1
|
Veiga GB, Zanini BM, Garcia DN, Hense JD, Barreto MM, Isola JVV, Mondadori RG, Masternak MM, Stout MB, Schneider A. Effects of calorie, protein, and branched chain amino acid restriction on ovarian aging in mice. Reprod Biol 2024; 24:100856. [PMID: 38295721 PMCID: PMC10978239 DOI: 10.1016/j.repbio.2024.100856] [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: 11/26/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 04/02/2024]
Abstract
Calorie restriction (CR) is an intervention that promotes longevity and preserves the ovarian reserve. Some studies have observed that the positive impacts of CR can be linked to restriction of protein (PR) and branched-chain amino acids (BCAAs) independent of calorie intake. The aim of this study was to compare the effects of protein and BCAA restriction to 30% CR on the ovarian reserve of female mice. For this, 3 month-old C57BL/6 female mice (n = 35) were randomized into four groups for four months dietary interventions including: control group (CTL; n = 8), 30% CR (CR; n = 9), protein restriction (PR; n = 9) and BCAA restriction (BCAAR; n = 9). Body mass gain, body composition, food intake, serum levels of BCAAs, ovarian reserve and estrous cyclicity were evaluated. We observed that CR, protein and BCAA restriction prevented weight gain and changed body composition compared to the CTL group. The BCAA restriction did not affect the ovarian reserve, while both PR and CR prevented activation of primordial follicles. This prevention occurred in PR group despite the lack of reduction of calorie intake compared to CTL group, and CR did not reduce protein intake in levels similar to the PR group. BCAA restriction resulted in increased calorie intake compared to CTL and PR mice, but only PR reduced serum BCAA levels compared to the CTL group. Our data indicates that PR has similar effects to CR on the ovarian reserve, whereas BCAA restriction alone did not affect it.
Collapse
Affiliation(s)
- Gabriel B Veiga
- Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Bianka M Zanini
- Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | | | - Jéssica D Hense
- Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | | | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA; Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | |
Collapse
|
2
|
Zhou X, He Y, Quan H, Pan X, Zhou Y, Zhang Z, Yuan X, Li J. HDAC1-Mediated lncRNA Stimulatory Factor of Follicular Development to Inhibit the Apoptosis of Granulosa Cells and Regulate Sexual Maturity through miR-202-3p- COX1 Axis. Cells 2023; 12:2734. [PMID: 38067162 PMCID: PMC10706290 DOI: 10.3390/cells12232734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Abnormal sexual maturity exhibits significant detrimental effects on adult health outcomes, and previous studies have indicated that targeting histone acetylation might serve as a potential therapeutic approach to regulate sexual maturity. However, the mechanisms that account for it remain to be further elucidated. Using the mouse model, we showed that Trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor, downregulated the protein level of Hdac1 in ovaries to promote the apoptosis of granulosa cells (GCs), and thus arrested follicular development and delayed sexual maturity. Using porcine GCs as a cell model, a novel sexual maturity-associated lncRNA, which was named as the stimulatory factor of follicular development (SFFD), transcribed from mitochondrion and mediated by HDAC1, was identified using RNA sequencing. Mechanistically, HDAC1 knockdown significantly reduced the H3K27ac level at the -953/-661 region of SFFD to epigenetically inhibit its transcription. SFFD knockdown released miR-202-3p to reduce the expression of cyclooxygenase 1 (COX1), an essential rate-limited enzyme involved in prostaglandin synthesis. This reduction inhibited the proliferation and secretion of 17β-estradiol (E2) while promoting the apoptosis of GCs. Consequently, follicular development was arrested and sexual maturity was delayed. Taken together, HDAC1 knockdown-mediated SFFD downregulation promoted the apoptosis of GCs through the miR-202-3p-COX1 axis and lead to delayed sexual maturity. Our findings reveal a novel regulatory network modulated by HDAC1, and HDAC1-mediated SFFD may be a promising new therapeutic target to treat delayed sexual maturity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaolong Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (Y.H.); (H.Q.); (X.P.); (Y.Z.); (Z.Z.)
| | - Jiaqi Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (Y.H.); (H.Q.); (X.P.); (Y.Z.); (Z.Z.)
| |
Collapse
|
3
|
Piau TB, de Queiroz Rodrigues A, Paulini F. Insulin-like growth factor (IGF) performance in ovarian function and applications in reproductive biotechnologies. Growth Horm IGF Res 2023; 72-73:101561. [PMID: 38070331 DOI: 10.1016/j.ghir.2023.101561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023]
Abstract
The role of the insulin-like growth factor (IGF) system has attracted close attention. The activity of IGF binding proteins (IGFBPs) within the ovary has not been fully elucidated to date. These proteins bind to IGF with an equal, or greater, affinity than to the IGF1 receptor, thus being in the main position to regulate IGF signalling, in addition to extending the half-life of IGFs within the bloodstream and promoting IGF storage in specific tissue niches. IGF1 has an important part in cell proliferation, differentiation and apoptosis. Considering the importance of IGFs in oocyte maturation, this review sought to elucidate aspects including: IGF production mechanisms; constituent members of their family and their respective functions; the role that these factors play during folliculogenesis, together with their functions during oocyte maturation and apoptosis, and their performance during luteal development. This review also explores the role of IGFs in biotechnological applications, focusing specifically on animal genetic gain.
Collapse
Affiliation(s)
- Tathyana Benetis Piau
- University of Brasília, Institute of Biological Sciences, Department of Physiological Sciences, Brasília, DF 70910-900, Brazil
| | - Aline de Queiroz Rodrigues
- University of Brasília, Institute of Biological Sciences, Department of Physiological Sciences, Brasília, DF 70910-900, Brazil
| | - Fernanda Paulini
- University of Brasília, Institute of Biological Sciences, Department of Physiological Sciences, Brasília, DF 70910-900, Brazil.
| |
Collapse
|
4
|
Effects of short-term in vitro heat stress on bovine preantral follicles. Livest Sci 2022. [DOI: 10.1016/j.livsci.2022.105076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
5
|
Bromoacetic acid impairs mouse oocyte in vitro maturation through affecting cytoskeleton architecture and epigenetic modification. Chem Biol Interact 2022; 368:110192. [PMID: 36174739 DOI: 10.1016/j.cbi.2022.110192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022]
Abstract
As a major public health achievement, disinfection of drinking water significantly decreases outbreaks of waterborne disease, but produces drinking water disinfection by-products (DBPs) unfortunately. The haloacetic acids (HAAs) including bromoacetic acid (BAA), the second major class of DBPs, are considered as a global public health concern. BAA has been identified as cytotoxic, genotoxic, mutagenic, carcinogenic, and teratogenic in somatic cells. However, the toxic effects of BAA on oocyte maturation remain obscure. Herein, we documented that exposure to BAA compromised mouse oocyte maturation in vitro, causing blocked polar body extrusion (PBE). Meiotic progression analysis demonstrated that exposure to BAA induced the activated spindle assembly checkpoint (SAC) mediated metaphase I (MI) arrest in oocytes. Further study revealed that exposure to BAA resulted in the hyperacetylation of α-tubulin, disrupting spindle assembly and chromosome alignment, which is responsible for the activation of SAC. Besides, the organization of actin, the other major component of cytoskeleton in oocytes, was disturbed after BAA exposure. In addition, exposure to BAA altered the status of histone H3 methylation and 5 mC, indicative of the damaged epigenetic modifications. Moreover, we found that exposure to BAA induced DNA damage in a dose-dependent manner in oocytes. Collectively, our study evidenced that exposure to BAA intervened mouse oocyte maturation via disrupting cytoskeletal dynamics, damaging epigenetic modifications and inducing accumulation of DNA damage.
Collapse
|
6
|
Dai S, Zhang H, Yang F, Shang W, Zeng S. Effects of IGF-1 on the Three-Dimensional Culture of Ovarian Preantral Follicles and Superovulation Rates in Mice. BIOLOGY 2022; 11:biology11060833. [PMID: 35741354 PMCID: PMC9219699 DOI: 10.3390/biology11060833] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 12/25/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) plays a crucial role during folliculogenesis, which has been demonstrated by previous research. However, the optimal IGF-1 dosage in the three-dimensional (3D) culture system is unknown. Mouse secondary follicles (140−150 µm) were cultured for 6 days within an alginate bead in a medium supplemented with 0 (G0), 5 ng/mL (G5), 10 ng/mL (G10), or 50 ng/mL IGF-1 (G50). Secretions of 17β-estradiol and progesterone were significantly increased in G10 and G50 (p < 0.05). However, G50 significantly inhibited follicular growth (p < 0.05), while G10 showed a higher oocyte maturation rate. Thus, the 10 ng/mL IGF-1 was used in subsequent experiments. IGF-1 enhanced the function of granulosa cells (GCs) by upregulating expressions of Star, Cyp19a1, Hsd3b1, Fshr, and Lhcgr. Oocyte secretory function was promoted by upregulating expressions of Bmp-15, Gdf-9, and Fgf-8. Addition of IGF-1 showed anti-apoptotic effect. However, G10 did not improve fertilization rate of MII oocytes compared to G0. In an intraperitoneal injection experiment in mice, IGF-1 significantly increased the number of ovulated oocytes (p < 0.05). In conclusion, 10 ng/mL IGF-1 can promote the production of mature oocytes in the 3D culture medium and injection of IGF-1 before superovulation increases the number of ovulated oocytes.
Collapse
Affiliation(s)
- Shizhen Dai
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.D.); (H.Z.); (F.Y.)
| | - Hanxue Zhang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.D.); (H.Z.); (F.Y.)
| | - Feng Yang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.D.); (H.Z.); (F.Y.)
| | - Wei Shang
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Center for Reproductive Medicine, The Sixth Medical Center, Beijing 100037, China
- Correspondence: (W.S.); (S.Z.)
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.D.); (H.Z.); (F.Y.)
- Correspondence: (W.S.); (S.Z.)
| |
Collapse
|
7
|
Wang H, Liu L, Liu C, Wang L, Chen J, Wang H, Heng D, Zeng M, Liu C, Zhou Z, Ye X, Wan Y, Li H, Liu L. Induction of meiosis by embryonic gonadal somatic cells differentiated from pluripotent stem cells. Stem Cell Res Ther 2021; 12:607. [PMID: 34930450 PMCID: PMC8686525 DOI: 10.1186/s13287-021-02672-4] [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: 08/20/2021] [Accepted: 12/02/2021] [Indexed: 11/13/2022] Open
Abstract
Background Depletion of oocytes leads to ovarian aging-associated infertility, endocrine disruption and related diseases. Excitingly, unlimited oocytes can be generated by differentiation of primordial germ cell like cells (PGCLCs) from pluripotent stem cells. Nevertheless, development of oocytes and follicles from PGCLCs relies on developmentally matched gonadal somatic cells, only available from E12.5 embryos in mice. It is therefore imperative to achieve an in vitro source of E12.5 gonadal somatic cells. Methods We explored to identify small molecules, which can induce female embryonic stem cells (ESCs) into gonadal somatic cell like cells. Results Using RNA-sequencing, we identified signaling pathways highly upregulated in E12.5_gonadal somatic cells (E12.5_GSCs). Through searching for the activators of these pathways, we identified small-molecule compounds Vitamin C (Vc) and AM580 in combination (V580) for inducing differentiation of female embryonic stem cells (ESCs) into E12.5_GSC-like cells (E12.5_GSCLCs). After V580 treatment for 6 days and sorted by a surface marker CD63, the cell population yielded a transcriptome profile similar to that of E12.5_GSCs, which promoted meiosis progression and folliculogenesis of primordial germ cells. This approach will contribute to the study of germ cell and follicle development and oocyte production and have implications in potentially treating female infertility. Conclusion ESCs can be induced into embryonic gonadal somatic cell like cells by small molecules. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02672-4.
Collapse
Affiliation(s)
- Haiying Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Linlin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Chang Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Lingling Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Jiyu Chen
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Huasong Wang
- Department of Cell Biology, College of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dai Heng
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Ming Zeng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chun Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Zhongcheng Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510655, China
| | - Xiaoying Ye
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Yajuan Wan
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Huiyu Li
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China. .,Department of Cell Biology and Genetics, College of Life Sciences; The Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, China. .,The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
8
|
Bulletti C, Allegra A, Mignini Renzini M, Vaiarelli A. How fixed versus variable gonadotropin dose during controlled ovarian stimulation could influence the management of infertility patients undergoing IVF treatment: a national Delphi consensus. Gynecol Endocrinol 2021; 37:255-263. [PMID: 32588675 DOI: 10.1080/09513590.2020.1770214] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/27/2020] [Accepted: 05/13/2020] [Indexed: 01/30/2023] Open
Abstract
AIM Define how and when fixed starting gonadotropin doses can be used in current clinical ART practices in Italy. METHODS A Delphi conference consisting of three rounds was performed in order to define the ideal clinical conditions in which fixed-gonadotropin-dose during COS should be applied. During the conference, 19 statements about the current ART practice were provided to a panel of twenty-nine national experts. Median score was 5 (IQ:4-6) in all Delphi rounds. RESULTS Eleven statements (57.9%) were classified as shareable with high-degree of convergence, 2 (10.5%) as shareable with low convergence and 6 (31.6%) as un-shareable with high convergence. The panel reached high consensus regarding some statements: (i) fixed FSH-dose in normoresponders and poor-responder, (ii) importance of predicting ovarian response before COS, considering multiple markers to select the right stimulation protocol for each patient, (iii) importance of therapy simplification and standardization to improve efficiency during COS. Moreover, a low-convergence was reached about use of GnRH antagonist as first treatment line and drug storage at room temperature. However from these findings, the debate remains open regarding some other statements: (a) usefulness of Bologna-criteria to define poor-responders; (b) efficacy to change always stimulation protocol after a failure IVF; (c) utility of AMH-dosed with standardized automatic mode to define normo-responder patients; (d) usefulness to modify the dosage of 12.5 IU/die during COS to improve stimulation effectiveness. CONCLUSION Controlled ovarian stimulation remains a challenging clinical step in Assisted Reproductive Technique, especially in some specific patient groups for which no clinical consensus is available. This study is the first attempt to describe the shared clinical opinion regarding the fixed versus variable gonadotropin dose in the real IVF practice.
Collapse
Affiliation(s)
- Carlo Bulletti
- Ostetricia e Ginecologia, EXTRA OMNES Medicina e Salute Riproduttiva, Cattolica, Italy
| | | | | | - Alberto Vaiarelli
- GENERA Centers for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| |
Collapse
|
9
|
Place NJ, Prado AM, Faykoo-Martinez M, Brieño-Enriquez MA, Albertini DF, Holmes MM. Germ cell nests in adult ovaries and an unusually large ovarian reserve in the naked mole-rat. Reproduction 2021; 161:89-98. [PMID: 33151901 DOI: 10.1530/rep-20-0304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/05/2020] [Indexed: 01/25/2023]
Abstract
The naked mole-rat (NMR, Heterocephalus glaber) is renowned for its eusociality and exceptionally long lifespan (> 30 y) relative to its small body size (35-40 g). A NMR phenomenon that has received far less attention is that females show no decline in fertility or fecundity into their third decade of life. The age of onset of reproductive decline in many mammalian species is closely associated with the number of germ cells remaining at the age of sexual maturity. We quantified ovarian reserve size in NMRs at the youngest age (6 months) when subordinate females can begin to ovulate after removal from the queen's suppression. We then compared the NMR ovarian reserve size to values for 19 other mammalian species that were previously reported. The NMR ovarian reserve at 6 months of age is exceptionally large at 108,588 ± 69,890 primordial follicles, which is more than 10-fold larger than in mammals of a comparable size. We also observed germ cell nests in ovaries from 6-month-old NMRs, which is highly unusual since breakdown of germ cell nests and the formation of primordial follicles is generally complete by early postnatal life in other mammals. Additionally, we found germ cell nests in young adult NMRs between 1.25 and 3.75 years of age, in both reproductively activated and suppressed females. The unusually large NMR ovarian reserve provides one mechanism to account for this species' protracted fertility. Whether germ cell nests in adult ovaries contribute to the NMR's long reproductive lifespan remains to be determined.
Collapse
Affiliation(s)
- Ned J Place
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, New York, USA
| | - Alexandra M Prado
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, New York, USA
| | | | - Miguel Angel Brieño-Enriquez
- Department of Obstetrics, Gynecology & Reproductive Medicine, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David F Albertini
- Department of Reproductive Biology, Bedford Research Foundation, Bedford, Massachusetts, USA
| | - Melissa M Holmes
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada.,Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
10
|
Zhang C, Shen J, Kong S, Zhang M, Zhang Q, Zhou J, Zhen X, Kang N, Jiang Y, Ding L, Sun H, Yan G. MicroRNA-181a promotes follicular granulosa cell apoptosis via sphingosine-1-phosphate receptor 1 expression downregulation†. Biol Reprod 2020; 101:975-985. [PMID: 31359035 DOI: 10.1093/biolre/ioz135] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/25/2019] [Accepted: 07/17/2019] [Indexed: 01/08/2023] Open
Abstract
Oxidative stress induces granulosa cell (GC) apoptosis and subsequent follicular atresia. Since our previous studies indicate that microRNA-181a (miR-181a) expression is increased in GCs undergoing apoptosis, the present study was designed to define the relationship between exposure to oxidative stressors in GCs and changes in miR-181a expression and function. To achieve this, we employed an H2O2-induced in vitro model and a 3-nitropropionic acid-induced in vivo model of ovarian oxidative stress. We demonstrated that in vitro miR-181a overexpression promoted GC apoptosis in a dose-dependent manner; sphingosine-1-phosphate (S1P) significantly reversed both H2O2-induced and miR-181a-induced apoptosis in GCs. Moreover, we identified sphingosine-1-phosphate receptor 1 (S1PR1), a critical receptor of S1P, as a novel target of miR-181a in GCs. MicroRNA-181a induced GC apoptosis by repressing S1PR1 expression in vitro. Importantly, increased miR-181a expression and decreased S1PR1 expression were detected in the in vivo ovarian oxidative stress model by Western blot analysis and immunohistochemistry. Furthermore, we found similar expression patterns of miR-181a and S1PR1 in GCs from patients with premature ovarian insufficiency. In conclusion, our results suggest that miR-181a directly suppresses expression of S1PR1, which has critical roles in mediating oxidative stress-induced GC apoptosis both in vitro and in vivo.
Collapse
Affiliation(s)
- Chunxue Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jingtao Shen
- Department of Nuclear Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shuangbo Kong
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Mei Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qun Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jidong Zhou
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xin Zhen
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Nannan Kang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yue Jiang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Lijun Ding
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Haixiang Sun
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,State Key Laboratory of Pharmaceutical Biotechnology, Department of Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guijun Yan
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Carnevale EM, Catandi GD, Fresa K. Equine Aging and the Oocyte: A Potential Model for Reproductive Aging in Women. J Equine Vet Sci 2020; 89:103022. [PMID: 32563447 DOI: 10.1016/j.jevs.2020.103022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/25/2022]
Abstract
Numerous similarities in reproductive aging have been documented between the mare and woman. Aging is associated with a decline in fertility. In mares and women, oocyte transfer procedures were initially used to establish that oocyte donor age is associated with oocyte quality. Age-associated differences in oocytes include altered morphology, gene expression, and developmental potential. Reactive oxygen species and mitochondrial dysfunction are thought to be important contributors to loss of oocyte quality. In the woman, aneuploidy is a primary consideration with maternal aging. Although misalignment of chromosomes during meiosis has been observed in the mare, less is known in this area. Reproductive aging will be reviewed in the mare and compared with the woman with emphasis on factors that affect oocyte quality and developmental potential. Areas in which the mare could be used as a research model to study reproductive aging in women will be highlighted.
Collapse
Affiliation(s)
- Elaine M Carnevale
- Equine Reproduction Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO.
| | - Giovana D Catandi
- Equine Reproduction Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Kyle Fresa
- Equine Reproduction Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| |
Collapse
|
12
|
Hu S, Yang S, Lu Y, Deng Y, Li L, Zhu J, Zhang Y, Hu B, Hu J, Xia L, He H, Han C, Liu H, Kang B, Li L, Wang J. Dynamics of the Transcriptome and Accessible Chromatin Landscapes During Early Goose Ovarian Development. Front Cell Dev Biol 2020; 8:196. [PMID: 32309280 PMCID: PMC7145905 DOI: 10.3389/fcell.2020.00196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
In contrast to the situation in mammals, very little is known about the molecular mechanisms regulating early avian ovarian development. This study aimed to investigate the dynamic changes in the histomorphology as well as the genome-wide transcriptome and chromatin accessibility landscapes of the goose ovary during late embryonic and early post-hatching stages. Results from hematoxylin-eosin, periodic acid-Schiff, and anti-CVH immunohistochemical stainings demonstrated that programmed oocyte loss, oocyte nest breakdown and primordial follicle formation, and the primordial-to-secondary follicle transition occur during the periods from embryonic day 15 (E15) to post-hatching day 0 (P0), from P0 to P4, and from P4 to P28, respectively. RNA-seq and ATAC-seq analyses revealed dynamic changes in both the ovarian transcriptome and accessible chromatin landscapes during early ovarian development, exhibiting the most extensive changes during peri-hatching oocyte loss, and moreover, differences were also identified in the genomic distribution of the differential ATAC-seq peaks between different developmental stages, suggesting that chromatin-level regulation of gene expression is facilitated by modulating the accessibility of different functional genomic regions to transcription factors. Motif analysis of developmental stage-selective peak regions identified hundreds of potential cis-regulatory elements that contain binding sites for many transcription factors, including SF1, NR5A2, ESRRβ, NF1, and THRβ, as well as members of the GATA, SMAD, and LHX families, whose expression fluctuated throughout early goose ovarian development. Integrated ATAC-seq and RNA-seq analysis suggested that the number and genomic distribution of the newly appeared and disappeared peaks differed according to developmental stage, and in combination with qRT-PCR validation potentiated the critical actions of the DEGs enriched in cell cycle, MAPK signaling, and FoxO signaling pathways during peri-hatching oocyte loss and those in ligand-receptor interaction, tissue remodeling, lipid metabolism, and Wnt signaling during primordial follicle formation and development. In conclusion, our study provides a framework for understanding the transcriptome and accessible chromatin dynamics during early avian ovarian development and a new avenue to unravel the transcriptional regulatory mechanisms that facilitate the occurrence of relevant molecular events.
Collapse
Affiliation(s)
- Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shuang Yang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yao Lu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yan Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jiaran Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yuan Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Lu Xia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Bo Kang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
13
|
Bezerra MÉS, Barberino RS, Menezes VG, Gouveia BB, Macedo TJS, Santos JMS, Monte APO, Barros VRP, Matos MHT. Insulin-like growth factor-1 (IGF-1) promotes primordial follicle growth and reduces DNA fragmentation through the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signalling pathway. Reprod Fertil Dev 2019; 30:1503-1513. [PMID: 29843892 DOI: 10.1071/rd17332] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 04/18/2018] [Indexed: 11/23/2022] Open
Abstract
We investigated the effects of insulin-like growth factor 1 (IGF-1) on the morphology and follicular activation of ovine preantral follicles cultured in situ and whether the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway is involved in IGF-1 action in the sheep ovary. Ovine ovarian fragments were fixed for histological and terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) analyses (fresh control) or cultured in supplemented alpha-minimum essential medium (α-MEM+; control) or α-MEM+ with IGF-1 (1, 10, 50, 100 or 200ngmL-1) for 7 days. Follicles were classified as normal or atretic, primordial or growing and the oocyte and follicle diameters were measured. DNA fragmentation was evaluated by TUNEL assay. Proliferating cell nuclear antigen (PCNA) immunohistochemistry was performed on the fresh control, α-MEM+ and 100ngmL-1 IGF-1 samples. Inhibition of PI3K activity was performed through pretreatment with the PI3K inhibitor LY294002 and phosphorylated AKT (pAKT) expression was analysed after culture in the absence or presence of LY294002. IGF-1 at 100ngmL-1 increased (P<0.05) follicular activation compared with α-MEM+ and decreased TUNEL-positive cells (P<0.05) compared with other treatments. PCNA-positive cells also increased (P<0.05) in 100ngmL-1 IGF-1. LY294002 significantly inhibited follicular activation stimulated by α-MEM+ and 100ngmL-1 IGF-1 and reduced pAKT expression in follicles. Overall, IGF-1 at 100ngmL-1 promoted primordial follicle activation, cell proliferation and reduced DNA fragmentation after in situ culture through the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Maria É S Bezerra
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Vanúzia G Menezes
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Bruna B Gouveia
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Taís J S Macedo
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Jamile M S Santos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Alane P O Monte
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Vanessa R P Barros
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| | - Maria H T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Rodovia BR 407, Km 12, Lote 543, Projeto C1, CEP: 56300-990, Petrolina, PE, Brazil
| |
Collapse
|
14
|
Zhe J, Chen S, Chen X, Liu Y, Li Y, Zhou X, Zhang J. A novel heterozygous splice-altering mutation in HFM1 may be a cause of premature ovarian insufficiency. J Ovarian Res 2019; 12:61. [PMID: 31279343 PMCID: PMC6612105 DOI: 10.1186/s13048-019-0537-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/01/2019] [Indexed: 12/23/2022] Open
Abstract
Background Premature ovarian insufficiency (POI) leads to early loss of ovarian function in women aged < 40 years and is highly heterogeneous in etiology. The genetic etiology of this disorder remains unknown in most women with POI. Methods Whole-exome sequencing (WES) was used to analyze genetic factors within a Chinese POI pedigree. Bioinformatic analysis was applied to identify the potential genetic cause, and Sanger sequencing confirmed the existence of a mutation within the pedigree. A minigene assay was performed to validate the effect of the mutation on pre-mRNA splicing. Results A novel heterozygous missense mutation in HFM1 (c.3470G > A) associated with POI was identified by whole-exome sequencing. This mutation was heterozygous in the affected family members and was absent in the unaffected family members. In silico analysis predicted that the mutation was potentially pathogenic. Bioinformatic splice prediction tools revealed that the mutation was very likely to have a strong impact on splice site function. Results of the minigene assay revealed that the mutation changed the mRNA splicing repertory. Conclusions The missense mutation of the HFM1 gene (c.3470G > A) may be a cause of POI. The mutation altered mRNA splicing in cells. This study can provide geneticists with deeper insight into the pathogenesis of POI and aid clinicians in making early diagnoses in affected women.
Collapse
Affiliation(s)
- Jing Zhe
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shiling Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Xin Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yudong Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ying Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xingyu Zhou
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| |
Collapse
|
15
|
Ge W, Li L, Dyce PW, De Felici M, Shen W. Establishment and depletion of the ovarian reserve: physiology and impact of environmental chemicals. Cell Mol Life Sci 2019; 76:1729-1746. [PMID: 30810760 PMCID: PMC11105173 DOI: 10.1007/s00018-019-03028-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/19/2019] [Accepted: 01/28/2019] [Indexed: 01/09/2023]
Abstract
The reproductive life span in women starts at puberty and ends at menopause, following the exhaustion of the follicle stockpile termed the ovarian reserve. Increasing data from experimental animal models and epidemiological studies indicate that exposure to a number of ubiquitously distributed reproductively toxic environmental chemicals (RTECs) can contribute to earlier menopause and even premature ovarian failure. However, the causative relationship between environmental chemical exposure and earlier menopause in women remains poorly understood. The present work, is an attempt to review the current evidence regarding the effects of RTECs on the main ovarian activities in mammals, focusing on how such compounds can affect the ovarian reserve at any stages of ovarian development. We found that in rodents, strong evidence exists that in utero, neonatal, prepubescent and even adult exposure to RTECs leads to impaired functioning of the ovary and a shortening of the reproductive lifespan. Regarding human, data from cross-sectional surveys suggest that human exposure to certain environmental chemicals can compromise a woman's reproductive health and in some cases, correlate with earlier menopause. In conclusion, evidences exist that exposure to RTECs can compromise a woman's reproductive health. However, human exposures may date back to the developmental stage, while the adverse effects are usually diagnosed decades later, thus making it difficult to determine the association between RTECs exposure and human reproductive health. Therefore, epidemiological surveys and more experimental investigation on humans, or alternatively primates, are needed to determine the direct and indirect effects caused by RTECs exposure on the ovary function, and to characterize their action mechanisms.
Collapse
Affiliation(s)
- Wei Ge
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Lan Li
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Paul W Dyce
- Department of Animal Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Wei Shen
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
16
|
Uliani RC, Conley AJ, Corbin CJ, Friso AM, Maciel LFS, Alvarenga MA. Anti-Müllerian hormone and ovarian aging in mares. J Endocrinol 2019; 240:147-156. [PMID: 30400031 DOI: 10.1530/joe-18-0391] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 10/26/2018] [Indexed: 12/31/2022]
Abstract
Anti-Müllerian hormone (AMH) is used as a marker of follicle population numbers and potential fertility in several species including horses but limited data exist across the lifespan. No one has decreased ovarian reserve experimentally to investigate whether a corresponding, quantitative decrease in AMH results. Concentrations of AMH across the lifespan were compiled from 1101 equine females sampled from birth to >33 years of age. Young and old mares (averaging 6 and 19 years) were hemi-ovariectomized and circulating AMH was assessed before and daily thereafter for 15 days. The remaining ovary was removed later and blood was drawn again before and after this second surgery for AMH determination. Polynomial regression analysis and analysis of mares grouped by 5-year intervals of age demonstrated AMH concentrations to be higher in mares aged 5-10 and 10-15 years than 0-5 years of age and lower in mares after 20 years of age. There was high variability in AMH concentrations among neonatal fillies, some of which had concentrations typical of males. Hemi-ovariectomy was followed by a decrease of AMH, almost exactly halving concentrations in intact mares. Concentrations of AMH had returned to intact levels in old mares before complete ovariectomy, as if exhibiting ovarian compensatory hypertrophy, but recovery of AMH was not evident in young mares. AMH may reflect ovarian senescence in mares after 20 years of age but is too variable to do so in the first two decades of life. The ovarian endocrine response to hemi-ovariectomy in mares appears to change with age.
Collapse
Affiliation(s)
- Renata C Uliani
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Alan J Conley
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - C Jo Corbin
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Aimê M Friso
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Luciana F S Maciel
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Marco A Alvarenga
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
17
|
Hiratsuka K, Monkawa T, Akiyama T, Nakatake Y, Oda M, Goparaju SK, Kimura H, Chikazawa-Nohtomi N, Sato S, Ishiguro K, Yamaguchi S, Suzuki S, Morizane R, Ko SBH, Itoh H, Ko MSH. Induction of human pluripotent stem cells into kidney tissues by synthetic mRNAs encoding transcription factors. Sci Rep 2019; 9:913. [PMID: 30696889 PMCID: PMC6351687 DOI: 10.1038/s41598-018-37485-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 12/05/2018] [Indexed: 01/10/2023] Open
Abstract
The derivation of kidney tissues from human pluripotent stem cells (hPSCs) and its application for replacement therapy in end-stage renal disease have been widely discussed. Here we report that consecutive transfections of two sets of synthetic mRNAs encoding transcription factors can induce rapid and efficient differentiation of hPSCs into kidney tissues, termed induced nephron-like organoids (iNephLOs). The first set - FIGLA, PITX2, ASCL1 and TFAP2C, differentiated hPSCs into SIX2+SALL1+ nephron progenitor cells with 92% efficiency within 2 days. Subsequently, the second set - HNF1A, GATA3, GATA1 and EMX2, differentiated these cells into PAX8+LHX1+ pretubular aggregates in another 2 days. Further culture in both 2-dimensional and 3-dimensional conditions produced iNephLOs containing cells characterized as podocytes, proximal tubules, and distal tubules in an additional 10 days. Global gene expression profiles showed similarities between iNephLOs and the human adult kidney, suggesting possible uses of iNephLOs as in vitro models for kidneys.
Collapse
Affiliation(s)
- Ken Hiratsuka
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
- Department of Nephrology, Endocrinology, and Metabolism, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Toshiaki Monkawa
- Department of Nephrology, Endocrinology, and Metabolism, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
- Medical Education Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Tomohiko Akiyama
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Yuhki Nakatake
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Mayumi Oda
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Sravan Kumar Goparaju
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Hiromi Kimura
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Nana Chikazawa-Nohtomi
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Saeko Sato
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Keiichiro Ishiguro
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
- Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Shintaro Yamaguchi
- Department of Nephrology, Endocrinology, and Metabolism, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Sayuri Suzuki
- Department of Nephrology, Endocrinology, and Metabolism, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Ryuji Morizane
- Department of Nephrology, Endocrinology, and Metabolism, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Shigeru B H Ko
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Hiroshi Itoh
- Department of Nephrology, Endocrinology, and Metabolism, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Minoru S H Ko
- Department of Systems Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan.
| |
Collapse
|
18
|
Western PS. Epigenomic drugs and the germline: Collateral damage in the home of heritability? Mol Cell Endocrinol 2018; 468:121-133. [PMID: 29471014 DOI: 10.1016/j.mce.2018.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 02/07/2023]
Abstract
The testis and ovary provide specialised environments that nurture germ cells and facilitate their maturation, culminating in the production of mature gametes that can found the following generation. The sperm and egg not only transmit genetic information, but also epigenetic modifications that affect the development and physiology of offspring. Importantly, the epigenetic information contained in mature sperm and oocytes can be influenced by a range of environmental factors, such as diet, chemicals and drugs. An increasing range of studies are revealing how gene-environment interactions are mediated through the germline. Outside the germline, altered epigenetic state is common in a range of diseases, including many cancers. As epigenetic modifications are reversible, pharmaceuticals that directly target epigenetic modifying proteins have been developed and are delivering substantial benefits to patients, particularly in oncology. While providing the most effective patient treatment is clearly the primary concern, some patients will want to conceive children after treatment. However, the impacts of epigenomic drugs on the male and female gametes are poorly understood and whether these drugs will have lasting effects on patients' germline epigenome and subsequent offspring remains largely undetermined. Currently, evidence based clinical guidelines for use of epigenomic drugs in patients of reproductive age are limited in this context. Developing a deeper understanding of the epigenetic mechanisms regulating the germline epigenome and its impact on inherited traits and disease susceptibility is required to determine how specific epigenomic drugs might affect the germline and inheritance. Understanding these potential effects will facilitate the development of informed clinical guidelines appropriate for the use of epigenomic drugs in patients of reproductive age, ultimately improving the safety of these therapies in the clinic.
Collapse
Affiliation(s)
- Patrick S Western
- Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
19
|
Nguyen QN, Zerafa N, Liew SH, Morgan FH, Strasser A, Scott CL, Findlay JK, Hickey M, Hutt KJ. Loss of PUMA protects the ovarian reserve during DNA-damaging chemotherapy and preserves fertility. Cell Death Dis 2018; 9:618. [PMID: 29795269 PMCID: PMC5966424 DOI: 10.1038/s41419-018-0633-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/15/2018] [Accepted: 04/26/2018] [Indexed: 12/14/2022]
Abstract
Female gametes are stored in the ovary in structures called primordial follicles, the supply of which is non-renewable. It is well established that DNA-damaging cancer treatments can deplete the ovarian reserve of primordial follicles, causing premature ovarian failure and infertility. The precise mechanisms underlying this chemotherapy-driven follicle loss are unclear, and this has limited the development of targeted ovarian-protective agents. To address this fundamental knowledge gap, we used gene deletion mouse models to examine the role of the DNA damage-induced pro-apoptotic protein, PUMA, and its transcriptional activator TAp63, in primordial follicle depletion caused by treatment with cyclophosphamide or cisplatin. Cyclophosphamide caused almost complete destruction of the primordial follicle pool in adult wild-type (WT) mice, and a significant destructive effect was also observed for cisplatin. In striking contrast, Puma-/- mice retained 100% of their primordial follicles following either genotoxic treatment. Furthermore, elimination of PUMA alone completely preserved fertility in cyclophosphamide-treated mice, indicating that oocytes rescued from DNA damage-induced death can repair themselves sufficiently to support reproductive function and offspring health. Primordial follicles were also protected in TAp63-/- mice following cisplatin treatment, but not cyclophosphamide, suggesting mechanistic differences in the induction of apoptosis and depletion of the ovarian reserve in response to these different chemotherapies. These studies identify PUMA as a crucial effector of apoptosis responsible for depletion of primordial follicles following exposure to cyclophosphamide or cisplatin, and this indicates that inhibition of PUMA may be an effective ovarian-protective strategy during cancer treatment in women.
Collapse
Affiliation(s)
- Quynh-Nhu Nguyen
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Nadeen Zerafa
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Seng H Liew
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - F Hamish Morgan
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Clare L Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Jock K Findlay
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Monash University, Clayton, VIC, Australia
| | - Martha Hickey
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- The Royal Womens Hospital, Parkville, VIC, 3052, Australia
| | - Karla J Hutt
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
20
|
Brandão FAS, Alves BG, Alves KA, Souza SS, Silva YP, Freitas VJF, Teixeira DIA, Gastal EL. Laparoscopic ovarian biopsy pick-up method for goats. Theriogenology 2017; 107:219-225. [PMID: 29179058 DOI: 10.1016/j.theriogenology.2017.10.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 10/23/2017] [Accepted: 10/30/2017] [Indexed: 11/25/2022]
Abstract
Biopsy pick-up (BPU) has been considered a safe method to harvest ovarian fragments from live animals. However, no studies have been reported on the use of BPU to collect in vivo ovarian tissue in goats. The goals of this study were: (i) to test different biopsy needle sizes to collect ovarian tissue in situ using the BPU method (Experiment 1), and (ii) to study ovarian tissue features such as preantral follicle density, morphology, class distribution, and stromal cell density in ovarian fragments obtained in vivo through a laparoscopic BPU method (Experiment 2). In Experiment 1, goat ovaries (n = 20) were collected in a slaughterhouse and subjected to in situ BPU. Three needles (16, 18, and 20G) were tested. In Experiment 2, the most efficient biopsy needle from Experiment 1 was used to perform laparoscopic BPU in goats (n = 8). In Experiment 1, the recovery rate was greater (P < 0.05; range 50-62%) with 16G and 18G needles than the 20G (17%) needle. The mean weight of ovarian fragments collected by the 16G needle was greater (P < 0.05) than the 18G and the 20G needle. In Experiment 2, 62 biopsy attempts were performed and 52 ovarian fragments were collected (90% success rate). Overall, 2054 preantral follicles were recorded in 5882 histological sections analyzed. Mean preantral follicular density was 28.4 ± 1.3 follicles per cm2. The follicular density differed (P < 0.05) among animals and ovarian fragments within the same animal. The mean stromal cell density in the ovarian fragments was 37.1 ± 0.5 cells per 2500 μm2, and differed (P < 0.05) among animals. Moreover, preantral follicle density and stromal cell density were associated (P < 0.001). The percentage of morphologically normal follicles was 70.1 ± 1.2, and differed (P < 0.05) among animals. The majority (79%) of the morphologically normal follicles was classified as primordial follicles, and differed (P < 0.05) among animals and between ovaries. In summary, a laparoscopic BPU method has been developed to harvest ovarian tissue in vivo with a satisfactory success rate in goats. Furthermore, this study described for the first time that goat ovarian biopsy fragments have a high heterogeneity in follicular density, morphology, class distribution, and stromal cell density.
Collapse
Affiliation(s)
- Fabiana A S Brandão
- Laboratory of Physiology and Control of Reproduction, State University of Ceará, Fortaleza, CE, Brazil
| | - Benner G Alves
- Laboratory of Manipulation of Oocytes and Preantral Follicles, State University of Ceará, Fortaleza, CE, Brazil
| | - Kele A Alves
- Laboratory of Manipulation of Oocytes and Preantral Follicles, State University of Ceará, Fortaleza, CE, Brazil
| | - Samara S Souza
- Laboratory of Physiology and Control of Reproduction, State University of Ceará, Fortaleza, CE, Brazil
| | - Yago P Silva
- Laboratory of Physiology and Control of Reproduction, State University of Ceará, Fortaleza, CE, Brazil
| | - Vicente J F Freitas
- Laboratory of Physiology and Control of Reproduction, State University of Ceará, Fortaleza, CE, Brazil
| | - Dárcio I A Teixeira
- Laboratory of Physiology and Control of Reproduction, State University of Ceará, Fortaleza, CE, Brazil
| | - Eduardo L Gastal
- Department of Animal Science, Food and Nutrition, Southern Illinois University, Carbondale, IL, USA.
| |
Collapse
|
21
|
miR-181a increases FoxO1 acetylation and promotes granulosa cell apoptosis via SIRT1 downregulation. Cell Death Dis 2017; 8:e3088. [PMID: 28981116 PMCID: PMC5680589 DOI: 10.1038/cddis.2017.467] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 08/06/2017] [Accepted: 08/17/2017] [Indexed: 12/23/2022]
Abstract
Oxidative stress impairs follicular development by inducing granulosa cell (GC) apoptosis, which involves enhancement of the transcriptional activity of the pro-apoptotic factor Forkhead box O1 (FoxO1). However, the mechanism by which oxidative stress promotes FoxO1 activity is still unclear. Here, we found that miR-181a was upregulated in hydrogen peroxide (H2O2)-treated GCs and a 3-nitropropionic acid (NP)-induced in vivo model of ovarian oxidative stress. miR-181a overexpression promoted GC apoptosis, whereas knockdown of endogenous miR-181a blocked H2O2-induced cell apoptosis. Moreover, we identified that Sirtuin 1 (SIRT1), a deacetylase that suppresses FoxO1 acetylation in GCs, was downregulated by miR-181a and reversed the promoting effects of H2O2 and miR-181a on FoxO1 acetylation and GC apoptosis. Importantly, decreased miR-181a expression in the in vivo ovarian oxidative stress model inhibited apoptosis by upregulating SIRT1 expression and FoxO1 deacetylation. Together, our results suggest that miR-181a mediates oxidative stress-induced FoxO1 acetylation and GC apoptosis by targeting SIRT1 both in vitro and in vivo.
Collapse
|
22
|
Abstract
Recently, the existence of a mechanism for neo-oogenesis in the ovaries of adult mammals has generated much controversy within reproductive biology. This mechanism, which proposes that the ovary has cells capable of renewing the follicular reserve, has been described for various species of mammals. The first evidence was found in prosimians and humans. However, these findings were not considered relevant because the predominant dogma for reproductive biology at the time was that of Zuckerman. This dogma states that female mammals are born with finite numbers of oocytes that decline throughout postnatal life. Currently, the concept of neo-oogenesis has gained momentum due to the discovery of cells with mitotic activity in adult ovaries of various mammalian species (mice, humans, rhesus monkeys, domestic animals such as pigs, and wild animals such as bats). Despite these reports, the concept of neo-oogenesis has not been widely accepted by the scientific community, generating much criticism and speculation about its accuracy because it has been impossible to reproduce some evidence. This controversy has led to the creation of two positions: one in favour of neo-oogenesis and the other against it. Various animal models have been used in support of both camps, including both classic laboratory animals and domestic and wild animals. The aim of this review is to critically present the current literature on the subject and to evaluate the arguments pro and contra neo-oogenesis in mammals.
Collapse
|
23
|
Patiño LC, Beau I, Carlosama C, Buitrago JC, González R, Suárez CF, Patarroyo MA, Delemer B, Young J, Binart N, Laissue P. New mutations in non-syndromic primary ovarian insufficiency patients identified via whole-exome sequencing. Hum Reprod 2017; 32:1512-1520. [DOI: 10.1093/humrep/dex089] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/13/2017] [Indexed: 12/11/2022] Open
|
24
|
Hill CE, Sapouckey SA, Suvorov A, Vandenberg LN. Developmental exposures to bisphenol S, a BPA replacement, alter estrogen-responsiveness of the female reproductive tract: A pilot study. COGENT MEDICINE 2017. [DOI: 10.1080/2331205x.2017.1317690] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Corinne E. Hill
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts – Amherst, 171A Goessmann, 686 N. Pleasant Street, Amherst, MA 01003, USA
| | - Sarah A. Sapouckey
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts – Amherst, 171A Goessmann, 686 N. Pleasant Street, Amherst, MA 01003, USA
| | - Alexander Suvorov
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts – Amherst, 171A Goessmann, 686 N. Pleasant Street, Amherst, MA 01003, USA
| | - Laura N. Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts – Amherst, 171A Goessmann, 686 N. Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
25
|
Abstract
The somatic component of follicular structure is a mixture of different cell types, represented by Granulosa cells (GCs) that are the paracrine regulators of the oocyte growth. GCs finely support this process by a continuous bidirectional talk with oocyte, which ensure oocyte quality and competence. Specific pathways are involved in the cross-talk and in both GCs and oocyte development. This review summarizes data from GCs gene expression analysis concerning both their physiological role and their interaction with oocyte. We also explore the CGs transcriptome modifications induced by controlled ovarian stimulation (COS) or pathological conditions and their impact in reproduction. The transcriptome analysis of GCs could be a powerful tool to improve our knowledge about the pathways involved in oocyte development. This approach, associated with new technologies as RNA-seq could allow the identifications of new noninvasive biological markers of oocyte quality to increase the efficiency of clinical IVF. Moreover, GCs expression analysis could be useful to shed light on new therapeutic targets by providing new options for the treatment of infertility.
Collapse
Affiliation(s)
- Marco D'Aurora
- a Department of Psychological , Health and Territorial Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
| | - Samantha Sperduti
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- c Department of Medical , Oral and Biotechnological Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Italy
| | - Giovanna Di Emidio
- d Department of Health , Life and Environmental Sciences, University of L'Aquila , L'Aquila , Italy Coppito , and
| | - Liborio Stuppia
- a Department of Psychological , Health and Territorial Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
| | - Paolo Giovanni Artini
- e Department of Experimental and Clinical Medicine , Division of Gynecology and Obstetrics, University of Pisa , Pisa , Italy
| | - Valentina Gatta
- a Department of Psychological , Health and Territorial Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
| |
Collapse
|
26
|
Pu D, Wang C, Cao J, Shen Y, Jiang H, Liu J, Wu BL, Zhang W, Wu J. Association analysis between HFM1 variation and primary ovarian insufficiency in Chinese women. Clin Genet 2016; 89:597-602. [PMID: 26679638 DOI: 10.1111/cge.12718] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 11/13/2015] [Accepted: 11/13/2015] [Indexed: 01/18/2023]
Abstract
HFM1 is a meiosis-specific gene and expressed in germ-line tissues. More recently, evidence has indicated that variations in HFM1 gene could be causative for primary ovarian insufficiency (POI), also known as premature ovarian failure. The aim of this study was to investigate the association between HFM1 gene variants and sporadic POI in Chinese women. A total of 138 POI patients and 316 healthy controls (matched for ethnic background, sex, and age of the patients) were recruited in this study. We screened the entire HFM1 coding region by direct sequencing in all subjects and identified six variants of HFM1 gene in POI group, namely c.148G>A/p.Glu50Lys, c.1241A>C/p.His414Pro, c.2325C>A/p.Phe775Leu, c.3367T>C/p.Ser1123Pro, c.3580C>T/p.Arg1194Cys, and c.1686-1G>C. The variation rate of HFM1 in POI group is significantly higher than control group (p < 0.01). The p.His414Pro and p.Arg1194Cys were predicted to be probably damaging to the HFM1 protein function, while p.Glu50Lys, p.Phe775Leu and p.Ser1123Pro mutants might not have any deleterious effect on the structure or function of the protein by online predictors. Taken together, our data suggested that HFM1 gene might be associated with primary ovarian insufficiency in Chinese population.
Collapse
Affiliation(s)
- D Pu
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University / Jiangsu Province Hospital / Jiangsu Women and Children Health Hospital, Nanjing, China
| | - C Wang
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University / Jiangsu Province Hospital / Jiangsu Women and Children Health Hospital, Nanjing, China
| | - J Cao
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University / Jiangsu Province Hospital / Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Y Shen
- Departments of Laboratory Medicine and Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - H Jiang
- Reproductive Medicine Center, the 105th hospital of PLA, Hefei, China
| | - J Liu
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University / Jiangsu Province Hospital / Jiangsu Women and Children Health Hospital, Nanjing, China
| | - B L Wu
- Departments of Laboratory Medicine and Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.,Children's Hospital of Fudan University, Department of Pathology and Institute of Biomedical Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - W Zhang
- Departments of Laboratory Medicine and Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.,Reproductive Medicine Center, the 105th hospital of PLA, Hefei, China
| | - J Wu
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University / Jiangsu Province Hospital / Jiangsu Women and Children Health Hospital, Nanjing, China
| |
Collapse
|
27
|
Monniaux D, Michel P, Postel M, Clément F. Multi-scale modelling of ovarian follicular development: From follicular morphogenesis to selection for ovulation. Biol Cell 2016; 108:149-60. [DOI: 10.1111/boc.201500087] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/02/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Danielle Monniaux
- INRA; UMR85 Physiologie de la Reproduction et des Comportements; Nouzilly F-37380 France
- CNRS; UMR7247; Nouzilly F-37380 France
- Université François Rabelais de Tours; Tours F-37041 France
- IFCE; Nouzilly F-37380 France
| | - Philippe Michel
- Université de Lyon; CNRS; UMR5208; Ecole Centrale de Lyon; Institut Camille Jordan; Ecully Cedex 69134 France
| | - Marie Postel
- Sorbonne Universités; UPMC Univ Paris 06; UMR7598; Laboratoire Jacques-Louis Lions; Paris F-75005 France
- Project-team Mycenae; Centre Inria de Paris; 75589 Paris Cedex 12 France
| | - Frédérique Clément
- Project-team Mycenae; Centre Inria de Paris; 75589 Paris Cedex 12 France
| |
Collapse
|
28
|
Gaytan F, Morales C, Leon S, Garcia-Galiano D, Roa J, Tena-Sempere M. Crowding and Follicular Fate: Spatial Determinants of Follicular Reserve and Activation of Follicular Growth in the Mammalian Ovary. PLoS One 2015; 10:e0144099. [PMID: 26642206 PMCID: PMC4671646 DOI: 10.1371/journal.pone.0144099] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/12/2015] [Indexed: 01/07/2023] Open
Abstract
Initiation of growth of resting ovarian follicles is a key phenomenon for providing an adequate number of mature oocytes in each ovulation, while preventing premature exhaustion of primordial follicle reserve during the reproductive lifespan. Resting follicle dynamics strongly suggest that primordial follicles are under constant inhibitory influences, by mechanisms and factors whose nature remains ill defined. In this work, we aimed to assess the influence of spatial determinants, with special attention to clustering patterns and crowding, on the fate of early follicles in the adult mouse and human ovary. To this end, detailed histological and morphometric analyses, targeting resting and early growing follicles, were conducted in ovaries from mice, either wild type (WT) or genetically modified to lack kisspeptin receptor expression (Kiss1r KO), and healthy adult women. Kiss1r KO mice were studied as model of persistent hypogonadotropism and anovulation. Different qualitative and quantitative indices of the patterns of spatial distribution of resting and early growing follicles in the mouse and human ovary, including the Morisita’s index of clustering, were obtained. Our results show that resting primordial follicles display a clear-cut clustered pattern of spatial distribution in adult mouse and human ovaries, and that resting follicle aggrupation is inversely correlated with the proportion of follicles initiating growth and entering into the growing pool. As a whole, our data suggest that resting follicle crowding, defined by changes in density and clustered pattern of distribution, is a major determinant of follicular activation and the fate of ovarian reserve. Uneven follicle crowding would constitute the structural counterpart of the major humoral regulators of early follicular growth, with potential implications in ovarian ageing and pathophysiology.
Collapse
Affiliation(s)
- Francisco Gaytan
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, 14004, Córdoba, Spain
- * E-mail: (FG); (MT-S)
| | | | - Silvia Leon
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, 14004, Córdoba, Spain
| | - David Garcia-Galiano
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
| | - Juan Roa
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, 14004, Córdoba, Spain
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, 14004, Córdoba, Spain
- FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland
- * E-mail: (FG); (MT-S)
| |
Collapse
|
29
|
Teng Z, Wang C, Wang Y, Huang K, Xiang X, Niu W, Feng L, Zhao L, Yan H, Zhang H, Xia G. S100A8, An Oocyte-Specific Chemokine, Directs the Migration of Ovarian Somatic Cells During Mouse Primordial Follicle Assembly. J Cell Physiol 2015; 230:2998-3008. [PMID: 25953201 DOI: 10.1002/jcp.25032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/04/2015] [Indexed: 01/22/2023]
Abstract
In the mammalian ovaries, the primordial follicle pool determines the reproductive capability over the lifetime of a female. The primordial follicle is composed of two cell members, namely the oocyte and the pre-granulosa cells that encircle the oocyte. However, it is unclear what factors are involved in the reorganization of the two distinct cells into one functional unit. This study was performed to address this issue. Firstly, in an in vitro reconstruction system, dispersed ovarian cells from murine fetal ovaries at 19.0 days post coitum (dpc) reassembled into follicle-like structures, independent of the physical distance between the cells, implying that either oocytes or ovarian somatic cells (OSCs) were motile. We then carried out a series of transwell assay experiments, and determined that it was in fact 19.0 dpc OSCs (as opposed to oocytes), which exhibited a significant chemotactic response to both fetal bovine serum and oocytes themselves. We observed that S100A8, a multi-functional chemokine, may participate in the process as it is mainly expressed in oocytes within the cysts/plasmodia. S100A8 significantly promoted the number of migrating OSCs by 2.5 times in vitro, of which 66.9% were FOXL2 protein-positive cells, implying that the majority of motile OSCs were pre-granulosa cells. In addition, an S100A8-specific antibody inhibited the formation of follicle-like reconstruction cell mass in vitro. And, the primordial follicle formation was reduced when S100a8-specific siRNA was applied onto in vitro cultured 17.5 dpc ovary. Therefore, S100A8 could be a chemokine of oocyte origin, which attracts OSCs to form the primordial follicles.
Collapse
Affiliation(s)
- Zhen Teng
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Chao Wang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Yijing Wang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Kun Huang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Xi Xiang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Wanbao Niu
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Lizhao Feng
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Lihua Zhao
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Hao Yan
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Hua Zhang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Guoliang Xia
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| |
Collapse
|
30
|
Lack of Cytosolic Carboxypeptidase 1 Leads to Subfertility due to the Reduced Number of Antral Follicles in pcd3J-/- Females. PLoS One 2015; 10:e0139557. [PMID: 26452267 PMCID: PMC4599934 DOI: 10.1371/journal.pone.0139557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022] Open
Abstract
Females homozygous for the Purkinje cell degeneration mutation (pcd) are fertile, although the success rate is much lower than in the wild type. We performed detailed analysis of reproductive abnormalities of pcd females. The number of oocytes produced following exogenous gonadotropin treatment was much lower in pcd3J-/- females than in pcd3J+/+ females. Furthermore, the estrous cyclicity of pcd3J-/- females according to the appearance of the vagina was almost undetectable comparing to that of the wild type. Histological analyses and follicle counting of 4- and 8-week-old pcd3J-/- ovaries showed an increase in the number of secondary follicles and a decrease in the number of antral follicles, indicating that AGTPBP1/ CCP1 plays an important role in the development of secondary follicles into antral follicles. Consistent with a previous analysis of the pcd cerebellum, pcd3J-/- ovaries also showed a clear increase in the level of polyglutamylation. Gene expression analysis showed that both oocytes and cumulus cells express CCP1. However, Ccp4 and CCP6, which can compensate the function of CCP1, were not expressed in mouse ovaries. Failure of microtubule deglutamylation did not affect the structure and function of the meiotic spindle in properly aligning chromosomes in the center of the nucleus during meiosis in pcd3J-/- females. We also showed that the pituitary-derived growth and reproduction-related endocrine system functions normally in pcd3J-/- mice. The results of this study provide insight into additional functions of CCP1, which cannot be fully explained by the side chain deglutamylation of microtubules alone.
Collapse
|
31
|
Findlay JK, Hutt KJ, Hickey M, Anderson RA. How Is the Number of Primordial Follicles in the Ovarian Reserve Established? Biol Reprod 2015; 93:111. [PMID: 26423124 DOI: 10.1095/biolreprod.115.133652] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022] Open
Abstract
The number of primordial follicles in the ovarian reserve is an important determinant of the length of the ovarian lifespan, and therefore the fertility of an individual. This reserve contains all of the oocytes potentially available for fertilization throughout the fertile lifespan. The maximum number is set during pregnancy or just after birth in most mammalian species; current evidence does not support neofolliculogenesis after the ovarian reserve is established, although this is increasingly being reexamined. Under physiological circumstances, this number will be influenced by the number of primordial germ cells initially specified in the epiblast of the developing embryo, their proliferation during and after migration to the developing gonads, and their death during oogenesis and formation of primordial follicles at nest breakdown. Death of germ cells during the establishment of the ovarian reserve occurs principally by autophagy or apoptosis, although the triggers that initiate these remain elusive. This review outlines the regulatory steps that determine the number of primordial follicles and thus the number of oocytes in the ovarian reserve at birth, using the mouse as the model, interspersed with human data where available. This information has application for understanding the variability in duration of fertility that occurs between normal individuals and with age, in premature ovarian insufficiency, and after chemotherapy or radiotherapy.
Collapse
Affiliation(s)
- John K Findlay
- Centre for Reproductive Biology, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Obstetrics & Gynaecology, Monash University, Clayton, Victoria, Australia Department of Obstetrics & Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Karla J Hutt
- Centre for Reproductive Biology, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Anatomy & Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Martha Hickey
- Department of Obstetrics & Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
32
|
Sirtuin Functions in Female Fertility: Possible Role in Oxidative Stress and Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:659687. [PMID: 26075037 PMCID: PMC4436464 DOI: 10.1155/2015/659687] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 10/13/2014] [Accepted: 11/04/2014] [Indexed: 12/15/2022]
Abstract
In search for strategies aimed at preventing oxidative threat to female fertility, a possible role of sirtuins has emerged. Sirtuins (silent information regulator 2 (Sir2) proteins), NAD(+) dependent enzymes with deacetylase and/or mono-ADP-ribosyltransferase activity, are emerging as key antiaging molecules and regulators in many diseases. Recently, a crucial role for SIRT1 and SIRT3, the main components of sirtuin family, as sensors and guardians of the redox state in oocytes, granulosa cells, and early embryos has emerged. In this context, the aim of the present review is to summarize current knowledge from research papers on the role of sirtuins in female fertility with particular emphasis on the impairment of SIRT1 signalling with oocyte aging. On this basis, the authors wish to build up a framework to promote research on the possible role of sirtuins as targets for future strategies for female fertility preservation.
Collapse
|
33
|
Tenenbaum-Rakover Y, Weinberg-Shukron A, Renbaum P, Lobel O, Eideh H, Gulsuner S, Dahary D, Abu-Rayyan A, Kanaan M, Levy-Lahad E, Bercovich D, Zangen D. Minichromosome maintenance complex component 8 (MCM8) gene mutations result in primary gonadal failure. J Med Genet 2015; 52:391-9. [DOI: 10.1136/jmedgenet-2014-102921] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/25/2015] [Indexed: 11/03/2022]
|
34
|
Abstract
I present evidence that humans have evolved convergently to social insects with regard to a large suite of social, ecological, and reproductive phenotypes. Convergences between humans and social insects include: (1) groups with genetically and environmentally defined structures; (2) extensive divisions of labor; (3) specialization of a relatively restricted set of females for reproduction, with enhanced fertility; (4) extensive extramaternal care; (5) within-group food sharing; (6) generalized diets composed of high-nutrient-density food; (7) solicitous juveniles, but high rates of infanticide; (8) ecological dominance; (9) enhanced colonizing abilities; and (10) collective, cooperative decision-making. Most of these convergent phenotypic adaptations stem from reorganization of key life-history trade-offs due to behavioral, physiological, and life-historical specializations. Despite their extensive socioreproductive overlap with social insects, humans differ with regard to the central aspect of eusociality: reproductive division of labor. This difference may be underpinned by the high energetic costs of producing offspring with large brains.
Collapse
Affiliation(s)
- Bernard Crespi
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada, V5A 1S6,
| |
Collapse
|
35
|
Chu Z, Niu B, Li N, Hu Y, Li J, Yu P, Wu C, Yan X, Lei A, Hua J. A lentiviral vector visualizing the germ cell specification in vitro under the control of Figla promoter. Appl Biochem Biotechnol 2015; 176:66-75. [PMID: 25652828 DOI: 10.1007/s12010-015-1523-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 01/21/2015] [Indexed: 01/30/2023]
Abstract
Premature ovarian failure (POF) is affecting more and more women, which is the loss of function of the ovaries before age 40. To elucidate the underlying mechanisms of the oogenesis is of importance to understand the causes of impaired fertility and POF. However, mammalian oogenesis in vivo is a complex process. Thus, building an oogenesis visualizing system is beneficial for the study of oogenesis. In this study, we found that Figla is specifically expressed in female mice oocyte. Then, we constructed a lentiviral vector (pTRIP-Figla-EGFP-puro) under the control of Figla promoter, which drived enhanced green fluorescent protein (EGFP) as an indicator and used the lentiviral vector transduction the ovarian cells and induced germ cells derived from human umbilical cord mesenchymal stem cells (hUC-MSCs), and the results showed that the lentiviral vector we constructed was able to specifically express green fluorescent protein (GFP) in the ovarian oocyte and induced oocyte-like cells derived from hUC-MSCs, which was Figla-positive cells. These results suggest that pTRIP-Figla-EGFP vector provides a new system to study the role of Figla in oogenesis, and an approach to study the development and the differentiation of germ cells derived from stem cells.
Collapse
Affiliation(s)
- Zhili Chu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Rastetter RH, Bernard P, Palmer JS, Chassot AA, Chen H, Western PS, Ramsay RG, Chaboissier MC, Wilhelm D. Marker genes identify three somatic cell types in the fetal mouse ovary. Dev Biol 2014; 394:242-52. [DOI: 10.1016/j.ydbio.2014.08.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/12/2014] [Accepted: 08/15/2014] [Indexed: 12/28/2022]
|
37
|
Modina SC, Tessaro I, Lodde V, Franciosi F, Corbani D, Luciano AM. Reductions in the number of mid-sized antral follicles are associated with markers of premature ovarian senescence in dairy cows. Reprod Fertil Dev 2014; 26:235-44. [PMID: 23327793 DOI: 10.1071/rd12295] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 11/26/2012] [Indexed: 01/31/2023] Open
Abstract
High-producing dairy cows are subfertile; however, the mechanisms responsible for the decreased fertility are unknown. The aim of the present study was to test the hypothesis that culled dairy cows (4-8 years old) characterised by 'Lo' ovaries (i.e. those with <10 mid-antral follicles) are affected by premature ovarian senescence. Cows in which both ovaries were 'Lo' ovaries represented 5% of the total population analysed, and exhibited reduced ovarian size (P<0.001) and increased perifollicular stroma (P<0.05) compared with age-matched controls (i.e. cows in which both ovaries had >10 mid-antral follicles; 'Hi' ovaries). The total number of follicles, including healthy and atretic primordial, primary, secondary and small antral follicles, was lower in Lo ovaries (P<0.01). Interestingly, the primordial follicle population in Lo ovaries was lower (P<0.05) than in the control. Finally, the follicular fluid of mid-antral follicles from Lo ovaries had reduced oestradiol and anti-Müllerian hormone levels (P<0.05), but increased progesterone concentrations (P<0.05). Together, these data account for the reduced fertility of cows with Lo ovaries and are in agreement with previous observations that oocytes isolated from Lo ovaries have reduced embryonic developmental competence. Cows with a specific Lo ovary condition may represent a suitable model to address the causes of low fertility in high-yielding dairy cows, as well as the condition of premature ovarian aging in single-ovulating species.
Collapse
Affiliation(s)
- Silvia C Modina
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, Università degli Studi di Milano, via G. Celoria 10, 20133, Milano, Italy
| | - Irene Tessaro
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, Università degli Studi di Milano, via G. Celoria 10, 20133, Milano, Italy
| | - Valentina Lodde
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, Università degli Studi di Milano, via G. Celoria 10, 20133, Milano, Italy
| | - Federica Franciosi
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, Università degli Studi di Milano, via G. Celoria 10, 20133, Milano, Italy
| | - Davide Corbani
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, Università degli Studi di Milano, via G. Celoria 10, 20133, Milano, Italy
| | - Alberto M Luciano
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Sciences and Food Safety, Università degli Studi di Milano, via G. Celoria 10, 20133, Milano, Italy
| |
Collapse
|
38
|
Monniaux D, Clément F, Dalbiès-Tran R, Estienne A, Fabre S, Mansanet C, Monget P. The ovarian reserve of primordial follicles and the dynamic reserve of antral growing follicles: what is the link? Biol Reprod 2014; 90:85. [PMID: 24599291 DOI: 10.1095/biolreprod.113.117077] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The growing follicles develop from a reserve of primordial follicles constituted early in life. From this pre-established reserve, a second ovarian reserve is formed, which consists of gonadotropin-responsive small antral growing follicles and is a dynamic reserve for ovulation. Its size, evaluated by direct antral follicular count or endocrine markers, determines the success of assisted reproductive technologies in humans and embryo production biotechnologies in animals. Strong evidence indicates that these two reserves are functionally related. The size of both reserves appears to be highly variable between individuals of similar age, but the equilibrium size of the dynamic reserve in adults seems to be specific to each individual. The dynamics of both follicular reserves appears to result from the fine tuning of regulations involving two main pathways, the phosphatase and tensin homolog (PTEN)/phosphatidylinositol-3 kinase (PI3K)/3-phosphoinositide-dependent protein kinase-1 (PDPK1)/v-akt murine thymoma viral oncogene homolog 1 (AKT1) and the bone morphogenetic protein (BMP)/anti-Müllerian hormone (AMH)/SMAD signaling pathways. Mutations in genes encoding the ligands, receptors, or signaling effectors of these pathways can accelerate or modulate the exhaustion rate of the ovarian reserves, causing premature ovarian insufficiency (POI) or increase in reproductive longevity, respectively. With female aging, the decline in primordial follicle numbers parallels the decrease in the size of the dynamic reserve of small antral follicles and the deterioration of oocyte quality. Recent progress in our knowledge of signaling pathways and their environmental and hormonal control during adult and fetal life opens new perspectives to improve the management of the ovarian reserves.
Collapse
Affiliation(s)
- Danielle Monniaux
- Institut National de la Recherche Agronomique (INRA), UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | | | | | | | | | | | | |
Collapse
|
39
|
Úbeda F, Ohtsuki H, Gardner A, Baalen M. Ecology drives intragenomic conflict over menopause. Ecol Lett 2014; 17:165-74. [PMID: 24320989 PMCID: PMC3912906 DOI: 10.1111/ele.12208] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/19/2013] [Accepted: 09/30/2013] [Indexed: 12/28/2022]
Abstract
Menopause is the transition from reproductive to non-reproductive life well before natural death. Rather than involving a smooth, rapid change, it is normally preceded by a long period of erratic hormonal fluctuation that is accompanied by a plethora of unpleasant symptoms. Here, we (1) suggest that this turbulent period owes to conflict, between a woman's maternally inherited (MI) and paternally inherited (PI) genes, over the trade-off between reproduction and communal care; (2) perform a theoretical analysis to show that this conflict is resolved either through silencing or fluctuating expression of one of the genes; (3) highlight which of the symptoms preceding menopause may result from antagonistic co-evolution of MI and PI genes; (4) argue that ecological differences between ancestral human populations may explain the variability in menopause among different ethnic groups; (5) discuss how these insights may be used to inform family planning and cancer risk assessment based on a woman's ancestral background.
Collapse
Affiliation(s)
- Francisco Úbeda
- School of Biological Sciences, Royal Holloway University of LondonEgham, Surrey, TW20 0EX, UK
| | - Hisashi Ohtsuki
- Department of Evolutionary Studies of Biosystems, The Graduate University for Advanced StudiesShonan Village, Kanagawa, Hayama, 240-0193, Japan
| | - Andy Gardner
- School of Biology, University of St AndrewsSt Andrews, KY16 9TH, UK
| | - Minus Baalen
- School of Biological Sciences, Royal Holloway University of LondonEgham, Surrey, TW20 0EX, UK
- Department of Evolutionary Studies of Biosystems, The Graduate University for Advanced StudiesShonan Village, Kanagawa, Hayama, 240-0193, Japan
- School of Biology, University of St AndrewsSt Andrews, KY16 9TH, UK
| |
Collapse
|
40
|
Bonnet A, Cabau C, Bouchez O, Sarry J, Marsaud N, Foissac S, Woloszyn F, Mulsant P, Mandon-Pepin B. An overview of gene expression dynamics during early ovarian folliculogenesis: specificity of follicular compartments and bi-directional dialog. BMC Genomics 2013; 14:904. [PMID: 24350644 PMCID: PMC3890531 DOI: 10.1186/1471-2164-14-904] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 12/10/2013] [Indexed: 12/03/2022] Open
Abstract
Background Successful early folliculogenesis is crucial for female reproductive function. It requires appropriate gene specific expression of the different types of ovarian cells at different developmental stages. To date, most gene expression studies on the ovary were conducted in rodents and did not distinguish the type of cell. In mono-ovulating species, few studies have addressed gene expression profiles and mainly concerned human oocytes. Results We used a laser capture microdissection method combined with RNA-seq technology to explore the transcriptome in oocytes and granulosa cells (GCs) during development of the sheep ovarian follicle. We first documented the expression profile of 15 349 genes, then focused on the 5 129 genes showing differential expression between oocytes and GCs. Enriched functional categories such as oocyte meiotic arrest and GC steroid synthesis reflect two distinct cell fates. We identified the implication of GC signal transduction pathways such as SHH, WNT and RHO GTPase. In addition, signaling pathways (VEGF, NOTCH, IGF1, etc.) and GC transzonal projections suggest the existence of complex cell-cell interactions. Finally, we highlighted several transcription regulators and specifically expressed genes that likely play an important role in early folliculogenesis. Conclusions To our knowledge, this is the first comprehensive exploration of transcriptomes derived from in vivo oocytes and GCs at key stages in early follicular development in sheep. Collectively, our data advance our understanding of early folliculogenesis in mono-ovulating species and will be a valuable resource for unraveling human ovarian dysfunction such as premature ovarian failure (POF).
Collapse
Affiliation(s)
- Agnes Bonnet
- INRA, UMR444 Génétique Cellulaire, Auzeville, BP52627, F-31326, Castanet-Tolosan, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The female germline comprises a reserve population of primordial (non-growing) follicles containing diplotene oocytes arrested in the first meiotic prophase. By convention, the reserve is established when all individual oocytes are enclosed by granulosa cells. This commonly occurs prior to or around birth, according to species. Histologically, the ‘reserve’ is the number of primordial follicles in the ovary at any given age and is ultimately depleted by degeneration and progression through folliculogenesis until exhausted. How and when the reserve reaches its peak number of follicles is determined by ovarian morphogenesis and germ cell dynamics involving i) oogonial proliferation and entry into meiosis producing an oversupply of oocytes and ii) large-scale germ cell death resulting in markedly reduced numbers surviving as the primordial follicle reserve. Our understanding of the processes maintaining the reserve comes primarily from genetically engineered mouse models, experimental activation or destruction of oocytes, and quantitative histological analysis. As the source of ovulated oocytes in postnatal life, the primordial follicle reserve requires regulation of i) its survival or maintenance, ii) suppression of development (dormancy), and iii) activation for growth and entry into folliculogenesis. The mechanisms influencing these alternate and complex inter-related phenomena remain to be fully elucidated. Drawing upon direct and indirect evidence, we discuss the controversial concept of postnatal oogenesis. This posits a rare population of oogonial stem cells that contribute new oocytes to partially compensate for the age-related decline in the primordial follicle reserve.
Collapse
|
42
|
La Marca A, Sunkara SK. Individualization of controlled ovarian stimulation in IVF using ovarian reserve markers: from theory to practice. Hum Reprod Update 2013; 20:124-40. [PMID: 24077980 DOI: 10.1093/humupd/dmt037] [Citation(s) in RCA: 353] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The main objective of individualization of treatment in IVF is to offer every single woman the best treatment tailored to her own unique characteristics, thus maximizing the chances of pregnancy and eliminating the iatrogenic and avoidable risks resulting from ovarian stimulation. Personalization of treatment in IVF should be based on the prediction of ovarian response for every individual. The starting point is to identify if a woman is likely to have a normal, poor or a hyper response and choose the ideal treatment protocol tailored to this prediction. The objective of this review is to summarize the predictive ability of ovarian reserve markers, such as antral follicle count (AFC) and anti-Mullerian hormone (AMH), and the therapeutic strategies that have been proposed in IVF after this prediction. METHODS A systematic review of the existing literature was performed by searching Medline, EMBASE, Cochrane library and Web of Science for publications in the English language related to AFC, AMH and their incorporation into controlled ovarian stimulation (COS) protocols in IVF. Literature available to May 2013 was included. RESULTS The search generated 305 citations of which 41 and 25 studies, respectively, reporting the ability of AMH and AFC to predict response to COS were included in this review. The literature review demonstrated that AFC and AMH, the most sensitive markers of ovarian reserve identified to date, are ideal in planning personalized COS protocols. These sensitive markers permit prediction of the whole spectrum of ovarian response with reliable accuracy and clinicians may use either of the two markers as they can be considered interchangeable. Following the categorization of expected ovarian response to stimulation clinicians can adopt tailored therapeutic strategies for each patient. Current scientific trend suggests the elective use of the GnRH antagonist based regimen for hyper-responders, and probably also poor responders, as likely to be beneficial. The selection of the appropriate and individualized gonadotrophin dose is also of paramount importance for effective COS and subsequent IVF outcomes. CONCLUSION Personalized IVF offers several benefits; it enables clinicians to give women more accurate information on their prognosis thus facilitating counselling especially in cases of extremes of ovarian response. The deployment of therapeutic strategies based on selective use of GnRH analogues and the fine tuning of the gonadotrophin dose on the basis of potential ovarian response in every single woman can allow for a safer and more effective IVF practice.
Collapse
Affiliation(s)
- Antonio La Marca
- Mother-Infant Department, Institute of Obstetrics and Gynaecology, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | | |
Collapse
|
43
|
Luciano AM, Franciosi F, Lodde V, Tessaro I, Corbani D, Modina SC, Peluso JJ. Oocytes isolated from dairy cows with reduced ovarian reserve have a high frequency of aneuploidy and alterations in the localization of progesterone receptor membrane component 1 and aurora kinase B. Biol Reprod 2013; 88:58. [PMID: 23325810 DOI: 10.1095/biolreprod.112.106856] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Oocytes isolated from cows of reproductive age with reduced antral follicle counts (AFC) have a diminished capacity of embryonic development, which may be related to alterations in the mechanism that directs the proper segregation of chromosomes. Because we demonstrated that progesterone receptor membrane component 1 (PGRMC1) is involved in chromosome congression and metaphase II (MII) plate formation, the present study was designed to determine 1) if the decrease in oocyte developmental competence observed in dairy cows with a reduced AFC is due to a higher incidence of aneuploidy and 2) whether alterations in PGRMC1 contributes to the incidence of aneuploidy. Oocytes from ovaries with reduced AFC and age-matched controls were matured in vitro and the occurrence of aneuploidy determined as well as the mRNA level and localization of PGRMC1. Although oocytes from ovaries with reduced AFC were capable of undergoing meiosis in vitro, these oocytes showed a 3-fold increase in aneuploidy compared to oocytes isolated from control ovaries (P < 0.05). Although Pgrmc1 mRNA levels were not altered, PGRMC1 and aurora kinase B (AURKB) failed to localize to precise focal points on MII chromosomes of oocytes from ovaries with reduced AFC. Furthermore, when oocytes of control ovaries were cultured with an inhibitor of AURKB activity, their MII plate was disrupted and PGRMC1 was not properly localized to the chromosomes. These results suggest that alterations in PGRMC1 and/or AURKB localization account in part for the increased aneuploidy and low development competence of oocytes from ovaries with reduced AFC.
Collapse
Affiliation(s)
- Alberto Maria Luciano
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
44
|
Xu J, Gridley T. Notch2 is required in somatic cells for breakdown of ovarian germ-cell nests and formation of primordial follicles. BMC Biol 2013; 11:13. [PMID: 23406467 PMCID: PMC3606475 DOI: 10.1186/1741-7007-11-13] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 02/13/2013] [Indexed: 12/22/2022] Open
Abstract
Background In the mouse ovary, oocytes initially develop in clusters termed germ-cell nests. Shortly after birth, these germ-cell nests break apart, and the oocytes individually become surrounded by somatic granulosa cells to form primordial follicles. Notch signaling plays essential roles during oogenesis in Drosophila, and recent studies have suggested that Notch signaling also plays an essential role during oogenesis and ovary development in mammals. However, no in vivo loss-of-function studies have been performed to establish whether Notch family receptors have an essential physiological role during normal ovarian development in mutant mice. Results Female mice with conditional deletion of the Notch2 gene in somatic granulosa cells of the ovary exhibited reduced fertility, accompanied by the formation of multi-oocyte follicles, which became hemorrhagic by 7 weeks of age. Formation of multi-oocyte follicles resulted from defects in breakdown of the primordial germ-cell nests. The ovaries of the Notch2 conditional mutant mice had increased numbers of oocytes, but decreased numbers of primordial follicles. Oocyte numbers in the Notch2 conditional mutants were increased not by excess or extended cellular proliferation, but as a result of decreased oocyte apoptosis. Conclusions Our work demonstrates that Notch2-mediated signaling in the somatic-cell lineage of the mouse ovary regulates oocyte apoptosis non-cell autonomously, and is essential for regulating breakdown of germ-cell nests and formation of primordial follicles. This model provides a new resource for studying the developmental and physiological roles of Notch signaling during mammalian reproductive biology.
Collapse
Affiliation(s)
- Jingxia Xu
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
45
|
Albamonte MI, Albamonte MS, Stella I, Zuccardi L, Vitullo AD. The infant and pubertal human ovary: Balbiani's body-associated VASA expression, immunohistochemical detection of apoptosis-related BCL2 and BAX proteins, and DNA fragmentation. Hum Reprod 2013; 28:698-706. [PMID: 23315064 DOI: 10.1093/humrep/des453] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION How do apoptosis-related BCL2 and BAX genes, known to regulate death or survival of germ cells in fetal and adult life, and germ-cell-specific VASA protein behave from birth to puberty in the human ovary? SUMMARY ANSWER In resting primordial follicles in both infant and pubertal ovaries, BCL2 family members and germ-cell-specific VASA behave as in fetal life. After birth, once follicles leave the resting reserve to enter the growing follicular pool, detection of apoptosis-related genes moves from the germ cell to granulosa cells and VASA expression is lost. WHAT IS KNOWN ALREADY In the human ovary, around 85% of the 7 × 10⁶ potential oocytes produced at mid-gestation are lost before birth, an extra 10% before puberty, and loss continues throughout reproductive life until germinal exhaustion of the ovary. Oocyte loss is mainly driven through a balanced expression of BCL2 gene family members. Apoptosis-inducing BAX gene shows a sustained expression throughout fetal and adult life, whereas apoptosis-inhibiting BCL2 is detectable during the proliferative stage of primordial germ cells and oogonia in the fetal ovary and proliferation of granulosa cells in growing follicles in the adult ovary. The germ-cell marker VASA is detectable in the fetal ovary from early oogenesis and is conspicuously expressed in primordial follicles, where in late pregnancy it is associated with the Balbiani's vitelline space. VASA expression is not detectable in the adult ovary. STUDY DESIGN, SIZE, DURATION This is a qualitative analysis involving infant/pubertal paraffin-embedded human ovaries screened for apoptosis-related proteins, DNA fragmentation and germ-cell identity. PARTICIPANTS/MATERIALS, SETTING, METHODS Ovaries from 13 patients ranging in age from 4 to 16 years, undergoing gynaecological surgical procedures due to benign pathology, were studied. Tissues were fixed in 10% formalin, paraffin-embedded and processed for immunohistochemistry to screen the temporal and cellular localization of germ-cell-specific VASA protein and BCL2 and BAX apoptosis-related proteins. In addition, a terminal deoxynucleotidyl transferase-mediated deoxiuridinetriphosphate nick-end labelling (TUNEL) assay was performed to detect DNA fragmentation. General histology and tissue integrity were assessed by haematoxylin-eosin staining. MAIN RESULTS AND THE ROLE OF CHANCE VASA showed a differential pattern of expression; in the resting primordial follicle reserve in infant and pubertal ovaries, it was associated with the Balbiani's body space in the germ cell. VASA remained detectable in primary follicles leaving the resting reserve, but once follicles entered the growing pool it became undetectable. This pattern of VASA expression is the same as in the fetal ovary. BAX was expressed both in the resting primordial reserve and in the pool of growing follicles, whereas BCL2 was detected only in granulosa cells in antral follicles in the growing pool. Apoptosis-related protein expression moved from the germ cell to the somatic stratum when primordial follicles left the resting reserve to enter the pool of growing follicles, irrespective of female age. Most TUNEL-positive cells were detected in the granulosa cells of antral follicles. No TUNEL-positive cells were found in resting primordial follicles. LIMITATIONS, REASONS FOR CAUTION The study was limited by the qualitative nature of the immuno-histochemical analysis and the TUNEL assay. The results neither quantify the levels of germ-cell death nor exclude other concurrent cell death mechanisms that could act in the regulation of female germ-cell number. WIDER IMPLICATIONS OF THE FINDING This study provides missing knowledge about apoptosis and germ-cell-specific VASA expression in the human ovary between birth and puberty and the participation of BCL2 and BAX genes in the balance between death and survival throughout female germ-line development. Intracellular localization of VASA in resting follicles emerges as a possible marker with prognostic value that needs further investigation, especially in infant patients entering ovarian cryo-preservation programmes. This knowledge will be valuable in optimizing the rescue and clinical use of germ cells to restore fertility in women. STUDY FUNDING/COMPETING INTEREST(S) No external funding was obtained for this study. The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- María Itatí Albamonte
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
46
|
Pangas SA. Regulation of the ovarian reserve by members of the transforming growth factor beta family. Mol Reprod Dev 2012; 79:666-79. [PMID: 22847922 DOI: 10.1002/mrd.22076] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 07/13/2012] [Indexed: 11/08/2022]
Abstract
Genetic or environmental factors that affect the endowment of oocytes, their assembly into primordial follicles, or their subsequent entry into the growing follicle pool can disrupt reproductive function and may underlie disorders such as primary ovarian insufficiency. Mouse models have been instrumental in identifying genes important in ovarian development, and a number of genes now associated with ovarian dysfunction in women were first identified as causing reproductive defects in knockout mice. The transforming growth factor beta (TGFB) family consists of developmentally important growth factors that include the TGFBs, anti-Müllerian hormone (AMH), activins, bone morphogenetic proteins (BMPs), and growth and differentiation factor 9 (GDF9). The ovarian primordial follicle pool is the source of oocytes in adults. Development of this pool can be grossly divided into three key processes: (1) establishment of oocytes during embryogenesis followed by (2) assembly and (3) activation of the primordial follicle. Disruptions in any of these processes may cause reproductive dysfunction. Most members of the TGFB family show pivotal roles in each of these areas. Understanding the phenotypes of various mouse models for this protein family will be directly relevant to understanding how disruptions in TGFB family signaling result in reproductive diseases in women and will present new areas for development of tailored diagnostics and interventions for infertility.
Collapse
Affiliation(s)
- Stephanie A Pangas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
47
|
Weghofer A, Kim A, Barad DH, Gleicher N. The impact of androgen metabolism and FMR1 genotypes on pregnancy potential in women with dehydroepiandrosterone (DHEA) supplementation. Hum Reprod 2012; 27:3287-93. [PMID: 22811313 DOI: 10.1093/humrep/des265] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND For decades androgens have been considered detrimental to follicle maturation. Animal studies now suggest that they are essential for normal folliculogenesis. Especially in women with premature ovarian aging (POA), recent IVF data in humans are supportive. The literature also suggests an association between recently reported ovarian genotypes of the FMR1 gene and ovarian aging patterns. We, therefore, attempted to determine a potential difference in androgen concentrations and androgen interactions in women with POA who do or do not become pregnant while undergoing androgen supplementation, and whether androgen concentrations and pregnancy chances are affected by FMR1 genotypes. METHODS We longitudinally assessed androgen metabolism in 91 women with POA, following pre-supplementation with micronized dehydroepiandrosterone (DHEA) prior to IVF. IVF outcomes were assessed based on androgen levels and ovarian FMR1 genotypes. RESULTS The mean age of the women was 39.8 ± 4.4 years; the clinical pregnancy rate was 25.3%. Total androgen concentrations were not associated with pregnancy; however, in women with abnormal FMR1 genotypes, but not those with the normal genotype, free testosterone significantly affected clinical pregnancy potential (β = 1.101, SE ± 0.508, P = 0.03). At the start of the IVF cycle, interactions of DHEA with total and free testosterone also significantly affected subsequent pregnancy rates (β = -0.058, SE ± 0.023, P = 0.01 and β = -0.496, SE ± 0.197, P = 0.012). CONCLUSIONS Androgen interactions significantly influence IVF pregnancy rates in women with POA, with the impact of total androgens on cycle outcomes varying according to FMR1 genotypes. These observations suggest that the effectiveness of androgen supplementation in women with POA varies based on FMR1 genotypes, and defines androgen deficiency as a subset of diminished ovarian reserve.
Collapse
Affiliation(s)
- Andrea Weghofer
- Department of Obstetrics and Gynecology, Medical University Vienna, Waehringer Guertel 18-20, Vienna A-1090, Austria.
| | | | | | | |
Collapse
|