1
|
Özden Akkaya Ö, Yağci A, Zik B, Kibria ASMG, Güler S, Çelik S, Altunbaş K. The effect of bisphenol A on the Notch (Notch2 and Jagged2) signaling pathway in the follicular development of the neonatal rat ovary. Biotech Histochem 2024:1-22. [PMID: 39382141 DOI: 10.1080/10520295.2024.2361313] [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: 10/10/2024] Open
Abstract
The formation of primordial follicles determines the pool size of follicles in the ovary, and is crucial for female reproductivity. Oocyte nest breakdown, and the formation of primordial follicles, largely depend upon the communication between oocytes and the surrounding pregranulosa cells. The neurogenic locus notch homolog protein (Notch) signaling pathway is the key player for this cell-to-cell communication, and is responsible for primordial folliculogenesis. However, different endocrine disruptors, including bisphenol A (BPA; a plasticizer and a constituent of reusable plastic containers) may affect the Notch signaling pathway, and might induce ovary dysfunction via Notch signaling. Consequently, we investigated the possible influence of BPA treatment on the proportional distribution of the follicular stages, follicle numbers, levels of apoptosis, and on Notch2 and Jagged2 expressions in the ovary. BPA was administered at doses of either 50 µg/kg/day or 50 mg/kg/day, at different time intervals, during neonatal and fetal periods in vivo. After collecting the ovaries from the various experimental groups, follicles were counted, and frequency of apoptosis was determined by TUNEL assay. In addition, Notch2 and Jagged2 expressions were assessed by immunohistochemical staining and qPCR. In summary, BPA treatment affected the follicle numbers and apoptosis level, and Notch2 and Jagged2 expressions varied with follicular stage. It was also observed that these parameters were dose and time dependent with respect to BPA exposure.
Collapse
Affiliation(s)
- Özlem Özden Akkaya
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Artay Yağci
- Department of Histology and Embryology, Milas Veterinary Faculty, Muğla Sıtkı Koçman University, Muğla, Türkiye
| | - Berrin Zik
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa, Türkiye
| | - A S M Golam Kibria
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Chattogram, Veterinary and Animal Sciences University, Chattogram, Bangladesh
| | - Sabire Güler
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa, Türkiye
| | - Sefa Çelik
- Department of Biochemistry, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Türkiye
| | - Korhan Altunbaş
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| |
Collapse
|
2
|
Wadood AA, Xiquan Z. Unraveling the mysteries of chicken proteomics: Insights into follicle development and reproduction. J Proteomics 2024; 308:105281. [PMID: 39154802 DOI: 10.1016/j.jprot.2024.105281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Chicken proteomics is a valuable method for comprehending the many mechanisms involved in follicle growth and reproduction in birds. This study offers a thorough summary of the latest progress in chicken proteomics research, specifically highlighting the knowledge obtained regarding follicle development and reproductive physiology. Proteomic studies have revealed essential proteins and pathways that play a role in follicle development, including those that control oocyte size, maturation, and ovulation. Proteomic investigations have provided insight into the molecular pathways that govern reproductive processes. By utilizing advanced proteomic technologies, including mass spectrometry and protein microarray analysis, we have been able to identify and measure many proteins in chicken follicles at their different developmental stages. The utilization of proteomic methods has enabled the identification of previously unknown biomarkers for reproductive efficiency that expedited the creation of innovative diagnostic instruments for monitoring reproductive health in chicken. Chicken proteomics not only offers insights into follicle growth and reproduction but also uncovers the effects of environmental influences on reproductive function. This provides new opportunities for exploring the molecular pathways that cause these effects. The integration of current data with upcoming proteomic technologies offers the potential for innovative strategies to enhance chicken reproduction.
Collapse
Affiliation(s)
- Armughan Ahmed Wadood
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Zhang Xiquan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
3
|
Wei X, Bjarkadottir BD, Nadjaja D, Sheikh S, Fatum M, Lane S, Williams SA. Effect of AMH on primordial follicle populations in mouse ovaries and human pre-pubertal ovarian xenografts during doxorubicin treatment. Front Cell Dev Biol 2024; 12:1449156. [PMID: 39258229 PMCID: PMC11383774 DOI: 10.3389/fcell.2024.1449156] [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: 06/14/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Survival rates of the childhood cancer patients are improving, however cancer treatments such as chemotherapy may lead to infertility due to loss of the primordial follicle (PMF) reserve. Doxorubicin (DXR) is a gonadotoxic chemotherapy agent commonly used in childhood cancers. Anti-Müllerian Hormone (AMH) has been reported to have a protective effect on the mouse ovarian reserve against DXR in vivo. However, whether AMH can prevent PMF loss in conjunction with DXR in human ovarian tissue in vivo has not been determined. Methods In order to investigate this, we first established an optimum dose of DXR that induced PMF loss in cultured mouse ovaries and investigated the efficacy of AMH on reducing DXR-induced PMF loss in mice in vitro. Second, we investigated the effects of DXR on pre-pubertal human ovarian tissue and the ability of AMH to prevent DXR-induced damage comparing using a mouse xenograft model with different transplantation sites. Results Mouse ovaries treated with DXR in vitro and in vivo had reduced PMF populations and damaged follicle health. We did not observe effect of DXR-induced PMF loss or damage to follicle/stromal health in human ovarian cortex, this might have been due to an insufficient dose or duration of DXR. Although AMH does not prevent DXR-induced PMF loss in pre-pubertal and adult mouse ovaries, in mouse ovaries treated with higher concentration of AMH in vitro, DXR did not cause a significant loss in PMFs. This is the first study to illustrate an effect of AMH on DXR-induced PMF loss on pre-pubertal mouse ovaries. However, more experiments with higher doses of AMH and larger sample size are needed to confirm this finding. Discussion We did not observe that AMH could prevent DXR-induced PMF loss in mouse ovaries in vivo. Further studies are warranted to investigate whether AMH has a protective effect against DXR in xenotransplanted human ovarian tissue. Thus, to obtain robust evidence about the potential of AMH in fertility preservation during chemotherapy treatment, alternative AMH administration strategies need to be explored alongside DXR administration to fully interrogate the effect of DXR and AMH on human xenografted tissues.
Collapse
Affiliation(s)
- Xi Wei
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Briet D Bjarkadottir
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Devi Nadjaja
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Sairah Sheikh
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Muhammad Fatum
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Fertility, Institute of Reproductive Sciences, Oxford, United Kingdom
| | - Sheila Lane
- Department of Paediatric Oncology and Haematology, Children's Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Suzannah A Williams
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Lee EB, Chakravarthi VP, Mohamadi R, Dahiya V, Vo K, Ratri A, Fields PE, Marsh CA, Rumi MAK. Loss of ERβ Disrupts Gene Regulation in Primordial and Primary Follicles. Int J Mol Sci 2024; 25:3202. [PMID: 38542176 PMCID: PMC10970686 DOI: 10.3390/ijms25063202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 11/03/2024] Open
Abstract
Loss of ERβ increases primordial follicle growth activation (PFGA), leading to premature ovarian follicle reserve depletion. We determined the expression and gene regulatory functions of ERβ in dormant primordial follicles (PdFs) and activated primary follicles (PrFs) using mouse models. PdFs and PrFs were isolated from 3-week-old Erβ knockout (Erβnull) mouse ovaries, and their transcriptomes were compared with those of control Erβfl/fl mice. We observed a significant (≥2-fold change; FDR p-value ≤ 0.05) deregulation of approximately 5% of genes (866 out of 16,940 genes, TPM ≥ 5) in Erβnull PdFs; ~60% (521 out of 866) of the differentially expressed genes (DEGs) were upregulated, and 40% were downregulated, indicating that ERβ has both transcriptional enhancing as well as repressing roles in dormant PdFs. Such deregulation of genes may make the Erβnull PdFs more susceptible to increased PFGA. When the PdFs undergo PFGA and form PrFs, many new genes are activated. During PFGA of Erβfl/fl follicles, we detected a differential expression of ~24% genes (4909 out of 20,743; ≥2-fold change; FDR p-value ≤ 0.05; TPM ≥ 5); 56% upregulated and 44% downregulated, indicating the gene enhancing and repressing roles of Erβ-activated PrFs. In contrast, we detected a differential expression of only 824 genes in Erβnull follicles during PFGA (≥2-fold change; FDR p-value ≤ 0.05; TPM ≥ 5). Moreover, most (~93%; 770 out of 824) of these DEGs in activated Erβnull PrFs were downregulated. Such deregulation of genes in Erβnull activated follicles may impair their inhibitory role on PFGA. Notably, in both Erβnull PdFs and PrFs, we detected a significant number of epigenetic regulators and transcription factors to be differentially expressed, which suggests that lack of ERβ either directly or indirectly deregulates the gene expression in PdFs and PrFs, leading to increased PFGA.
Collapse
Affiliation(s)
- Eun Bee Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| | - V. Praveen Chakravarthi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| | - Ryan Mohamadi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| | - Vinesh Dahiya
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| | - Kevin Vo
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| | - Anamika Ratri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| | - Patrick E. Fields
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| | - Courtney A. Marsh
- Department of Obstetrics and Gynecology, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA;
| | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center (KUMC), Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.); (R.M.); (V.D.); (K.V.); (A.R.); (P.E.F.)
| |
Collapse
|
5
|
Henríquez S, Valdivia MJ, Mainigi M, Villarroel C, Velasquez L, Strauss Iii JF, Devoto L. The role of estrogen metabolites in human ovarian function. Steroids 2024; 203:109368. [PMID: 38278282 DOI: 10.1016/j.steroids.2024.109368] [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: 08/04/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024]
Abstract
Estrogens produced by the ovary play diverse roles in controlling physiological changes in the function of the female reproductive system. Although estradiol acts through classical nuclear receptors, its metabolites (EMs) act by alternative pathways. It has been postulated that EMs act through paracrine-autocrine pathways to regulate key processes involved in normal follicular growth, corpus luteum (CL) development, function, and regression. The present review describes recent advances in understanding the role of EMs in human ovarian physiology during the menstrual cycle, including their role in anovulatory disorders and their action in other target tissues.
Collapse
Affiliation(s)
- Soledad Henríquez
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile; Institute of Interdisciplinary Research in Biomedical Sciences (I3CBSEK), Faculty of Health Sciences, SEK University, Santiago, Chile.
| | - Maria Jose Valdivia
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Monica Mainigi
- Department of Obstetrics and Gynecology and Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Claudio Villarroel
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis Velasquez
- Institute of Interdisciplinary Research in Biomedical Sciences (I3CBSEK), Faculty of Health Sciences, SEK University, Santiago, Chile
| | - Jerome F Strauss Iii
- Department of Obstetrics and Gynecology and Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Luigi Devoto
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
6
|
Rosario R, Stewart HL, Spears N, Telfer EE, Anderson RA. Anti-Mullerian hormone attenuates both cyclophosphamide-induced damage and PI3K signalling activation, while rapamycin attenuates only PI3K signalling activation, in human ovarian cortex in vitro. Hum Reprod 2024; 39:382-392. [PMID: 38070496 PMCID: PMC10833070 DOI: 10.1093/humrep/dead255] [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: 05/05/2023] [Revised: 11/13/2023] [Indexed: 02/02/2024] Open
Abstract
STUDY QUESTION What are the effects of cyclophosphamide exposure on the human ovary and can anti-Mullerian hormone (AMH) and rapamycin protect against these? SUMMARY ANSWER Exposure to cyclophosphamide compromises the health of primordial and transitional follicles in the human ovarian cortex and upregulates PI3K signalling, indicating both direct damage and increased follicular activation; AMH attenuates both of these chemotherapy-induced effects, while rapamycin attenuates only PI3K signalling upregulation. WHAT IS KNOWN ALREADY Studies primarily in rodents demonstrate that cyclophosphamide causes direct damage to primordial follicles or that the primordial follicle pool is depleted primarily through excessive initiation of follicle growth. This increased follicular activation is mediated via upregulated PI3K signalling and/or reduced local levels of AMH production due to lost growing follicles. Furthermore, while rodent data show promise regarding the potential benefits of inhibitors/protectants alongside chemotherapy treatment to preserve female fertility, there is no information about the potential for this in humans. STUDY DESIGN, SIZE, DURATION Fresh ovarian cortical biopsies were obtained from 17 healthy women aged 21-41 years (mean ± SD: 31.8 ± 4.9 years) at elective caesarean section. Biopsies were cut into small fragments and cultured for 24 h with either vehicle alone (DMSO), the active cyclophosphamide metabolite 4-hydroperoxycyclophosphamide (4-HC) alone, 4-HC + rapamycin or 4-HC+AMH. Two doses of 4-HC were investigated, 0.2 and 2 μM in separate experiments, using biopsies from seven women (aged 27-41) and six women (aged 21-34), respectively. Biopsies from four women (aged 28-38) were used to investigate the effect of rapamycin or AMH only. PARTICIPANTS/MATERIALS, SETTING, METHODS Histological analysis of ovarian tissue was undertaken for follicle staging and health assessment. Western blotting and immunostaining were used to assess activation of PI3K signalling by measuring phosphorylation of AKT and phosphorylated FOXO3A staining intensity, respectively. MAIN RESULTS AND THE ROLE OF CHANCE Exposure to either dose of 4-HC caused an increase in the proportion of unhealthy primordial (P < 0.0001, both doses) and transitional follicles (P < 0.01 for low dose and P < 0.01 for high dose) compared to vehicle. AMH significantly reduced follicle damage by approximately half in both of the investigated doses of 4-HC (P < 0.0001), while rapamycin had no protective effect on the health of the follicles. Culture with AMH or rapamycin alone had no effect on follicle health. Activation of PI3K signalling following 4-HC exposure was demonstrated by both Western blotting data showing that 4-HC increased in AKT phosphorylation and immunostaining showing increased phosphorylated FOXO3A staining of non-growing oocytes. Treatment with rapamycin reduced the activation of PI3K signalling in experiments with low doses of 4-HC while culture with AMH reduced PI3K activation (both AKT phosphorylation and phosphorylated FOXO3A staining intensity) across both doses investigated. LIMITATIONS, REASONS FOR CAUTION These in vitro studies may not replicate in vivo exposures. Furthermore, longer experiment durations are needed to determine whether the effects observed translate into irreparable deficits of ovarian follicles. WIDER IMPLICATIONS OF THE FINDINGS These data provide a solid foundation on which to explore the efficacy of AMH in protecting non-growing ovarian follicles from gonadotoxic chemotherapies. Future work will require consideration of the sustained effects of chemotherapy treatment and potential protectants to ensure these agents do not impair the developmental competence of oocytes or lead to the survival of oocytes with accumulated DNA damage, which could have adverse consequences for potential offspring. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from TENOVUS Scotland, the Academy of Medical Sciences (to R.R.), the Medical Research Council (G1100357 to R.A.A., MR/N022556/1 to the MRC Centre for Reproductive Health), and Merck Serono UK (to R.A.A.). R.R., H.L.S., N.S., and E.E.T. declare no conflicts of interest. R.A.A. reports grants and personal fees from Roche Diagnostics and Ferring Pharmaceuticals, and personal fees from IBSA and Merck outside the submitted work. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Roseanne Rosario
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Hazel L Stewart
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Norah Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Evelyn E Telfer
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Institute of Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
8
|
Chen X, Liu S, Peng X, Zong X. The roles of anti-Müllerian hormone in breast cancer. Endocr Relat Cancer 2023; 30:e230060. [PMID: 37410375 PMCID: PMC10448580 DOI: 10.1530/erc-23-0060] [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: 03/08/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
Anti-Müllerian hormone (AMH) is produced and secreted by granulosa cells of growing follicles, and its main role is to inhibit the recruitment of primordial follicles, reduce the sensitivity of follicles to follicle-stimulating hormone (FSH), and regulate FSH-dependent preantral follicle growth. It has become an effective indicator of ovarian reserve in clinical practice. Research on AMH and its receptors in recent years has led to a better understanding of its role in breast cancer. AMH specifically binds to anti-Müllerian hormone receptor II (AMHRII) to activate downstream pathways and regulate gene transcription. Since AMHRII is expressed in breast cancer cells and triggers apoptosis, AMH/AMHRII may play an important role in the occurrence, treatment, and prognosis of breast cancer, which needs further research. The AMH level is a potent predictor of ovarian function after chemotherapy in premenopausal breast cancer patients older than 35 years, either for ovarian function injury or ovarian function recovery. Moreover, AMHRII has the potential to be a new marker for the molecular typing of breast cancer and a new target for breast cancer treatment, which may be a link in the downstream pathway after TP53 mutation.
Collapse
Affiliation(s)
- Xuan Chen
- Department of Breast Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sixuan Liu
- Department of Breast Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Peng
- Department of Breast Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyun Zong
- Department of Breast Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Fraire-Zamora JJ, Sharma K, Ammar OF, Massarotti C, Ali ZE, Telfer EE, Williams S, Ata B, Liperis G. Mind the gap: deciphering the role of anti-Müllerian hormone in follicular development-from animal studies toward clinical application. Hum Reprod 2023:7131353. [PMID: 37075310 DOI: 10.1093/humrep/dead075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Indexed: 04/21/2023] Open
Affiliation(s)
| | - Kashish Sharma
- HealthPlus Fertility and Women's Health Centre, Abu Dhabi, UAE
| | - Omar F Ammar
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Claudia Massarotti
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- DINOGMI Department, University of Genova, Genova, Italy
| | - Zoya E Ali
- Research & Development Department, Hertility Health Limited, London, UK
| | - Evelyn E Telfer
- University of Edinburgh Institute of Cell Biology, Edinburgh, UK
| | - Suzannah Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Baris Ata
- Obstetrics and Gynecology Department, Koc University, Istanbul, Turkey
- ART Fertility Clinics, Dubai, United Arab Emirates
| | | |
Collapse
|
10
|
Ataei-Nazari S, Amoushahi M, Madsen JF, Jensen J, Heuck A, Mohammadi-Sangcheshmeh A, Lykke-Hartmann K. Cyclin-dependent kinase 6 (CDK6) as a potent regulator of the ovarian primordial-to-primary follicle transition. Front Cell Dev Biol 2022; 10:1036917. [PMID: 36619863 PMCID: PMC9816807 DOI: 10.3389/fcell.2022.1036917] [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: 09/05/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction: Ovarian follicle development requires tight coordination between several factors to initiate folliculogenesis to generate a mature and fertile egg. Studies have shown that cell cycle factors might contribute to follicle development, hover specific knowledge on individual CDKs and follicle activation has not been investigated. Among cell cycle regulators, CDK6 is a key player through binding to cyclin D resulting DNA synthesis and genome duplication. Interestingly, the CDK6 gene is differentially expressed in oocytes and granulosa cells from human primordial and primary follicles, which suggest a potential role of CDK6 in the primordial-to-primary transition. In this study, we investigated the potential regulatory role of CDK6 in progression of primordial to primary follicle transition using BSJ-03-123 (BSJ), a CDK6-specific degrader. Methods: In mouse ovarian in vitro culture, BSJ reduced the activation of primordial follicles, and reduced follicle development. As a next step, we examined the egg maturation read-out and found that BSJ-treated follicles matured to competent MII eggs with resumption of first meiosis, comparable with the control group. Results: Noteworthy, it appears that inhibition of CDK6 did increase number of apotoptic cells, articular in the granulosa cells, but had no impact on ROS level of cultured ovaries compared to control group, indicating that the cells were not stressed. Oocyte quality thus appeared safe. Discussion: The results of this study indicate that CDK6 plays a role in the primordial-to-primary transition, suggesting that cell cycle regulation is an essential part of ovarian follicle development.
Collapse
Affiliation(s)
- S. Ataei-Nazari
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - M. Amoushahi
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - JF. Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - J. Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - A. Heuck
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - K. Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark,Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark,*Correspondence: K. Lykke-Hartmann,
| |
Collapse
|
11
|
Aydogan Mathyk B, Cetin E, Yildiz BO. Use of anti-Müllerian hormone for understanding ovulatory dysfunction in polycystic ovarian syndrome. Curr Opin Endocrinol Diabetes Obes 2022; 29:528-534. [PMID: 36218229 DOI: 10.1097/med.0000000000000772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to understand how anti-Müllerian hormone (AMH) contributes to ovulatory dysfunction in polycystic ovarian syndrome (PCOS). RECENT FINDINGS In the last few years, new findings have emerged on AMH and its role on the central nervous system causing ovulatory dysfunction. SUMMARY Anovulation is a prominent feature of PCOS. Women with anovulatory PCOS have higher AMH levels than in ovulatory PCOS. Higher levels of AMH may contribute to the pathophysiology of PCOS through central and peripheral actions. Once universal standardization is achieved to measure serum AMH, the benefits would be significant in diagnosing women with PCOS.
Collapse
Affiliation(s)
- Begum Aydogan Mathyk
- Department of Obstetrics and Gynecology, HCA/University of South Florida Morsani College of Medicine GME, Brandon Regional Hospital, Brandon, Florida
| | - Esra Cetin
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, Illinois, USA
| | - Bulent O Yildiz
- Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Hacettepe, Ankara, Turkey
| |
Collapse
|
12
|
Zhou Y, Richard S, Batchelor NJ, Oorschot DE, Anderson GM, Pankhurst MW. Anti-Müllerian hormone-mediated preantral follicle atresia is a key determinant of antral follicle count in mice. Hum Reprod 2022; 37:2635-2645. [PMID: 36107143 PMCID: PMC9627584 DOI: 10.1093/humrep/deac204] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 08/23/2022] [Indexed: 07/21/2023] Open
Abstract
STUDY QUESTION Does anti-Müllerian hormone (AMH) induce preantral follicle atresia in mice? SUMMARY ANSWER The present findings suggest that AMH-mediated follicle atresia only occurs in early follicles before they become sensitive to FSH. WHAT IS KNOWN ALREADY Most prior studies have investigated the ability of AMH to inhibit primordial follicle activation. Our previous study showed that AMH-overexpressing mice had fewer preantral follicles than expected after accounting for primordial follicle inhibition but the reason for this was not determined. STUDY DESIGN, SIZE, DURATION Cross-sectional-control versus transgenic/knockout mouse studies were carried out. PARTICIPANTS/MATERIALS, SETTING, METHODS Studies were conducted on female wild-type (Amh+/+), AMH-knockout (Amh-/-) and AMH overexpressing (Thy1.2-AMHTg/0) mice on a C57Bl/6J background (age: 42-120 days). The follicle counts were conducted for primordial, transitioning, primary, secondary and antral follicles in Amh-/- and Amh+/+ mice. After confirming that follicle development speeds were identical (proliferating cell nuclear antigen immunohistochemistry), the ratio of follicles surviving beyond each stage of folliculogenesis was determined in both genotypes. Evidence for increased rates of preantral follicle atresia was assessed by active caspase-3 immunohistochemistry in wild-type and Thy1.2-AMHTg/0 mice. MAIN RESULTS AND THE ROLE OF CHANCE Amh -/- mice at 100-120 days of age had lower primordial follicle counts but higher primordial follicle activation rates compared to Amh+/+ mice. These counteracting effects led to equivalent numbers of primordial follicles transitioning to the primary stage in Amh+/+ and Amh-/- mice. Despite this, Amh+/+ mice had fewer primary, secondary, small antral and medium antral follicles than Amh-/- mice indicating differing rates of developing follicle atresia between genotypes. Cleaved caspase-3 immunohistochemistry in Thy1.2-AMHTg/0 ovaries revealed high rates of granulosa cell and oocyte apoptosis in late primary/early secondary follicles of Thy1.2-AMHTg/0 mice. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The findings were shown only in one species and additional research will be required to determine generalizability to other species. WIDER IMPLICATIONS OF THE FINDINGS This study is consistent with prior studies showing that Amh-/- mice have increased primordial follicle activation but these new findings demonstrate that AMH-mediated preantral follicle atresia is a predominant cause of the increased small antral follicle counts in Amh-/- mice. This suggests that the role of AMH is not to conserve the ovarian reserve to prolong fertility, but instead to prevent the antral follicle pool from becoming too large. While this study may demonstrate a new function for AMH, the biological purpose of this function requires further investigation, particularly in mono-ovulatory species. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the Health Research Council of New Zealand and the University of Otago. No competing interests to declare.
Collapse
Affiliation(s)
- Y Zhou
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - S Richard
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - N J Batchelor
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - D E Oorschot
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - G M Anderson
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - M W Pankhurst
- Correspondence address. Department of Anatomy, University of Otago, PO Box 56, Dunedin 9054, New Zealand. Tel: +64-3-479-7440; E-mail:
| |
Collapse
|
13
|
Liu X, Han Y, Wang X, Zhang Y, Du A, Yao R, Lv J, Luo H. Serum anti-Müllerian hormone levels are associated with early miscarriage in the IVF/ICSI fresh cycle. BMC Pregnancy Childbirth 2022; 22:279. [PMID: 35366830 PMCID: PMC8976292 DOI: 10.1186/s12884-022-04591-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Anti-Müllerian hormone (AMH) is used as a biomarker to estimate ovarian reserve. The relationship between AMH and early miscarriage of in vitro fertilization (IVF) is still inconclusive. This study aimed to explore whether serum AMH levels are associated with early miscarriage rates after in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) with fresh embryo transfer (ET). METHODS This retrospective cohort study included 2246 patients undergoing their first oocyte retrievals for IVF/ICSI with fresh embryos transferred to Tianjin Central Hospital of Gynecology Obstetrics between May 2018 and March 2020. The serum AMH levels of the patients were measured within 12 months before the IVF/ICSI cycles. All women were divided into a low-AMH group, medium-AMH group and high-AMH group. Binary logistic regression was applied to confirm whether the serum AMH level was associated with the risk of early miscarriage independent of potential confounders, such as age, body mass index (BMI), duration of infertility, main diagnosis, history of internal medicine diseases, number of oocytes retrieved and high-quality embryo rate. RESULTS The early miscarriage rate was significantly lower in the medium-AMH group than in either the low-AMH or high-AMH group among young (< 35 years) women (P = 0.015). In women above 35 years of age, the early miscarriage rates in the three AMH groups were not significantly different. Young women with high serum AMH levels had a significantly higher risk of early miscarriage regardless of age or other potential confounders (adjusted odds ratio (OR) 2.382, 95% confidence interval (CI) 1.246 to 4.553, P = 0.009). The results remained similar after restricting the analysis to women without polycystic ovary syndrome (PCOS). CONCLUSIONS With a high AMH level, young women had a higher risk of early miscarriage than women with a medium AMH level in their first IVF/ICSI treatment. In young women, serum AMH levels were independently associated with the risk of early miscarriage after IVF-ET treatment. Serum AMH levels might be a valuable marker to estimate the risk of early miscarriage. It is worth noting to the clinical value of AMH.
Collapse
Affiliation(s)
- Xin Liu
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China
| | - Ying Han
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China
| | - Xinyan Wang
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China
| | - Yunshan Zhang
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China
| | - Aijun Du
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China
| | - Ruqiang Yao
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China
| | - Jiabei Lv
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China
| | - Haining Luo
- Centre for Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nankai San Ma Road, Tianjin, 300100, China.
| |
Collapse
|
14
|
Huang QY, Chen SR, Chen JM, Shi QY, Lin S. Therapeutic options for premature ovarian insufficiency: an updated review. Reprod Biol Endocrinol 2022; 20:28. [PMID: 35120535 PMCID: PMC8815154 DOI: 10.1186/s12958-022-00892-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/15/2022] [Indexed: 11/16/2022] Open
Abstract
Primary ovarian insufficiency (POI) is a rare gynecological condition. This disease causes menstrual disturbances, infertility, and various health problems. Historically, hormone replacement therapy is the first-line treatment for this disorder. Women diagnosed with POI are left with limited therapeutic options. In order to remedy this situation, a new generation of therapeutic approaches, such as in vitro activation, mitochondrial activation technique, stem cell and exosomes therapy, biomaterials strategies, and platelet-rich plasma intra-ovarian infusion, is being developed. However, these emerging therapies are yet in the experimental stage and require precise design components to accelerate their conversion into clinical treatments. Thus, each medical practitioner bears responsibility for selecting suitable therapies for individual patients. In this article, we provide a timely analysis of the therapeutic strategies that are available for POI patients and discuss the prospects of POI therapy.
Collapse
Affiliation(s)
- Qiao-Yi Huang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Shao-Rong Chen
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Jia-Ming Chen
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Qi-Yang Shi
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China.
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China.
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
| |
Collapse
|
15
|
Wang YW, Wu CH, Lin TY, Luo CW. Expression profiling of ovarian BMP antagonists reveals the potential interaction between TWSG1 and the chordin subfamily in the ovary. Mol Cell Endocrinol 2021; 538:111457. [PMID: 34517078 DOI: 10.1016/j.mce.2021.111457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022]
Abstract
The TGF-β superfamily members and their antagonists comprise an indispensable system that controls mammalian ovarian development in a sophisticated manner. In contrast to a plethora of studies on the ovary-expressed TGF-β superfamily members, knowledge regarding their antagonists, including their expression profiles and antagonism preferences, is still lacking. Using quantitative PCR in rats and transcriptomic dataset comparisons in mice and humans, we set out to characterize the relative expression levels of most antagonists in the mammalian ovary. We found that Twsg1 and Nbl1 are the most abundant BMP antagonists expressed in the rodent and human ovaries, respectively. TWSG1 has been reported to have synergistic action with the chordin subfamily, including CHRD and CHRDL1, the genes of which also showed moderate expression in the mammalian ovary. Therefore, their ovarian expression profiles and antagonisms against the ovary-expressed TGF-β superfamily members were further characterized. Bioactivity tests indicated that TWSG1 alone can directly inhibit the signaling of BMP6 or BMP7. In addition, it can further enhance the antagonizing ability of CHRD towards BMP2, BMP4, BMP7 and GDF5, or CHRDL1's antagonism towards BMP2, BMP4, GDF5 and activin A. In combination with their distinct transcript profiles in ovarian compartments, our findings suggest that TWSG1 may work coordinately with CHRD within theca/interstitial shells and also with CHRDL1 in developing granulosa cells; these interactions would modulate the intraovarian functions of the TGF-β superfamily members, such as the control of progesterone production.
Collapse
Affiliation(s)
- Ying-Wen Wang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chia-Hsu Wu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Ting-Yu Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Ching-Wei Luo
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
16
|
di Clemente N, Racine C, Pierre A, Taieb J. Anti-Müllerian Hormone in Female Reproduction. Endocr Rev 2021; 42:753-782. [PMID: 33851994 DOI: 10.1210/endrev/bnab012] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Joëlle Taieb
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| |
Collapse
|
17
|
Hao X, Anastácio A, Viñals-Ribé L, Santamaria Lacuesta A, Diakaki C, Alonso de Mena S, Liu K, Rodriguez-Wallberg KA. Follicle Rescue From Prepubertal Ovaries After Recent Treatment With Cyclophosphamide-An Experimental Culture System Using Mice to Achieve Mature Oocytes for Fertility Preservation. Front Oncol 2021; 11:682470. [PMID: 34631518 PMCID: PMC8497963 DOI: 10.3389/fonc.2021.682470] [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: 03/18/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Ovarian tissue cryopreservation is the only feasible method for fertility preservation in prepubertal girls that will undergo gonadotoxic chemotherapy. To date, the only clinical use of cryopreserved tissue is by a later tissue retransplantation to the patient. Clinical challenges in fertility preservation of very young patients with cancer include time constraints that do not allow to retrieve the tissue for cryopreservation before starting chemotherapy and the preclusion of future ovarian tissue transplantation due to the risk of reintroduction of malignant cells in patients with systemic diseases. To overcome these two challenges, we investigated using an experimental model the feasibility of retrieving secondary follicles from ovaries of prepubertal mice after cyclophosphamide (CPA) treatment in increasing doses of 50, 75, and 100 mg/kg. The follicles were thereafter cultured and matured in vitro. The main outcomes included the efficiency of the method in terms of obtained matured oocytes and the safety of these potentially fertility preservative procedures in terms of analyses of oocyte competence regarding normality of the spindle and chromosome configurations. Our findings demonstrated that it was feasible to isolate and culture secondary follicles and to obtain mature oocytes from prepubertal mice ovaries recently treated with CPA. The efficiency of this method was highly demonstrated in the 100 mg/kg CPA group, with near 90% follicle survival rate after 12 days' culture, similarly to control. Around 80% of the follicles met the criteria to put into maturation, and more than 40% of them achieved metaphase II, with normal spindle and chromosome configurations observed. Suboptimal results were obtained in the 50 and 75 mg/kg CPA groups. These paradoxical findings towards CPA dose might probably reflect a more difficult selection of damaged growing follicles from ovaries recently treated with lower doses of CPA and a hampered ability to identify and discard those with reduced viability for the culture.
Collapse
Affiliation(s)
- Xia Hao
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Amandine Anastácio
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Laia Viñals-Ribé
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Ana Santamaria Lacuesta
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Christina Diakaki
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Sara Alonso de Mena
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Kui Liu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong, SAR, China
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden.,Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
18
|
Harris BS, Steiner AZ, Jukic AM. Ovarian Reserve Biomarkers and Menstrual Cycle Length in a Prospective Cohort Study. J Clin Endocrinol Metab 2021; 106:e3748-e3759. [PMID: 33772306 PMCID: PMC8372629 DOI: 10.1210/clinem/dgab204] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT While age-related changes in menstrual cycle length are well known, it is unclear whether anti-Müllerian (AMH) or other ovarian reserve biomarkers have a direct association with cycle length. OBJECTIVE To determine the association between biomarkers of ovarian reserve and menstrual cycle length. METHODS Secondary analysis using data from time to conceive (TTC), a prospective time to pregnancy cohort study. The age-independent association between cycle length and biomarkers of ovarian reserve was analyzed using linear mixed and marginal models. Study participants were TTC-enrolled women aged 30-44 years with no history of infertility who were attempting to conceive for <3 months were enrolled. Serum AMH, follicle-stimulating hormone, and inhibin B levels were measured on cycle day 2, 3, or 4. Participants recorded daily menstrual cycle data for ≤4 months. The primary outcome was menstrual cycle length; follicular and luteal phase lengths were secondary outcomes. RESULTS Multivariable analysis included 1880 cycles from 632 women. Compared with AMH levels of 1.6 to 3.4 ng/mL, women with AMH <1.6 ng/mL had cycles and follicular phases that were 0.98 (95% CI -1.46, -0.50) and 1.58 days shorter (95% CI -2.53, -0.63), respectively, while women with AMH >8 ng/mL had cycles that were 2.15 days longer (95% CI 1.46, 2.83), follicular phases that were 2 days longer (95% CI 0.77, 3.24), and luteal phases that were 1.80 days longer (95% CI 0.71, 2.88). CONCLUSION Increasing AMH levels are associated with longer menstrual cycles due to both a lengthening of the follicular and the luteal phase independent of age.
Collapse
Affiliation(s)
- Benjamin S Harris
- Division of Reproductive Endocrinology and Infertility, Duke University Medical Center, Durham, NC 27703, USA
| | - Anne Z Steiner
- Division of Reproductive Endocrinology and Infertility, Duke University Medical Center, Durham, NC 27703, USA
| | - Anne Marie Jukic
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), Durham, NC 27703, USA
| |
Collapse
|
19
|
Song Y, Liu H. A review on the relationship between anti-mullerian hormone and fertility in treating young breast cancer patients. BMC WOMENS HEALTH 2021; 21:295. [PMID: 34376160 PMCID: PMC8353739 DOI: 10.1186/s12905-021-01420-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/06/2021] [Indexed: 11/18/2022]
Abstract
Despite the fact that the long-term survival rate of breast cancer patients had been significantly improved owing to the systemic breast cancer therapies, there are still some side effects such as amenorrhea and fertility retention to be resolved, leaving it an important thing to understand the possible side effects on fertility and fertility preservation strategies while undergoing breast cancer treatment, due to the fact that most young patients hope to become pregnant and have children after breast cancer treatment. With anti-müllerian hormone (AMH) being the most sensitive marker for predicting ovarian function in young premenopausal women with breast cancer, this review is aimed to provide the additional guidance for clinical application of AMH by exploring the impacts of AMH on the fertility of young breast cancer patients, the relationship between AMH and metabolism, and the relationship between BRAC gene mutation and fertility protection strategies.
Collapse
Affiliation(s)
- Yixuan Song
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
20
|
Current Understandings of Core Pathways for the Activation of Mammalian Primordial Follicles. Cells 2021; 10:cells10061491. [PMID: 34199299 PMCID: PMC8231864 DOI: 10.3390/cells10061491] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
The mammalian ovary has two main functions-producing mature oocytes for fertilization and secreting hormones for maintaining the ovarian endocrine functions. Both functions are vital for female reproduction. Primordial follicles are composed of flattened pre-granulosa cells and a primary oocyte, and activation of primordial follicles is the first step in follicular development and is the key factor in determining the reproductive capacity of females. The recent identification of the phosphatidylinositol 3 kinase (PI3K)/phosphatase and tensin homolog deleted on chromosome 10 (PTEN) signaling pathway as the key controller for follicular activation has made the study of primordial follicle activation a hot research topic in the field of reproduction. This review systematically summarizes the roles of the PI3K/PTEN signaling pathway in primordial follicle activation and discusses how the pathway interacts with various other molecular networks to control follicular activation. Studies on the activation of primordial follicles have led to the development of methods for the in vitro activation of primordial follicles as a treatment for infertility in women with premature ovarian insufficiency or poor ovarian response, and these are also discussed along with some practical applications of our current knowledge of follicular activation.
Collapse
|
21
|
Henríquez S, Kohen P, Xu X, Villarroel C, Muñoz A, Godoy A, Strauss JF, Devoto L. Significance of pro-angiogenic estrogen metabolites in normal follicular development and follicular growth arrest in polycystic ovary syndrome. Hum Reprod 2021; 35:1655-1665. [PMID: 32558920 DOI: 10.1093/humrep/deaa098] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Do alterations in pro- and anti-angiogenic estrogen metabolites in follicular fluid (FF) contribute to the follicular growth arrest and anovulation associated with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER FF of PCOS women with anovulation have reduced levels of pro-angiogenic estrogen metabolites (EMs) and vascular endothelial growth factor (VEGF) compared to that of fertile women with regular menstrual cycles, but exogenous gonadotropins increase the pro-angiogenic EMs and VEGF levels in PCOS women. WHAT IS KNOWN ALREADY PCOS is characterized by the arrest of follicular development that leads to chronic anovulation. Follicular arrest is generally associated with elevated plasma levels of luteinizing hormone (LH), androgens and anti-Mullerian hormone (AMH). There is also reduced angiogenesis in the follicles of PCOS women compared to those of normal cycling women. It is known that angiogenesis is a critical factor during follicular development. We and other investigators have explored the role of EMs in ovarian angiogenesis, particularly in human corpus luteum function, showing that 4-hydroxyestrone (4-OHE1) and 16-ketoestradiol (16-kE2) have pro-angiogenic effects while 2-methoxyestradiol (2-ME2) and 2-methoxyestrone (2-ME1) have anti-angiogenic effects. Additionally, 2-hydroxyestradiol (2-OHE2), which is produced in the ovary, has proliferative and pro-angiogenic properties. We hypothesized that EMs could be involved in angiogenesis necessary for ovarian follicular development in fertile women, and that dysregulation of these factors may contribute to follicular arrest in PCOS. The relationship between EMs, VEGF and AMH in the pathophysiology of follicular arrest in PCOS has not been previously studied at a follicular level in anovulatory women without ovulation induction. STUDY DESIGN, SIZE, DURATION This is a comparative experimental study of serum and FF collected from different sized follicles (antral ˂10 mm and dominant ˃16 mm) of women with and without ovarian stimulation. The study included women with regular menstrual cycles who were proven to be fertile (n = 20) and PCOS women with follicular arrest who were candidates for ovarian drilling (n = 17), as well as other patients requiring ovarian stimulation, i.e. control women undergoing IVF for male factor infertility (n = 12) and PCOS women undergoing IVF (n = 17). In vitro studies were carried out on granulosa-lutein cells (GCs) obtained from subsets of women undergoing IVF for male factor infertility (n = 6) and PCOS women undergoing IVF (n = 6). GCs were maintained in culture for up to 6 days. PARTICIPANTS/MATERIALS, SETTING, METHODS Intrafollicular estradiol, estrone and EMs concentrations were determined by high performance liquid chromatography-mass spectrometry. Testosterone in serum was measured by RIA, and LH, FSH and sex hormone-binding globulin in serum were measured with IRMA kits. AMH was determined in serum and FF by enzyme linked immunosorbant assay (ELISA). VEGF levels were measured in FF and conditioned medium by ELISA. Conditioned medium were obtained from cultured GCs. The angiogenic potential was assessed by in vitro angiogenic assays. MAIN RESULTS AND THE ROLE OF CHANCE Pro-angiogenic EMs (4-OHE1, 16-kE2 and 2-OHE2) and VEGF were lower in FF of antral follicles of PCOS women with follicular arrest compared those of fertile women with ovulatory cycles (P < 0.05). In contrast, higher concentrations of AMH were found in FF of antral follicles from PCOS women with follicular arrest compared to those of fertile women with ovulatory cycles (P < 0.05). Exogenous gonadotropins used in IVF increased pro-angiogenic EMs and VEGF production in PCOS women, reaching similar profiles compared to control women receiving gonadotropins in their IVF treatment for male factor infertility. The pro-angiogenic EM 2-OHE2 increased the angiogenic potential and VEGF levels of GCs from PCOS women compared to the basal condition (P < 0.05). These findings suggest that there is a role for pro-angiogenic EMs in the control of follicular VEGF production. LIMITATIONS, REASONS FOR CAUTION The limitations include the possibility that in vitro analysis of GCs might not reflect the in vivo mechanisms involved in the pro-angiogenic action of 2-OHE2 since GCs obtained at the time of oocyte retrieval belong to a very early stage of the luteal phase and might not be representative of GCs during follicular growth. Therefore, our findings do not conclusively rule out the possibility that other in vivo mechanisms also account for defective angiogenesis observed in PCOS. WIDER IMPLICATIONS OF THE FINDINGS The present study highlights the significance of EMs, angiogenic factors and AMH and their interaction in the pathophysiology of follicular development in PCOS. This study provides new insights into the role of pro-angiogenic factors in follicular arrest in PCOS. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by CONICYT/FONDECYT 1140693 and NIH grant R01HD083323. All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Soledad Henríquez
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile.,Institute of Interdisciplinary Research in Biomedical Sciences (I3CBSEK), Faculty of Health Sciences, SEK University, Santiago, Chile
| | - Paulina Kohen
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Xia Xu
- Research Technology Program, Biomedical Research (formerly SAIC-Frederick), Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Claudio Villarroel
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Obstetrics and Gynecology, Faculty of Medicine, San Borja-Arriaran Clinical Hospital, University of Chile, Santiago, Chile
| | - Alex Muñoz
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Obstetrics and Gynecology, Faculty of Medicine, San Borja-Arriaran Clinical Hospital, University of Chile, Santiago, Chile
| | - Ana Godoy
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Luigi Devoto
- Institute for Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Obstetrics and Gynecology, Faculty of Medicine, San Borja-Arriaran Clinical Hospital, University of Chile, Santiago, Chile
| |
Collapse
|
22
|
Vo KCT, Kawamura K. In Vitro Activation Early Follicles: From the Basic Science to the Clinical Perspectives. Int J Mol Sci 2021; 22:ijms22073785. [PMID: 33917468 PMCID: PMC8038686 DOI: 10.3390/ijms22073785] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/29/2021] [Accepted: 04/03/2021] [Indexed: 12/16/2022] Open
Abstract
Development of early follicles, especially the activation of primordial follicles, is strictly modulated by a network of signaling pathways. Recent advance in ovarian physiology has been allowed the development of several therapies to improve reproductive outcomes by manipulating early folliculogenesis. Among these, in vitro activation (IVA) has been recently developed to extend the possibility of achieving genetically related offspring for patients with premature ovarian insufficiency and ovarian dysfunction. This method was established based on basic science studies of the intraovarian signaling pathways: the phosphoinositide 3-kinase (PI3K)/Akt and the Hippo signaling pathways. These two pathways were found to play crucial roles in folliculogenesis from the primordial follicle to the early antral follicle. Following the results of rodent experiments, IVA was implemented in clinical practice. There have been multiple recorded live births and ongoing pregnancies. Further investigations are essential to confirm the efficacy and safety of IVA before used widely in clinics. This review aimed to summarize the published literature on IVA and provide future perspectives for its improvement.
Collapse
|
23
|
Robertson DM, Lee CH, Baerwald A. Interactions between serum FSH, inhibin B and antral follicle count in the decline of serum AMH during the menstrual cycle in late reproductive age. Endocrinol Diabetes Metab 2021; 4:e00172. [PMID: 33855196 PMCID: PMC8029535 DOI: 10.1002/edm2.172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/30/2020] [Accepted: 07/04/2020] [Indexed: 11/15/2022] Open
Abstract
Objective To investigate the hormonal interrelationships during the menstrual cycle in women of late reproductive age with suppressed serum AMH and antral follicle count (AFC). Methods Serum hormones (AMH, FSH, LH, estradiol, progesterone, inhibin A, inhibin B), AFC (2-10 mm) and AMH/AFC ratio (an estimate of AMH/follicle) were assessed every 2-3 days across the menstrual cycle in 26 healthy ovulatory women aged 18-50 years. Results An 11-fold fall in AMH/AFC was observed in women aged ≥45 years compared to those 18-45 years (P < .001). Although women ≥45 years exhibited normal menstrual cycle patterns of serum estradiol, progesterone, LH and inhibin A, FSH was elevated (P < .001) and inhibin B suppressed (P < .001) compared to the younger group. Overall FSH was inversely correlated (r = .55, P < .05) and AMH directly correlated (r = .88, P < .01) with AFC; however, these relationships were curvilinear and more pronounced when AFC was low. Inhibin B was directly linearly correlated (r = .70, P < .01) with AFC across both high and low AMH/follicle groups. Conclusions It is hypothesized that the marked fall in AMH/follicle in late reproductive age is attributed to the change in the hormonal interplay between the pituitary and ovary. The fall in AFC leads to a decrease in inhibin B and a concomitant increase in FSH by a recognized feedback mechanism. It is postulated the elevated FSH suppresses AMH either directly or indirectly through oocyte-specific growth factors leading to a marked fall in AMH/follicle. We propose that pituitary-ovarian and intra-ovarian regulatory systems underpin the accelerated fall in AMH/follicle during the transition to menopause.
Collapse
Affiliation(s)
- David M. Robertson
- Centre for Endocrinology and MetabolismHudson Institute of Medical Research, Clayton, VictoriaMonash UniversityClaytonVictoriaAustralia
- School of Women’s and Children’s HealthUniversity of New South WalesKensingtonAustralia
| | - Chel Hee Lee
- Department of Mathematics and StatisticsUniversity of CalgaryAlbertaCanada
| | - Angela Baerwald
- Department of Academic Family MedicineCollege of MedicineUniversity of SaskatchewanSaskatchewanCanada
| |
Collapse
|
24
|
Pourjafari F, Haghpanah T, Sharififar F, Nematollahi-Mahani SN, Afgar A, Ezzatabadipour M. Evaluation of expression and serum concentration of anti-Mullerian hormone as a follicle growth marker following consumption of fennel and flaxseed extract in first-generation mice pups. BMC Complement Med Ther 2021; 21:90. [PMID: 33711998 PMCID: PMC7953604 DOI: 10.1186/s12906-021-03267-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/28/2021] [Indexed: 11/22/2022] Open
Abstract
Background The aim of the present study was to assess the expression and serum level of AMH in first-generation female mice pups following fennel and flaxseed consumption. Methods Twenty pregnant NMRI mice were allocated into four groups including control (CTL), fennel (FV), flaxseed (LU) and FV+ LU. Sixty-four female offsprings after lactation period, received the same regimen as their mothers for 56 and 240 days. The ovarian follicles development, serum concentration of AMH, as well as gene and protein expression of AMH were evaluated in the female offsprings at post-natal day 56 (PND56) and 240 (PND240). Results The number of total growing follicles were raised in the FV group in compression to the all experimental groups. In contrast, LU group showed a marked decrease in their numbers. The highest level of serum AMH was seen in the FV-diet mice, whereas LU negatively affected it. The expression level of AMH also increased in the FV and FV + LU groups, while a reduction was observed in the LU group. As well, IHC data showed that the number of AMH-positive cells in almost ovarian follicles of FV and FV + LU-treated mice was in compared to those of the LU group. Conclusions The overall effect of fennel treatment (alone and in combination with flaxseed) on ovary might be maintain primordial follicle storage through increased expression and serum level of AMH.
Collapse
Affiliation(s)
- Fahimeh Pourjafari
- Anatomical Sciences Department, School of Medicine, Kerman University of Medical Sciences, P.O. Box:76169-14115, Kerman, Iran
| | - Tahereh Haghpanah
- Anatomical Sciences Department, School of Medicine, Kerman University of Medical Sciences, P.O. Box:76169-14115, Kerman, Iran
| | - Fariba Sharififar
- Herbal and Traditional Medicines Research Center, Department of Pharmacognosy, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Ali Afgar
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Massood Ezzatabadipour
- Anatomical Sciences Department, School of Medicine, Kerman University of Medical Sciences, P.O. Box:76169-14115, Kerman, Iran.
| |
Collapse
|
25
|
Racine C, Genêt C, Bourgneuf C, Dupont C, Plisson-Petit F, Sarry J, Hennequet-Antier C, Vigouroux C, Mathieu d'Argent E, Pierre A, Monniaux D, Fabre S, di Clemente N. New Anti-Müllerian Hormone Target Genes Involved in Granulosa Cell Survival in Women With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2021; 106:e1271-e1289. [PMID: 33247926 DOI: 10.1210/clinem/dgaa879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE A protective effect of anti-Müllerian hormone (AMH) on follicle atresia was recently demonstrated using long-term treatments, but this effect has never been supported by mechanistic studies. This work aimed to gain an insight into the mechanism of action of AMH on follicle atresia and on how this could account for the increased follicle pool observed in women with polycystic ovary syndrome (PCOS). METHODS In vivo and in vitro experiments were performed to study the effects of AMH on follicle atresia and on the proliferation and apoptosis of granulosa cells (GCs). RNA-sequencing was carried out to identify new AMH target genes in GCs. The expression of some of these genes in GCs from control and PCOS women was compared using microfluidic real time quantitative RT-PCR. RESULTS A short-term AMH treatment prevented follicle atresia in prepubertal mice. Consistent with this result, AMH inhibited apoptosis and promoted proliferation of different models of GCs. Moreover, integrative biology analyses of 965 AMH target genes identified in 1 of these GC models, confirmed that AMH had initiated a gene expression program favoring cell survival and proliferation. Finally, on 43 genes selected among the most up- and down-regulated AMH targets, 8 were up-regulated in GCs isolated from PCOS women, of which 5 are involved in cell survival. MAIN CONCLUSIONS Our results provide for the first time cellular and molecular evidence that AMH protects follicles from atresia by controlling GC survival and suggest that AMH could participate in the increased follicle pool of PCOS patients.
Collapse
Affiliation(s)
- Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Carine Genêt
- GenPhySE, Université de Toulouse, INRAE, INP, ENVT, Castanet-Tolosan, France
| | - Camille Bourgneuf
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Charlotte Dupont
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | | | - Julien Sarry
- GenPhySE, Université de Toulouse, INRAE, INP, ENVT, Castanet-Tolosan, France
| | - Christelle Hennequet-Antier
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Corinne Vigouroux
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Emmanuelle Mathieu d'Argent
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Danielle Monniaux
- Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Stéphane Fabre
- GenPhySE, Université de Toulouse, INRAE, INP, ENVT, Castanet-Tolosan, France
| | - Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| |
Collapse
|
26
|
Vo KCT, Kawamura K. Ovarian Fragmentation and AKT Stimulation for Expansion of Fertile Lifespan. FRONTIERS IN REPRODUCTIVE HEALTH 2021; 3:636771. [PMID: 36304045 PMCID: PMC9580792 DOI: 10.3389/frph.2021.636771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/22/2021] [Indexed: 11/13/2022] Open
Abstract
Since the first baby was born after in vitro fertilization, the female infertility treatment has been well-developed, yielding successful outcomes. However, successful pregnancies for patients with premature ovarian insufficiency and diminished ovarian reserve are still difficult and diverse therapies have been suggested to improve the chances to have their genetically linked offspring. Recent studies demonstrated that the activation Akt pathway by using a phosphatase and tensin homolog enzyme inhibitor and a phosphatidylinositol-3 kinase stimulator can activate dormant primordial follicles in both mice and human ovaries. Subsequent researches suggested that the disruption of Hippo signaling pathway by ovarian fragmentation increased the expression of downstream growth factors and secondary follicle growth. Based on the combination of ovarian fragmentation and Akt stimulation, the in vitro activation (IVA) approach has resulted in successful follicle growth and live births in premature ovarian insufficiency patients. The approach with disruption of Hippo signaling only was also shown to be effective for treating poor ovarian responders with diminishing ovarian reserve, including advanced age women and cancer patients undergoing sterilizing treatments. This review aims to summarize the effectiveness of ovarian fragmentation and Akt stimulation on follicle growth and the potential of IVA in extending female fertile lifespan.
Collapse
|
27
|
Hu S, Zhu M, Wang J, Li L, He H, Hu B, Hu J, Xia L. Histomorphology and gene expression profiles during early ovarian folliculogenesis in duck and goose. Poult Sci 2021; 100:1098-1108. [PMID: 33518069 PMCID: PMC7858004 DOI: 10.1016/j.psj.2020.10.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/15/2020] [Accepted: 10/01/2020] [Indexed: 11/24/2022] Open
Abstract
In contrast to the later stages of follicle development, little is known about the characteristics and mechanisms associated with early folliculogenesis in avian species. The objectives of the present study were to examine and compare the histomorphological and molecular changes of primordial, primary, and secondary follicles from duck and goose ovaries during the first 6 post-hatching week. Morphological analysis showed that the length and width of both duck and goose ovaries increased steadily during weeks 1 to 5 but increased acutely at week 6, whereas a greater increment was observed in the ovarian length of ducks than that of geese during weeks 4 to 5. Furthermore, smaller diameters of the 3 categories of follicles were observed in ducks than those in geese at the first appearance, but they reached a similar size at week 6. More importantly, secondary follicles were found in the ovaries of ducks 1 wk earlier than in those of geese. These results indicated a more rapid growth rate for ovarian follicles in ducks than in geese during early post-hatching development. At the molecular level, it was found that the mRNAs encoding follicle stimulating hormone receptor (FSHR), anti-Müllerian hormone (AMH), B-cell leukemia/lymphoma 2, and cysteine-dependent aspartate specific protease 3 (CASPASE3) were ubiquitously expressed in all ovarian follicles of ducks and geese with different expression profiles in each follicular category during the first 6 post-hatching week. Notably, transcript levels of FSHR, AMH, and CASPASE3 changed differently between ducks and geese during weeks 5 to 6, which was postulated to be one of the mechanisms inducing more rapid growth of ovarian follicles in ducks rather than in geese. In conclusion, our results revealed, for the first time, differences in early folliculogenesis, including the rate of growth of each follicular category and the timing of transition of primary to secondary follicles, between ducks and geese, and these differences could result from different expression profiles of FSHR, AMH, and CASPASE3 during early post-hatching development.
Collapse
Affiliation(s)
- Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Mou Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China.
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Lu Xia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| |
Collapse
|
28
|
Grosbois J, Devos M, Demeestere I. Implications of Nonphysiological Ovarian Primordial Follicle Activation for Fertility Preservation. Endocr Rev 2020; 41:5882019. [PMID: 32761180 DOI: 10.1210/endrev/bnaa020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
In recent years, ovarian tissue cryopreservation has rapidly developed as a successful method for preserving the fertility of girls and young women with cancer or benign conditions requiring gonadotoxic therapy, and is now becoming widely recognized as an effective alternative to oocyte and embryo freezing when not feasible. Primordial follicles are the most abundant population of follicles in the ovary, and their relatively quiescent metabolism makes them more resistant to cryoinjury. This dormant pool represents a key target for fertility preservation strategies as a resource for generating high-quality oocytes. However, development of mature, competent oocytes derived from primordial follicles is challenging, particularly in larger mammals. One of the main barriers is the substantial knowledge gap regarding the regulation of the balance between dormancy and activation of primordial follicles to initiate their growing phase. In addition, experimental and clinical factors also affect dormant follicle demise, while the mechanisms involved remain largely to be elucidated. Moreover, most of our basic knowledge of these processes comes from rodent studies and should be extrapolated to humans with caution, considering the differences between species in the reproductive field. Overcoming these obstacles is essential to improving both the quantity and the quality of mature oocytes available for further fertilization, and may have valuable biological and clinical applications, especially in fertility preservation procedures. This review provides an update on current knowledge of mammalian primordial follicle activation under both physiological and nonphysiological conditions, and discusses implications for fertility preservation and priorities for future research.
Collapse
Affiliation(s)
- Johanne Grosbois
- Research Laboratory in Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.,Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Melody Devos
- Research Laboratory in Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory in Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.,Obstetrics and Gynecology Department, Erasme Hospital, Brussels, Belgium
| |
Collapse
|
29
|
Dai X, Wang Y, Yang H, Gao T, Yu C, Cao F, Xia X, Wu J, Zhou X, Chen L. AMH has no role in predicting oocyte quality in women with advanced age undergoing IVF/ICSI cycles. Sci Rep 2020; 10:19750. [PMID: 33184364 PMCID: PMC7661530 DOI: 10.1038/s41598-020-76543-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
It has been widely acknowledged that anti-Müllerian hormone (AMH) is a golden marker of ovarian reserve. Declined ovarian reserve (DOR), based on experience from reproductive-aged women, refers to both the quantitative and qualitative reduction in oocytes. This view is challenged by a recent study clearly showing that the quality of oocytes is similar in young women undergoing IVF cycles irrespective of the level of AMH. However, it remains elusive whether AMH indicates oocyte quality in women with advanced age (WAA). The aim of this study was to investigate this issue. In the present study, we retrospectively analysed the data generated from a total of 492 IVF/ICSI cycles (from January 2017 to July 2020), and these IVF/ICSI cycles contributed 292 embryo transfer (ET) cycles (from June 2017 to September 2019, data of day 3 ET were included for analysis) in our reproductive centre. Based on the level of AMH, all patients (= > 37 years old) were divided into 2 groups: the AMH high (H) group and the AMH low (L) group. The parameters of in vitro embryo development and clinical outcomes were compared between the two groups. The results showed that women in the L group experienced severe DOR, as demonstrated by a higher rate of primary diagnosis of DOR, lower antral follicle count (AFC), higher level of basal follicle stimulating hormone (FSH) and cancelation cycles, lower level of E2 production on the day of surge, and fewer oocytes and MII oocytes retrieved. Compared with women in the H group, women in the L group showed slightly reduced top embryo formation rate but a similar normal fertilization rate and blastocyst formation rate. More importantly, we found that the rates of implantation, spontaneous miscarriage and livebirth were similar between the two groups, while the pregnancy rate was significantly reduced in the L group compared with the H group. Further analysis indicated that the higher pregnancy rate of women in the H group may be due to more top embryos transferred per cycle. Due to an extremely low implantation potential for transfer of non-top embryos from WAA (= > 37 years old) in our reproductive centre, we assumed that all the embryos that implanted may result from the transfer of top embryos. Based on this observation, we found that the ratio of embryos that successfully implanted or eventually led to a livebirth to top embryos transferred was similar between the H and the L groups. Furthermore, women with clinical pregnancy or livebirth in the H or L group did not show a higher level of serum AMH but were younger than women with non-pregnancy or non-livebirth. Taken together, this study showed that AMH had a limited role in predicting in vitro embryo developmental potential and had no role in predicting the in vivo embryo developmental potential, suggesting that in WAA, AMH should not be used as a marker of oocyte quality. This study supports the view that the accumulation of top embryos via multiple oocyte retrieval times is a good strategy for the treatment of WAA.
Collapse
Affiliation(s)
- Xiuliang Dai
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Yufeng Wang
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Haiyan Yang
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Tingting Gao
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Chunmei Yu
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Fang Cao
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Xiyang Xia
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Jun Wu
- Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, 210029, China.
| | - Xianju Zhou
- Department of Neurology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China. .,Department of Neurology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| | - Li Chen
- Department of Reproductive Medicine Center, The Affiliated Changzhou Maternal and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| |
Collapse
|
30
|
Luo Y, Sun L, Dong M, Zhang X, Huang L, Zhu X, Nong Y, Liu F. The best execution of the DuoStim strategy (double stimulation in the follicular and luteal phase of the same ovarian cycle) in patients who are poor ovarian responders. Reprod Biol Endocrinol 2020; 18:102. [PMID: 33059712 PMCID: PMC7566062 DOI: 10.1186/s12958-020-00655-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 09/24/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Patients found to be poor ovarian responders (POR) are a challenging patient population for any assisted reproduction technology. Despite attempts at various controlled ovarian stimulation schemes, reproductive outcomes in this patient population have not improved. In recent years, the DuoStim protocol (both follicular and luteal phase stimulation during the same menstrual cycle) has shown a potential for use in patients with POR. METHODS This retrospective study reviewed the medical records of 304 women who were diagnosed as POR and underwent the DuoStim protocol. We compared follicular phase stimulation (FPS) data and luteal phase stimulation (LPS) data of the same patients. We also compared the effects of different trigger drugs including urine human chorionic gonadotropin (uHCG; 10,000 IU), recombinant human chorionic gonadotropin (rHCG; 250 μg), and gonadotropin-releasing hormone agonist (GnRH-a; 0.2 mg) at the FPS and LPS stages. RESULTS POR undergoing the DuoStim protocol resulted in a significantly higher number of oocytes retrieved, normal fertilised oocytes, cleaved embryos, cryopreserved embryos, and good quality embryos at the LPS stage than at the FPS stage. Trigger drugs at the FPS stage did not affect the FPS stage data. Regardless of the stage, rHCG and GnRH-a yielded significantly more cryopreserved embryos and good quality embryos than uHCG. CONCLUSION The use of GnRH-a or rHCG as the trigger drug may be better than uHCG in both the FPS and LPS stages for POR undergoing the DuoStim protocol. This will increase the number of good quality embryos at the LPS stage. We found that the LPS stage results in more oocytes (and therefore more embryos) than the FPS stage.
Collapse
Affiliation(s)
- Yanqun Luo
- grid.412601.00000 0004 1760 3828The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Tianhe District, Guangzhou, 510630 Guangdong Province China
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| | - Li Sun
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| | - Mei Dong
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| | - Xiqian Zhang
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| | - Li Huang
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| | - Xiulan Zhu
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| | - Yingqi Nong
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| | - Fenghua Liu
- grid.459579.3Department of Reproductive Medical Center, Guangdong Women and Children Hospital, No. 521 Xingnan Road, Guangzhou, 511400 Guangdong Province China
| |
Collapse
|
31
|
Chen Y, Yang W, Shi X, Zhang C, Song G, Huang D. The Factors and Pathways Regulating the Activation of Mammalian Primordial Follicles in vivo. Front Cell Dev Biol 2020; 8:575706. [PMID: 33102482 PMCID: PMC7554314 DOI: 10.3389/fcell.2020.575706] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022] Open
Abstract
Mammalian ovaries consist of follicles as basic functional units. Each follicle comprised an innermost oocyte and several surrounding flattened granulosa cells. Unlike males, according to the initial size of the primordial follicle pool and the rate of its activation and depletion, a female's reproductive life has been determined early in life. Primordial follicles, once activated, will get into an irreversible process of development. Most follicles undergo atretic degeneration, and only a few of them could mature and ovulate. Although there are a lot of researches contributing to exploring the activation of primordial follicles, little is known about its underlying mechanisms. Thus, in this review, we collected the latest papers and summarized the signaling pathways as well as some factors involved in the activation of primordial follicles, hoping to lead to a more profound understanding of the cellular and molecular mechanisms of primordial follicle activation.
Collapse
Affiliation(s)
- Yao Chen
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weina Yang
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Shi
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlu Zhang
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ge Song
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, Guangzhou, China
| | - Donghui Huang
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Jewgenow K, Zahmel J. Preservation of female genetic resources in feline species. Theriogenology 2020; 156:124-129. [PMID: 32698038 DOI: 10.1016/j.theriogenology.2020.06.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 06/27/2020] [Indexed: 02/05/2023]
Abstract
The development of assisted reproduction techniques (ART) specifically for felids has been propagated for two main reasons: (i) most felids are threatened and faced with extinction in all or part of their native habitats (IUCN Red List of Threatened Species, www.catsg.org), and (ii) the domestic cat (Felis catus) can serve as a research model for the implementation of advanced assisted reproductive techniques (ART) to be applied in exotic cats. Domestic cat ovaries can be freshly obtained from veterinary clinics and are frequently used for research on preservation of genetic resources in feline species. The presented review will summarize recent advances and obstacles in biobanking of female genetic resources and discuss alternative approaches which are under investigation.
Collapse
Affiliation(s)
- Katarina Jewgenow
- Leibniz-Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 17, D-10315, Berlin, Germany.
| | - Jennifer Zahmel
- Leibniz-Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 17, D-10315, Berlin, Germany
| |
Collapse
|
33
|
New insights into anti-Müllerian hormone role in the hypothalamic-pituitary-gonadal axis and neuroendocrine development. Cell Mol Life Sci 2020; 78:1-16. [PMID: 32564094 PMCID: PMC7867527 DOI: 10.1007/s00018-020-03576-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/08/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022]
Abstract
Research into the physiological actions of anti-Müllerian hormone (AMH) has rapidly expanded from its classical role in male sexual differentiation to the regulation of ovarian function, routine clinical use in reproductive health and potential use as a biomarker in the diagnosis of polycystic ovary syndrome (PCOS). During the past 10 years, the notion that AMH could act exclusively at gonadal levels has undergone another paradigm shift as several exciting studies reported unforeseen AMH actions throughout the Hypothalamic–Pituitary–Gonadal (HPG) axis. In this review, we will focus on these findings reporting novel AMH actions across the HPG axis and we will discuss their potential impact and significance to better understand human reproductive disorders characterized by either developmental alterations of neuroendocrine circuits regulating fertility and/or alterations of their function in adult life. Finally, we will summarize recent preclinical studies suggesting that elevated levels of AMH may potentially be a contributing factor to the central pathophysiology of PCOS and other reproductive diseases.
Collapse
|
34
|
Vatanparast M, Moshrefi M, Yari N, Khalili MA, Macchiarelli G, Palmerini MG. Effective dosage of growth differentiation factor‐9β in folliculogenesis and angiogenesis in the sheep ovarian tissues grafted onto chick embryo chorioallantoic membrane. J Obstet Gynaecol Res 2020; 46:1384-1392. [DOI: 10.1111/jog.14266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/14/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Mahboubeh Vatanparast
- Molecular Medicine Research CenterRafsanjan University of Medical Sciences Rafsanjan Iran
- Research and Clinical Center for InfertilityYazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences Yazd Iran
| | - Mojgan Moshrefi
- Research and Clinical Center for InfertilityYazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences Yazd Iran
- Medical Nanotechnology and Tissue Engineering Research CenterYazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences Yazd Iran
| | - Nahid Yari
- Research and Clinical Center for InfertilityYazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences Yazd Iran
| | - Mohammad Ali Khalili
- Research and Clinical Center for InfertilityYazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences Yazd Iran
| | - Guido Macchiarelli
- Department of LifeHealth and Environmental Sciences, University of L'Aquila L'Aquila Italy
| | - Maria Grazia Palmerini
- Department of LifeHealth and Environmental Sciences, University of L'Aquila L'Aquila Italy
| |
Collapse
|
35
|
Pargianas M, Salta S, Apostolopoulou K, Lazaros L, Kyrgiou M, Tinelli A, Malvasi A, Kalogiannidis I, Georgiou I, Kosmas IP. Pathways Involved in Premature Ovarian Failure: A Systematic Review of Experimental Studies. Curr Pharm Des 2020; 26:2087-2095. [PMID: 32175834 DOI: 10.2174/1381612826666200316160145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/10/2020] [Indexed: 11/22/2022]
Abstract
Premature ovarian failure (POF), which may be undetectable for a long time, is associated with impaired fertility. The mechanisms involved in the pathogenesis of POF as well as the concomitant treatments are still unclear. Although many data exist, mainly produced by the study of transgenic animals under various experimental conditions, they remain fragmented. A systematic review of the pathways involved in premature ovarian failure was conducted. Data extraction was performed from experimental studies until 2019. The molecular processes and their correlation with the follicular developmental stage have been described. Furthermore, the effects in other cells, such as oocytes, granulosa and theca cells have been reported. An overall estimation was conducted.
Collapse
Affiliation(s)
- Michail Pargianas
- Department of Obstetrics and Gynecology, Ioannina State General Hospital G. Chatzikosta, Ioannina, Greece
| | - Styliani Salta
- University Hospitals of Leicester, Haemophilia Centre, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Katerina Apostolopoulou
- Department of Biological Applications and Technologies, Ioannina University, Ioannina, Greece
| | - Leandros Lazaros
- Genetics and IVF Unit, Department of Obstetrics and Gynecology, Medical School, Ioannina University, Ioannina, Greece
| | - Maria Kyrgiou
- West London Gynecological Cancer Center, Queen Charlotte's and Chelsea-Hammersmith Hospital, Imperial Healthcare NHS Trust, London, United Kingdom
| | - Andrea Tinelli
- Moscow Institute of Physics and Technology (State University), Moscow Region, Russian Federation.,Department of Obstetrics and Gynecology, Division of Experimental Endoscopic Surgery, Imaging, Technology and Minimally Invasive Therapy, Vito Fazzi Hospital, Lecce, Italy
| | - Antonio Malvasi
- Moscow Institute of Physics and Technology (State University), Moscow Region, Russian Federation.,Department of Gynecology and Obstetrics, Santa Maria Hospital, Bari, Italy
| | - Ioannis Kalogiannidis
- Third Department of Obstetrics and Gynaecology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Georgiou
- Genetics and IVF Unit, Department of Obstetrics and Gynecology, Medical School, Ioannina University, Ioannina, Greece
| | - Ioannis P Kosmas
- Department of Obstetrics and Gynecology, Ioannina State General Hospital G. Chatzikosta, Ioannina, Greece.,Moscow Institute of Physics and Technology (State University), Moscow Region, Russian Federation
| |
Collapse
|
36
|
Wu W, Wang X, Li Y, Zhang Y. Analysis of the women with the AMH concentrations below the limit of reference range but with the ideal number of retrieved oocytes. Arch Gynecol Obstet 2020; 301:1089-1094. [PMID: 32179967 DOI: 10.1007/s00404-020-05491-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/05/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE Serum anti-Mullerian hormone (AMH) shows a strong positive correlation to the number of oocytes retrieved but the patients undergoing assisted reproductive technology (ART) with lower AMH concentrations also could retrieve an ideal number of oocytes sometimes. The aim of the current study was to assess the performance of this population. METHODS This retrospective study included a total of 44 in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) treatment cycles as test group, in which their AMH levels were below the limit of reference range but more than 7 eggs were retrieved, and 103 control cases enrolled from May 2016 to December 2018 after matched with the same range of age from 24 to 38 years old and the same number range of retrieved oocytes from 7 to 18 and chosen randomly according to clinical ovarian stimulation protocols by the ratio of 3:7. Several parameters, which include age, basal endocrine status, number of oocytes, the maturation rate of eggs, 2PN fertilization rate, 3PN rate, total fertilization rate, cleavage rate, 8-cell embryo rate of D3 and the utilization rate of embryo, were compared and evaluated between two groups by Wilcoxon rank-sum test and t test for two independent samples. RESULTS Although the same age range from 24 to 38 years and the same retrieval eggs from 7 to 18, compared with the control group, the test group showed less number of oocytes collected (9.455 vs 10.767, p = 0.016), younger ages (28.36 vs 32.59, p = 0.000), and higher FSH of basal endocrine status (9.783 vs 7.338, p = 0.021). Between the two groups, there were no significant differences in parameters such as the mature eggs (7.05 vs 7.92, p = 0.079), the mature oocyte rate (74.189% vs 73.916%, p = 0.924), the number of 2PNs (5.36 vs 5.91, p = 0.236), 2PN rate (73.678% vs 75.125%, p = 0.769), the number of 3PNs (0.39 vs 0.50, p = 0.773), 3PN rate (5.104% vs 5.592%, p = 0.697), number of total fertilizations (6.77 vs 7.35, p = 0.241), total fertilization rate (96.461% vs 93.166%, p = 0.332), cleavage rate (82.003% vs 81.382%, p = 0.673), the number of 8-cells on D3 (1.59 vs 1.91, p = 0.227), the rate of 8-cell on D3 (36.259% vs 41.084%, p = 0.551) and the utilization rate of embryo (62.853% vs 61.824%, p = 0.806). CONCLUSIONS These findings indicate that there are no significant differences in the maturation of oocytes and the embryo quality for the women with low AMH level and ideal retrieval eggs but they should undergo IVF treatment and achieve successful pregnancy as soon as possible due to the higher basal FSH, the relatively fewer number of retrieval eggs and the possibility of coming diminished ovarian reserve (DOR) or poor respond (PR) despite their younger ages.
Collapse
Affiliation(s)
- Wenbin Wu
- Reproductive Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Xingling Wang
- Reproductive Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yushan Li
- Reproductive Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuchao Zhang
- Reproductive Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
37
|
Fu YX, Wang FM, Ou-Yang XE, Yang HM, Hu T, Wang YF, Wang YF, Wang H, Hu R. Anti-Müllerian Hormone Regulates Stem Cell Factor via cAMP/PKA Signaling Pathway in Human Granulosa Cells by Inhibiting the Phosphorylation of CREB. Reprod Sci 2020; 27:325-333. [PMID: 32046389 DOI: 10.1007/s43032-019-00033-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/03/2019] [Indexed: 10/25/2022]
Abstract
Anti-Müllerian hormone (AMH) downregulates the level of stem cell factor (SCF) via the cAMP/PKA signaling pathway in human granulosa cells (GCs). Little information is available on the molecular mechanism underlying the interaction. This study is aimed at determining whether AMH regulates expression of SCF via the cAMP-PKA-CREB signaling pathway in human GCs. In the present study, we verified the binding of cAMP-response element-binding protein (CREB) to promoter of SCF in human GCs. Furthermore, the effect of CREB was tested on the SCF promoter, and the site of CREB binding to SCF promoter was identified using truncations as well as assays of SCF-promoted mutation and CREB mutation. To investigate the correlation among AMH, SCF promoter, and CREB, pGL-Basic-SCF+CREB was transfected into overexpressed AMH GCs (AMH-high GCs), low expressed AMH GCs (AMH-low GCs), and normal GCs (GCs), respectively. Finally, immunofluorescence, double immunostaining, and Western blot were carried out in AMH-high and AMH-low GCs to confirm the AMH-mediated regulation of SCF expression by inhibiting the phosphorylation of CREB (pCREB) in GCs. Results indicated CREB interacted with SCF promoter and significantly enhanced the transcription level of SCF. The CREB binding site was localized at 318-321 bp of SCF gene promote. AMH inhibits the expression of SCF by phosphorylation of CREB via the PKA signaling pathway in GCs. These findings provide an in-depth understanding of the molecular mechanism underlying AMH suppressing the follicle growth, which would aid in the development of a novel therapy.
Collapse
Affiliation(s)
- Yun-Xing Fu
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Fei-Miao Wang
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | | | - Hui-Min Yang
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ting Hu
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ya-Fei Wang
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yan-Fei Wang
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Hui Wang
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Rong Hu
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
38
|
KHARCHE SURESHDINKAR, JENA DAYANIDHI, GANGWAR CHETNA, KHATTI AMIT, BALAMURUGAN B. Role of certain growth factors and hormones in folliculogenesis. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2020. [DOI: 10.56093/ijans.v89i12.96617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Folliculogenesis is an inextricable process associated with female fertility and infertility cases. This process involves many events at cellular and molecular level in a highly orchestrated fashion which culminates with ovulation. Various factors like hormonal factors, growth factors, role of ovarian micro environment, diseases of reproductive tract etc. influence the process of folliculogenesis in systematic manner. The function and mechano-biology of these growth factors and hormones have been studied by many researchers. This review discusses about those hormonal and growth factors which are involved in folliculogenesis process.
Collapse
|
39
|
Guo R, Pankhurst MW. Accelerated ovarian reserve depletion in female anti-Müllerian hormone knockout mice has no effect on lifetime fertility†. Biol Reprod 2019; 102:915-922. [DOI: 10.1093/biolre/ioz227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/12/2019] [Accepted: 12/12/2019] [Indexed: 11/14/2022] Open
Abstract
Abstract
Anti-Müllerian hormone (AMH) inhibits the activation of primordial follicles in the ovary. This causes an increased rate of ovarian reserve depletion in Amh−/− mice. The depletion of the ovarian reserve is responsible for the onset of menopause but age-related infertility occurs in advance of ovarian reserve depletion. To determine whether accelerated loss of primordial follicles leads to earlier onset infertility, Amh−/− and Amh+/+ females were paired with Amh+/+ stud males and birth rates were recorded across the females’ reproductive lifespan. The number of primordial follicles remaining in the ovaries of Amh−/− and Amh+/+ females were quantified in two cohorts at 11–12 and 12–13 months of age. As expected, the ovarian reserve in the Amh−/− females became depleted approximately 1 month earlier than Amh+/+ females. However, no difference was observed in the cumulative number of births over the lifespan, nor were there any differences in mean littersize at any age. It is possible that the reproductive lifespan of mice is too short for sufficient divergence of primordial follicles numbers to cause differences in Amh−/− and Amh+/+ female fertility. An alternative explanation contradicts current thinking; the function of AMH may be unrelated to the longevity of the reproductive lifespan in female mice.
Collapse
Affiliation(s)
- Ruikang Guo
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Michael W Pankhurst
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
40
|
Thanatsis N, Kaponis A, Koika V, Georgopoulos NA, Decavalas GO. Reduced Foxo3a, FoxL2, and p27 mRNA expression in human ovarian tissue in premature ovarian insufficiency. Hormones (Athens) 2019; 18:409-415. [PMID: 31637660 DOI: 10.1007/s42000-019-00134-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/10/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE Previous studies have suggested that deletion of Foxo3a, FoxL2, PTEN, p27, and AMH leads to early exhaustion of the primordial follicle pool and premature ovarian insufficiency (POI) in transgenic mice. Our aim was to assess for the first time, to our knowledge, messenger RNA (mRNA) expression of these genes and AMHR2 in human ovarian tissue from women with POI. We hypothesized that these genes would be underexpressed in POI women compared with healthy controls. METHODS mRNA levels were evaluated by quantitative reverse transcription-polymerase chain reaction and real-time polymerase chain reaction in cortical ovarian tissue obtained by laparoscopy from Caucasian Greek women with POI (n = 5) and healthy women with normal menstruation (n = 6). Morphological analysis of the ovarian biopsies was also performed to assess the presence of primordial or other types of growing follicles. RESULTS Ovarian tissue from POI patients showed lower Foxo3a, FoxL2, and p27 mRNA expression compared with controls (p = 0.017, p = 0.017, and p = 0.030, respectively). mRNA expression of AMH, PTEN, and AMHR2 was reduced in ovarian biopsies from POI patients as well. However, these differences were not statistically significant (p = 0.143, p = 0.247, and p = 0.662, respectively). Morphological analysis showed complete lack of follicular structures in all POI biopsies. CONCLUSIONS Our findings suggest a possible role of Foxo3a, FoxL2, and p27 in the pathogenesis of human POI, which may prove to be of great diagnostic-therapeutic value. Further larger studies are needed to identify a similar pattern for AMH, PTEN, and AMHR2 and to investigate gene expression at a protein level.
Collapse
Affiliation(s)
- Nikolaos Thanatsis
- Department of Obstetrics and Gynecology, Patras University School of Medicine, 26504, Patras, Greece.
| | - Apostolos Kaponis
- Department of Obstetrics and Gynecology, Patras University School of Medicine, 26504, Patras, Greece
| | - Vasiliki Koika
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, Patras University School of Medicine, 26504, Patras, Greece
| | - Neoklis A Georgopoulos
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, Patras University School of Medicine, 26504, Patras, Greece
| | - George O Decavalas
- Department of Obstetrics and Gynecology, Patras University School of Medicine, 26504, Patras, Greece
| |
Collapse
|
41
|
Li Y, Gao D, Xu T, Adur MK, Zhang L, Luo L, Zhu T, Tong X, Zhang D, Wang Y, Ning W, Qi X, Cao Z, Zhang Y. Anti-Müllerian hormone inhibits luteinizing hormone-induced androstenedione synthesis in porcine theca cells. Theriogenology 2019; 142:421-432. [PMID: 31711705 DOI: 10.1016/j.theriogenology.2019.10.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 11/28/2022]
Abstract
AMH (Anti-Müllerian Hormone) is involved in the regulation of follicle growth initiation and inhibits FSH-induced aromatase expression and estrogen production in granulosa cells. However, the function of AMH in steroidogenesis by theca cells remains unclear. The aim of this study is to investigate the role of AMH as a regulator of the basal and stimulated steroid production by pig granulosa cells (pGCs) and theca cells (pTCs). PGCs and pTCs were incubated with hormones AMH, LH (luteinizing hormone), FSH (follicle stimulating hormone), individually or in combination. The expression of CYP19A1, HSD3B1, CYP11A1, LHCGR, and CYP17A1 mRNA were evaluated by quantitative reverse transcriptase PCR. In pGCs, 10 ng/mL AMH significantly decreased the FSH-stimulated effect on FSHR and CYP19A1 expression and estradiol production. In pTCs, LH treatment significantly increased the expression of HSD3B1, CYP11A1, LHCGR, and androstenedione or progesterone production (P < 0.05). Additionally, 10 ng/mL AMH also significantly decreased the LH-stimulated effects on the expression of HSD3B1, CYP11A1, CYP17A1, LHCGR and androstenedione production. Transfection with siAMHR2-I abolished the suppressive effects of AMH on LH-induced HSD3B1 expression and androstenedione production. Taken together, these results demonstrate that AMH is involved in FSH induced estradiol production in pGCs and LH induced androstenedione production in pTCs by regulating the steroidogenesis pathway.
Collapse
Affiliation(s)
- Yunsheng Li
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Di Gao
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Tengteng Xu
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Malavika K Adur
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Ling Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Lei Luo
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Ting Zhu
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Xu Tong
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Dandan Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Yiqing Wang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Wei Ning
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Xin Qi
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Zubing Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| | - Yunhai Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
42
|
Lemcke RA, Stephens CS, Hildebrandt KA, Johnson PA. Anti-Müllerian hormone type II receptor in avian follicle development. Biol Reprod 2019; 99:1227-1234. [PMID: 29931109 DOI: 10.1093/biolre/ioy140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 06/18/2018] [Indexed: 11/13/2022] Open
Abstract
Anti-Müllerian hormone (AMH) helps maintain the ovarian reserve by regulating primordial follicle activation and follicular selection in mammals, although its role within the avian ovary is unknown. In mammals, AMH is primarily produced in granulosa cells of preantral and early antral follicles. Similarly, in the hen, the granulosa cells of smaller follicles are the predominant source of AMH. The importance of AMH in mammalian ovarian dynamics suggests the protein and its specific Type II receptor, AMHRII, may have conserved functions in the hen. AMHRII mRNA expression is highest (P < 0.01) in small follicles of the hen and decreases as follicle size increases. Similarly, expression of AMHRII and AMH is highest in granulosa cells from small follicles as compared to larger follicles. Dissection of 3-5 mm follicles into ooplasm and granulosa components shows that AMHRII mRNA levels are greater in ooplasm than granulosa cells. Furthermore, immunohistochemistry also revealed AMHRII staining in the oocyte and granulosa cells. AMH expression in mammals is elevated during periods of reproductive dormancy, possibly protecting the ovarian reserve. AMHRII and AMH mRNA were significantly higher (P < 0.05) in nonlaying ovaries of broiler hens. In molting layer hens, AMHRII mRNA was significantly greater (P < 0.05) compared to nonmolting hen ovaries. These results suggest that AMH may have a direct effect on the oocyte and, thereby, contribute to bidirectional communication between oocyte and granulosa cells. Enhanced expression of AMHRII and AMH during reproductive quiescence supports a potential role of AMH in protecting the ovarian reserve in hens.
Collapse
Affiliation(s)
- R A Lemcke
- Department of Animal Science, Cornell University, Ithaca, New York, USA
| | - C S Stephens
- Department of Animal Science, Cornell University, Ithaca, New York, USA
| | - K A Hildebrandt
- Department of Animal Science, Cornell University, Ithaca, New York, USA
| | - P A Johnson
- Department of Animal Science, Cornell University, Ithaca, New York, USA
| |
Collapse
|
43
|
Ovarian Follicle Depletion Induced by Chemotherapy and the Investigational Stages of Potential Fertility-Protective Treatments-A Review. Int J Mol Sci 2019; 20:ijms20194720. [PMID: 31548505 PMCID: PMC6801789 DOI: 10.3390/ijms20194720] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 01/10/2023] Open
Abstract
Ovarian follicle pool depletion, infertility, and premature menopause are all known sequelae of cancer treatment that negatively impact the quality of life of young cancer survivors. The mechanisms involved in this undesired iatrogenic ovarian damage have been intensively studied, but many of them remain unclear. Several chemotherapeutic drugs have been shown to induce direct and indirect DNA-damage and/or cellular stress, which are often followed by apoptosis and/or autophagy. Damage to the ovarian micro-vessel network induced by chemotherapeutic agents also seems to contribute to ovarian dysfunction. Another proposed mechanism behind ovarian follicle pool depletion is the overactivation of primordial follicles from the quiescent pool; however, current experimental data are inconsistent regarding these effects. There is great interest in characterizing the mechanisms involved in ovarian damage because this might lead to the identification of potentially protective substances as possible future therapeutics. Research in this field is still at an experimental stage, and further investigations are needed to develop effective and individualized treatments for clinical application. This review provides an overview of the current knowledge and the proposed hypothesis behind chemotherapy-induced ovarian damage, as well as current knowledge on possible co-treatments that might protect the ovary and the follicles from such damages.
Collapse
|
44
|
Kano M, Hsu JY, Saatcioglu HD, Nagykery N, Zhang L, Morris Sabatini ME, Donahoe PK, Pépin D. Neoadjuvant Treatment With Müllerian-Inhibiting Substance Synchronizes Follicles and Enhances Superovulation Yield. J Endocr Soc 2019; 3:2123-2134. [PMID: 31687639 PMCID: PMC6821214 DOI: 10.1210/js.2019-00190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/16/2019] [Indexed: 12/25/2022] Open
Abstract
Müllerian-inhibiting substance (MIS), also known as anti-Müllerian hormone, is thought to be a negative regulator of primordial follicle activation. We have previously reported that treatment with exogenous MIS can induce complete ovarian suppression within 5 weeks of treatment in mice. To investigate the kinetics of the return of folliculogenesis following the reversal of suppression, we treated animals with recombinant human MIS (rhMIS) protein for 40 days in adult female Nu/Nu mice and monitored the recovery of each follicle type over time. Following cessation of MIS therapy, secondary, and antral follicles returned within 30 days, along with the normalization of reproductive hormones, including LH, FSH, MIS, and Inhibin B. Furthermore, 30 days following MIS pretreatment, the number of antral follicles were significantly higher than controls, and superovulation with timed pregnant mare serum gonadotropin and human chorionic gonadotropin stimulation at this time point resulted in an approximately threefold increased yield of eggs. Use of the combined rhMIS-gonadotropin superovulation regimen in a diminished ovarian reserve (DOR) mouse model, created by 4-vinylcyclohexene dioxide treatment, also resulted in a twofold improvement in the yield of eggs. In conclusion, treatment with rhMIS can induce a reversible ovarian suppression, following which a rapid and synchronized large initial wave of growing follicles can be harnessed to enhance the response to superovulation. Therapies modulating MIS signaling may therefore augment the response to current ovarian stimulation protocols and could be particularly useful to women with DOR or poor responders to controlled ovarian hyperstimulation during in vitro fertilization.
Collapse
Affiliation(s)
- Motohiro Kano
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Y Hsu
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
| | - Hatice D Saatcioglu
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Nicholas Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - LiHua Zhang
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Mary E Morris Sabatini
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Sonigo C, Beau I, Binart N, Grynberg M. Anti-Müllerian Hormone in Fertility Preservation: Clinical and Therapeutic Applications. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119854755. [PMID: 31258345 PMCID: PMC6585130 DOI: 10.1177/1179558119854755] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022]
Abstract
Anti-Müllerian hormone (AMH) is a member of the transforming growth factor
(TGF)-beta family and a key regulator of sexual differentiation and
folliculogenesis. While the serum AMH level has been used in reproductive
medicine as a biomarker of quantitative ovarian reserve for more than 20 years,
new potential therapeutic applications of recombinant AMH are emerging, notably
in the field of oncofertility. Indeed, it is well known that chemotherapy, used
to treat cancer, induces ovarian follicular depletion and subsequent
infertility. Animal models have been used widely to understand the effects of
different cytotoxic agents on ovarian function, and several hypotheses regarding
chemotherapy gonadotoxicity have been proposed, that is, it might have a direct
detrimental effect on the primordial follicles constituting the ovarian reserve
and/or on the pool of growing follicles secreting AMH. Recently, a new mechanism
of chemotherapy-induced follicular depletion, called the “burn-out effect,” has
been proposed. According to this theory, chemotherapeutic agents may lead to a
massive growth of dormant follicles which are then destroyed. As AMH is one of
the factors regulating the recruitment of primordial follicles from the ovarian
reserve, recombinant AMH administration concomitant with chemotherapy might
limit follicular depletion, therefore representing a promising option for
preserving fertility in women suffering from cancer. This review reports on the
potential usefulness of AMH measurement as well as AMH’s role as a therapeutic
agent in the field of female fertility preservation.
Collapse
Affiliation(s)
- Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Antoine Béclère, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Clamart, France.,Inserm U1185, Université Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Isabelle Beau
- Inserm U1185, Université Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Nadine Binart
- Inserm U1185, Université Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Antoine Béclère, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Clamart, France.,Université Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France.,Inserm U1133, Université Paris Diderot, Paris, France
| |
Collapse
|
46
|
Oh SR, Choe SY, Cho YJ. Clinical application of serum anti-Müllerian hormone in women. Clin Exp Reprod Med 2019; 46:50-59. [PMID: 31181872 PMCID: PMC6572668 DOI: 10.5653/cerm.2019.46.2.50] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/22/2019] [Indexed: 12/18/2022] Open
Abstract
Anti-Müllerian hormone (AMH), a peptide growth factor of the transforming growth factor-β family, is a reliable marker of ovarian reserve. Regarding assisted reproductive technology, AMH has been efficiently used as a marker to predict ovarian response to stimulation. The clinical use of AMH has recently been extended and emphasized. The uses of AMH as a predictive marker of menopause onset, diagnostic tool for polycystic ovary syndrome, and assessment of ovarian function before and after gynecologic surgeries or gonadotoxic agents such as chemotherapy have been investigated. Serum AMH levels can also be affected by environmental and genetic factors; thus, the effects of factors that may alter AMH test results should be considered. This review summarizes the findings of recent studies focusing on the clinical application of AMH and factors that influence the AMH level and opinions on the use of the AMH level to assess the probability of conception before reproductive life planning as a “fertility test.”
Collapse
Affiliation(s)
- So Ra Oh
- Department of Obstetrics and Gynecology, Dong-A University Medical Center, Dong-A University College of Medicine, Busan, Korea
| | - Sun Yi Choe
- Department of Obstetrics and Gynecology, Dong-A University Medical Center, Dong-A University College of Medicine, Busan, Korea
| | - Yeon Jean Cho
- Department of Obstetrics and Gynecology, Dong-A University Medical Center, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
47
|
Cozzolino M, Cruz M, Patel A, Patel J, Pacheco A, Garcia-Velasco JA. Serum and follicular fluid Stem Cell Factor assay in IVF poor responder and normal responder patients: a predictive biomarker of oocyte retrieval. Arch Gynecol Obstet 2019; 300:447-454. [PMID: 31062149 DOI: 10.1007/s00404-019-05172-2] [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: 10/04/2018] [Accepted: 04/24/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The aim of the study is to investigate serum stem cell factor (SCF) concentrations as potential biomarker for oocyte retrieval efficiency in IVF patients with poor prognosis. METHODS A pilot case-control study was performed on 30 poor and 30 normal responders that were stimulated with antagonist protocol. SCF concentrations were evaluated in samples of serum and follicular fluid obtained by all patients on the day of oocyte retrieval. At the time of oocyte retrieval, follicular fluid from at least two follicles ≥ 14 mm and two follicles < 14 mm was collected for SCF determination. RESULTS We did not find any statistical difference when comparing serum and follicular fluid levels of SCF in both poor- and normal-responder patients, the same results were achieved when poor-responder patients were stratified according to the number of MII oocytes retrieved. Moreover, levels of SCF (OR 1.000, 0.994-1.006) or in follicular fluid from ovarian follicles ≥ 14 mm (OR 0.995, CI 0.989-1.001) or from ovarian follicles < 14 mm (OR 1.003, CI 0.999-1.0069), were not significantly associated with the chances of ongoing pregnancies in poor-responder patients. CONCLUSION SCF was unable to predict oocyte retrieval efficiency or the chances of reaching embryo transfer.
Collapse
Affiliation(s)
- Mauro Cozzolino
- IVIRMA Madrid, Avenida del Talgo 68-70, 28023, Madrid, Spain. .,Rey Juan Carlos University, Madrid, Spain.
| | - Maria Cruz
- IVIRMA Madrid, Avenida del Talgo 68-70, 28023, Madrid, Spain
| | - Azadeh Patel
- Department of Reproductive Medicine, Nova IVI Fertility, Ahmedabad, India
| | - Jayesh Patel
- Department of Reproductive Medicine, Nova IVI Fertility, Ahmedabad, India
| | - Alberto Pacheco
- IVIRMA Madrid, Avenida del Talgo 68-70, 28023, Madrid, Spain
| | - Juan Antonio Garcia-Velasco
- IVIRMA Madrid, Avenida del Talgo 68-70, 28023, Madrid, Spain.,Rey Juan Carlos University, Madrid, Spain.,Department of Reproductive Medicine, Nova IVI Fertility, Ahmedabad, India
| |
Collapse
|
48
|
Gowkielewicz M, Lipka A, Piotrowska A, Szadurska-Noga M, Nowakowski JJ, Dzięgiel P, Majewski MK, Jozwik M, Majewska M. Anti-Müllerian Hormone Expression in Endometrial Cancer Tissue. Int J Mol Sci 2019; 20:ijms20061325. [PMID: 30884769 PMCID: PMC6471522 DOI: 10.3390/ijms20061325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/29/2022] Open
Abstract
Anti-Müllerian hormone (AMH) is a commonly known factor secreted by Sertoli cells, responsible for regression of the Müllerian ducts in male fetuses. AMH has also other functions in humans. In vivo and in vitro studies have shown that AMH inhibits cell cycle and induces apoptosis in cancers with AMH receptors. The aim of the study was to assess whether the tissue of pre-cancerous states of endometrium (PCS) and various histopathologic types of endometrial cancer (EC) exhibit the presence of AMH. We aimed to investigate whether the potential presence of the protein concerns menopausal women or those regularly menstruating, and whether is related to cancers with a good or a bad prognosis, as well as what other factors may influence AMH expression. The undertaken analysis was carried out on tissues retrieved from 232 women who underwent surgical treatment for PCS and EC. Tissues were prepared for immunohistochemical assessment with the use of a tissue microarrays method. AMH expression was confirmed in 23 patients with well differentiated endometrioid adenocarcinoma (G1), moderately differentiated endometrioid adenocarcinoma (G2), clear cell carcinoma (CCA) and nonatypical hyperplasia. AMH was not found in EC tissues in regularly menstruating women. An appropriately long mean period of breastfeeding in line with a prolonged period of hormonal activity had a positive effect on AMH expression. Our results may suggest that AMH is a factor which protects the organism against cancer, and should be further investigated as a potential prognosis marker and a therapeutic agent.
Collapse
Affiliation(s)
- Marek Gowkielewicz
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-045 Olsztyn, Poland.
| | - Aleksandra Lipka
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-045 Olsztyn, Poland.
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland.
| | - Marta Szadurska-Noga
- Department of Pathomorphology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland.
| | - Jacek J Nowakowski
- Department of Ecology & Environmental Protection, University of Warmia and Mazury in Olsztyn, 10⁻727 Olsztyn, Poland.
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland.
- Department of Physiotherapy, Wroclaw University School of Physical Education, 51-612 Wroclaw, Poland.
| | - Mariusz Krzysztof Majewski
- Department of Human Physiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland.
| | - Marcin Jozwik
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-045 Olsztyn, Poland.
| | - Marta Majewska
- Department of Human Physiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland.
| |
Collapse
|
49
|
Pépin D, Sabatini ME, Donahoe PK. Müllerian inhibiting substance/anti-Müllerian hormone as a fertility preservation agent. Curr Opin Endocrinol Diabetes Obes 2018; 25:399-405. [PMID: 30320617 DOI: 10.1097/med.0000000000000442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE OF REVIEW The nascent field of oncofertility is quickly gaining traction as novel experimental treatments are being developed, driving a renewed interest in Müllerian inhibiting substance (MIS) as an ovarian fertoprotectant. RECENT FINDINGS MIS is unique in its mechanisms of ovarian protection by virtue of acting directly on granulosa cells of primordial follicles and for being a benign reproductive hormone, with few side effects. We will explore in this review how it may be utilized to protect the ovary from chemotherapy, or to enhance ovarian tissue cryopreservation therapy. We will also examine potential mechanisms of action of MIS across multiple cell types, as well as current limitations in our understanding of the pharmacology of recombinant MIS. SUMMARY The usefulness of MIS as a fertoprotectant may be dependent on the mechanisms of gonadotoxicity of each chemotherapeutic. Further investigation is needed to determine how to best deliver and combine MIS treatment to existing fertility management strategies.
Collapse
Affiliation(s)
- David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston
- Department of Surgery, Harvard Medical School, Cambridge
| | - Mary E Sabatini
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston
- Department of Surgery, Harvard Medical School, Cambridge
| |
Collapse
|
50
|
Peluso JJ, Liu X, Uliasz T, Pru CA, Kelp NC, Pru JK. PGRMC1/2 promotes luteal vascularization and maintains the primordial follicles of mice. Reproduction 2018; 156:365-373. [PMID: 30306772 PMCID: PMC6348134 DOI: 10.1530/rep-18-0155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/20/2018] [Accepted: 07/27/2018] [Indexed: 01/05/2023]
Abstract
To determine whether conditional depletion of progesterone receptor membrane component (PGRMC) 1 and PGRMC2 affected ovarian follicle development, follicle distribution was assessed in ovaries of young (≈3-month-old) and middle-aged (≈6-month-old) control (Pgrmc1/2fl/fl) and double conditional PGRMC1/2-knockout (Pgrmc1/2d/d) mice. This study revealed that the distribution of primary, preantral and antral follicles was not altered in Pgrmc1/2d/d mice, regardless of the age. Although the number of primordial follicles was similar at ≈3 months of age, their numbers were reduced by ≈80% in 6-month-old Pgrmc1/2d/d mice compared to age-matched Pgrmc1/2fl/fl mice. The Pgrmc1/2d/d mice were generated using Pgr-cre mice, so ablation of Pgrmc1 and Pgrmc2 in the ovary was restricted to peri-ovulatory follicles and subsequent corpora lutea (CL). In addition, the vascularization of CL was attenuated in Pgrmc1/2d/d mice, although mRNA levels of vascular endothelial growth factor A (Vegfa) were elevated. Moreover, depletion of Pgrmc1 and Pgrmc2 altered the gene expression profile in the non-luteal component of the ovary such that Vegfa expression, a stimulator of primordial follicle growth, was elevated; Kit Ligand expression, another stimulator of primordial follicle growth, was suppressed and anti-Mullerian hormone, an inhibitor of primordial follicle growth, was enhanced compared to Pgrmc1/2fl/fl mice. These data reveal that luteal cell depletion of Pgrmc1 and 2 alters the expression of growth factors within the non-luteal component of the ovary, which could account for the premature demise of the adult population of primordial follicles. In summary, the survival of adult primordial follicles is dependent in part on progesterone receptor membrane component 1 and 2.
Collapse
Affiliation(s)
- John J. Peluso
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030
- Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, CT 06030
| | - Xiufang Liu
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030
| | - Tracy Uliasz
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030
| | - Cindy A. Pru
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| | - Nicole C. Kelp
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| | - James K. Pru
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| |
Collapse
|