various cellular and molecular methods to have a better understanding of the mechanism of these cells, some limitations still exist in GC culture such as low primary cell yield and proliferation capability. Therefore, immortalization of primary cells is an approach to overcome these limitations. In the current study, GCs were obtained from two females, one with PCOS and one with normal folliculogenesis. In the first stage, we established two human GC (hGC) lines by immortalizing them through retrovirus-mediated transfer of the human telomerase reverse transcriptase (hTERT) and c-Myc genes. Subsequently, the normal and PCOS cell lines were characterized and were investigated for their growth features. The cell lines were also examined in terms of immortal markers of hTERT, follicle stimulating hormone receptor (FSHR), aromatase, anti-Müllerian hormone (AMH), growth differentiation factor 9 (GDF9), bone morphogenetic protein 15 (BMP15), estrogen, and progesterone. Our results indicated that the normal and PCOS cell lines both showed similar characteristics to GCs during the follicular stage in normal and PCOS women. The normal and PCOS cell lines demonstrate molecular mechanisms similar to that of GCs such as folliculogenesis, oogenesis, and steroidogenesis, which enable researchers to perform further investigations in future.
Collapse
Affiliation(s)
- Zohreh Hashemian
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Parvaneh Afsharian
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Parvaneh Farzaneh
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Faezeh Vakhshiteh
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | | | - Ahmad Nasimian
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran.
| |
Collapse
|
23
|
Liu X, Xiao H, Jie M, Dai S, Wu X, Li M, Wang D. Amh regulate female folliculogenesis and fertility in a dose-dependent manner through Amhr2 in Nile tilapia. Mol Cell Endocrinol 2020; 499:110593. [PMID: 31560938 DOI: 10.1016/j.mce.2019.110593] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 10/26/2022]
Abstract
In the present study, Amh was found to be abundantly expressed in the granulosa cells of the primary growth follicles, and Amhr2 in the granulosa cells, oogonia and phase I oocytes in tilapia by immunohistochemistry. In addition, Amh and Amhr2 were also found to be expressed in the brain and pituitary. Heterozygous mutation of either amh or amhr2 resulted in increased primary growth follicles and decreased fertility, and homozygous mutation resulted in hypertrophic ovaries with significantly increased primary follicles and failed transition from primary to vitellogenic follicles. Expression of gnrh3 in the brain, fsh and lh in the pituitary and serum E2 concentration were significantly decreased in both mutants. Significantly increased apoptosis of follicle cells was observed in both mutants. However, administration of E2 failed to rescue the folliculogenesis defects of the mutants. Our results suggested that Amh acts in a dose-dependent manner by binding Amhr2 in tilapia.
Collapse
Affiliation(s)
- Xingyong Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hesheng Xiao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Mimi Jie
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Shengfei Dai
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Xin Wu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Minghui Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
24
|
Puttabyatappa M, Matiller V, Stassi AF, Salvetti NR, Ortega HH, Padmanabhan V. Developmental Programming: Prenatal Testosterone Excess on Ovarian SF1/DAX1/FOXO3. Reprod Sci 2020; 27:342-354. [PMID: 32046386 DOI: 10.1007/s43032-019-00029-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/09/2019] [Indexed: 12/22/2022]
Abstract
Prenatal testosterone (T) excess, partly via androgenic programming, enhances follicular recruitment/persistence in sheep as in women with polycystic ovarian syndrome (PCOS). Decreased anti-Mullerian hormone (AMH) in early growing and increased AMH in antral follicles may underlie enhanced recruitment and persistence, respectively. Changes in AMH may be mediated by steroidogenic factor 1 (SF1), an enhancer of AMH, and dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1 (DAX1), that antagonizes SF1. Another mediator could be forkhead box 03 (FOXO3) which regulates follicular recruitment/atresia. To test if androgen-programmed changes in SF1, DAX1, and FOXO3 proteins contribute to follicular defects in prenatal T-treated sheep, ovaries from control, prenatal T-, and dihydrotestosterone (DHT)-treated (days 30-90 of gestation) animals at fetal day (FD) 90, FD140, and 1 and 2 years-of-age were studied. Prenatal T increased DAX1 in granulosa cells of primordial through large preantral and theca cells of large preantral follicles at FD140 and increased SF1 in the granulosa cells of preantral and antral and theca cells of large preantral follicle at 2 years-of-age. Prenatal T increased FOXO3 only in theca cells of preantral (FD140) and antral (2 years-of-age) follicles. Prenatal DHT increased DAX1 in granulosa cells from small preantral follicles at FD140 while increasing SF1 in granulosa cells from antral follicles at 1 year-of-age. These age-dependent changes in DAX1/SF1 partly via androgen-programming are consistent with changes in AMH and may contribute to the enhanced follicular recruitment/persistence, and multifollicular phenotype of prenatal T-treated females and may be of translational relevance to PCOS.
Collapse
Affiliation(s)
- Muraly Puttabyatappa
- Department of Pediatrics and the Reproductive Sciences Program, University of Michigan, Room 7510 MSRB I, 1150 Medical Center Drive, Ann Arbor, MI, 48109-5718, USA
| | - Valentina Matiller
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina
| | - Antonela F Stassi
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina
| | - Natalia R Salvetti
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina
| | - Hugo H Ortega
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL) - Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina
| | - Vasantha Padmanabhan
- Department of Pediatrics and the Reproductive Sciences Program, University of Michigan, Room 7510 MSRB I, 1150 Medical Center Drive, Ann Arbor, MI, 48109-5718, USA.
| |
Collapse
|
25
|
Rodriguez A, Briley SM, Patton BK, Tripurani SK, Rajapakshe K, Coarfa C, Rajkovic A, Andrieux A, Dejean A, Pangas SA. Loss of the E2 SUMO-conjugating enzyme Ube2i in oocytes during ovarian folliculogenesis causes infertility in mice. Development 2019; 146:dev.176701. [PMID: 31704792 PMCID: PMC6918767 DOI: 10.1242/dev.176701] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 10/29/2019] [Indexed: 01/25/2023]
Abstract
The number and quality of oocytes within the ovarian reserve largely determines fertility and reproductive lifespan in mammals. An oocyte-specific transcription factor cascade controls oocyte development, and some of these transcription factors, such as newborn ovary homeobox gene (NOBOX), are candidate genes for primary ovarian insufficiency in women. Transcription factors are frequently modified by the post-translational modification SUMOylation, but it is not known whether SUMOylation is required for function of the oocyte-specific transcription factors or if SUMOylation is required in oocytes during their development within the ovarian follicle. To test this, the sole E2 SUMO-conjugating enzyme, Ube2i, was ablated in mouse oocytes beginning in primordial follicles. Loss of oocyte Ube2i resulted in female infertility with major defects in stability of the primordial follicle pool, ovarian folliculogenesis, ovulation and meiosis. Transcriptomic profiling of ovaries suggests that loss of oocyte Ube2i caused defects in both oocyte- and granulosa cell-expressed genes, including NOBOX and some of its known target genes. Together, these studies show that SUMOylation is required in the mammalian oocyte during folliculogenesis for both oocyte development and communication with ovarian somatic cells.
Collapse
Affiliation(s)
- Amanda Rodriguez
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shawn M. Briley
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bethany K. Patton
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Swamy K. Tripurani
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kimal Rajapakshe
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aleksander Rajkovic
- Department of Pathology, University of California, San Francisco, CA 94134, USA
| | - Alexandra Andrieux
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Pasteur Institute, 75015 Paris, France
| | - Anne Dejean
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Pasteur Institute, 75015 Paris, France
| | - Stephanie A. Pangas
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA,Author for correspondence ()
| |
Collapse
|
26
|
Valer JA, Sánchez-de-Diego C, Pimenta-Lopes C, Rosa JL, Ventura F. ACVR1 Function in Health and Disease. Cells 2019; 8:cells8111366. [PMID: 31683698 PMCID: PMC6912516 DOI: 10.3390/cells8111366] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Activin A receptor type I (ACVR1) encodes for a bone morphogenetic protein type I receptor of the TGFβ receptor superfamily. It is involved in a wide variety of biological processes, including bone, heart, cartilage, nervous, and reproductive system development and regulation. Moreover, ACVR1 has been extensively studied for its causal role in fibrodysplasia ossificans progressiva (FOP), a rare genetic disorder characterised by progressive heterotopic ossification. ACVR1 is linked to different pathologies, including cardiac malformations and alterations in the reproductive system. More recently, ACVR1 has been experimentally validated as a cancer driver gene in diffuse intrinsic pontine glioma (DIPG), a malignant childhood brainstem glioma, and its function is being studied in other cancer types. Here, we review ACVR1 receptor function and signalling in physiological and pathological processes and its regulation according to cell type and mutational status. Learning from different functions and alterations linked to ACVR1 is a key step in the development of interdisciplinary research towards the identification of novel treatments for these pathologies.
Collapse
Affiliation(s)
- José Antonio Valer
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Cristina Sánchez-de-Diego
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| |
Collapse
|
27
|
Karakas Alkan K, Ceylan A, Alkan H, Ozen D, Bayraktaroglu AG, Kaymaz M. Immunohistochemical and qPCR determination of the expression and serum level of anti-Müllerian hormone in pre-pubertal, intact and ovarian remnant syndrome detected bitches. Reprod Domest Anim 2019; 54:979-986. [PMID: 31050839 DOI: 10.1111/rda.13451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/26/2019] [Indexed: 11/30/2022]
Abstract
The aim of this study was to determine the serum concentrations, ovarian presence and expression of anti-Müllerian hormone (AMH) in pre-pubertal, bitches with signs of ovarian remnant syndrome (ORS) and intact bitches. In addition, we aimed to verify the suitability of serum AMH concentrations for diagnostic purposes in sterilized bitches and/or in suspected cases of ORS in the field of veterinary medicine. For this purpose, 36 healthy female dogs divided into six groups: proestrus, oestrus, dioestrus, anoestrus, pre-pubertal and ORS. Serum AMH concentrations were determined by electrochemiluminescence immunoassay, and ovarian presence and distribution of AMH was confirmed by immunohistochemical and qPCR techniques. According to the results of qPCR, while the expression values of AMH were at the highest concentrations in the proestrus and oestrus, there was a statistically significant decrease in these values at the later stages of the cycle (p < 0.05). According to hormone analysis, the serum AMH values of the ORS group had decreased significantly compared with the proestrus and oestrus (p < 0.05). Although serum AMH levels of ORS group were increased compared with anestrus and pre-pubertal groups, this increase was statistically non-significant (p > 0.05). Immunohistochemically, AMH expression was first observed in the granulosa cells of primordial follicles in folliculogenesis. Expression values were the highest in the proestrous and oestrus groups, but values from bitches in later stages of the cycle were statistically significant decrease in comparison with these groups (p < 0.05). As a result, AMH concentration and expression were found to be higher in proestrus and oestrus than in other periods (p < 0.05). In addition, the measurable level of AMH concentration in bitches with ORS is an indication that it can be used in the diagnosis of ORS.
Collapse
Affiliation(s)
- Kubra Karakas Alkan
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Ahmet Ceylan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Hasan Alkan
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Selcuk University, Konya, Turkey
| | - Dogukan Ozen
- Department of Biostatistics, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Alev Gurol Bayraktaroglu
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Mustafa Kaymaz
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
28
|
Oskayli MC, Gulcin N, Ozatman E, Gercel G, Mutus M, Aksu B, Durakbasa CU. Assessment of ovarian reserve using serum anti-Müllerian hormone after ovarian torsion surgery. Pediatr Int 2019; 61:504-507. [PMID: 30825401 DOI: 10.1111/ped.13818] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/30/2018] [Accepted: 02/05/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND The aim of this study was to determine ovarian reserve using serum anti-Müllerian hormone (AMH) level in children who had undergone either ovarian-preserving surgery or oophorectomy because of ovarian torsion. METHODS Patients aged > 10 years who had undergone surgery for unilateral ovarian torsion were contacted for the study with ethics committee approval. Seventeen patients agreed to be included. RESULTS A total of 10 patients had undergone ovarian detorsion and seven had undergone oophorectomy. Mean age at operation was 11.6 ± 2.23 years (range, 8-15 years) and 13.2 ± 2.17 years (range, 10-16 years), respectively (P = 0.46). Ovarian torsion was isolated in four patients in the first group, and in three in the second. The remainder had associated benign masses. At the time of this study, mean patient age was 18 ± 2.11 years (range, 14-21 years) with a mean postoperative follow up of 5.9 ± 2.8 years (range, 2-10.5 years). Echogenicity of all preserved ovaries was normal on pelvic Doppler ultrasonography, with presence of antral follicles in six. Three ovaries were smaller than expected for age, although two of these had antral follicles. Mean AMH was 5.54 ± 2.25 ng/mL in the detorsion group and 2.70 ± 2.11 ng/mL in the oophorectomy group (P = 0.04). CONCLUSIONS The presence of follicles in preserved ovaries after detorsion has been reported previously. AMH is expressed in granulosa cells of growing follicles and its serum level is valuable in assessing the quantitative aspects of ovarian reserve. Preservation of the ovary in children with torsion is justified in terms of future ovarian reserve.
Collapse
Affiliation(s)
- Meltem Caglar Oskayli
- Department of Pediatric Surgery, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Neslihan Gulcin
- Department of Pediatric Surgery, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Erdem Ozatman
- Department of Pediatric Surgery, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Gonca Gercel
- Department of Pediatric Surgery, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Murat Mutus
- Department of Pediatric Surgery, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Burhan Aksu
- Department of Pediatric Surgery, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Cigdem Ulukaya Durakbasa
- Department of Pediatric Surgery, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
29
|
Harzif AK, Wiweko B, Addina P, Iswaranti K, Silvia M, Mariana A, Mutia K, Sumapraja K, Muharam R, Pratama G. Anti-Mullerian hormone levels in female cancer patients of reproductive age in Indonesia: A cross-sectional study. F1000Res 2019; 8:159. [PMID: 32185016 PMCID: PMC7059784 DOI: 10.12688/f1000research.15728.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2020] [Indexed: 11/25/2022] Open
Abstract
Background: Efforts in reproductive preservation for cancer patients have become one of the important aspects of cancer management. In fact, decline in reproductive function is known to occur after exposure to anti-cancer treatments. Measuring anti-Müllerian hormone (AMH) levels is known to be the best parameter in predicting ovarian reserves, which indicates reproductive function. In total, 68% of cancer survivors of reproductive age who underwent anti-cancer treatments suffer from infertility. Meanwhile, ovarian reserves also decrease with increasing age. There is ongoing debate on whether the ovarian reserves of cancer patients could be reduced long before exposure to anti-cancer therapy. Therefore, it is important to know whether ovarian reserves in cancer patients decrease before or after anti-cancer therapy. This can help predict the reproductive function in such cases and the effectiveness of ovarian preservation efforts. Methods: A cross-sectional study was conducted, comparing the AMH levels of 44 female cancer patients of reproductive age before cancer therapy, to 44 non-cancer patients of reproductive age (age matched) . The AMH was determined from blood.The biological ages from both groups were adjusted using the Indonesian Kalkulator of Oocytes. Results: The median age in both groups was 28 years old. The AMH levels in the blood of the cancer group were found to be significantly lower in contrast to those in the non-cancer group (1.11 [0.08-4.65] ng/ml vs. 3.99 [1.19- 8.7]; p- value <0.001). Therefore, the biological age in the cancer group was 10 years older than that of the non-cancer group, indicating that ovarian aging occurs earlier in cancer patients. Conclusions: AMH levels of cancer patients of reproductive age were already reduced before cancer therapy, given an older biological age, in contrast to that of the non-cancer patients. Proper counseling and implementation of fertility-preserving methods is highly recommended in this group of patients.
Collapse
Affiliation(s)
- Achmad Kemal Harzif
- Division of Reproductive Endocrionolgy and Infertility, Department of Obstetrics and Gynecology Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Budi Wiweko
- Division of Reproductive Endocrionolgy and Infertility, Department of Obstetrics and Gynecology Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Putri Addina
- Department of Obstetrics and Gynecology Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Kartika Iswaranti
- Department of Obstetrics and Gynecology Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Melisa Silvia
- Indonesian Reproductive Medicine Research and Training Center (INA-REPROMED) of Faculty of Medicine Universitas Indonesia,, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Ana Mariana
- Indonesian Reproductive Medicine Research and Training Center (INA-REPROMED) of Faculty of Medicine Universitas Indonesia,, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Kresna Mutia
- Indonesian Reproductive Medicine Research and Training Center (INA-REPROMED) of Faculty of Medicine Universitas Indonesia,, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Kanadi Sumapraja
- Division of Reproductive Endocrionolgy and Infertility, Department of Obstetrics and Gynecology Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - R Muharam
- Division of Reproductive Endocrionolgy and Infertility, Department of Obstetrics and Gynecology Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| | - Gita Pratama
- Division of Reproductive Endocrionolgy and Infertility, Department of Obstetrics and Gynecology Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, DKI Jakarta Province, Indonesia
| |
Collapse
|
30
|
Żelaźniewicz A, Bielawski T, Nowak J, Pawłowski B. Body symmetry and reproductive hormone levels in women. Women Health 2018; 59:391-405. [PMID: 29979937 DOI: 10.1080/03630242.2018.1492499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Fluctuating asymmetry (FA), a morphological marker of developmental stability, may be related to an individual's biological condition, e.g., health or fertility. The aim of this study was to test if the level of a woman's FA was related to her fertility and reproductive potential as measured by reproductive hormone levels. Fifty-three healthy, non-pregnant, naturally cycling women (mean age = 23.42, SD = 1.85 years), participated in the study, conducted in Wrocław (Poland) in May 2015. Early-follicular phase serum levels of anti-Müllerian hormone (AMH), follicle-stimulating hormone (FSH), luteinizing hormone (LH), and estradiol (E2) were measured. FA was calculated based on anthropometric measures of six bilateral body traits, and the composite FA index was used in statistical analyses. No relationship was observed between FA and the levels of FSH, LH, and AMH (p > .05), controlled for potential confounders. However, the level of E2 was positively correlated with FA (p < .05). Thus, in young women, FA was not related to hormones levels related to ovarian reserve, but more symmetrical women had lower E2 levels. As FA is an index of developmental stability, environmental, and genetic stress, the results of the study confirm previous research suggesting that developmental conditions may be related to women's endogenous estrogen levels.
Collapse
Affiliation(s)
| | - Tomasz Bielawski
- a Department of Human Biology , University of Wrocław , Wrocław , Poland
| | - Judyta Nowak
- a Department of Human Biology , University of Wrocław , Wrocław , Poland
| | - Bogusław Pawłowski
- a Department of Human Biology , University of Wrocław , Wrocław , Poland
| |
Collapse
|
31
|
Oocyte stage-specific effects of MTOR determine granulosa cell fate and oocyte quality in mice. Proc Natl Acad Sci U S A 2018; 115:E5326-E5333. [PMID: 29784807 PMCID: PMC6003357 DOI: 10.1073/pnas.1800352115] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
MTOR (mechanistic target of rapamycin), an integrator of pathways important for cellular metabolism, proliferation, and differentiation, is expressed at all stages of oocyte development. Primordial oocytes constitute a nonproliferating, nongrowing reserve of potential eggs maintained for the entire reproductive lifespan of mammalian females. Using conditional knockouts, we determined the role of MTOR in both primordial and growing oocytes. MTOR-dependent pathways in primordial oocytes are not needed to sustain the viability of the primordial oocyte pool or their recruitment into the cohort of growing oocytes but are essential later for maintenance of oocyte genomic integrity, sustaining ovarian follicular development, and fertility. In growing oocytes, MTOR-dependent pathways are required for processes that promote completion of meiosis and enable embryonic development. MTOR (mechanistic target of rapamycin) is a widely recognized integrator of signals and pathways key for cellular metabolism, proliferation, and differentiation. Here we show that conditional knockout (cKO) of Mtor in either primordial or growing oocytes caused infertility but differentially affected oocyte quality, granulosa cell fate, and follicular development. cKO of Mtor in nongrowing primordial oocytes caused defective follicular development leading to progressive degeneration of oocytes and loss of granulosa cell identity coincident with the acquisition of immature Sertoli cell-like characteristics. Although Mtor was deleted at the primordial oocyte stage, DNA damage accumulated in oocytes during their later growth, and there was a marked alteration of the transcriptome in the few oocytes that achieved the fully grown stage. Although oocyte quality and fertility were also compromised when Mtor was deleted after oocytes had begun to grow, these occurred without overtly affecting folliculogenesis or the oocyte transcriptome. Nevertheless, there was a significant change in a cohort of proteins in mature oocytes. In particular, down-regulation of PRC1 (protein regulator of cytokinesis 1) impaired completion of the first meiotic division. Therefore, MTOR-dependent pathways in primordial or growing oocytes differentially affected downstream processes including follicular development, sex-specific identity of early granulosa cells, maintenance of oocyte genome integrity, oocyte gene expression, meiosis, and preimplantation developmental competence.
Collapse
|
32
|
Graves JS, Henry RG, Cree BAC, Lambert-Messerlian G, Greenblatt RM, Waubant E, Cedars MI, Zhu A, Bacchetti P, Hauser SL, Oksenberg JR. Ovarian aging is associated with gray matter volume and disability in women with MS. Neurology 2017; 90:e254-e260. [PMID: 29273686 DOI: 10.1212/wnl.0000000000004843] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 10/02/2017] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To determine if ovarian aging as measured by levels of anti-Müllerian hormone (AMH) is associated with pattern of multiple sclerosis (MS) progression in women. METHODS Women with MS and healthy controls were included from a longitudinal research cohort with up to 10 years follow-up. Plasma AMH levels were measured by ELISA for baseline and years 3, 5, and 8-10. Mixed effects logistic and linear regression models were employed, with adjustments for age, disease duration, and other covariables as appropriate. RESULTS AMH levels were similar (0.98-fold difference, 95% confidence interval [CI] 0.69-1.37, p = 0.87) in women with MS (n = 412, mean age 42.6 years) and healthy controls (n = 180, mean age 44 years). In a multivariable model of women with MS, including adjustments for age, body mass index, and disease duration, 10-fold lower AMH level was associated with 0.43-higher Expanded Disability Status Scale (EDSS) score (95% CI 0.15-0.70, p = 0.003), 0.25-unit worse MS Functional Composite z score (95% CI -0.40 to -0.10, p = 0.0015), and 7.44 mm3 lower cortical gray matter volume (95% CI -14.6 to -0.30; p = 0.041) at baseline. In a multivariable random-intercept-random-slope model using all observations over time, 10-fold decrease in AMH was associated with a 0.27 increase in EDSS (95% CI 0.11-0.43, p = 0.006) and 5.48 mm3 (95% CI 11.3-0.33, p = 0.065) and 4.55 mm3 (95% CI 9.33-0.23, p = 0.062) decreases in total gray and cortical gray matter, respectively. CONCLUSION As a marker of ovarian aging, lower AMH levels were associated with greater disability and gray matter loss in women with MS independent of chronological age and disease duration.
Collapse
Affiliation(s)
- Jennifer S Graves
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI.
| | - Roland G Henry
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Bruce A C Cree
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Geralyn Lambert-Messerlian
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Ruth M Greenblatt
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Emmanuelle Waubant
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Marcelle I Cedars
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Alyssa Zhu
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | | | - Peter Bacchetti
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Stephen L Hauser
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| | - Jorge R Oksenberg
- From the Departments of Neurology (J.S.G., R.G.H., B.A.C.C., E.W., A.Z., S.L.H., J.R.O.), Pharmacology (R.M.G.), Obstetrics, Gynecology and Reproductive Sciences (M.I.C.), and Epidemiology and Biostatistics (P.B.), University of California, San Francisco; and Women and Infants Hospital and the Alpert Medical School at Brown University (G.L.-M.), Providence, RI
| |
Collapse
|
33
|
Sacchi S, Marinaro F, Xella S, Marsella T, Tagliasacchi D, La Marca A. The anti-Müllerian hormone (AMH) induces forkhead box L2 (FOXL2) expression in primary culture of human granulosa cells in vitro. J Assist Reprod Genet 2017; 34:1131-1136. [PMID: 28660501 DOI: 10.1007/s10815-017-0980-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/13/2017] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Anti-Müllerian hormone (AMH) and forkhead box L2 (FOXL2) are two pivotal genes expressed in human granulosa cells (hGCs) where both genes share similar inhibitory functions on activation and follicular growth in order to preserve the ovarian follicle reserve. Furthermore, AMH and FOXL2 contribute to inhibit steroidogenesis, decreasing or preventing the activation of gonadotrophin-dependent aromatase CYP19A1 cytochrome P450 family 19 subfamily A member 1 (CYP19A1). The purpose of this study is to evaluate the role of AMH in regulating the expression of FOXL2. METHODS Primary cultures of hGCs were treated with increasing concentrations of recombinant human AMH (rhAMH; range 10-100 ng/ml) for 3 h. Negative controls were performed using corresponding amounts of AMH vehicle. Total RNA or proteins were purified and quantified by spectrophotometry. FOXL2 and CYP19A1 gene expression, normalized by reference gene ribosomal protein S7 (RpS7), was evaluated by RT-qPCR. Each reaction was repeated in triplicate. Statistical analysis was performed. Extracted proteins were analyzed by immunoblot using anti-FOXL2 and anti-β-actin as primary antibodies. RESULTS rhAMH treatments tested did not modulate the basal expression of aromatase CYP19A1 gene. rhAMH (50 ng/ml) was able to increase FOXL2 gene expression and its intracellular content. CONCLUSIONS This study demonstrated the existence of an AMH-FOXL2 relationship in hGCs. AMH is capable of increasing both gene and protein expression of FOXL2. Because FOXL2 induces AMH transcription, these ovarian factors could be finely regulated by a positive feedback loop mechanism to preserve the ovarian follicle reserve.
Collapse
Affiliation(s)
- Sandro Sacchi
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia and Clinica Eugin Modena, Via del Pozzo 71, 41100, Modena, Italy
| | - Federica Marinaro
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia and Clinica Eugin Modena, Via del Pozzo 71, 41100, Modena, Italy
| | - Susanna Xella
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia and Clinica Eugin Modena, Via del Pozzo 71, 41100, Modena, Italy
| | - Tiziana Marsella
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia and Clinica Eugin Modena, Via del Pozzo 71, 41100, Modena, Italy
| | - Daniela Tagliasacchi
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia and Clinica Eugin Modena, Via del Pozzo 71, 41100, Modena, Italy
| | - Antonio La Marca
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia and Clinica Eugin Modena, Via del Pozzo 71, 41100, Modena, Italy.
| |
Collapse
|
34
|
Jung S, Allen N, Arslan AA, Baglietto L, Brinton LA, Egleston BL, Falk R, Fortner RT, Helzlsouer KJ, Idahl A, Kaaks R, Lundin E, Merritt M, Onland-Moret C, Rinaldi S, Sánchez MJ, Sieri S, Schock H, Shu XO, Sluss PM, Staats PN, Travis RC, Tjønneland A, Trichopoulou A, Tworoger S, Visvanathan K, Krogh V, Weiderpass E, Zeleniuch-Jacquotte A, Zheng W, Dorgan JF. Demographic, lifestyle, and other factors in relation to antimüllerian hormone levels in mostly late premenopausal women. Fertil Steril 2017; 107:1012-1022.e2. [PMID: 28366409 PMCID: PMC5426228 DOI: 10.1016/j.fertnstert.2017.02.105] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 02/09/2017] [Accepted: 02/20/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To identify reproductive, lifestyle, hormonal, and other correlates of circulating antimüllerian hormone (AMH) concentrations in mostly late premenopausal women. DESIGN Cross-sectional study. SETTING Not applicable. PATIENT(S) A total of 671 premenopausal women not known to have cancer. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Concentrations of AMH were measured in a single laboratory using the picoAMH ELISA. Multivariable-adjusted median (and interquartile range) AMH concentrations were calculated using quantile regression for several potential correlates. RESULT(S) Older women had significantly lower AMH concentrations (≥40 [n = 444] vs. <35 years [n = 64], multivariable-adjusted median 0.73 ng/mL vs. 2.52 ng/mL). Concentrations of AMH were also significantly lower among women with earlier age at menarche (<12 [n = 96] vs. ≥14 years [n = 200]: 0.90 ng/mL vs. 1.12 ng/mL) and among current users of oral contraceptives (n = 27) compared with never or former users (n = 468) (0.36 ng/mL vs. 1.15 ng/mL). Race, body mass index, education, height, smoking status, parity, and menstrual cycle phase were not significantly associated with AMH concentrations. There were no significant associations between AMH concentrations and androgen or sex hormone-binding globulin concentrations or with factors related to blood collection (e.g., sample type, time, season, and year of blood collection). CONCLUSION(S) Among premenopausal women, lower AMH concentrations are associated with older age, a younger age at menarche, and currently using oral contraceptives, suggesting these factors are related to a lower number or decreased secretory activity of ovarian follicles.
Collapse
Affiliation(s)
- Seungyoun Jung
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - Naomi Allen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Alan A Arslan
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, New York; Departments of Population Health and Environmental Medicine and Perlmuttr Cancer Center, New York University School of Medicine, New York, New York
| | - Laura Baglietto
- Cancer Epidemiology Centre, Cancer Council of Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | | | - Roni Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Renée T Fortner
- Division of Cancer Epidemiology, German Cancer Research Cancer, Heidelberg, Germany
| | - Kathy J Helzlsouer
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Annika Idahl
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - Rudolph Kaaks
- Division of Cancer Epidemiology, German Cancer Research Cancer, Heidelberg, Germany
| | - Eva Lundin
- Department of Medical Biosciences, Pathology, and Public Health and Clinical Medicine: Nutritional Research, Umeå University, Umeå, Sweden
| | - Melissa Merritt
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Charlotte Onland-Moret
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sabina Rinaldi
- International Agency for Research on Cancer, Lyon, France
| | - María-José Sánchez
- Escuela Andaluza de Salud Pública, Instituto de Investigación Biosanitaria ibs, GRANADA, Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Helena Schock
- Division of Cancer Epidemiology, German Cancer Research Cancer, Heidelberg, Germany
| | - Xiao-Ou Shu
- Department of Epidemiology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Patrick M Sluss
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - Paul N Staats
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ruth C Travis
- Cancer Epidemiology Unit, University of Oxford, Oxford, United Kingdom
| | | | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece; World Health Organization Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
| | - Shelley Tworoger
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Baltimore, Maryland; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Vittorio Krogh
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway; Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
| | - Anne Zeleniuch-Jacquotte
- Departments of Population Health and Environmental Medicine and Perlmuttr Cancer Center, New York University School of Medicine, New York, New York
| | - Wei Zheng
- Department of Epidemiology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joanne F Dorgan
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
35
|
Pankhurst MW. A putative role for anti-Müllerian hormone (AMH) in optimising ovarian reserve expenditure. J Endocrinol 2017; 233:R1-R13. [PMID: 28130407 DOI: 10.1530/joe-16-0522] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/24/2017] [Indexed: 12/31/2022]
Abstract
The mammalian ovary has a finite supply of oocytes, which are contained within primordial follicles where they are arrested in a dormant state. The number of primordial follicles in the ovary at puberty is highly variable between females of the same species. Females that enter puberty with a small ovarian reserve are at risk of a shorter reproductive lifespan, as their ovarian reserve is expected to be depleted faster. One of the roles of anti-Müllerian hormone (AMH) is to inhibit primordial follicle activation, which slows the rate at which the ovarian reserve is depleted. A simple interpretation is that the function of AMH is to conserve ovarian reserve. However, the females with the lowest ovarian reserve and the greatest risk of early reserve depletion have the lowest levels of AMH. In contrast, AMH apparently strongly inhibits primordial follicle activation in females with ample ovarian reserve, for reasons that remain unexplained. The rate of primordial follicle activation determines the size of the developing follicle pool, which in turn, determines how many oocytes are available to be selected for ovulation. This review discusses the evidence that AMH regulates the size of the developing follicle pool by altering the rate of primordial follicle activation in a context-dependent manner. The expression patterns of AMH across life are also consistent with changing requirements for primordial follicle activation in the ageing ovary. A potential role of AMH in the fertility of ageing females is proposed herein.
Collapse
Affiliation(s)
- Michael W Pankhurst
- Department of AnatomySchool of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
36
|
Snoeck F, Sarrazin S, Wydooghe E, Van Soom A. Age and anti-Müllerian hormone levels predict the success of in vitro maturation of cat oocytes. Reprod Domest Anim 2016; 52 Suppl 2:98-102. [DOI: 10.1111/rda.12827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- F Snoeck
- Department of Reproduction, Obstetrics and Herd Health; Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - S Sarrazin
- Department of Reproduction, Obstetrics and Herd Health; Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - E Wydooghe
- Department of Reproduction, Obstetrics and Herd Health; Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - A Van Soom
- Department of Reproduction, Obstetrics and Herd Health; Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| |
Collapse
|
37
|
The comparison of animal models for premature ovarian failure established by several different source of inducers. Regul Toxicol Pharmacol 2016; 81:223-232. [PMID: 27612992 DOI: 10.1016/j.yrtph.2016.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/27/2016] [Accepted: 09/05/2016] [Indexed: 01/03/2023]
Abstract
The objective of this study was to compare premature ovarian failure animal models established by several different source of inducers. Female ICR mice, KM mice, and SD rats were treated by cyclophosphamide at 120 mg/kg, busulfan at 12 mg/kg, cisplatin at 3 or 4 mg/kg, 4-vinylcyclohexene diepoxide at 160 mg/kg, 35% galactose food pellet, and tripterygium glycosides at 50 mg/kg, respectively. Parameters were analyzed by body weight, serum concentration level of related hormones, ovarian and uterine pathological examination. The results indicated the body weight of mice increased very slowly following single dose of cyclophosphamide (p < 0.05) with damaged ovary; repeated doses of cisplatin could induce body weight significantly decreased (p < 0.01) with a rising trend of serum LH concentration, declining tendency of serum E2 concentration and injured ovary and uterus; 4-vinylcyclohexene diepoxide also hindered the mice growing (p < 0.05) with damaged ovary and uterus; the body weight of mice feed by 35% galactose food pellet increased slowly (p < 0.05) with dramatically higher serum concentration level of galactose, albumin, and total protein (p < 0.001) and injured ovary. Busulfan and tripterygium glycosides did not present obvious evidences. In conclusion, the inducers presented their respective features in such animal models and should be appropriately applied in preventive methods.
Collapse
|
38
|
Abstract
In mammals, ovulation is a multistep physiological process that includes preovulatory follicle growth, oocyte meiotic maturation, cumulus-oocyte complex (COC) expansion, follicle rupture, and luteinization. TGF-β signaling pathway has multiple functions in mammalian ovary, as its complexity in ovarian function has been demonstrated by mouse models with knockouts of TGF-β receptors and SMADs. We describe the protocol that we use to study functions of TGF-β signaling pathway in follicle development and ovulation. Because total knockout of TGF-β pathway components often causes embryonic lethality, which prevents further investigation of these genes in ovarian functions, people have generated ovarian cell type-specific knockout mouse strains for TGF-β signaling pathway genes. These mouse models are also described.
Collapse
Affiliation(s)
- Chao Yu
- Life Science Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China
| | - Jian-Jie Zhou
- Life Science Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China
| | - Heng-Yu Fan
- Life Science Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
39
|
Pirgon O, Sivrice C, Demirtas H, Dundar B. Assessment of ovarian reserve in euthyroid adolescents with Hashimoto thyroiditis. Gynecol Endocrinol 2016; 32:306-10. [PMID: 26608409 DOI: 10.3109/09513590.2015.1116510] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
AIM We aimed to investigate the ovarian function and reserve in euthyroid adolescents (TSH < 2.5 mIU/L) diagnosed with Hashimoto thyroiditis (HT). METHODS This case-control study included 30 adolescent girls (mean age 15.1 ± 1.4 years) newly diagnosed as HT with presence of high thyroid antibodies with gland heterogeneity in ultrasound and age-matched 30 healthy female subjects. Anti-ovarian antibody (AOAb), LH/FSH ratio, estradiol, anti-mullerian hormone (AMH), inhibin-B, total testosterone, antral follicle count, ovarian volumes and uterine length were measured. The clinical, laboratory, and ultrasound data of the HT and control groups were compared. RESULTS There were no significant differences between the girls with HT and healthy controls in relation to LH/FSH ratio, estradiol and inhibin-B levels. AOAb (p = 0.02), AMH (p = 0.007) and total testosterone levels were higher in HT group than the control group (p = 0.03). AOAb level was found to be positively correlated with LH/FSH ratio (p = 0.03), AMH (p = 0.01) and inhibin-B (p < 0.001) in HT group. CONCLUSION This study demonstrated that the adolescent girls diagnosed with autoimmune thyroiditis had normal ovarian reserve based on measurements of AMH, inhibin B, FSH, LH/FSH ratio, estradiol and antral follicle counts.
Collapse
Affiliation(s)
- Ozgur Pirgon
- a Department of Pediatric Endocrinology and Diabetes
| | | | - Hakan Demirtas
- b Department of Pediatric Radiology, Faculty of Medicine , S. Demirel University , Isparta , Turkey , and
| | - Bumin Dundar
- c Department of Pediatric Endocrinology and Diabetes, Faculty of Medicine , Katip Celebi University , Izmir , Turkey
| |
Collapse
|
40
|
Dumont A, Robin G, Catteau-Jonard S, Dewailly D. Role of Anti-Müllerian Hormone in pathophysiology, diagnosis and treatment of Polycystic Ovary Syndrome: a review. Reprod Biol Endocrinol 2015; 13:137. [PMID: 26691645 PMCID: PMC4687350 DOI: 10.1186/s12958-015-0134-9] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/13/2015] [Indexed: 11/10/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of chronic anovulation and hyperandrogenism in young women. Excessive ovarian production of Anti-Müllerian Hormone, secreted by growing follicles in excess, is now considered as an important feature of PCOS. The aim of this review is first to update the current knowledge about the role of AMH in the pathophysiology of PCOS. Then, this review will discuss the improvement that serum AMH assay brings in the diagnosis of PCOS. Last, this review will explain the utility of serum AMH assay in the management of infertility in women with PCOS and its utility as a marker of treatment efficiency on PCOS symptoms. It must be emphasized however that the lack of an international standard for the serum AMH assay, mainly because of technical issues, makes it difficult to define consensual thresholds, and thus impairs the widespread use of this new ovarian marker. Hopefully, this should soon improve.
Collapse
Affiliation(s)
- Agathe Dumont
- Service de Gynécologie Endocrinienne et de Médecine de la Reproduction, Hôpital Jeanne de Flandre, CHRU, 2 Avenue Eugène Avinée, 59037, Lille, France.
| | - Geoffroy Robin
- Service de Gynécologie Endocrinienne et de Médecine de la Reproduction, Hôpital Jeanne de Flandre, CHRU, 2 Avenue Eugène Avinée, 59037, Lille, France.
| | - Sophie Catteau-Jonard
- Service de Gynécologie Endocrinienne et de Médecine de la Reproduction, Hôpital Jeanne de Flandre, CHRU, 2 Avenue Eugène Avinée, 59037, Lille, France.
| | - Didier Dewailly
- Service de Gynécologie Endocrinienne et de Médecine de la Reproduction, Hôpital Jeanne de Flandre, CHRU, 2 Avenue Eugène Avinée, 59037, Lille, France.
| |
Collapse
|
41
|
Pelusi C, Stancampiano M, Fanelli F, Pariali M, Gambineri A, Pasquali R. Anti-müllerian hormone and insulin-like 3 levels in healthy normal-weight ovulatory and anovulatory eumenorrheic late adolescent females: potential early biomarkers of ovarian dysfunction? Eur J Obstet Gynecol Reprod Biol 2015; 195:188-192. [PMID: 26579638 DOI: 10.1016/j.ejogrb.2015.09.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 09/08/2015] [Accepted: 09/28/2015] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate differences in anti-müllerian hormone (AMH) and insulin-like 3 (INSL3) levels and their association with gonadotropin and ovarian steroid hormones, as expression of ovarian function, between healthy normal-weight ovulatory and anovulatory eumenorrheic late adolescent females. STUDY DESIGN This study analyzed AMH and INSL3 levels in forty healthy eumenorrheic late adolescent females (aged 16-19 ys), selected from a cross-sectional epidemiological study performed on the prevalence of hyperandrogenic states. The subjects were divided into ovulatory (n: 28) and anovulatory (n: 12) groups in accordance to a previous cluster analysis based on progesterone (P) distribution measured once in the latter part of the cycle. Both groups were compared for anthropometric, biochemical and hormonal parameters. RESULTS INSL3 and AMH were detectable in all samples. Testosterone (P=0.01), the free-androgen index (FAI) (P=0.051), gonadotropins (LH: P=0.02; FSH: P=0.004) and AMH (P=0.02) levels were significantly higher in the anovulatory group with respect to their ovulatory counterpart. A trend toward significantly higher INSL3 concentrations (P=0.08) was also shown in the anovulatory group. A positive correlation between INSL3 levels and androgens such as androstenedione (r=0.38; P=0.02), testosterone (r=0.44; P=0.004) and FAI (r=0.42; P=0.006) and a negative borderline significant correlation (r=-0.30; P=0.055) between AMH and P were shown in all subjects. CONCLUSION Healthy eumenorrheic late adolescent females with sporadic anovulation display higher AMH and INSL-3 blood concentrations in association with higher androgen levels compared with age- and BMI-matched subjects with ovulatory cycle, suggesting evidence of an earlier ovarian dysfunction.
Collapse
Affiliation(s)
- Carla Pelusi
- Division of Endocrinology, Department of Medical and Surgical Science (DIMEC), and Center of Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University Alma Mater Studiorum, Bologna, Italy
| | - Marianna Stancampiano
- Division of Endocrinology, Department of Medical and Surgical Science (DIMEC), and Center of Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University Alma Mater Studiorum, Bologna, Italy
| | - Flaminia Fanelli
- Division of Endocrinology, Department of Medical and Surgical Science (DIMEC), and Center of Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University Alma Mater Studiorum, Bologna, Italy
| | - Milena Pariali
- Division of Endocrinology, Department of Medical and Surgical Science (DIMEC), and Center of Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University Alma Mater Studiorum, Bologna, Italy
| | - Alessandra Gambineri
- Division of Endocrinology, Department of Medical and Surgical Science (DIMEC), and Center of Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University Alma Mater Studiorum, Bologna, Italy
| | - Renato Pasquali
- Division of Endocrinology, Department of Medical and Surgical Science (DIMEC), and Center of Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University Alma Mater Studiorum, Bologna, Italy.
| |
Collapse
|
42
|
Salmassi A, Mettler L, Hedderich J, Jonat W, Deenadayal A, von Otte S, Eckmann-Scholz C, Schmutzler AG. Cut-Off Levels of Anti-Mullerian Hormone for The Prediction of Ovarian Response, In Vitro Fertilization Outcome and Ovarian Hyperstimulation Syndrome. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2015; 9:157-67. [PMID: 26246873 PMCID: PMC4518483 DOI: 10.22074/ijfs.2015.4236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/31/2014] [Indexed: 01/09/2023]
Abstract
Background Evaluation of anti-mullerian hormone (AMH) cut-off levels in as-
sisted reproductive technology (ART) as predictive factor for individualization of
stimulation protocols and to avoid ovarian hyperstimulation syndrome (OHSS). Materials and Methods In a retrospective study, 177 infertile patients were as-
sessed for AMH in serum and follicular fluid (FF) on the day of follicular puncture
(FP), between 2012 and 2013 in Kiel, Germany. AMH levels and pregnancy rates
were compared between low, moderate and high responders and cut-off levels of
low and high responders. AMH cut-off levels in pathological cases were evaluated
in analysis 1 (OHSS) and in analysis 2 [polycystic ovarian syndrome, (PCOS)] and
compared in analysis 3 to normal endocrinological parameters. Results AMH levels in FF were higher than in serum (P<0.001). AMH levels in serum
and FF increased from low through moderate to high responders (P<0.001). Pregnancy
rates were 14.7, 23.3 and 44.9% (P=0.009), respectively. AMH cut-off level for poor
responders was 0.61 ng/ml in serum with a pregnancy rate of 13.8 and 37.1% for below
and above of this level, respectively. For FF, it was 1.43 ng/ml. AMH levels in analysis
1 and 2 were significantly higher than in analysis 3 (P=0.001). AMH cut-off level for
OHSS was 1.5 ng/ml in serum with OHSS rates of 80.8 and 19.2 % for above and below
of the level, respectively. For FF, it was 2.7 ng/ml. PCOS patients had an AMH cut-off
level of 3.9 ng/ml in serum and 6.8 ng/ml in FF, resulting in a PCOS rate of 100% above
this level. Conclusion AMH levels can help to assess ovarian response potential and guide ovarian
stimulation while avoiding OHSS.
Collapse
Affiliation(s)
- Ali Salmassi
- Department of Gynaecology and Obstetrics, Center of Reproductive Medicine, University Hospitals Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Liselotte Mettler
- Department of Gynaecology and Obstetrics, Center of Reproductive Medicine, University Hospitals Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jurgen Hedderich
- Institute of Medical Informatics and Statistics, University of Kiel, Kiel, Germany
| | - Walter Jonat
- Department of Gynaecology and Obstetrics, Center of Reproductive Medicine, University Hospitals Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Anupama Deenadayal
- Department of Gynaecology and Obstetrics, Center of Reproductive Medicine, University Hospitals Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Soeren von Otte
- Department of Gynaecology and Obstetrics, Center of Reproductive Medicine, University Hospitals Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christel Eckmann-Scholz
- Department of Gynaecology and Obstetrics, Center of Reproductive Medicine, University Hospitals Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Andreas Gerd Schmutzler
- Department of Gynaecology and Obstetrics, Center of Reproductive Medicine, University Hospitals Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
43
|
Vernunft A, Schwerhoff M, Viergutz T, Diederich M, Kuwer A. Anti-Muellerian hormone levels in plasma of Holstein-Friesian heifers as a predictive parameter for ovum pick-up and embryo production outcomes. J Reprod Dev 2014; 61:74-9. [PMID: 25482112 PMCID: PMC4354234 DOI: 10.1262/jrd.2014-091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The aim of this study was to investigate whether plasma anti-Muellerian hormone (AMH) levels of Holstein-Friesian heifers could be used to predict ovum pick-up (OPU) and embryo production outcomes. Plasma samples and data were collected from 64 heifers, which underwent repeated OPU with subsequent in vitro embryo production followed by embryo flushing after superovulation. AMH levels were significantly positively correlated with the number of follicles aspirated per OPU session (r = 0.45), recovered oocytes per OPU (r =0.43) and in vitro produced embryos per OPU (r = 0.28). No significant correlations between AMH and in vivo produced embryos were ascertained. Our results suggest that correlations between AMH and outcomes of an OPU-IVF program are too low to use AMH as a precise predictive parameter for the success of a particular OPU procedure in Holstein-Friesian heifers. However, AMH can help to identify groups of
very good or very poor oocyte donors.
Collapse
Affiliation(s)
- Andreas Vernunft
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany
| | | | | | | | | |
Collapse
|
44
|
Li J, Li R, Yu H, Zhao S, Yu Y, Qiao J. The relationship between serum anti-Müllerian hormone levels and the follicular arrest for women with polycystic ovary syndrome. Syst Biol Reprod Med 2014; 61:103-9. [DOI: 10.3109/19396368.2014.973123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
45
|
Feeney A, Nilsson E, Skinner MK. Cytokine (IL16) and tyrphostin actions on ovarian primordial follicle development. Reproduction 2014; 148:321-31. [PMID: 24970835 DOI: 10.1530/rep-14-0246] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
An ovarian follicle is composed of an oocyte and surrounding theca and granulosa cells. Oocytes are stored in an arrested state within primordial follicles until they are signaled to re-initiate development by undergoing primordial-to-primary follicle transition. Previous gene bionetwork analyses of primordial follicle development identified a number of critical cytokine signaling pathways and genes potentially involved in the process. In the current study, candidate regulatory genes and pathways from the gene network analyses were tested for their effects on the formation of primordial follicles (follicle assembly) and on primordial follicle transition using whole ovary organ culture experiments. Observations indicate that the tyrphostin inhibitor (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one increased follicle assembly significantly, supporting a role for the MAPK signaling pathway in follicle assembly. The cytokine interleukin 16 (IL16) promotes primordial-to-primary follicle transition as compared with the controls, where as Delta-like ligand 4 (DLL4) and WNT-3A treatments have no effect. Immunohistochemical experiments demonstrated the localization of both the cytokine IL16 and its receptor CD4 in the granulosa cells surrounding each oocyte within the ovarian follicle. The tyrphostin LDN193189 (LDN) is an inhibitor of the bone morphogenic protein receptor 1 within the TGFB signaling pathway and was found to promote the primordial-to-primary follicle transition. Observations support the importance of cytokines (i.e., IL16) and cytokine signaling pathways in the regulation of early follicle development. Insights into regulatory factors affecting early primordial follicle development are provided that may associate with ovarian disease and translate to improved therapy in the future.
Collapse
Affiliation(s)
- Amanda Feeney
- School of Biological SciencesCenter for Reproductive Biology, Washington State University, Pullman, Washington 99164-4236, USA
| | - Eric Nilsson
- School of Biological SciencesCenter for Reproductive Biology, Washington State University, Pullman, Washington 99164-4236, USA
| | - Michael K Skinner
- School of Biological SciencesCenter for Reproductive Biology, Washington State University, Pullman, Washington 99164-4236, USA
| |
Collapse
|
46
|
Anti-mullerian hormone in the management of infertility. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2014. [DOI: 10.1016/j.mefs.2014.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
47
|
Visser JA, Themmen APN. Role of anti-Müllerian hormone and bone morphogenetic proteins in the regulation of FSH sensitivity. Mol Cell Endocrinol 2014; 382:460-465. [PMID: 23994017 DOI: 10.1016/j.mce.2013.08.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/18/2013] [Indexed: 10/26/2022]
Abstract
The ovary is under control of the hypothalamus and pituitary through the glycoprotein hormones LH and FSH. These hormones undergo a cyclic variation which results in the selection of the species-specific number of follicles that will ovulate during the cycle. Where LH is the main ovulatory hormone and regulator of corpus luteum function, FSH plays an essential role in the cyclic recruitment of the follicles. Within the microenvironment of the ovary, growth factors affect this dominant control of FSH by regulating the FSH sensitivity of individual follicles. In this review we discuss the role of anti-Müllerian hormone (AMH) and bone morphogenetic proteins (BMPs) in this process.
Collapse
Affiliation(s)
- Jenny A Visser
- Dept. of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Axel P N Themmen
- Dept. of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
48
|
Dewailly D, Andersen CY, Balen A, Broekmans F, Dilaver N, Fanchin R, Griesinger G, Kelsey TW, La Marca A, Lambalk C, Mason H, Nelson SM, Visser JA, Wallace WH, Anderson RA. The physiology and clinical utility of anti-Müllerian hormone in women. Hum Reprod Update 2014; 20:370-85. [DOI: 10.1093/humupd/dmt062] [Citation(s) in RCA: 577] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
49
|
Zhang Y, Ding JX, Tao X, Lu ZY, Wang JJ, Feng WW, Hua KQ. Goserelin can inhibit ovarian cancer proliferation and simultaneously protect ovarian function from cisplatin: anin vitroandin vivostudy. J Chemother 2013; 25:96-103. [DOI: 10.1179/1973947813y.0000000069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
50
|
Chong YH, Campbell AJ, Farrand S, McLennan IS. Anti-Müllerian hormone level in older women: detection of granulosa cell tumor recurrence. Int J Gynecol Cancer 2013; 22:1497-9. [PMID: 23051961 DOI: 10.1097/igc.0b013e318270ac69] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE To determine whether anti-Müllerian hormone (AMH) production resumes during normal late menopausal aging. Anti-Müllerian hormone has been proposed as a specific serum marker for adult granulosa cell tumors. MATERIALS AND METHODS Serum AMH from 21 elderly postmenopausal women (mean age, 77 years) and 9 young women (mean age, 22 years) were measured by ultrasensitive immunoassay. RESULTS Both median (0 pmol/L) and mean (0.48 pmol/L) serum AMH values for the elderly women were below the level of detection for the immunoassay kit. Three of the 21 participants had minimally detectable level of AMH (1.13-2.76 pmol/L). The cohort of young women had expected normal values of AMH as measurable by the same immunoassay kit. CONCLUSIONS Serum AMH values were negligible for postmenopausal women older than 65 years. This extends the normative data for AMH to 108 years old, providing a reference range for the detection of granulosa cell tumors in postmenopausal women.
Collapse
Affiliation(s)
- Yih Harng Chong
- Department of Anatomy, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | | | | | | |
Collapse
|