1
|
Zhao ZH, Gu LJ, Zhang XG, Wang ZB, Ou XH, Sun QY. Single-cell and spatial transcriptomes reveal the impact of maternal low protein diet on follicular cell composition and ovarian micro-environment in the offspring. J Nutr Biochem 2025; 136:109789. [PMID: 39490908 DOI: 10.1016/j.jnutbio.2024.109789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/06/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Maternal low protein diet around pregnancy reduces the primordial follicles in offspring ovary. Resolving cellular and molecular mechanisms associated with low protein diet is therefore urgently needed for the guidance of dietary interventions. Here, we utilized single-cell and spatial RNA-seq to create transcriptomic atlases of offspring ovaries from maternal low protein diet mice. Analysis of cell type specific low protein diet associated transcriptional changes revealed increased unfolded protein and decreased oxidative phosphorylation defense as a hallmark of low protein diet effects. Altered pathways included hedgehog signaling in granulosa cells, BMP signaling in theca cells and PTN signaling in early theca cells. Notably, the disordered follicular cell function and ovarian microenvironment may closely corelated with decreased follicular number and quality. Collectively, our findings depict the transcriptomic atlases of the offspring ovary derived from maternal low protein diet group and provide candidate molecular mechanisms underlying the complex ovarian cell changes conferred by low protein diet.
Collapse
Affiliation(s)
- Zheng-Hui Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Lin-Jian Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Guohui Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiang-Hong Ou
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
| |
Collapse
|
2
|
Yu Y, Guo Y, Zhu J, Shen R, Tang J. Chemotherapy drug combinations induced maternal ovarian damage and long-term effect on fetal reproductive system in mice. Eur J Pharm Sci 2024; 201:106860. [PMID: 39043317 DOI: 10.1016/j.ejps.2024.106860] [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: 03/28/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
With the postponement of female reproductive age and the higher incidence of cancer in young people, fertility preservation has become increasingly important in childbearing age. Chemotherapy during pregnancy is crucial for maternal cancer treatments and fetal outcomes. It is a need to further study ovarian damage caused by chemotherapy drug combinations and long-term effects on offspring development, and a detailed understanding of side effects of chemotherapy drugs. In this study, chemotherapy drug combinations significantly impacted on ovarian function, especially epirubicin/cyclophosphamide (EC) combination led to an unbalance in the development of the left and right ovary. Exposure to EC and cisplatin/paclitaxel (TP) increased the number of progenitor follicles while decreased the count of antral follicles and corpora luteum. As to the estrus cycle, EC exposure resulted in a longer estrus period and diestrus period, while TP exposure only extended the diestrus period. EC and TP affected steroid biosynthesis by reducing the expression of SF1 and P450arom.γ-H2AX was detected in both EC and TP exposure groups. As to the impact on the offspring from 4T1 tumor-bearing pregnant mice injected with EC, no significant difference was observed in the physical and neurological development compared to the control, but the ovarian weights, estrus cycles of the offspring were significantly different. Chemotherapy drug combinations exhibit ovarian toxicity, not only causing direct damage on the follicle cells but also disrupting steroid biosynthesis. The reproductive system of offspring from maternal tumor-bearing mice exposed to chemotherapy drugs was observed disorder, but the concrete mechanism still needs further exploration.
Collapse
Affiliation(s)
- Yang Yu
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Yang Guo
- Shanghai Laboratory Animal Research Center, 3577 Road, Pudong District, Shanghai 201203, China
| | - Jialei Zhu
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, 3577 Road, Pudong District, Shanghai 201203, China.
| | - Jing Tang
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200090, China.
| |
Collapse
|
3
|
Mercier A, Johnson J, Kallen AN. Prospective solutions to ovarian reserve damage during the ovarian tissue cryopreservation and transplantation procedure. Fertil Steril 2024; 122:565-573. [PMID: 39181229 DOI: 10.1016/j.fertnstert.2024.08.330] [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: 07/20/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Birth rates continue to decline as more women experience fertility issues. Assisted reproductive technologies are available for patients seeking fertility treatment, including cryopreservation techniques. Cryopreservation can be performed on gametes, embryos, or gonadal tissue and can be used for patients who desire to delay in vitro fertilization treatment. This review focuses on ovarian tissue cryopreservation, the freezing of ovarian cortex containing immature follicles. Ovarian tissue cryopreservation is the only available treatment for the restoration of ovarian function in patients who undergo gonadotoxic treatments, and its wide adoption has led to its recent designation as "no longer experimental" by the American Society for Reproductive Medicine. Ovarian tissue cryopreservation and subsequent transplantation can restore native endocrine function and can support the possibility of pregnancy and live birth for the patient. Importantly, there are multiple steps in the procedure that put the ovarian reserve at risk of damage. The graft is highly susceptible to ischemic reperfusion injury and mass primordial follicle growth activation, resulting in a "burnout" phenomenon. In this review, we summarize current efforts to combat the loss of primordial follicles in grafts through improvements in freeze and thaw protocols, transplantation techniques, and pharmacologic adjuvant treatments. We conducted a review of the literature, with emphasis on emergent research in the last 5 years. Regarding freeze and thaw protocols, we discuss the widely accepted slow freezing approach and newer vitrification protocols. Discussion of improved transplantation techniques includes consideration of the transplantation location of the ovarian tissue and the importance of graft sites in promoting neovascularization. Finally, we discuss pharmacologic treatments being studied to improve tissue performance postgraft. Of note, there is significant research into the efficacy of adjuvants used to reduce ischemic injury, improve neovascularization, and inhibit hyperactivation of primordial follicle growth activations. Although the "experimental" label has been removed from ovarian tissue cryopreservation and subsequent transplantation, there is a significant need for further research to better understand sources of ovarian reserve damage to improve outcomes. Future research directions are provided as we consider how to reach the most hopeful results for women globally.
Collapse
Affiliation(s)
- Abigail Mercier
- Divisions of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Vermont Larner College of Medicine, Burlington, Vermont
| | - Joshua Johnson
- Divisions of Reproductive Endocrinology and Infertility and Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Amanda N Kallen
- Divisions of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Vermont Larner College of Medicine, Burlington, Vermont.
| |
Collapse
|
4
|
Madsen JF, Ernst EH, Amoushahi M, Dueholm M, Ernst E, Lykke-Hartmann K. Dorsomorphin inhibits AMPK, upregulates Wnt and Foxo genes and promotes the activation of dormant follicles. Commun Biol 2024; 7:747. [PMID: 38902324 PMCID: PMC11190264 DOI: 10.1038/s42003-024-06418-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
AMPK is a well-known energy sensor regulating cellular metabolism. Metabolic disorders such as obesity and diabetes are considered detrimental factors that reduce fecundity. Here, we show that pharmacologically induced in vitro activation (by metformin) or inhibition (by dorsomorphin) of the AMPK pathway inhibits or promotes activation of ovarian primordial follicles in cultured murine ovaries and human ovarian cortical chips. In mice, activation of primordial follicles in dorsomorphin in vitro-treated ovaries reduces AMPK activation and upregulates Wnt and FOXO genes, which, interestingly, is associated with decreased phosphorylation of β-catenin. The dorsomorphin-treated ovaries remain of high quality, with no detectable difference in reactive oxygen species production, apoptosis or mitochondrial cytochrome c oxidase activity, suggesting safe activation. Subsequent maturation of in vitro-treated follicles, using a 3D alginate cell culture system, results in mature metaphase eggs with protruding polar bodies. These findings demonstrate that the AMPK pathway can safely regulate primordial follicles by modulating Wnt and FOXO genes, and reduce β-catenin phosphorylation.
Collapse
Affiliation(s)
- Julie Feld Madsen
- Department of Biomedicine, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Emil Hagen Ernst
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, DK-8000, Aarhus C, Denmark
- Department of Gynaecology and Obstetrics, Gødstrup Hospital, DK-7400, Herning, Denmark
| | | | - Margit Dueholm
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, DK-8000, Aarhus C, Denmark
| | - Erik Ernst
- Department of Biomedicine, Aarhus University, DK-8000, Aarhus C, Denmark
- Fertility Clinic Regional Hospital Horsens, DK-8700, Horsens, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, DK-8000, Aarhus C, Denmark.
- Department of Clinical Genetics, Aarhus University Hospital, DK-8200, Aarhus N, Denmark.
| |
Collapse
|
5
|
Ñaupas LVS, Gomes FDR, Ferreira ACA, Morais SM, Alves DR, Teixeira DIA, Alves BG, Watanabe Y, Figueiredo JR, Tetaping GM, Rodrigues APR. Alpha lipoic acid controls degeneration and ensures follicular development in ovine ovarian tissue cultured in vitro. Theriogenology 2024; 225:55-66. [PMID: 38795511 DOI: 10.1016/j.theriogenology.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/28/2024]
Abstract
This study aims to evaluate the effects of adding alpha lipoic acid (ALA) to the in vitro ovarian tissue culture medium, either fresh or after vitrification/warming. For this purpose, 10 ovaries from five adult sheep were used. Each pair of ovaries gave rise to 16 fragments and were randomly distributed into two groups: fresh (n = 8) and vitrified (n = 8). Two fresh fragments were fixed immediately and considered the control, while another six were cultured in vitro for 14 days in the absence; presence of a constant (100 μM/0-14 day) or dynamic (50 μM/day 0-7 and 100 μM/day 8-14) concentration of ALA. As for the vitrified fragments, two were fixed and the other six were cultured in vitro under the same conditions described for the fresh group. All the fragments were subjected to morphological evaluation, follicular development and stromal density (classical histology), DNA fragmentation (TUNEL), senescence (Sudan Black), fibrosis (Masson's Trichome), and endoplasmic reticulum stress (immunofluorescence). Measurements of the antioxidant capacity against the free radicals 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and 2,2-diphenyl-1-picryl-hydrazyl-hydrate (DPPH) and estradiol (E2) levels in the culture medium was performed. The results showed that in the absence of ALA, in vitro culture of vitrified ovarian fragments showed a significant reduction (P < 0.05) in follicular morphology and increased the presence of senescence and tissue fibrosis (P < 0.05). Dynamic ALA maintained E2 levels unchanged (P > 0.05) until the end of vitrified ovarian tissue culture and controlled the levels of ABTS and DPPH radicals in fresh or vitrified cultures. Therefore, it is concluded that ALA should be added to the vitrified ovarian tissue in vitro culture medium to reduce the damage that leads to loss of ovarian function. To ensure steroidogenesis during in vitro culture, ALA should be added dynamically (different concentrations throughout culture).
Collapse
Affiliation(s)
- L V S Ñaupas
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - F D R Gomes
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A C A Ferreira
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - S M Morais
- Laboratory of Natural Products Chemistry, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - D R Alves
- Laboratory of Natural Products Chemistry, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - D I A Teixeira
- Laboratory of Image Diagnosis Applied to Animal Reproduction, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, E, Brazil
| | - B G Alves
- Ovid Research Company, Berkeley, CA, United States
| | - Y Watanabe
- Vitrogen YVF Biotech, Cravinhos, SP, Brazil
| | - J R Figueiredo
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - G M Tetaping
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A P R Rodrigues
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
6
|
Ren P, Tong X, Li J, Jiang H, Liu S, Li X, Lai M, Yang W, Rong Y, Zhang Y, Jin J, Ma Y, Pan W, Fan HY, Zhang S, Zhang YL. CRL4 DCAF13 E3 ubiquitin ligase targets MeCP2 for degradation to prevent DNA hypermethylation and ensure normal transcription in growing oocytes. Cell Mol Life Sci 2024; 81:165. [PMID: 38578457 PMCID: PMC10997554 DOI: 10.1007/s00018-024-05185-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
The DNA methylation is gradually acquired during oogenesis, a process sustained by successful follicle development. However, the functional roles of methyl-CpG-binding protein 2 (MeCP2), an epigenetic regulator displaying specifical binding with methylated DNA, remains unknown in oogenesis. In this study, we found MeCP2 protein was highly expressed in primordial and primary follicle, but was almost undetectable in secondary follicles. However, in aged ovary, MeCP2 protein is significantly increased in both oocyte and granulosa cells. Overexpression of MeCP2 in growing oocyte caused transcription dysregulation, DNA hypermethylation, and genome instability, ultimately leading to follicle growth arrest and apoptosis. MeCP2 is targeted by DCAF13, a substrate recognition adaptor of the Cullin 4-RING (CRL4) E3 ligase, and polyubiquitinated for degradation in both cells and oocytes. Dcaf13-null oocyte exhibited an accumulation of MeCP2 protein, and the partial rescue of follicle growth arrest induced by Dcaf13 deletion was observed following MeCP2 knockdown. The RNA-seq results revealed that large amounts of genes were regulated by the DCAF13-MeCP2 axis in growing oocytes. Our study demonstrated that CRL4DCAF13 E3 ubiquitin ligase targets MeCP2 for degradation to ensure normal DNA methylome and transcription in growing oocytes. Moreover, in aged ovarian follicles, deceased DCAF13 and DDB1 protein were observed, indicating a potential novel mechanism that regulates ovary aging.
Collapse
Affiliation(s)
- Peipei Ren
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Junjian Li
- College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Huifang Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Siya Liu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiang Li
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Mengru Lai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yan Rong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yingyi Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Jiamin Jin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yerong Ma
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weiwei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Heng-Yu Fan
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China.
| | - Yin-Li Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
7
|
Kashi O, Meirow D. Overactivation or Apoptosis: Which Mechanisms Affect Chemotherapy-Induced Ovarian Reserve Depletion? Int J Mol Sci 2023; 24:16291. [PMID: 38003481 PMCID: PMC10671775 DOI: 10.3390/ijms242216291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Dormant primordial follicles (PMF), which constitute the ovarian reserve, are recruited continuously into the cohort of growing follicles in the ovary throughout female reproductive life. Gonadotoxic chemotherapy was shown to diminish the ovarian reserve pool, to destroy growing follicle population, and to cause premature ovarian insufficiency (POI). Three primary mechanisms have been proposed to account for this chemotherapy-induced PMF depletion: either indirectly via over-recruitment of PMF, by stromal damage, or through direct toxicity effects on PMF. Preventative pharmacological agents intervening in these ovotoxic mechanisms may be ideal candidates for fertility preservation (FP). This manuscript reviews the mechanisms that disrupt follicle dormancy causing depletion of the ovarian reserve. It describes the most widely studied experimental inhibitors that have been deployed in attempts to counteract these affects and prevent follicle depletion.
Collapse
Affiliation(s)
- Oren Kashi
- The Morris Kahn Fertility Preservation Center, Sheba Medical Center, Ramat Gan 5262000, Israel;
| | - Dror Meirow
- The Morris Kahn Fertility Preservation Center, Sheba Medical Center, Ramat Gan 5262000, Israel;
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
8
|
Lopez J, Hohensee G, Liang J, Sela M, Johnson J, Kallen AN. The Aging Ovary and the Tales Learned Since Fetal Development. Sex Dev 2023; 17:156-168. [PMID: 37598664 PMCID: PMC10841896 DOI: 10.1159/000532072] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND While the term "aging" implies a process typically associated with later life, the consequences of ovarian aging are evident by the time a woman reaches her forties, and sometimes earlier. This is due to a gradual decline in the quantity and quality of oocytes which occurs over a woman's reproductive lifespan. Indeed, the reproductive potential of the ovary is established even before birth, as the proper formation and assembly of the ovarian germ cell population during fetal life determines the lifetime endowment of oocytes and follicles. In the ovary, sophisticated molecular processes have been identified that regulate the timing of ovarian aging and these are critical to ensuring follicular maintenance. SUMMARY The mechanisms thought to contribute to overall aging have been summarized under the term the "hallmarks of aging" and include such processes as DNA damage, mitochondrial dysfunction, telomere attrition, genomic instability, and stem cell exhaustion, among others. Similarly, in the ovary, molecular processes have been identified that regulate the timing of ovarian aging and these are critical to ensuring follicular maintenance. In this review, we outline critical processes involved in ovarian aging, highlight major achievements for treatment of ovarian aging, and discuss ongoing questions and areas of debate. KEY MESSAGES Ovarian aging is recognized as what may be a complex process in which age, genetics, environment, and many other factors contribute to the size and depletion of the follicle pool. The putative hallmarks of reproductive aging outlined herein include a diversity of plausible processes contributing to the depletion of the ovarian reserve. More research is needed to clarify if and to what extent these putative regulators do in fact govern follicle and oocyte behavior, and how these signals might be integrated in order to control the overall pattern of ovarian aging.
Collapse
Affiliation(s)
- Jesus Lopez
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Gabe Hohensee
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Jing Liang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Meirav Sela
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Joshua Johnson
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, CO, USA
| | - Amanda N. Kallen
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
9
|
Peng Y, Guo R, Shi B, Li D. The role of long non-coding RNA H19 in infertility. Cell Death Discov 2023; 9:268. [PMID: 37507391 PMCID: PMC10382492 DOI: 10.1038/s41420-023-01567-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Infertility is defined as the failure to conceive after at least one year of unprotected intercourse. Long non-coding RNAs (lncRNAs) are transcripts that contain more than 200 nucleotides but do not convert into proteins. LncRNAs, particularly lncRNA H19, have been linked to the emergence and progression of various diseases. This review focuses on the role of H19 in infertility caused by polycystic ovary syndrome, endometriosis, uterine fibroids, diminished ovarian reserve, male factor, and assisted reproductive technology-related pathology, highlighting the potential of H19 as a molecular target for the future treatment of infertility.
Collapse
Affiliation(s)
- Yuanyuan Peng
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China
| | - Renhao Guo
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Bei Shi
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
| |
Collapse
|
10
|
Sills ES, Harrity C, Wood SH, Tan SL. mTOR Inhibition via Low-Dose, Pulsed Rapamycin with Intraovarian Condensed Platelet Cytokines: An Individualized Protocol to Recover Diminished Reserve? J Pers Med 2023; 13:1147. [PMID: 37511761 PMCID: PMC10381109 DOI: 10.3390/jpm13071147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/03/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
No major breakthroughs have entered mainstream clinical fertility practice since egg donation and intracytoplasmic sperm injection decades ago, and oocyte deficits secondary to advanced age continue as the main manifestation of diminished ovarian reserve. In the meantime, several unproven IVF 'accessories' have emerged including so-called ovarian rejuvenation which entails placing fresh autologous platelet-rich plasma (PRP) directly into ovarian tissue. Among cellular responses attributed to this intervention are reduced oxidative stress, slowed apoptosis and improved metabolism. Besides having an impact on the existing follicle pool, platelet growth factors might also facilitate de novo oocyte recruitment by specified gene upregulation targeting uncommitted ovarian stem cells. Given that disordered activity at the mechanistic target of rapamycin (mTOR) has been shown to exacerbate or accelerate ovarian aging, PRP-discharged plasma cytokines combined with mTOR suppression by pulsed/cyclic rapamycin represents a novel fusion technique to enhance ovarian function. While beneficial effects have already been observed experimentally in oocytes and embryos with mTOR inhibition alone, this proposal is the first to discuss intraovarian platelet cytokines followed by low-dose, phased rapamycin. For refractory cases, this investigational, tailored approach could amplify or sustain ovarian capacity sufficient to permit retrieval of competent oocytes via distinct but complementary pathways-thus reducing dependency on oocyte donation.
Collapse
Affiliation(s)
- E Scott Sills
- Plasma Research Section, Regenerative Biology Group/CAG, San Clemente, CA 92673, USA
- Department of Obstetrics & Gynecology, Palomar Medical Center, Escondido, CA 92029, USA
| | - Conor Harrity
- Department of Obstetrics & Gynaecology, Royal College of Surgeons in Ireland, D02 HC66 Dublin, Ireland
| | - Samuel H Wood
- Department of Obstetrics & Gynecology, Palomar Medical Center, Escondido, CA 92029, USA
- Gen 5 Fertility Center, San Diego, CA 92121, USA
| | - Seang Lin Tan
- OriginElle Fertility Clinic, Montreal, QC H4A 3J3, Canada
- Department of Obstetrics & Gynecology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
11
|
Xia L, Shen Y, Liu S, Du J. Iron overload triggering ECM-mediated Hippo/YAP pathway in follicle development: a hypothetical model endowed with therapeutic implications. Front Endocrinol (Lausanne) 2023; 14:1174817. [PMID: 37223010 PMCID: PMC10200985 DOI: 10.3389/fendo.2023.1174817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/12/2023] [Indexed: 05/25/2023] Open
Abstract
Disruption of iron homeostasis plays a negative role in follicle development. The dynamic changes in follicle growth are dependent on Hippo/YAP signaling and mechanical forces. However, little is known about the liaison between iron overload and the Hippo/YAP signalling pathway in term of folliculogenesis. Here, based on the available evidence, we established a hypothesized model linking excessive iron, extracellular matrix (ECM), transforming growth factor-β (TGF-β) and Hippo/Yes-associated protein (YAP) signal regarding follicle development. Hypothetically, the TGF-β signal and iron overload may play a synergistic role in ECM production via YAP. We speculate that the dynamic homeostasis of follicular iron interacts with YAP, increasing the risk of ovarian reserve loss and may enhance the sensitivity of follicles to accumulated iron. Hence, therapeutic interventions targeting iron metabolism disorders, and Hippo/YAP signal may alter the consequences of the impaired developmental process based on our hypothesis, which provides potential targets and inspiration for further drug discovery and development applied to clinical treatment.
Collapse
Affiliation(s)
- Lingjin Xia
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Yupei Shen
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Du
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Nakamura N, Yoshida N, Suwa T. Three major reasons why transgenerational effects of radiation are difficult to detect in humans. Int J Radiat Biol 2023; 100:1297-1311. [PMID: 36880868 DOI: 10.1080/09553002.2023.2187478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/18/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE Ionizing radiation can induce mutations in germ cells in various organisms, including fruit flies and mice. However, currently, there is no clear evidence for the transgenerational effects of radiation in humans. This review is an effort to identify possible reasons for the lack of such observations. METHODS Literature search and narrative review. RESULTS 1) In both mice and humans, resting oocytes locate primarily in the cortical region of the ovary where the number of blood vessels is very low especially when young and extra-cellular material is rich, and this region is consequently hypoxic, which probably leads to immature oocytes being resistant to the cell killing and mutagenic effects of radiation. 2) In studies of spermatogonia, the mouse genes used for specific locus test (SLT) studies, which include coat color genes, were hypermutable when compared to many other genes. Recent studies which examined over 1000 segments of genomic DNA indicate that the induction rate of deletion mutation per segment was on the order of 10-6 per Gy, which is one order of magnitude lower than that obtained from the SLT data. Therefore, it appears possible that detecting any transgenerational effects of radiation following human male exposures will be difficult due to a lack of mutable marker genes. 3) Fetal malformations were examined in studies in humans, but the genetic component in such malformations is low, and abnormal fetuses are prone to undergo miscarriage which does not occur in mice, and which leads to difficulties in detecting transgenerational effects. CONCLUSION The lack of clear evidence for radiation effects in humans probably does not result from any problem in the methodologies used but may be due largely to biological properties. Currently, whole genome sequencing studies of exposed parents and offspring are planned, but ethical guidelines need to be followed to avoid discrimination, which had once happened to the atomic bomb survivors.
Collapse
Affiliation(s)
- Nori Nakamura
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Noriaki Yoshida
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Tatsuya Suwa
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
13
|
Portillo AM, Varela E, García-Velasco JA. Influence of telomerase activity and initial distribution on human follicular aging: Moving from a discrete to a continuum model. Math Biosci 2023; 358:108985. [PMID: 36828232 DOI: 10.1016/j.mbs.2023.108985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023]
Abstract
A discrete model is proposed for the temporal evolution of a population of cells sorted according to their telomeric length. This model assumes that, during cell division, the distribution of the genetic material to daughter cells is asymmetric, i.e. chromosomes of one daughter cell have the same telomere length as the mother, while in the other daughter cell telomeres are shorter. Telomerase activity and cell death are also taken into account. The continuous model is derived from the discrete model by introducing the generational age as a continuous variable in [0,h], being h the Hayflick limit, i.e. the number of times that a cell can divide before reaching the senescent state. A partial differential equation with boundary conditions is obtained. The solution to this equation depends on the initial telomere length distribution. The initial and boundary value problem is solved exactly when the initial distribution is of exponential type. For other types of initial distributions, a numerical solution is proposed. The model is applied to the human follicular growth from preantral to preovulatory follicle as a case study and the aging rate is studied as a function of telomerase activity, the initial distribution and the Hayflick limit. Young, middle and old cell-aged initial normal distributions are considered. In all cases, when telomerase activity decreases, the population ages and the smaller the h value, the higher the aging rate becomes. However, the influence of these two parameters is different depending on the initial distribution. In conclusion, the worst-case scenario corresponds to an aged initial telomere distribution.
Collapse
Affiliation(s)
- A M Portillo
- Instituto de Investigación en Matemáticas de la Universidad de Valladolid, Valladolid, Spain; Departamento de Matemática Aplicada, Escuela de Ingenierías Industriales, Universidad de Valladolid, Pso. Prado de la Magdalena 3-5, Valladolid, 47011, Spain.
| | - E Varela
- IVI Foundation, The Health Research Institute La Fe (IIS La Fe), Edificio Biopolo. Av. Fernando Abril Martorell, 106 - Torre A, Planta 1, Valencia, 46026, Spain; Rey Juan Carlos University, Edificio Departamental II. Av. de Atenas, s/n, Alcorcón, Madrid, 28922, Spain.
| | - J A García-Velasco
- IVI Foundation, The Health Research Institute La Fe (IIS La Fe), Edificio Biopolo. Av. Fernando Abril Martorell, 106 - Torre A, Planta 1, Valencia, 46026, Spain; IVIRMA Madrid, Av. del Talgo, 68, Madrid, 28023, Spain; Rey Juan Carlos University, Edificio Departamental II. Av. de Atenas, s/n, Alcorcón, Madrid, 28922, Spain.
| |
Collapse
|
14
|
Zhang T, He M, Zhang J, Tong Y, Chen T, Wang C, Pan W, Xiao Z. Mechanisms of primordial follicle activation and new pregnancy opportunity for premature ovarian failure patients. Front Physiol 2023; 14:1113684. [PMID: 36926197 PMCID: PMC10011087 DOI: 10.3389/fphys.2023.1113684] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Primordial follicles are the starting point of follicular development and the basic functional unit of female reproduction. Primordial follicles are formed around birth, and most of the primordial follicles then enter a dormant state. Since primordial follicles are limited in number and can't be renewed, dormant primordial follicles cannot be reversed once they enter the growing state. Thus, the orderly occurrence of primordial follicles selective activation directly affects the rate of follicle consumption and thus determines the length of female reproductive lifespan. Studies have found that appropriately inhibiting the activation rate of primordial follicles can effectively slow down the rate of follicle consumption, maintain fertility and delay ovarian aging. Based on the known mechanisms of primordial follicle activation, primordial follicle in vitro activation (IVA) technique has been clinically developed. IVA can help patients with premature ovarian failure, middle-aged infertile women, or infertile women due to gynecological surgery treatment to solve infertility problems. The study of the mechanism of selective activation of primordial follicles can contribute to the development of more efficient and safe IVA techniques. In this paper, recent mechanisms of primordial follicle activation and its clinical application are reviewed.
Collapse
Affiliation(s)
- Tuo Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Meina He
- College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jingjing Zhang
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yuntong Tong
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tengxiang Chen
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wei Pan
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ziwen Xiao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
15
|
Albertini DF. Factoring attrition into natural and ART-based human fecundity. J Assist Reprod Genet 2022; 39:1967-1968. [PMID: 36089626 PMCID: PMC9474985 DOI: 10.1007/s10815-022-02615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
16
|
Xue L, Li X, Zhu X, Zhang J, Zhou S, Tang W, Chen D, Chen Y, Dai J, Wu M, Wu M, Wang S. Carbon tetrachloride exposure induces ovarian damage through oxidative stress and inflammatory mediated ovarian fibrosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113859. [PMID: 35816842 DOI: 10.1016/j.ecoenv.2022.113859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/02/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
Carbon tetrachloride (CCL4) is widely used as a chemical intermediate and as a feedstock in the production of chlorofluorocarbons. CCL4 is highly toxic in the liver, kidney, testicle, brain and other tissues. However, the effect of CCL4 on ovarian function has not been reported. In this study, we found that the mice treated with CCL4 showed decreased ovarian function with disturbed estrus cycle, decreased serum level of 17β-estradiol and the reduced number of healthy follicles. Ovarian damage was accompanied by oxidative stress and the production of proinflammatory cytokines, especially interleukins. The indicators of oxidative stress, 4-Hydroxynonenal (4-HNE), 8-hydroxy-2´-deoxyguanosine (8-OHdG), 3-Nitrotyrosine (3-NT) and malondialdehyde (MDA), and the levels of proinflammatory cytokines IL-1α, IL-1β, IL-6 and IL-11 were increased, while the antioxidants, including superoxide dismutase (SOD), nuclear factor erythroid2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1), were decreased in the CCL4 group. In the CCL4 treated group, the results of Sirius Red staining, immunohistochemistry and qPCR indicated that proinflammatory cytokines caused further ovarian fibrosis. And CCL4 could also promote ovarian thecal cells to secrete inflammatory cytokines, resulting in fibrosis in vitro. In addition, CCL4 inhibited oocyte development and triggered oocyte apoptosis. In conclusion, CCL4 exposure causes ovarian damage by strong oxidative stress and the high expression of the proinflammatory cytokine mediated ovarian fibrosis.
Collapse
Affiliation(s)
- Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Xiang Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China; Department of Obstetrics and Gynecology, Xiehe Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Yingying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| | - Mingfu Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, 430030 Wuhan, Hubei, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, 430030 Wuhan, Hubei, China.
| |
Collapse
|
17
|
Johnson J, Emerson JW, Lawley SD. Recapitulating human ovarian aging using random walks. PeerJ 2022; 10:e13941. [PMID: 36032944 PMCID: PMC9406804 DOI: 10.7717/peerj.13941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/02/2022] [Indexed: 01/19/2023] Open
Abstract
Mechanism(s) that control whether individual human primordial ovarian follicles (PFs) remain dormant, or begin to grow, are all but unknown. One of our groups has recently shown that activation of the Integrated Stress Response (ISR) pathway can slow follicular granulosa cell proliferation by activating cell cycle checkpoints. Those data suggest that the ISR is active and fluctuates according to local conditions in dormant PFs. Because cell cycle entry of (pre)granulosa cells is required for PF growth activation (PFGA), we propose that rare ISR checkpoint resolution allows individual PFs to begin to grow. Fluctuating ISR activity within individual PFs can be described by a random process. In this article, we model ISR activity of individual PFs by one-dimensional random walks (RWs) and monitor the rate at which simulated checkpoint resolution and thus PFGA threshold crossing occurs. We show that the simultaneous recapitulation of (i) the loss of PFs over time within simulated subjects, and (ii) the timing of PF depletion in populations of simulated subjects equivalent to the distribution of the human age of natural menopause can be produced using this approach. In the RW model, the probability that individual PFs grow is influenced by regionally fluctuating conditions, that over time manifests in the known pattern of PFGA. Considered at the level of the ovary, randomness appears to be a key, purposeful feature of human ovarian aging.
Collapse
Affiliation(s)
- Joshua Johnson
- Department of Obstetrics and Gynecology, University of Colorado-Anschutz Medical Center, Aurora, Colorado, United States
| | - John W. Emerson
- Department of Statistics and Data Science, Yale University, New Haven, Connecticut, United States
| | - Sean D. Lawley
- Department of Mathematics, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
18
|
Huang YS, Lin CY. Stimulatory Effects of Androgens on Eel Primary Ovarian Development - from Phenotypes to Genotypes. Vet Med Sci 2022. [DOI: 10.5772/intechopen.99582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Androgens stimulate primary ovarian development in Vertebrate. Japanese eels underwent operation to sample the pre- and post-treated ovarian tissues from the same individual. Ovarian phenotypic or genotypic data were mined in a pair. A correlation between the initial ovarian status (determined by kernel density estimation (KDE), presented as a probability density of oocyte size) and the consequence of androgen (17MT) treatment (change in ovary) has been showed. The initial ovarian status appeared to be important to influence ovarian androgenic sensitivity. The initial ovary was important to the outcomes of androgen treatments, and ePAV (expression presence-absence variation) is existing in Japanese eel by analyze DEGs; core, unique, or accessory genes were identified, the sensitivities of initial ovaries were correlated with their gene expression profiles. We speculated the importance of genetic differential expression on the variations of phenotypes by 17MT, and transcriptomic approach seems to allow extracting multiple layers of genomic data.
Collapse
|
19
|
Roberts JF, Jeff Huang CC. Bovine models for human ovarian diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:101-154. [PMID: 35595347 DOI: 10.1016/bs.pmbts.2022.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
During early embryonic development, late fetal growth, puberty, adult reproductive years, and advanced aging, bovine and human ovaries closely share molecular pathways and hormonal signaling mechanisms. Other similarities between these species include the size of ovaries, length of gestation, ovarian follicular and luteal dynamics, and pathophysiology of ovarian diseases. As an economically important agriculture species, cattle are a foundational species in fertility research with decades of groundwork using physiologic, genetic, and therapeutic experimental techniques. Many technologies used in modern reproductive medicine, such as ovulation induction using hormonal therapy, were first used in cows before human trials. Human ovarian diseases with naturally occurring bovine correlates include premature ovary insufficiency (POI), polycystic ovarian syndrome (PCOS), and sex-cord stromal tumors (SCSTs). This article presents an overview of bovine ovary research related to causes of infertility, ovarian diseases, diagnostics, and therapeutics, emphasizing where the bovine model can offer advantages over other lab animals for translational applications.
Collapse
Affiliation(s)
- John F Roberts
- Department of Comparative, Diagnostic & Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States.
| | - Chen-Che Jeff Huang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
20
|
Xue W, Xue F, Jia T, Hao A. Research and experimental verification of the molecular mechanism of berberine in improving premature ovarian failure based on network pharmacology. Bioengineered 2022; 13:9885-9900. [PMID: 35420511 PMCID: PMC9161839 DOI: 10.1080/21655979.2022.2062104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Based on the research methods of network pharmacology, this study analyzed the improvement effect of berberine (BBR) on premature ovarian failure (POF) and its molecular mechanism. Carry out GO and KEGG enrichment analysis by R language to obtain the potential targets and pathways of BBR in the improvement of POF. Use SD rats and ovarian granulosa cells (GCs) for experimental verification. ELISA was used to measure the content of related hormones in the serum, CCK-8 was used to measure cell viability, western blot was used to measure the content of the target protein in the ovaries and GCs, and q-RT-PCR was used to detect the expression of the target genes in the ovaries and GCs. Predicted by network pharmacology: PTEN, AKT1, FoxO1, FasL, and Bim are the targets with the highest relative correlation between BBR and POF. The results of experiments show that the treatment of low and medium doses of BBR can increase the ovarian index of rats; BBR can increase the levels of Estradiol (E2) and Anti-Mullerian hormone (AMH) in the serum of rats and reduce the levels of Follicle stimulating hormone (FSH) and Luteinizing hormone (LH). BBR can increase the cell viability of GCs; BBR can inhibit the PTEN/AKT1/FoxO1 signaling pathway and its phosphorylation level and reduce the expression of Fas/FasL and Bim mRNA. Overall, BBR can promote the ovarian to maintain normal hormone levels, protect GCs, and enhance the function of POF.
Collapse
Affiliation(s)
- Wu Xue
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning, China.,Key Laboratory of Follicular Development and Reproductive Health of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Fan Xue
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Tao Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ai Hao
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning, China.,Key Laboratory of Follicular Development and Reproductive Health of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning Province, China.,Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
21
|
Mei Q, Li H, Liu Y, Wang X, Xiang W. Advances in the study of CDC42 in the female reproductive system. J Cell Mol Med 2021; 26:16-24. [PMID: 34859585 PMCID: PMC8742232 DOI: 10.1111/jcmm.17088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
CDC42 is a member of the Rho‐GTPase family and is involved in a variety of cellular functions including regulation of cell cycle progression, constitution of the actin backbone and membrane transport. In particular, CDC42 plays a key role in the establishment of polarity in female vertebrate oocytes, and essential to this major regulatory role is its local occupation of specific regions of the cell to ensure that the contractile ring is assembled at the right time and place to ensure proper gametogenesis. The multifactor controlled ‘inactivation‐activation’ process of CDC42 also allows it to play an important role in the multilevel signalling network, and the synergistic regulation of multiple genes ensures maximum precision during gametogenesis. The purpose of this paper is to review the role of CDC42 in the control of gametogenesis and to explore its related mechanisms, with the aim of further understanding the great research potential of CDC42 in female vertebrate germ cells and its future clinical translation.
Collapse
Affiliation(s)
- Qiaojuan Mei
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiying Li
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Liu
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Wang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenpei Xiang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Llerena Cari E, Hagen-Lillevik S, Giornazi A, Post M, Komar AA, Appiah L, Bitler B, Polotsky AJ, Santoro N, Kieft J, Lai K, Johnson J. Integrated stress response control of granulosa cell translation and proliferation during normal ovarian follicle development. Mol Hum Reprod 2021; 27:gaab050. [PMID: 34314477 PMCID: PMC8660582 DOI: 10.1093/molehr/gaab050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/06/2021] [Indexed: 11/13/2022] Open
Abstract
Mechanisms that directly control mammalian ovarian primordial follicle (PF) growth activation and the selection of individual follicles for survival are largely unknown. Follicle cells produce factors that can act as potent inducers of cellular stress during normal function. Consistent with this, we show here that normal, untreated ovarian cells, including pre-granulosa cells of dormant PFs, express phenotype and protein markers of the activated integrated stress response (ISR), including stress-specific protein translation (phospho-Serine 51 eukaryotic initiation factor 2α; P-EIF2α), active DNA damage checkpoints, and cell-cycle arrest. We further demonstrate that mRNAs upregulated in primary (growing) follicles versus arrested PFs mostly include stress-responsive upstream open reading frames (uORFs). Treatment of a granulosa cell (GC) line with the PF growth trigger tumor necrosis factor alpha results in the upregulation of a 'stress-dependent' translation profile. This includes further elevated P-eIF2α and a shift of uORF-containing mRNAs to polysomes. Because the active ISR corresponds to slow follicle growth and PF arrest, we propose that repair and abrogation of ISR checkpoints (e.g. checkpoint recovery) drives the GC cell cycle and PF growth activation (PFGA). If cellular stress is elevated beyond a threshold(s) or, if damage occurs that cannot be repaired, cell and follicle death ensue, consistent with physiological atresia. These data suggest an intrinsic quality control mechanism for immature and growing follicles, where PFGA and subsequent follicle growth and survival depend causally upon ISR resolution, including DNA repair and thus the proof of genomic integrity.
Collapse
Affiliation(s)
- Evelyn Llerena Cari
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Aurora, CO, USA
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Aurora, CO, USA
| | - Synneva Hagen-Lillevik
- University of Utah School of Medicine, Department of Pediatrics and Department of Nutrition and Integrative Physiology, Salt Lake City, UT, USA
| | | | - Miriam Post
- University of Colorado-Anschutz Medical Campus, Department of Pathology, Aurora, CO, USA
| | - Anton A Komar
- Cleveland State University, Center for Gene Regulation in Health and Disease (GRHD), Cleveland, OH, USA
| | - Leslie Appiah
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Academic Specialists in Obstetrics and Gynecology, Aurora, CO, USA
| | - Benjamin Bitler
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Aurora, CO, USA
| | - Alex J Polotsky
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Aurora, CO, USA
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Aurora, CO, USA
| | - Nanette Santoro
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Aurora, CO, USA
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Aurora, CO, USA
| | - Jeffrey Kieft
- University of Colorado-Anschutz Medical Campus, Department of Biochemistry and Molecular Genetics, Aurora, CO, USA
| | - Kent Lai
- University of Utah School of Medicine, Department of Pediatrics and Department of Nutrition and Integrative Physiology, Salt Lake City, UT, USA
| | - Joshua Johnson
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Aurora, CO, USA
- University of Colorado-Anschutz Medical Campus, Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Aurora, CO, USA
| |
Collapse
|
23
|
Lins TLBG, Barberino RS, Monte APO, Pinto JGC, Campinho DSP, Palheta RC, Matos MHT. Rutin promotes activation and reduces apoptosis of primordial follicles by regulating Akt phosphorylation after in vitro culture of ovine ovarian tissue. Theriogenology 2021; 173:64-72. [PMID: 34339905 DOI: 10.1016/j.theriogenology.2021.07.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/09/2021] [Accepted: 07/25/2021] [Indexed: 12/22/2022]
Abstract
The aims of this study were to analyze the effects of different concentrations of rutin on primordial follicle survival and development after in vitro culture of sheep ovarian tissue, and to verify the possible involvement of the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) pathway in the rutin actions. Ovarian fragments were fixed for histological analysis (fresh control) or cultured in α-minimum essential medium alone (α-MEM+: control medium) or in α-MEM+supplemented with different concentrations of rutin (0.1; 1 or 10 μg/mL) for 7 days. Inhibition of the PI3K activity was performed in fragments cultured with 50 μM LY294002. Thereafter, immunohistochemistry was performed to evaluate the expression of cleaved caspase-3 (apoptosis) and Akt phosphorylation (p-Akt). The results showed that 1 μg/mL rutin has a greater percentage of normal follicles (P < 0.05) than those of α-MEM+ and other rutin treatments. In addition, 1 μg/mL rutin maintained the follicular apoptosis similar (P > 0.05) to that of the fresh control and lower than α-MEM+ and 10 μg/mL rutin. All rutin concentrations increased (P < 0.05) follicular activation compared to fresh control and α-MEM+. Furthermore, follicular and oocyte diameters increased (P < 0.05) only after culture with 1 μg/mL rutin. After PI3K inhibition, there was a reduction (P < 0.05) of rutin follicular effects. In conclusion, rutin at 1 μg/mL reduces apoptosis, promotes activation and growth of sheep primordial follicles through the modulation of the PI3K/Akt signaling pathway after in vitro culture of ovine ovarian tissue.
Collapse
Affiliation(s)
- T L B G Lins
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, Brazil
| | - R S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, Brazil
| | - A P O Monte
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, Brazil
| | - J G C Pinto
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, Brazil
| | - D S P Campinho
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, Brazil
| | - R C Palheta
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Federal University of São Francisco Valley, Petrolina, PE, Brazil
| | - M H T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE, Brazil.
| |
Collapse
|
24
|
Pors SE, Harðardóttir L, Olesen HØ, Riis ML, Jensen LB, Andersen AS, Cadenas J, Grønning AP, Colmorn LB, Dueholm M, Andersen CY, Kristensen SG. Effect of sphingosine-1-phosphate on activation of dormant follicles in murine and human ovarian tissue. Mol Hum Reprod 2021; 26:301-311. [PMID: 32202615 DOI: 10.1093/molehr/gaaa022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 02/09/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022] Open
Abstract
In vitro activation of resting ovarian follicles, with the use of mechanical stress and/or pharmacological compounds, is an emerging and novel approach for infertility treatment. The aim of this study was to assess the sphingolipid, sphingosine-1-phosphate (S1P), as a potential in vitro activation agent in murine and human ovarian tissues and isolated follicles. Juvenile murine ovaries and donated human ovarian tissues, from 10 women undergoing ovarian tissue cryopreservation for fertility preservation, were incubated with or without 12 μM S1P for 3 h for quantitative PCR analysis, and 12 h for xenotransplantation or culture studies. Gene expression analyses were performed for genes downstream of the Hippo signaling pathway. Murine ovaries and isolated murine and human preantral follicles showed significantly increased mRNA expression levels of Ccn2/CCN2 following S1P treatment compared to controls. This increase was shown to be specific for the Hippo signaling pathway and for the S1P2 receptor, as co-treatment with Hippo-inhibitor, verteporfin and S1PR2 antagonist, JTE-013, reduced the S1P-induced Ccn2 gene expression in murine ovaries. Histological evaluation of human cortical tissues (5 × 5 × 1 mm; n = 30; three pieces per patient) xenografted for 6 weeks and juvenile murine ovaries cultured for 4 days (n = 9) or allografted for 2 weeks (n = 48) showed no differences in the distribution of resting or growing follicles in S1P-treated ovarian tissues compared to controls. Collectively, S1P increased Ccn2/CCN2 gene expression in isolated preantral follicles and ovarian tissue from mice and human, but it did not promote follicle activation or growth in vivo. Thus, S1P does not appear to be a potent in vitro activation agent under these experimental conditions.
Collapse
Affiliation(s)
- Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Lilja Harðardóttir
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark.,Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark.,Current workplace: Center for Obstetrics and Pediatrics, Department of Obstetrics and Fetal Medicine, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Hanna Ørnes Olesen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Malene Lundgaard Riis
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Lea Bejstrup Jensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Astrid Sten Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark.,Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Jesús Cadenas
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Annika Patricia Grønning
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark.,Department of Technology, Faculty of Health, University College Copenhagen, Copenhagen, Denmark
| | - Lotte Berdiin Colmorn
- The Fertility Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Margit Dueholm
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark.,Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Frost ER, Ford EA, Peters AE, Reed NL, McLaughlin EA, Baker MA, Lovell-Badge R, Sutherland JM. Signal transducer and activator of transcription (STAT) 1 and STAT3 are expressed in the human ovary and have Janus kinase 1-independent functions in the COV434 human granulosa cell line. Reprod Fertil Dev 2021; 32:1027-1039. [PMID: 32758351 DOI: 10.1071/rd20098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/29/2020] [Indexed: 01/03/2023] Open
Abstract
Ovarian granulosa cells are fundamental for oocyte maintenance and maturation. Recent studies have demonstrated the importance of members of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling pathway in the granulosa cell population of mouse and horse ovaries, with perturbation of JAK1 signalling in the mouse shown to impair oocyte maintenance and accelerate primordial follicle activation. The presence and role of the JAK/STAT pathway in human granulosa cells has yet to be elucidated. In this study, expression of JAK1, STAT1 and STAT3 was detected in oocytes and granulosa cells of human ovarian sections from fetal (40 weeks gestation) and premenopausal ovaries (34-41 years of age; n=3). To determine the effects of JAK1 signalling in granulosa cells, the human granulosa-like cell line COV434 was used, with JAK1 inhibition using ruxolitinib. Chemical inhibition of JAK1 in COV434 cells with 100nM ruxolitinib for 72h resulted in significant increases in STAT3 mRNA (P=0.034) and p-Y701-STAT1 protein (P=0.0117), demonstrating a role for JAK1 in modulating STAT in granulosa cells. This study implicates a conserved role for JAK/STAT signalling in human ovary development, warranting further investigation of this pathway in human granulosa cell function.
Collapse
Affiliation(s)
- E R Frost
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; and Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; and Corresponding author.
| | - E A Ford
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - A E Peters
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - N L Reed
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - E A McLaughlin
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and School of Science, Western Sydney University, Penrith, NSW 2751, Australia; and School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand
| | - M A Baker
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - R Lovell-Badge
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - J M Sutherland
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
26
|
Huang YS, Cheng WC, Lin CY. Androgenic Sensitivities and Ovarian Gene Expression Profiles Prior to Treatment in Japanese Eel (Anguilla japonica). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:430-444. [PMID: 34191211 DOI: 10.1007/s10126-021-10035-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/28/2021] [Indexed: 06/13/2023]
Abstract
Androgens stimulate ovarian development in eels. Our previous report indicated a correlation between the initial (debut) ovarian status (determined by kernel density estimation (KDE), presented as a probability density of oocyte size) and the consequence of 17MT treatment (change in ovary). The initial ovarian status appeared to be an important factor influencing ovarian androgenic sensitivity. We postulated that the sensitivities of initial ovaries are correlated with their gene expression profiles. Japanese eels underwent operation to sample the initial ovarian tissues, and the samples were stored in liquid nitrogen. Using high-throughput next-generation sequencing (NGS) technology, ovarian transcriptomic data were mined and analyzed based on functional gene classification with cutoff-based differentially expressed genes (DEGs); the ovarian status was transformed into gene expression profiles globally or was represented by a set of gene list. Our results also implied that the initial ovary might be an important factor influencing the outcomes of 17MT treatments, and the genes related with neuronal activities or neurogenesis seemed to play an essential role in the positive effect.
Collapse
Affiliation(s)
- Yung-Sen Huang
- Department of Life Science, National University of Kaohsiung, No. 700 Kaohsiung University Road, Nan Tzu Dist, 811, Kaohsiung, Taiwan.
| | - Wen-Chih Cheng
- Institute of Information Science, Academia Sinica, No. 128 Academia Road, Section 2, Nankang Dist., 115, Taipei, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, No. 128 Academia Road, Section 2, Nankang Dist., 115, Taipei, Taiwan
| |
Collapse
|
27
|
Frost ER, Taylor G, Baker MA, Lovell-Badge R, Sutherland JM. Establishing and maintaining fertility: the importance of cell cycle arrest. Genes Dev 2021; 35:619-634. [PMID: 33888561 PMCID: PMC8091977 DOI: 10.1101/gad.348151.120] [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: 01/01/2023]
Abstract
In this review, Frost et al. summarize the current knowledge on the Cip/Kip family of cyclin-dependent kinase inhibitors in mouse gonad development and highlight new roles for cell cycle inhibitors in controlling and maintaining female fertility. Development of the ovary or testis is required to establish reproductive competence. Gonad development relies on key cell fate decisions that occur early in embryonic development and are actively maintained. During gonad development, both germ cells and somatic cells proliferate extensively, a process facilitated by cell cycle regulation. This review focuses on the Cip/Kip family of cyclin-dependent kinase inhibitors (CKIs) in mouse gonad development. We particularly highlight recent single-cell RNA sequencing studies that show the heterogeneity of cyclin-dependent kinase inhibitors. This diversity highlights new roles for cell cycle inhibitors in controlling and maintaining female fertility.
Collapse
Affiliation(s)
- Emily R Frost
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia.,Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Güneş Taylor
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Mark A Baker
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| | - Robin Lovell-Badge
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| |
Collapse
|
28
|
Telfer EE, Andersen CY. In vitro growth and maturation of primordial follicles and immature oocytes. Fertil Steril 2021; 115:1116-1125. [PMID: 33823993 DOI: 10.1016/j.fertnstert.2021.03.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/18/2022]
Abstract
Cryopreservation of ovarian tissue to preserve the fertility of girls and young women at high risk of sterility is now widely practiced. Pieces of cryopreserved ovarian cortex can be thawed and autografted to restore fertility, but because of the risks of reintroduction of the cancer, transplantation may not be possible for girls and women with blood-borne leukemias or cancers with a high risk of ovarian metastasis. Cryopreserved ovarian tissue contains mainly primordial follicles but also provides access to immature oocytes from small antral follicles, which may be matured in vitro to provide an additional source of mature oocytes. So in cases in which transplantation is contraindicated, fertility restoration could be safely achieved in the laboratory either by in vitro maturation (IVM) of oocytes aspirated from growing follicles or by the complete in vitro growth (IVG) and maturation (IVM) of primordial follicles to produce fertile metaphase II (MII) oocytes. The development of IVM and IVG methods to support all stages of oocytes available within ovarian tissue will maximize the potential for all patients undergoing fertility preservation.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, The University of Edinburgh, Edinburgh EH8 8XE, Scotland.
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Faculty of Health and Medical Science, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
29
|
Yao J, Huang R, Li M, Jiang Y, Wu P, Li Y, Peng W, Hua C, Huang Y, You H, Chen Y, Lin D, Yang X. PTEN Expression in Human Granulosa Cells Is Associated with Ovarian Responses and Clinical Outcomes in IVF. Reprod Sci 2021; 28:1910-1921. [PMID: 33439476 DOI: 10.1007/s43032-020-00429-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/10/2020] [Indexed: 12/30/2022]
Abstract
The ovarian reserve determines the success of in vitro fertilization (IVF) and embryo transfer treatment. It predicts the ovarian response in controlled ovarian hyperstimulation cycles. Apoptosis in granulosa cells surrounding oocytes is important for ovarian function and has been closely associated with follicular atresia. PTEN (encoding phosphatase and tensin homolog) is a well-known tumor suppressor gene that functions as a mediator of apoptosis and is crucial for mammal reproduction. In the present study, we analyzed the expression level of PTEN in human granulosa cells and aimed to investigate its association with the ovarian response and clinical outcomes in IVF. Apoptosis in granulosa cells were analyzed using Annexin V-Allophycocyanin staining after PTEN short hairpin RNA lentivirus transfection. Real-time fluorescent quantitative PCR analysis showed that the PTEN transcript level was significantly higher in poor responders and significantly lower in high responders, compared with that in normal responders. However, PTEN expression in the pregnancy group decreased slightly, but not significantly, compared with that in the non-pregnancy group. The apoptosis rate of granulosa cells declined significantly after 24-h transfection of the PTEN-shRNA lentivirus. These results suggest a fundamental role of PTEN in the regulation of follicular development, and that it might be involved in the pathogenesis of follicular dysplasia and ovarian dysfunction.
Collapse
Affiliation(s)
- Jianfeng Yao
- Quanzhou Maternity & Child Healthcare Hospital, Quanzhou, People's Republic of China
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Rongfu Huang
- The Second Affiliated Hospital, Fujian Medical University, Quanzhou, People's Republic of China
| | - Ming Li
- Department of Histology and Embryology, Hunan University of Medicine, Huaihua, People's Republic of China
| | - Yi Jiang
- Quanzhou Center for Disease Control and Prevention, Quanzhou, People's Republic of China
| | - Peiya Wu
- Quanzhou Maternity & Child Healthcare Hospital, Quanzhou, People's Republic of China
| | - Youzhu Li
- The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Weilin Peng
- Quanzhou Maternity & Child Healthcare Hospital, Quanzhou, People's Republic of China
| | - Chengzhou Hua
- Quanzhou Maternity & Child Healthcare Hospital, Quanzhou, People's Republic of China
| | - Yanfang Huang
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Huifang You
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, People's Republic of China
| | - Yuanyuan Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Dianliang Lin
- Fuzhou Maternity & Child Healthcare Hospital, Fuzhou, People's Republic of China.
| | - Xiaoyu Yang
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, People's Republic of China.
- Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, People's Republic of China.
| |
Collapse
|
30
|
Zhang R, Wesevich V, Chen Z, Zhang D, Kallen AN. Emerging roles for noncoding RNAs in female sex steroids and reproductive disease. Mol Cell Endocrinol 2020; 518:110875. [PMID: 32668269 PMCID: PMC7609472 DOI: 10.1016/j.mce.2020.110875] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 02/08/2023]
Abstract
The "central dogma" of molecular biology, that is, that DNA blueprints encode messenger RNAs which are destined for translation into protein, has been challenged in recent decades. In actuality, a significant portion of the genome encodes transcripts that are transcribed into functional RNA. These noncoding RNAs (ncRNAs), which are not transcribed into protein, play critical roles in a wide variety of biological processes. A growing body of evidence derived from mouse models and human data demonstrates that ncRNAs are dysregulated in various reproductive pathologies, and that their expression is essential for female gametogenesis and fertility. Yet in many instances it is unclear how dysregulation of ncRNA expression leads to a disease process. In this review, we highlight new observations regarding the roles of ncRNAs in the pathogenesis of disordered female steroid hormone production and disease, with an emphasis on long noncoding RNAs (lncRNAs) and microRNAs (miRNAs). We will focus our discussion in the context of three ovarian disorders which are characterized in part by altered steroid hormone biology - diminished ovarian reserve, premature ovarian insufficiency, and polycystic ovary syndrome. We will also discuss the limitations and challenges faced in studying noncoding RNAs and sex steroid hormone production. An enhanced understanding of the role of ncRNAs in sex hormone regulatory networks is essential in order to advance the development of potential diagnostic markers and therapeutic targets for diseases, including those in reproductive health. Our deepened understanding of ncRNAs has the potential to uncover new applications and therapies; however, in many cases, the next steps will involve distinguishing critical ncRNAs from those which are merely changing in response to a particular disease state, or which are altogether unrelated to disease pathophysiology.
Collapse
Affiliation(s)
- Runju Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1st Xueshi Road, Hangzhou, Zhejiang, China
| | - Victoria Wesevich
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Zhaojuan Chen
- Department of Gynecology. Beijing Haidian Hospital of Traditional Chinese Medicine, Beijing, China.
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1st Xueshi Road, Hangzhou, Zhejiang, China.
| | - Amanda N Kallen
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
31
|
Chen Y, Yang W, Shi X, Zhang C, Song G, Huang D. The Factors and Pathways Regulating the Activation of Mammalian Primordial Follicles in vivo. Front Cell Dev Biol 2020; 8:575706. [PMID: 33102482 PMCID: PMC7554314 DOI: 10.3389/fcell.2020.575706] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022] Open
Abstract
Mammalian ovaries consist of follicles as basic functional units. Each follicle comprised an innermost oocyte and several surrounding flattened granulosa cells. Unlike males, according to the initial size of the primordial follicle pool and the rate of its activation and depletion, a female's reproductive life has been determined early in life. Primordial follicles, once activated, will get into an irreversible process of development. Most follicles undergo atretic degeneration, and only a few of them could mature and ovulate. Although there are a lot of researches contributing to exploring the activation of primordial follicles, little is known about its underlying mechanisms. Thus, in this review, we collected the latest papers and summarized the signaling pathways as well as some factors involved in the activation of primordial follicles, hoping to lead to a more profound understanding of the cellular and molecular mechanisms of primordial follicle activation.
Collapse
Affiliation(s)
- Yao Chen
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weina Yang
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Shi
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlu Zhang
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ge Song
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, Guangzhou, China
| | - Donghui Huang
- Institute of Reproduction Health Research (Institute of Family Planning Research), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Abstract
Primary ovarian insufficiency (POI) is an uncommon yet devastating occurrence that results from a premature depletion of the ovarian pool of primordial follicles. Our understanding of both putative and plausible mechanisms underlying POI, previously considered to be largely "idiopathic", has been furthered over the past several years, largely due to advances in the field of genetics and through expansion of translational models for experimental research. In this review, our goal is to familiarize the multidisciplinary readers of the F1000 platform with the strides made in the field of reproductive medicine that hold both preventative and therapeutic implications for those women who are at risk for or who have POI.
Collapse
Affiliation(s)
- Victoria Wesevich
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Amanada N Kellen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Lubna Pal
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
33
|
Control of Murine Primordial Follicle Growth Activation by IκB/NFκB Signaling. Reprod Sci 2020; 27:2063-2074. [PMID: 32542534 DOI: 10.1007/s43032-020-00225-3] [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: 02/20/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023]
Abstract
The transcription factor NFκB has been associated with the timing of menopause in a large human genome-wide association study. Furthermore, preclinical studies demonstrate that loss of Tumor necrosis factor alpha (Tnfα) or its receptor Tnfr2 slows primordial follicle growth activation (PFGA). Although Tnfα:receptor signaling stimulates NFκB and may mechanistically link these findings, very little is known about NFκB signaling in PFGA. Because signaling downstream of Tnfα/Tnfr2 ligand/receptor interaction has not been interrogated as relates to PFGA, we evaluated the expression of key NFκB signaling proteins in primordial and growing follicles, as well as during ovarian aging. We show that key members of the NFκB pathway, including subunits, activating kinases, and inhibitory proteins, are expressed in the murine ovary. Furthermore, the subunits p65 and p50, and the cytosolic inhibitory proteins IκBα and IκBβ, are present in ovarian follicles, including at the primordial stage. Finally, we assessed PFGA in genetically modified mice (AKBI) previously demonstrated to be resistant to inflammatory stress-induced NFκB activation due to overexpression of the NFκB inhibitory protein IκBβ. Consistent with the hypothesis that NFκB plays a key role in PFGA, AKBI mice exhibit slower PGFA than wild-type (WT) controls, and their ovaries contain nearly twice the number of primordial follicles as WT both at early and late reproductive ages. These data provide mechanistic insight on the control of PFGA and suggest that targeting NFκB at the level of IκB proteins may be a tractable route to slowing the rate of PFGA in women faced with early ovarian demise.
Collapse
|
34
|
Xia X, Burn MS, Chen Y, Karakaya C, Kallen A. The relationship between H19 and parameters of ovarian reserve. Reprod Biol Endocrinol 2020; 18:46. [PMID: 32404103 PMCID: PMC7218823 DOI: 10.1186/s12958-020-00578-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
CONTEXT The H19 long noncoding RNA (lncRNA) belongs to a highly conserved, imprinted gene cluster involved in embryonic development and growth control. We previously described a novel mechanism whereby the Anti-mullerian hormone (Amh) appears to be regulated by H19. However, the relationship between circulating H19 and markers of ovarian reserve including AMH not been investigated. OBJECTIVE To determine whether H19 expression is altered in women with decreased ovarian reserve. DESIGN Experimental study. SETTING Yale School of Medicine (New Haven, USA) and Gazi University School of Medicine (Ankara, Turkey). PATIENTS OR OTHER PARTICIPANTS A total of 141 women undergoing infertility evaluation and treatment. INTERVENTION Collection of discarded blood samples and cumulus cells at the time of baseline infertility evaluation and transvaginal oocyte retrieval, respectively. MAIN OUTCOME MEASURE Serum and cumulus cell H19 expression. RESULTS Women with diminished ovarian reserve (as determined by AMH) had significantly lower serum H19 expression levels as compared to controls (p < 0.01). Serum H19 was moderately positively correlated with serum AMH. H19 expression was increased 3.7-fold in cumulus cells of IVF patients who demonstrated a high response to gonadotropins, compared to low responders (p < 0.05). CONCLUSION In this study, we show that downregulation of H19 in serum and cumulus cells is closely associated with decreased ovarian reserve, as measured by decreased AMH levels and reduced oocyte yield at oocyte retrieval. Further study with expanded sample sizes is necessary to determine whether H19 may be of use as a novel biomarker for diminished ovarian reserve.
Collapse
Affiliation(s)
- Xi Xia
- Reproductive Center, Peking University Shenzhen Hospital, Shenzhen 518000, China
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
| | - Martina S. Burn
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
| | - Yong Chen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province 350122 P.R. China
| | - Cengiz Karakaya
- Department of Medical Biochemistry, Gazi University School of Medicine, Ankara, Turkey
| | - Amanda Kallen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Yale School of Medicine, Gynecology, and Reproductive Sciences, New Haven, Connecticut 06512 USA
| |
Collapse
|
35
|
Xie Y, Li S, Zhou L, Lin H, Jiao X, Qiu Q, Liang Y, Zhang Q. Rapamycin preserves the primordial follicle pool during cisplatin treatment in vitro and in vivo. Mol Reprod Dev 2020; 87:442-453. [PMID: 32112509 DOI: 10.1002/mrd.23330] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 01/16/2020] [Indexed: 12/22/2022]
Abstract
Rapamycin has been proven to effectively inhibit the activation of primordial follicles while cisplatin-induced the loss of primordial follicles due to the over-activation of the primordial follicle stockpile. Whether rapamycin could inhibit the loss of primordial follicles induced by cisplatin is still unknown. The ovaries of neonatal Sprague Dawley rats were cultured in vitro in different doses of rapamycin (0.08, 0.16, and 0.32 μg/ml) and cisplatin (0.1, 0.4, and 0.8 μg/ml). The immature BALB/c mice were administered cisplatin with or without rapamycin by intraperitoneal injection. Ovaries were collected to analyze the histomorphology, the messenger RNA (mRNA) expression of anti-Mullerian hormone (AMH), growth differentiation factor 9 (GDF9), and bone morphogenetic protein 15 (BMP15) and the expression of key proteins of mammalian target of rapamycin (mTOR) pathway. Growing follicle counts of ovaries cultured in vitro in the R0.16 and R0.32 groups were decreased and the ratio of growing to primordial follicles was also decreased in a dose-dependent manner. In the C0.8 group, growing follicles were decreased compared with the other groups while the ratio was substantially increased in the C0.4 and C0.8 group. Co-treatment attenuated primordial follicle loss and reduced the upregulated ratio induced by cisplatin. Ovarian follicle dynamics in vivo was consistent with the in vitro results. Primordial follicles counts were statistically increased and the ratio was reduced in the rapamycin group compared with the control group. Primordial follicle counts were dramatically reduced in the cisplatin group whereas co-treatment with rapamycin slightly recovered its counts. There was no obvious difference in the number of growing follicles between the cisplatin group and other groups. The ratio was significantly increased in cisplatin-treated mice whereas decreased in the co-treatment group. The apoptosis rate of antral follicles in cisplatin-treated mice was higher than the other groups while the apoptosis rate was decreased in the co-treatment group in vivo. Compared with the control and rapamycin group, the mRNA expression of AMH, GDF9, and BMP15 were downregulated in the cisplatin group. The co-treatment group recovered the mRNA expression of BMP15. In addition, the expression of key protein of mTOR pathway rpS6 and its phosphorylated forms were increased in the cisplatin-treated group while co-treatment decreased their expression. Rapamycin attenuated the loss of primordial follicles induced by cisplatin through the inhibitory effect of rapamycin on the mTOR pathway. These results suggest that rapamycin may be an effective drug for the protection of ovarian function during chemotherapy.
Collapse
Affiliation(s)
- Yanqiu Xie
- Department of Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Gongdong, China.,Department of Obstetrics and Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Gongdong, China
| | - Song Li
- Department of Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Gongdong, China
| | - Linyan Zhou
- Department of Reproductive Medicine Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Gongdong, China
| | - Haiyan Lin
- Department of Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Gongdong, China
| | - Xuedan Jiao
- Department of Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Gongdong, China
| | - Qi Qiu
- Department of Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Gongdong, China
| | - Yihua Liang
- Department of Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Gongdong, China
| | - Qingxue Zhang
- Department of Reproductive Medicine Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Gongdong, China
| |
Collapse
|
36
|
Lee HN, Chang EM. Primordial follicle activation as new treatment for primary ovarian insufficiency. Clin Exp Reprod Med 2019; 46:43-49. [PMID: 31181871 PMCID: PMC6572666 DOI: 10.5653/cerm.2019.46.2.43] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/16/2019] [Indexed: 12/24/2022] Open
Abstract
Primordial follicle activation is a process in which individual primordial follicles leave their dormant state and enter a growth phase. While existing hormone stimulation strategies targeted the growing follicles, the remaining dormant primordial follicles were ruled out from clinical use. Recently, in vitro activation (IVA), which is a method for controlling primordial follicle activation, has provided an innovative technology for primary ovarian insufficiency (POI) patients. IVA was developed based on Hippo signaling and phosphatase and tensin homolog (PTEN)/phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/forkhead box O3 (FOXO3) signaling modulation. With this method, dormant primordial follicles are activated to enter growth phase and developed into competent oocytes. IVA has been successfully applied in POI patients who only have a few remaining remnant primordial follicles in the ovary, and healthy pregnancies and deliveries have been reported. IVA may also provide a promising option for fertility preservation in cancer patients and prepubertal girls whose fertility preservation choices are limited to tissue cryopreservation. Here, we review the basic mechanisms, translational studies, and current clinical results for IVA. Limitations and further study requirements that could potentially optimize IVA for future use will also be discussed.
Collapse
Affiliation(s)
- Hye Nam Lee
- Department of Obstetrics and Gynecology, Fertility Center, CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Eun Mi Chang
- Department of Obstetrics and Gynecology, Fertility Center, CHA Gangnam Medical Center, CHA University, Seoul, Korea
| |
Collapse
|
37
|
Abstract
Mammalian target of rapamycin (mTOR) is a conserved serine/threonine kinase of the phosphatidylinositol kinase-related kinase family that regulates cell growth, metabolism, and autophagy. Extensive research has linked mTOR to several human diseases including cancer, neurodegenerative disorders, and aging. In this review, recent publications regarding the mechanisms underlying the role of mTOR in female reproduction under physiological and pathological conditions are summarized. Moreover, we assess whether strategies to improve or suppress mTOR expression could have therapeutic potential for reproductive diseases like premature ovarian failure, polycystic ovarian syndrome, and endometriosis.
Collapse
|
38
|
A (micro)environmental perspective on the evolution of female reproductive aging. J Assist Reprod Genet 2018; 35:2129-2131. [PMID: 30374734 DOI: 10.1007/s10815-018-1355-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 10/22/2018] [Indexed: 01/08/2023] Open
|
39
|
Garcia-Guerra A, Wiltbank MC, Battista SE, Kirkpatrick BW, Sartori R. Mechanisms regulating follicle selection in ruminants: lessons learned from multiple ovulation models. Anim Reprod 2018; 15:660-679. [PMID: 36249844 PMCID: PMC9536070 DOI: 10.21451/1984-3143-ar2018-0027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/12/2018] [Indexed: 12/02/2022] Open
Abstract
Selection of a single dominant follicle from a cohort of growing follicles is a unique biological process, a key step in female reproductive function in monovular species, and lies at the core of reproductive technologies in cattle. Follicle growth and the number of follicles that ovulate are regulated by precise endocrine, paracrine, and autocrine mechanisms. Most of our current understanding about follicle selection focuses on the role of FSH, LH, and the IGF family in follicle growth and selection of the dominant follicle. However, more recently the role of members of the TGF-ß family has been highlighted, particularly in high fecundity genotypes in sheep. Intercellular signaling between the oocyte and granulosa cells (GC) regulates proliferation and differentiation due to actions of bone morphogenetic protein 15 (BMP15) and growth and differentiation factor 9 (GDF9) within the follicle. Mutations that either knockout or reduce the activity of BMP15 or GDF9 have been found to increase ovulation rate in heterozygotes and generally cause severe follicle abnormalities in homozygotes. A mutation in the intracellular kinase domain of the BMPR1B receptor (Booroola fecundity gene) increases ovulation rate in heterozygotes with further increases in ovulation in homozygotes. The physiological mechanisms linking these mutations to increased ovulation rates are still not well defined. A recently identified high fecundity bovine genotype, Trio, causes increased expression of SMAD6, an intracellular inhibitor of the BMP15/GDF9 signalling pathways. This bovine model has provided insights into the mechanisms associated with selection of multiple dominant follicles and multiple ovulations in carriers of fecundity alleles. The present review focuses on the mechanisms involved in follicle selection in ruminants with a special emphasis on the contribution made by multiple ovulation models in both cattle and sheep. The evaluation of multiple ovulation models in ruminants has allowed us to construct a new physiological model that relates changes in the BMP15/GDF9 signalling pathways to the physiological changes that result in selection of multiple dominant follicles. This model is characterized by acquisition of dominance at a smaller follicle size but at a similar time in the follicular wave with multiple follicles acquiring dominance in a hierarchal sequence, delaying FSH suppression and, thus allowing additional follicles to continue to grow and acquire dominance.
Collapse
Affiliation(s)
- Alvaro Garcia-Guerra
- Department of Animal Sciences, The Ohio State University, Columbus, OH, 43210 USA
| | - Milo C. Wiltbank
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sarah E. Battista
- Department of Animal Sciences, The Ohio State University, Columbus, OH, 43210 USA
| | - Brian W. Kirkpatrick
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Roberto Sartori
- Department of Animal Science, University of São Paulo, Piracicaba, SP, 13418-900, Brazil
| |
Collapse
|