1
|
Lima EA, Carvalho LR, Orlandi RE, Simões LMS, Bottino MP, Santos APC, de Oliveira Scarpa F, Sales JNDS. Effect of maternal heat stress at different stages of pregnancy on the reproductive performance and antral follicle count of the progeny of Holstein cows. Anim Reprod Sci 2025; 272:107665. [PMID: 39667294 DOI: 10.1016/j.anireprosci.2024.107665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/08/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
The objective was to evaluate the effect of maternal exposure to heat stress conditions at different stages of pregnancy on the reproductive performance and antral follicle count of progeny. The study was conducted at five commercial dairy farms located in southeastern Brazil. Retrospective data (2002 and 2015) on reproductive indices of daughters of Holstein cows that were exposed to heat stress conditions in the first (n = 447), second (n = 729) or third (n = 746) trimester of pregnancy and daughters of cows not exposed to heat stress (n = 359) were analyzed. Antral follicle count (AFC) and ovarian size were determined in a subgroup of animals (n = 242) by transrectal ultrasound. Statistical analysis was performed using the SAS GLIMMIX procedure. The number of services after first calving was lower in daughters of cows not exposed to heat stress conditions than in those exposed during different trimesters of pregnancy (P = 0.03). The calving interval was longer for daughters of cows exposed to heat stress conditions in the second and third trimesters of pregnancy than for those not exposed to heat stress (P = 0.01). In addition, the probability of pregnancy at first AI, (P = 0.01) and the AFC (P = 0.001) were greater in daughters of cows not exposed to heat stress conditions than in those exposed during the different phases of the gestational period. These findings indicate that maternal heat stress in different stages of pregnancy negatively affect the future fertility and AFC of bovine offspring.
Collapse
Affiliation(s)
- Eduardo Alves Lima
- Faculty of Animal Science and Veterinary Medicine, UFLA, Lavras, MG 37200-900, Brazil
| | - Laís Reis Carvalho
- Faculty of Animal Science and Veterinary Medicine, UFLA, Lavras, MG 37200-900, Brazil.
| | | | | | | | | | | | - Jose Nelio de Sousa Sales
- Faculty of Animal Science and Veterinary Medicine, UFLA, Lavras, MG 37200-900, Brazil; Department of Veterinary Medicine, UFJF, Juiz de Fora, MG 36036-900, Brazil
| |
Collapse
|
2
|
Torkzadeh T, Asadi Z, Jafari Atrabi M, Khodadi M, Eivazkhani F, Hajiaghalou S, Akbarinejad V, Fathi R. Combination of FSH and testosterone could enhance activation of primordial follicles and growth of activated follicles in 1-day-old mice ovaries in vitro cultured for 12 days. ZYGOTE 2024:1-9. [PMID: 39710995 DOI: 10.1017/s0967199424000479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Treatment with follicle-stimulating hormone (FSH) and testosterone (T2) and their combination have been observed to be influential on ovarian follicles of 1-day-old mice ovaries cultured for 8 days. Given that extension of the culture period could positively impact the development of follicles in cultured ovaries, the present study was conducted to evaluate the main and interaction effects of FSH by T2 on the development of ovarian follicles in 1-day-old mice ovaries cultured for 12 days. One-day-old mice ovaries were initially cultured with base medium for 4 days; thereafter, different hormonal treatments were added to the culture media, and the culture was continued for 8 additional days until day 12. Ovaries were collected for histological and molecular assessments on day 12. The greatest activation of primordial follicles and progression of activated follicles to the preantral stage was detected in ovaries treated with the combination of FSH and T2 (P < 0.05). This positive effect on the morphology of ovarian follicles was accompanied by upregulation of Pi3k, Gdf9, Bmp15, Cx37 and Fshr in the ovaries cultured with the combination of FSH and T2 (P < 0.05). Nonetheless, treatment with FSH and T2 led to a diminished proportion of intact follicles (P < 0.05), even though Bax/Bcl2 gene expression ratio, as an apoptotic index, was less in hormone-treated ovaries (P < 0.05). In conclusion, the combination of FSH and T2 could improve the activation of primordial follicles and the growth of activated follicles towards the preantral stage. This positive effect of FSH plus T2 appeared to be at least partly mediated through the upregulation of Pi3k and oocyte-derived growth factors including Gdf9 and Bmp15.
Collapse
Affiliation(s)
- Tahoura Torkzadeh
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zahra Asadi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73014, USA
| | - Mohammad Jafari Atrabi
- Institute of Pharmacology and Toxicology, University Medical Center, Georg August University, Göttingen, Germany
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research (DPZ), Göttingen, Germany
| | - Maryam Khodadi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farideh Eivazkhani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Samira Hajiaghalou
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
3
|
Baldini GM, Lot D, Malvasi A, Laganà AS, Vimercati A, Dellino M, Cicinelli E, Baldini D, Trojano G. Abnormalities of Oocyte Maturation: Mechanisms and Implications. Int J Mol Sci 2024; 25:12197. [PMID: 39596263 PMCID: PMC11595025 DOI: 10.3390/ijms252212197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
The elucidation of oocyte maturation mechanisms is paramount for advancing embryo development within the scope of assisted reproductive technologies (ART). Both cytoplasmic and nuclear maturation represent intricate processes governed by tightly regulated cellular pathways, which are essential for ensuring the oocyte's competence for fertilization and subsequent embryogenesis. A comprehensive grasp of these mechanisms is vital, as the maturation stage of the oocyte significantly influences chromosomal integrity, spindle formation, and its ability to support the initial stages of embryonic development. By leveraging this knowledge, we can enhance in vitro fertilization (IVF) protocols, refining ovarian stimulation regimens and culture conditions to improve oocyte quality. This, in turn, has the potential to boost pregnancy rates and outcomes. Further research in this area will contribute to the development of novel interventions that aim to increase the efficacy of preimplantation embryonic development, offering new opportunities for individuals undergoing fertility treatments.
Collapse
Affiliation(s)
- Giorgio Maria Baldini
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70121 Bari, Italy; (G.M.B.); (A.M.); (A.V.); (M.D.); (E.C.)
| | - Dario Lot
- IVF Center, Momo Fertilife, 76011 Bisceglie, Italy;
| | - Antonio Malvasi
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70121 Bari, Italy; (G.M.B.); (A.M.); (A.V.); (M.D.); (E.C.)
| | - Antonio Simone Laganà
- Unit of Obstetrics and Gynecology “Paolo Giacone” Hospital, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities (PROMISE), University of Palermo, 90135 Palermo, Italy;
| | - Antonella Vimercati
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70121 Bari, Italy; (G.M.B.); (A.M.); (A.V.); (M.D.); (E.C.)
| | - Miriam Dellino
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70121 Bari, Italy; (G.M.B.); (A.M.); (A.V.); (M.D.); (E.C.)
| | - Ettore Cicinelli
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70121 Bari, Italy; (G.M.B.); (A.M.); (A.V.); (M.D.); (E.C.)
| | | | - Giuseppe Trojano
- Department of Maternal and Child Health, Madonna delle Grazie Hospital, 75010 Matera, Italy;
| |
Collapse
|
4
|
Balali H, Morabbi A, Karimian M. Concerning influences of micro/nano plastics on female reproductive health: focusing on cellular and molecular pathways from animal models to human studies. Reprod Biol Endocrinol 2024; 22:141. [PMID: 39529078 PMCID: PMC11552210 DOI: 10.1186/s12958-024-01314-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
The female reproductive system can face serious disorders and show reproductive abnormalities under the influence of environmental pollutants. Microplastics (MPs) and nanoplastics (NPs) as emerging pollutants, by affecting different components of this system, may make female fertility a serious challenge. Animal studies have demonstrated that exposure to these substances weakens the function of ovaries and causes a decrease in ovarian reserve capacity. Also, continuous exposure to micro/nano plastics (MNPs) leads to increased levels of reactive oxygen species, induction of oxidative stress, inflammatory responses, apoptosis of granulosa cells, and reduction of the number of ovarian follicles. Furthermore, by interfering with the hypothalamic-pituitary-ovarian axis, these particles disturb the normal levels of ovarian androgens and endocrine balance and delay the growth of gonads. Exposure to MNPs can accelerate carcinogenesis in the female reproductive system in humans and animal models. Animal studies have determined that these particles can accumulate in the placenta, causing metabolic changes, disrupting the development of the fetus, and endangering the health of future generations. In humans, the presence of micro/nanoplastics in placenta tissue, infant feces, and breast milk has been reported. These particles can directly affect the health of the mother and fetus, increasing the risk of premature birth and other pregnancy complications. This review aims to outline the hazardous effects of micro/nano plastics on female reproductive health and fetal growth and discuss the results of animal experiments and human research focusing on cellular and molecular pathways.
Collapse
Affiliation(s)
- Hasti Balali
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran
| | - Ali Morabbi
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran.
| |
Collapse
|
5
|
Abazarikia A, So W, Xiao S, Kim SY. Oocyte death is triggered by the stabilization of TAp63α dimers in response to cisplatin. Cell Death Dis 2024; 15:799. [PMID: 39511162 PMCID: PMC11544165 DOI: 10.1038/s41419-024-07202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
The TAp63α protein is highly expressed in primordial follicle oocytes, where it typically exists in an inactive dimeric form. Upon DNA damage, TAp63α undergoes hyperphosphorylation, transitioning from a dimeric to a tetrameric structure, which initiates oocyte apoptosis by upregulating pro-apoptotic gene. Our results demonstrate that cisplatin, an alkylating anti-cancer agent, predominantly produced the TAp63α dimer rather than the tetramer. We further observed that TAp63α protein accumulation occurred in primordial follicle oocytes following cisplatin treatment, and this accumulation was significantly reduced by cycloheximide, a protein synthesis inhibitor. These findings suggest that TAp63α accumulation is driven primarily by de novo protein synthesis in response to DNA damage. Notably, cycloheximide protected oocytes from cisplatin-induced apoptosis, as evidenced by reduced levels of both PUMA, a known pro-apoptotic target gene of TAp63α, and TAp63α itself. Additionally, TAp63α turnover appears to be regulated by ubiquitination and proteasome degradation, as evidenced by TAp63α accumulation without oocyte death when treated with PYR-41, a pharmacological inhibitor. However, when TAp63α was stabilized by PYR-41 and subsequently activated by cisplatin, oocyte death occurred, marked by increased γH2AX and Cleaved PARP. Moreover, the Casein kinase 1 inhibitor PF-670462 effectively blocked cisplatin-induced oocyte death, indicating that CK1-mediated phosphorylation is essential for TAp63α activation, even in the absence of tetramer formation. The ATR inhibitor BEZ235 prevented cisplatin-induced TAp63α accumulation, suggesting that TAp63α accumulation precedes its phosphorylation-driven activation. Collectively, our study reveals a novel mechanism of cisplatin-induced apoptosis in primordial follicle oocyte through TAp63α stabilization and accumulation, independent of tetramerization.
Collapse
Affiliation(s)
- Amirhossein Abazarikia
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wonmi So
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers Unversity, Piscataway, NJ, USA
| | - So-Youn Kim
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
6
|
Dong MZ, Ouyang YC, Gao SC, Gu LJ, Guo JN, Sun SM, Wang ZB, Sun QY. Protein phosphatase 4 maintains the survival of primordial follicles by regulating autophagy in oocytes. Cell Death Dis 2024; 15:658. [PMID: 39245708 PMCID: PMC11381532 DOI: 10.1038/s41419-024-07051-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
In mammalian ovary, the primordial follicle pool serves as the source of developing follicles and fertilizable ova. To maintain the normal length of female reproductive life, the primordial follicles must have adequate number and be kept in a quiescent state before menopause. However, the molecular mechanisms underlying primordial follicle survival are poorly understood. Here, we provide genetic evidence showing that lacking protein phosphatase 4 (PPP4) in oocytes, a member of PP2A-like subfamily, results in infertility in female mice. A large quantity of primordial follicles has been depleted around the primordial follicle pool formation phase and the ovarian reserve is exhausted at about 7 months old. Further investigation demonstrates that depletion of PPP4 causes the abnormal activation of mTOR, which suppresses autophagy in primordial follicle oocytes. The abnormal primordial follicle oocytes are eventually erased by pregranulosa cells in the manner of lysosome invading. These results show that autophagy prevents primordial follicles over loss and PPP4-mTOR pathway governs autophagy during the primordial follicle formation and dormant period.
Collapse
Affiliation(s)
- Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shi-Cai Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lin-Jian Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia-Ni Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si-Min Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
| |
Collapse
|
7
|
Alarcón R, Alegre AL, Rivera O, Dioguardi G, Zenclussen ML, Muñoz-de-Toro M, Luque EH, Ingaramo PI. Altered ovarian reserve in Ewe lambs exposed to a glyphosate-based herbicide. CHEMOSPHERE 2024; 363:142895. [PMID: 39067823 DOI: 10.1016/j.chemosphere.2024.142895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
Glyphosate-based herbicides (GBHs) are considered endocrine disruptors that affect the female reproductive tract of rats and ewe lambs. The present study aimed to investigate the impact of neonatal exposure to a low dose of a GBH on the ovarian follicular reserve of ewe lambs and the response to a gonadotropic stimulus with porcine FSH (pFSH). To this end, ewe lambs were orally exposed to an environmentally relevant GBH dose (1 mg/kg/day) or vehicle (Control) from postnatal day (PND) 1 to PND14, and then some received pFSH (50 mg/day) between PND41 and 43. The ovaries were dissected, and follicular types and gene expression were assessed via RT-PCR. The treatments did not affect the body weight of animals, but pFSH increased ovarian weight, not observed in GBH-exposed lambs. GBH-exposed lambs showed decreased Estrogen receptor-alpha (56%), Progesterone receptor (75%), Activin receptor II (ACVRII) (85%), and Bone morphogenetic protein 15 (BMP15) (88%) mRNA levels. Control lambs treated with pFSH exhibited downregulation of Follistatin (81%), ACVRII (77%), BMP15 (93%), and FSH receptor (FSHr) (72%). GBH-exposed lambs treated with pFSH displayed reduced ACVRII (68%), BMP15 (81%), and FSHr (50%). GBH-exposed lambs also exhibited decreased Anti-Müllerian hormone expression in primordial and antral follicles (27%) and (54%) respectively) and reduced Bone morphogenetic protein 4 (31%) expression in primordial follicles. Results suggest that GBH disrupts key follicular development molecules and interferes with pFSH action in ovarian receptors, decreasing the ovarian reserve. Future studies should explore whether this decreased ovarian reserve impairs adult ovarian function and its response to superovulation stimuli.
Collapse
Affiliation(s)
- Ramiro Alarcón
- Instituto de Salud y Ambiente del Litoral (ISAL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Bioquímica y Ciencias Biológicas Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ana Laura Alegre
- Instituto de Salud y Ambiente del Litoral (ISAL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Bioquímica y Ciencias Biológicas Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Oscar Rivera
- Instituto de Investigación sobre Producción Agropecuaria, Ambiente y Salud (IIPAAS), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - Gisela Dioguardi
- Instituto de Investigación sobre Producción Agropecuaria, Ambiente y Salud (IIPAAS), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - María Laura Zenclussen
- Instituto de Salud y Ambiente del Litoral (ISAL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Bioquímica y Ciencias Biológicas Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Bioquímica y Ciencias Biológicas Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Bioquímica y Ciencias Biológicas Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Paola I Ingaramo
- Instituto de Salud y Ambiente del Litoral (ISAL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Bioquímica y Ciencias Biológicas Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
8
|
Winship AL, Alesi LR, Stringer JM, Cao Y, Lewis YM, Tu L, Swindells EOK, Giridharan S, Cai X, Griffiths MJ, Zerafa N, Gilham L, Hickey M, Hutt KJ. Conditional loss of Brca1 in oocytes causes reduced litter size, ovarian reserve depletion and impaired oocyte in vitro maturation with advanced reproductive age in mice. EBioMedicine 2024; 106:105262. [PMID: 39084071 PMCID: PMC11342213 DOI: 10.1016/j.ebiom.2024.105262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND An estimated 1 in 350 women carry germline BRCA1/2 mutations, which confer an increased risk of developing breast and ovarian cancer, and may also contribute to subfertility. All mature, sex steroid-producing ovarian follicles are drawn from the pool of non-renewable primordial follicles, termed the 'ovarian reserve'. The clinical implications of early ovarian reserve exhaustion extend beyond infertility, to include the long-term adverse health consequences of loss of endocrine function and premature menopause. We aimed to determine whether conditional loss of Brca1 in oocytes impacts ovarian follicle numbers, oocyte quality and fertility in mice with advancing maternal age. We also aimed to determine the utility of AMH as a marker of ovarian function, by assessing circulating AMH levels in mice and women with BRCA1/2 mutations, and correlating this with ovarian follicle counts. METHODS In this study, we addressed a longstanding question in the field regarding the functional consequences of BRCA1 inactivation in oocytes. To recapitulate loss of BRCA1 protein function in oocytes, we generated mice with conditional gene deletion of Brca1 in oocytes using Gdf9-Cre recombinase (WT: Brca1fl/flGdf9+/+; cKO: Brca1fl/flGdf9cre/+). FINDINGS While the length of the fertile lifespan was not altered between groups after a comprehensive breeding trial, conditional loss of Brca1 in oocytes led to reduced litter size in female mice. Brca1 cKO animals had a reduced ovarian reserve and oocyte maturation was impaired with advanced maternal age at postnatal day (PN)300, compared to WT animals. Serum anti-Müllerian hormone (AMH) concentrations (the gold-standard indirect marker of the ovarian reserve used in clinical practice) were not predictive of reduced primordial follicle number in Brca1 cKO mice versus WT. Furthermore, we found no correlation between follicle number or density and serum AMH concentrations in matched samples from a small cohort of premenopausal women with BRCA1/2 mutations. INTERPRETATION Together, our data demonstrate that BRCA1 is a key regulator of oocyte number and quality in females and suggest that caution should be used in relying on AMH as a reliable marker of the ovarian reserve in this context. FUNDING This work was made possible through Victorian State Government Operational Infrastructure Support and Australian Government NHMRC IRIISS. This work was supported by funding from the Australian Research Council (ALW - DE21010037 and KJH - FT190100265), as well as the National Breast Cancer Foundation (IIRS-22-092) awarded to ALW and KJH. LRA, YML, LT, EOKS and MG were supported by Australian Government Research Training Program Scholarships. LRA, YML and LT were also supported by a Monash Graduate Excellence Scholarship. YC, SG and XC were supported by Monash Biomedicine Discovery Institute PhD Scholarships. LRA was also supported by a Monash University ECPF24-6809920940 Fellowship. JMS was supported by NHMRC funding (2011299). MH was supported by an NHMRC Investigator Grant (1193838).
Collapse
Affiliation(s)
- Amy L Winship
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia.
| | - Lauren R Alesi
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Jessica M Stringer
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Yujie Cao
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Yasmin M Lewis
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Lisa Tu
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Elyse O K Swindells
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Saranya Giridharan
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Xuebi Cai
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Meaghan J Griffiths
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia; University of Edinburgh, MRC Centre for Reproductive Health, Queens Medical Research Institute, Edinburgh, UK
| | - Nadeen Zerafa
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Leslie Gilham
- Breast Cancer Network Australia and Breast Cancer Trials Australia, Camberwell, VIC, Australia
| | - Martha Hickey
- Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
| | - Karla J Hutt
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
9
|
Li S, Wang Q, Mi J, Chen H, Yuan T, Wang Y, Zhao L, Ma Q, Huang S. Lactobacillus crispatus-Mediated Gut-Reproductive Tract Axis-Alleviated Microbial Dysbiosis and Oviductal Inflammation in a Laying Hen Model. Microorganisms 2024; 12:1559. [PMID: 39203401 PMCID: PMC11356123 DOI: 10.3390/microorganisms12081559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Oviductal inflammation (OI) significantly reduces the egg production and economic returns in poultry farming. While Lactobacillus crispatus (LAC) is effective against inflammation, its role in treating or preventing oviductal inflammation is understudied. In this study, we investigated the therapeutic mechanisms of LAC on oviductal inflammation, with a focus on reproductive tract health, microbiome, gene expression, and cytokine levels. This study involved 24 Jingfen No. 6 laying hens aged 60 weeks, divided into four groups: the CON, OI, OI + LAC, and OI + heat-killed Lactobacillus crispatus (HLAC) groups. And it included a 10-day adaptation, a 7-day period for the development of OI using inflammation-inducing drugs (the control received saline), followed by an 8-day treatment in which the CON and OI groups received 1 mL of MRS broth daily, and the OI + LAC and OI + HLAC groups were treated with live and heat-killed Lactobacillus crispatus (109 CFUs/mL), respectively, with six hens in each group. This study showed that Lactobacillus crispatus supplementation significantly reduced the oviductal inflammation and atrophy in the hens, with the affected hens showing markedly lower egg production rates (p < 0.001) compared to the control and treated groups (OI + HLAC and OI + LAC). The daily intake of fresh (OI + LAC, p = 0.076) or heat-killed (OI + HLAC, p < 0.01) Lactobacillus crispatus notably enhanced the feed conversion efficiency. The OI group suffered significant ovarian damage and vascular rupture, more so than the CON group, while Lactobacillus crispatus supplementation mitigated this damage. The IL-1β, IL-6, and IL-8 levels were significantly elevated in the OI group compared to those in the OI + LAC group (p < 0.05), with a significant reduction in the TNF-α levels in the latter (p < 0.001). The supplementation improved the microbial composition in the cecum, isthmus, and shell gland, enriching the cecum with beneficial bacteria, such as Ruminococcus_torques_group and Megamonas. This approach fostered ovarian health and follicle differentiation and preserved the epithelial cell barrier function in the shell gland, reducing inflammatory damage in the genital tract. This dual efficacy underscores the role of the probiotic in diminishing oviductal inflammation, regardless of its state.
Collapse
Affiliation(s)
- Shinuo Li
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Qingfeng Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Jinqiu Mi
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Haotian Chen
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Tianhao Yuan
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Yue Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Lihong Zhao
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Qiugang Ma
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| | - Shimeng Huang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (S.L.); (Q.W.); (T.Y.); (Y.W.); (L.Z.)
- Laboratory of Feedgrain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing 101206, China
| |
Collapse
|
10
|
Li F, Zhu F, Wang S, Hu H, Zhang D, He Z, Chen J, Li X, Cheng L, Zhong F. Icariin alleviates cisplatin-induced premature ovarian failure by inhibiting ferroptosis through activation of the Nrf2/ARE pathway. Sci Rep 2024; 14:17318. [PMID: 39068256 PMCID: PMC11283570 DOI: 10.1038/s41598-024-67557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
Cisplatin is a widely used chemotherapeutic drug that can induce ovarian damage. Icariin (ICA), a natural antioxidant derived from Epimedium brevicornum Maxim., has been found to protect against organ injury. The aim of the present study was to investigate whether ICA can exert an ovarian-protective effect on cisplatin induced premature ovarian failure (POF) and the underlying mechanism involved. The preventive effect of ICA was evaluated using body weight, the oestrous cycle, ovarian histological analysis, and follicle counting. ICA treatment increased body weight, ovarian weight, and the number of follicles and improved the oestrous cycle in POF mice. ICA reduced cisplatin-induced oxidative damage and upregulated the protein expression levels of Nrf2, GPX4 and HO-1. Moreover, ICA reduced the expression levels of Bax and γH2AX and inhibited ovarian apoptosis. In addition, ICA activated the Nrf2 pathway in vitro and reversed changes in the viability of cisplatin-induced KGN cells, reactive oxygen species (ROS) levels, lipid peroxidation, and apoptosis, and these effects were abrogated when Nrf2 was knocked down or inhibited. Molecular docking confirmed that ICA promotes the release of Nrf2 by competing with Nrf2 for binding to Keap1. The inhibitory effects of ICA on cisplatin-induced oxidative stress, ferroptosis, and apoptosis may be mediated by its modulatory effects on the Nrf2 pathway, providing a novel perspective on the potential mechanisms by which ICA prevents POF.
Collapse
Affiliation(s)
- Fangfang Li
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fengyu Zhu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Ministry of Education of the People's Republic of China, Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Siyuan Wang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Huiqing Hu
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Di Zhang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhouying He
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jiaqi Chen
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xuqing Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Ministry of Education of the People's Republic of China, Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Linghui Cheng
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Ministry of Education of the People's Republic of China, Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
11
|
Cakir C, Kuspinar G, Aslan K, Bozyigit C, Kasapoglu I, Dirican M, Uncu G, Avci B. Dehydroepiandrosterone modulates the PTEN/PI3K/AKT signaling pathway to alleviate 4-vinylcyclohexene diepoxide-induced premature ovarian insufficiency in rats. Exp Anim 2024; 73:319-335. [PMID: 38494723 PMCID: PMC11254495 DOI: 10.1538/expanim.23-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/09/2024] [Indexed: 03/19/2024] Open
Abstract
Dehydroepiandrosterone (DHEA) is frequently integrated as an adjuvant in over a quarter of controlled ovarian hyperstimulation (COH) protocols, despite the ongoing debate regarding its impact. This study aimed to evaluate the efficacy and mechanism of action of DHEA on ovarian follicular development and ovarian response in rats with varying ovarian reserves. The study involved 75 rats categorized into 15 distinct groups. The ovarian tissues of rats in both the normal ovarian reserve group and the premature ovarian insufficiency (POI) group, induced by 4-vinylcyclohexene diepoxide (VCD) injection, were subjected to histomorphological and biochemical analyses following the administration of DHEA, either alone or in combination with COH. Follicle counting was performed on histological sections obtained from various tissues. Serum concentrations of anti-Müllerian hormone (AMH) and the quantification of specific proteins in ovarian tissue, including phosphatase and tensin homolog of chromosome 10 (PTEN), phosphoinositide 3-kinase (PI3K), phosphorylated protein kinase B (pAKT), cyclooxygenase 2 (COX-2), caspase-3, as well as assessments of total antioxidant status and total oxidant status, were conducted employing the ELISA method. The impact of DHEA exhibited variability based on ovarian reserve. In the POI model, DHEA augmented follicular development and ovarian response to the COH protocol by upregulating the PTEN/PI3K/AKT signaling pathway, mitigating apoptosis, inflammation, and oxidative stress, contrary to its effects in the normal ovarian reserve group. In conclusion, it has been determined that DHEA may exert beneficial effects on ovarian stimulation response by enhancing the initiation of primordial follicles and supporting antral follicle populations.
Collapse
Affiliation(s)
- Cihan Cakir
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Goktan Kuspinar
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Kiper Aslan
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Cengiz Bozyigit
- Department of Medical Biochemistry, Bursa City Hospital, Doğanköy District, Nilüfer Bursa, 16110, Türkiye
| | - Isil Kasapoglu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Melahat Dirican
- Department of Medical Biochemistry, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Gurkan Uncu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Berrin Avci
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| |
Collapse
|
12
|
Song C, Qin Y, Li Y, Yang B, Guo T, Ma W, Xu D, Xu K, Fu F, Jin L, Wu Y, Tang S, Chen X, Zhang F. Deleterious variants in RNF111 impair female fertility and induce premature ovarian insufficiency in humans and mice. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1325-1337. [PMID: 38874713 DOI: 10.1007/s11427-024-2606-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 06/15/2024]
Abstract
Premature ovarian insufficiency (POI) is a heterogeneous female disorder characterized by the loss of ovarian function before the age of 40. It represents a significant detriment to female fertility. However, the known POI-causative genes currently account for only a fraction of cases. To elucidate the genetic factors underlying POI, we conducted whole-exome sequencing on a family with three fertile POI patients and identified a deleterious missense variant in RNF111. In a subsequent replication study involving 1,030 POI patients, this variant was not only confirmed but also accompanied by the discovery of three additional predicted deleterious RNF111 variants. These variants collectively account for eight cases, representing 0.78% of the study cohort. A further study involving 500 patients with diminished ovarian reserve also identified two additional RNF111 variants. Notably, RNF111 encodes an E3 ubiquitin ligase with a regulatory role in the TGF-β/BMP signaling pathway. Our analysis revealed that RNF111/RNF111 is predominantly expressed in the oocytes of mice, monkeys, and humans. To further investigate the functional implications of RNF111 variants, we generated two mouse models: one with a heterozygous missense mutation (Rnf111+/M) and another with a heterozygous null mutation (Rnf111+/-). Both mouse models exhibited impaired female fertility, characterized by reduced litter sizes and small ovarian reserve. Additionally, RNA-seq and quantitative proteomics analysis unveiled that Rnf111 haploinsufficiency led to dysregulation in female gonad development and negative regulation of the BMP signaling pathway within mouse ovaries. In conclusion, our findings strongly suggest that monoallelic deleterious variants in RNF111 can impair female fertility and induce POI in both humans and mice.
Collapse
Affiliation(s)
- Chengcheng Song
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Reproductive Endocrinology of Ministry of Education, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, 250021, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bingyi Yang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China
| | - Ting Guo
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Reproductive Endocrinology of Ministry of Education, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, 250021, China
| | - Wenqing Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dian Xu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Keyan Xu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Key Laboratory of Reproductive Endocrinology of Ministry of Education, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, 250021, China
| | - Fangfang Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Jin
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China
| | - Yanhua Wu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China
- National Demonstration Center for Experimental Biology Education, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Shuyan Tang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China
| | - Xiaojun Chen
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China.
- Department of Gynecology, the Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200011, China.
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203, China.
- Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, 200030, China.
| |
Collapse
|
13
|
Ballif G, Clément F, Yvinec R. Nonlinear compartmental modeling to monitor ovarian follicle population dynamics on the whole lifespan. J Math Biol 2024; 89:9. [PMID: 38844702 DOI: 10.1007/s00285-024-02108-6] [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: 07/18/2022] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 06/28/2024]
Abstract
In this work, we introduce a compartmental model of ovarian follicle development all along lifespan, based on ordinary differential equations. The model predicts the changes in the follicle numbers in different maturation stages with aging. Ovarian follicles may either move forward to the next compartment (unidirectional migration) or degenerate and disappear (death). The migration from the first follicle compartment corresponds to the activation of quiescent follicles, which is responsible for the progressive exhaustion of the follicle reserve (ovarian aging) until cessation of reproductive activity. The model consists of a data-driven layer embedded into a more comprehensive, knowledge-driven layer encompassing the earliest events in follicle development. The data-driven layer is designed according to the most densely sampled experimental dataset available on follicle numbers in the mouse. Its salient feature is the nonlinear formulation of the activation rate, whose formulation includes a feedback term from growing follicles. The knowledge-based, coating layer accounts for cutting-edge studies on the initiation of follicle development around birth. Its salient feature is the co-existence of two follicle subpopulations of different embryonic origins. We then setup a complete estimation strategy, including the study of structural identifiability, the elaboration of a relevant optimization criterion combining different sources of data (the initial dataset on follicle numbers, together with data in conditions of perturbed activation, and data discriminating the subpopulations) with appropriate error models, and a model selection step. We finally illustrate the model potential for experimental design (suggestion of targeted new data acquisition) and in silico experiments.
Collapse
Affiliation(s)
- Guillaume Ballif
- Inria, Centre Inria de Saclay, Université Paris-Saclay, 91120, Palaiseau, France.
| | - Frédérique Clément
- Inria, Centre Inria de Saclay, Université Paris-Saclay, 91120, Palaiseau, France
| | - Romain Yvinec
- Inria, Centre Inria de Saclay, Université Paris-Saclay, 91120, Palaiseau, France
- PRC, INRAE, CNRS, Université de Tours, 37380, Nouzilly, France
| |
Collapse
|
14
|
Folts L, Martinez AS, Bunce C, Capel B, McKey J. OoCount: A Machine-Learning Based Approach to Mouse Ovarian Follicle Counting and Classification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593993. [PMID: 38798456 PMCID: PMC11118501 DOI: 10.1101/2024.05.13.593993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The number and distribution of ovarian follicles in each growth stage provides a reliable readout of ovarian health and function. Leveraging techniques for three-dimensional (3D) imaging of ovaries in toto has the potential to uncover total, accurate ovarian follicle counts. However, because of the size and holistic nature of these images, counting oocytes is time consuming and difficult. The advent of deep-learning algorithms has allowed for the rapid development of ultra-fast, automated methods to analyze microscopy images. In recent years, these pipelines have become more user-friendly and accessible to non-specialists. We used these tools to create OoCount, a high-throughput, open-source method for automatic oocyte segmentation and classification from fluorescent 3D microscopy images of whole mouse ovaries using a deep-learning convolutional neural network (CNN) based approach. We developed a fast tissue-clearing and spinning disk confocal-based imaging protocol to obtain 3D images of whole mount perinatal and adult mouse ovaries. Fluorescently labeled oocytes from 3D images of ovaries were manually annotated in Napari to develop a machine learning training dataset. This dataset was used to retrain StarDist using a CNN within DL4MicEverywhere to automatically label all oocytes in the ovary. In a second phase, we utilize Accelerated Pixel and Object Classification, a Napari plugin, to classify labeled oocytes and sort them into growth stages. Here, we provide an end-to-end protocol for producing high-quality 3D images of the perinatal and adult mouse ovary, obtaining follicle counts and staging. We also demonstrate how to customize OoCount to fit images produced in any lab. Using OoCount, we can obtain accurate counts of oocytes in each growth stage in the perinatal and adult ovary, improving our ability to study ovarian function and fertility. Summary sentence This protocol introduces OoCount, a high-throughput, open-source method for automatic oocyte segmentation and classification from fluorescent 3D microscopy images of whole mouse ovaries using a machine learning-based approach.
Collapse
|
15
|
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
|
16
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [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: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
17
|
Lawley SD, Sammel MD, Santoro N, Johnson J. Mathematical recapitulation of the end stages of human ovarian aging. SCIENCE ADVANCES 2024; 10:eadj4490. [PMID: 38215196 PMCID: PMC10786411 DOI: 10.1126/sciadv.adj4490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024]
Abstract
Ovarian aging in women can be described as highly unpredictable within individuals but predictable across large populations. We showed previously that modeling an individual woman's ovarian reserve of primordial follicles using mathematical random walks replicates the natural pattern of growing follicles exiting the reserve. Compiling many simulations yields the observed population distribution of the age at natural menopause (ANM). Here, we have probed how stochastic control of primordial follicle loss might relate to the distribution of the preceding menopausal transition (MT), when women begin to experience menstrual cycle irregularity. We show that identical random walk model conditions produce both the reported MT distribution and the ANM distribution when thresholds are set for growing follicle availability. The MT and ANM are shown to correspond to gaps when primordial follicles fail to grow for 7 and 12 days, respectively. Modeling growing follicle supply is shown to precisely recapitulate epidemiological data and provides quantitative criteria for the MT and ANM in humans.
Collapse
Affiliation(s)
- Sean D. Lawley
- Department of Mathematics, University of Utah, 155 S 1400 E, JWB 233, Salt Lake City, UT 84112, USA
| | - Mary D. Sammel
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO 80045, USA
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine (AMC) Building RC2, Room P15 3103, Aurora, CO 80045, USA
| | - Nanette Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine (AMC) Building RC2, Room P15 3103, Aurora, CO 80045, USA
| | - Joshua Johnson
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine (AMC) Building RC2, Room P15 3103, Aurora, CO 80045, USA
| |
Collapse
|
18
|
Thomson EE, Beltman ME, Crowe MA, McAloon CG, Furlong JG, Duane MM, Brennan JPN, McDonald MM, Kelly ET. Association between maternal growth in the pre-conception and early gestational period of nulliparous dairy heifers with anti-Müllerian hormone in their female offspring. Reprod Domest Anim 2024; 59:e14498. [PMID: 37902253 DOI: 10.1111/rda.14498] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/10/2023] [Accepted: 10/18/2023] [Indexed: 10/31/2023]
Abstract
Anti-Müllerian hormone (AMH) is secreted by granulosa cells of healthy, growing follicles and is positively correlated with the ovarian reserve. Maternal and environmental factors, such as nutrition, disease, parity and endocrine disruptors, are thought to have a profound impact on ovarian reserve development during early foetal life. For genetic progress, it can be advantageous to breed dairy replacements from heifers to expedite the generation interval; however, there is some evidence that nulliparous animals produce female offspring with smaller ovarian reserves compared with multiparous animals. The objective of this prospective, observational study was to determine whether maternal growth in the pre-conception and early gestational period of nulliparous dairy heifers is associated with pre-weaning AMH concentrations in their female offspring. Our hypothesis was that excessive growth in this period would negatively impact AMH concentrations. Seasonal, pasture-based dairy heifer calves (n = 156) born from nulliparous dams, from six Irish farms, were blood sampled at an average of 60 days of age in spring 2022 and tested for AMH. Mixed-effects linear regression models were constructed with Box-Cox transformed AMH concentration as the dependent variable. The independent variables tested included maternal average daily gain (ADG) from pre-breeding examination (PBE) to pregnancy diagnosis (PD) between 30 and 60 days in calf (DIC), ADG from PBE to PD over 60 DIC and ADG between the two PDs. Calf breed and age at sampling were forced into the models, and the farm was treated as a random effect in all models. We found that as ADG increased from the pre-breeding period to their first PD visit, the AMH concentration in their offspring reduced. However, ADG explained only a small amount of the variation in AMH concentrations (marginal R2 = 0.041). In conclusion, the results of our study suggest that excessive growth prior to conception and in early gestation of nulliparous heifers could impact the ovarian reserve of their female offspring, and may imply that farmers should avoid excessive growth in the immediate pre-breeding and early gestational periods.
Collapse
Affiliation(s)
- Eilidh E Thomson
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - Marijke E Beltman
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - Mark A Crowe
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - Conor G McAloon
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - John G Furlong
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - Mary M Duane
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | - Jane P N Brennan
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | | | - Emmet T Kelly
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| |
Collapse
|
19
|
Chen Y, Yang S, Yu K, Zhang J, Wu M, Zheng Y, Zhu Y, Dai J, Wang C, Zhu X, Dai Y, Sun Y, Wu T, Wang S. Spatial omics: An innovative frontier in aging research. Ageing Res Rev 2024; 93:102158. [PMID: 38056503 DOI: 10.1016/j.arr.2023.102158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Disentangling the impact of aging on health and disease has become critical as population aging progresses rapidly. Studying aging at the molecular level is complicated by the diverse aging profiles and dynamics. However, the examination of cellular states within aging tissues in situ is hampered by the lack of high-resolution spatial data. Emerging spatial omics technologies facilitate molecular and spatial analysis of tissues, providing direct access to precise information on various functional regions and serving as a favorable tool for unraveling the heterogeneity of aging. In this review, we summarize the recent advances in spatial omics application in multi-organ aging research, which has enhanced the understanding of aging mechanisms from multiple standpoints. We also discuss the main challenges in spatial omics research to date, the opportunities for further developing the technology, and the potential applications of spatial omics in aging and aging-related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Shuhao Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Kaixu Yu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Centre, Sun Yat-sen University, Guangzhou, China
| | - Yun Zhu
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL 62702, USA
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Chunyan Wang
- College of Science & Engineering Jinan University, Guangzhou, China
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yunhong Sun
- Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| |
Collapse
|
20
|
Dey P, Monferini N, Donadini L, Lodde V, Franciosi F, Luciano AM. Method of Isolation and In Vitro Culture of Primordial Follicles in Bovine Animal Model. Methods Mol Biol 2024; 2770:171-182. [PMID: 38351454 DOI: 10.1007/978-1-0716-3698-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
The mammalian ovary is a substantial source of oocytes arranged into follicles at various stages of folliculogenesis, from the primordial to the ovulatory ones. Primordial follicles constitute the most abundant source of gametes inside the mammalian ovary at any given time.The isolation of a high number of primordial follicles, together with the development of protocols for in vitro follicle growth, would provide a powerful tool to fully exploit the female reproductive potential and boost the rescue and restoration of fertility in assisted reproduction technologies in human medicine, animal breeding, and preservation of threatened species. However, the most significant limitation is the lack of efficient methods for isolating a healthy and homogeneous population of viable primordial follicles suitable for in vitro culture. Here, we provide a fast and high-yield strategy for the mechanical isolation of primordial follicles from limited portions of the ovarian cortex in the bovine animal model.
Collapse
Affiliation(s)
- Pritha Dey
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
| | - Noemi Monferini
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
| | - Ludovica Donadini
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
| | - Valentina Lodde
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
| | - Federica Franciosi
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
| | - Alberto Maria Luciano
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
21
|
Hu L, Bai L, Qin R, Wang X, Zhou J, Yu C, Chen Y, Wang S, Zhao S, Chen L, Lu R. Optimizing FSH Concentration Modulation in the Short-Acting GnRH-a Long Protocol for IVF/ICSI: A Retrospective Study. Adv Ther 2024; 41:215-230. [PMID: 37884809 DOI: 10.1007/s12325-023-02702-y] [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: 09/04/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023]
Abstract
INTRODUCTION Exogenous gonadotropin (Gn) is given to regulate follicle-stimulating hormone (FSH) levels to achieve optimal ovarian response in in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI). The objective of this study was to analyze the optimal degree of change in FSH blood concentration with ovarian responsiveness in a short-acting gonadotropin-releasing hormone agonist (GnRH-a) long protocol for IVF/ICSI. METHODS This retrospective study was conducted at Changzhou Maternity and Child Health Hospital's Reproductive Center from May 2017 to May 2023. A total of 794 ovarian stimulation cycles for IVF/ICSI using the short-acting GnRH-a long protocol was included. Ovarian responsiveness was assessed based on the number of follicles > 14 mm on human chorionic gonadotropin (HCG) trigger day, refine-follicular output rate (Refine-FORT) and good quality embryos. Delta 1 referred to the change in FSH level between days 6-8 of gonadotropin usage and baseline FSH, while Delta 2 referred to the change in FSH level between HCG trigger day and days 6-8 of gonadotropin usage. Simple and multiple linear regression analysis were performed to adjust for confounding factors. RESULTS The number of follicles > 14 mm on HCG trigger day was found to be the most suitable indicator for evaluating ovarian responsiveness compared to the number of follicles > 16 mm and the number of retrieved oocytes. When Delta 1 ranged from 1.94 to 3.37, the number of follicles > 14 mm on HCG trigger day was the highest. When Delta 1 ranged from 3.37 to 5.90, the Refine-FORT was the highest. However, when Delta 1 exceeded 5.90, the number of follicles > 14 mm on HCG trigger day, Refine-FORT and good quality embryo all significantly decreased. On the other hand, when Delta 2 was ≤ - 1.58, the number of follicles > 14 mm on HCG trigger day and the Refine-FORT were both the highest. CONCLUSION This study identifies optimal Delta 1 and Delta 2 ranges for effective ovarian responsiveness in a short-acting GnRH-a long protocol for IVF/ICSI and introduces the novel measure of the number of follicles > 14 mm on HCG trigger day. The optimal range for Delta 1 was 1.94 to 3.37, and Delta 2 should be < - 1.58 for achieving a higher number and quality of oocytes.
Collapse
Affiliation(s)
- Lingmin Hu
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Lijing Bai
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Rui Qin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Scientific Research and Education Department, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Xiaoyu Wang
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Jing Zhou
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Chunmei Yu
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Yang Chen
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Shuxian Wang
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Shenyu Zhao
- Department of Neurology, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Li Chen
- Department of Reproduction, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| | - Renjie Lu
- Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| |
Collapse
|
22
|
Walsh PT, Martínez-Marchal A, Brieño-Enríquez MA. Culture of the Intact Postnatal Naked Mole-Rat Ovary: From Meiotic Prophase to Single-Cell RNASeq. Methods Mol Biol 2024; 2818:179-194. [PMID: 39126475 DOI: 10.1007/978-1-0716-3906-1_12] [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] [Indexed: 08/12/2024]
Abstract
Recently, we reported that, in the naked mole-rat (Heterocephalus glaber) ovary, there is mitotic expansion of the primordial germ cells (PGCs), and the initiation of the meiotic program occurs postnatally. This is opposite to almost all other mammals, including humans and mice, whose reproductive cycle begins very early in development. In both mouse and human, the ovaries become populated with PGCs in utero; these PGCs will later generate the oogonia. After mitotic proliferation, these cells will trigger the meiotic program and initiate meiotic prophase I. Given that all these processes happen in utero, their analysis has been very challenging; so the ability to study them postnatally and to manipulate them with inhibitors or other substances, in the naked mole-rat, opens new possibilities in the field. In this chapter, we present a comprehensive collection of protocols that permit the culture of whole naked mole-rat ovaries, followed by analysis of germ cells, from PGCs to oocytes, in meiotic prophase I, as well the obtention of single-cell suspension or single-nuclei suspension for RNASeq.
Collapse
Affiliation(s)
- Patrick T Walsh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana Martínez-Marchal
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Miguel Angel Brieño-Enríquez
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
23
|
Torkzadeh T, Asadi Z, Jafari Atrabi M, Eivazkhani F, Khodadi M, Hajiaghalou S, Akbarinejad V, Fathi R. Optimisation of hormonal treatment to improve follicular development in one-day-old mice ovaries cultured under in vitro condition. Reprod Fertil Dev 2023; 35:733-749. [PMID: 37995332 DOI: 10.1071/rd23027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
CONTEXT Base medium containing knock-out serum replacement (KSR) has been found to support formation and maintenance of follicles in one-day-old mice ovaries, but has not been shown to properly support activation and growth of primordial follicles. AIMS The present study was conducted to tailor the hormonal content of base medium containing KSR to enhance development of primordial follicles in neonatal ovaries. METHODS One-day-old mice ovaries were initially cultured with base medium for four days, and then, different hormonal treatments were added to the culture media and the culture was proceeded for four additional days until day eight. Ovaries were collected for histological and molecular assessments on days four and eight. KEY RESULTS In experiment I, the main and interactive effects of FSH and testosterone were investigated and FSH promoted activation of primordial follicles and development of primary and preantral follicles, and upregulated genes of phosphoinositide 3-kinase (Pi3k ), KIT ligand (Kitl ), growth differentiation factor 9 (Gdf9 ) and follicle stimulating hormone receptor (Fshr ) (P Bmp15 ), Connexin-43 (Cx43 ) and luteinising hormone and choriogonadotropin receptor (Lhcgr ) (P P Lhcgr (P P >0.05). CONCLUSIONS Supplementation of culture medium containing KSR with gonadotropins, particularly hMG, could improve follicular growth and expression of factors regulating follicular development. IMPLICATIONS This study was a step forward in formulating an optimal medium for development of follicles in cultured one-day-old mice ovaries.
Collapse
Affiliation(s)
- Tahoura Torkzadeh
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zahra Asadi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran; and Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA
| | - Mohammad Jafari Atrabi
- Institute of Pharmacology and Toxicology, University Medical Center, Georg August University, Göttingen, Germany; and Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research (DPZ), Göttingen, Germany
| | - Farideh Eivazkhani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maryam Khodadi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Samira Hajiaghalou
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
24
|
Appeltant R, Hermes R, Holtze S, Modina SC, Galli C, Bjarkadottir BD, Adeniran BV, Wei X, Swegen A, Hildebrandt TB, Williams SA. The neonatal southern white rhinoceros ovary contains oogonia in germ cell nests. Commun Biol 2023; 6:1049. [PMID: 37848538 PMCID: PMC10582104 DOI: 10.1038/s42003-023-05256-5] [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: 12/09/2022] [Accepted: 08/18/2023] [Indexed: 10/19/2023] Open
Abstract
The northern white rhinoceros is functionally extinct with only two females left. Establishing methods to culture ovarian tissues, follicles, and oocytes to generate eggs will support conservation efforts using in vitro embryo production. To the best of our knowledge, this is the first description of the structure and molecular signature of any rhinoceros, more specifically, we describe the neonatal and adult southern white rhinoceros (Ceratotherium simum simum) ovary; the closest relation of the northern white rhinoceros. Interestingly, all ovaries contain follicles despite advanced age. Analysis of the neonate reveals a population of cells molecularly characterised as mitotically active, pluripotent with germ cell properties. These results indicate that unusually, the neonatal ovary still contains oogonia in germ cell nests at birth, providing an opportunity for fertility preservation. Therefore, utilising ovaries from stillborn and adult rhinoceros can provide cells for advanced assisted reproductive technologies and investigating the neonatal ovaries of other endangered species is crucial for conservation.
Collapse
Affiliation(s)
- Ruth Appeltant
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
- Gamete Research Centre, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Robert Hermes
- Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str 17, D-10315, Berlin, Germany
| | - Susanne Holtze
- Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str 17, D-10315, Berlin, Germany
| | - Silvia Clotilde Modina
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy
| | - Cesare Galli
- Avantea srl, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100, Cremona, Italy
- Fondazione Avantea, 26100, Cremona, Italy
| | - Briet D Bjarkadottir
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
| | - Babatomisin V Adeniran
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
| | - Xi Wei
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
| | - Aleona Swegen
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, 2308, NSW, Australia
| | - Thomas Bernd Hildebrandt
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy
- Freie Universität Berlin, D-14195, Berlin, Germany
| | - Suzannah A Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
25
|
Cramer DW. Incessant ovulation: a review of its importance in predicting cancer risk. Front Oncol 2023; 13:1240309. [PMID: 37869082 PMCID: PMC10588628 DOI: 10.3389/fonc.2023.1240309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/16/2023] [Indexed: 10/24/2023] Open
Abstract
Estrous cycles are recurring changes in therian mammals induced by estrogen, progesterone, and other hormones culminating in endometrial proliferation, ovulation, and implantation if fertilization occurred. In women, the estrous cycle is the menstrual cycle; but, unlike most mammals, the end of an infertile cycle is marked by endometrial sloughing and the start of another without an anestrous phase. Women stop cycling at menopause, while in most mammals, cycles continue until death. Epidemiologic studies identified menarche, menopause, births, lactation, and oral contraceptive (OC) use as key risk factors for ovarian, breast, and endometrial cancers. A composite variable was created to estimate the number of cycles not interrupted by events that stop ovulation. Captured by the phrase "incessant ovulation", repetitive cycles were first postulated to affect ovarian cancer risk and later extended to breast and endometrial cancers. These associations could be explained by cumulative effects of repetitive tissue changes within reproductive organs, immune consequences of repetitive ovulation through the glycoprotein mucin 1, and residual effects of past ovulations that enhance ovarian production of testosterone. The latter two pathways could affect the risk for cancers in other organs not considered "reproductive".
Collapse
Affiliation(s)
- Daniel W. Cramer
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
26
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
27
|
Xu W, Yang Y, Yu Y, Wen C, Zhao S, Cao L, Zhao S, Qin Y, Chen ZJ. FAAP100 is required for the resolution of transcription-replication conflicts in primordial germ cells. BMC Biol 2023; 21:174. [PMID: 37580696 PMCID: PMC10426154 DOI: 10.1186/s12915-023-01676-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND The maintenance of genome stability in primordial germ cells (PGCs) is crucial for the faithful transmission of genetic information and the establishment of reproductive reserve. Numerous studies in recent decades have linked the Fanconi anemia (FA) pathway with fertility, particularly PGC development. However, the role of FAAP100, an essential component of the FA core complex, in germ cell development is unexplored. RESULTS We find that FAAP100 plays an essential role in R-loop resolution and replication fork protection to counteract transcription-replication conflicts (TRCs) during mouse PGC proliferation. FAAP100 deletion leads to FA pathway inactivation, increases TRCs as well as cotranscriptional R-loops, and contributes to the collapse of replication forks and the generation of DNA damage. Then, the activated p53 signaling pathway triggers PGC proliferation defects, ultimately resulting in insufficient establishment of reproductive reserve in both sexes of mice. CONCLUSIONS Our findings suggest that FAAP100 is required for the resolution of TRCs in PGCs to safeguard their genome stability.
Collapse
Affiliation(s)
- Weiwei Xu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yajuan Yang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yongze Yu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Canxin Wen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Simin Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Lili Cao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Shidou Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| | - Yingying Qin
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, 250012, Shandong, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.
| |
Collapse
|
28
|
Cramer DW, Vitonis AF, Huang T, Shafrir AL, Eliassen AH, Barbieri RL, Hankinson SE. Estimated Ovulatory Years Prior to Menopause and Postmenopausal Endogenous Hormone Levels. Cancer Epidemiol Biomarkers Prev 2023; 32:976-985. [PMID: 37127868 PMCID: PMC10630892 DOI: 10.1158/1055-9965.epi-23-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/30/2023] [Accepted: 04/27/2023] [Indexed: 05/03/2023] Open
Abstract
BACKGROUND Lifetime ovulatory years (LOY) is estimated by the difference between ages at menopause and menarche subtracting time for events interrupting ovulation. We tested whether LOY influences sex hormone levels in postmenopausal women with at least one intact ovary not using hormones. METHODS Estradiol, estrone, estrone sulfate, total testosterone, dehydroepiandrostendione sulfate, prolactin, and sex hormone binding globulin were measured in 1,976 postmenopausal women from the Nurses' Health Study. Associations of age, body mass index (BMI), smoking, alcohol use, and other factors on hormones were assessed by t tests and ANOVA. Linear regression was used to assess multivariable adjusted associations between LOY and hormones and trends in hormone levels per 5-year increases in LOY were estimated. RESULTS Women averaged 61.4 years old, 11.0 years since menopause, with BMI of 25.8 kg/m2. A total of 13.6% had irregular cycles, 17.5% hysterectomy, 6.4% unilateral oophorectomy, and 13.8% were current smokers. Variables associated with one or more hormone levels were included as covariates. Each 5-year increase in LOY was significantly associated with a 5.2% increase in testosterone in women with BMI < 25 kg/m2 and a 7.4% increase in testosterone and 7.3% increase in estradiol in women with above-average BMI. CONCLUSIONS This is the first study to show that greater LOY is associated with higher testosterone in postmenopausal women and higher estradiol in those with elevated BMI, suggesting accumulation of functioning stromal and thecal cells from repeated ovulations and peripheral conversion of testosterone. IMPACT A possible explanation for why greater LOY increases risk for breast, endometrial, and ovarian cancer is offered.
Collapse
Affiliation(s)
- Daniel W. Cramer
- Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, 221 Longwood Ave, Boston, Massachusetts 02115
- Harvard Medical School, 260 Longwood Avenue, Boston, Massachusetts 02115
| | - Allison F. Vitonis
- Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, 221 Longwood Ave, Boston, Massachusetts 02115
| | - Tianyi Huang
- Harvard Medical School, 260 Longwood Avenue, Boston, Massachusetts 02115
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115
| | - Amy L. Shafrir
- Division of Adolescent/Young Adult Medicine, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115
| | - A. Heather Eliassen
- Harvard Medical School, 260 Longwood Avenue, Boston, Massachusetts 02115
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115
- Departments of Nutrition and Epidemiology, Harvard TH Chan School of Public Health, 655 Huntington Avenue, Boston, Massachusetts 02115
| | - Robert L. Barbieri
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, 75 Francis Street, Boston, Massachusetts 02115
| | - Susan E. Hankinson
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst MA 01003
| |
Collapse
|
29
|
Stringer JM, Alesi LR, Winship AL, Hutt KJ. Beyond apoptosis: evidence of other regulated cell death pathways in the ovary throughout development and life. Hum Reprod Update 2023; 29:434-456. [PMID: 36857094 PMCID: PMC10320496 DOI: 10.1093/humupd/dmad005] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/06/2022] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Regulated cell death is a fundamental component of numerous physiological processes; spanning from organogenesis in utero, to normal cell turnover during adulthood, as well as the elimination of infected or damaged cells throughout life. Quality control through regulation of cell death pathways is particularly important in the germline, which is responsible for the generation of offspring. Women are born with their entire supply of germ cells, housed in functional units known as follicles. Follicles contain an oocyte, as well as specialized somatic granulosa cells essential for oocyte survival. Follicle loss-via regulated cell death-occurs throughout follicle development and life, and can be accelerated following exposure to various environmental and lifestyle factors. It is thought that the elimination of damaged follicles is necessary to ensure that only the best quality oocytes are available for reproduction. OBJECTIVE AND RATIONALE Understanding the precise factors involved in triggering and executing follicle death is crucial to uncovering how follicle endowment is initially determined, as well as how follicle number is maintained throughout puberty, reproductive life, and ovarian ageing in women. Apoptosis is established as essential for ovarian homeostasis at all stages of development and life. However, involvement of other cell death pathways in the ovary is less established. This review aims to summarize the most recent literature on cell death regulators in the ovary, with a particular focus on non-apoptotic pathways and their functions throughout the discrete stages of ovarian development and reproductive life. SEARCH METHODS Comprehensive literature searches were carried out using PubMed and Google Scholar for human, animal, and cellular studies published until August 2022 using the following search terms: oogenesis, follicle formation, follicle atresia, oocyte loss, oocyte apoptosis, regulated cell death in the ovary, non-apoptotic cell death in the ovary, premature ovarian insufficiency, primordial follicles, oocyte quality control, granulosa cell death, autophagy in the ovary, autophagy in oocytes, necroptosis in the ovary, necroptosis in oocytes, pyroptosis in the ovary, pyroptosis in oocytes, parthanatos in the ovary, and parthanatos in oocytes. OUTCOMES Numerous regulated cell death pathways operate in mammalian cells, including apoptosis, autophagic cell death, necroptosis, and pyroptosis. However, our understanding of the distinct cell death mediators in each ovarian cell type and follicle class across the different stages of life remains the source of ongoing investigation. Here, we highlight recent evidence for the contribution of non-apoptotic pathways to ovarian development and function. In particular, we discuss the involvement of autophagy during follicle formation and the role of autophagic cell death, necroptosis, pyroptosis, and parthanatos during follicle atresia, particularly in response to physiological stressors (e.g. oxidative stress). WIDER IMPLICATIONS Improved knowledge of the roles of each regulated cell death pathway in the ovary is vital for understanding ovarian development, as well as maintenance of ovarian function throughout the lifespan. This information is pertinent not only to our understanding of endocrine health, reproductive health, and fertility in women but also to enable identification of novel fertility preservation targets.
Collapse
Affiliation(s)
- Jessica M Stringer
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lauren R Alesi
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Amy L Winship
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Karla J Hutt
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
30
|
Zhu M, Xu M, Zhang J, Zheng C. The role of Hippo pathway in ovarian development. Front Physiol 2023; 14:1198873. [PMID: 37334049 PMCID: PMC10275494 DOI: 10.3389/fphys.2023.1198873] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
The follicle is the functional unit of the ovary, whereby ovarian development is largely dependent on the development of the follicles themselves. The activation, growth, and progression of follicles are modulated by a diverse range of factors, including reproductive endocrine system and multiple signaling pathways. The Hippo pathway exhibits a high degree of evolutionary conservation between both Drosophila and mammalian systems, and is recognized for its pivotal role in regulating cellular proliferation, control of organ size, and embryonic development. During the process of follicle development, the components of the Hippo pathway show temporal and spatial variations. Recent clinical studies have shown that ovarian fragmentation can activate follicles. The mechanism is that the mechanical signal of cutting triggers actin polymerization. This process leads to the disruption of the Hippo pathway and subsequently induces the upregulation of downstream CCN and apoptosis inhibitors, thereby promoting follicle development. Thus, the Hippo pathway plays a crucial role in both the activation and development of follicles. In this article, we focused on the development and atresia of follicles and the function of Hippo pathway in these processes. Additionally, the physiological effects of Hippo pathway in follicle activation are also explored.
Collapse
|
31
|
Su C, Zhang R, Zhang X, Lv M, Liu X, Ao K, Hao J, Mu YL. Dingkun Pill modulate ovarian function in chemotherapy-induced premature ovarian insufficiency mice by regulating PTEN/PI3K/AKT/FOXO3a signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023:116703. [PMID: 37257704 DOI: 10.1016/j.jep.2023.116703] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dingkun Pill (DKP) is a traditional Chinese medicine that has been shown to have beneficial effects on reproductive function. However, the specific mechanism underlying its effect on POI is not well understood. AIM OF THE STUDY To investigate the effect of different doses of Dingkun Pill on ovarian function in cyclophosphamide (CTX)-induced premature ovarian insufficiency (POI) mice and to explore its molecular mechanism through PTEN/PI3K/AKT/FOXO3a signaling pathway. This study will provide valuable insights into the potential clinical application of Dingkun Pill for the treatment of POI. MATERIALS AND METHODS Fifty female ICR mice were randomly divided into normal control (NC) group, model control (MC) group, and Dingkun Pill low, medium, high dose (DKP-L, M, H) groups. Mice were injected with CTX to construct the POI model. Mice in the DKP-L, M, and H groups were given 0.9 g/kg, 1.8 g/kg, and 3.6 g/kg of Dingkun Pill suspension for 21 days, respectively. Mice in the NC and MC groups were given the same amount of normal saline by gavage. Changes in body weight, estrous cycle and gonadal index were observed in each group of mice. Serum levels of FSH, LH, E2 and AMH were detected by ELISA. Hematoxylin-eosin (HE) staining observed the changes of ovarian pathological morphology and follicle counts at all levels. qRT-PCR was used to measure the levels of the PTEN and FOXO3a genes in ovarian tissue. The expression of PTEN/PI3K/AKT/FOXO3a signaling pathway related proteins were detected by Western-blot and immunohistochemistry(IHC). RESULTS In POI mice, Dingkun Pill increased body weight, promoted the recovery of estrous cycle, increased ovarian index, and improved pathological morphology of the ovaries. The FSH level decreased in the medium dose group (P < 0.05), the LH level reduced significantly in the medium and high dose groups (P < 0.01), and the E2 level in the high dose group increased (P < 0.05). There was no significant difference in AMH levels across all dose groups. The number of growing follicles improved at all levels in the low and medium dose groups, but declined significantly in the high dose group. However, the number of corpus luteum increased significantly in the high dose group (P < 0.001), and the atretic follicles in the three dose groups decreased. Results from qRT-PCR, Western-blot and IHC showed that the moderate dose of Dingkun Pill suppressed the levels of the p-PI3K and p-AKT proteins by upregulating the expression of PTEN in the ovarian tissues of POI mice, thereby inhibiting the expression of the key protein p-FOXO3a. However, the inhibitory effect of the higher dose may be less than that of the lower and intermediate dose groups. CONCLUSIONS The Dingkun Pill modulated hormonal levels, promoted follicle growth and induced ovulation in mice with CTX-induced POI, with better results in the low and moderate dose groups. Its mechanism may be related to the regulation of the PTEN/PI3K/AKT/FOXO3a signaling pathway.
Collapse
Affiliation(s)
- Chan Su
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China; The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Ruihong Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan, 250012, China
| | - Xiujuan Zhang
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China
| | - Mengxiao Lv
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China
| | - Xiang Liu
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Kai Ao
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Jing Hao
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan, 250012, China.
| | - Yu-Lan Mu
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China.
| |
Collapse
|
32
|
Yu Y, Xu W, Wen C, Zhao S, Li G, Liu R, Chen ZJ, Qin Y, Ma J, Yang Y, Zhao S. UBE2T resolves transcription-replication conflicts and protects common fragile sites in primordial germ cells. Cell Mol Life Sci 2023; 80:92. [PMID: 36928776 PMCID: PMC11072727 DOI: 10.1007/s00018-023-04733-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/04/2023] [Accepted: 02/22/2023] [Indexed: 03/18/2023]
Abstract
The proper development of primordial germ cells (PGCs) is an essential prerequisite for gametogenesis and mammalian fertility. The Fanconi anemia (FA) pathway functions in maintaining the development of PGCs. FANCT/UBE2T serves as an E2 ubiquitin-conjugating enzyme that ubiquitylates the FANCD2-FANCI complex to activate the FA pathway, but its role in the development of PGCs is not clear. In this study, we found that Ube2t knockout mice showed defects in PGC proliferation, leading to severe loss of germ cells after birth. Deletion of UBE2T exacerbated DNA damage and triggered the activation of the p53 pathway. We further demonstrated that UBE2T counteracted transcription-replication conflicts by resolving R-loops and stabilizing replication forks, and also protected common fragile sites by resolving R-loops in large genes and promoting mitotic DNA synthesis to maintain the genome stability of PGCs. Overall, these results provide new insights into the function and regulatory mechanisms of the FA pathway ensuring normal development of PGCs.
Collapse
Affiliation(s)
- Yongze Yu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Weiwei Xu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Canxin Wen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Simin Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Guangyu Li
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Ran Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Jinan, 250021, Shandong, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
- Center for Reproductive Medicine, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, 200135, China
| | - Yingying Qin
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Jinlong Ma
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| | - Yajuan Yang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| | - Shidou Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
33
|
Brieño-Enríquez MA, Faykoo-Martinez M, Goben M, Grenier JK, McGrath A, Prado AM, Sinopoli J, Wagner K, Walsh PT, Lopa SH, Laird DJ, Cohen PE, Wilson MD, Holmes MM, Place NJ. Postnatal oogenesis leads to an exceptionally large ovarian reserve in naked mole-rats. Nat Commun 2023; 14:670. [PMID: 36810851 PMCID: PMC9944903 DOI: 10.1038/s41467-023-36284-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 01/24/2023] [Indexed: 02/24/2023] Open
Abstract
In the long-lived naked mole-rat (NMR), the entire process of oogenesis occurs postnatally. Germ cell numbers increase significantly in NMRs between postnatal days 5 (P5) and P8, and germs cells positive for proliferation markers (Ki-67, pHH3) are present at least until P90. Using pluripotency markers (SOX2 and OCT4) and the primordial germ cell (PGC) marker BLIMP1, we show that PGCs persist up to P90 alongside germ cells in all stages of female differentiation and undergo mitosis both in vivo and in vitro. We identified VASA+ SOX2+ cells at 6 months and at 3-years in subordinate and reproductively activated females. Reproductive activation was associated with proliferation of VASA+ SOX2+ cells. Collectively, our results suggest that highly desynchronized germ cell development and the maintenance of a small population of PGCs that can expand upon reproductive activation are unique strategies that could help to maintain the NMR's ovarian reserve for its 30-year reproductive lifespan.
Collapse
Affiliation(s)
- Miguel Angel Brieño-Enríquez
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Mariela Faykoo-Martinez
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Mississauga, Mississauga, ON, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Meagan Goben
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer K Grenier
- RNA sequencing core and Center for Reproductive Genomics, College of Veterinary, Cornell University, Ithaca, NY, USA
| | - Ashley McGrath
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | - Alexandra M Prado
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | - Jacob Sinopoli
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | - Kate Wagner
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | - Patrick T Walsh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Samia H Lopa
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diana J Laird
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Paula E Cohen
- Center for Reproductive Genomics, Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | - Michael D Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Melissa M Holmes
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Mississauga, Mississauga, ON, Canada
| | - Ned J Place
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
34
|
Song W, Li A, Sha QQ, Liu SY, Zhou Y, Zhou CY, Zhang X, Li XZ, Jiang JX, Li F, Li C, Schatten H, Ou XH, Sun QY. Maternal exposure to 4-vinylcyclohexene diepoxide during pregnancy induces subfertility and birth defects of offspring in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 859:160431. [PMID: 36423845 DOI: 10.1016/j.scitotenv.2022.160431] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/13/2022] [Accepted: 11/19/2022] [Indexed: 06/16/2023]
Abstract
4-vinylcyclohexene diepoxide (VCD), widely used in industry, is a hazardous compound that can cause premature ovarian failure, but whether maternal VCD exposure affects the health and reproduction of offspring is unknown. Here we focused on the effects of VCD on fertility and physical health of F1 and F2 offspring in mice. The pregnant mice were injected intraperitoneally with different dosages of VCD once every day from 6.5 to 18.5 days post-coitus (dpc). We showed that maternal exposure to VCD during pregnancy significantly reduced the litter size and ovarian reserve, while increasing microtia occurrences of F1 mice. The cytospread staining showed a significant inhibition of meiotic prophase I progression from the zygotene stage to the pachytene stage. Mechanistically, the expression level of DNA damage marker (γ-H2AX) and BAX/BCL2 ratios were significantly increased, and RAD51 and DMC1 were extensively recruited to DNA double strand breaks sites in the oocytes of offspring from VCD-exposed mothers. Overall, our results provide solid evidence showing that maternal exposure to VCD during pregnancy has intergenerational deleterious effects on the offspring.
Collapse
Affiliation(s)
- Wei Song
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China; College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Ang Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Qian-Qian Sha
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Shao-Yuan Liu
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Yong Zhou
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Chang-Yin Zhou
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Xue Zhang
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Xiao-Zhen Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Jia-Xin Jiang
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Fei Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Chao Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Xiang-Hong Ou
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China.
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou 510317, China; College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China.
| |
Collapse
|
35
|
Dela Cruz C, Kinnear HM, Hashim PH, Wandoff A, Nimmagadda L, Chang FL, Padmanabhan V, Shikanov A, Moravek MB. A mouse model mimicking gender-affirming treatment with pubertal suppression followed by testosterone in transmasculine youth. Hum Reprod 2023; 38:256-265. [PMID: 36484619 PMCID: PMC10167862 DOI: 10.1093/humrep/deac257] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 11/04/2022] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Can mice serve as a translational model to examine the reproductive consequences of pubertal suppression with GnRH agonist (GnRHa) followed by testosterone (T) administration, a typical therapy in peripubertal transmasculine youth? SUMMARY ANSWER An implanted depot with 3.6 mg of GnRHa followed by T enanthate at 0.45 mg weekly can be used in peripubertal female mice for investigating the impact of gender-affirming hormone therapy in transmasculine youth. WHAT IS KNOWN ALREADY There is limited knowledge available in transgender medicine to provide evidence-based fertility care, with the current guidelines being based on the assumption of fertility loss. We recently successfully developed a mouse model to investigate the reproductive consequences of T therapy given to transgender men. On the other hand, to our knowledge, there is no mouse model to assess the reproductive outcomes in peripubertal transmasculine youth. STUDY DESIGN, SIZE, DURATION A total of 80 C57BL/6N female mice were used in this study, with n = 7 mice in each experimental group. PARTICIPANTS/MATERIALS, SETTING, METHODS We first assessed the effectiveness of GnRHa in arresting pubertal development in the female mice. In this experiment, 26-day-old female mice were subcutaneously implanted with a GnRHa (3.6 mg) depot. Controls underwent a sham surgery. Animals were euthanized at 3, 9, 21 and 28 days after the day of surgery. In the second experiment, we induced a transmasculine youth mouse model. C57BL/6N female mice were subcutaneously implanted with a 3.6 mg GnRHa depot on postnatal day 26 for 21 days and this was followed by weekly injections of 0.45 mg T enanthate for 6 weeks. The control for the GnRH treatment was sham surgery and the control for T treatment was sesame oil vehicle injections. Animals were sacrificed 0.5 weeks after the last injection. The data collected included the day of the vaginal opening and first estrus, daily vaginal cytology, weekly and terminal reproductive hormones levels, body/organ weights, ovarian follicular distribution and corpora lutea (CL) counts. MAIN RESULTS AND THE ROLE OF CHANCE GnRHa implanted animals remained in persistent diestrus and had reduced levels of FSH (P = 0.0013), LH (P = 0.0082) and estradiol (P = 0.0155), decreased uterine (P < 0.0001) and ovarian weights (P = 0.0002), and a lack of CL at 21 days after GnRHa implantation. T-only and GnRHa+T-treated animals were acyclic throughout the treatment period, had sustained elevated levels of T, suppressed LH levels (P < 0.0001), and an absence of CL compared to controls (P < 0.0001). Paired ovarian weights were reduced in the T-only and GnRHa+T groups compared with the control and GnRHa-only groups. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Although it is an appropriate tool to provide relevant findings, precaution is needed to extrapolate mouse model results to mirror human reproductive physiology. WIDER IMPLICATIONS OF THE FINDINGS To our knowledge, this study describes the first mouse model mimicking gender-affirming hormone therapy in peripubertal transmasculine youth. This model provides a tool for researchers studying the effects of GnRHa-T therapy on other aspects of reproduction, other organ systems and transgenerational effects. The model is supported by GnRHa suppressing puberty and maintaining acyclicity during T treatment, lower LH levels and absence of CL. The results also suggest GnRHa+T therapy in peripubertal female mice does not affect ovarian reserve, since the number of primordial follicles was not affected by treatment. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Michigan Institute for Clinical and Health Research grants KL2 TR 002241 and UL1 TR 002240 (C.D.C.); National Institutes of Health grants F30-HD100163 and T32-HD079342 (H.M.K.); University of Michigan Office of Research funding U058227 (A.S.); American Society for Reproductive Medicine/Society for Reproductive Endocrinology and Infertility grant (M.B.M.); and National Institutes of Health R01-HD098233 (M.B.M.). The University of Virginia Center for Research in Reproduction Ligand Assay and Analysis Core Facility was supported by the Eunice Kennedy Shriver NICHD/NIH grants P50-HD028934 and R24-HD102061. The authors declare that they have no competing interests.
Collapse
Affiliation(s)
- Cynthia Dela Cruz
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Postdoctoral Translational Scholar Program, Michigan Institute for Clinical & Health Research, University of Michigan, Ann Arbor, MI, USA
| | - Hadrian M Kinnear
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Prianka H Hashim
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Abigail Wandoff
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Likitha Nimmagadda
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Faith L Chang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - Ariella Shikanov
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Molly B Moravek
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Division of Reproductive Endocrinology and Infertility, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Pei J, Xiong L, Guo S, Wang X, Bao P, Wu X, Yan P, Guo X. A single-cell transcriptomic atlas characterizes cell types and their molecular features in yak ovarian cortex. FASEB J 2023; 37:e22718. [PMID: 36527406 DOI: 10.1096/fj.202201176rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/21/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
The ovary as one of the most dynamic organs produces steroids to orchestrate female secondary sexual characteristics, harbors ovarian reserve for oocytes, releases mature oocytes for fertilization, and maintains pregnancy. Yak (Bos grunniens) is the only bovid animal that can adapt to the harsh climatic conditions on the Qinghai-Tibetan Plateau (altitudes of over 3000 m above sea level). However, the cellular atlas is composed of oocytes and other somatic cells, and their individual molecular characteristics remain to be elucidated in the yak ovary. Here, single-cell RNA sequencing (scRNA-seq) was performed to delineate the molecular signature of various cell types in the yak ovarian cortex. A cellular atlas of yak ovarian cortex was constructed successfully on the basis of the differentially expressed genes (DEGs) from the distinct cell types and their functional enrichment analysis, comprising endothelial cells, nature kill cells, stromal cells, smooth muscle cells, oocytes, macrophages, epithelial cells, and granulosa cells. Meanwhile, the signature genes were determined based on their expression specificity in each cell type. A cell-to-cell communication network was built in light of the differentially overexpressed ligand and receptor genes from each cell type. Further, the oocytes were subdivided into four subtypes based on their individual DEGs and the functional enrichment of the DEGs. FST and TOP2A were identified as maker genes for oocytes by immunostaining in the yak ovarian cortex. The cellular atlas reveals the biological characteristics of the ovarian cortex at the cellular molecular level and provides insights into female reproductive biology via cellular communications in the yak.
Collapse
Affiliation(s)
- Jie Pei
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Lin Xiong
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Shaoke Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengjia Bao
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Xiaoyun Wu
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Xian Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| |
Collapse
|
37
|
Li J, Li Q, Zhang L, Zhang S, Dai Y. Poly-ADP-ribose polymerase (PARP) inhibitors and ovarian function. Biomed Pharmacother 2023; 157:114028. [PMID: 36410122 DOI: 10.1016/j.biopha.2022.114028] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/19/2022] Open
Abstract
Poly-ADP-ribose polymerase (PARP) plays an important role in DNA damage detection and repair. PARP inhibitors (PARPi) are a novel class of targeted agents used widely in the treatment of female cancer patients with BRCA mutations, including younger patients. However, the impact of PARPi on ovarian function remains a considerable problem in clinical practice. In this review article, we summarize the current understanding of PARPi's effects on the function of ovary and discuss their potential underlying mechanisms, highlighting the significance of further investigation on the criterion for ovarian failure and its preventive approaches during PARPi treatment.
Collapse
Affiliation(s)
- Jiajia Li
- Gynecologic Oncology Department, First Hospital of Jilin University, Changchun, Jilin, China; Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Qingchao Li
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Lingyi Zhang
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China; Gynecology and Obstetrics Department, Second Hospital of Jilin University, Changchun, Jilin, China
| | - Songling Zhang
- Gynecologic Oncology Department, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
38
|
Brieño-Enríquez MA. Characterization of the Postnatal Naked Mole-Rat Ovary: From Primordial Germ Cells to Meiotic Prophase I Oocytes. Methods Mol Biol 2023; 2677:185-201. [PMID: 37464243 DOI: 10.1007/978-1-0716-3259-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The mammalian reproductive cycle, including those of humans and mice, begins very early in development. In utero, the ovaries become populated with primordial germ cells (PGCs) that will generate the oogonia. First, these cells proliferate mitotically, and then they trigger the meiotic program and initiate meiotic prophase I. Since these processes happen during gestation, their study had been very limited and challenging. Recently, we reported that, in the naked mole-rat (Heterocephalus glaber) ovary, there is mitotic expansion of the PGCs, and the initiation of the meiotic program occurs postnatally. In this chapter, we present a comprehensive collection of protocols that permit the analysis of naked mole-rat germ cells, from PGCs to oocytes, in meiotic prophase I, using in vivo and in vitro approaches.
Collapse
Affiliation(s)
- Miguel Angel Brieño-Enríquez
- Magee-Women's Research Institute, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
39
|
EFFECTS OF DIOSMIN ADMINISTRATION ON CISPLATIN-INDUCED PREMATURE OVARIAN FAILURE IN A RAT MODEL. JOURNAL OF CONTEMPORARY MEDICINE 2022. [DOI: 10.16899/jcm.1166435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
m
We aimed to examine the potential beneficial effects of diosmin administration on cisplatin - induced premature ovarian failure (POF) in a rat model
Material and Methods
Twenty-eight rats were divided into four groups. Group A rats (n:7) were determined as the sham group. The remaining rats received an intraperitoneal injection of 1.5 mg/kg/day cisplatin for 10 days to create a POF model. Then, they were randomly divided into 3 subgroups.
Group B was determined as POF group. Group C rats were given 100mg/kg/day diosmin for 10 days simultaneously while creating POF model. Group D rats were given 100mg/kg/day diosmin for 10 days after POF model was created. Twentieth day blood samples were taken and left ovaries were resected for examination.
Results
CIS-induced rats showed reduced levels of SOD, AMH and E2 compared to sham group rats (p0.05) between the sham group, group C and D. No significant (p>0.05) difference in FSH value was observed between group C, D and sham groups.
There was no significant (p>0.05) difference in the number of secondary and antral follicles between group C and D compared to the sham group. Primordial follicle count was significantly higher in group C than group B (p
Collapse
|
40
|
Oocyte Casein kinase 1α deletion causes defects in primordial follicle formation and oocyte loss by impairing oocyte meiosis and enhancing autophagy in developing mouse ovary. Cell Death Dis 2022; 8:388. [PMID: 36115846 PMCID: PMC9482644 DOI: 10.1038/s41420-022-01184-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022]
Abstract
Casein kinase 1α is a member of CK1 family, which is ubiquitously expressed and plays multiple functions, including its potential roles in regulating cell division. But the functions of CK1α in mammalian oogenesis and folliculogenesis remain elusive. In this study, we assayed the cell type of CK1α expression in the developing mouse ovary and confirmed that CK1α was highly expressed in ovaries after birth. The oocyte-specific CK1α knockout (cKO) mouse model was then established by crossing Ddx4-Cre mice with Csnk1a1-floxp mice, and the effects of CK1α deletion on oogenesis and folliculogenesis were identified. The results showed that oocyte CK1α deletion impaired the progression of oocyte meiosis and primordial follicle formation during meiotic prophase I, which subsequently caused oocyte loss and mouse infertility. Further, the in vivo CK1α deletion and in vitro inhibition of CK1 activity resulted in the defects of DNA double-strand break (DSB) repair, whereas apoptosis and autophagy were enhanced in the developing ovary. These may contribute to oocyte loss and infertility in cKO mice. It is thus concluded that CK1α is essential for mouse oogenesis and folliculogenesis by involving in regulating the processes of oocyte meiosis and DNA DSB repair during meiotic prophase I of mouse oocytes. However, the related signaling pathway and molecular mechanisms need to be elucidated further.
Collapse
|
41
|
The programmed death of fetal oocytes and the correlated surveillance mechanisms. REPRODUCTIVE AND DEVELOPMENTAL MEDICINE 2022. [DOI: 10.1097/rd9.0000000000000016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
42
|
Malott KF, Leon Parada K, Lee M, Swanson E, Luderer U. Gestational Benzo[a]pyrene Exposure Destroys F1 Ovarian Germ Cells Through Mitochondrial Apoptosis Pathway and Diminishes Surviving Oocyte Quality. Toxicol Sci 2022; 190:23-40. [PMID: 35993611 PMCID: PMC9960072 DOI: 10.1093/toxsci/kfac086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Polycyclic aromatic hydrocarbons, including benzo[a]pyrene (BaP), are products of incomplete combustion. In female mouse embryos primordial germ cells proliferate before and after arriving at the gonadal ridge around embryonic (E) 10 and begin entering meiosis at E13.5. Now oocytes, they arrest in the first meiotic prophase beginning at E17.5. We previously reported dose-dependent depletion of ovarian follicles in female mice exposed to 2 or 10 mg/kg-day BaP E6.5-15.5. We hypothesized that embryonic ovaries are more sensitive to gestational BaP exposure during the mitotic developmental window, and that this exposure results in persistent oxidative stress in ovaries and oocytes of exposed F1 female offspring. We orally dosed timed-pregnant female mice with 0 or 2 mg/kg-day BaP in oil from E6.5-11.5 (mitotic window) or E12.5-17.5 (meiotic window). Cultured E13.5 ovaries were utilized to investigate the mechanism of BaP-induced germ cell death. We observed statistically significant follicle depletion and increased ovarian lipid peroxidation in F1 pubertal ovaries following BaP exposure during either prenatal window. Culture of E13.5 ovaries with BaP induced germ cell DNA damage and release of cytochrome c from the mitochondria in oocytes, confirming that BaP exposure induced apoptosis via the mitochondrial pathway. Mitochondrial membrane potential, oocyte lipid droplet (LD) volume, and mitochondrial-LD colocalization were decreased and mitochondrial superoxide levels were increased in the MII oocytes of F1 females exposed gestationally to BaP. Results demonstrate similar sensitivity to germ cell depletion and persistent oxidative stress in F1 ovaries and oocytes following gestational BaP exposure during mitotic or meiotic windows.
Collapse
Affiliation(s)
- Kelli F Malott
- Environmental Health Sciences Graduate Program, University of California, Irvine, Irvine, California 92617, USA,Department of Environmental and Occupational Health, University of California, Irvine, Irvine, California 92617, USA
| | - Kathleen Leon Parada
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California 92617, USA
| | - Melody Lee
- Department of Medicine, University of California, Irvine, Irvine, California 92617, USA
| | - Edward Swanson
- Department of Medicine, University of California, Irvine, Irvine, California 92617, USA
| | - Ulrike Luderer
- To whom correspondence should be addressed at Center for Occupational and Environmental Health, 100 Theory Drive, Suite 100, Irvine, CA 92617, USA. E-mail:
| |
Collapse
|
43
|
Ma X, Zhang W, Song J, Li F, Liu J. Lifelong exposure to pyrethroid insecticide cypermethrin at environmentally relevant doses causes primary ovarian insufficiency in female mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 298:118839. [PMID: 35031408 DOI: 10.1016/j.envpol.2022.118839] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/29/2021] [Accepted: 01/08/2022] [Indexed: 06/14/2023]
Abstract
Pyrethroids are a class of widely used insecticides. Our recent epidemiological study of Chinese women reported that pyrethroid exposure was positively associated with the risk of primary ovarian insufficiency (POI). In this study, we utilized cypermethrin (CP), the most frequently detected pyrethroid in the environment, to recognize how lifelong and low-dose exposure to pyrethroids affects ovarian functions and the underlying mechanism(s). Female mice were exposed to CP at doses of human dietary intake of 6.7 μg/kg/day, an acceptable daily intake (ADI) of 20 μg/kg/day, or the chronic reference dose (RfD) of 60 μg/kg/day, starting from gestational day 0.5 until 44-week-old. We assessed effects on fertility, serum hormone levels, ovarian follicular development and ovarian transcriptomic profiles. Chronic exposure to CP at doses of ADI and RfD caused a significant reduction in the size of the primordial follicle pool on postnatal day (PND) 5 and the number of all types of follicles in 44-week-old mice, lower estrogen and higher gonadotropin levels, as well as decreased fertility. Significant increase in apoptosis and decrease in cell proliferation were observed in CP-exposed ovarian follicles from PND 5 and 44-week-old mice. Ovarian transcriptomic data showed that the pro-apoptotic protein BMF and the cell cycle inhibitor p27 were significantly up-regulated in CP-exposed ovaries. Cyp17a1, Cyp19a1 and Hsd17b1 genes involved in the key steps of steroidogenesis were down-regulated in the ovaries of female mice exposed to CP. This study first reported that lifelong exposure to CP at doses of ADI or RfD caused an ovarian phenotype similar to human POI in female mice and provided a mechanistic explanation. Our findings suggest that lifelong exposure to pyrethroids of low doses, which are recommended as 'safe' dosages, may have a significant impact on the ovarian health of female mammals and humans.
Collapse
Affiliation(s)
- Xiaochen Ma
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Wei Zhang
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, 310036 Hangzhou, China
| | - Jingyi Song
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Feixue Li
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, 310036 Hangzhou, China
| | - Jing Liu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, 310058 Hangzhou, China.
| |
Collapse
|
44
|
Nwachukwu CU, Woad KJ, Barnes N, Gardner DS, Robinson RS. Maternal protein restriction affects fetal ovary development in sheep. REPRODUCTION AND FERTILITY 2022; 2:161-171. [PMID: 35128451 PMCID: PMC8815062 DOI: 10.1530/raf-20-0073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/24/2021] [Indexed: 02/05/2023] Open
Abstract
Maternal malnutrition has important developmental consequences for the foetus. Indeed, adverse fetal ovarian development could have lifelong impact, with potentially reduced ovarian reserve and fertility of the offspring. This study investigated the effect of maternal protein restriction on germ cell and blood vessel development in the fetal sheep ovary. Ewes were fed control (n = 7) or low protein (n = 8) diets (17.0 g vs 8.7 g crude protein/MJ metabolizable energy) from conception to day 65 of gestation (gd65). On gd65, fetal ovaries were subjected to histological and immunohistochemical analysis to quantify germ cells (OCT4, VASA, DAZL), proliferation (Ki67), apoptosis (caspase 3) and vascularisation (CD31). Protein restriction reduced the fetal ovary weight (P < 0.05) but had no effect on fetal weight (P > 0.05). The density of germ cells was unaffected by maternal diet (P > 0.05). In the ovarian cortex, OCT4+ve cells were more abundant than DAZL+ve (P < 0.001) and VASA+ve cells (P < 0.001). The numbers, density and estimated total weight of OCT4, DAZL, and VASA+ve cells within the ovigerous cords were similar in both dietary groups (P > 0.05). Similarly, maternal protein restriction had no effect on germ cell proliferation or apoptotic indices (P > 0.05) and the number, area and perimeter of medullary blood vessels and degree of microvascularisation in the cortex (P > 0.05). In conclusion, maternal protein restriction decreased ovarian weight despite not affecting germ cell developmental progress, proliferation, apoptosis, or ovarian vascularity. This suggests that reduced maternal protein has the potential to regulate ovarian development in the offspring.
Collapse
Affiliation(s)
- Chinwe U Nwachukwu
- School of Veterinary Medicine and Science, Sutton Bonington campus, The University of Nottingham, Loughborough, Leicestershire, UK.,Department of Agricultural Science, School of Agriculture and Vocational Studies, Alvan Ikoku Federal College of Education, Owerri, Imo State, Nigeria
| | - Kathryn J Woad
- School of Veterinary Medicine and Science, Sutton Bonington campus, The University of Nottingham, Loughborough, Leicestershire, UK
| | - Nicole Barnes
- School of Veterinary Medicine and Science, Sutton Bonington campus, The University of Nottingham, Loughborough, Leicestershire, UK.,Medivet Oxted, Oxted, UK
| | - David S Gardner
- School of Veterinary Medicine and Science, Sutton Bonington campus, The University of Nottingham, Loughborough, Leicestershire, UK
| | - Robert S Robinson
- School of Veterinary Medicine and Science, Sutton Bonington campus, The University of Nottingham, Loughborough, Leicestershire, UK
| |
Collapse
|
45
|
Sulfasalazine exposure during pregnancy and lactation induces alterations in reproductive behavior in adult female rat offspring. Life Sci 2022; 293:120303. [PMID: 35051419 DOI: 10.1016/j.lfs.2022.120303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 11/21/2022]
Abstract
AIMS Sulfasalazine (SAS) is the first line drug in the treatment of chronic inflammatory bowel diseases in pregnant women. SAS and its metabolites cross the placenta and can be transferred through the milk. However, the long-term consequences to the reproductive system of offspring from dams exposed to SAS have not yet been studied. Thus, our study investigated the effects of SAS treatment during gestational and lactational periods on maternal care in F0 and reproductive outcomes in F1 females. MAIN METHODS Wistar female rats (n = 10/group) received 300 mg/kg/day of SAS dissolved in carboxymethyl cellulose (CMC), by gavage, from gestational day 0 to lactation day 21 and 3 mg/kg/day of folic acid during gestation. The control group received CMC only. On PND 21, the female pups were selected for reproductive evaluation at different time points: infancy and adulthood. The reproductive parameters evaluated were installation of puberty (vaginal opening and first estrus), estrous cyclicity, reproductive organs weight, histological analysis of the ovary follicles and uterus, analysis of oxidative stress in ovarian tissue, reproductive behavior (sexual and maternal), and fertility. KEY FINDINGS SAS treatment decreased the retrieving behavior in F0 females. The F1 females presented an increase in the lordosis score, frequency of lordosis of magnitude 3, and lipid peroxidation of ovarian tissues in both infancy and adult life. SIGNIFICANCE The SAS effects observed in the current study represent a relevant concern for public health, as they demonstrated that treatment with SAS compromised the maternal motivation of dams and induced reproductive alterations in F1 females.
Collapse
|
46
|
Luderer U, Lim J, Ortiz L, Nguyen JD, Shin JH, Allen BD, Liao LS, Malott K, Perraud V, Wingen LM, Arechavala RJ, Bliss B, Herman DA, Kleinman MT. Exposure to environmentally relevant concentrations of ambient fine particulate matter (PM 2.5) depletes the ovarian follicle reserve and causes sex-dependent cardiovascular changes in apolipoprotein E null mice. Part Fibre Toxicol 2022; 19:5. [PMID: 34996492 PMCID: PMC8740366 DOI: 10.1186/s12989-021-00445-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Fine particulate matter (PM2.5) exposure accelerates atherosclerosis and contains known ovotoxic chemicals. However, effects of exposure to PM2.5 on the finite ovarian follicle pool have hardly been investigated, nor have interactions between ovarian and cardiovascular effects. We hypothesized that subchronic inhalation exposure to human-relevant concentrations of PM2.5 results in destruction of ovarian follicles via apoptosis induction, as well as accelerated recruitment of primordial follicles into the growing pool. Further, we hypothesized that destruction of ovarian follicles enhances the adverse cardiovascular effects of PM2.5 in females. RESULTS Hyperlipidemic apolipoprotein E (Apoe) null ovary-intact or ovariectomized female mice and testis-intact male mice were exposed to concentrated ambient PM2.5 or filtered air for 12 weeks, 5 days/week for 4 h/day using a versatile aerosol concentration enrichment system. Primordial, primary, and secondary ovarian follicle numbers were decreased by 45%, 40%, and 17%, respectively, in PM2.5-exposed ovary-intact mice compared to controls (P < 0.05). The percentage of primary follicles with granulosa cells positive for the mitosis marker Ki67 was increased in the ovaries from PM2.5-exposed females versus controls (P < 0.05), consistent with increased recruitment of primordial follicles into the growing pool. Exposure to PM2.5 increased the percentages of primary and secondary follicles with DNA damage, assessed by γH2AX immunostaining (P < 0.05). Exposure to PM2.5 increased the percentages of apoptotic antral follicles, determined by TUNEL and activated caspase 3 immunostaining (P < 0.05). Removal of the ovaries and PM2.5-exposure exacerbated the atherosclerotic effects of hyperlipidemia in females (P < 0.05). While there were statistically significant changes in blood pressure and heart rate variability in PM2.5-compared to Air-exposed gonad-intact males and females and ovariectomized females, the changes were not consistent between exposure years and assessment methods. CONCLUSIONS These results demonstrate that subchronic PM2.5 exposure depletes the ovarian reserve by increasing recruitment of primordial follicles into the growing pool and increasing apoptosis of growing follicles. Further, PM2.5 exposure and removal of the ovaries each increase atherosclerosis progression in Apoe-/- females. Premature loss of ovarian function is associated with increased risk of osteoporosis, cardiovascular disease and Alzheimer's disease in women. Our results thus support possible links between PM2.5 exposure and other adverse health outcomes in women.
Collapse
Affiliation(s)
- Ulrike Luderer
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Center for Occupational and Environmental Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - Jinhwan Lim
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA
| | - Laura Ortiz
- grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - Johnny D. Nguyen
- grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - Joyce H. Shin
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - Barrett D. Allen
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA
| | - Lisa S. Liao
- grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - Kelli Malott
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92617 USA
| | - Veronique Perraud
- grid.266093.80000 0001 0668 7243Department of Chemistry, University of California Irvine, Irvine, CA 92617 USA
| | - Lisa M. Wingen
- grid.266093.80000 0001 0668 7243Department of Chemistry, University of California Irvine, Irvine, CA 92617 USA
| | - Rebecca J. Arechavala
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - Bishop Bliss
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - David A. Herman
- grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| | - Michael T. Kleinman
- grid.266093.80000 0001 0668 7243Department of Environmental and Occupational Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Center for Occupational and Environmental Health, University of California Irvine, 100 Theory Drive, Suite 100, Irvine, CA 92617 USA ,grid.266093.80000 0001 0668 7243Department of Medicine, University of California Irvine, Irvine, CA 92617 USA
| |
Collapse
|
47
|
Umeno K, Sasaki A, Kimura N. The impact of oocyte death on mouse primordial follicle formation and ovarian reserve. Reprod Med Biol 2022; 21:e12489. [PMID: 36329711 PMCID: PMC9623396 DOI: 10.1002/rmb2.12489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
Background Ovaries, the source of oocytes, maintain the numbers of primordial follicles, develop oocytes for fertilization and embryonic development. Although it is well known that about two-thirds of oocytes are lost during the formation of primordial follicles through cyst fragmentation and the aggregation of oocytes within the cyst, the mechanism responsible for this remains unclear. Methods We provide an overview of cell death that is associated with the oocyte cyst breakdown and primordial follicle assembly along with our recent findings for mice that had been treated with a TNFα ligand inhibitor. Main Findings It is generally accepted that apoptosis is the major mechanism responsible for the depletion of germ cells. In fact, a gene deficiency or the overexpression of apoptosis regulators can have a great effect on follicle numbers and/or fertility. Apoptosis, however, may not be the only cause of the large-scale oocyte attrition during oocyte cyst breakdown, and other mechanisms, such as aggregation, may also be involved in this process. Conclusion The continued study of oocyte death during primordial follicle formation could lead to the development of novel strategies for manipulating the primordial follicle pool, leading to improved fertility by enhancing the ovarian reserve.
Collapse
Affiliation(s)
- Ken Umeno
- Laboratory of Animal Reproduction, Graduate School of Agricultural ScienceYamagata UniversityTsuruokaJapan
| | - Ayana Sasaki
- Laboratory of Animal Reproduction, Graduate School of Agricultural ScienceYamagata UniversityTsuruokaJapan
| | - Naoko Kimura
- Laboratory of Animal Reproduction, Graduate School of Agricultural ScienceYamagata UniversityTsuruokaJapan
| |
Collapse
|
48
|
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
|
49
|
Xu PC, Luan Y, Yu SY, Xu J, Coulter DW, Kim SY. Effects of PD-1 blockade on ovarian follicles in a prepubertal female mouse. J Endocrinol 2021; 252:15-30. [PMID: 34647523 PMCID: PMC8630981 DOI: 10.1530/joe-21-0209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
Immunotherapy has emerged at the forefront of cancer treatment. Checkpoint inhibitor pembrolizumab (KEYTRUDA), a chimeric antibody which targets programmed cell death protein 1 (PD-1), has been approved by the Food and Drug Administration (FDA) for use in pediatric patients with relapsed or refractory classical Hodgkin's lymphoma. However, there is currently no published data regarding the effects of pembrolizumab on the ovary of female pediatric patients. In this study, prepubertal immunocompetent and immunodeficient female mice were injected with pembrolizumab or anti-mouse PD-1 antibody. The number of primordial follicles significantly decreased post-injection of both pembrolizumab and anti-mouse PD-1 antibody in immunocompetent mice. However, no changes in follicle numbers were observed in immunodeficient nude mice. Superovulation test and vaginal opening experiments suggest that there is no difference in the number of cumulus-oocyte complexes (COCs) and the timing of puberty onset between the control and anti-mouse PD-1 antibody treatment groups, indicating that there is no effect on short-term fertility. Elevation of pro-inflammatory cytokine TNF-α following COX-2 upregulation was observed in the ovary. CD3+ T-cell infiltration was detected within some ovarian follicles and between stromal cells of the ovaries in mice following treatment with anti-mouse PD-1 antibody. Thus, PD-1 immune checkpoint blockade affects the ovarian reserve through a mechanism possibly involving inflammation following CD3+ T-cell infiltration.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacology
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/pharmacology
- Cell Count
- Female
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/pharmacology
- Infertility, Female/chemically induced
- Infertility, Female/pathology
- Mice
- Mice, Nude
- Oocytes/cytology
- Oocytes/drug effects
- Ovarian Follicle/drug effects
- Ovarian Reserve/drug effects
- Ovary/drug effects
- Ovary/physiology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Sexual Maturation/drug effects
Collapse
Affiliation(s)
- Pauline C. Xu
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yi Luan
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Seok-Yeong Yu
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jing Xu
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon; Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Donald W. Coulter
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - So-Youn Kim
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
50
|
Schindler K. Inhibition of BIN2 extends reproductive lifespan. NATURE AGING 2021; 1:977-979. [PMID: 37118339 DOI: 10.1038/s43587-021-00132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Karen Schindler
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA.
- Department of Genetics, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|