1
|
Li Q, Zheng T, Chen J, Li B, Zhang Q, Yang S, Shao J, Guan W, Zhang S. Exploring melatonin's multifaceted role in female reproductive health: From follicular development to lactation and its therapeutic potential in obstetric syndromes. J Adv Res 2025; 70:223-242. [PMID: 38692429 PMCID: PMC11976432 DOI: 10.1016/j.jare.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Melatonin is mainly secreted by the pineal gland during darkness and regulates biological rhythms through its receptors in the suprachiasmatic nucleus of the hypothalamus. In addition, it also plays a role in the reproductive system by affecting the function of the hypothalamic-pituitary-gonadal axis, and by acting as a free radical scavenger thus contributing to the maintenance of the optimal physiological state of the gonads. Besides, melatonin can freely cross the placenta to influence fetal development. However, there is still a lack of overall understanding of the role of melatonin in the reproductive cycle of female mammals. AIM OF REVIEW Here we focus the role of melatonin in female reproduction from follicular development to delivery as well as the relationship between melatonin and lactation. We further summarize the potential role of melatonin in the treatment of preeclampsia, polycystic ovary syndrome, endometriosis, and ovarian aging. KEY SCIENTIFIC CONCEPTS OF REVIEW Understanding the physiological role of melatonin in female reproductive processes will contribute to the advancement of human fertility and reproductive medicine research.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
2
|
Hong P, Lu Y, Li H, Liu Z, Ou J, Li T, Shu Y. Predicting the risk of a high proportion of three/multiple pronuclei (3PN/MPN) zygotes in individual IVF cycles using comparative machine learning algorithms. Eur J Obstet Gynecol Reprod Biol 2025; 306:139-146. [PMID: 39826276 DOI: 10.1016/j.ejogrb.2025.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND The majority of machine learning applications in assisted reproduction have been focused on predicting the likelihood of pregnancy. In the present study, we aim to investigate which machine learning models are most effective in predicting the occurrence of a high proportion (>30 %) of 3PN/MPN zygotes in individual IVF cycles. METHODS Eight machine learning algorithms were trained and compared, including the AdaBoost and Gaussian NB. Data from IVF cycles carried out from September 2015 to September 2019 were used as a training set. Cycle data from October 2019 to June 2020 were used as a validation set to verify the training model. Cycles with a 3PN/MPN zygote proportion higher than 30 % were classified as high 3PN/MPN zygote proportion cycles. RESULTS The AdaBoost algorithm was the best model for model construction and external validation. In both the training and validation sets, age, basal FSH, FSH and E2 level on the day of Gonadotrophin (GN) stimulation, and FSH and LH levels on the day of HCG were statistically higher in patients with 3PN/MPN > 30 % than in patients with 3PN/MPN ≤ 30 %; AFC, AMH, E2 level on HCG day and total number of oocytes were lower in patients with 3PN/MPN > 30 % than in patients with 3PN/MPN ≤ 30 %. The top five predictors were the number of oocytes retrieved, age, male factor infertility, AFC, and total days of GN stimulation. CONCLUSION By applying a suitable machine learning algorithm, we can potentially predict the risk of a high proportion of 3PN/MPN zygotes in individual IVF cycles before insemination and avoid polyspermy fertilization by ICSI fertilization method.
Collapse
Affiliation(s)
- Pingping Hong
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Yaxin Lu
- Centre for Big Data and Artificial Intelligence, The third Affiliated Hospital of Sun Yat-sen University, No.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, China.
| | - Haiyang Li
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Zifeng Liu
- Centre for Big Data and Artificial Intelligence, The third Affiliated Hospital of Sun Yat-sen University, No.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, China.
| | - Jianpin Ou
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Tao Li
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Yimin Shu
- Center for Advanced Reproductive Medicine, Department of Obstetrics & Gynecology, University of Kansas Medical Center, Overland Park, KS 66211, USA.
| |
Collapse
|
3
|
Zhang F, Zhu M, Chen Y, Wang G, Yang H, Lu X, Li Y, Chang HM, Wu Y, Ma Y, Yuan S, Zhu W, Dong X, Zhao Y, Yu Y, Wang J, Mu L. Harnessing omics data for drug discovery and development in ovarian aging. Hum Reprod Update 2025:dmaf002. [PMID: 39977580 DOI: 10.1093/humupd/dmaf002] [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: 09/29/2024] [Revised: 12/02/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Ovarian aging occurs earlier than the aging of many other organs and has a lasting impact on women's overall health and well-being. However, effective interventions to slow ovarian aging remain limited, primarily due to an incomplete understanding of the underlying molecular mechanisms and drug targets. Recent advances in omics data resources, combined with innovative computational tools, are offering deeper insight into the molecular complexities of ovarian aging, paving the way for new opportunities in drug discovery and development. OBJECTIVE AND RATIONALE This review aims to synthesize the expanding multi-omics data, spanning genome, transcriptome, proteome, metabolome, and microbiome, related to ovarian aging, from both tissue-level and single-cell perspectives. We will specially explore how the analysis of these emerging omics datasets can be leveraged to identify novel drug targets and guide therapeutic strategies for slowing and reversing ovarian aging. SEARCH METHODS We conducted a comprehensive literature search in the PubMed database using a range of relevant keywords: ovarian aging, age at natural menopause, premature ovarian insufficiency (POI), diminished ovarian reserve (DOR), genomics, transcriptomics, epigenomics, DNA methylation, RNA modification, histone modification, proteomics, metabolomics, lipidomics, microbiome, single-cell, genome-wide association studies (GWAS), whole-exome sequencing, phenome-wide association studies (PheWAS), Mendelian randomization (MR), epigenetic target, drug target, machine learning, artificial intelligence (AI), deep learning, and multi-omics. The search was restricted to English-language articles published up to September 2024. OUTCOMES Multi-omics studies have uncovered key mechanisms driving ovarian aging, including DNA damage and repair deficiencies, inflammatory and immune responses, mitochondrial dysfunction, and cell death. By integrating multi-omics data, researchers can identify critical regulatory factors and mechanisms across various biological levels, leading to the discovery of potential drug targets. Notable examples include genetic targets such as BRCA2 and TERT, epigenetic targets like Tet and FTO, metabolic targets such as sirtuins and CD38+, protein targets like BIN2 and PDGF-BB, and transcription factors such as FOXP1. WIDER IMPLICATIONS The advent of cutting-edge omics technologies, especially single-cell technologies and spatial transcriptomics, has provided valuable insights for guiding treatment decisions and has become a powerful tool in drug discovery aimed at mitigating or reversing ovarian aging. As technology advances, the integration of single-cell multi-omics data with AI models holds the potential to more accurately predict candidate drug targets. This convergence offers promising new avenues for personalized medicine and precision therapies, paving the way for tailored interventions in ovarian aging. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Fengyu Zhang
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ming Zhu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yi Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Guiquan Wang
- Xiamen Key Laboratory of Reproduction and Genetics, Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinmei Lu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Li
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Yang Wu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuai Yuan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Wencheng Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jia Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Zhan CL, Zhou D, Sun MH, Jiang WJ, Lee SH, Li XH, Lu QY, Kim JD, Lee GH, Sim JM, Chung HJ, Cho ES, Sa SJ, Cui XS. In Vivo-Matured Oocyte Resists Post-Ovulatory Aging through the Hub Genes DDX18 and DNAJC7 in Pigs. Antioxidants (Basel) 2024; 13:867. [PMID: 39061935 PMCID: PMC11274268 DOI: 10.3390/antiox13070867] [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: 03/28/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Assisted reproduction technology (ART) procedures are often impacted by post-ovulatory aging (POA), which can lead to reduced fertilization rates and impaired embryo development. This study used RNA sequencing analysis and experimental validation to study the similarities and differences between in vivo- and vitro-matured porcine oocytes before and after POA. Differentially expressed genes (DEGs) between fresh in vivo-matured oocyte (F_vivo) and aged in vivo-matured oocyte (A_vivo) and DEGs between fresh in vitro-matured oocyte (F_vitro) and aged in vitro-matured oocyte (A_vitro) were intersected to explore the co-effects of POA. It was found that "organelles", especially "mitochondria", were significantly enriched Gene Ontology (GO) terms. The expression of genes related to the "electron transport chain" and "cell redox homeostasis" pathways related to mitochondrial function significantly showed low expression patterns in both A_vivo and A_vitro groups. Weighted correlation network analysis was carried out to explore gene expression modules specific to A_vivo. Trait-module association analysis showed that the red modules were most associated with in vivo aging. There are 959 genes in the red module, mainly enriched in "RNA binding", "mRNA metabolic process", etc., as well as in GO terms, and "spliceosome" and "nucleotide excision repair" pathways. DNAJC7, IK, and DDX18 were at the hub of the gene regulatory network. Subsequently, the functions of DDX18 and DNAJC7 were verified by knocking down their expression at the germinal vesicle (GV) and Metaphase II (MII) stages, respectively. Knockdown at the GV stage caused cell cycle disorders and increase the rate of abnormal spindle. Knockdown at the MII stage resulted in the inefficiency of the antioxidant melatonin, increasing the level of intracellular oxidative stress, and in mitochondrial dysfunction. In summary, POA affects the organelle function of oocytes. A_vivo oocytes have some unique gene expression patterns. These genes may be potential anti-aging targets. This study provides a better understanding of the detailed mechanism of POA and potential strategies for improving the success rates of assisted reproductive technologies in pigs and other mammalian species.
Collapse
Affiliation(s)
- Cheng-Lin Zhan
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Dongjie Zhou
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Ming-Hong Sun
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Wen-Jie Jiang
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Song-Hee Lee
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Xiao-Han Li
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Qin-Yue Lu
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Ji-Dam Kim
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Gyu-Hyun Lee
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Jae-Min Sim
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| | - Hak-Jae Chung
- The Center for Reproductive Control, TNT Research Co., Ltd., Jiphyeonjungang 3-gil 13, Sejong-si 30141, Republic of Korea;
| | - Eun-Seok Cho
- Swine Science Division, National Institute of Animal Science, RDA, Cheonan-si 31000, Republic of Korea;
| | - Soo-Jin Sa
- Planning and Coordination Division, National Institute of Animal Science, Iseo-myeon, Wanju-gun 55365, Republic of Korea;
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-L.Z.); (D.Z.); (M.-H.S.); (W.-J.J.); (S.-H.L.); (X.-H.L.); (Q.-Y.L.); (J.-D.K.); (G.-H.L.); (J.-M.S.)
| |
Collapse
|
5
|
Huang W, Li X, Yang H, Huang H. The impact of maternal age on aneuploidy in oocytes: Reproductive consequences, molecular mechanisms, and future directions. Ageing Res Rev 2024; 97:102292. [PMID: 38582380 DOI: 10.1016/j.arr.2024.102292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/12/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Age-related aneuploidy in human oocytes is a major factor contributing to decreased fertility and adverse reproductive outcomes. As females age, their oocytes are more prone to meiotic chromosome segregation errors, leading primarily to aneuploidy. Elevated aneuploidy rates have also been observed in oocytes from very young, prepubertal conceptions. A key barrier to developing effective treatments for age-related oocyte aneuploidy is our incomplete understanding of the molecular mechanisms involved. The challenge is becoming increasingly critical as more people choose to delay childbearing, a trend that has significant societal implications. In this review, we summarize current knowledge regarding the process of oocyte meiosis and folliculogenesis, highlighting the relationship between age and chromosomal aberrations in oocytes and embryos, and integrate proposed mechanisms of age-related meiotic disturbances across structural, protein, and genomic levels. Our goal is to spur new research directions and therapeutic avenues.
Collapse
Affiliation(s)
- Weiwei Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Xinyuan Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Hongbo Yang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Department of Obstetrics and Gynecology, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
Kakinuma K, Kakinuma T. Significance of oxidative stress and antioxidant capacity tests as biomarkers of premature ovarian insufficiency: A case control study. World J Clin Cases 2024; 12:479-487. [PMID: 38322464 PMCID: PMC10841946 DOI: 10.12998/wjcc.v12.i3.479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/27/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a condition that causes secondary amenorrhea owing to ovarian hypofunction at an early stage. Early follicular depletion results in intractable infertility, thereby considerably reducing the quality of life of females. Given the continuum in weakened ovarian function, progressing from incipient ovarian failure (IOF) to transitional ovarian failure and further to POI, it is necessary to develop biomarkers for predicting POI. The oxidative stress states in IOF and POI were comprehensively evaluated via oxidative stress [diacron-reactive oxygen metabolites (d-ROMs)] test and antioxidant capacity [biological antioxidant potential (BAP)]. AIM To explore the possibilities of oxidative stress and antioxidant capacity as biomarkers for the early detection of POI. METHODS Females presenting with secondary amenorrhea over 4 mo and a follicle stimulating hormone level of > 40 mIU/mL were categorized into the POI group. Females presenting with a normal menstrual cycle and a follicle stimulating hormone level of > 10.2 mIU/mL were categorized into the IOF group. Healthy females without ovarian hypofunction were categorized into the control group. Among females aged < 40 years who visited our hospital from January 2021 to June 2022, we recruited 11 patients into both POI and IOF groups. For the potential antioxidant capacity, the relative oxidative stress index (BAP/d-ROMs × 100) was calculated, and the oxidative stress defense system was comprehensively evaluated. RESULTS d-ROMs were significantly higher in the POI and IOF groups than in the control group, (478.2 ± 58.7 U.CARR, 434.5 ± 60.6 U.CARR, and 341.1 ± 35.1 U.CARR, respectively) (U.CARR is equivalent to 0.08 mg/dL of hydrogen peroxide). However, no significant difference was found between the POI and IOF groups. Regarding BAP, no significant difference was found between the control, IOF, and POI groups (2078.5 ± 157.4 μmol/L, 2116.2 ± 240.2 μmol/L, and 2029.0 ± 186.4 μmol/L, respectively). The oxidative stress index was significantly higher in the POI and IOF groups than in the control group (23.7 ± 3.3, 20.7 ± 3.6, and 16.5 ± 2.1, respectively). However, no significant difference was found between the POI and IOF groups. CONCLUSION High levels of oxidative stress suggest that evaluating the oxidative stress state may be a useful indicator for the early detection of POI.
Collapse
Affiliation(s)
- Kaoru Kakinuma
- Department of Obstetrics and Gynecology, International University of Health and Welfare Hospital, Tochigi 329-2763, Japan
- Graduate School of Medicine, International University of Health and Welfare, Tokyo 107-8402, Japan
| | - Toshiyuki Kakinuma
- Department of Obstetrics and Gynecology, International University of Health and Welfare Hospital, Tochigi 329-2763, Japan
| |
Collapse
|
7
|
Ducreux B, Patrat C, Trasler J, Fauque P. Transcriptomic integrity of human oocytes used in ARTs: technical and intrinsic factor effects. Hum Reprod Update 2024; 30:26-47. [PMID: 37697674 DOI: 10.1093/humupd/dmad025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/04/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Millions of children have been born throughout the world thanks to ARTs, the harmlessness of which has not yet been fully demonstrated. For years, efforts to evaluate the specific effects of ART have focused on the embryo; however, it is the oocyte quality that mainly dictates first and foremost the developmental potential of the future embryo. Ovarian stimulation, cryopreservation, and IVM are sometimes necessary steps to obtain a mature oocyte, but they could alter the appropriate expression of the oocyte genome. Additionally, it is likely that female infertility, environmental factors, and lifestyle have a significant influence on oocyte transcriptomic quality, which may interfere with the outcome of an ART attempt. OBJECTIVE AND RATIONALE The objective of this review is to identify transcriptomic changes in the human oocyte caused by interventions specific to ART but also intrinsic factors such as age, reproductive health issues, and lifestyle. We also provide recommendations for future good practices to be conducted when attempting ART. SEARCH METHODS An in-depth literature search was performed on PubMed to identify studies assessing the human oocyte transcriptome following ART interventions, or in the context of maternal aging, suboptimal lifestyle, or reproductive health issues. OUTCOMES ART success is susceptible to external factors, maternal aging, lifestyle factors (smoking, BMI), and infertility due to endometriosis or polycystic ovary syndrome. Indeed, all of these are likely to increase oxidative stress and alter mitochondrial processes in the foreground. Concerning ART techniques themselves, there is evidence that different ovarian stimulation regimens shape the oocyte transcriptome. The perturbation of processes related to the mitochondrion, oxidative phosphorylation, and metabolism is observed with IVM. Cryopreservation might dysregulate genes belonging to transcriptional regulation, ubiquitination, cell cycle, and oocyte growth pathways. For other ART laboratory factors such as temperature, oxygen tension, air pollution, and light, the evidence remains scarce. Focusing on genes involved in chromatin-based processes such as DNA methylation, heterochromatin modulation, histone modification, and chromatin remodeling complexes, but also genomic imprinting, we observed systematic dysregulation of such genes either after ART intervention or lifestyle exposure, as well as due to internal factors such as maternal aging and reproductive diseases. Alteration in the expression of such epigenetic regulators may be a common mechanism linked to adverse oocyte environments, explaining global transcriptomic modifications. WIDER IMPLICATIONS Many IVF factors and additional external factors have the potential to impair oocyte transcriptomic integrity, which might not be innocuous for the developing embryo. Fortunately, it is likely that such dysregulations can be minimized by adapting ART protocols or reducing adverse exposure.
Collapse
Affiliation(s)
- Bastien Ducreux
- Université Bourgogne Franche-Comtés-Equipe Génétique des Anomalies du Développement (GAD) INSERM UMR1231, Dijon, France
| | - Catherine Patrat
- Université de Paris Cité, Faculty of Medicine, Inserm 1016, Paris, France
- Department of Reproductive Biology-CECOS, aphp.centre-Université Paris Cité, Paris, France
| | - Jacquetta Trasler
- Department of Pediatrics, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Human Genetics, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pharmacology & Therapeutics, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Patricia Fauque
- Université Bourgogne Franche-Comtés-Equipe Génétique des Anomalies du Développement (GAD) INSERM UMR1231, Dijon, France
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction-CECOS, Dijon, France
| |
Collapse
|
8
|
Dias Nunes J, Demeestere I, Devos M. BRCA Mutations and Fertility Preservation. Int J Mol Sci 2023; 25:204. [PMID: 38203374 PMCID: PMC10778779 DOI: 10.3390/ijms25010204] [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: 11/28/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Hereditary cancers mostly affect the adolescent and young adult population (AYA) at reproductive age. Mutations in BReast CAncer (BRCA) genes are responsible for the majority of cases of hereditary breast and ovarian cancer. BRCA1 and BRCA2 act as tumor suppressor genes as they are key regulators of DNA repair through homologous recombination. Evidence of the accumulation of DNA double-strand break has been reported in aging oocytes, while BRCA expression decreases, leading to the hypothesis that BRCA mutation may impact fertility. Moreover, patients exposed to anticancer treatments are at higher risk of fertility-related issues, and BRCA mutations could exacerbate the treatment-induced depletion of the ovarian reserve. In this review, we summarized the functions of both genes and reported the current knowledge on the impact of BRCA mutations on ovarian ageing, premature ovarian insufficiency, female fertility preservation strategies and insights about male infertility. Altogether, this review provides relevant up-to-date information on the impact of BRCA1/2 mutations on fertility. Notably, BRCA-mutated patients should be adequately counselled for fertility preservation strategies, considering their higher sensitivity to chemotherapy gonadotoxic effects.
Collapse
Affiliation(s)
- Joana Dias Nunes
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
- Fertility Clinic, HUB-Erasme Hospital, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Melody Devos
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
| |
Collapse
|
9
|
Ducreux B, Ferreux L, Patrat C, Fauque P. Overview of Gene Expression Dynamics during Human Oogenesis/Folliculogenesis. Int J Mol Sci 2023; 25:33. [PMID: 38203203 PMCID: PMC10778858 DOI: 10.3390/ijms25010033] [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: 11/20/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
The oocyte transcriptome follows a tightly controlled dynamic that leads the oocyte to grow and mature. This succession of distinct transcriptional states determines embryonic development prior to embryonic genome activation. However, these oocyte maternal mRNA regulatory events have yet to be decoded in humans. We reanalyzed human single-oocyte RNA-seq datasets previously published in the literature to decrypt the transcriptomic reshuffles ensuring that the oocyte is fully competent. We applied trajectory analysis (pseudotime) and a meta-analysis and uncovered the fundamental transcriptomic requirements of the oocyte at any moment of oogenesis until reaching the metaphase II stage (MII). We identified a bunch of genes showing significant variation in expression from primordial-to-antral follicle oocyte development and characterized their temporal regulation and their biological relevance. We also revealed the selective regulation of specific transcripts during the germinal vesicle-to-MII transition. Transcripts associated with energy production and mitochondrial functions were extensively downregulated, while those associated with cytoplasmic translation, histone modification, meiotic processes, and RNA processes were conserved. From the genes identified in this study, some appeared as sensitive to environmental factors such as maternal age, polycystic ovary syndrome, cryoconservation, and in vitro maturation. In the future, the atlas of transcriptomic changes described in this study will enable more precise identification of the transcripts responsible for follicular growth and oocyte maturation failures.
Collapse
Affiliation(s)
- Bastien Ducreux
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD) INSERM UMR1231, 2 Rue Angélique Ducoudray, F-21000 Dijon, France;
| | - Lucile Ferreux
- Faculty of Medicine, Inserm 1016, Université de Paris Cité, F-75014 Paris, France; (L.F.); (C.P.)
- Department of Reproductive Biology-CECOS, Aphp.Centre-Université Paris Cité, Cochin, F-75014 Paris, France
| | - Catherine Patrat
- Faculty of Medicine, Inserm 1016, Université de Paris Cité, F-75014 Paris, France; (L.F.); (C.P.)
- Department of Reproductive Biology-CECOS, Aphp.Centre-Université Paris Cité, Cochin, F-75014 Paris, France
| | - Patricia Fauque
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD) INSERM UMR1231, 2 Rue Angélique Ducoudray, F-21000 Dijon, France;
- Laboratoire de Biologie de la Reproduction-CECOS, CHU Dijon Bourgogne, 14 Rue Gaffarel, F-21000 Dijon, France
| |
Collapse
|
10
|
Qu J, Hu H, Niu H, Sun X, Li Y. Melatonin restores the declining maturation quality and early embryonic development of oocytes in aged mice. Theriogenology 2023; 210:110-118. [PMID: 37490796 DOI: 10.1016/j.theriogenology.2023.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/03/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023]
Abstract
With increase in women's age, the reproductive capability of female mammals decreases dramatically caused by age-related oxidative stress, coinciding with the decline in the ovarian reserve, and the quality and quantity of oocytes, which is the main determinant of female fertility. Melatonin, as an effective antioxidant and antiaging substance, is secreted by the pineal gland and been found in the follicular fluid as well, which has been turned out to enable to protect oocytes from oxidative stress during ovulation. However, the beneficial effects of melatonin on meiotic maturation in vitro and early embryo development of aged oocytes are still not fully understood. Thus, the aim of this study is to explore the potential mechanism of melatonin to improve the oocytes maturation and early embryonic development. The results suggested that oocyte quality decreased with age, whereas 10-6 M melatonin supplementation can significantly prompt the maturation quality of oocytes, the rate of fertilization and the formation rate of blastocyst. Mechanistic investigation indicated that melatonin supplementation not only restored the function of mitochondria by reducing reactive oxygen species (ROS) generation and early apoptosis, but also increased the level of ATP and total GSH through enhancing the mRNA expression levels of SIRT1, SIRT3, GPX4, SOD1 and SOD2. In conclusion, melatonin could alleviate the impairment of age-related oxidative stress to meiotic maturation and early embryonic development of oocytes. This study may provide a potential remediation strategy to improve the quality of oocytes from aged women and the efficiency of assisted reproductive technologies.
Collapse
Affiliation(s)
- Jingwen Qu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China; The Department of Animal and Veterinary Science, University of Vermont, Burlington, VT, 05405, USA.
| | - Huiru Hu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| | - Haoyuan Niu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| | - Xiaomei Sun
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| | - Yongjun Li
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
11
|
Shen J, Liu Y, Teng X, Jin L, Feng L, Sun X, Zhao F, Huang B, Zhong J, Chen Y, Wang L. Spatial Transcriptomics of Aging Rat Ovaries Reveals Unexplored Cell Subpopulations with Reduced Antioxidative Defense. Gerontology 2023; 69:1315-1329. [PMID: 37717573 DOI: 10.1159/000533922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
INTRODUCTION Ovarian aging is characterized by a gradual decline in quantity and quality of oocytes and lower chance of fertility. Better understanding the genetic modulation during ovarian aging can further address available treatment options for aging-related ovarian diseases and fertility preservation. METHODS A novel technique spatial transcriptomics (ST) was used to investigate the spatial transcriptome features of rat ovaries. Transcriptomes from ST spots in the young and aged ovaries were clustered using differentially expressed genes. These data were analyzed to determine the spatial organization of age-induced heterogeneity and potential mechanisms underlying ovarian aging. RESULTS In this study, ST technology was applied to profile the comprehensive spatial imaging in young and aged rat ovary. Fifteen ovarian cell clusters with distinct gene-expression signatures were identified. The gene expression dynamics of granulosa cell clusters revealed three sub-types with sequential developmental stages. Aged ovary showed a significant decrease in the number of granulosa cells from the antral follicle. Besides, a remarkable rearrangement of interstitial gland cells was detected in aging ovary. Further analysis of aging-associated transcriptional changes revealed that the disturbance of oxidative pathway was a crucial factor in ovarian aging. CONCLUSIONS This study firstly described an aging-related spatial transcriptome changes in ovary and identified the potential targets for prevention of ovarian aging. These data may provide the basis for further investigations of the diagnosis and treatment of aging-related ovarian disorders.
Collapse
Affiliation(s)
- Jiayu Shen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China,
| | - Yuanyuan Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyuan Teng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ligui Jin
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Feng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiwen Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bao Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinjie Zhong
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingying Chen
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liquan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Chen H, Feng Y, Chen C, Yu S. Both early and late maternal age at childbirth is associated with increasing odds of central obesity in offspring. Am J Hum Biol 2023; 35:e23898. [PMID: 36932653 DOI: 10.1002/ajhb.23898] [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: 10/10/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
OBJECTIVE Despite studies on offspring obesity and delayed parenthood, little attention has been paid to the central obesity of offspring. The purpose of this study was to test the hypothesis that maternal age at childbirth (MAC) was associated with central obesity in offspring among the adult population, and fasting insulin may play a role in this association as a mediating factor. METHODS A total of 423 adults (mean age 37.9 years, 37.1% female) were included. Information about maternal variables and other confounders was collected by face-to-face interview. Waist circumference and insulin were determined through physical measurements and biochemical examinations. Logistic regression model and restricted cubic spline model were used to analyze the relationship between MAC and central obesity of offspring. The mediating effect of fasting insulin levels on association between MAC and offspring waist circumference was also analyzed. RESULTS There was a nonlinear relationship between MAC and central obesity in offspring. Compared with subjects with MAC 27-32 years, those with MAC 21-26 years (OR = 1.814, 95% CI: 1.129-2.915) and MAC ≥33 years (OR = 3.337, 95% CI: 1.638-6.798) had higher odds to develop central obesity. Offspring fasting insulin was also higher in MAC 21-26 years and MAC ≥33 years compared with those with MAC 27-32 years. Taking the group MAC 27-32 years as reference, the mediating effect of fasting insulin levels on the waist circumference was 20.6% and 12.4% for MAC 21-26 years and ≥ 33 years, respectively. CONCLUSION MAC 27-32 years has the lowest odds of central obesity in offspring. Fasting insulin levels may have a partial mediating effect on the association between MAC and central obesity.
Collapse
Affiliation(s)
- Hongyun Chen
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yinhua Feng
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Changying Chen
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Academic Training Center, Institute for Hospital Management of Henan Province, Zhengzhou, China
| | - Songcheng Yu
- College of Public Health, Zhengzhou University, Zhengzhou, China
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Academic Training Center, Institute for Hospital Management of Henan Province, Zhengzhou, China
| |
Collapse
|
13
|
Bai K, Hao E, Huang CX, Yue QX, Wang DH, Shi L, Chen YF, Chen H, Huang RL. Melatonin alleviates ovarian function damage and oxidative stress induced by dexamethasone in the laying hens through FOXO1 signaling pathway. Poult Sci 2023; 102:102745. [PMID: 37302326 PMCID: PMC10276286 DOI: 10.1016/j.psj.2023.102745] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 06/13/2023] Open
Abstract
Oxidative stress can trigger follicular atresia, and decrease follicles quantity in each development stage, thereby alleviating reproductive activity. The induction of oxidative stress in chickens through intraperitoneal injection of dexamethasone is a reliable and stable method. Melatonin has been shown to mitigate oxidative stress in this model, but the underlying mechanism remains unclear. Therefore, this study aimed to investigate whether melatonin can recover aberrant antioxidant status induced by dexamethasone and the specific mechanism behind melatonin-dependent protection. A total of 150 healthy 40-wk-old Dawu Jinfeng laying hens with similar body weights and laying rates were randomly divided into three groups, with five replicates per group and 10 hens per replicate. The hens in the control group (NS) received intraperitoneal injections of normal saline for 30 d, the dexamethasone group (Dex+NS) received 20 mg/kg dose of dexamethasone for the first 15 d, followed by the 15 d of normal saline treatment. While in the melatonin group (Dex+Mel), dexamethasone (20 mg/kg dose) was injected intraperitoneally in the first 15 d, and melatonin (20 mg/kg/d) was injected in the last 15 d. The results showed that dexamethasone treatment significantly enhanced oxidative stress (P < 0.05), while melatonin not only inhibited the oxidative stress but also notably enhanced the antioxidant enzymes superoxide dismutase (SOD), catalase activity (CAT), glutathione peroxidase (GSH-Px), and antioxidant genes CAT, superoxide dismutase 1 (SOD1), glutathione peroxidase 3 (GPX3), and recombinant peroxiredoxin 3 (PRDX3) expression (P < 0.05). Melatonin treatment also markedly reduced 8-hydroxy deoxyguanosine (8-OHdG), malondialdehyde (MDA), and reactive oxygen species (ROS) levels (P < 0.05) and apoptotic genes Caspase-3, Bim, and Bax in the follicle. In the Dex+Mel group, the Bcl-2 and SOD1 protein levels were also increased (P < 0.05). Melatonin inhibited the forkhead Box Protein O1 (FOXO1) gene and its protein expression (P < 0.05). In general, this investigation revealed that melatonin might decrease oxidative stress and ROS by enhancing antioxidant enzymes and genes, activating the antiapoptotic genes, and inhibiting the FOXO1 pathway in laying hens.
Collapse
Affiliation(s)
- Kang Bai
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China
| | - Erying Hao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China
| | - Chen-Xuan Huang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China; Department of Animal Nutrition and Management, Swedish University of Agricultural Science, Uppsala 75007, Sweden
| | - Qiao-Xian Yue
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China; Department of Animal Breeding and Genetics, Swedish University of Agricultural Science, Uppsala 75007, Sweden
| | - De-He Wang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China
| | - Lei Shi
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China
| | - Yi-Fan Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China
| | - Hui Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China.
| | - Ren-Lu Huang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding Hebei 071001, China
| |
Collapse
|
14
|
Kakinuma K, Kakinuma T. Analysis of oxidative stress and antioxidative potential in premature ovarian insufficiency. World J Clin Cases 2023; 11:2684-2693. [PMID: 37214574 PMCID: PMC10198121 DOI: 10.12998/wjcc.v11.i12.2684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/20/2023] [Accepted: 03/23/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is characterized by an early decline in ovarian function, inducing secondary amenorrhea. While the cause of POI has not yet been identified, the function of mitochondria in the ovaries and the cytotoxicity associated with reactive oxygen species (ROS) have been implicated in follicle pool depletion and a decline in follicle quality. Recently developed tests have enabled easy measurement of diacron-reactive oxygen metabolites (d-ROMs) and biological antioxidant potential (BAP). The combination of these two tests is used to comprehensively assess oxidative stress in the blood.
AIM To comprehensively assess the oxidative stress of d-ROMs and BAP in POI.
METHODS Participants were classified into two groups: A POI group of 11 women aged < 40 years examined between January 2021 and June 2022 with a history of secondary amenorrhea for at least 4 mo in our hospital and an FSH value of ≥ 40 mIU/mL; and a control group of healthy women of the same age with normal ovarian function in our hospital. Plasma d-ROMs and BAP were measured in both these groups underwent. Differences between groups were assessed using the t-test.
RESULTS The mean age and mean body mass index (BMI) were 35.8 ± 3.0 years and 20.1 ± 1.9 kg/m2 in the control group and 35.8 ± 2.7 years and 19.4 ± 2.5 kg/m2 in the POI group, respectively. The mean gravidity and parity in control and POI groups were 0.6 ± 0.7 and 0.4 ± 0.5 and 0.6 ± 0.9 and 0.3 ± 0.5, respectively. The two groups did not differ significantly in terms of mean age, BMI, gravidity, or parity. The d-ROMs level was significantly higher in the POI group than in the control group (478.2 ± 58.7 vs 341.1 ± 35.1 U.CARR; P < 0.001); however, the BAP level did not significantly differ between the two groups (2078.5 ± 157.4 vs 2029.0 ± 186.4 μmol/L). The oxidase stress index (d-ROMs/BAP × 100) was significantly higher in the POI group than in the control group (23.7 ± 3.3 vs 16.5 ± 2.1; P < 0.001).
CONCLUSION Oxidative stress was significantly greater in the POI group than in the control group, suggesting oxidative stress as a factor that can serve as a POI biomarker.
Collapse
Affiliation(s)
- Kaoru Kakinuma
- Department of Obstetrics and Gynecology, International Health and Welfare Hospital, Nasushiobara 327-2763, Japan
| | - Toshiyuki Kakinuma
- Department of Obstetrics and Gynecology, International Health and Welfare Hospital, Nasushiobara 327-2763, Japan
| |
Collapse
|
15
|
Czajkowska K, Ajduk A. Mitochondrial activity and redox status in oocytes from old mice: The interplay between maternal and postovulatory aging. Theriogenology 2023; 204:18-30. [PMID: 37031516 DOI: 10.1016/j.theriogenology.2023.03.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Maternal aging has been reported to reduce oocyte quality and, in turn, lower the developmental potential of the resulting embryos. Here, we show that maternally aged oocytes display two strikingly different phenotypes: some have normal morphology, whereas others have significantly shrunk cytoplasm. The latter phenotype usually prevails in aged females. Our objective was to characterize both types of maternally aged oocytes and investigate the origins of this diversity. Importantly, our experiments indicate that shrunk maternally aged oocytes are severely compromised in terms of mitochondrial functionality as compared to their young or morphologically normal maternally aged counterparts: they display significantly decreased mitochondrial activity and lower amounts of ROS. In contrast, morphologically normal maternally aged oocytes had the same mitochondrial activity as young ones, while their ROS levels were higher. Surprisingly, the shrunk phenotype was completely absent in maternally aged oocytes that matured in vitro, suggesting that it is not caused inherently by maternal aging, but may be related to other factors, like postovulatory aging. Indeed, an additional culture of in vitro matured young and old oocytes (i.e., in vitro postovulatory aging) significantly decreased their mitochondrial activity and led to cytoplasm shrinkage. In vivo postovulatory aging had a similar effect on oocytes from both young and old females. Finally, we examined the developmental potential of oocytes obtained from aged females. Shrunk (i.e., most likely postovulatory aged) oocytes failed to become fertilized, whereas morphologically normal ones (i.e., most likely not subjected to postovulatory aging) underwent fertilization and subsequent cleavage divisions, although they achieved the 2-cell stage less frequently than morphologically normal oocytes from young females. Importantly, the quality of blastocysts as well as the live birth rate for morphologically normal oocytes from old and young females were similar. In summary, our data clearly indicate that two pools of oocytes present in oviducts of aged females differ significantly in their quality and developmental potential and that the more severely affected phenotype results most likely from a synergistic action of maternal and postovulatory aging.
Collapse
|
16
|
Takahashi N, Franciosi F, Daldello EM, Luong XG, Althoff P, Wang X, Conti M. CPEB1-dependent disruption of the mRNA translation program in oocytes during maternal aging. Nat Commun 2023; 14:416. [PMID: 36697412 PMCID: PMC9877008 DOI: 10.1038/s41467-023-35994-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
The molecular causes of deteriorating oocyte quality during aging are poorly defined. Since oocyte developmental competence relies on post-transcriptional regulations, we tested whether defective mRNA translation contributes to this decline in quality. Disruption in ribosome loading on maternal transcripts is present in old oocytes. Using a candidate approach, we detect altered translation of 3'-UTR-reporters and altered poly(A) length of the endogenous mRNAs. mRNA polyadenylation depends on the cytoplasmic polyadenylation binding protein 1 (CPEB1). Cpeb1 mRNA translation and protein levels are decreased in old oocytes. This decrease causes de-repression of Ccnb1 translation in quiescent oocytes, premature CDK1 activation, and accelerated reentry into meiosis. De-repression of Ccnb1 is corrected by Cpeb1 mRNA injection in old oocytes. Oocyte-specific Cpeb1 haploinsufficiency in young oocytes recapitulates all the translation phenotypes of old oocytes. These findings demonstrate that a dysfunction in the oocyte translation program is associated with the decline in oocyte quality during aging.
Collapse
Affiliation(s)
- Nozomi Takahashi
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Federica Franciosi
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,Reproductive and Developmental Biology Lab, Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 20133, Milan, Italy
| | - Enrico Maria Daldello
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Xuan G Luong
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Peter Althoff
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Xiaotian Wang
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Marco Conti
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA. .,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA. .,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
17
|
Yu CM, Fei-Liu, Zhang JH, Dai XL, Wang YF, Chen L. Analysis of the split insemination (IVF+ICSI) treatment in patients with borderline semen in first cycle. J Gynecol Obstet Hum Reprod 2022; 51:102491. [DOI: 10.1016/j.jogoh.2022.102491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
|
18
|
Hua L, Chen W, Meng Y, Qin M, Yan Z, Yang R, Liu Q, Wei Y, Zhao Y, Yan L, Qiao J. The combination of DNA methylome and transcriptome revealed the intergenerational inheritance on the influence of advanced maternal age. Clin Transl Med 2022; 12:e990. [PMID: 36103411 PMCID: PMC9473489 DOI: 10.1002/ctm2.990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/03/2022] [Accepted: 07/08/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The number of women delivering at advanced maternal age (AMA; > = 35) continuously increases in developed and high-income countries. Large cohort studies have associated AMA with increased risks of various pregnancy complications and adverse pregnancy outcomes, which raises great concerns about the adverse effect of AMA on the long-term health of offspring. Specific acquired characteristics of parents can be passed on to descendants through certain molecular mechanisms, yet the underlying connection between AMA-related alterations in parents and that in offspring remains largely uncharted. METHODS We profiled the DNA methylomes of paired parental peripheral bloods and cord bloods from 20 nuclear families, including 10 AMA and 10 Young, and additional transcriptomes of 10 paired maternal peripheral bloods and cord bloods. RESULTS We revealed that AMA induced aging-like changes in DNA methylome and gene expression in both parents and offspring. The expression changes in several genes, such as SLC28A3, were highly relevant to the disorder in DNA methylation. In addition, AMA-related differentially methylated regions (DMRs) identified in mother and offspring groups showed remarkable similarities in both genomic locations and biological functions, mainly involving neuron differentiation, metabolism, and histone modification pathways. AMA-related differentially expressed genes (DEGs) shared by mother and offspring groups were highly enriched in the processes of immune cell activation and mitotic nuclear division. We further uncovered developmental-dependent dynamics for the DNA methylation of intergenerationally correlated DMRs during pre-implantation embryonic development, as well as diverse gene expression patterns during gametogenesis and early embryonic development for those common AMA-related DEGs presenting intergenerational correlation, such as CD24. Moreover, some intergenerational DEGs, typified by HTRA3, also showed the same significant alterations in AMA MII oocyte or blastocyst. CONCLUSIONS Our results reveal potential intergenerational inheritance of both AMA-related DNA methylome and transcriptome and provide new insights to understand health problems in AMA offspring.
Collapse
Affiliation(s)
- Lingyue Hua
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Wei Chen
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Yan Meng
- Department of Obstetrics and GynecologyBeijing Jishuitan Hospital, Fourth Clinical College of Peking UniversityBeijingChina
| | - Meng Qin
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Zhiqiang Yan
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Rui Yang
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Qiang Liu
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Yuan Wei
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Center for Healthcare Quality Management in ObstetricsBeijingChina
| | - Yangyu Zhao
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Center for Healthcare Quality Management in ObstetricsBeijingChina
| | - Liying Yan
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Jie Qiao
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Peking UniversityMinistry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
- Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- Beijing Advanced Innovation Center for GenomicsBeijingChina
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
19
|
Derisoud E, Jouneau L, Dubois C, Archilla C, Jaszczyszyn Y, Legendre R, Daniel N, Peynot N, Dahirel M, Auclair-Ronzaud J, Wimel L, Duranthon V, Chavatte-Palmer P. Maternal age affects equine day 8 embryo gene expression both in trophoblast and inner cell mass. BMC Genomics 2022; 23:443. [PMID: 35705916 PMCID: PMC9199136 DOI: 10.1186/s12864-022-08593-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Breeding a mare until she is not fertile or even until her death is common in equine industry but the fertility decreases as the mare age increases. Embryo loss due to reduced embryo quality is partly accountable for this observation. Here, the effect of mare's age on blastocysts' gene expression was explored. Day 8 post-ovulation embryos were collected from multiparous young (YM, 6-year-old, N = 5) and older (OM, > 10-year-old, N = 6) non-nursing Saddlebred mares, inseminated with the semen of one stallion. Pure or inner cell mass (ICM) enriched trophoblast, obtained by embryo bisection, were RNA sequenced. Deconvolution algorithm was used to discriminate gene expression in the ICM from that in the trophoblast. Differential expression was analyzed with embryo sex and diameter as cofactors. Functional annotation and classification of differentially expressed genes and gene set enrichment analysis were also performed. RESULTS Maternal aging did not affect embryo recovery rate, embryo diameter nor total RNA quantity. In both compartments, the expression of genes involved in mitochondria and protein metabolism were disturbed by maternal age, although more genes were affected in the ICM. Mitosis, signaling and adhesion pathways and embryo development were decreased in the ICM of embryos from old mares. In trophoblast, ion movement pathways were affected. CONCLUSIONS This is the first study showing that maternal age affects gene expression in the equine blastocyst, demonstrating significant effects as early as 10 years of age. These perturbations may affect further embryo development and contribute to decreased fertility due to aging.
Collapse
Affiliation(s)
- Emilie Derisoud
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France.
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France.
| | - Luc Jouneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Cédric Dubois
- IFCE, Plateau technique de Chamberet, 19370, Chamberet, France
| | - Catherine Archilla
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Yan Jaszczyszyn
- Institute for Integrative Biology of the Cell (I2BC), UMR 9198 CNRS, CEA, Paris-Sud University F-91198, Gif-sur-Yvette, France
| | - Rachel Legendre
- Institut Pasteur-Bioinformatics and Biostatistics Hub-Department of Computational Biology, Paris, France
| | - Nathalie Daniel
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Nathalie Peynot
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Michèle Dahirel
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | | | - Laurence Wimel
- IFCE, Plateau technique de Chamberet, 19370, Chamberet, France
| | - Véronique Duranthon
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France.
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France.
| |
Collapse
|
20
|
Lombardo SD, Wangsaputra IF, Menche J, Stevens A. Network Approaches for Charting the Transcriptomic and Epigenetic Landscape of the Developmental Origins of Health and Disease. Genes (Basel) 2022; 13:764. [PMID: 35627149 PMCID: PMC9141211 DOI: 10.3390/genes13050764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 02/04/2023] Open
Abstract
The early developmental phase is of critical importance for human health and disease later in life. To decipher the molecular mechanisms at play, current biomedical research is increasingly relying on large quantities of diverse omics data. The integration and interpretation of the different datasets pose a critical challenge towards the holistic understanding of the complex biological processes that are involved in early development. In this review, we outline the major transcriptomic and epigenetic processes and the respective datasets that are most relevant for studying the periconceptional period. We cover both basic data processing and analysis steps, as well as more advanced data integration methods. A particular focus is given to network-based methods. Finally, we review the medical applications of such integrative analyses.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, 1030 Vienna, Austria;
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Ivan Fernando Wangsaputra
- Maternal and Fetal Health Research Group, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK;
| | - Jörg Menche
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, 1030 Vienna, Austria;
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1030 Vienna, Austria
- Faculty of Mathematics, University of Vienna, 1030 Vienna, Austria
| | - Adam Stevens
- Maternal and Fetal Health Research Group, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK;
| |
Collapse
|
21
|
Chromosome Segregation in the Oocyte: What Goes Wrong during Aging. Int J Mol Sci 2022; 23:ijms23052880. [PMID: 35270022 PMCID: PMC8911062 DOI: 10.3390/ijms23052880] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 12/13/2022] Open
Abstract
Human female fertility and reproductive lifespan decrease significantly with age, resulting in an extended post-reproductive period. The central dogma in human female reproduction contains two important aspects. One is the pool of oocytes in the human ovary (the ovarian reserve; approximately 106 at birth), which diminishes throughout life until menopause around the age of 50 (approximately 103 oocytes) in women. The second is the quality of oocytes, including the correctness of meiotic divisions, among other factors. Notably, the increased rate of sub- and infertility, aneuploidy, miscarriages, and birth defects are associated with advanced maternal age, especially in women above 35 years of age. This postponement is also relevant for human evolution; decades ago, the female aging-related fertility drop was not as important as it is today because women were having their children at a younger age. Spindle assembly is crucial for chromosome segregation during each cell division and oocyte maturation, making it an important event for euploidy. Consequently, aberrations in this segregation process, especially during the first meiotic division in human eggs, can lead to implantation failure or spontaneous abortion. Today, human reproductive medicine is also facing a high prevalence of aneuploidy, even in young females. However, the shift in the reproductive phase of humans and the strong increase in errors make the problem much more dramatic at later stages of the female reproductive phase. Aneuploidy in human eggs could be the result of the non-disjunction of entire chromosomes or sister chromatids during oocyte meiosis, but partial or segmental aneuploidies are also relevant. In this review, we intend to describe the relevance of the spindle apparatus during oocyte maturation for proper chromosome segregation in the context of maternal aging and the female reproductive lifespan.
Collapse
|
22
|
Bebbere D, Coticchio G, Borini A, Ledda S. Oocyte aging: looking beyond chromosome segregation errors. J Assist Reprod Genet 2022; 39:793-800. [PMID: 35212880 PMCID: PMC9051005 DOI: 10.1007/s10815-022-02441-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/18/2022] [Indexed: 11/30/2022] Open
Abstract
The age-associated decline in female fertility is largely ascribable to a decrease in oocyte quality. This phenomenon is multifaceted and influenced by numerous interconnected maternal and environmental factors. An increase in the rate of meiotic errors is the major cause of the decline in oocyte developmental competence. However, abnormalities in the ooplasm accumulating with age - including altered metabolism, organelle dysfunction, and aberrant gene regulation - progressively undermine oocyte quality. Stockpiling of maternal macromolecules during folliculogenesis is crucial, as oocyte competence to achieve maturation, fertilization, and the earliest phases of embryo development occur in absence of transcription. At the same time, crucial remodeling of oocyte epigenetics during oogenesis is potentially exposed to interfering factors, such as assisted reproduction technologies (ARTs) or environmental changes, whose impact may be enhanced by reproductive aging. As the effects of maternal aging on molecular mechanisms governing the function of the human oocyte remain poorly understood, studies in animal models are essential to deepen current understanding, with translational implications for human ARTs. The present mini review aims at offering an updated and consistent view of cytoplasmic alterations occurring in oocytes during aging, focusing particularly on gene and epigenetic regulation. Appreciation of these mechanisms could inspire solutions to mitigate/control the phenomenon, and thus benefit modern ARTs.
Collapse
Affiliation(s)
- Daniela Bebbere
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy.
| | | | | | - Sergio Ledda
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| |
Collapse
|
23
|
Pailas A, Niaka K, Zorzompokou C, Marangos P. The DNA Damage Response in Fully Grown Mammalian Oocytes. Cells 2022; 11:cells11050798. [PMID: 35269420 PMCID: PMC8909749 DOI: 10.3390/cells11050798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
DNA damage in cells can occur physiologically or may be induced by exogenous factors. Genotoxic damage may cause cancer, ageing, serious developmental diseases and anomalies. If the damage occurs in the germline, it can potentially lead to infertility or chromosomal and genetic aberrations in the developing embryo. Mammalian oocytes, the female germ cells, are produced before birth, remaining arrested at the prophase stage of meiosis over a long period of time. During this extensive state of arrest the oocyte may be exposed to different DNA-damaging insults for months, years or even decades. Therefore, it is of great importance to understand how these cells respond to DNA damage. In this review, we summarize the most recent developments in the understanding of the DNA damage response mechanisms that function in fully grown mammalian oocytes.
Collapse
Affiliation(s)
- Alexandros Pailas
- Department of Biological Applications and Technology, School of Health Sciences, Institute of Biosciences, University Research Centre, University of Ioannina, 45110 Ioannina, Greece
| | - Konstantina Niaka
- Department of Biological Applications and Technology, School of Health Sciences, Institute of Biosciences, University Research Centre, University of Ioannina, 45110 Ioannina, Greece
| | - Chrysoula Zorzompokou
- Department of Biological Applications and Technology, School of Health Sciences, Institute of Biosciences, University Research Centre, University of Ioannina, 45110 Ioannina, Greece
| | - Petros Marangos
- Department of Biological Applications and Technology, School of Health Sciences, Institute of Biosciences, University Research Centre, University of Ioannina, 45110 Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology, University of Ioannina Campus, 45115 Ioannina, Greece
| |
Collapse
|
24
|
Potabattula R, Trapphoff T, Dittrich M, Fic K, Ptak GE, Dieterle S, Haaf T. Ribosomal DNA methylation in human and mouse oocytes increases with age. Aging (Albany NY) 2022; 14:1214-1232. [PMID: 35157611 PMCID: PMC8876901 DOI: 10.18632/aging.203891] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
An age-dependent increase in ribosomal DNA (rDNA) methylation has been observed across a broad spectrum of somatic tissues and the male mammalian germline. Bisulfite pyrosequencing (BPS) was used to determine the methylation levels of the rDNA core promoter and the rDNA upstream control element (UCE) along with two oppositely genomically imprinted control genes (PEG3 and GTL2) in individual human germinal vesicle (GV) oocytes from 90 consenting women undergoing fertility treatment because of male infertility. Apart from a few (4%) oocytes with single imprinting defects (in either PEG3 or GTL2), the analyzed GV oocytes displayed correct imprinting patterns. In 95 GV oocytes from 42 younger women (26-32 years), the mean methylation levels of the rDNA core promoter and UCE were 7.4±4.0% and 9.3±6.1%, respectively. In 79 GV oocytes from 48 older women (33-39 years), methylation levels increased to 9.3±5.3% (P = 0.014) and 11.6±7.4% (P = 0.039), respectively. An age-related increase in oocyte rDNA methylation was also observed in 123 mouse GV oocytes from 29 4-16-months-old animals. Similar to the continuously mitotically dividing male germline, ovarian aging is associated with a gain of rDNA methylation in meiotically arrested oocytes. Oocytes from the same woman can exhibit varying rDNA methylation levels and, by extrapolation, different epigenetic ages.
Collapse
Affiliation(s)
- Ramya Potabattula
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | | | - Marcus Dittrich
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
- Department of Bioinformatics, Julius Maximilians University, Würzburg, Germany
| | - Kinga Fic
- Malopolska Centre of Biotechnology (MCB), Jagiellonian University, Krakow, Poland
| | - Grazyna E. Ptak
- Malopolska Centre of Biotechnology (MCB), Jagiellonian University, Krakow, Poland
| | - Stefan Dieterle
- Fertility Center, Dortmund, Germany
- Division of Reproductive Medicine and Infertility, Department of Obstetrics and Gynecology, Witten/Herdecke University, Dortmund, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| |
Collapse
|
25
|
Yang H, Kolben T, Kessler M, Meister S, Paul C, van Dorp J, Eren S, Kuhn C, Rahmeh M, Herbst C, Fink SG, Weimer G, Mahner S, Jeschke U, von Schönfeldt V. FAM111A Is a Novel Molecular Marker for Oocyte Aging. Biomedicines 2022; 10:257. [PMID: 35203468 PMCID: PMC8869572 DOI: 10.3390/biomedicines10020257] [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: 11/18/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Aging is the main cause of decline in oocyte quality, which can further trigger the failure of assisted reproductive technology (ART). Exploring age-related genes in oocytes is an important way to investigate the molecular mechanisms involved in oocyte aging. To provide novel insight into this field, we performed a pooled analysis of publicly available datasets, using the overlapping results of two statistical methods on two Gene Expression Omnibus (GEO) datasets. The methods utilized in the current study mainly include Spearman rank correlation, the Wilcoxon signed-rank test, t-tests, Venn diagrams, Gene Ontology (GO), Protein-Protein Interaction (PPI), Gene Set Enrichment Analysis (GSEA), Gene Set Variation Analysis (GSVA), and receiver operating characteristic (ROC) curve analysis. We identified hundreds of age-related genes across different gene expression datasets of in vitro maturation-metaphase II (IVM-MII) oocytes. Age-related genes in IVM-MII oocytes were involved in the biological processes of cellular metabolism, DNA replication, and histone modifications. Among these age-related genes, FAM111A expression presented a robust correlation with age, seen in the results of different statistical methods and different datasets. FAM111A is associated with the processes of chromosome segregation and cell cycle regulation. Thus, this enzyme is potentially an interesting novel marker for the aging of oocytes, and warrants further mechanistic study.
Collapse
Affiliation(s)
- Huixia Yang
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Thomas Kolben
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Mirjana Kessler
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Sarah Meister
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Corinna Paul
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Julia van Dorp
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Sibel Eren
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Martina Rahmeh
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Cornelia Herbst
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Sabine Gabriele Fink
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Gabriele Weimer
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Viktoria von Schönfeldt
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (M.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (C.H.); (S.G.F.); (G.W.); (S.M.); (V.v.S.)
| |
Collapse
|
26
|
He X, Wang Y, Wu M, Wei J, Sun X, Wang A, Hu G, Jia J. Secoisolariciresinol Diglucoside Improves Ovarian Reserve in Aging Mouse by Inhibiting Oxidative Stress. Front Mol Biosci 2022; 8:806412. [PMID: 35059437 PMCID: PMC8764264 DOI: 10.3389/fmolb.2021.806412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/13/2021] [Indexed: 12/31/2022] Open
Abstract
Ovarian reserve is a key factor in the reproductive function of the ovaries. Ovarian aging is characterized by a gradual decline in the quantity and quality of follicles. The underlying mechanism of ovarian aging is complex and age-related oxidative stress is considered one of the most likely factors. Secoisolariciresinol diglucoside (SDG) has been shown to have good scavenging ability against reactive oxygen species (ROS) which slowly accumulates in ovarian tissues. However, it is unknown whether SDG had beneficial effects on aging ovaries. In this study, we used 37-week-old female C57BL/6J mouse as a natural reproductive aging model to evaluate the role of SDG in ovarian aging. SDG (7 and 70 mg/kg) intragastric administration was performed in the mice daily. After 8 weeks, the effects of SDG on aging ovaries were evaluated by counting the number of follicles and the expression of follicle-stimulating hormone receptors (FSHR) in the ovary. The mechanism of SDG on the aging ovaries was further explored through ovarian metabolomics. It was found that SDG can effectively increase the number of growing follicles and increase the expression of the FSHR protein. The metabolomics results showed that the ovaries in the SDG intervention group achieved better uptake and transport of nutrients, including amino acids and glucose that are necessary for the development of oocytes. At the same time, the ovaries of the SDG intervention group showed that the drug reduced ROS generation. Additionally, we found that ovarian telomere length and ovarian mitochondrial DNA copy number that are highly susceptible to ROS damage and are also related to aging. The results showed that SDG can significantly increase mitochondrial DNA copy number and slow down the process of telomere shortening. These data indicate that SDG improves ovarian reserve by inhibiting oxidative stress.
Collapse
Affiliation(s)
- XueLai He
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yong Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - MeiQi Wu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - JiangChun Wei
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - XianDuo Sun
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - AnHua Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - GaoSheng Hu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - JingMing Jia
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
27
|
Huang R, Dong Y, Levy E, Julien P, Marc I, He H, Xu YJ, Wei SQ, Fraser WD, Luo ZC. Large-for-Gestational-Age, Leptin, and Adiponectin in Infancy. J Clin Endocrinol Metab 2022; 107:e688-e697. [PMID: 34477199 DOI: 10.1210/clinem/dgab642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Fetal overgrowth "programs" an elevated risk of obesity and type 2 diabetes in adulthood. Plausibly, adipokines may be involved in programming metabolic health. OBJECTIVE This work aimed to evaluate whether large-for-gestational-age (LGA), an indicator of fetal overgrowth, is associated with altered circulating leptin and adiponectin levels in infancy, and assess the determinants. METHODS In the Canadian 3D birth cohort, we studied 70 LGA (birth weight > 90th percentile) and 140 optimal-for-gestational-age (OGA, 25th-75th percentiles) infants matched by maternal ethnicity, smoking, and gestational age at delivery. The primary outcomes were fasting leptin, and total and high-molecular-weight (HMW) adiponectin concentrations at age 2 years. RESULTS LGA infants had higher body mass index (BMI) than OGA infants. However, there were no significant differences in leptin, and total and HMW adiponectin concentrations. Leptin concentrations were positively associated with female sex, weight (z score) gain 0 to 24 months, current BMI, and the sum of triceps and subscapular skinfold thickness, and negatively associated with maternal age and White ethnicity. Female sex was associated with lower total and HMW adiponectin concentrations. Weight (z score) gain 0 to 24 months and current BMI were positively correlated with total and HMW adiponectin concentrations in LGA infants only. CONCLUSION This study is the first to demonstrate that LGA does not matter for circulating leptin and adiponectin concentrations in infancy, and there may be LGA-specific positive associations between weight gain or current BMI and adiponectin concentrations in infancy, suggesting dysfunction in establishing the adiposity-adiponectin negative feedback loop in LGA individuals.
Collapse
Affiliation(s)
- Rong Huang
- Department of Obstetrics and Gynecology, Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, M5G 1X5, Canada
- Sainte-Justine Hospital Research Center, University of Montreal, Montreal, H3T 1C5, Canada
| | - Yu Dong
- Department of Obstetrics and Gynecology, Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, M5G 1X5, Canada
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200092, China
| | - Emile Levy
- Sainte-Justine Hospital Research Center, University of Montreal, Montreal, H3T 1C5, Canada
| | - Pierre Julien
- CHU de Quebec-Laval University Research Center, Laval University, Quebec City G1V 4G2, Canada
| | - Isabelle Marc
- CHU de Quebec-Laval University Research Center, Laval University, Quebec City G1V 4G2, Canada
| | - Hua He
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200092, China
| | - Ya-Jie Xu
- Department of Obstetrics and Gynecology, Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, M5G 1X5, Canada
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200092, China
| | - Shu-Qin Wei
- Sainte-Justine Hospital Research Center, University of Montreal, Montreal, H3T 1C5, Canada
| | - William D Fraser
- Sainte-Justine Hospital Research Center, University of Montreal, Montreal, H3T 1C5, Canada
- Department of Obstetrics and Gynecology, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, University of Sherbrooke, Sherbrooke J1H 5N4, Canada
| | - Zhong-Cheng Luo
- Department of Obstetrics and Gynecology, Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, M5G 1X5, Canada
- Sainte-Justine Hospital Research Center, University of Montreal, Montreal, H3T 1C5, Canada
| |
Collapse
|
28
|
Finelli R, Moreira BP, Alves MG, Agarwal A. Unraveling the Molecular Impact of Sperm DNA Damage on Human Reproduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1358:77-113. [DOI: 10.1007/978-3-030-89340-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
29
|
Repalle D, Saritha KV, Bhandari S, Chittora M, Choudhary J. Role of Female Age in Regulating the Effect of Sperm DNA Fragmentation on the Live Birth Rates in Intracytoplasmic Sperm Injection Cycles with Own and Donor Oocytes. J Hum Reprod Sci 2022; 15:64-71. [PMID: 35494199 PMCID: PMC9053350 DOI: 10.4103/jhrs.jhrs_150_21] [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: 10/23/2021] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Sperm DNA integrity assessment has been progressively used as an unfettered measure of sperm as it proffers more prognostic and diagnostic information than routine semen analysis. The contentious effect of sperm DNA fragmentation (SDF) on clinical outcomes can be attributed to female factors such as age, oocyte quality and ovarian reserve. Aims The study is mainly aimed to know the influence of SDF on the live birth rates in intracytoplasmic sperm injection (ICSI) cycles with own and donor oocytes. Second, to know the role of female age in regulating the effect of SDF on the live birth rates in ICSI cycles with own and donor oocytes. Setting and Design A prospective cohort study was done at our tertiary care centre attached to the reproductive medicine unit in medical college. Materials and Methods The study included 356 patients who underwent first ICSI cycles either with own or donor-oocytes along with day 5 fresh embryo transfers only. The main outcome measures were live birth rates and miscarriage rates. Statistical Analysis Used Chi-squared test was used to compare the categorical variables between the groups. The receiver operating characteristic curve was developed to correlate the female age with the live birth rate. Results A significant decrease in the live birth rates (42.85% vs. 26.15%, P = 0.023) and an increase in the miscarriage rates (12.30% vs. 34.61%, P = 0.013) were observed in the high-SDF group ICSI cycles of own-oocyte patients. However, there was no significant difference in the live birth rates and miscarriage rates in the low- and high-SDF groups of donor oocyte ICSI cycle patients (P > 0.05). The own-oocyte ICSI cycle patients were further stratified based on the female age. In the female age group ≤30 years there was no significant difference in the live birth and miscarriage rates (P > 0.05) similar to donor oocyte ICSI cycles. Whereas, there was a significant difference in the live birth rates in the females of age >30 years (13.79% vs. 34.37%, P = 0.040). Conclusion In conclusion, high-SDF has a negative influence on the live birth rates and a positive influence on the miscarriage rates in patients with own-oocyte ICSI cycles. A similar influence was not observed in patients with donor-oocyte ICSI cycles and in young female patients (age ≤30 years) with own-oocyte ICSI cycles.
Collapse
Affiliation(s)
- Deepthi Repalle
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| | - K. V. Saritha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| | - Shilpa Bhandari
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Megha Chittora
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Jitendra Choudhary
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| |
Collapse
|
30
|
Saritha KV, Repalle D, Bhandari S. Sperm DNA fragmentation does not affect the clinical outcomes in the cumulative transfers of an ICSI cycle along with blastocyst transfers in couples with normozoospermic male patients. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2022. [DOI: 10.4103/2305-0500.346090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
31
|
Blengini CS, Nguyen AL, Aboelenain M, Schindler K. Age-dependent integrity of the meiotic spindle assembly checkpoint in females requires Aurora kinase B. Aging Cell 2021; 20:e13489. [PMID: 34704342 PMCID: PMC8590096 DOI: 10.1111/acel.13489] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/09/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
A hallmark of advanced maternal age is a significant increase in meiotic chromosome segregation errors, resulting in early miscarriages and congenital disorders. These errors most frequently occur during meiosis I (MI). The spindle assembly checkpoint (SAC) prevents chromosome segregation errors by arresting the cell cycle until proper chromosome alignment is achieved. Unlike in mitosis, the SAC in oocytes is desensitized, allowing chromosome segregation in the presence of improperly aligned chromosomes. Whether SAC integrity further deteriorates with advancing maternal age, and if this decline contributes to increased segregation errors remains a fundamental question. In somatic cells, activation of the SAC depends upon Aurora kinase B (AURKB), which functions to monitor kinetochore–microtubule attachments and recruit SAC regulator proteins. In mice, oocyte‐specific deletion of AURKB (Aurkb cKO) results in an increased production of aneuploid metaphase II‐arrested eggs and premature age‐related infertility. Here, we aimed to understand the cause of the short reproductive lifespan and hypothesized that SAC integrity was compromised. In comparing oocytes from young and sexually mature Aurkb cKO females, we found that SAC integrity becomes compromised rapidly with maternal age. We show that the increased desensitization of the SAC is driven by reduced expression of MAD2, ZW10 and Securin proteins, key contributors to the SAC response pathway. The reduced expression of these proteins is the result of altered protein homeostasis, likely caused by the accumulation of reactive oxygen species. Taken together, our results demonstrate a novel function for AURKB in preserving the female reproductive lifespan possibly by protecting oocytes from oxidative stress.
Collapse
Affiliation(s)
- Cecilia S. Blengini
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
| | - Alexandra L. Nguyen
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
| | - Mansour Aboelenain
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
- Department of Theriogenology Faculty of Veterinary Medicine Mansoura University Mansoura Egypt
| | - Karen Schindler
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
| |
Collapse
|
32
|
In vitro fertilization-embryo transfer in patients with unexplained recurrent pregnancy loss. Chin Med J (Engl) 2021; 134:2421-2429. [PMID: 34669635 PMCID: PMC8654439 DOI: 10.1097/cm9.0000000000001657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: Empiric therapy for patients with unexplained recurrent pregnancy loss (URPL) is not precise. Some patients will ask for assisted reproductive technology due to secondary infertility or advanced maternal age. The clinical outcomes of URPL patients who have undergone in vitro fertilization-embryo transfer (IVF-ET) require elucidation. The IVF outcome and influencing factors of URPL patients need further study. Methods: A retrospective cohort study was designed, and 312 infertile patients with URPL who had been treated during January 2012 to December 2015 in the Reproduction Center of Peking University Third Hospital were included. By comparing clinical outcomes between these patients and those with tubal factor infertility (TFI), the factors affecting the clinical outcomes of URPL patients were analyzed. Results: The clinical pregnancy rate (35.18% vs. 34.52% in fresh ET cycles, P = 0.877; 34.48% vs. 40.27% in frozen-thawed ET cycles, P = 0.283) and live birth rate (LBR) in fresh ET cycles (27.67% vs. 26.59%, P = 0.785) were not significantly different between URPL group and TFI group. URPL group had lower LBR in frozen-thawed ET cycles than that of TFI group (23.56% vs. 33.56%, P = 0.047), but the cumulative LBRs (34.69% vs. 38.26%, P = 0.368) were not significantly different between the two groups. The increased endometrial thickness (EMT) on the human chorionic gonadotropin day (odds ratio [OR]: 0.848, 95% confidence interval [CI]: 0.748–0.962, P = 0.010) and the increased number of eggs retrieved (OR: 0.928, 95% CI: 0.887–0.970, P = 0.001) were protective factors for clinical pregnancy in stimulated cycles. The increased number of eggs retrieved (OR: 0.875, 95% CI: 0.846–0.906, P < 0.001), the increased two-pronucleus rate (OR: 0.151, 95% CI: 0.052–0.437, P < 0.001), and increased EMT (OR: 0.876, 95% CI: 0.770–0.997, P = 0.045) in ET day were protective factors for the cumulative live birth outcome. Conclusion: After matching ages, no significant differences in clinical outcomes were found between the patients with URPL and the patients with TFI. A thicker endometrium and more retrieved oocytes increase the probability of pregnancy in fresh transfer cycles, but a better normal fertilization potential will increase the possibility of a live birth.
Collapse
|
33
|
Palla G, Pollner P, Börcsök J, Major A, Molnár B, Csabai I. Hierarchy and control of ageing-related methylation networks. PLoS Comput Biol 2021; 17:e1009327. [PMID: 34534207 PMCID: PMC8480875 DOI: 10.1371/journal.pcbi.1009327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/29/2021] [Accepted: 08/05/2021] [Indexed: 11/28/2022] Open
Abstract
DNA methylation provides one of the most widely studied biomarkers of ageing. Since the methylation of CpG dinucleotides function as switches in cellular mechanisms, it is plausible to assume that by proper adjustment of these switches age may be tuned. Though, adjusting hundreds of CpG methylation levels coherently may never be feasible and changing just a few positions may lead to biologically unstable state. A prominent example of methylation-based age estimators is provided by Horvath's clock, based on 353 CpG dinucleotides, showing a high correlation (not necessarily causation) with chronological age across multiple tissue types. On this small subset of CpG dinucleotides we demonstrate how the adjustment of one methylation level leads to a cascade of changes at other sites. Among the studied subset, we locate the most important CpGs (and related genes) that may have a large influence on the rest of the sub-system. According to our analysis, the structure of this network is way more hierarchical compared to what one would expect based on ensembles of uncorrelated connections. Therefore, only a handful of CpGs is enough to modify the system towards a desired state. When propagation of the change over the network is taken into account, the resulting modification in the predicted age can be significantly larger compared to the effect of isolated CpG perturbations. By adjusting the most influential single CpG site and following the propagation of methylation level changes we can reach up to 5.74 years in virtual age reduction, significantly larger than without taking into account of the network control. Extending our approach to the whole methylation network may identify key nodes that have controller role in the ageing process.
Collapse
Affiliation(s)
- Gergely Palla
- Health Services Management Training Centre, Semmelweis University, Budapest, Hungary
- MTA-ELTE Statistical and Biological Physics Research Group, Dept. of Biological Physics, Eötvös University, Budapest, Hungary
| | - Péter Pollner
- Health Services Management Training Centre, Semmelweis University, Budapest, Hungary
- MTA-ELTE Statistical and Biological Physics Research Group, Dept. of Biological Physics, Eötvös University, Budapest, Hungary
| | - Judit Börcsök
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - András Major
- Dept. of Physics of Complex Systems, ELTE Eötvös University, Budapest, Hungary
| | - Béla Molnár
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - István Csabai
- Dept. of Physics of Complex Systems, ELTE Eötvös University, Budapest, Hungary
| |
Collapse
|
34
|
van der Reest J, Nardini Cecchino G, Haigis MC, Kordowitzki P. Mitochondria: Their relevance during oocyte ageing. Ageing Res Rev 2021; 70:101378. [PMID: 34091076 DOI: 10.1016/j.arr.2021.101378] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022]
Abstract
The oocyte is recognised as the largest cell in mammalian species and other multicellular organisms. Mitochondria represent a high proportion of the cytoplasm in oocytes and mitochondrial architecture is different in oocytes than in somatic cells, characterised by a rounder appearance and fragmented network. Although the number of mitochondria per oocyte is higher than in any other mammalian cell, their number and activity decrease with advancing age. Mitochondria integrate numerous processes essential for cellular function, such as metabolic processes related to energy production, biosynthesis, and waste removal, as well as Ca2+ signalling and reactive oxygen species (ROS) homeostasis. Further, mitochondria are responsible for the cellular adaptation to different types of stressors such as oxidative stress or DNA damage. When these stressors outstrip the adaptive capacity of mitochondria to restore homeostasis, it leads to mitochondrial dysfunction. Decades of studies indicate that mitochondrial function is multifaceted, which is reflected in the oocyte, where mitochondria support numerous processes during oocyte maturation, fertilization, and early embryonic development. Dysregulation of mitochondrial processes has been consistently reported in ageing and age-related diseases. In this review, we describe the functions of mitochondria as bioenergetic powerhouses and signal transducers in oocytes, how dysfunction of mitochondrial processes contributes to reproductive ageing, and whether mitochondria could be targeted to promote oocyte rejuvenation.
Collapse
|
35
|
Benammar A, Derisoud E, Vialard F, Palmer E, Ayoubi JM, Poulain M, Chavatte-Palmer P. The Mare: A Pertinent Model for Human Assisted Reproductive Technologies? Animals (Basel) 2021; 11:2304. [PMID: 34438761 PMCID: PMC8388489 DOI: 10.3390/ani11082304] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/28/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022] Open
Abstract
Although there are large differences between horses and humans for reproductive anatomy, follicular dynamics, mono-ovulation, and embryo development kinetics until the blastocyst stage are similar. In contrast to humans, however, horses are seasonal animals and do not have a menstrual cycle. Moreover, horse implantation takes place 30 days later than in humans. In terms of artificial reproduction techniques (ART), oocytes are generally matured in vitro in horses because ovarian stimulation remains inefficient. This allows the collection of oocytes without hormonal treatments. In humans, in vivo matured oocytes are collected after ovarian stimulation. Subsequently, only intra-cytoplasmic sperm injection (ICSI) is performed in horses to produce embryos, whereas both in vitro fertilization and ICSI are applied in humans. Embryos are transferred only as blastocysts in horses. In contrast, four cells to blastocyst stage embryos are transferred in humans. Embryo and oocyte cryopreservation has been mastered in humans, but not completely in horses. Finally, both species share infertility concerns due to ageing and obesity. Thus, reciprocal knowledge could be gained through the comparative study of ART and infertility treatments both in woman and mare, even though the horse could not be used as a single model for human ART.
Collapse
Affiliation(s)
- Achraf Benammar
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
- Department of Gynaecology and Obstetrics, Foch Hospital, 92150 Suresnes, France
| | - Emilie Derisoud
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - François Vialard
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Eric Palmer
- Académie d’Agriculture de France, 75007 Paris, France;
| | - Jean Marc Ayoubi
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
- Department of Gynaecology and Obstetrics, Foch Hospital, 92150 Suresnes, France
| | - Marine Poulain
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
- Department of Gynaecology and Obstetrics, Foch Hospital, 92150 Suresnes, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| |
Collapse
|
36
|
Munisha M, Schimenti JC. Genome maintenance during embryogenesis. DNA Repair (Amst) 2021; 106:103195. [PMID: 34358805 DOI: 10.1016/j.dnarep.2021.103195] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/25/2022]
Abstract
Genome maintenance during embryogenesis is critical, because defects during this period can be perpetuated and thus have a long-term impact on individual's health and longevity. Nevertheless, genome instability is normal during certain aspects of embryonic development, indicating that there is a balance between the exigencies of timely cell proliferation and mutation prevention. In particular, early embryos possess unique cellular and molecular features that underscore the challenge of having an appropriate balance. Here, we discuss genome instability during embryonic development, the mechanisms used in various cell compartments to manage genomic stress and address outstanding questions regarding the balance between genome maintenance mechanisms in key cell types that are important for adulthood and progeny.
Collapse
Affiliation(s)
- Mumingjiang Munisha
- Dept. of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, United States
| | - John C Schimenti
- Dept. of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, United States.
| |
Collapse
|
37
|
Chen J, Cheng Y, Fu W, Peng X, Sun X, Chen H, Chen X, Yu M. PPOS Protocol Effectively Improves the IVF Outcome Without Increasing the Recurrence Rate in Early Endometrioid Endometrial Cancer and Atypical Endometrial Hyperplasia Patients After Fertility Preserving Treatment. Front Med (Lausanne) 2021; 8:581927. [PMID: 34386503 PMCID: PMC8354380 DOI: 10.3389/fmed.2021.581927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 06/28/2021] [Indexed: 01/21/2023] Open
Abstract
Objective: To investigate the effectiveness and recurrence risk of different ovulation stimulation protocols in early-stage endometrioid endometrial cancer (EEC) and atypical endometrial hyperplasia (AEH) patients after successful fertility preserving treatment. Design: A retrospective review of clinical files between June 2012 and July 2018. Setting: University hospital. Patients: Ninety seven women (74 AEH and 23 early-stage EEC patients) underwent in vitro fertilization (IVF) and frozen-thawed embryo transfer (FET) after successful fertility preserving treatment. All patients received megestrol acetate which was initiated immediately after AEH or EEC diagnosis by hysteroscopy. Fertility treatment was initiated after confirmation of complete response by two consecutive hysteroscopic evaluations and endometrium biopsy in a 3-month interval. Women with tubal factors underwent IVF treatment directly. Women who failed to conceive spontaneously within 12 months or after other infertility treatments like ovulation induction for 6 consecutive months or 2 consecutive artificial insemination failures were also offered IVF treatment. Main Outcome Measure (s): The clinical and laboratory embryo data, clinical pregnancy outcomes and endometrial disease recurrence rates. Results: Compared with the standard regimen group, the good-quality embryo rate was higher in progestin primed ovarian stimulation (PPOS) regimen group (P = 0.034). Univariate analysis showed significant differences in age (P = 0.033), treatment time of endometrial lesions (P < 0.001), and duration of Gn treatment (P = 0.018) between the recurrent and non-recurrent groups. In the adjusted model of multivariate logistic regression analysis, the age (P = 0.014) at ovulation induction and treatment time of endometrial lesions (P < 0.001) were significantly correlated with the recurrence of endometrial disease. Conclusions: The PPOS protocol is a feasible and safe strategy to stimulate ovulation during IVF after fertility preservation therapy, and the age at ovulation induction and treatment time of endometrial lesions are two stable predictors of recurrence in endometrial diseases.
Collapse
Affiliation(s)
- Jiazhou Chen
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yali Cheng
- Shanghai Key Laboratory of Female Reproductive and Endocrine-Related Diseases, Shanghai, China
| | - Wei Fu
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiandong Peng
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaoxi Sun
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive and Endocrine-Related Diseases, Shanghai, China
| | - Hua Chen
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaojun Chen
- Shanghai Key Laboratory of Female Reproductive and Endocrine-Related Diseases, Shanghai, China.,Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Min Yu
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
38
|
Yang Q, Mumusoglu S, Qin Y, Sun Y, Hsueh AJ. A kaleidoscopic view of ovarian genes associated with premature ovarian insufficiency and senescence. FASEB J 2021; 35:e21753. [PMID: 34233068 DOI: 10.1096/fj.202100756r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022]
Abstract
Ovarian infertility and subfertility presenting with premature ovarian insufficiency (POI) and diminished ovarian reserve are major issues facing the developed world due to the trend of delaying childbirth. Ovarian senescence and POI represent a continuum of physiological/pathophysiological changes in ovarian follicle functions. Based on advances in whole exome sequencing, evaluation of gene copy variants, together with family-based and genome-wide association studies, we discussed genes responsible for POI and ovarian senescence. We used a gene-centric approach to sort out literature deposited in the Ovarian Kaleidoscope database (http://okdb.appliedbioinfo.net) by sub-categorizing candidate genes as ligand-receptor signaling, meiosis and DNA repair, transcriptional factors, RNA metabolism, enzymes, and others. We discussed individual gene mutations found in POI patients and verification of gene functions in gene-deleted model organisms. Decreased expression of some of the POI genes could be responsible for ovarian senescence, especially those essential for DNA repair, meiosis and mitochondrial functions. We propose to set up a candidate gene panel for targeted sequencing in POI patients together with studies on mitochondria-associated genes in middle-aged subfertile patients.
Collapse
Affiliation(s)
- Qingling Yang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sezcan Mumusoglu
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Obstetrics and Gynecology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yingpu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Aaron J Hsueh
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
39
|
Li J, Hu T, Wang Y, Fu Y, Wang F, Hu R. Development a nomogram to predict fertilisation rate of infertile males with borderline semen by using semen parameters combined with AMH and INHB. Andrologia 2021; 53:e14182. [PMID: 34270116 PMCID: PMC8519038 DOI: 10.1111/and.14182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/10/2021] [Accepted: 06/19/2021] [Indexed: 11/30/2022] Open
Abstract
The sperm quality of some males is in a critical state, making it hard for clinicians to choose the suitable fertilisation methods. This study aimed to develop an intelligent nomogram for predicting fertilisation rate of infertile males with borderline semen. 160 males underwent in vitro fertilisation (IVF), 58 of whom received rescue ICSI (R‐ICSI) due to fertilisation failure (fertilisation rate of IVF ≤30%). A least absolute shrinkage and selection operator (LASSO) regression analysis identified sperm concentration, progressively motile spermatozoa (PMS), seminal plasma anti‐Müllerian hormone (spAMH), seminal plasma inhibin (spINHB), serum AMH (serAMH) and serum INHB (serINHB) as significant predictors. The nomogram was plotted by multivariable logistic regression. This nomogram‐illustrated model showed good discrimination, calibration and clinical value. The area under the receiver operating characteristic curve (AUC) of the nomogram was 0.762 (p < .001). Calibration curve and Hosmer–Lemeshow test (p = .5261) showed good consistency between the predictions of the nomogram and the actual observations, and decision curve analysis showed that the nomogram was clinically useful. This nomogram may be useful in predicting fertilisation rate, mainly focused on new biomarkers, INHB and AMH. It could assist clinicians and laboratory technicians select appropriate fertilisation methods (IVF or ICSI) for male patients with borderline semen.
Collapse
Affiliation(s)
- Jialing Li
- Ningxia Medical University, Yinchuan, China.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Ting Hu
- Gansu Provincial Maternity and Child-care Hospital, Lanzhou, Gansu, China
| | - Yanfei Wang
- Gansu Provincial Maternity and Child-care Hospital, Lanzhou, Gansu, China
| | - Yuxing Fu
- Ningxia Medical University, Yinchuan, China.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Feimiao Wang
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Rong Hu
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
40
|
Oocyte ability to repair sperm DNA fragmentation: the impact of maternal age on intracytoplasmic sperm injection outcomes. Fertil Steril 2021; 116:123-129. [DOI: 10.1016/j.fertnstert.2020.10.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022]
|
41
|
Yuan L, Yin P, Yan H, Zhong X, Ren C, Li K, Chin Heng B, Zhang W, Tong G. Single-cell transcriptome analysis of human oocyte ageing. J Cell Mol Med 2021; 25:6289-6303. [PMID: 34037315 PMCID: PMC8256362 DOI: 10.1111/jcmm.16594] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/18/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Oocyte ageing is a key bottleneck and intractable challenge for in vitro fertilization treatment of aged female patients. The underlying molecular mechanisms of human oocyte ageing remain to be elucidated. Hence, this study aims to investigate the key genes and relevant biological signalling pathways involved in human oocyte ageing. We isolated mRNA for single-cell RNA sequencing from MII human oocytes donated by patients undergoing intracytoplasmic sperm injection. Nine RNA-seq datasets were analyzed, which included 6 older patients(average 42.67±2.25 years) and 3 younger patients (average 25.67±2.08 years). 481 differentially expressed genes (DEGs) were identified, including 322 upregulated genes enriched in transcription, ubiquitination, epigenetic regulation, and cellular processes, and 159 downregulated genes enriched in ubiquitination, cell cycle, signalling pathway, and DNA repair. The STRING database was used to analyse protein-protein interactions, and the Cytoscape software was used to identify hub genes. From these DEGs, 17 hub genes were identified including 12 upregulated genes (UBE2C, UBC, CDC34, UBR1, KIF11, ASF1B, PRC1, ESPL1, GTSE1, EXO1, UBA1, KIF4A) and 5 downregulated genes (UBA52, UBE2V2, SKP1, CCNB1, MAD2L1). The significant key biological processes that are associated with these hub genes include ubiquitin-mediated proteolysis, ubiquitination-related pathways, oocyte meiosis, and cell cycle. Among these, UBE2C may play a crucial role in human oocyte ageing.
Collapse
Affiliation(s)
- Lihua Yuan
- Shuguang Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Ping Yin
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hua Yan
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiufang Zhong
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chunxia Ren
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Kai Li
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | | | - Wuwen Zhang
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Guoqing Tong
- Center of Reproductive MedicineShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
42
|
Advanced maternal age perturbs mouse embryo development and alters the phenotype of derived embryonic stem cells. J Dev Orig Health Dis 2021; 13:395-405. [PMID: 34193331 DOI: 10.1017/s2040174421000325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Advanced maternal age (AMA) is known to reduce fertility, increases aneuploidy in oocytes and early embryos and leads to adverse developmental consequences which may associate with offspring lifetime health risks. However, investigating underlying effects of AMA on embryo developmental potential is confounded by the inherent senescence present in maternal body systems further affecting reproductive success. Here, we describe a new model for the analysis of early developmental mechanisms underlying AMA by the derivation and characterisation of mouse embryonic stem cell (mESC-like) lines from naturally conceived embryos. Young (7-8 weeks) and Old (7-8 months) C57BL/6 female mice were mated with young males. Preimplantation embryos from Old dams displayed developmental retardation in blastocyst morphogenesis. mESC lines established from these blastocysts using conventional techniques revealed differences in genetic, cellular and molecular criteria conserved over several passages in the standardised medium. mESCs from embryos from AMA dams displayed increased incidence of aneuploidy following Giemsa karyotyping compared with those from Young dams. Moreover, AMA caused an altered pattern of expression of pluripotency markers (Sox2, OCT4) in mESCs. AMA further diminished mESC survival and proliferation and reduced the expression of cell proliferation marker, Ki-67. These changes coincided with altered expression of the epigenetic marker, Dnmt3a and other developmental regulators in a sex-dependent manner. Collectively, our data demonstrate the feasibility to utilise mESCs to reveal developmental mechanisms underlying AMA in the absence of maternal senescence and with reduced animal use.
Collapse
|
43
|
Zhou Z, Yang X, Pan Y, Shang L, Chen S, Yang J, Jin L, Zhang F, Wu Y. Temporal transcriptomic landscape of postnatal mouse ovaries reveals dynamic gene signatures associated with ovarian aging. Hum Mol Genet 2021; 30:1941-1954. [PMID: 34137841 PMCID: PMC8522635 DOI: 10.1093/hmg/ddab163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 11/17/2022] Open
Abstract
The ovary is the most important organ for maintaining female reproductive health, but it fails before most other organs. Aging-associated alterations in gene expression patterns in mammalian ovaries remain largely unknown. In this study, the transcriptomic landscape of postnatal mouse ovaries over the reproductive lifespan was investigated using bulk RNA sequencing in C57BL/6 mice. Gene expression dynamics revealed that the lifespan of postnatal mouse ovaries comprised four sequential stages, during which 2517 genes were identified as differentially enriched. Notably, the DNA repair pathway was found to make a considerable and specific contribution to the process of ovarian aging. Temporal gene expression patterns were dissected to identify differences in gene expression trajectories over the lifespan. In addition to DNA repair, distinct biological functions (including hypoxia response, epigenetic modification, fertilization, mitochondrial function, etc.) were overrepresented in particular clusters. Association studies were further performed to explore the relationships between known genes responsible for ovarian function and differentially expressed genes identified in this work. We found that the causative genes of human premature ovarian insufficiency were specifically enriched in distinct gene clusters. Taken together, our findings reveal a comprehensive transcriptomic landscape of the mouse ovary over the lifespan, providing insights into the molecular mechanisms underlying mammalian ovarian aging and supporting future etiological studies of aging-associated ovarian disorders.
Collapse
Affiliation(s)
- Zixue Zhou
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China
| | - Xi Yang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China.,Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Yuncheng Pan
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China
| | - Lingyue Shang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China
| | - Siyuan Chen
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China
| | - Jialin Yang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China
| | - Li Jin
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China.,Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Yanhua Wu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), State Key Laboratory of Genetic Engineering at School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China.,National Demonstration Center for Experimental Biology Education, School of Life Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
44
|
Li CJ, Lin LT, Tsai HW, Chern CU, Wen ZH, Wang PH, Tsui KH. The Molecular Regulation in the Pathophysiology in Ovarian Aging. Aging Dis 2021; 12:934-949. [PMID: 34094652 PMCID: PMC8139203 DOI: 10.14336/ad.2020.1113] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/13/2020] [Indexed: 12/23/2022] Open
Abstract
The female reproductive system is of great significance to women’s health. Aging of the female reproductive system occurs approximately 10 years prior to the natural age-associated functional decline of other organ systems. With an increase in life expectancy worldwide, reproductive aging has gradually become a key health issue among women. Therefore, an adequate understanding of the causes and molecular mechanisms of ovarian aging is essential towards the inhibition of age-related diseases and the promotion of health and longevity in women. In general, women begin to experience a decline in ovarian function around the age of 35 years, which is mainly manifested as a decrease in the number of ovarian follicles and the quality of oocytes. Studies have revealed the occurrence of mitochondrial dysfunction, reduced DNA repair, epigenetic changes, and metabolic alterations in the cells within the ovaries as age increases. In the present work, we reviewed the possible factors of aging-induced ovarian insufficiency based on its clinical diagnosis and performed an in-depth investigation of the relevant molecular mechanisms and potential targets to provide novel approaches for the effective improvement of ovarian function in older women.
Collapse
Affiliation(s)
- Chia-Jung Li
- 1Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,2Institute of BioPharmaceutical sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Li-Te Lin
- 1Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,2Institute of BioPharmaceutical sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.,3Department of Obstetrics and Gynaecology, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Hsiao-Wen Tsai
- 1Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,2Institute of BioPharmaceutical sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chyi-Uei Chern
- 1Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- 4Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Peng-Hui Wang
- 3Department of Obstetrics and Gynaecology, National Yang-Ming University School of Medicine, Taipei, Taiwan.,5Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,6Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,7Female Cancer Foundation, Taipei, Taiwan
| | - Kuan-Hao Tsui
- 1Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,2Institute of BioPharmaceutical sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.,3Department of Obstetrics and Gynaecology, National Yang-Ming University School of Medicine, Taipei, Taiwan.,8Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, Taiwan
| |
Collapse
|
45
|
Llonch S, Barragán M, Nieto P, Mallol A, Elosua‐Bayes M, Lorden P, Ruiz S, Zambelli F, Heyn H, Vassena R, Payer B. Single human oocyte transcriptome analysis reveals distinct maturation stage-dependent pathways impacted by age. Aging Cell 2021; 20:e13360. [PMID: 33908703 PMCID: PMC8135014 DOI: 10.1111/acel.13360] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/09/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Female fertility is inversely correlated with maternal age due to a depletion of the oocyte pool and a reduction in oocyte developmental competence. Few studies have addressed the effect of maternal age on the human mature oocyte (MII) transcriptome, which is established during oocyte growth and maturation, however, the pathways involved remain unclear. Here, we characterize and compare the transcriptomes of a large cohort of fully grown germinal vesicle stage (GV) and in vitro matured (IVM‐MII) oocytes from women of varying reproductive age. First, we identified two clusters of cells reflecting the oocyte maturation stage (GV and IVM‐MII) with 4445 and 324 putative marker genes, respectively. Furthermore, we identified genes for which transcript representation either progressively increased or decreased with age. Our results indicate that the transcriptome is more affected by age in IVM‐MII oocytes (1219 genes) than in GV oocytes (596 genes). In particular, we found that transcripts of genes involved in chromosome segregation and RNA splicing significantly increased representation with age, while genes related to mitochondrial activity showed a lower representation. Gene regulatory network analysis facilitated the identification of potential upstream master regulators of the genes involved in those biological functions. Our analysis suggests that advanced maternal age does not globally affect the oocyte transcriptome at GV or IVM‐MII stages. Nonetheless, hundreds of genes displayed altered transcript representation, particularly in IVM‐MII oocytes, which might contribute to the age‐related quality decline in human oocytes.
Collapse
Affiliation(s)
- Sílvia Llonch
- Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
| | | | - Paula Nieto
- CNAG‐CRG Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
| | - Anna Mallol
- Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
| | - Marc Elosua‐Bayes
- CNAG‐CRG Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
| | - Patricia Lorden
- CNAG‐CRG Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
| | - Sara Ruiz
- CNAG‐CRG Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
| | | | - Holger Heyn
- CNAG‐CRG Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
| | | | - Bernhard Payer
- Centre for Genomic Regulation (CRG) The Barcelona Institute of Science and Technology Barcelona Spain
- Universitat Pompeu Fabra (UPF) Barcelona Spain
| |
Collapse
|
46
|
Horta F, Catt S, Ramachandran P, Vollenhoven B, Temple-Smith P. Female ageing affects the DNA repair capacity of oocytes in IVF using a controlled model of sperm DNA damage in mice. Hum Reprod 2021; 35:529-544. [PMID: 32108237 DOI: 10.1093/humrep/dez308] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/17/2019] [Indexed: 01/07/2023] Open
Abstract
STUDY QUESTION Does female ageing have a negative effect on the DNA repair capacity of oocytes fertilised by spermatozoa with controlled levels of DNA damage? SUMMARY ANSWER Compared to oocytes from younger females, oocytes from older females have a reduced capacity to repair damaged DNA introduced by spermatozoa. WHAT IS KNOWN ALREADY The reproductive lifespan in women declines with age predominantly due to poor oocyte quality. This leads to decreased reproductive outcomes for older women undergoing assisted reproductive technology (ART) treatments, compared to young women. Ageing and oocyte quality have been clearly associated with aneuploidy, but the range of factors that influence this change in oocyte quality with age remains unclear. The DNA repair activity prior to embryonic genomic activation is considered to be of maternal origin, with maternal transcripts and proteins controlling DNA integrity. With increasing maternal age, the number of mRNAs stored in oocytes decreases. This could result in diminished efficiency of DNA repair and/or negative effects on embryo development, especially in the presence of DNA damage. STUDY DESIGN, SIZE, DURATION Oocytes from two age groups of 30 super-ovulated female mice (young: 5-8 weeks old, n = 15; old: 42-45 weeks old, n = 15) were inseminated with sperm from five males with three different controlled DNA damage levels; control: ≤10%, 1 Gray (Gy): 11-30%, and 30 Gy: >30%. Inseminated oocytes (young: 125, old: 78) were assessed for the formation of zygotes (per oocyte) and blastocysts (per zygote). Five replicates of five germinal vesicles (GVs) and five MII oocytes from each age group were analysed for gene expression. The DNA damage response (DDR) was assessed in a minimum of three IVF replicates in control and 1 Gy zygotes and two-cell embryos using γH2AX labelling. PARTICIPANTS/MATERIALS, SETTING, METHODS Swim-up sperm samples from the cauda epididymidis of C57BL6 mice were divided into control (no irradiation) and 1- and 30-Gy groups. Treated spermatozoa were irradiated at 1 and 30 Gy, respectively, using a linear accelerator Varian 21iX. Following irradiation, samples were used for DNA damage assessment (Halomax) and for insemination. Presumed zygotes were cultured in a time-lapse incubator (MIRI, ESCO). Gene expression of 91 DNA repair genes was assessed using the Fluidigm Biomark HD system. The DNA damage response in zygotes (6-8 h post-fertilisation) and two-cell embryos (22-24 h post-fertilisation) was assessed by immunocytochemical analysis of γH2AX using confocal microscopy (Olympus FV1200) and 3D volumetric analysis using IMARIS software. MAIN RESULTS AND THE ROLE OF CHANCE The average sperm DNA damage for the three groups was statistically different (control: 6.1%, 1 Gy: 16.1%, 30 Gy: 53.1%, P < 0.0001), but there were no significant differences in fertilisation rates after IVF within or between the two age groups [(young; control: 86.79%, 1 Gy: 82.75%, 30 Gy: 76.74%) (old; control: 93.1%, 1 Gy: 70.37%, 30 Gy: 68.18%) Fisher's exact]. However, blastocyst rates were significantly different (P < 0.0001) among the groups [(young; control: 86.95%, 1 Gy: 33.33%, 30 Gy: 0.0%) (old; control: 70.37%, 1 Gy: 0.0%, 30 Gy: 0.0%)]. Between the age groups, 1-Gy samples showed a significant decrease in the blastocyst rate in old females compared to young females (P = 0.0166). Gene expression analysis revealed a decrease in relative expression of 21 DNA repair genes in old GV oocytes compared to young GV oocytes (P < 0.05), and similarly, old MII oocytes showed 23 genes with reduced expression compared to young MII oocytes (P < 0.05). The number of genes with decreased expression in older GV and MII oocytes significantly affected pathways such as double strand break (GV: 5; MII: 6), nucleotide excision repair (GV: 8; MII: 5) and DNA damage response (GV: 4; MII: 8). There was a decreased DDR in zygotes and in two-cell embryos from old females compared to young regardless of sperm treatment (P < 0.05). The decrease in DNA repair gene expression of oocytes and decreased DDR in embryos derived from older females suggests that ageing results in a diminished DNA repair capacity. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Ionising radiation was used only for experimental purposes, aiming at controlled levels of sperm DNA damage; however, it can also damage spermatozoa proteins. The female age groups selected in mice were intended to model effects in young and old women, but clinical studies are required to demonstrate a similar effect. WIDER IMPLICATIONS OF THE FINDINGS Fertilisation can occur with sperm populations with medium and high DNA damage, but subsequent embryo growth is affected to a greater extent with aging females, supporting the theory that oocyte DNA repair capacity decreases with age. Assessment of the oocyte DNA repair capacity may be a useful diagnostic tool for infertile couples. STUDY FUNDING/COMPETING INTEREST(S) Funded by the Education Program in Reproduction and Development, Department of Obstetrics and Gynaecology, Monash University. None of the authors has any conflict of interest to report.
Collapse
Affiliation(s)
- F Horta
- Education Program in Reproduction & Development, Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia
| | - S Catt
- Education Program in Reproduction & Development, Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia
| | - P Ramachandran
- Peter MacCallum Cancer Centre, Monash Health, Melbourne, VIC 3164, Australia
| | - B Vollenhoven
- Monash IVF, Melbourne, VIC 3168, Australia.,Women's and Newborn Program, Monash Health, VIC 3169, Australia.,Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia
| | - P Temple-Smith
- Education Program in Reproduction & Development, Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia
| |
Collapse
|
47
|
Cafe SL, Nixon B, Ecroyd H, Martin JH, Skerrett-Byrne DA, Bromfield EG. Proteostasis in the Male and Female Germline: A New Outlook on the Maintenance of Reproductive Health. Front Cell Dev Biol 2021; 9:660626. [PMID: 33937261 PMCID: PMC8085359 DOI: 10.3389/fcell.2021.660626] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 01/07/2023] Open
Abstract
For fully differentiated, long lived cells the maintenance of protein homeostasis (proteostasis) becomes a crucial determinant of cellular function and viability. Neurons are the most well-known example of this phenomenon where the majority of these cells must survive the entire course of life. However, male and female germ cells are also uniquely dependent on the maintenance of proteostasis to achieve successful fertilization. Oocytes, also long-lived cells, are subjected to prolonged periods of arrest and are largely reliant on the translation of stored mRNAs, accumulated during the growth period, to support meiotic maturation and subsequent embryogenesis. Conversely, sperm cells, while relatively ephemeral, are completely reliant on proteostasis due to the absence of both transcription and translation. Despite these remarkable, cell-specific features there has been little focus on understanding protein homeostasis in reproductive cells and how/whether proteostasis is "reset" during embryogenesis. Here, we seek to capture the momentum of this growing field by highlighting novel findings regarding germline proteostasis and how this knowledge can be used to promote reproductive health. In this review we capture proteostasis in the context of both somatic cell and germline aging and discuss the influence of oxidative stress on protein function. In particular, we highlight the contributions of proteostasis changes to oocyte aging and encourage a focus in this area that may complement the extensive analyses of DNA damage and aneuploidy that have long occupied the oocyte aging field. Moreover, we discuss the influence of common non-enzymatic protein modifications on the stability of proteins in the male germline, how these changes affect sperm function, and how they may be prevented to preserve fertility. Through this review we aim to bring to light a new trajectory for our field and highlight the potential to harness the germ cell's natural proteostasis mechanisms to improve reproductive health. This manuscript will be of interest to those in the fields of proteostasis, aging, male and female gamete reproductive biology, embryogenesis, and life course health.
Collapse
Affiliation(s)
- Shenae L. Cafe
- Priority Research Centre for Reproductive Science, Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Heath Ecroyd
- Molecular Horizons, School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Jacinta H. Martin
- Department of Human Genetics, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - David A. Skerrett-Byrne
- Priority Research Centre for Reproductive Science, Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Elizabeth G. Bromfield
- Priority Research Centre for Reproductive Science, Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
48
|
Wartosch L, Schindler K, Schuh M, Gruhn JR, Hoffmann ER, McCoy RC, Xing J. Origins and mechanisms leading to aneuploidy in human eggs. Prenat Diagn 2021; 41:620-630. [PMID: 33860956 PMCID: PMC8237340 DOI: 10.1002/pd.5927] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/02/2021] [Accepted: 02/21/2021] [Indexed: 11/18/2022]
Abstract
The gain or loss of a chromosome-or aneuploidy-acts as one of the major triggers for infertility and pregnancy loss in humans. These chromosomal abnormalities affect more than 40% of eggs in women at both ends of the age spectrum, that is, young girls as well as women of advancing maternal age. Recent studies in human oocytes and embryos using genomics, cytogenetics, and in silico modeling all provide new insight into the rates and potential genetic and cellular factors associated with aneuploidy at varying stages of development. Here, we review recent studies that are shedding light on potential molecular mechanisms of chromosome missegregation in oocytes and embryos across the entire female reproductive life span.
Collapse
Affiliation(s)
- Lena Wartosch
- Department of MeiosisMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Karen Schindler
- Department of GeneticsRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
- Human Genetics Institute of New JerseyRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| | - Melina Schuh
- Department of MeiosisMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Jennifer R. Gruhn
- DNRF Center for Chromosome StabilityDepartment of Cellular and Molecular MedicineFaculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Eva R. Hoffmann
- DNRF Center for Chromosome StabilityDepartment of Cellular and Molecular MedicineFaculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Rajiv C. McCoy
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jinchuan Xing
- Department of GeneticsRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
- Human Genetics Institute of New JerseyRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| |
Collapse
|
49
|
Deng Z, Wang W, Xu X, Nie Y, Liu Y, Gould OEC, Ma N, Lendlein A. Biofunction of Polydopamine Coating in Stem Cell Culture. ACS APPLIED MATERIALS & INTERFACES 2021; 13:10748-10759. [PMID: 33594879 DOI: 10.1021/acsami.0c22565] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
High levels of reactive oxygen species (ROS) during stem cell expansion often lead to replicative senescence. Here, a polydopamine (PDA)-coated substrate was used to scavenge extracellular ROS for mesenchymal stem cell (MSC) expansion. The PDA-coated substrate could reduce the oxidative stress and mitochondrial damage in replicative senescent MSCs. The expression of senescence-associated β-galactosidase of MSCs from three human donors (both bone marrow- and adipose tissue-derived) was suppressed on PDA. The MSCs on the PDA-coated substrate showed a lower level of interleukin 6 (IL-6), one of the senescence-associated inflammatory components. Cellular senescence-specific genes, such as p53 and p21, were downregulated on the PDA-coated substrate, while the stemness-related gene, OCT4, was upregulated. The PDA-coated substrate strongly promoted the proliferation rate of MSCs, while the stem cell character and differentiation potential were retained. Large-scale expansion of stem cells would greatly benefit from the PDA-coated substrate.
Collapse
Affiliation(s)
- Zijun Deng
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, 14195 Berlin, Germany
| | - Weiwei Wang
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
| | - Xun Xu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
| | - Yan Nie
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Yue Liu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Oliver E C Gould
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
| | - Nan Ma
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, 14195 Berlin, Germany
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, 14195 Berlin, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| |
Collapse
|
50
|
Gebremedhn S, Ali A, Hossain M, Hoelker M, Salilew-Wondim D, Anthony RV, Tesfaye D. MicroRNA-Mediated Gene Regulatory Mechanisms in Mammalian Female Reproductive Health. Int J Mol Sci 2021; 22:938. [PMID: 33477832 PMCID: PMC7832875 DOI: 10.3390/ijms22020938] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/12/2022] Open
Abstract
Mammalian reproductive health affects the entire reproductive cycle starting with the ovarian function through implantation and fetal growth. Various environmental and physiological factors contribute to disturbed reproductive health status leading to infertility problems in mammalian species. In the last couple of decades a significant number of studies have been conducted to investigate the transcriptome of reproductive tissues and organs in relation to the various reproductive health issues including endometritis, polycystic ovarian syndrome (PCOS), intrauterine growth restriction (IUGR), preeclampsia, and various age-associated reproductive disorders. Among others, the post-transcriptional regulation of genes by small noncoding miRNAs contributes to the observed transcriptome dysregulation associated with reproductive pathophysiological conditions. MicroRNAs as a class of non-coding RNAs are also known to be involved in various pathophysiological conditions either in cellular cytoplasm or they can be released to the extracellular fluid via membrane-bounded extracellular vesicles and proteins. The present review summarizes the cellular and extracellular miRNAs and their association with the etiology of major reproductive pathologies including PCOS, endometritis, IUGR and age-associated disorders in various mammalian species.
Collapse
Affiliation(s)
- Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Rd, Fort Collins, CO 80523, USA; (S.G.); (A.A.); (R.V.A.)
| | - Asghar Ali
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Rd, Fort Collins, CO 80523, USA; (S.G.); (A.A.); (R.V.A.)
| | - Munir Hossain
- Department of Animal Breeding and Genetics, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh;
| | - Michael Hoelker
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, 53115 Bonn, Germany; (M.H.); (D.S.-W.)
| | - Dessie Salilew-Wondim
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, 53115 Bonn, Germany; (M.H.); (D.S.-W.)
| | - Russell V. Anthony
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Rd, Fort Collins, CO 80523, USA; (S.G.); (A.A.); (R.V.A.)
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Rd, Fort Collins, CO 80523, USA; (S.G.); (A.A.); (R.V.A.)
| |
Collapse
|