1
|
Zhou J, Dong Q. Testicular aging: mechanism, management and future therapy. Exp Cell Res 2025; 449:114603. [PMID: 40373850 DOI: 10.1016/j.yexcr.2025.114603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/03/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Testicular aging results in degeneration in testicular function, including decreased testosterone production and quality of sperm. Decreased testosterone level is associated with a range of systemic diseases and comorbidities, including cardiovascular disease, cognitive decline, depression, osteoporosis, frailty, increased body fat, and metabolic syndrome. In addition, with the rapid development of industrialization and increasing environmental pollution, the quality of male semen continues to decline globally. Currently, the average age of first marriage and childbirth for men is delayed, and the birth rate has been declining year by year. At present, the therapies for testosterone level decline in clinical practice are relatively limited. Therefore, studying the triggering and delaying mechanisms of testicular aging is significant for improving male health and protecting male fertility. This review summarizes the mechanisms and treatment strategies for male reproductive aging.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, 610000, Sichuan Province, China
| | - Qiang Dong
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, 610000, Sichuan Province, China.
| |
Collapse
|
2
|
Moazamian A, Saez F, Drevet JR, Aitken RJ, Gharagozloo P. Redox-Driven Epigenetic Modifications in Sperm: Unraveling Paternal Influences on Embryo Development and Transgenerational Health. Antioxidants (Basel) 2025; 14:570. [PMID: 40427452 PMCID: PMC12108309 DOI: 10.3390/antiox14050570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Male-factor infertility accounts for nearly half of all infertility cases, and mounting evidence points to oxidative stress as a pivotal driver of sperm dysfunction, genetic instability, and epigenetic dysregulation. In particular, the oxidative DNA lesion 8-hydroxy-2'-deoxyguanosine (8-OHdG) has emerged as a central mediator at the interface of DNA damage and epigenetic regulation. We discuss how this lesion can disrupt key epigenetic mechanisms such as DNA methylation, histone modifications, and small non-coding RNAs, thereby influencing fertilization outcomes, embryo development, and offspring health. We propose that the interplay between oxidative DNA damage and epigenetic reprogramming is further exacerbated by aging in both the paternal and maternal germlines, creating a "perfect storm" that increases the risk of heritable (epi)mutations. The consequences of unresolved oxidative lesions can thus persist beyond fertilization, contributing to transgenerational health risks. Finally, we explore the promise and potential pitfalls of antioxidant therapy as a strategy to mitigate sperm oxidative damage. While antioxidant supplementation may hold significant therapeutic value for men with subfertility experiencing elevated oxidative stress, a careful, personalized approach is essential to avoid reductive stress and unintended epigenetic disruptions. Recognizing the dual role of oxidative stress in shaping both the genome and the epigenome underscores the need for integrating redox biology into reproductive medicine, with the aim of improving fertility treatments and safeguarding the health of future generations.
Collapse
Affiliation(s)
- Aron Moazamian
- EVALSEM, GReD Institute, CRBC, Faculté de Médecine, Université Clermont Auvergne, 28 Place Henri Dunant, 6300 Clermont-Ferrand, France; (F.S.); (J.R.D.)
- CellOxess Biotechnology, Research & Development, Ewing, NJ 08638, USA
| | - Fabrice Saez
- EVALSEM, GReD Institute, CRBC, Faculté de Médecine, Université Clermont Auvergne, 28 Place Henri Dunant, 6300 Clermont-Ferrand, France; (F.S.); (J.R.D.)
| | - Joël R. Drevet
- EVALSEM, GReD Institute, CRBC, Faculté de Médecine, Université Clermont Auvergne, 28 Place Henri Dunant, 6300 Clermont-Ferrand, France; (F.S.); (J.R.D.)
| | - Robert John Aitken
- Priority Research Centre for Reproductive Science, University of Newcastle, Newcastle 2308, Australia;
| | | |
Collapse
|
3
|
Wan Y, Wen Z, Liu W, Jiang X, Wu L, Jiang H, Zhang X, Shi Q, Hua J. High risk of low birth weight in couples with advanced paternal age in in vitro fertilization treatment. Andrology 2025; 13:830-839. [PMID: 39195011 DOI: 10.1111/andr.13745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/22/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND With the development of socio-economic conditions and a shift in attitudes towards fertility, there has been a gradual increase in delayed childbearing since the 2000s. Age plays a significant role in the decline of fertility. However, we know very little about the association of paternal age with reproductive outcomes. OBJECTIVES To investigate the correlation between advanced paternal age and semen quality, embryo quality, pregnancy, and neonatal outcomes in IVF cycles. MATERIALS AND METHODS In this study, after excluding female partners aged ≥35 years, we analyzed data from 761 infertile couples who underwent in vitro fertilization cycles at the First Affiliated Hospital of USTC between June 2020 and March 2023. Cases were classified into three groups according to the age of the male: <35 years (530 infertile couples), 35 years ≤ paternal age <40 years (125 infertile couples), and ≥40 years (106 infertile couples). Then, we compared the general clinical data arising from in vitro fertilization cycles between the three groups, including semen parameters, embryonic parameters, and pregnancy and neonatal birth outcomes. RESULTS Data analysis showed that the duration of infertility and the incidence of secondary infertility were significantly higher in paternal age ≥35 years groups than those aged <35 years (all p < 0.05). We also observed a significant difference between ≥40 years and <35 years groups in terms of the normal fertilization rate, high-quality embryo rate, clinical pregnancy rate, miscarriage rate, live birth rate, Apgar scores, and the low birth weight neonatal rate (all p < 0.05). The group with paternal age ≥40 years showed statistically significant differences in terms of clinical pregnancy rate, miscarriage rate, live birth rate, and low birth weight on multivariable logistic regression (all p < 0.05). CONCLUSION The results of our study indicate that advanced paternal age (≥40 years) has a significant impact on the embryo quality, pregnancy outcome, and neonatal outcome. Paternal age over 40 years is a risk for in vitro fertilization success rate.
Collapse
Affiliation(s)
- Yangyang Wan
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Zhu Wen
- Department of Urology, The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenjing Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiaohua Jiang
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Limin Wu
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Hui Jiang
- Department of Urology, Peking University First Hospital Institute of Urology, Peking University Andrology Center, Beijing, China
| | - Xiansheng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qinghua Shi
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Juan Hua
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
4
|
Gupta N, Joseph T, Karuppusami R, Kunjummen AT, Kamath MS. Predictive factors for spontaneous conception in women with moderate and severe endometriosis following operative laparoscopy: A cohort study. Eur J Obstet Gynecol Reprod Biol 2025; 309:192-199. [PMID: 40168751 DOI: 10.1016/j.ejogrb.2025.03.052] [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: 02/21/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
STUDY OBJECTIVE To externally validate the Endometriosis Fertility Index (EFI) and to assess cumulative spontaneous pregnancy rates in women with moderate to severe endometriosis following fertility-enhancing minimally invasive surgery. DESIGN Retrospective cohort study. SETTING Tertiary-care center for infertility treatment. PARTICIPANTS Women diagnosed with r-ASRM Stage III or IV endometriosis following fertility-sparing surgery between June 2013 and July 2023, and who desired for fertility. INTERVENTION Minimally invasive laparoscopy. MEASUREMENTS AND MAIN RESULTS The follow-up rate was 76.3 %, with a median duration of 15 months [Range (1-130 months)]. The mean age of women was 28.5 ± 3.8 years. Women with low EFI scores (0-3) had lower cumulative spontaneous pregnancy rates (11.4 % at six months, 17 % at three years) compared to those with higher EFI scores (7-8), who had significantly higher rates (35 % at six months, 84.5 % after three years) (P < 0.001). A similar trend was observed for overall pregnancy rates. Factors negatively correlated with spontaneous pregnancy included lower least function scores (P < 0.001), increasing maternal age (P = 0.003), rASRM scores of 16-40 vs. ≥ 71 (P < 0.001), lower antral follicle count (P = 0.02), and increasing paternal age (P < 0.001). Cystectomy (P = 0.001) was positively associated with spontaneous pregnancy post-surgery. Among the 518 women identified with moderate and severe endometriosis, 395 were followed up, out of which 370 attempted spontaneous conception for at least six months, resulting in a pregnancy rate of 37.3 % (138/370). CONCLUSION The current study demonstrated that EFI scores correlate with pregnancy rates, validating the scoring system in a South Asian population. Additionally, clinical factors such as low antral follicle count, and advanced paternal age may be considered to make informed clinical decisions post-surgery for women with advanced endometriosis who wish to conceive.
Collapse
Affiliation(s)
- Nivedita Gupta
- Department of Reproductive Medicine and Surgery, Christian Medical College, Vellore, India
| | - Treasa Joseph
- Department of Reproductive Medicine and Surgery, Christian Medical College, Vellore, India
| | - Reka Karuppusami
- Department of Biostatistics, Christian Medical College, Vellore, India
| | - Aleyamma T Kunjummen
- Department of Reproductive Medicine and Surgery, Christian Medical College, Vellore, India
| | - Mohan S Kamath
- Department of Reproductive Medicine and Surgery, Christian Medical College, Vellore, India.
| |
Collapse
|
5
|
Aoki Y, Wakamatsu M, Sono N, Xiao W, Ishii E, Nagai T, Nagai Y, Fujiwara Y, Kunieda T, Otsuki J. Impact of aging on spermatogenic function and reproductive outcomes in repro57 heterozygous male mice: A model for age-related infertility. J Assist Reprod Genet 2025:10.1007/s10815-025-03481-x. [PMID: 40257706 DOI: 10.1007/s10815-025-03481-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 04/08/2025] [Indexed: 04/22/2025] Open
Abstract
PURPOSE This study aims to investigate the histological changes, sperm parameters, and their impact on embryo development rates and offspring numbers in advanced-age male repro57 heterozygous mice, corresponding to approximately 40 years of age in humans. METHODS Sperm parameters were assessed in both young and advanced-age repro57 heterozygous mice, as well as in young and advanced-age wild-type mice. Additionally, testis weight and histological analysis of seminiferous tubules were conducted to identify degenerative changes. Male mice from each group were mated with young wild-type females to compare offspring numbers, and in vitro fertilization (IVF) was used to evaluate fertilization and blastocyst formation rates. RESULTS No significant differences in sperm concentration and motility were observed between young and aged wild-type mice or between young wild-type and young repro57 heterozygous mice. However, advanced-age repro57 heterozygous mice exhibited significantly lower sperm parameters and testis weight compared to advanced-age wild-type mice. Histological analysis revealed increased Sertoli cell vacuolation in the seminiferous tubules of advanced-age repro57 heterozygous mice. Additionally, these advanced-age mice exhibited significantly lower blastocyst formation rates and produced fewer offspring compared to advanced-age wild-type mice. CONCLUSION Advanced reproductive aging in repro57 heterozygous male mice is associated with marked senescence-like degenerative changes, leading to a decline in offspring numbers, attributed to increased Sertoli cell vacuolation and diminished sperm quality.
Collapse
Affiliation(s)
- Yuto Aoki
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Misaki Wakamatsu
- Department of Animal Science, Faculty of Agriculture, Okayama University, 1 - 1- 1 Tsushimanaka, Tsushimanaka, KitaKita, OkayamaOkayama, 700 - 8530, Japan
| | - Nanami Sono
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Wei Xiao
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Emi Ishii
- Nagai Mother's Hospital, Kamihikona, Misato, Saitama, 341 - 0004, Japan
| | - Takeshi Nagai
- Nagai Mother's Hospital, Kamihikona, Misato, Saitama, 341 - 0004, Japan
| | - Yasushi Nagai
- Nagai Mother's Hospital, Kamihikona, Misato, Saitama, 341 - 0004, Japan
| | - Yasuhiro Fujiwara
- Laboratory of Pathology and Development, Institute for Quantitative Biosciences, The University of Tokyo, YayoiTokyo, Bunkyo, 113 - 0032, Japan
| | - Tetsuo Kunieda
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Junko Otsuki
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan.
- Assisted Reproductive Technology Center, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan.
| |
Collapse
|
6
|
Cui L, Nie X, Guo Y, Ren P, Guo Y, Wang X, Li R, Hotaling JM, Cairns BR, Guo J. Single-cell transcriptomic atlas of the human testis across the reproductive lifespan. NATURE AGING 2025; 5:658-674. [PMID: 40033047 PMCID: PMC12003174 DOI: 10.1038/s43587-025-00824-2] [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] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025]
Abstract
Testicular aging is associated with declining reproductive health, but the molecular mechanisms are unclear. Here we generate a dataset of 214,369 single-cell transcriptomes from testicular cells of 35 individuals aged 21-69, offering a resource for studying testicular aging and physiology. Machine learning analysis reveals a stronger aging response in somatic cells compared to germ cells. Two waves of aging-related changes are identified: the first in peritubular cells of donors in their 30s, marked by increased basement membrane thickness, indicating a priming state for aging. In their 50s, testicular cells exhibit functional changes, including altered steroid metabolism in Leydig cells and immune responses in macrophages. Further analyses reveal the impact of body mass index on spermatogenic capacity as age progresses, particularly after age 45. Altogether, our findings illuminate molecular alterations during testis aging and their relationship with body mass index, providing a foundation for future research and offering potential diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Lina Cui
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xichen Nie
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
- Division of Urology, Department of Surgery, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Yixuan Guo
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Pengcheng Ren
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yifei Guo
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Ran Li
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - James M Hotaling
- Division of Urology, Department of Surgery, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Bradley R Cairns
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA.
| | - Jingtao Guo
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of the Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
7
|
Guitart-Solanes A, Romero M, Fernandez-Duran I, Niedenberger BA, Madrid-Sandín C, Roig I, Geyer CB, Vaquero A, Schindler K, Vazquez BN. SIRT7 links H3K36ac epigenetic regulation with genome maintenance in the aging mouse testis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.645534. [PMID: 40236082 PMCID: PMC11996300 DOI: 10.1101/2025.03.31.645534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Reproductive aging is an increasing health concern affecting family planning and overall well-being. While extensively studied in females, the mechanisms driving male reproductive aging remain largely unexamined. Here we found that mammalian Sirtuin 7 (SIRT7) sustains spermatogenesis in an age-dependent manner through the control of histone 3 lysine 36 acetylation (H3K36ac). SIRT7 deficiency in mice resulted in increased levels of H3K36ac in spermatogonia and spermatocytes. In a germ cell line, SIRT7 deficiency disrupted nucleosome stability and increased vulnerability to genotoxic stress. Importantly, undifferentiated spermatogonia, which are required for continuous sperm production, decreased prematurely in Sirt7 -/- mice and showed genome damage accumulation. These changes were concurrent with age-dependent defects in homologous chromosome synapsis and partial meiotic arrest. Taken together, our results indicate that SIRT7 connects H3K36ac epigenetic regulation to long-term genome stability in male germ cells, ensuring steady-state spermatogenesis during the lengthy male reproductive lifespan.
Collapse
|
8
|
Kaltsas A, Markou E, Kyrgiafini MA, Zikopoulos A, Symeonidis EN, Dimitriadis F, Zachariou A, Sofikitis N, Chrisofos M. Oxidative-Stress-Mediated Epigenetic Dysregulation in Spermatogenesis: Implications for Male Infertility and Offspring Health. Genes (Basel) 2025; 16:93. [PMID: 39858640 PMCID: PMC11765119 DOI: 10.3390/genes16010093] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Male reproductive health is governed by an intricate interplay of genetic, epigenetic, and environmental factors. Epigenetic mechanisms-encompassing DNA methylation, histone modifications, and non-coding RNA activity-are crucial both for spermatogenesis and sperm maturation. However, oxidative stress, driven by excessive reactive oxygen species, disrupts these processes, leading to impaired sperm function and male infertility. This disruption extends to epigenetic modifications, resulting in abnormal gene expression and chromatin remodeling that compromise genomic integrity and fertilization potential. Importantly, oxidative-stress-induced epigenetic alterations can be inherited, affecting the health and fertility of offspring and future generations. This review investigates how oxidative stress influences epigenetic regulation in male reproduction by modifying DNA methylation, histone modifications, and non-coding RNAs, ultimately compromising spermatogenesis. Additionally, it discusses the transgenerational implications of these epigenetic disruptions and their potential role in hereditary infertility and disease predisposition. Understanding these mechanisms is vital for developing therapeutic strategies that mitigate oxidative damage and restore epigenetic homeostasis in the male germline. By integrating insights from molecular, clinical, and transgenerational research, this work emphasizes the need for targeted interventions to enhance male reproductive health and prevent adverse outcomes in progeny. Furthermore, elucidating the dose-response relationships between oxidative stress and epigenetic changes remains a critical research priority, informing personalized diagnostics and therapeutic interventions. In this context, future studies should adopt standardized markers of oxidative damage, robust clinical trials, and multi-omic approaches to capture the complexity of epigenetic regulation in spermatogenesis. Such rigorous investigations will ultimately reduce the risk of transgenerational disorders and optimize reproductive health outcomes.
Collapse
Affiliation(s)
- Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Eleftheria Markou
- Department of Microbiology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Maria-Anna Kyrgiafini
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece;
| | - Athanasios Zikopoulos
- Obstetrics and Gynecology, Royal Devon and Exeter Hospital, Barrack Rd, Exeter EX2 5DW, UK;
| | | | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Athanasios Zachariou
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Nikolaos Sofikitis
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| |
Collapse
|
9
|
Li W, Yu Z, Xu S, Li Z, Xia W. Extracellular Vesicles in the Aging Male Reproductive System: Progresses and Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:375-394. [PMID: 40301265 DOI: 10.1007/978-3-031-82990-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Extracellular vesicles (EVs) serve as crucial mediators of intercellular communication in spermatogenesis, steroidogenesis, and age-related pathophysiological processes within the male reproductive system. These EVs exhibit promising prospects for disease diagnosis and therapeutic administration. This review explores the impact of advanced paternal age on male fertility and testosterone decline, shedding light on the underlying mechanisms. It highlights the decline in semen quality, DNA damage, and alterations in sperm miRNA profiles associated with aging. The interplay between oxidative stress and antioxidants crucially regulates male reproductive aging. Currently, most studies focus on Sertoli cell-derived EVs, while understanding of Leydig cell-derived vesicles remains limited. Multi-omics integration will enhance the understanding of male reproductive aging and guide personalized interventions, revealing potential biomarkers and targets in the future.
Collapse
Affiliation(s)
- Wenbo Li
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Depart. of Andrology, Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Lab of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Depart. of ART, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziwen Yu
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Shuai Xu
- Depart. of Andrology, Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Lab of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Depart. of ART, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Li
- Depart. of Andrology, Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Lab of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Depart. of ART, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Bengbu Hospital of Shanghai General Hospital (The Second Affiliated Hospital of Bengbu Medical University), Bengbu, Anhui, China.
| | - Weiliang Xia
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
10
|
Aitken RJ. Paternal age, de novo mutations, and offspring health? New directions for an ageing problem. Hum Reprod 2024; 39:2645-2654. [PMID: 39361588 PMCID: PMC11630042 DOI: 10.1093/humrep/deae230] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/07/2024] [Indexed: 10/05/2024] Open
Abstract
This Directions article examines the mechanisms by which a father's age impacts the health and wellbeing of his children. Such impacts are significant and include adverse birth outcomes, dominant genetic conditions, neuropsychiatric disorders, and a variety of congenital developmental defects. As well as age, a wide variety of environmental and lifestyle factors are also known to impact offspring health via changes mediated by the male germ line. This picture of a dynamic germ line responsive to a wide range of intrinsic and extrinsic factors contrasts with the results of trio studies indicating that the incidence of mutations in the male germ line is low and exhibits a linear, monotonic increase with paternal age (∼two new mutations per year). While the traditional explanation for this pattern of mutation has been the metronomic plod of replication errors, an alternative model pivots around the 'faulty male' hypothesis. According to this concept, the genetic integrity of the male germ line can be dynamically impacted by age and a variety of other factors, and it is the aberrant repair of such damage that drives mutagenesis. Fortunately, DNA proofreading during spermatogenesis is extremely effective and these mutant cells are either repaired or deleted by apoptosis/ferroptosis. There appear to be only two mechanisms by which mutant germ cells can escape this apoptotic fate: (i) if the germ cells acquire a mutation that by enhancing proliferation or suppressing apoptosis, permits their clonal expansion (selfish selection hypothesis) or (ii) if a genetically damaged spermatozoon manages to fertilize an oocyte, which then fixes the damage as a mutation (or epimutation) as a result of defective DNA repair (oocyte collusion hypothesis). Exploration of these proposed mechanisms should not only help us better understand the aetiology of paternal age effects but also inform potential avenues of remediation.
Collapse
Affiliation(s)
- Robert John Aitken
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, School of Environmental and Life Sciences, College of Engineering Science and Environment, University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
11
|
Wang S, Ren J, Jing Y, Qu J, Liu GH. Perspectives on biomarkers of reproductive aging for fertility and beyond. NATURE AGING 2024; 4:1697-1710. [PMID: 39672897 DOI: 10.1038/s43587-024-00770-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/29/2024] [Indexed: 12/15/2024]
Abstract
Reproductive aging, spanning an age-related functional decline in the female and male reproductive systems, compromises fertility and leads to a range of health complications. In this Perspective, we first introduce a comprehensive framework for biomarkers applicable in clinical settings and discuss the existing repertoire of biomarkers used in practice. These encompass functional, imaging-based and biofluid-based biomarkers, all of which reflect the physiological characteristics of reproductive aging and help to determine the reproductive biological age. Next, we delve into the molecular alterations associated with aging in the reproductive system, highlighting the gap between these changes and their potential as biomarkers. Finally, to enhance the precision and practicality of assessing reproductive aging, we suggest adopting cutting-edge technologies for identifying new biomarkers and conducting thorough validations in population studies before clinical applications. These advancements will foster improved comprehension, prognosis and treatment of subfertility, thereby increasing chances of preserving reproductive health and resilience in populations of advanced age.
Collapse
Affiliation(s)
- Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Jie Ren
- Aging Biomarker Consortium, Beijing, China
- Key Laboratory of RNA Science and Engineering, China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying Jing
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Qu
- Aging Biomarker Consortium, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, CAS, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, CAS, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| |
Collapse
|
12
|
Benonisdottir S, Straub VJ, Kong A, Mills MC. Genetics of female and male reproductive traits and their relationship with health, longevity and consequences for offspring. NATURE AGING 2024; 4:1745-1759. [PMID: 39672892 DOI: 10.1038/s43587-024-00733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 12/15/2024]
Abstract
Substantial shifts in reproductive behaviors have recently taken place in many high-income countries including earlier age at menarche, advanced age at childbearing, rising childlessness and a lower number of children. As reproduction shifts to later ages, genetic factors may become increasingly important. Although monogenic genetic effects are known, the genetics underlying human reproductive traits are complex, with both causal effects and statistical bias often confounded by socioeconomic factors. Here, we review genome-wide association studies (GWASs) of 44 reproductive traits of both female and male individuals from 2007 to early 2024, examining reproductive behavior, reproductive lifespan and aging, infertility and hormonal concentration. Using the GWAS Catalog as a basis, from 159 relevant studies, we isolate 37 genes that harbor association signals for four or more reproductive traits, more than half of which are linked to rare Mendelian disorders, including ten genes linked to reproductive-related disorders: FSHB, MCM8, DNAH2, WNT4, ESR1, IGSF1, THRB, BRWD1, CYP19A1 and PTPRF. We also review the relationship of reproductive genetics to related health and behavioral traits, aging and longevity and the effect of parental age on offspring outcomes as well as reflecting on limitations, open questions and challenges in this fast-moving field.
Collapse
Affiliation(s)
- Stefania Benonisdottir
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
- Institute of Physical Science, University of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Vincent J Straub
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Augustine Kong
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Melinda C Mills
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK.
- Department of Genetics, University Medical Centre Groningen, Groningen, the Netherlands.
- Department of Economics, Econometrics and Finance, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
13
|
Xia K, Luo P, Yu J, He S, Dong L, Gao F, Chen X, Ye Y, Gao Y, Ma Y, Yang C, Zhang Y, Yang Q, Han D, Feng X, Wan Z, Cai H, Ke Q, Wang T, Li W, Tu X, Sun X, Deng C, Xiang AP. Single-cell RNA sequencing reveals transcriptomic landscape and potential targets for human testicular ageing. Hum Reprod 2024; 39:2189-2209. [PMID: 39241251 PMCID: PMC11447013 DOI: 10.1093/humrep/deae199] [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: 05/24/2023] [Revised: 07/05/2024] [Indexed: 09/08/2024] Open
Abstract
STUDY QUESTION What is the molecular landscape underlying the functional decline of human testicular ageing? SUMMARY ANSWER The present study provides a comprehensive single-cell transcriptomic atlas of testes from young and old humans and offers insights into the molecular mechanisms and potential targets for human testicular ageing. WHAT IS KNOWN ALREADY Testicular ageing is known to cause male age-related fertility decline and hypogonadism. Dysfunction of testicular cells has been considered as a key factor for testicular ageing. STUDY DESIGN, SIZE, DURATION Human testicular biopsies were collected from three young individuals and three old individuals to perform single-cell RNA sequencing (scRNA-seq). The key results were validated in a larger cohort containing human testicular samples from 10 young donors and 10 old donors. PARTICIPANTS/MATERIALS, SETTING, METHODS scRNA-seq was used to identify gene expression signatures for human testicular cells during ageing. Ageing-associated changes of gene expression in spermatogonial stem cells (SSCs) and Leydig cells (LCs) were analysed by gene set enrichment analysis and validated by immunofluorescent and functional assays. Cell-cell communication analysis was performed using CellChat. MAIN RESULTS AND THE ROLE OF CHANCE The single-cell transcriptomic landscape of testes from young and old men was surveyed, revealing age-related changes in germline and somatic niche cells. In-depth evaluation of the gene expression dynamics in germ cells revealed that the disruption of the base-excision repair pathway is a prominent characteristic of old SSCs, suggesting that defective DNA repair in SSCs may serve as a potential driver for increased de novo germline mutations with age. Further analysis of ageing-associated transcriptional changes demonstrated that stress-related changes and cytokine pathways accumulate in old somatic cells. Age-related impairment of redox homeostasis in old LCs was identified and pharmacological treatment with antioxidants alleviated this cellular dysfunction of LCs and promoted testosterone production. Lastly, our results revealed that decreased pleiotrophin signalling was a contributing factor for impaired spermatogenesis in testicular ageing. LARGE SCALE DATA The scRNA-seq sequencing and processed data reported in this paper were deposited at the Genome Sequence Archive (https://ngdc.cncb.ac.cn/), under the accession number HRA002349. LIMITATIONS, REASONS FOR CAUTION Owing to the difficulty in collecting human testis tissue, the sample size was limited. Further in-depth functional and mechanistic studies are warranted in future. WIDER IMPLICATIONS OF THE FINDINGS These findings provide a comprehensive understanding of the cell type-specific mechanisms underlying human testicular ageing at a single-cell resolution, and suggest potential therapeutic targets that may be leveraged to address age-related male fertility decline and hypogonadism. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Key Research and Development Program of China (2022YFA1104100), the National Natural Science Foundation of China (32130046, 82171564, 82101669, 82371611, 82371609, 82301796), the Natural Science Foundation of Guangdong Province, China (2022A1515010371), the Major Project of Medical Science and Technology Development Research Center of National Health Planning Commission, China (HDSL202001000), the Open Project of NHC Key Laboratory of Male Reproduction and Genetics (KF202001), the Guangdong Province Regional Joint Fund-Youth Fund Project (2021A1515110921, 2022A1515111201), and the China Postdoctoral Science Foundation (2021M703736). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Kai Xia
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Peng Luo
- Reproductive Medicine Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Jiajie Yu
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Siyuan He
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Lin Dong
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Feng Gao
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Reproductive Medicine Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xuren Chen
- Reproductive Medicine Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yunlin Ye
- Department of Urology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Yong Gao
- Reproductive Medicine Centre, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Yuanchen Ma
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Cuifeng Yang
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yadong Zhang
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiyun Yang
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dayu Han
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xin Feng
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zi Wan
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongcai Cai
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiong Ke
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiang'an Tu
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiangzhou Sun
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chunhua Deng
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
14
|
Frungieri MB, Calandra RS, Matzkin ME, Rossi SP. Melatonin as a natural anti-inflammatory and anti-oxidant therapy in the testis: a focus on infertility and aging†. Biol Reprod 2024; 111:543-556. [PMID: 38869910 DOI: 10.1093/biolre/ioae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Melatonin is a pineal hormone that regulates testicular activity (i.e., steroidogenesis and spermatogenesis) through two complementary mechanisms, indirect effects exerted via the hypothalamic-adenohypophyseal axis and direct actions that take place on the different cell populations of the male gonad. The effects of increased age on the testis and the general mechanisms involved in testicular pathology leading to infertility are still only poorly understood. However, there is growing evidence that link testicular aging and idiopathic male infertility to local inflammatory and oxidative stress events. Because literature data strongly indicate that melatonin exhibits anti-inflammatory and anti-oxidant properties, this review focuses on the potential benefits exerted by this indoleamine at testicular level in male reproductive fertility and aging. Taking into account that the effects of melatonin supplementation on testicular function are currently being investigated, the overview covers not only promising prospects but also many questions concerning the future therapeutic value of this indoleamine as an anti-aging drug as well as in the management of cases of male infertility for which there are no medical treatments currently available.
Collapse
Affiliation(s)
- Mónica Beatriz Frungieri
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
| | - Ricardo Saúl Calandra
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
| | - María Eugenia Matzkin
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
- Cátedra 1, Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Soledad Paola Rossi
- Laboratorio de Neuro-Inmuno-Endocrinología Testicular, Instituto de Biología y Medicina Experimental, Fundación Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires, Argentina
- Cátedra 1, Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
15
|
Al-Salihy AARS. Longitudinal trends and correlation between autism spectrum disorder prevalence and sperm quality parameters (2000-2024): a comprehensive statistical analysis. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 6:1438049. [PMID: 39239154 PMCID: PMC11374721 DOI: 10.3389/frph.2024.1438049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/31/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction Over the past few decades, there has been growing concern about the concurrent trends of increasing Autism Spectrum Disorder (ASD) prevalence and declining sperm quality. These trends represent significant public health challenges that warrant thorough investigation of their underlying causes and implications. Objectives The primary objectives of this study are to analyze trends in ASD prevalence and sperm quality parameters from 2000 to 2024, assess the statistical significance and effect size of these trends, explore potential correlations between ASD prevalence and sperm quality parameters, and identify significant predictors among sperm quality parameters that influence ASD prevalence. Methods This study employed a longitudinal approach using multiple regression, time series analysis, ANOVA, Principal Component Analysis (PCA), hierarchical clustering, logistic regression, and cross-correlation analysis. Data on ASD prevalence were sourced from the CDC Autism and Developmental Disabilities Monitoring Network, while sperm quality data were collected from various published studies. Results The findings reveal significant negative associations between ASD prevalence and sperm quality parameters such as sperm concentration and motility, suggesting that better sperm quality is linked to lower ASD rates. Conversely, parameters like sperm DNA fragmentation (SDF), volume of ejaculate, pH level, and semen viscosity show positive associations with ASD prevalence, indicating higher values in these parameters correlate with higher ASD rates. Conclusion The study highlights the importance of maintaining reproductive health to potentially mitigate ASD risk and calls for further research to elucidate the underlying mechanisms driving these trends. These findings support the hypothesis that reproductive health factors play a crucial role in ASD etiology and suggest potential biological markers for assessing ASD risk.
Collapse
|
16
|
Ajayi AF, Oyovwi MO, Olatinwo G, Phillips AO. Unfolding the complexity of epigenetics in male reproductive aging: a review of therapeutic implications. Mol Biol Rep 2024; 51:881. [PMID: 39085654 DOI: 10.1007/s11033-024-09823-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Epigenetics studies gene expression changes influenced by environmental and lifestyle factors, linked to health conditions like reproductive aging. Male reproductive aging causes sperm decline, conceiving difficulties, and increased genetic abnormalities. Recent research focuses on epigenetics' role in male reproductive aging. OBJECTIVES This review explores epigenetics and male reproductive aging, focusing on sperm quality, environmental and lifestyle factors' impact, and potential health implications for offspring. METHODS An extensive search of the literature was performed applying multiple databases, such as PubMed and Google Scholar. The search phrases employed were: epigenetics, male reproductive ageing, sperm quality, sperm quantity, environmental influences, lifestyle factors, and offspring health. This review only included articles that were published in English and had undergone a peer-review process. The literature evaluation uncovered that epigenetic alterations have a substantial influence on the process of male reproductive ageing. RESULT Research has demonstrated that variations in the quality and quantity of sperm that occur with ageing are linked to adjustments in DNA methylation and histone. Moreover, there is evidence linking epigenetic alterations in sperm to environmental and lifestyle factors, including smoking, alcohol intake, and exposure to contaminants. These alterations can have enduring impacts on the well-being of descendants, since they can shape the activation of genes and potentially elevate the likelihood of genetic disorders. In conclusion, epigenetics significantly influences male reproductive aging, with sperm quality and quantity influenced by environmental and lifestyle factors. CONCLUSION This underscores the need for comprehensive approaches to managing male reproductive health, and underscores the importance of considering epigenetics in diagnosis and treatment.
Collapse
Affiliation(s)
- Ayodeji Folorunsho Ajayi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Anchor Biomed Research Institute, Ogbomoso, Oyo State, Nigeria
- Department of Physiology, Adeleke University, Ede, Osun State, Nigeria
| | | | - Goodness Olatinwo
- Department of Physiology, School of Basic Medical Sciences, Babcock University, Ilishan Remo, Ogun State, Nigeria
| | - Akano Oyedayo Phillips
- Department of Physiology, School of Basic Medical Sciences, Babcock University, Ilishan Remo, Ogun State, Nigeria
| |
Collapse
|
17
|
Castellini C, Cordeschi G, Tienforti D, Barbonetti A. Relationship between male aging and semen quality: a retrospective study on over 2500 men. Arch Gynecol Obstet 2024; 309:2843-2852. [PMID: 38551703 PMCID: PMC11147833 DOI: 10.1007/s00404-024-07448-8] [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: 11/16/2023] [Accepted: 02/25/2024] [Indexed: 06/04/2024]
Abstract
PURPOSE We aimed to evaluate whether and to what extent an association exists between male aging and worsening of semen parameters and to determine whether a threshold age can be identified above which the decline in semen quality becomes statistically significant. METHODS 2612 men (age: 16-56 years) attending an andrology outpatient clinic for semen analysis and clinical evaluation were studied. Semen analyses were performed according to the ongoing WHO-recommended procedures. Total motile count (TMC) and total progressive motile count (TPMC) were calculated by multiplying total sperm number by total motility and progressive motility, respectively. RESULTS Significant negative correlations were found between age and total motility (r = - 0.131, p < 0.0001), progressive motility (r = - 0.112, p < 0.0001), TPMC (r = - 0.042, p = 0.037), and normal sperm morphology (r = - 0.053, p = 0.007). All these associations persisted in multivariate regression models adjusted for abstinence time, smoking, history of male accessory gland infections, varicocele and the year in which semen analysis was performed. When comparisons were performed among quartiles of increasing age, the fourth quartile, corresponding to the age group > 40 years, was associated with a significant decrease in total and progressive motility. An earlier decline in the TPMC and percentage of normal forms was also observed. CONCLUSION Advancing male age exhibits an independent association with a decrease in the percentage of motile and morphologically normal spermatozoa, with greater evidence from the age of > 40 years. Further studies are warranted to elucidate the mechanisms and clinical reflections of these associations.
Collapse
Affiliation(s)
- Chiara Castellini
- Andrology Unit, Department of Clinical Medicine, Life, Health and Environmental Sciences, University of L'Aquila, Coppito, 67100, L'Aquila, Italy
| | - Giuliana Cordeschi
- Andrology Unit, Department of Clinical Medicine, Life, Health and Environmental Sciences, University of L'Aquila, Coppito, 67100, L'Aquila, Italy
| | - Daniele Tienforti
- Andrology Unit, Department of Clinical Medicine, Life, Health and Environmental Sciences, University of L'Aquila, Coppito, 67100, L'Aquila, Italy
| | - Arcangelo Barbonetti
- Andrology Unit, Department of Clinical Medicine, Life, Health and Environmental Sciences, University of L'Aquila, Coppito, 67100, L'Aquila, Italy.
| |
Collapse
|
18
|
Feuz MB, Nelson DC, Miller LB, Zwerdling AE, Meyer RG, Meyer-Ficca ML. Reproductive Ageing: Current insights and a potential role of NAD in the reproductive health of aging fathers and their children. Reproduction 2024; 167:e230486. [PMID: 38471307 PMCID: PMC11075800 DOI: 10.1530/rep-23-0486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/12/2024] [Indexed: 03/14/2024]
Abstract
In brief In light of the increasing age of first-time fathers, this article summarizes the current scientific knowledge base on reproductive aging in the male, including sperm quality and health impacts for the offspring. The emerging role of NAD decline in reproductive aging is highlighted. Abstract Over the past decades, the age of first-time fathers has been steadily increasing due to socio-economic pressures. While general mechanisms of aging are subject to intensive research, male reproductive aging has remained an understudied area, and the effects of increased age on the male reproductive system are still only poorly understood, despite new insights into the potential dire consequences of advanced paternal age for the health of their progeny. There is also growing evidence that reproductive aging is linked to overall health in men, but this review mainly focuses on pathophysiological consequences of old age in men, such as low sperm count and diminished sperm genetic integrity, with an emphasis on mechanisms underlying reproductive aging. The steady decline of NAD levels observed in aging men represents one of the emerging concepts in that regard. Because it offers some mechanistic rationale explaining the effects of old age on the male reproductive system, some of the NAD-dependent functions in male reproduction are briefly outlined in this review. The overview also provides many questions that remain open about the basic science of male reproductive aging.
Collapse
Affiliation(s)
- Morgan B. Feuz
- Department of Veterinary, Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT, United States
- These authors contributed equally
| | - D. Colton Nelson
- Department of Veterinary, Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT, United States
- These authors contributed equally
| | - Laura B. Miller
- Department of Veterinary, Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT, United States
- These authors contributed equally
| | - Alexie E Zwerdling
- Department of Veterinary, Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT, United States
- These authors contributed equally
| | - Ralph G. Meyer
- Department of Veterinary, Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Mirella L. Meyer-Ficca
- Department of Veterinary, Clinical and Life Sciences, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| |
Collapse
|
19
|
Kanoh T, Mizoguchi T, Tonoki A, Itoh M. Modeling of age-related neurological disease: utility of zebrafish. Front Aging Neurosci 2024; 16:1399098. [PMID: 38765773 PMCID: PMC11099255 DOI: 10.3389/fnagi.2024.1399098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
Many age-related neurological diseases still lack effective treatments, making their understanding a critical and urgent issue in the globally aging society. To overcome this challenge, an animal model that accurately mimics these diseases is essential. To date, many mouse models have been developed to induce age-related neurological diseases through genetic manipulation or drug administration. These models help in understanding disease mechanisms and finding potential therapeutic targets. However, some age-related neurological diseases cannot be fully replicated in human pathology due to the different aspects between humans and mice. Although zebrafish has recently come into focus as a promising model for studying aging, there are few genetic zebrafish models of the age-related neurological disease. This review compares the aging phenotypes of humans, mice, and zebrafish, and provides an overview of age-related neurological diseases that can be mimicked in mouse models and those that cannot. We presented the possibility that reproducing human cerebral small vessel diseases during aging might be difficult in mice, and zebrafish has potential to be another animal model of such diseases due to their similarity of aging phenotype to humans.
Collapse
Affiliation(s)
- Tohgo Kanoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Takamasa Mizoguchi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ayako Tonoki
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Motoyuki Itoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Health and Disease Omics Center, Chiba University, Chiba, Japan
| |
Collapse
|
20
|
Aitken RJ. What is driving the global decline of human fertility? Need for a multidisciplinary approach to the underlying mechanisms. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 6:1364352. [PMID: 38726051 PMCID: PMC11079147 DOI: 10.3389/frph.2024.1364352] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
An intense period of human population expansion over the past 250 years is about to cease. Total fertility rates are falling dramatically all over the world such that highly industrialized nations, including China and the tiger economies of SE Asia, will see their populations decline significantly in the coming decades. The socioeconomic, geopolitical and environmental ramifications of this change are considerable and invite a multidisciplinary consideration of the underlying mechanisms. In the short-term, socioeconomic factors, particularly urbanization and delayed childbearing are powerful drivers of reduced fertility. In parallel, lifestyle factors such as obesity and the presence of numerous reproductive toxicants in the environment, including air-borne pollutants, nanoplastics and electromagnetic radiation, are seriously compromising reproductive health. In the longer term, it is hypothesized that the reduction in family size that accompanies the demographic transition will decrease selection pressure on high fertility genes leading to a progressive loss of human fecundity. Paradoxically, the uptake of assisted reproductive technologies at scale, may also contribute to such fecundity loss by encouraging the retention of poor fertility genotypes within the population. Since the decline in fertility rate that accompanies the demographic transition appears to be ubiquitous, the public health implications for our species are potentially devastating.
Collapse
Affiliation(s)
- Robert John Aitken
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, School of Environmental and Life Sciences, College of Engineering Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
21
|
Liao M, Xu Q, Mao X, Zhang J, Wu L, Chen Q. Paternal age does not jeopardize the live birth rate and perinatal outcomes after in vitro fertilization: an analysis based on 56,113 frozen embryo transfer cycles. Am J Obstet Gynecol 2024; 230:354.e1-354.e13. [PMID: 37952870 DOI: 10.1016/j.ajog.2023.11.1224] [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: 06/20/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND The global trend of delaying childbearing has led to an increasing number of couples seeking in vitro fertilization. The adverse effects of advanced maternal age on pregnancy and perinatal outcomes are well documented, regardless of the conception method. In addition, advanced paternal age may contribute to poor reproductive potential because of high levels of sperm DNA fragmentation. However, it remains challenging to guide older men regarding the effect of paternal age on pregnancy and birth outcomes in the field of assisted reproduction. OBJECTIVE This study aimed to investigate the association of paternal age with live birth and perinatal outcomes following in vitro fertilization-frozen embryo transfer. STUDY DESIGN A retrospective study was performed at a university-affiliated fertility center, involving women who were younger than 36 years and had undergone frozen embryo transfer from January 2011 to June 2021. Subjects were categorized into 6 groups based on paternal age: <25, 25 to 29, 30 to 34, 35 to 39, 40 to 44, and ≥45 years. A generalized estimating equation logistic regression model was used to account for the clustered nature of data and to adjust for confounders. Paternal age between 25 and 29 years served as the reference group in the logistic regression models. RESULTS A total of 56,113 cycles who met the inclusion criteria were included in the final analysis. On unadjusted analyses, the reproductive outcome parameters showed a considerable decline with increasing male age. The live birth rate decreased from 47.9% for men aged 25 to 29 years to 40.3% among men aged ≥40 years. Similarly, the clinical pregnancy rate decreased from 54.4% in the reference group to 47.8% in the ≥40 years age group. Conversely, the miscarriage rate increased as male age increased, from 10.2% among men aged 25 to 29 years to 13.5% among men aged ≥45 years. However, the differences in the reproductive outcomes mentioned above were no longer significant in the multivariable models. Compared with the younger controls, advanced paternal age was not associated with a lower chance of live birth (males aged 40-44 years: adjusted odds ratio, 0.94; 95% confidence interval, 0.85-1.04; males aged ≥45 years: adjusted odds ratio, 0.93; 95% confidence interval, 0.79-1.10). In addition, the rates of clinical pregnancy (males aged 40-44 years: adjusted odds ratio, 0.95; 95% confidence interval, 0.85-1.05; males aged ≥45 years: adjusted odds ratio, 0.94; 95% confidence interval, 0.79-1.12) and miscarriage (males aged 40-44 years: adjusted odds ratio, 1.05; 95% confidence interval, 0.85-1.31; males aged ≥45 years: adjusted odds ratio, 1.07; 95% confidence interval, 0.77-1.50) were comparable between the reference and advanced paternal age groups. Furthermore, men in the youngest age group (<25 years) did not have worse pregnancy outcomes than those in the reference group. Regarding perinatal outcomes, there was no difference among the study cohorts in terms of preterm birth, low birthweight, macrosomia, small for gestational age, and large for gestational age, both in the unadjusted and confounder-adjusted models. CONCLUSION This study did not demonstrate a significant association between paternal age and live birth and perinatal outcomes after in vitro fertilization-frozen embryo transfer when the female partners were younger than 36 years. With the global trend toward delaying childbirth, our findings provide useful information for counseling patients that increasing paternal age may not adversely affect pregnancy and perinatal outcomes in assisted reproduction.
Collapse
Affiliation(s)
- Maokun Liao
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyu Xu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Mao
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qiuju Chen
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Sengupta P, Dutta S, Liew FF, Dhawan V, Das B, Mottola F, Slama P, Rocco L, Roychoudhury S. Environmental and Genetic Traffic in the Journey from Sperm to Offspring. Biomolecules 2023; 13:1759. [PMID: 38136630 PMCID: PMC10741607 DOI: 10.3390/biom13121759] [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: 09/12/2023] [Revised: 11/04/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Recent advancements in the understanding of how sperm develop into offspring have shown complex interactions between environmental influences and genetic factors. The past decade, marked by a research surge, has not only highlighted the profound impact of paternal contributions on fertility and reproductive outcomes but also revolutionized our comprehension by unveiling how parental factors sculpt traits in successive generations through mechanisms that extend beyond traditional inheritance patterns. Studies have shown that offspring are more susceptible to environmental factors, especially during critical phases of growth. While these factors are broadly detrimental to health, their effects are especially acute during these periods. Moving beyond the immutable nature of the genome, the epigenetic profile of cells emerges as a dynamic architecture. This flexibility renders it susceptible to environmental disruptions. The primary objective of this review is to shed light on the diverse processes through which environmental agents affect male reproductive capacity. Additionally, it explores the consequences of paternal environmental interactions, demonstrating how interactions can reverberate in the offspring. It encompasses direct genetic changes as well as a broad spectrum of epigenetic adaptations. By consolidating current empirically supported research, it offers an exhaustive perspective on the interwoven trajectories of the environment, genetics, and epigenetics in the elaborate transition from sperm to offspring.
Collapse
Affiliation(s)
- Pallav Sengupta
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Sulagna Dutta
- School of Life Sciences, Manipal Academy of Higher Education (MAHE), Dubai 345050, United Arab Emirates
| | - Fong Fong Liew
- Department of Preclinical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom 42610, Selangor, Malaysia
| | - Vidhu Dhawan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Biprojit Das
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011, India
| | - Filomena Mottola
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, 613 00 Brno, Czech Republic
| | - Lucia Rocco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | | |
Collapse
|
23
|
Lahimer M, Gherissi O, Ben Salem N, Ben Mustapha H, Bach V, Khorsi-Cauet H, Khairi H, Ben Ali H, BenKhalifa M, Ajina M. Effect of Micronutrients and L-Carnitine as Antioxidant on Sperm Parameters, Genome Integrity, and ICSI Outcomes: Randomized, Double-Blind, and Placebo-Controlled Clinical Trial. Antioxidants (Basel) 2023; 12:1937. [PMID: 38001791 PMCID: PMC10669279 DOI: 10.3390/antiox12111937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
The evaluation of sperm DNA integrity is recommended in the sixth edition of the 2021 World Health Organization guidelines. Oxidative stress has been identified as a crucial factor leading to genome decay, lipid peroxidation, and nucleoprotein oxidation. This double-blind, placebo-controlled clinical trial aimed to assess the effect of oral antioxidant treatment (Fertilis), which contains L-carnitine and some micronutrients, in the improvement of conventional sperm parameters, sperm DNA integrity and in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) outcomes. A total of 263 participants were enrolled and randomly divided into two groups: 131 participants received the antioxidant treatment, while 132 participants received a placebo. The male partners in both groups underwent the antioxidant treatment or the placebo for a duration of three months. For each participant, we performed a hormonal test, an infectious test, a spermogram, a TUNEL assay for sperm DNA fragmentation, a toluidine blue staining for sperm DNA decondensation, and an IVF/ICSI procedure. Sperm characteristics analysis (volume, count, motility, and vitality), sperm DNA fragmentation, and sperm DNA decondensation were assessed and compared to the results preceding the antioxidant treatment. The study outcome revealed a significant decrease in the DNA fragmentation index and a significant increase in sperm motility after 3 months of treatment (p = 0.01 and p = 0.02, respectively). Additionally, a significant improvement in clinical pregnancy rate (p = 0.01) and life birth rate (p = 0.031) was observed. No significant changes were observed in conventional sperm parameters (volume, count, and vitality) or sperm DNA decondensation (SDI). Antioxidant therapy has a beneficial impact on achieving pregnancy, whether through spontaneous conception or assisted reproductive procedures (ART).
Collapse
Affiliation(s)
- Marwa Lahimer
- Service of Reproductive Biology, University Hospital Farhat Hached, Sousse, University of Sousse, Sousse 4000, Tunisia; (O.G.); (N.B.S.); (H.B.M.)
- Exercise Physiology and Physiopathology: From Integrated to Molecular “Biology, Medicine and Health” (Code: LR19ES09), Sousse 4002, Tunisia
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, 80025 Amiens, France; (V.B.); (H.K.-C.); (M.B.)
| | - Oumaima Gherissi
- Service of Reproductive Biology, University Hospital Farhat Hached, Sousse, University of Sousse, Sousse 4000, Tunisia; (O.G.); (N.B.S.); (H.B.M.)
| | - Nesrine Ben Salem
- Service of Reproductive Biology, University Hospital Farhat Hached, Sousse, University of Sousse, Sousse 4000, Tunisia; (O.G.); (N.B.S.); (H.B.M.)
| | - Henda Ben Mustapha
- Service of Reproductive Biology, University Hospital Farhat Hached, Sousse, University of Sousse, Sousse 4000, Tunisia; (O.G.); (N.B.S.); (H.B.M.)
| | - Véronique Bach
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, 80025 Amiens, France; (V.B.); (H.K.-C.); (M.B.)
| | - Hafida Khorsi-Cauet
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, 80025 Amiens, France; (V.B.); (H.K.-C.); (M.B.)
- ART and Reproductive Biology Laboratory, University Hospital and School of Medicine, Picardie University Jules Verne, CHU Sud, 80025 Amiens, France
| | - Hedi Khairi
- Faculty of Medicine Ibn Eljazzar of Sousse, Department of Obstetrics & Gynecology Sousse University, Sousse 4000, Tunisia;
| | - Habib Ben Ali
- Laboratory Histology Embryologiy, Faculty of Medicine Sousse, University of Sousse, Sousse 4000, Tunisia;
| | - Moncef BenKhalifa
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, 80025 Amiens, France; (V.B.); (H.K.-C.); (M.B.)
- ART and Reproductive Biology Laboratory, University Hospital and School of Medicine, Picardie University Jules Verne, CHU Sud, 80025 Amiens, France
| | - Mounir Ajina
- Service of Reproductive Biology, University Hospital Farhat Hached, Sousse, University of Sousse, Sousse 4000, Tunisia; (O.G.); (N.B.S.); (H.B.M.)
- Exercise Physiology and Physiopathology: From Integrated to Molecular “Biology, Medicine and Health” (Code: LR19ES09), Sousse 4002, Tunisia
| |
Collapse
|