1
|
Racine C, Fraissinet F, Tolu S, Pereira T, Gil S, Badel A, Bailbé D, Fève B, Movassat J, Cate R, di Clemente N. A blocking antibody against anti-Müllerian hormone restores ovulation and normal androgen levels in a spontaneous rat model of polycystic ovary syndrome. EBioMedicine 2025; 115:105716. [PMID: 40252252 PMCID: PMC12032919 DOI: 10.1016/j.ebiom.2025.105716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Polycystic Ovary Syndrome (PCOS), the leading cause of infertility worldwide, is characterised by oligo-anovulation, hyperandrogenism, polycystic ovarian morphology and high Anti-Müllerian hormone (AMH) levels, associated with severe metabolic disturbances. However, the role of AMH in the physiopathology of this syndrome remains poorly understood and strategies to block its effects have never been investigated in animal models of PCOS. METHODS We used Western-blotting, ELISA and gene reporter approaches to evaluate the blocking efficacy, interspecificity and mechanism of action of an antibody against human AMH, Mab22A2. Then, we investigated the ability of a rat version of Mab22A2, rMab22A2, to alleviate reproductive dysfunction in Goto-Kakizaki (GK) rats, which spontaneously exhibit all the features of women with PCOS. FINDINGS We showed that Mab22A2 was interspecific, did not prevent AMH from binding to its receptor and was able to block the effects of AMH in gonadal cell lines. In addition, treatment of anovulatory GK rats with rMab22A2 reduced their bioavailable serum AMH levels and normalised their androgen concentrations. Finally, this treatment also induced ovulation in 84% of the rats and resulted in 66% of pregnancies. INTERPRETATION Our results show that AMH is a major driver of reproductive and hormonal dysfunction in PCOS and provide proof of concept that a blocking antibody against AMH can reverse the major reproductive dysfunction observed in PCOS, opening up promising avenues for the treatment of patients with PCOS. FUNDING Inserm, Sorbonne University, Inserm Transfert, the French Endocrine Society and the Medical Research Foundation (grant agreement n°EQU201903007868).
Collapse
Affiliation(s)
- Chrystèle Racine
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France; Université Paris Cité, Paris 75013, France
| | - François Fraissinet
- Department of General Biochemistry, Rouen University Hospital, Rouen 76000, France
| | - Stefania Tolu
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Tony Pereira
- Department of General Biochemistry, Rouen University Hospital, Rouen 76000, France
| | | | - Anne Badel
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Danielle Bailbé
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Bruno Fève
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France; Department of Endocrinology, Hôpital Saint-Antoine, APHP, Paris 75012, France
| | | | - Richard Cate
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Nathalie di Clemente
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France.
| |
Collapse
|
2
|
Detti L, Mari MC, Diamond MP, Saed GM. Anti-Mullerian hormone (AMH) protects ovarian follicle loss by downregulating granulosa cell function in in vitro and in vivo models. J Assist Reprod Genet 2025:10.1007/s10815-025-03473-x. [PMID: 40198512 DOI: 10.1007/s10815-025-03473-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
PURPOSE AMH inhibits hormone production in luteinized granulosa cells (GCs) and stalls ovarian follicle development in vitro and in vivo. We sought to confirm AMH's mechanism of action through SMAD activation and investigate AMH inhibition of follicle development and function, in vitro and in vivo. METHODS A primary culture of GCs isolated from follicular fluid was used, and cells were treated with recombinant AMH (rAMH) or placebo for 24 h. For the mouse model, 18-weeks old C57BL female mice were either euthanized at the beginning or treated with rAMH or normal saline for 3 weeks. Primordial (PDF), primary follicle (PRF), secondary (SEF), and tertiary follicles (TEF) were calculated. Real-time RT-PCR and ELISA were performed to quantify GC gene expression and protein translation of human SMAD 1, 5, and 8, FSH-R and mouse FSH-R, inhibin B, caspase 3, Ki67, BMP15, GDF9, and the epigenetic regulators miRNAa and b. RESULTS In vitro, rAMH-treated GC showed activation of the SMAD 1, 5 and downregulation of SMAD 8, with greater magnitude at increasing rAMH doses (p < 0.04) and consequential control of downstream regulators. In vivo, the rAMH-treated mice showed increased SEFs and decreased PRFs while PDFs, TEFs, were unchanged compared with baseline. Compared with Placebo, the rAMH group showed increased PDFs, while PRFs, and TEFs were significantly decreased, and SEFs were unchanged. CONCLUSIONS AMH caused SMAD activation in a dose-dependent manner, with downstream downregulation of cell function and replication, also through activation of miRNAs. These mechanisms were confirmed by the in vivo findings with ultimate downregulation of follicular development and preservation of the ovarian follicle number. Counteracting follicular depletion, AMH could be used to protect the ovarian follicle reservoir.
Collapse
Affiliation(s)
- Laura Detti
- Department of Obstetrics and Gynecology, Baylor College of Medicine, 6651 Main Street, Suite F1020, Houston, TX, 77030, USA.
| | - Michael C Mari
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
| | | | - Ghassan M Saed
- The C. S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
3
|
Balough JL, Dipali SS, Velez K, Kumar TR, Duncan FE. Hallmarks of female reproductive aging in physiologic aging mice. NATURE AGING 2024; 4:1711-1730. [PMID: 39672896 DOI: 10.1038/s43587-024-00769-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
The female reproductive axis is one of the first organ systems to age, which has consequences for fertility and overall health. Here, we provide a comprehensive overview of the biological process of female reproductive aging across reproductive organs, tissues and cells based on research with widely used physiologic aging mouse models, and describe the mechanisms that underpin these phenotypes. Overall, aging is associated with dysregulation of the hypothalamic-pituitary-ovarian axis, perturbations of the ovarian stroma, reduced egg quantity and quality, and altered uterine morphology and function that contributes to reduced capacity for fertilization and impaired embryo development. Ultimately, these age-related phenotypes contribute to altered pregnancy outcomes and adverse consequences in offspring. Conserved mechanisms of aging, as well as those unique to the reproductive system, underlie these phenotypes. The knowledge of such mechanisms will lead to development of therapeutics to extend female reproductive longevity and support endocrine function and overall health.
Collapse
Affiliation(s)
- Julia L Balough
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA
| | - Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karen Velez
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - T Rajendra Kumar
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Francesca E Duncan
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA.
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
4
|
Gowkielewicz M, Lipka A, Zdanowski W, Waśniewski T, Majewska M, Carlberg C. Anti-Müllerian hormone: biology and role in endocrinology and cancers. Front Endocrinol (Lausanne) 2024; 15:1468364. [PMID: 39351532 PMCID: PMC11439669 DOI: 10.3389/fendo.2024.1468364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Anti-Müllerian hormone (AMH) is a peptide belonging to the transforming growth factor beta superfamily and acts exclusively through its receptor type 2 (AMHR2). From the 8th week of pregnancy, AMH is produced by Sertoli cells, and from the 23rd week of gestation, it is produced by granulosa cells of the ovary. AMH plays a critical role in regulating gonadotropin secretion, ovarian tissue responsiveness to pituitary hormones, and the pathogenesis of polycystic ovarian syndrome. It inhibits the transition from primordial to primary follicles and is considered the best marker of ovarian reserve. Therefore, measuring AMH concentration of the hormone is valuable in managing assisted reproductive technologies. AMH was initially discovered through its role in the degeneration of Müllerian ducts in male fetuses. However, due to its ability to inhibit the cell cycle and induce apoptosis, it has also garnered interest in oncology. For example, antibodies targeting AMHR2 are being investigated for their potential in diagnosing and treating various cancers. Additionally, AMH is present in motor neurons and functions as a protective and growth factor. Consequently, it is involved in learning and memory processes and may support the treatment of Alzheimer's disease. This review aims to provide a comprehensive overview of the biology of AMH and its role in both endocrinology and oncology.
Collapse
Affiliation(s)
- Marek Gowkielewicz
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Lipka
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Wojciech Zdanowski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tomasz Waśniewski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
5
|
Kontogiannis S, Markantes G, Stamou M, Tsagkarakis M, Mamali I, Giannitsas K, Perimenis P, Georgopoulos N, Athanasopoulos A. Anti-Müllerian hormone: a novel biomarker for aggressive prostate cancer? Emerging evidence from a prospective study of radical prostatectomies. Hormones (Athens) 2024; 23:297-304. [PMID: 38127275 PMCID: PMC11190032 DOI: 10.1007/s42000-023-00520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE Prostate cancer patients are a heterogeneous group as regards the aggressiveness of the disease. The relationship of steroid hormones with the aggressiveness of prostate cancer is unclear. It is known that the anti-Müllerian hormone (AMH) inhibits prostate cancer cell lines in vitro. The aim of this study is to investigate the relationship of AMH and steroid hormones with the aggressiveness of prostate cancer. METHODS This was a prospective study of consecutive radical prostatectomy patients. We measured the following hormones: total testosterone, sex hormone-binding globulin, albumin, luteinizing hormone, follicle-stimulating hormone, estradiol, dehydroepiandrosterone sulfate, androstenedione, and AMH. The minimum follow-up after radical prostatectomy was 5 years. For the aggressiveness of prostate cancer, we considered the following three variables: post-operative Gleason score (GS) ≥ 8, TNM pΤ3 disease, and prostate-specific antigen (PSA) biochemical recurrence (BCR). RESULTS In total, 91 patients were enrolled. The mean age and PSA were 64.8 years and 9.3 ng/dl, respectively. The median post-operative GS was 7. Low AMH blood levels were correlated with higher post-operative GS (p = 0.001), as well as with PSA BCR (p = 0.043). With pT3 disease, only albumin was (negatively) correlated (p = 0.008). ROC analysis showed that AMH is a good predictor of BCR (AUC 0.646, 95% CI 0.510-0.782, p = 0.043); a cutoff value of 3.06 ng/dl had a positive prognostic value of 71.4% and a negative prognostic value of 63.3% for BCR. Cox regression analysis showed that AMH is a statistically significant and independent prognostic marker for BCR (p = 0.013). More precisely, for every 1 ng/ml of AMH rise, the probability for PSA BCR decreases by 20.8% (HR = 0.792). Moreover, in Kaplan-Meier analysis, disease-free survival is more probable in patients with AMΗ ≥ 3.06 ng/ml (p = 0.004). CONCLUSIONS Low AMH blood levels were correlated with aggressive prostate cancer in this radical prostatectomy cohort of patients. Therefore, AMH could be a prognostic biomarker for the aggressiveness of the disease.
Collapse
Affiliation(s)
| | | | - Maria Stamou
- Harvard Reproductive Sciences Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Irini Mamali
- Endocrinology Department, Patras University Hospital, Patras, Greece
| | | | | | | | | |
Collapse
|
6
|
Elias KM, Ng NW, Dam KU, Milne A, Disler ER, Gockley A, Holub N, Seshan ML, Church GM, Ginsburg ES, Anchan RM. Fertility restoration in mice with chemotherapy induced ovarian failure using differentiated iPSCs. EBioMedicine 2023; 94:104715. [PMID: 37482511 PMCID: PMC10435842 DOI: 10.1016/j.ebiom.2023.104715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Treatment options for premature ovarian insufficiency (POI) are limited to hormone replacement and donor oocytes. A novel induced pluripotent stem cell (iPSC) transplant paradigm in a mouse model has potential translational applications for management of POI. METHODS Mouse ovarian granulosa cell derived-iPSCS were labelled with green fluorescent protein (GFP) reporter and differentiated in vitro into oocytes. Differentiated cells were assayed for estradiol and progesterone secretion by enzyme-linked immunosorbent assays. After Fluorescence-Activated Cell Sorting (FACS) for the cell surface marker anti-Mullerian hormone receptor (AMHR2), enriched populations of differentiated cells were surgically transplanted into ovaries of mice that had POI secondary to gonadotoxic pre-treatment with alkylating agents. A total of 100 mice were used in these studies in five separate experiments with 56 animals receiving orthotopic ovarian injections of either FACS sorted or unsorted differentiated iPSCSs and the remaining animals receiving sham injections of PBS diluent. Following transplantation surgery, mice were stimulated with gonadotropins inducing oocyte development and underwent oocyte retrieval. Nine transplanted mice were cross bred with wild-type mice to assess fertility. Lineage tracing of resultant oocytes, F1 (30 pups), and F2 (42 pups) litters was interrogated by GFP expression and validation by short tandem repeat (STR) lineage tracing. FINDINGS [1] iPSCs differentiate into functional oocytes and steroidogenic ovarian cells which [2] express an ovarian (GJA1) and germ cell (ZP1) markers. [3] Endocrine function and fertility were restored in mice pretreated with gonadotoxic alkylating agents via orthotopic transplantation of differentiated iPSCS, thus generating viable, fertile mouse pups. INTERPRETATION iPSC-derived ovarian tissue can reverse endocrine and reproductive sequelae of POI. FUNDING Center for Infertility and Reproductive Surgery Research Award, Siezen Foundation award (RMA). Reproductive Scientist Development Program, Marriott Foundation, Saltonstall Foundation, Brigham Ovarian Cancer Research Fund (K.E).
Collapse
Affiliation(s)
- Kevin M Elias
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Nicholas W Ng
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Kh U Dam
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Ankrish Milne
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Emily R Disler
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Alison Gockley
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Nicole Holub
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Maya L Seshan
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Elizabeth S Ginsburg
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Raymond M Anchan
- Division of Reproductive Endocrinology and Infertility, Center for Infertility and Reproductive Surgery, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA.
| |
Collapse
|
7
|
Pru JK. Low-serum anti-Müllerian hormone in middle-aged women associates with obesity markers. Menopause 2023; 30:237-238. [PMID: 36811962 DOI: 10.1097/gme.0000000000002164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- James K Pru
- From the Program in Reproductive Biology, University of Wyoming, Laramie, WY
| |
Collapse
|
8
|
Royland Marpaung F, Surya Priyanto A, Ayu Kusumawati F, Soehita S, Aryati. Determination of serum anti-Mullerian hormone levels in a low-prognosis women treated in-vitro fertilization/intracytoplasmic sperm injection: A cohort study. Int J Reprod Biomed 2023; 21:255-262. [PMID: 37122893 PMCID: PMC10133738 DOI: 10.18502/ijrm.v21i3.13201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 09/10/2022] [Accepted: 02/14/2023] [Indexed: 09/18/2024] Open
Abstract
Background Outcome prediction of participants treated with in-vitro fertilization or intracytoplasmic sperm injection (IVF/ICSI) using anti-Mullerian hormone (AMH) concentration has been widely used. According to the patient-oriented strategies encompassing individualized oocyte number (POSEIDON) definition, low prognosis Bologna responders have changed from poor. This definition divides low prognosis into 4 groups. Objective The purpose of this study was to assess blood AMH levels in the group of women treated with IVF/ICSI who were thought to have a low prognosis. Materials and Methods A retrospective cohort study among 252 suspected low-prognosis group participants was assessed between January 2016 and December 2019 at Morula IVF, National hospital, Surabaya, Indonesia. Observed AMH serum levels and pregnancy rates were compared among 4 subgroups. Results The AMH cutoff value was 1.7 ng/mL with a sensitivity of 86.7% and a specificity of 70% for diagnosing low-prognosis women using POSEIDON criteria. There was no difference in the pregnancy rate between those groups (p > 0.05). Conclusion AMH levels may indicate a poor prognosis for women having IVF/ICSI in accordance with POSEIDON guidelines. To predict the poor prognosis in women, the cutoff value must be identified.
Collapse
Affiliation(s)
- Ferdy Royland Marpaung
- Department of Clinical Pathology, Faculty of Medicine, Dr Soetomo Academic Hospital, Universitas Airlangga, Surabaya, Indonesia
| | | | | | - Sidarti Soehita
- Department of Clinical Pathology, Faculty of Medicine, Dr Soetomo Academic Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Aryati
- Department of Clinical Pathology, Faculty of Medicine, Dr Soetomo Academic Hospital, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
9
|
Bedenk J, Režen T, Jančar N, Geršak K, Virant Klun I. Effect of In Vitro Maturation of Human Oocytes Obtained After Controlled Ovarian Hormonal Stimulation on the Expression of Development- and Zona Pellucida-Related Genes and Their Interactions. Reprod Sci 2023; 30:667-677. [PMID: 35915350 DOI: 10.1007/s43032-022-01047-1] [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: 05/23/2022] [Accepted: 07/18/2022] [Indexed: 11/26/2022]
Abstract
In an in vitro fertilization program, approximately 10-15% of oocytes obtained after controlled ovarian stimulation are immature, with germinal vesicles (GVs). These oocytes are usually discarded in clinical practice; however, an in vitro maturation (IVM) procedure can be applied to mature them. There are scarce data in the literature on the effect of IVM on the expression of important development- and zona pellucida (ZP)-related genes in human oocytes; therefore, we wanted to determine this. One hundred nine human oocytes were collected from patients enrolled in an intracytoplasmic sperm injection program. The expression of the BMP4, GDF9, ZP1, ZP2, ZP3, and ZP4 genes was analyzed using RT-qPCR in oocytes matured in vitro with different reproductive hormones in the IVM medium (AMH, FSH + hCG, FSH + hCG + AMH), in in vivo matured oocytes and in immature oocytes with GVs. No statistically significant differences in the expression of selected genes in oocytes were observed among groups with different reproductive hormones in IVM medium. However, several interesting significant correlations were found between BMP4 and GDF9, and ZP1 and ZP4; between GDF9 and ZP1, and ZP2 and ZP4; and between ZP1 and ZP3 and ZP4 in the in vitro matured oocytes, while no such correlations were present in other groups of oocytes. The type of reproductive hormone in the maturation medium does not affect the expression of the analyzed genes in oocytes during the maturation process. However, the in vitro maturation procedure itself generated correlations among analyzed genes that were otherwise not present in in vivo matured and immature oocytes.
Collapse
Affiliation(s)
- Jure Bedenk
- Clinical Research Centre, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia.
| | - Tadeja Režen
- Institute of Biochemistry and Molecular Genetics, Centre for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Nina Jančar
- Department of Gynaecology and Obstetrics, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| | - Ksenija Geršak
- Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Irma Virant Klun
- Clinical Research Centre, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| |
Collapse
|
10
|
Sinha N, Driscoll CS, Qi W, Huang B, Roy S, Knott JG, Wang J, Sen A. Anti-Müllerian hormone treatment enhances oocyte quality, embryonic development and live birth rate†. Biol Reprod 2022; 107:813-822. [PMID: 35657015 PMCID: PMC9476226 DOI: 10.1093/biolre/ioac116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/24/2022] [Accepted: 05/24/2022] [Indexed: 03/13/2025] Open
Abstract
The anti-Müllerian hormone (AMH) produced by the granulosa cells of growing follicles is critical for folliculogenesis and is clinically used as a diagnostic and prognostic marker of female fertility. Previous studies report that AMH-pretreatment in mice creates a pool of quiescent follicles that are released following superovulation, resulting in an increased number of ovulated oocytes. However, the quality and developmental competency of oocytes derived from AMH-induced accumulated follicles as well as the effect of AMH treatment on live birth are not known. This study reports that AMH priming positively affects oocyte maturation and early embryonic development culminating in higher number of live births. Our results show that AMH treatment results in good-quality oocytes with greater developmental competence that enhances embryonic development resulting in blastocysts with higher gene expression. The transcriptomic analysis of oocytes from AMH-primed mice compared with those of control mice reveal that AMH upregulates a large number of genes and pathways associated with oocyte quality and embryonic development. Mitochondrial function is the most affected pathway by AMH priming, which is supported by more abundant active mitochondria, mitochondrial DNA content and adenosine triphosphate levels in oocytes and embryos isolated from AMH-primed animals compared with control animals. These studies for the first time provide an insight into the overall impact of AMH on female fertility and highlight the critical knowledge necessary to develop AMH as a therapeutic option to improve female fertility.
Collapse
Affiliation(s)
- Niharika Sinha
- Reproductive and Developmental Sciences Program, Department of Animal Science, East Lansing, MI, USA
| | - Chad S Driscoll
- Reproductive and Developmental Sciences Program, Department of Animal Science, East Lansing, MI, USA
| | - Wenjie Qi
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Binbin Huang
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Sambit Roy
- Reproductive and Developmental Sciences Program, Department of Animal Science, East Lansing, MI, USA
| | - Jason G Knott
- Reproductive and Developmental Sciences Program, Department of Animal Science, East Lansing, MI, USA
| | - Jianrong Wang
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Aritro Sen
- Reproductive and Developmental Sciences Program, Department of Animal Science, East Lansing, MI, USA
| |
Collapse
|
11
|
Farhat SA, Jabbari F, Jabbari P, Rezaei N. Targeting signaling pathways involved in primordial follicle growth or dormancy: potential application in prevention of follicular loss and infertility. Expert Opin Biol Ther 2022; 22:871-881. [PMID: 35658707 DOI: 10.1080/14712598.2022.2086042] [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] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Premature ovarian failure (POF) is one of the important causes of infertility in females. To date, no efficient preventive pharmacological treatment has been offered to prevent POF. Therefore, it is necessary to focus on strategies that provide a normal reproductive lifespan to females at risk of developing POF. AREAS COVERED Recently, attention has been drawn to discovering pathways involved in primordial follicle activation, as the inhibition of this process might maintain the stock of primordial follicles and therefore, prevent POF. In vitro and animal studies have resulted in the discovery of several of these pathways that can be used to develop new treatments for POF. These studies show crosstalk of these pathways at different levels. One of the important crossing points of many of these pathways involves anti-Mullerian hormone (AMH). Herein, we discuss different aspects of this topic by reviewing related published articles indexed in PubMed and Web of Science as of December 2021. EXPERT OPINION Although the findings seem promising, most of the studies were conducted on animals, and the interaction between these factors and the possible outcomes of their administration in the long term are still unknown. Therefore, further investigation is necessary to assess these aspects.
Collapse
Affiliation(s)
- Sara Ali Farhat
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Forouq Jabbari
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parnian Jabbari
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Bedenk J, Režen T, Železnik Ramuta T, Jančar N, Vrtačnik Bokal E, Geršak K, Virant Klun I. Recombinant anti-Müllerian hormone in the maturation medium improves the in vitro maturation of human immature (GV) oocytes after controlled ovarian hormonal stimulation. Reprod Biol Endocrinol 2022; 20:18. [PMID: 35073905 PMCID: PMC8785574 DOI: 10.1186/s12958-022-00895-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/16/2022] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND In vitro maturation (IVM) of oocytes is a laboratory method that allows the maturation of immature (GV) oocytes retrieved from patients enrolled in the in vitro fertilization (IVF) programme. However, this method is still sparsely researched and used in clinical practice, leading to suboptimal clinical results. Anti-Müllerian hormone (AMH) is an important hormone with known effects on human ovaries, especially on follicles (follicular cells) during folliculogenesis. In contrast, the effect of AMH on the human oocyte itself is unknown. Therefore, we wanted to determine whether human oocytes express AMH receptor 2 (AMHR2) for this hormone. Recombinant AMH was added to the IVM medium to determine whether it affected oocyte maturation. METHODS In total, 247 human oocytes (171 immature and 76 mature) were collected from patients enrolled in the intracytoplasmic sperm injection (ICSI) programme who were aged 20 to 43 years and underwent a short antagonist protocol of ovarian stimulation. The expression of AMHR2 protein and AMHR2 gene was analysed in immature and mature oocytes. Additionally, maturation of GV oocytes was performed in vitro in different maturation media with or without added AMH to evaluate the effect of AMH on the oocyte maturation rate. RESULTS Immunocytochemistry and confocal microscopy revealed that AMHR2 protein is expressed in both immature and mature human oocytes. AMHR2 was expressed in a spotted pattern throughout the whole oocyte. The IVM procedure revealed that AMH in maturation medium improved GV oocyte maturation in vitro, as all oocytes were successfully matured in maturation medium containing recombinant AMH only. Furthermore, antagonism between AMH and follicle-stimulating hormone (FSH) during the maturation process was observed, with fewer oocytes maturing when both AMH and FSH were added to the maturation medium. Finally, AMHR2 gene expression was found in immature and in vitro matured oocytes but absent in mature oocytes. CONCLUSIONS The positive AMHR2 protein and AMHR2 gene expression in human oocytes shows that AMH could directly act on human oocytes. This was further functionally confirmed by the IVM procedure. These findings suggest the potential clinical application of recombinant AMH to improve IVM of human oocytes in the future.
Collapse
Affiliation(s)
- Jure Bedenk
- Clinical Research Centre, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia.
| | - Tadeja Režen
- Institute of Biochemistry and Molecular Genetics, Centre for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Taja Železnik Ramuta
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Nina Jančar
- Department of Gynaecology and Obstetrics, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| | - Eda Vrtačnik Bokal
- Department of Gynaecology and Obstetrics, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| | - Ksenija Geršak
- Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Irma Virant Klun
- Clinical Research Centre, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| |
Collapse
|
13
|
Kanakatti Shankar R, Dowlut-McElroy T, Dauber A, Gomez-Lobo V. Clinical Utility of Anti-Mullerian Hormone in Pediatrics. J Clin Endocrinol Metab 2022; 107:309-323. [PMID: 34537849 PMCID: PMC8764360 DOI: 10.1210/clinem/dgab687] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT Anti-Mullerian hormone (AMH) was originally described in the context of sexual differentiation in the male fetus but has gained prominence now as a marker of ovarian reserve and fertility in females. In this mini-review, we offer an updated synopsis on AMH and its clinical utility in pediatric patients. DESIGN AND RESULTS A systematic search was undertaken for studies related to the physiology of AMH, normative data, and clinical role in pediatrics. In males, AMH, secreted by Sertoli cells, is found at high levels prenatally and throughout childhood and declines with progression through puberty to overlap with levels in females. Thus, serum AMH has clinical utility as a marker of testicular tissue in males with differences in sexual development and cryptorchidism and in the evaluation of persistent Mullerian duct syndrome. In females, serum AMH has been used as a predictive marker of ovarian reserve and fertility, but prepubertal and adolescent AMH assessments need to be interpreted cautiously. AMH is also a marker of tumor burden, progression, and recurrence in germ cell tumors of the ovary. CONCLUSIONS AMH has widespread clinical diagnostic utility in pediatrics but interpretation is often challenging and should be undertaken in the context of not only age and sex but also developmental and pubertal stage of the child. Nonstandardized assays necessitate the need for assay-specific normative data. The recognition of the role of AMH beyond gonadal development and maturation may usher in novel diagnostic and therapeutic applications that would further expand its utility in pediatric care.
Collapse
Affiliation(s)
- Roopa Kanakatti Shankar
- Division of Endocrinology, Children’s National Hospital, Washington DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Correspondence: Roopa Kanakatti Shankar, MBBS, MS, George Washington University School of Medicine, Endocrinologist, Children’s National Hospital, 111 Michigan Ave NW, Washington DC, 20010, USA.
| | - Tazim Dowlut-McElroy
- Pediatric and Adolescent Gynecology Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Andrew Dauber
- Division of Endocrinology, Children’s National Hospital, Washington DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Veronica Gomez-Lobo
- Pediatric and Adolescent Gynecology Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| |
Collapse
|
14
|
Wang L, Yao Y, Xu C, Wang X, Wu D, Hong Z. Exploration of the Tumor Mutational Burden as a Prognostic Biomarker and Related Hub Gene Identification in Prostate Cancer. Technol Cancer Res Treat 2021; 20:15330338211052154. [PMID: 34806485 PMCID: PMC8606726 DOI: 10.1177/15330338211052154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To explore the signature function of the tumor mutational burden (TMB) and
potential biomarkers in prostate cancer (PCa), transcriptome profiles, somatic
mutation data, and clinicopathologic feature information were downloaded from
The Cancer Genome Atlas (TCGA) database. R software package was used to generate
a waterfall plot to summarize the specific mutation information and calculate
the TMB value of PCa. Least absolute shrinkage and selection operator (LASSO)
Cox regression analysis was used to select the hub genes related to the TMB from
the ImmPort network to build a risk score (RS) model to evaluate prognostic
values and plot Kaplan–Meier (K-M) curves to predict PCa patients survival. The
results showed that PCa patients with a high TMB exhibited higher infiltration
of CD8+ T cells and CD4+ T cells and better overall survival (OS) than those
with a low TMB. The anti-Mullerian hormone (AMH), baculoviral IAP
repeat-containing 5 (BIRC5), and opoid receptor kappa 1 (OPRK1) genes were three
hub genes and their copy number variation (CNV) was relatively likely to affect
the infiltration of immune cells. Moreover, PCa patients with low AMH or BIRC5
expression had a longer survival time and lower cancer recurrence, while
elevated AMH or BIRC5 expression favored PCa progression. In contrast, PCa
patients with low OPRK1 expression had poorer OS in the early stage of PCa and a
higher recurrent rate than those with high expression. Taken together, these
results suggest that the TMB may be a promising prognostic biomarker for PCa and
that AMH, OPRK1, and BIRC5 are hub genes affecting the TMB; AMH, OPRK1, and
BIRC5 could serve as potential immunotherapeutic targets for PCa treatment.
Collapse
Affiliation(s)
- Licheng Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yicong Yao
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinan Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhe Hong
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
15
|
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: 101] [Impact Index Per Article: 25.3] [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
|
16
|
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
|
17
|
Barad DH. Antimüllerian hormone-levels not written in stone. Fertil Steril 2021; 116:83-84. [PMID: 34148596 DOI: 10.1016/j.fertnstert.2021.05.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Affiliation(s)
- David H Barad
- The Center for Human Reproduction, and the Foundation for Reproductive Medicine, New York, New York
| |
Collapse
|
18
|
Bahadur A, Verma N, Mundhra R, Chawla L, Ajmani M, Sri MS, Arora S. Correlation of Homeostatic Model Assessment-Insulin Resistance, Anti-Mullerian Hormone, and BMI in the Characterization of Polycystic Ovary Syndrome. Cureus 2021; 13:e16047. [PMID: 34336524 PMCID: PMC8321419 DOI: 10.7759/cureus.16047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2021] [Indexed: 11/16/2022] Open
Abstract
Objective: To evaluate the correlation of homeostatic model assessment-insulin resistance (HOMA-IR), anti-Mullerian hormone (AMH), and BMI and to compare their values across the different phenotypes in polycystic ovary syndrome (PCOS) women of the reproductive age group. Study design: A total of 307 PCOS-diagnosed women were included in the study and further classified in different phenotypes. BMI, HOMA-IR, and serum AMH values were noted and their association with different phenotypes was seen. The correlation of these variables was also noted. Results: Phenotype D was the most common phenotype followed by type A, type B, and type C. A total of 265 women had an AMH value of ≥4 mg/ml with the highest value in phenotype A followed by D, C, and B. HOMA-IR and BMI values did not vary significantly among different phenotypes. HOMA-IR and BMI had a statistically significant positive correlation and serum AMH was negatively correlated with HOMA-IR, but no significant correlation was seen between serum AMH and BMI. The biochemical parameters like luteinizing hormone (LH), follicle-stimulating hormone (FSH), LH: FSH ratio, and serum testosterone showed no correlation with phenotypes or any other clinical parameter. Conclusion: HOMA-IR and BMI showed a statistically significant positive correlation indicating the need for lifestyle modification and weight reduction in PCOS women, which can further help in decreasing insulin resistance. A strong correlation of serum AMH levels and phenotypes shows the importance of serum AMH levels for classifying different PCOS phenotypes.
Collapse
Affiliation(s)
- Anupama Bahadur
- Obstetrics and Gynecology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Neha Verma
- Obstetrics and Gynecology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Rajlaxmi Mundhra
- Obstetrics and Gynecology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Latika Chawla
- Obstetrics and Gynecology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Megha Ajmani
- Obstetrics and Gynecology, All India Institute of Medical Sciences, Rishikesh, IND
| | | | - Shivaani Arora
- Obstetrics and Gynecology, All India Institute of Medical Sciences, Rishikesh, IND
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW In this review, we will summarize research looking into anti-Müllerian hormone (AMH) as a measure of oocyte quality. RECENT FINDINGS AMH is a key factor involved in embryogenesis but also in the development of early follicles. Owing to its relatively small inter and intracycle variability, it has become a widely used method of ovarian reserve testing. In the realm of assisted reproductive technology, it has demonstrated a reliable ability to gauge the number of oocytes obtained during an in-vitro fertilization cycle. For these purposes, AMH is a readily measured quantitative tool. However, its qualitative role is as yet undefined. SUMMARY Although levels of this hormone have been associated with fertilization, blastulation, implantation, and clinical pregnancy rates, there is no clear link with live-birth rates. Furthermore, AMH levels do not appear to correspond with risk of fetal trisomy. AMH does show significant predictive value for the risk of premature ovarian insufficiency and time to onset of menopause.
Collapse
|
20
|
Lee S, Ozkavukcu S, Ku SY. Current and Future Perspectives for Improving Ovarian Tissue Cryopreservation and Transplantation Outcomes for Cancer Patients. Reprod Sci 2021; 28:1746-1758. [PMID: 33791995 PMCID: PMC8144135 DOI: 10.1007/s43032-021-00517-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/21/2021] [Indexed: 02/06/2023]
Abstract
Although advances in cancer treatment and early diagnosis have significantly improved cancer survival rates, cancer therapies can cause serious side effects, including ovarian failure and infertility, in women of reproductive age. Infertility following cancer treatment can have significant adverse effects on the quality of life. However, established methods for fertility preservation, including embryo or oocyte cryopreservation, are not always suitable for female cancer patients because of complicated individual conditions and treatment methods. Ovarian tissue cryopreservation and transplantation is a promising option for fertility preservation in pre-pubertal girls and adult patients with cancer who require immediate treatment, or who are not eligible to undergo ovarian stimulation. This review introduces various methods and strategies to improve ovarian tissue cryopreservation and transplantation outcomes, to help patients and clinicians choose the best option when considering the potential complexity of a patient's situation. Effective multidisciplinary oncofertility strategies, involving the inclusion of a highly skilled and experienced oncofertility team that considers cryopreservation methods, thawing processes and devices, surgical procedures for transplantation, and advances in technologies, are necessary to provide high-quality care to a cancer patient.
Collapse
Affiliation(s)
- Sanghoon Lee
- Moores Cancer Center, University of California San Diego, San Diego, CA, USA.
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Sinan Ozkavukcu
- Center for Assisted Reproduction, Department of Obstetrics and Gynecology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
21
|
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: 0.5] [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
|
22
|
Loubersac S, Dezellus A, Lefebvre T, Reignier A, Barriere P, Masson D, Freour T. Evolution of serum Anti-Müllerian Hormone (AMH) level in young women treated with chemotherapy for breast cancer according to basal AMH level. Eur J Obstet Gynecol Reprod Biol 2020; 254:132-137. [PMID: 32971432 DOI: 10.1016/j.ejogrb.2020.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Serum AMH level has been shown to decrease in women treated for breast cancer in several studies. However, whether basal AMH status affects AMH dynamics during chemotherapy remains to be clarified. The objective of this study was to compare serum AMH dynamics in young women with either low, normal or high basal serum AMH level at diagnosis, during and after treatment with chemotherapy for breast cancer. STUDY DESIGN In this secondary analysis of a prospective cohort study, serum AMH was measured during and after chemotherapy in 239 women of reproductive age diagnosed with breast cancer and treated with chemotherapy. The association between AMH dynamics throughout chemotherapy and during follow-up and basal AMH status, i.e. low AMH (<1 μg/l, <7 pmol/l), normal AMH (1-4.9 μg/l, 7-36 pmol/l) and high AMH (≥5 μg/l, >36 pmol/l), was evaluated. Menses occurrence was also recorded. RESULTS A total of 21 women had low, 154 had normal and 64 had high basal AMH level. Serum AMH rapidly decreased during chemotherapy in all groups, and its variation during chemotherapy and follow-up was not significantly different between the 3 groups. CONCLUSION No association was found between AMH variation during chemotherapy and follow-up, and basal AMH level at diagnosis. However, women with high basal AMH levels have significantly higher AMH levels throughout chemotherapy and follow-up than women with normal or low basal AMH levels at diagnosis.
Collapse
Affiliation(s)
- S Loubersac
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France
| | - A Dezellus
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Service d'oncologie, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain, France
| | - T Lefebvre
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France
| | - A Reignier
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Inserm, Université de Nantes, Centre de Recherche en Transplantation et Immunologie (ou CRTI), Nantes, France
| | - P Barriere
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Inserm, Université de Nantes, Centre de Recherche en Transplantation et Immunologie (ou CRTI), Nantes, France
| | - D Masson
- Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Laboratoire de biochimie, CHU de Nantes, place Alexis-Ricordeau, 44000, Nantes, France; INSERM UMR 913, rue Gaston Veil, 44035, Nantes, France
| | - T Freour
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Inserm, Université de Nantes, Centre de Recherche en Transplantation et Immunologie (ou CRTI), Nantes, France.
| | | |
Collapse
|
23
|
Detti L, Abuzeid MI, Peregrin-Alvarez I, Christiansen ME, Malekzadeh P, Sledge J, Saed GM. Recombinant Anti-Müllerian Hormone (rAMH) for Stalling In Vitro Granulosa Cell Replication. Reprod Sci 2020; 27:1873-1878. [PMID: 32617880 DOI: 10.1007/s43032-020-00206-6] [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: 02/17/2020] [Accepted: 04/16/2020] [Indexed: 11/28/2022]
Abstract
To investigate whether recombinant AMH (rAMH) is able to decrease cellular proliferation/apoptosis in luteinized granulosa cells (GCs) through hormonal regulation, a primary culture of GCs was established from GCs obtained at time of oocyte retrieval from follicular fluid of 3 patients. Cells were seeded in well cell culture plates at a density of 100,000 cells/well in medium and treated with rAMH 20 ng/ml (rAMH group), or phosphate-buffered saline (PBS-control group), for 24 h. Total RNA was extracted from all cells, followed by cDNA synthesis and real-time RT-PCR to quantify the expression levels of AMH, AMH-R2, FSH-R, inhibin B, cell proliferation (Ki67), and apoptosis (Caspase 3). We used independent sample t test (SPSS v25) and a p < 0.05 significance. Cellular expressions of AMH, AMH-R2, FSH-R, and inhibin B were reduced greater than 50% in the rAMH group, compared with that of the the control group (p ≤ 0.005 for all). Ki67 and Caspase3 were also reduced greater than 30% in the rAMH group (p ≤ 0.001 for both). Our findings show a direct inhibitory effect of AMH on luteinized GCs' expression of the major regulatory hormones, in addition to a significant decrease in markers of cell proliferation and apoptosis. These results confirm the inhibitory effects of AMH on follicular development.
Collapse
Affiliation(s)
- Laura Detti
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN, USA. .,Department of Obstetrics and Gynecology Subspecialties, Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA.
| | | | - Irene Peregrin-Alvarez
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mary E Christiansen
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Pouran Malekzadeh
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jennifer Sledge
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ghassan M Saed
- The C. S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
24
|
Bozhokin MS, Sopova YV, Kachkin DV, Rubel AA, Khotin MG. Mechanisms of TGFβ3 Action as a Therapeutic Agent for Promoting the Synthesis of Extracellular Matrix Proteins in Hyaline Cartilage. BIOCHEMISTRY (MOSCOW) 2020; 85:436-447. [PMID: 32569551 DOI: 10.1134/s0006297920040045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hyaline cartilage is a nonvascular connective tissue covering the joint surface. It consists mostly of the extracellular matrix proteins and a small number of highly differentiated chondrocytes. At present, various techniques for repairing joint surfaces damage, for example, the use of modified cell cultures and biodegradable scaffolds, are under investigation. Molecular mechanisms of cartilage tissue proliferation have been also actively studied in recent years. TGFβ3, which plays a critical role in the proliferation of normal cartilage tissue, is one of the most important protein among cytokines and growth factors affecting chondrogenesis. By interacting directly with receptors on the cell membrane surface, TGFβ3 triggers a cascade of molecular interactions involving transcription factor Sox9. In this review, we describe the effects of TGFβ3 on the receptor complex activation and subsequent intracellular trafficking of Smad proteins and analyze the relation between these processes and upregulation of expression of major extracellular matrix genes, such as col2a1 and acan.
Collapse
Affiliation(s)
- M S Bozhokin
- Vreden Russian Scientific Research Institute of Traumatology and Orthopedics, St. Petersburg, 195427, Russia. .,Institute of Cytology, Russian Academy of Science, St. Petersburg, 194064, Russia
| | - Y V Sopova
- Vavilov Institute of General Genetics, Russian Academy of Science, St. Petersburg Branch, St. Petersburg, 199034, Russia.,St. Petersburg State University, Faculty of Biology, St. Petersburg, 199034, Russia.,St. Petersburg State University, Laboratory of Amyloid Biology, St. Petersburg, 199034, Russia
| | - D V Kachkin
- St. Petersburg State University, Faculty of Biology, St. Petersburg, 199034, Russia.,St. Petersburg State University, Laboratory of Amyloid Biology, St. Petersburg, 199034, Russia
| | - A A Rubel
- St. Petersburg State University, Faculty of Biology, St. Petersburg, 199034, Russia.,St. Petersburg State University, Laboratory of Amyloid Biology, St. Petersburg, 199034, Russia
| | - M G Khotin
- Institute of Cytology, Russian Academy of Science, St. Petersburg, 194064, Russia
| |
Collapse
|
25
|
Hormonal Effects in Reproductive Technology with Focus on Diminished Ovarian Reserve. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32406026 DOI: 10.1007/978-3-030-38474-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Abstract
Modern use of reproductive technologies has revolutionized the treatment of infertile couples. Strategies to improve ovarian function in cases of diminished ovarian reserve are perhaps the least understood area in this field and will be the chief focus of this chapter.
Collapse
|
26
|
Anti-Müllerian hormone levels and risk of cancer: A systematic review. Maturitas 2020; 135:53-67. [DOI: 10.1016/j.maturitas.2020.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/21/2020] [Accepted: 03/07/2020] [Indexed: 02/06/2023]
|
27
|
Association of Anti-Mullerian Hormone with C-Reactive Protein in Men. Sci Rep 2019; 9:13081. [PMID: 31511566 PMCID: PMC6739398 DOI: 10.1038/s41598-019-49596-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/28/2019] [Indexed: 11/08/2022] Open
Abstract
AbstractWhile serum anti-mullerian hormone (AMH) levels are inversely associated with all-cause mortality in men, the underlying mechanisms are unclear. Elevated levels of inflammation, also associated with all-cause mortality, and may be the link between AMH and mortality. Hence, we examined the association of AMH with serum c-reactive protein (CRP), a biomarker of inflammation, in men. We included men ≥20 years from the National Health and Nutrition Examination Survey (1999–2004). We used survey weight-adjusted linear regression to examine the association between AMH and CRP without and with adjustment for age, race, body mass index (BMI), smoking, hypertension, diabetes, cholesterol, glomerular filtration rate (GFR), testosterone, androstenedione, and sex hormone binding globulin. Of the 949 men, 212 (22%) were elderly, 493 (52%) Caucasian, 254 (27%) current smokers, 100 (10%) diabetics, and 312 (33%) had hypertension. Mean (SD) AMH was 8.4 (7.2) ng/mL and median (IQR) CRP level was 0.17 (3) mg/L. Using linear regression, each 10 ng/mL rise in AMH was associated with 0.09 mg/dL (95%CI = −0.14 to −0.03; p = 0.002) decrease before and 0.08 mg/dL (95%CI = −0.13 to −0.02; p = 0.004) decrease in CRP after adjusting for potential confounders. Similarly, men in the highest quartile of AMH had significantly lower CRP compared to those in the lowest quartile (unadjusted difference = −0.19 mg/dL; 95%CI = −0.31 to −0.06; p = 0.006, adjusted difference = −0.16 mg/dL; 95%CI = −0.3 to −0.01; p = 0.035). We found an independent, robust, and inverse association between CRP and AMH in men. Effect of AMH on mortality may be through amelioration of inflammation.
Collapse
|
28
|
The role of anti-Müllerian hormone (AMH) in ovarian disease and infertility. J Assist Reprod Genet 2019; 37:89-100. [PMID: 31755000 DOI: 10.1007/s10815-019-01622-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
PURPOSE In this review, the current knowledge on anti-Müllerian hormone (AMH) is presented, concerning its value in disease and IVF treatment as well as in terms of its prospective clinical use. METHODS AMH is becoming the most appropriate biomarker for the ovarian reserve measured predominantly for assisted reproductive treatment (ART) patients in comparison to the currently used antral follicle count (AFC). However, this is not the only way AMH measurements can be used in the clinics. Because of this, we reviewed the current literature for the use of AMH in current or prospective clinical practice. RESULTS We found that AMH has a high predictive value in assessing the ovarian reserve, which can lead to a better efficiency of in vitro fertilization (IVF) procedures. It has a high potential to be developed as a staple diagnostic marker of ovarian disease, especially for ovarian cancers and even as a possible treatment tool for certain cancers. It could potentially be used to prevent oocyte loss due to chemo- or radiotherapy. CONCLUSION AMH is an important hormone especially in women reproductive organs and is currently seen as the best biomarker for a multitude of uses in reproductive medicine. Currently, the biggest issue lies in the lack of international standardization of AMH. However, it is encouraging to see that there is interest in AMH in the form of research on its action and use in reproductive medicine.
Collapse
|
29
|
Kereilwe O, Pandey K, Borromeo V, Kadokawa H. Anti-Müllerian hormone receptor type 2 is expressed in gonadotrophs of postpubertal heifers to control gonadotrophin secretion. Reprod Fertil Dev 2019. [PMID: 29533759 DOI: 10.1071/rd17377] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Preantral and small antral follicles may secret anti-Müllerian hormone (AMH) to control gonadotrophin secretion from ruminant gonadotrophs. The present study investigated whether the main receptor for AMH, AMH receptor type 2 (AMHR2), is expressed in gonadotrophs of postpubertal heifers to control gonadotrophin secretion. Expression of AMHR2 mRNA was detected in anterior pituitaries (APs) of postpubertal heifers using reverse transcription-polymerase chain reaction. An anti-AMHR2 chicken antibody was developed against the extracellular region near the N-terminus of bovine AMHR2. Western blotting using this antibody detected the expression of AMHR2 protein in APs. Immunofluorescence microscopy using the same antibody visualised colocalisation of AMHR2 with gonadotrophin-releasing hormone (GnRH) receptor on the plasma membrane of gonadotrophs. AP cells were cultured for 3.5 days and then treated with increasing concentrations (0, 1, 10, 100, or 1000pgmL-1) of AMH. AMH (10-1000pgmL-1) stimulated (P<0.05) basal FSH secretion. In addition, AMH (100-1000pgmL-1) weakly stimulated (P<0.05) basal LH secretion. AMH (100-1000pgmL-1) inhibited GnRH-induced FSH secretion, but not GnRH-induced LH secretion, in AP cells. In conclusion, AMHR2 is expressed in gonadotrophs of postpubertal heifers to control gonadotrophin secretion.
Collapse
Affiliation(s)
- Onalenna Kereilwe
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| | - Kiran Pandey
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| | - Vitaliano Borromeo
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, 26900, Italy
| | - Hiroya Kadokawa
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| |
Collapse
|
30
|
Roness H, Spector I, Leichtmann-Bardoogo Y, Savino AM, Dereh-Haim S, Meirow D. Pharmacological administration of recombinant human AMH rescues ovarian reserve and preserves fertility in a mouse model of chemotherapy, without interfering with anti-tumoural effects. J Assist Reprod Genet 2019; 36:1793-1803. [PMID: 31250176 PMCID: PMC6730972 DOI: 10.1007/s10815-019-01507-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To determine whether pharmacological administration of recombinant human anti-Mullerian hormone (rAMH) protects the ovarian reserve and preserves fertility without interfering with anti-tumoural cytotoxic action of chemotherapy. METHODS Intraperitoneal delivery of rAMH and ovarian post-receptor activity were assessed with immunohistochemistry and western blot. Differential follicle counts and reproductive outcomes were assessed after cyclophosphamide (Cy) administration, with/without concurrent administration of rAMH. Interference of rAMH with Cy chemotoxicity was assessed on a human breast cancer cell line and an in vivo mouse model of human leukaemia. RESULTS rAMH reached the ovary after intraperitoneal injection and demonstrated post-receptor bioactivity. Cy administration in mice caused primordial follicle activation, as shown by a decrease in primordial follicle population accompanied by an increase in early growing follicles and granulosa cell proliferation. Co-administration of rAMH reduced follicle activation, thereby protecting the primordial follicle reserve, and improving long-term fertility and reproductive outcomes. rAMH co-administration did not interfere with the cytotoxic actions of Cy in vitro on breast cancer cell line or in vivo in a model of human leukaemia. CONCLUSION This study demonstrates that rAMH is bioactive in the ovary for a limited time, and that pharmacological administration of rAMH during chemotherapy treatment reduces follicle activation and primordial follicle loss and significantly improves reproductive outcomes in a mouse model, and does not interfere with the therapeutic actions of the treatment. Further investigation is necessary to determine whether it has similar protective effects in the human ovary.
Collapse
Affiliation(s)
- H Roness
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - I Spector
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - Y Leichtmann-Bardoogo
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - A M Savino
- Department of Human Molecular Genetics and Biochemistry, Sheba Medical Centre, Ramat Gan, Israel
| | - Sanaz Dereh-Haim
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - Dror Meirow
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
31
|
Zamarin D. Novel therapeutics: response and resistance in ovarian cancer. Int J Gynecol Cancer 2019; 29:s16-s21. [PMID: 31462544 PMCID: PMC7368996 DOI: 10.1136/ijgc-2019-000456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/16/2019] [Indexed: 11/04/2022] Open
Abstract
Here we review the latest pre-clinical and clinical developments for treatment of ovarian cancer, presented at the American Association of Cancer Research/Rivkin Center Ovarian Cancer Research Symposium held at the University of Washington in September 2018. Abstracts and presentations pertaining to the 'Novel Therapeutics' session were reviewed and are summarized here. The session featured a keynote presentation from Dr Ursula Matulonis, who summarized the current state of the art of treatment of ovarian cancer, including recent clinical trials incorporating the use of novel agents, including poly-ADP-ribose polymerase (PARP) inhibitors, other DNA-damaging agents, vascular endothelial growth factor receptor inhibitors, mirvetuximab soravtansine, and immune checkpoint blockade. Dr Jung-Min Lee then summarized the rationale and the results of early studies for targeting cell cycle checkpoint kinases for anti-cancer therapy. Eight submissions were selected for oral presentations, and 36 abstracts were presented as posters. The topics covered a range of clinical and pre-clinical strategies and biomarkers, including immunotherapy, mechanisms of chemotherapy, and PARP inhibitor resistance, DNA-damaging agents, and other novel therapeutic strategies. Key studies have highlighted that resistance to chemotherapy and PARP inhibitors remain a major challenge in therapy of ovarian cancer. Cancer stem cells represent an important mechanism of chemoresistance and strategies to target these cells may be a pathway to prevention of ovarian cancer relapse. Advancement of novel therapeutics targeting DNA damage, cell metabolism, and endoplasmic reticulum present some of the novel strategies in the pipeline. Emerging compelling pre-clinical data with novel antibody-drug conjugates targeting various surface receptors in ovarian cancer alone and in combination with immune checkpoint blockade generate a strong enthusiasm for rapid translation of these strategies to clinic.
Collapse
Affiliation(s)
- Dmitriy Zamarin
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
32
|
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: 1.2] [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
|
33
|
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: 0.9] [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
|
34
|
Hsu JY, James KE, Bormann CL, Donahoe PK, Pépin D, Sabatini ME. Müllerian-Inhibiting Substance/Anti-Müllerian Hormone as a Predictor of Preterm Birth in Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2018; 103:4187-4196. [PMID: 30239805 DOI: 10.1210/jc.2018-01320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023]
Abstract
CONTEXT There is increasing evidence for Müllerian-inhibiting substance (MIS)/anti-Müllerian hormone (AMH) physiologic activity in the human uterus, so it is relevant to study how MIS/AMH levels impact pregnancy. OBJECTIVE To investigate the association of MIS/AMH levels with the risk of adverse obstetric outcomes. DESIGN Retrospective cohort study. SETTING Academic fertility center. PATIENTS Women who became pregnant through in vitro fertilization between January 2012 and October 2016. Exclusion criteria were: oocyte donation, gestational carrier, multiple gestations, miscarriage before 20 weeks, or medically indicated preterm deliveries. INTERVENTIONS None. MAIN OUTCOME MEASURES There were two primary outcomes, preterm birth and cesarean delivery for arrest of labor. Because MIS/AMH level is highly skewed by certain infertility diagnoses, the preterm birth analysis was stratified by polycystic ovary syndrome (PCOS) diagnosis, and the cesarean delivery for arrest of labor analysis was stratified by diminished ovarian reserve diagnosis. χ2, Mann-Whitney, and t tests were used as appropriate. A P value of <0.05 was considered statistically significant. RESULTS Among women with PCOS, those who delivered prematurely had substantially higher MIS/AMH levels (18 vs 6.4 ng/mL, P = 0.003) than did those who delivered at term. At the highest MIS/AMH values, preterm deliveries predominated; above the 90th percentile in women with PCOS, all deliveries were premature. No effect of MIS/AMH level was observed in women without PCOS. We found no association between MIS/AMH values and cesarean delivery for labor arrest. CONCLUSION In women with PCOS, substantially elevated MIS/AMH levels are significantly associated with preterm birth, suggesting closer follow-up and further studies to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Jennifer Y Hsu
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Kaitlyn E James
- Deborah Kelly Center for Outcomes Research, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
| | - Charles L Bormann
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Patricia K Donahoe
- Harvard Medical School, Boston, Massachusetts
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - David Pépin
- Harvard Medical School, Boston, Massachusetts
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Mary E Sabatini
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Roy S, Gandra D, Seger C, Biswas A, Kushnir VA, Gleicher N, Kumar TR, Sen A. Oocyte-Derived Factors (GDF9 and BMP15) and FSH Regulate AMH Expression Via Modulation of H3K27AC in Granulosa Cells. Endocrinology 2018; 159:3433-3445. [PMID: 30060157 PMCID: PMC6112599 DOI: 10.1210/en.2018-00609] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 07/20/2018] [Indexed: 12/15/2022]
Abstract
Anti-Müllerian hormone (AMH) produced by ovarian granulosa cells (GCs) plays a crucial role in ovarian function. It is used as a diagnostic and/or prognostic marker of fertility as well as for pathophysiological conditions in women. In this study, we investigated the underlying mechanism for regulation of AMH expression in GCs using primary mouse GCs and a human GC tumor-derived KGN cell line. We find that growth differentiation factor 9 (GDF9) and bone morphogenetic factor 15 (BMP15) together (GDF9 + BMP15), but not when tested separately, significantly induce AMH expression in vitro and in vivo (serum AMH). Our results show that GDF9 + BMP15 through the PI3K/Akt and Smad2/3 pathways synergistically recruit the coactivator p300 on the AMH promoter region that promotes acetylation of histone 3 lysine 27 (H3K27ac), facilitating AMH/Amh expression. Intriguingly, we also find that FSH inhibits GDF9 + BMP15-induced increase of AMH/Amh expression. This inhibition occurs through FSH-induced protein kinase A/SF1-mediated expression of gonadotropin inducible ovarian transcription factor 1, a transcriptional repressor, that recruits histone deacetylase 2 to deacetylate H3K27ac, resulting in the suppression of AMH/Amh expression. Furthermore, we report that ovarian Amh mRNA levels are significantly higher in Fshβ-null mice (Fshβ-/-) compared with those in wild-type (WT) mice. In addition, ovarian Amh mRNA levels are restored in Fshβ-null mice expressing a human WT FSHβ transgene (FSHβ-/-hFSHβWT). Our study provides a mechanistic insight into the regulation of AMH expression that has many implications in female reproduction/fertility.
Collapse
Affiliation(s)
- Sambit Roy
- Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, Michigan
| | - Divya Gandra
- Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, Michigan
| | - Christina Seger
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Anindita Biswas
- Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, Michigan
| | | | - Norbert Gleicher
- Center for Human Reproduction, New York, New York
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University, New York, New York
- Department of Obstetrics and Gynecology, Vienna University of Medicine, Vienna, Austria
| | - T Rajendra Kumar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz, Denver, Colorado
| | - Aritro Sen
- Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, Michigan
| |
Collapse
|
36
|
Gleicher N. Expected advances in human fertility treatments and their likely translational consequences. J Transl Med 2018; 16:149. [PMID: 29866181 PMCID: PMC5987489 DOI: 10.1186/s12967-018-1525-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/26/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Due to rapid research progress in reproductive biology and reproductive clinical endocrinology, many human infertility treatments are close to potential breakthroughs and translational applications. We here review current barriers, where such breakthroughs will likely come from, what they will entail, and their potential clinical applications. MAIN TEXT The radical nature of change will primarily benefit older women, reduce fertility treatment costs and thereby expand access to treatment. A still widely overlooked prerequisite for implantation and normal pregnancy maintenance is timely development of maternal immunological tolerance toward an implanting paternal semi-allograft, if malfunctioning associated with implantation failure and pregnancy loss, while premature termination of tolerance appears associated with premature labor, pre-eclampsia/eclampsia and gestoses of pregnancy. Common denominators between pregnancy and invasive malignancies have again been attracting attention, suggesting that, like in malignant tumors, degrees of embryo aneuploidy may affect invasiveness and ability to "disarm" the immune system's innate response against implanting embryos. Linking tolerance to implantation, we offer evidence that the so-called "implantation window" is likely immunological rather than hormonally defined. CONCLUSIONS Because many here outlined treatment changes will disproportionally benefit older women, they will exert a pronounced effect on society, as increasing numbers of women at grandparental ages will become mothers.
Collapse
Affiliation(s)
- Norbert Gleicher
- The CHR, 21 East 69th Street, New York, NY, 10021, USA.
- The Foundation for Reproductive Medicine, New York, NY, 10021, USA.
- Laboratory for Stem Cell Biology and Molecular Embryology, Rockefeller University, New York, NY, 10065, USA.
- Department of Obstetrics and Gynecology, Vienna Medical School, 1090, Vienna, Austria.
| |
Collapse
|