1
|
Xie K, Wang C, Scifo E, Pearson B, Ryan D, Henzel K, Markert A, Schaaf K, Mi X, Tian X, Jia J, Wang M, Bonn S, Schölling M, Möhl C, Bano D, Zhou Y, Ehninger D. Intermittent fasting boosts sexual behavior by limiting the central availability of tryptophan and serotonin. Cell Metab 2025; 37:1189-1205.e7. [PMID: 40157367 DOI: 10.1016/j.cmet.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 12/02/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Aging affects reproductive capabilities in males through physiological and behavioral alterations, including endocrine changes and decreased libido. In this study, we investigated the influence of intermittent fasting (IF) on these aging-related declines, using male C57BL/6J mice. Our findings revealed that IF significantly preserved reproductive success in aged mice, not by improving traditional reproductive metrics such as sperm quality or endocrine functions but by enhancing mating behavior. This behavioral improvement was attributed to IF's ability to counter age-dependent increases in serotonergic inhibition, primarily through the decreased supply of the serotonin precursor tryptophan from the periphery to the brain. Our research underscores the potential of dietary interventions like IF in mitigating age-associated declines in male reproductive health and suggests a novel approach to managing conditions related to reduced sexual desire, highlighting the complex interplay between diet, metabolism, and reproductive behavior.
Collapse
Affiliation(s)
- Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Chengfeng Wang
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Enzo Scifo
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Brandon Pearson
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Devon Ryan
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Kristin Henzel
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Astrid Markert
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Kristina Schaaf
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Xue Mi
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Xin Tian
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Jiajia Jia
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Meiqin Wang
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Stefan Bonn
- Institute of Medical Systems Biology, Hamburg Center for Biomedical AI (bAIome), Molecular Neurobiology Hamburg (ZMNH), and Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Manuel Schölling
- Image and Data Analysis Facility, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Christoph Möhl
- Image and Data Analysis Facility, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Yu Zhou
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China.
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany.
| |
Collapse
|
2
|
Liu YD, Teng XM, Bai DD, Xing FY, Chang QR, Li JL, Gao SR, Liu WQ, Guo Y. Inhibition of aging-induced DNA hypermethylation by si-Dnmt3a/3b in pre-implantation embryos improves aberrant social behavior in offspring. Int J Biol Macromol 2025; 307:142130. [PMID: 40107549 DOI: 10.1016/j.ijbiomac.2025.142130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 02/24/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Advanced paternal age constitutes a noteworthy risk factor for neurodevelopmental disorders in progeny, encompassing conditions such as schizophrenia, autism, and atypical social behavior. Nonetheless, the precise underlying mechanisms remain inadequately comprehended. In this study, we delved into the alterations of DNA methylation induced by aging in sperm and pre-implantation embryos of male mice. A total of 3909 Differentially Methylated Regions (DMRs) were identified in sperm from aged male mice compared to young group. In addition, the overall DNA methylation in androgenetic 2-cell embryos exhibited a pronounced increase in aged group, leading to altered expression of genes related to neuropsychiatric disorders. Interestingly, a total of 242 shared DMRs were identified through the overlap of the sperm DMR sets and the 2-cell embryo DMR sets. This finding suggests the plausible involvement of these shared DMRs in the intergenerational transmission of epigenetic traits. Furthermore, F1 male mice from aged group exhibited a marked decrease in sociability and displayed DNA methylation alterations associated with nerve signal transduction components in their brain tissues. Intriguingly, inhibition of DNA methyltransferases (DNMT3A/3B) by siRNA in pre-implantation embryos improved abnormal social behaviors in F1 males from aged fathers, with concomitant changes in the expression of genes related to nerve development detected in 4-cell embryos. Our study indicates that male aging exert an influence not only on DNA methylation modification in sperm and pre-implantation embryo, but also on neurobehavioral abnormalities of their offspring. Repairing DNA methylation in pre-implantation embryos may offer a promising avenue for ameliorating abnormal social behavior in offspring.
Collapse
Affiliation(s)
- Ying-Dong Liu
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiao-Ming Teng
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Dan-Dan Bai
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Feng-Ying Xing
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, China
| | - Qiu-Rong Chang
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jin-Li Li
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Shao-Rong Gao
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Wen-Qiang Liu
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Yi Guo
- Centre for Assisted Reproduction of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
3
|
Kopf RK, Banks S, Brent LJN, Humphries P, Jolly CJ, Lee PC, Luiz OJ, Nimmo D, Winemiller KO. Loss of Earth's old, wise, and large animals. Science 2025; 387:eado2705. [PMID: 39571003 DOI: 10.1126/science.ado2705] [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: 01/25/2024] [Accepted: 11/07/2024] [Indexed: 01/04/2025]
Abstract
Earth's old animals are in decline. Despite this, emerging research is revealing the vital contributions of older individuals to cultural transmission, population dynamics, and ecosystem processes and services. Often the largest and most experienced, old individuals are most valued by humans and make important contributions to reproduction, information acquisition and cultural transmission, trophic dynamics, and resistance and resilience to natural and anthropogenic disturbance. These observations contrast with the senescence-focused paradigm of old age that has dominated the literature for more than a century yet are consistent with findings from behavioral ecology and life history theory. In this work, we review why the global loss of old individuals can be particularly detrimental to long-lived animals with indeterminate growth; those with increasing reproductive output with age; and those dependent on migration, sociality, and cultural transmission for survival. Longevity conservation is needed to protect the important ecological roles and ecosystem services provided by old animals.
Collapse
Affiliation(s)
- R Keller Kopf
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Darwin, NT, Australia
| | - Sam Banks
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Darwin, NT, Australia
| | - Lauren J N Brent
- Centre for Research in Animal Behaviour, University of Exeter, Exeter, UK
| | - Paul Humphries
- School of Agricultural, Environmental and Veterinary Sciences and Gulbali Institute, Charles Sturt University, Albury, NSW, Australia
| | - Chris J Jolly
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Darwin, NT, Australia
- School of Natural Sciences, Macquarie University, Sydney, NSW, Australia
| | - Phyllis C Lee
- Amboseli Trust for Elephants, Langata, Nairobi, Kenya
- Behaviour and Evolution Research Group, Psychology, Faculty of Natural Sciences, University of Stirling, Stirling, UK
| | - Osmar J Luiz
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Darwin, NT, Australia
| | - Dale Nimmo
- School of Agricultural, Environmental and Veterinary Sciences and Gulbali Institute, Charles Sturt University, Albury, NSW, Australia
| | - Kirk O Winemiller
- Department of Ecology and Conservation Biology, Texas A&M University, College Station, TX, USA
| |
Collapse
|
4
|
Wang SH, Lee JT, Lin MC, Wu CS, Thompson WK, Fan CC. Associations of paternal age with offspring under-five mortality and perinatal outcomes: a cohort study using claims data in Taiwan. BMJ PUBLIC HEALTH 2024; 2:e001113. [PMID: 40018562 PMCID: PMC11816320 DOI: 10.1136/bmjph-2024-001113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 10/31/2024] [Indexed: 03/01/2025]
Abstract
Background The causal relationship between advanced paternal age and offspring health is unclear, owing to familial confounders. This study examined the association of paternal age with offspring's under-five mortality and perinatal outcomes, using sibling comparison analyses to account for familial confounding factors. Methods A nationwide birth cohort study was designed based on Taiwan's single-payer compulsory National Health Insurance programme. Individuals born between 2001 and 2015 were included, resulting in 2454 104 live-born singletons. Among them, 1513 222 individuals had full sibling(s) who were included in the sibling-comparison analyses. Logistic regression analyses were used to evaluate the main study cohort whereas conditional logistic regressions were used in the sibling-comparison analyses. Results In the main cohort, paternal age categories showed a U-shaped relationship with offspring's under-five mortality in the crude analysis, which attenuated towards the null hypothesis after accounting for the measured potential confounders. There was an increased risk of premature birth (gestational age <37 weeks), low birth weight (<2500 g), large for gestational age (90th percentile) and low 5 min Apgar Score (<7) in individuals with a paternal age of >35 years. Sibling-comparison analyses that accounted for unmeasured familial time-invariant confounders showed that younger siblings with older paternal age had a lower risk of under-five mortality, low birth weight, small for gestational age (10th percentile), congenital defects and low 5 min Apgar Score, and a higher risk of premature birth and large for gestational age. Conclusions Children with older fathers had lower risks of under-five mortality, low birth weight, small for gestational age, congenital defects and low 5 min Apgar Score.
Collapse
Affiliation(s)
- Shi-Heng Wang
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Jian-Te Lee
- Department of Pediatrics, National Taiwan University Hospital Yun-Lin Branch, Douliou, Taiwan
| | - Mei-Chen Lin
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chi-Shin Wu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Psychiatry, National Taiwan University Hospital, Yunlin Branch, Douliu, Taiwan
| | - Wesley K Thompson
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
| | - Chun-Chieh Fan
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
5
|
Luo P, Guo H, Liu B, Zhang Z, Xie Y, Yao J, Li X, Bian J, Zhuang J, Ouyang B, Wu J. Transcriptome analyses reveal key features of mouse seminal vesicle during aging. Syst Biol Reprod Med 2024; 70:249-260. [PMID: 39167124 DOI: 10.1080/19396368.2024.2388121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Despite the significant morphological changes that occur in the seminal vesicles with aging, the transcriptomic characteristics remain largely unexplored. To address this, we performed bulk RNA sequencing on seminal vesicle samples from mice aged 3, 13, and 21 months to uncover transcriptomic alterations. Our findings reveal that aged seminal vesicles display cystic dilatation, epithelial hypoplasia, disordered muscle layers, fibrosis, and reduced proliferation capability. A comparison between 3-month-old and 21-month-old mice indicated that leukocyte-mediated immunity and leukocyte migration were the most significantly upregulated biological processes among differentially expressed genes (DEGs). Notably, several DEGs associated with "leukocyte migration," such as Vcam1, Cxcl13, and Ccl8, exhibited an increasing trend in transcriptomic and protein expression at three different time points in the seminal vesicles of mice. Additionally, we identified multiple aging-associated DEGs, including P21 and Tnfrsf1b. Two genes (Cd209f and Ccl8) were consistently upregulated across all six regions of the male reproductive glands (testis, epididymis, and seminal vesicle) in the comparison of bulk RNA datasets from 3-month-old and 21-month-old mice. These analyses highlight an enhanced state of immune and inflammatory response in aged seminal vesicles. This study represents the first exploration of the overall transcriptome landscape of seminal vesicles in a murine model of natural aging, offering new insights into the mechanisms underlying aging-related seminal vesicle dysfunction.
Collapse
Affiliation(s)
- Peng Luo
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital, SunYat-sen University, Guangzhou, China
| | - Haibin Guo
- Department of Reproductive Medicine, Henan Province People's Hospital, Zhengzhou, China
| | - Baoning Liu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiqiang Zhang
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yun Xie
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiahui Yao
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangping Li
- Department of Urology and Andrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Bian
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jintao Zhuang
- Department of Urology, The Eastern Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bin Ouyang
- Center for Reproductive Medicine, Guangdong Women and Children Hospital, Guangzhou, China
- Department of Andrology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jinhua Wu
- Department of Andrology, Ganzhou People's Hospital of Jiangxi Province, Ganzhou, China
| |
Collapse
|
6
|
Trigg N, Schjenken JE, Martin JH, Skerrett-Byrne DA, Smyth SP, Bernstein IR, Anderson AL, Stanger SJ, Simpson ENA, Tomar A, Teperino R, Conine CC, De Iuliis GN, Roman SD, Bromfield EG, Dun MD, Eamens AL, Nixon B. Subchronic elevation in ambient temperature drives alterations to the sperm epigenome and accelerates early embryonic development in mice. Proc Natl Acad Sci U S A 2024; 121:e2409790121. [PMID: 39527742 PMCID: PMC11588121 DOI: 10.1073/pnas.2409790121] [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: 05/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Forecasted increases in the prevalence and severity of extreme weather events accompanying changes in climatic behavior pose potential risk to the reproductive capacity of humans and animals of ecological and agricultural significance. While several studies have revealed that heat stress induced by challenges such as testicular insulation can elicit a marked negative effect on the male reproductive system, and particularly the production of spermatozoa, less is known about the immediate impact on male reproductive function following subchronic whole-body exposure to elevated ambient temperature. To address this knowledge gap, we exposed unrestrained male mice to heat stress conditions that emulate a heat wave (daily cycle of 8 h at 35 °C followed by 16 h at 25 °C) for a period of 7 d. Neither the testes or epididymides of heat-exposed male mice exhibited evidence of gross histological change, and similarly, spermatozoa of exposed males retained their functionality and ability to support embryonic development. However, the embryos generated from heat-exposed spermatozoa experienced pronounced changes in gene expression linked to acceleration of early embryo development, aberrant blastocyst hatching, and increased fetal:placental weight ratio. Such changes were causally associated with an altered sperm small noncoding RNA (sncRNA) profile, such that these developmental phenotypes were recapitulated by microinjection of wild-type embryos sired by control spermatozoa with RNAs extracted from heat-exposed spermatozoa. Such data highlight that even relatively modest excursions in ambient temperature can affect male reproductive function and identify the sperm sncRNA profile as a particular point of vulnerability to this imposed environmental stress.
Collapse
Affiliation(s)
- Natalie Trigg
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- Department of Genetics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Department of Pediatrics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Center for Reproductive and Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - John E. Schjenken
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Jacinta H. Martin
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - David A. Skerrett-Byrne
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Shannon P. Smyth
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- School of BioSciences Bio21 Molecular Sciences and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, VIC3010, Australia
| | - Ilana R. Bernstein
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Amanda L. Anderson
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Simone J. Stanger
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Ewan N. A. Simpson
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Archana Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Colin C. Conine
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Department of Pediatrics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Center for Reproductive and Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Geoffry N. De Iuliis
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Shaun D. Roman
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- NSW Health Pathology, Newcastle, NSW2300, Australia
| | - Elizabeth G. Bromfield
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- School of BioSciences Bio21 Molecular Sciences and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, VIC3010, Australia
| | - Matthew D. Dun
- Cancer Signaling Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Andrew L. Eamens
- School of Health, University of the Sunshine Coast, Maroochydore, QLD4558, Australia
| | - Brett Nixon
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| |
Collapse
|
7
|
Amir S, Arowolo O, Mironova E, McGaunn J, Oluwayiose O, Sergeyev O, Pilsner JR, Suvorov A. Mechanistic target of rapamycin (mTOR) pathway in Sertoli cells regulates age-dependent changes in sperm DNA methylation. eLife 2024; 13:RP90992. [PMID: 39283662 PMCID: PMC11405012 DOI: 10.7554/elife.90992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Over the past several decades, a trend toward delayed childbirth has led to increases in parental age at the time of conception. Sperm epigenome undergoes age-dependent changes increasing risks of adverse conditions in offspring conceived by fathers of advanced age. The mechanism(s) linking paternal age with epigenetic changes in sperm remain unknown. The sperm epigenome is shaped in a compartment protected by the blood-testes barrier (BTB) known to deteriorate with age. Permeability of the BTB is regulated by the balance of two mTOR complexes in Sertoli cells where mTOR complex 1 (mTORC1) promotes the opening of the BTB and mTOR complex 2 (mTORC2) promotes its integrity. We hypothesized that this balance is also responsible for age-dependent changes in the sperm epigenome. To test this hypothesis, we analyzed reproductive outcomes, including sperm DNA methylation in transgenic mice with Sertoli cell-specific suppression of mTORC1 (Rptor KO) or mTORC2 (Rictor KO). mTORC2 suppression accelerated aging of the sperm DNA methylome and resulted in a reproductive phenotype concordant with older age, including decreased testes weight and sperm counts, and increased percent of morphologically abnormal spermatozoa and mitochondrial DNA copy number. Suppression of mTORC1 resulted in the shift of DNA methylome in sperm opposite to the shift associated with physiological aging - sperm DNA methylome rejuvenation and mild changes in sperm parameters. These results demonstrate for the first time that the balance of mTOR complexes in Sertoli cells regulates the rate of sperm epigenetic aging. Thus, mTOR pathway in Sertoli cells may be used as a novel target of therapeutic interventions to rejuvenate the sperm epigenome in advanced-age fathers.
Collapse
Affiliation(s)
- Saira Amir
- Department of Environmental Health Sciences, University of MassachusettsAmherstUnited States
| | - Olatunbosun Arowolo
- Department of Environmental Health Sciences, University of MassachusettsAmherstUnited States
| | - Ekaterina Mironova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State UniversityMoscowRussian Federation
| | - Joseph McGaunn
- Department of Environmental Health Sciences, University of MassachusettsAmherstUnited States
| | - Oladele Oluwayiose
- Department of Obstetrics and Gynecology, Wayne State UniversityDetroitUnited States
| | - Oleg Sergeyev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State UniversityMoscowRussian Federation
| | - J Richard Pilsner
- Department of Obstetrics and Gynecology, Wayne State UniversityDetroitUnited States
| | - Alexander Suvorov
- Department of Environmental Health Sciences, University of MassachusettsAmherstUnited States
| |
Collapse
|
8
|
Izadi M, Sadri N, Abdi A, Serajian S, Jalalei D, Tahmasebi S. Epigenetic biomarkers in aging and longevity: Current and future application. Life Sci 2024; 351:122842. [PMID: 38879158 DOI: 10.1016/j.lfs.2024.122842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
The aging process has been one of the most necessary research fields in the current century, and knowing different theories of aging and the role of different genetic, epigenetic, molecular, and environmental modulating factors in increasing the knowledge of aging mechanisms and developing appropriate diagnostic, therapeutic, and preventive ways would be helpful. One of the most conserved signs of aging is epigenetic changes, including DNA methylation, histone modifications, chromatin remodeling, noncoding RNAs, and extracellular RNAs. Numerous biological processes and hallmarks are vital in aging development, but epigenomic alterations are especially notable because of their importance in gene regulation and cellular identity. The mounting evidence points to a possible interaction between age-related epigenomic alterations and other aging hallmarks, like genome instability. To extend a healthy lifespan and possibly reverse some facets of aging and aging-related diseases, it will be crucial to comprehend global and locus-specific epigenomic modifications and recognize corresponding regulators of health and longevity. In the current study, we will aim to discuss the role of epigenomic mechanisms in aging and the most recent developments in epigenetic diagnostic biomarkers, which have the potential to focus efforts on reversing the destructive signs of aging and extending the lifespan.
Collapse
Affiliation(s)
- Mehran Izadi
- Department of Infectious and Tropical Diseases, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
| | - Nariman Sadri
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Abdi
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Sahar Serajian
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Dorsa Jalalei
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Safa Tahmasebi
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Fouéré C, Hozé C, Besnard F, Boussaha M, Boichard D, Sanchez MP. Investigating the impact of paternal age, paternal heat stress, and estimation of non-genetic paternal variance on dairy cow phenotype. Genet Sel Evol 2024; 56:46. [PMID: 38890567 PMCID: PMC11184688 DOI: 10.1186/s12711-024-00918-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Linear models that are commonly used to predict breeding values in livestock species consider paternal influence solely as a genetic effect. However, emerging evidence in several species suggests the potential effect of non-genetic semen-mediated paternal effects on offspring phenotype. This study contributes to such research by analyzing the extent of non-genetic paternal effects on the performance of Holstein, Montbéliarde, and Normande dairy cows. Insemination data, including semen Batch Identifier (BI, a combination of bull identification and collection date), was associated with various traits measured in cows born from the insemination. These traits encompassed stature, milk production (milk, fat, and protein yields), udder health (somatic cell score and clinical mastitis), and female fertility (conception rates of heifers and cows). We estimated (1) the effects of age at collection and heat stress during spermatogenesis, and (2) the variance components associated with BI or Weekly aggregated BI (WBI). RESULTS Overall, the non-genetic paternal effect estimates were small and of limited biological importance. However, while heat stress during spermatogenesis did not show significant associations with any of the traits studied in daughters, we observed significant effects of bull age at semen collection on the udder health of daughters. Indeed, cows born from bulls collected after 1500 days of age had higher somatic cell scores compared to those born from bulls collected at a younger age (less than 400 days old) in both Holstein and Normande breeds (+ 3% and + 5% of the phenotypic mean, respectively). In addition, across all breeds and traits analyzed, the estimates of non-genetic paternal variance were consistently low, representing on average 0.13% and 0.09% of the phenotypic variance for BI and WBI, respectively (ranging from 0 to 0.7%). These estimates did not significantly differ from zero, except for milk production traits (milk, fat, and protein yields) in the Holstein breed and protein yield in the Montbéliarde breed when WBI was considered. CONCLUSIONS Our findings indicate that non-genetic paternal information transmitted through semen does not substantially influence the offspring phenotype in dairy cattle breeds for routinely measured traits. This lack of substantial impact may be attributed to limited transmission or minimal exposure of elite bulls to adverse conditions.
Collapse
Affiliation(s)
- Corentin Fouéré
- Eliance, 75012, Paris, France.
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
| | - Chris Hozé
- Eliance, 75012, Paris, France
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Florian Besnard
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- Idele, 75012, Paris, France
| | - Mekki Boussaha
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Didier Boichard
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Marie-Pierre Sanchez
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| |
Collapse
|
10
|
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
|
11
|
Wu D, Zhang K, Guan K, Khan FA, Pandupuspitasari NS, Negara W, Sun F, Huang C. Future in the past: paternal reprogramming of offspring phenotype and the epigenetic mechanisms. Arch Toxicol 2024; 98:1685-1703. [PMID: 38460001 DOI: 10.1007/s00204-024-03713-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/20/2024] [Indexed: 03/11/2024]
Abstract
That certain preconceptual paternal exposures reprogram the developmental phenotypic plasticity in future generation(s) has conceptualized the "paternal programming of offspring health" hypothesis. This transgenerational effect is transmitted primarily through sperm epigenetic mechanisms-DNA methylation, non-coding RNAs (ncRNAs) and associated RNA modifications, and histone modifications-and potentially through non-sperm-specific mechanisms-seminal plasma and circulating factors-that create 'imprinted' memory of ancestral information. The epigenetic landscape in sperm is highly responsive to environmental cues, due to, in part, the soma-to-germline communication mediated by epididymosomes. While human epidemiological studies and experimental animal studies have provided solid evidences in support of transgenerational epigenetic inheritance, how ancestral information is memorized as epigenetic codes for germline transmission is poorly understood. Particular elusive is what the downstream effector pathways that decode those epigenetic codes into persistent phenotypes. In this review, we discuss the paternal reprogramming of offspring phenotype and the possible underlying epigenetic mechanisms. Cracking these epigenetic mechanisms will lead to a better appreciation of "Paternal Origins of Health and Disease" and guide innovation of intervention algorithms to achieve 'healthier' outcomes in future generations. All this will revolutionize our understanding of human disease etiology.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | | | - Windu Negara
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
12
|
Sfakianoudis K, Zikopoulos A, Grigoriadis S, Seretis N, Maziotis E, Anifandis G, Xystra P, Kostoulas C, Giougli U, Pantos K, Simopoulou M, Georgiou I. The Role of One-Carbon Metabolism and Methyl Donors in Medically Assisted Reproduction: A Narrative Review of the Literature. Int J Mol Sci 2024; 25:4977. [PMID: 38732193 PMCID: PMC11084717 DOI: 10.3390/ijms25094977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
One-carbon (1-C) metabolic deficiency impairs homeostasis, driving disease development, including infertility. It is of importance to summarize the current evidence regarding the clinical utility of 1-C metabolism-related biomolecules and methyl donors, namely, folate, betaine, choline, vitamin B12, homocysteine (Hcy), and zinc, as potential biomarkers, dietary supplements, and culture media supplements in the context of medically assisted reproduction (MAR). A narrative review of the literature was conducted in the PubMed/Medline database. Diet, ageing, and the endocrine milieu of individuals affect both 1-C metabolism and fertility status. In vitro fertilization (IVF) techniques, and culture conditions in particular, have a direct impact on 1-C metabolic activity in gametes and embryos. Critical analysis indicated that zinc supplementation in cryopreservation media may be a promising approach to reducing oxidative damage, while female serum homocysteine levels may be employed as a possible biomarker for predicting IVF outcomes. Nonetheless, the level of evidence is low, and future studies are needed to verify these data. One-carbon metabolism-related processes, including redox defense and epigenetic regulation, may be compromised in IVF-derived embryos. The study of 1-C metabolism may lead the way towards improving MAR efficiency and safety and ensuring the lifelong health of MAR infants.
Collapse
Affiliation(s)
- Konstantinos Sfakianoudis
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli, 15232 Athens, Greece; (K.S.); (K.P.)
| | - Athanasios Zikopoulos
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
- Obstetrics and Gynecology, Royal Cornwall Hospital, Treliske, Truro TR1 3LJ, UK
| | - Sokratis Grigoriadis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Nikolaos Seretis
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Evangelos Maziotis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - George Anifandis
- Department of Obstetrics and Gynecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41222 Larisa, Greece;
| | - Paraskevi Xystra
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Charilaos Kostoulas
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Urania Giougli
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Konstantinos Pantos
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli, 15232 Athens, Greece; (K.S.); (K.P.)
| | - Mara Simopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Ioannis Georgiou
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| |
Collapse
|
13
|
Aburada N, Ito J, Inoue Y, Yamamoto T, Hayashi M, Teramoto N, Okada Y, Koshiishi Y, Shirasuna K, Iwata H. Effect of paternal aging and vitrification on mitochondrial DNA copy number and telomere length of mouse blastocysts. J Reprod Dev 2024; 70:65-71. [PMID: 38267053 PMCID: PMC11017102 DOI: 10.1262/jrd.2023-079] [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: 09/14/2023] [Accepted: 12/31/2023] [Indexed: 01/26/2024] Open
Abstract
In this study, we examined the effects of paternal aging on the mitochondrial DNA copy number (mt-cn), telomere length (TL), and gene expression in mouse embryos. The effects of vitrification on the mt-cn and TL of the embryos derived from young and aged male parents (YF and AF, respectively) were examined. C57BL/6N male mice were used for embryo production at 13-23 and 50-55 weeks of age. Two-cell stage embryos were collected from the oviducts of superovulated female mice (8-15 weeks old) and cultured for 24 h until the 8-cell stage, followed by embryo vitrification. Fresh and vitrified-warmed embryos were incubated for 2 days until the blastocyst stage, and mt-cn and TL were investigated. The cell-free mitochondrial DNA copy number (cf-mt-cn) in the spent culture medium (SCM) of the embryos was then investigated. RNA sequencing of blastocysts revealed that metabolic pathways, including oxidative phosphorylation and mTOR pathways, were enriched in differentially expressed genes. The mt-cn and TL of AF-derived blastocysts were lower and shorter, respectively, than those of YF-derived blastocysts. Paternal aging did not affect the blastocyst rate after vitrification. Vitrification of the 8-cell stage embryos did not affect the mt-cn of the blastocysts. However, it increased the cf-mt-cn (cell-free mt-cn) in the SCM of both YF- and AF-derived embryos. Vitrification did not affect the TL of either YF- or AF-derived embryos. Thus, paternal aging affected the mt-cn and TL of the embryos, but vitrification did not affect these parameters in either age groups.
Collapse
Affiliation(s)
- Nao Aburada
- Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | - Jun Ito
- Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | - Yuki Inoue
- Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | | | | | - Noko Teramoto
- Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | - Yuri Okada
- Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | | | | | - Hisataka Iwata
- Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| |
Collapse
|
14
|
Miyahara K, Tatehana M, Kikkawa T, Osumi N. Investigating the impact of paternal aging on murine sperm miRNA profiles and their potential link to autism spectrum disorder. Sci Rep 2023; 13:20608. [PMID: 38062235 PMCID: PMC10703820 DOI: 10.1038/s41598-023-47878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Paternal aging has consistently been linked to an increased risk of neurodevelopmental disorders, including autism spectrum disorder (ASD), in offspring. Recent evidence has highlighted the involvement of epigenetic factors. In this study, we aimed to investigate age-related alterations in microRNA (miRNA) profiles of mouse sperm and analyze target genes regulated by differentially expressed miRNAs (DEmiRNAs). Microarray analyses were conducted on sperm samples from mice at different ages: 3 months (3 M), over 12 M, and beyond 20 M. We identified 26 miRNAs with differential expression between the 3 and 20 M mice, 34 miRNAs between the 12 and 20 M mice, and 2 miRNAs between the 3 and 12 M mice. The target genes regulated by these miRNAs were significantly associated with apoptosis/ferroptosis pathways and the nervous system. We revealed alterations in sperm miRNA profiles due to aging and suggest that the target genes regulated by these DEmiRNAs are associated with apoptosis and the nervous system, implying a potential link between paternal aging and an increased risk of neurodevelopmental disorders such as ASD. The observed age-related changes in sperm miRNA profiles have the potential to impact sperm quality and subsequently affect offspring development.
Collapse
Affiliation(s)
- Kazusa Miyahara
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Misako Tatehana
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takako Kikkawa
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
15
|
Fresnedo-Ramírez J, Anderson ES, D'Amico-Willman K, Gradziel TM. A review of plant epigenetics through the lens of almond. THE PLANT GENOME 2023; 16:e20367. [PMID: 37434488 DOI: 10.1002/tpg2.20367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 07/13/2023]
Abstract
While genomes were originally seen as static entities that stably held and organized genetic information, recent advances in sequencing have uncovered the dynamic nature of the genome. New conceptualizations of the genome include complex relationships between the environment and gene expression that must be maintained, regulated, and sometimes even transmitted over generations. The discovery of epigenetic mechanisms has allowed researchers to understand how traits like phenology, plasticity, and fitness can be altered without changing the underlying deoxyribonucleic acid sequence. While many discoveries were first made in animal systems, plants provide a particularly complex set of epigenetic mechanisms due to unique aspects of their biology and interactions with human selective breeding and cultivation. In the plant kingdom, annual plants have received the most attention; however, perennial plants endure and respond to their environment and human management in distinct ways. Perennials include crops such as almond, for which epigenetic effects have long been linked to phenomena and even considered relevant for breeding. Recent discoveries have elucidated epigenetic phenomena that influence traits such as dormancy and self-compatibility, as well as disorders like noninfectious bud failure, which are known to be triggered by the environment and influenced by inherent aspects of the plant. Thus, epigenetics represents fertile ground to further understand almond biology and production and optimize its breeding. Here, we provide our current understanding of epigenetic regulation in plants and use almond as an example of how advances in epigenetics research can be used to understand biological fitness and agricultural performance in crop plants.
Collapse
Affiliation(s)
| | - Elizabeth S Anderson
- Department of Horticulture and Crop Science, The Ohio State University, Wooster, OH, USA
| | | | - Thomas M Gradziel
- Department of Plant Sciences, University of California, Davis, Davis, CA, USA
| |
Collapse
|
16
|
Klutstein M, Gonen N. Epigenetic aging of mammalian gametes. Mol Reprod Dev 2023; 90:785-803. [PMID: 37997675 DOI: 10.1002/mrd.23717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/25/2023]
Abstract
The process of aging refers to physiological changes that occur to an organism as time progresses and involves changes to DNA, proteins, metabolism, cells, and organs. Like the rest of the cells in the body, gametes age, and it is well established that there is a decline in reproductive capabilities in females and males with aging. One of the major pathways known to be involved in aging is epigenetic changes. The epigenome is the multitude of chemical modifications performed on DNA and chromatin that affect the ability of chromatin to be transcribed. In this review, we explore the effects of aging on female and male gametes with a focus on the epigenetic changes that occur in gametes throughout aging. Quality decline in oocytes occurs at a relatively early age. Epigenetic changes constitute an important part of oocyte aging. DNA methylation is reduced with age, along with reduced expression of DNA methyltransferases (DNMTs). Histone deacetylases (HDAC) expression is also reduced, and a loss of heterochromatin marks occurs with age. As a consequence of heterochromatin loss, retrotransposon expression is elevated, and aged oocytes suffer from DNA damage. In sperm, aging affects sperm number, motility and fecundity, and epigenetic changes may constitute a part of this process. 5 methyl-cytosine (5mC) methylation is elevated in sperm from aged men, but methylation on Long interspersed nuclear elements (LINE) elements is reduced. Di and trimethylation of histone 3 lysine 9 (H3K9me2/3) is reduced in sperm from aged men and trimethylation of histone 3 lysine 27 (H3K27me3) is elevated. The protamine makeup of sperm from aged men is also changed, with reduced protamine expression and a misbalanced ratio between protamine proteins protamine P1 and protamine P2. The study of epigenetic reproductive aging is recently gaining interest. The current status of the field suggests that many aspects of gamete epigenetic aging are still open for investigation. The clinical applications of these investigations have far-reaching consequences for fertility and sociological human behavior.
Collapse
Affiliation(s)
- Michael Klutstein
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitzan Gonen
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
17
|
Malamitsi-Puchner A, Briana DD. Advanced parental age affects cardiometabolic risk in offspring. Acta Paediatr 2023; 112:2307-2311. [PMID: 37410550 DOI: 10.1111/apa.16902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/08/2023]
Abstract
Advanced age at conception usually refers to human mothers aged 35 years plus and fathers aged 40 years plus. Advanced parental age may be responsible for genetic and/or epigenetic alterations and may affect the health of offspring. Limited epidemiological and experimental studies have addressed the effect of advanced parental age on cardio-metabolic functions in human and rodent offspring. This mini review aimed to present the knowledge by focusing on adverse and favourable outcomes related to sex-specific risks and intergenerational inheritance. The outcomes identified by this review were mainly negative, but there were also some positive results.
Collapse
Affiliation(s)
- Ariadne Malamitsi-Puchner
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina D Briana
- Neonatal Intensive Care Unit, 3rd Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
18
|
Santiago J, Silva JV, Santos MAS, Fardilha M. Age-Dependent Alterations in Semen Parameters and Human Sperm MicroRNA Profile. Biomedicines 2023; 11:2923. [PMID: 38001924 PMCID: PMC10669352 DOI: 10.3390/biomedicines11112923] [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: 10/16/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The trend to delay parenthood is increasing, impacting fertility and reproductive outcomes. Advanced paternal age (APA), defined as men's age above 40 years at conception, has been linked with testicular impairment, abnormal semen parameters, and poor reproductive and birth outcomes. Recently, the significance of sperm microRNA for fertilization and embryonic development has emerged. This work aimed to investigate the effects of men's age on semen parameters and sperm microRNA profiles. The ejaculates of 333 Portuguese men were collected between 2018 and 2022, analyzed according to WHO guidelines, and a density gradient sperm selection was performed. For microRNA expression analysis, 16 normozoospermic human sperm samples were selected and divided into four age groups: ≤30, 31-35, 36-40, and >40 years. microRNA target genes were retrieved from the miRDB and TargetScan databases and Gene Ontology analysis was performed using the DAVID tool. No significant correlation was found between male age and conventional semen parameters, except for volume. Fifteen differentially expressed microRNAs (DEMs) between groups were identified. Enrichment analysis suggested the involvement of DEMs in the sperm of men with advanced age in critical biological processes like embryonic development, morphogenesis, and male gonad development. Targets of DEMs were involved in signaling pathways previously associated with the ageing process, including cellular senescence, autophagy, insulin, and mTOR pathways. These results suggest that although conventional semen parameters were not affected by men's age, alterations in microRNA regulation may occur and be responsible for poor fertility and reproductive outcomes associated with APA.
Collapse
Affiliation(s)
- Joana Santiago
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
| | - Joana V. Silva
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
| | - Manuel A. S. Santos
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
- Multidisciplinary Institute of Ageing, MIA-Portugal, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Margarida Fardilha
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (J.V.S.); (M.A.S.S.)
| |
Collapse
|
19
|
Zablocki-Thomas P, Rebout N, Karaskiewicz CL, Bales KL. Survival rates and mortality risks of Plecturocebus cupreus at the California National Primate Research Center. Am J Primatol 2023; 85:e23531. [PMID: 37424137 PMCID: PMC10921862 DOI: 10.1002/ajp.23531] [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: 05/13/2022] [Revised: 04/27/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023]
Abstract
This article describes survivorship and explores factors affecting mortality risks in a captive colony of coppery titi monkeys (Plecturocebus cupreus) housed at the California National Primate Research Center (CNPRC), at UC Davis, in Davis, CA. We analyzed data collected on individuals since the colony's creation in the 1960s, with a sample of 600 animals with partially complete information (date of birth, age at death, body mass, parental lineage). We used three methods: (1) Kaplan-Meier regressions followed by a log-rank test to compare survival in male and female titi monkeys, (2) a breakpoint analysis to identify shifts in the survival curves, and (3) Cox regressions to test the effect of body mass change, parental pair tenure, and parental age on mortality risk. We found that males tend to have a longer median lifespan than females (14.9 and 11.4 years; p = 0.094) and that survival decreases earlier in males than in females during adulthood (9.8 and 16.2 years). A body mass loss of 10% from adulthood to the time of death led to a 26% higher risk of dying (p < 0.001) as compared to an individual with stable body mass. We found no evidence of sociobiological factors on mortality risks (parental age, parental pair tenure), but an exploratory analysis suggested that a higher rate of offspring conceptions increases mortality risks. This description of factors influencing survival and mortality in titi monkeys is a first step toward understanding aging in this species to consider titi monkeys as a primate model for socioemotional aging.
Collapse
Affiliation(s)
| | | | - Chloe L. Karaskiewicz
- California National Primate Research Center, Davis CA
- Department of Psychology, University of California, Davis, United States of America
| | - Karen L. Bales
- California National Primate Research Center, Davis CA
- Department of Psychology, University of California, Davis, United States of America
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, United States of America
| |
Collapse
|
20
|
Xie K, Ehninger D. Ageing-associated phenotypes in mice. Mech Ageing Dev 2023; 214:111852. [PMID: 37454704 DOI: 10.1016/j.mad.2023.111852] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Ageing is a continuous process in life featuring progressive damage accumulation that leads to physiological decline, functional deterioration and ultimately death of an organism. Based on the relatively close anatomical and physiological similarity to humans, the mouse has been proven as a valuable model organism in ageing research over the last decades. In this review, we survey methods and tools currently in use to assess ageing phenotypes in mice. We summarize a range of ageing-associated alterations detectable at two major levels of analysis: (1) physiology and pathophysiology and (2) molecular biomarkers. Age-sensitive phenotypes provided in this article may serve to inform future studies targeting various aspects of organismal ageing in mice. In addition, we discuss conceptual and technical challenges faced by previous ageing studies in mice and, where possible, provide recommendations on how to resolve some of these issues.
Collapse
Affiliation(s)
- Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany.
| |
Collapse
|
21
|
Galkin F, Kovalchuk O, Koldasbayeva D, Zhavoronkov A, Bischof E. Stress, diet, exercise: Common environmental factors and their impact on epigenetic age. Ageing Res Rev 2023; 88:101956. [PMID: 37211319 DOI: 10.1016/j.arr.2023.101956] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Epigenetic aging clocks have gained significant attention as a tool for predicting age-related health conditions in clinical and research settings. They have enabled geroscientists to study the underlying mechanisms of aging and assess the effectiveness of anti-aging therapies, including diet, exercise and environmental exposures. This review explores the effects of modifiable lifestyle factors' on the global DNA methylation landscape, as seen by aging clocks. We also discuss the underlying mechanisms through which these factors contribute to biological aging and provide comments on what these findings mean for people willing to build an evidence-based pro-longevity lifestyle.
Collapse
Affiliation(s)
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Canada
| | | | - Alex Zhavoronkov
- Deep Longevity, Hong Kong; Insilico Medicine, Hong Kong; Buck Institute for Research on Aging, Novato, CA, USA
| | - Evelyne Bischof
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Shanghai University of Medicine and Health Sciences, Shanghai, China; Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Via S. Pansini, 580131, Naples, Italy
| |
Collapse
|
22
|
Kachhawaha AS, Mishra S, Tiwari AK. Epigenetic control of heredity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:25-60. [PMID: 37225323 DOI: 10.1016/bs.pmbts.2023.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Epigenetics is the field of science that deals with the study of changes in gene function that do not involve changes in DNA sequence and are heritable while epigenetics inheritance is the process of transmission of epigenetic modifications to the next generation. It can be transient, intergenerational, or transgenerational. There are various epigenetic modifications involving mechanisms such as DNA methylation, histone modification, and noncoding RNA expression, all of which are inheritable. In this chapter, we summarize the information on epigenetic inheritance, its mechanism, inheritance studies on various organisms, factors affecting epigenetic modifications and their inheritance, and the role of epigenetic inheritance in the heritability of diseases.
Collapse
Affiliation(s)
- Akanksha Singh Kachhawaha
- Laboratory of Forensic Chemistry & Toxicology, School of Forensic Sciences, National Forensic Sciences University (NFSU), Gandhinagar, Gujarat, India
| | - Sarita Mishra
- Laboratory of Forensic Chemistry & Toxicology, School of Forensic Sciences, National Forensic Sciences University (NFSU), Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India.
| |
Collapse
|
23
|
Fricke C, Sanghvi K, Emery M, Lindenbaum I, Wigby S, Ramm SA, Sepil I. Timeless or tainted? The effects of male ageing on seminal fluid. Front Ecol Evol 2023. [DOI: 10.3389/fevo.2023.1066022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Reproductive ageing can occur due to the deterioration of both the soma and germline. In males, it has mostly been studied with respect to age-related changes in sperm. However, the somatic component of the ejaculate, seminal fluid, is also essential for maintaining reproductive function. Whilst we know that seminal fluid proteins (SFPs) are required for male reproductive success across diverse taxa, age-related changes in SFP quantity and composition are little understood. Additionally, only few studies have explored the reproductive ageing of the tissues that produce SFPs, and the resulting reproductive outcomes. Here we provide a systematic review of studies addressing how advancing male age affects the production and properties of seminal fluid, in particular SFPs and oxidative stress, highlighting many open questions and generating new hypotheses for further research. We additionally discuss how declines in function of different components of seminal fluid, such as SFPs and antioxidants, could contribute to age-related loss of reproductive ability. Overall, we find evidence that ageing results in increased oxidative stress in seminal fluid and a decrease in the abundance of various SFPs. These results suggest that seminal fluid contributes towards important age-related changes influencing male reproduction. Thus, it is essential to study this mostly ignored component of the ejaculate to understand male reproductive ageing, and its consequences for sexual selection and paternal age effects on offspring.
Collapse
|
24
|
The effect of advanced paternal age on the lifespan of male offspring in an ancient Chinese genealogical data set. Maturitas 2023; 168:44-48. [PMID: 36442347 DOI: 10.1016/j.maturitas.2022.10.008] [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: 04/30/2022] [Revised: 10/02/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Advanced paternal age has been reported to be associated with a variety of short-term outcomes in offspring, but long-term effects are rarely examined. The present study evaluated the impact of advanced paternal age on offspring's longevity. METHODS We studied the effect of paternal reproductive age on the lifespan of male offspring using a Chinese genealogy data set that spans 226 years of the Qing Dynasty (1683-1909). Multivariable-adjusted Cox regression analyses of 1274 men with survival data were used to calculate hazard ratios (HRs) of advanced parental age at reproduction. We also evaluated whether the lifespan of brothers differed when they were born to the same parents at different ages. RESULTS In models adjusted for maternal age, advanced paternal age was negatively associated with the lifespan of male offspring. Individuals born to fathers aged >40 years had a 32 % higher HR of a lifespan shorter than those born to fathers aged 25-29 years (adjusted HR 1.320, 95 % CI: 1.066-1.634). The adjusted HR for offspring born to fathers aged 35-39 years was 1.232 (95 % CI: 1.013-1.500). Older brothers born to fathers aged 20-34 years had a significantly lower risk of a reduced lifespan compared with their younger brothers with fathers aged ≥35 years at reproduction (P < 0.01). CONCLUSION Advanced paternal age at reproduction is a negative factor for male offspring's life expectancy. With the sustained increase in paternal age over the past generation, further investigation is warranted into the impact on birth outcomes and public health.
Collapse
|
25
|
Aminzadeh-Gohari S, Kofler B, Herzog C. Dietary restriction in senolysis and prevention and treatment of disease. Crit Rev Food Sci Nutr 2022; 64:5242-5268. [PMID: 36484738 PMCID: PMC7616065 DOI: 10.1080/10408398.2022.2153355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging represents a key risk factor for a plethora of diseases. Targeting detrimental processes which occur during aging, especially before onset of age-related disease, could provide drastic improvements in healthspan. There is increasing evidence that dietary restriction (DR), including caloric restriction, fasting, or fasting-mimicking diets, extend both lifespan and healthspan. This has sparked interest in the use of dietary regimens as a non-pharmacological means to slow aging and prevent disease. Here, we review the current evidence on the molecular mechanisms underlying DR-induced health improvements, including removal of senescent cells, metabolic reprogramming, and epigenetic rejuvenation.
Collapse
Affiliation(s)
- Sepideh Aminzadeh-Gohari
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| |
Collapse
|
26
|
Xie K, Fuchs H, Scifo E, Liu D, Aziz A, Aguilar-Pimentel JA, Amarie OV, Becker L, da Silva-Buttkus P, Calzada-Wack J, Cho YL, Deng Y, Edwards AC, Garrett L, Georgopoulou C, Gerlini R, Hölter SM, Klein-Rodewald T, Kramer M, Leuchtenberger S, Lountzi D, Mayer-Kuckuk P, Nover LL, Oestereicher MA, Overkott C, Pearson BL, Rathkolb B, Rozman J, Russ J, Schaaf K, Spielmann N, Sanz-Moreno A, Stoeger C, Treise I, Bano D, Busch DH, Graw J, Klingenspor M, Klopstock T, Mock BA, Salomoni P, Schmidt-Weber C, Weiergräber M, Wolf E, Wurst W, Gailus-Durner V, Breteler MMB, Hrabě de Angelis M, Ehninger D. Deep phenotyping and lifetime trajectories reveal limited effects of longevity regulators on the aging process in C57BL/6J mice. Nat Commun 2022; 13:6830. [PMID: 36369285 PMCID: PMC9652467 DOI: 10.1038/s41467-022-34515-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022] Open
Abstract
Current concepts regarding the biology of aging are primarily based on studies aimed at identifying factors regulating lifespan. However, lifespan as a sole proxy measure for aging can be of limited value because it may be restricted by specific pathologies. Here, we employ large-scale phenotyping to analyze hundreds of markers in aging male C57BL/6J mice. For each phenotype, we establish lifetime profiles to determine when age-dependent change is first detectable relative to the young adult baseline. We examine key lifespan regulators (putative anti-aging interventions; PAAIs) for a possible countering of aging. Importantly, unlike most previous studies, we include in our study design young treated groups of animals, subjected to PAAIs prior to the onset of detectable age-dependent phenotypic change. Many PAAI effects influence phenotypes long before the onset of detectable age-dependent change, but, importantly, do not alter the rate of phenotypic change. Hence, these PAAIs have limited effects on aging.
Collapse
Affiliation(s)
- Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Enzo Scifo
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Dan Liu
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.,Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Juan Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Oana Veronica Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Julia Calzada-Wack
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Yi-Li Cho
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Yushuang Deng
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - A Cole Edwards
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Lillian Garrett
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Christina Georgopoulou
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Raffaele Gerlini
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Sabine M Hölter
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Tanja Klein-Rodewald
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | | | - Stefanie Leuchtenberger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Dimitra Lountzi
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Phillip Mayer-Kuckuk
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Lena L Nover
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Manuela A Oestereicher
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Clemens Overkott
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Brandon L Pearson
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.,Mailman School of Public Health, Columbia University, 630W. 168th St., New York, NY, 10032, USA
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Member of German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jan Rozman
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Member of German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Institute of Molecular Genetics of the Czech Academy of Sciences, Czech Centre for Phenogenomics, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Jenny Russ
- Nuclear Function Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Kristina Schaaf
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Adrián Sanz-Moreno
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Claudia Stoeger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Irina Treise
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München, 81675, Munich, Germany
| | - Jochen Graw
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, 85350, Freising-Weihenstephan, Germany
| | - Thomas Klopstock
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-University Munich, 80336, Munich, Germany.,DZNE, German Center for Neurodegenerative Diseases, 80336, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD, 20892, USA
| | - Paolo Salomoni
- Nuclear Function Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Carsten Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technische Universität München, and Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Marco Weiergräber
- Research Group Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices, 53175, Bonn, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,DZNE, German Center for Neurodegenerative Diseases, 80336, Munich, Germany.,Chair of Developmental Genetics, TUM School of Life Sciences (SoLS), Technische Universität München, Freising, Germany
| | - Valérie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.,Institute for Medical Biometry, Informatics and Epidemiology, Faculty of Medicine, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Member of German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Chair of Experimental Genetics, TUM School of Life Sciences (SoLS), Technische Universität München, 85354, Freising, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.
| |
Collapse
|
27
|
Sharman P, Young AJ, Wilson AJ. Evidence of maternal and paternal age effects on speed in thoroughbred racehorses. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220691. [PMID: 36249332 DOI: 10.5061/dryad.qbzkh18m0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/16/2022] [Indexed: 05/25/2023]
Abstract
Effects of parental age on offspring viability have been reported in a wide range of species. However, to what extent parental age influences offspring traits beyond viability remains unclear. Moreover, previous research has primarily focused on maternal age effects. The purpose of this study was to test for paternal and maternal age effects on offspring speed in thoroughbred racehorses. We analysed over 900 000 race performances by over 100 000 horses on British racecourses between 1996 and 2019. With knowledge of the age of all 41 107 dams and 2 887 sires at offspring conception, we jointly modelled maternal and paternal age effects using a 'within-individual centring' approach. Within-parents, we identified a significant effect of maternal age on offspring speed of -0.017 yards s-1 yr-1 and a corresponding paternal age effect of -0.011 yards s-1 yr-1. Although maternal age effects were stronger (more negative), the existence and magnitude of paternal effects is particularly noteworthy, given thoroughbred sires have no involvement in parental care. Our results also suggest that the selective disappearance of both sires and dams is ongoing. These findings could potentially be used to optimize thoroughbred racehorse breeding decisions, and more generally, add to the increasing body of evidence that both maternal and paternal age affect a range of offspring characteristics.
Collapse
Affiliation(s)
- Patrick Sharman
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| | - Andrew J Young
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| | - Alastair J Wilson
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| |
Collapse
|
28
|
Sharman P, Young AJ, Wilson AJ. Evidence of maternal and paternal age effects on speed in thoroughbred racehorses. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220691. [PMID: 36249332 PMCID: PMC9532991 DOI: 10.1098/rsos.220691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/16/2022] [Indexed: 05/10/2023]
Abstract
Effects of parental age on offspring viability have been reported in a wide range of species. However, to what extent parental age influences offspring traits beyond viability remains unclear. Moreover, previous research has primarily focused on maternal age effects. The purpose of this study was to test for paternal and maternal age effects on offspring speed in thoroughbred racehorses. We analysed over 900 000 race performances by over 100 000 horses on British racecourses between 1996 and 2019. With knowledge of the age of all 41 107 dams and 2 887 sires at offspring conception, we jointly modelled maternal and paternal age effects using a 'within-individual centring' approach. Within-parents, we identified a significant effect of maternal age on offspring speed of -0.017 yards s-1 yr-1 and a corresponding paternal age effect of -0.011 yards s-1 yr-1. Although maternal age effects were stronger (more negative), the existence and magnitude of paternal effects is particularly noteworthy, given thoroughbred sires have no involvement in parental care. Our results also suggest that the selective disappearance of both sires and dams is ongoing. These findings could potentially be used to optimize thoroughbred racehorse breeding decisions, and more generally, add to the increasing body of evidence that both maternal and paternal age affect a range of offspring characteristics.
Collapse
Affiliation(s)
- Patrick Sharman
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| | - Andrew J. Young
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| | - Alastair J. Wilson
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| |
Collapse
|
29
|
Sharman P, Young AJ, Wilson AJ. Evidence of maternal and paternal age effects on speed in thoroughbred racehorses. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220691. [PMID: 36249332 DOI: 10.6084/m9.figshare.c.6228607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/16/2022] [Indexed: 05/25/2023]
Abstract
Effects of parental age on offspring viability have been reported in a wide range of species. However, to what extent parental age influences offspring traits beyond viability remains unclear. Moreover, previous research has primarily focused on maternal age effects. The purpose of this study was to test for paternal and maternal age effects on offspring speed in thoroughbred racehorses. We analysed over 900 000 race performances by over 100 000 horses on British racecourses between 1996 and 2019. With knowledge of the age of all 41 107 dams and 2 887 sires at offspring conception, we jointly modelled maternal and paternal age effects using a 'within-individual centring' approach. Within-parents, we identified a significant effect of maternal age on offspring speed of -0.017 yards s-1 yr-1 and a corresponding paternal age effect of -0.011 yards s-1 yr-1. Although maternal age effects were stronger (more negative), the existence and magnitude of paternal effects is particularly noteworthy, given thoroughbred sires have no involvement in parental care. Our results also suggest that the selective disappearance of both sires and dams is ongoing. These findings could potentially be used to optimize thoroughbred racehorse breeding decisions, and more generally, add to the increasing body of evidence that both maternal and paternal age affect a range of offspring characteristics.
Collapse
Affiliation(s)
- Patrick Sharman
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| | - Andrew J Young
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| | - Alastair J Wilson
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, Cornwall TR10 9FE, UK
| |
Collapse
|
30
|
Early Development of Cardiac Fibrosis in Young Old-Father Offspring. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8770136. [PMID: 36193084 PMCID: PMC9526616 DOI: 10.1155/2022/8770136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022]
Abstract
Cardiac aging is characterized by progressive fibrosis. Epidemiological studies have found that advanced paternal age is associated with an increased risk of heart failure in the next generation. This study is aimed at evaluating the effect of paternal age, in the young male rat progeny, on cardiac phenotype under circulatory stress conditions. Offspring rats were obtained by mating old males (24 months old) with young females (two months old) and by mating young males (two months old) with the same young females. Hypertension was induced in old father offspring (OFO) rats and young old father (YFO) offspring rats using L-NAME (N(ω)-nitro-L-arginine methyl ester). The OFO L-NAME rats showed a high blood pressure phenotype associated with substantial cardiac hypertrophy and an exacerbation of cardiac fibrosis compared to the YFO L-NAME rats. Histological analysis of heart tissue showed an expansion of the extracellular matrix, with fibroblasts displaying markers of epicardial origin (Tcf21, Tbx18, and Wt1) in the OFO group. Moreover, western blot and protein phosphorylation antibody array identified the TGF-β2 receptor pathway as preferentially activated in aged hearts as well as in OFO cardiac tissue treated with L-NAME. In addition, old father offspring rats (OFO+OFO L-NAME) had increased cardiac DNA methylation. In young hypertensive progeny, advanced paternal age at conception may be a risk factor for early progression towards cardiac fibrosis. An intergenerational transmission may be behind the paternal age-related cardiac remodeling in the young offspring.
Collapse
|
31
|
Ashapkin V, Suvorov A, Pilsner JR, Krawetz SA, Sergeyev O. Age-associated epigenetic changes in mammalian sperm: implications for offspring health and development. Hum Reprod Update 2022; 29:24-44. [PMID: 36066418 PMCID: PMC9825272 DOI: 10.1093/humupd/dmac033] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 08/05/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Modern reproductive behavior in most developed countries is characterized by delayed parenthood. Older gametes are generally less fertile, accumulating and compounding the effects of varied environmental exposures that are modified by lifestyle factors. Clinicians are primarily concerned with advanced maternal age, while the influence of paternal age on fertility, early development and offspring health remains underappreciated. There is a growing trend to use assisted reproductive technologies for couples of advanced reproductive age. Thus, the number of children born from older gametes is increasing. OBJECTIVE AND RATIONALE We review studies reporting age-associated epigenetic changes in mammals and humans in sperm, including DNA methylation, histone modifications and non-coding RNAs. The interplay between environment, fertility, ART and age-related epigenetic signatures is explored. We focus on the association of sperm epigenetics on epigenetic and phenotype events in embryos and offspring. SEARCH METHODS Peer-reviewed original and review articles over the last two decades were selected using PubMed and the Web of Science for this narrative review. Searches were performed by adopting the two groups of main terms. The first group included 'advanced paternal age', 'paternal age', 'postponed fatherhood', 'late fatherhood', 'old fatherhood' and the second group included 'sperm epigenetics', 'sperm', 'semen', 'epigenetic', 'inheritance', 'DNA methylation', 'chromatin', 'non-coding RNA', 'assisted reproduction', 'epigenetic clock'. OUTCOMES Age is a powerful factor in humans and rodent models associated with increased de novo mutations and a modified sperm epigenome. Age affects all known epigenetic mechanisms, including DNA methylation, histone modifications and profiles of small non-coding (snc)RNA. While DNA methylation is the most investigated, there is a controversy about the direction of age-dependent changes in differentially hypo- or hypermethylated regions with advanced age. Successful development of the human sperm epigenetic clock based on cross-sectional data and four different methods for DNA methylation analysis indicates that at least some CpG exhibit a linear relationship between methylation levels and age. Rodent studies show a significant overlap between genes regulated through age-dependent differentially methylated regions and genes targeted by age-dependent sncRNA. Both age-dependent epigenetic mechanisms target gene networks enriched for embryo developmental, neurodevelopmental, growth and metabolic pathways. Thus, age-dependent changes in the sperm epigenome cannot be described as a stochastic accumulation of random epimutations and may be linked with autism spectrum disorders. Chemical and lifestyle exposures and ART techniques may affect the epigenetic aging of sperm. Although most epigenetic modifications are erased in the early mammalian embryo, there is growing evidence that an altered offspring epigenome and phenotype is linked with advanced paternal age due to the father's sperm accumulating epigenetic changes with time. It has been hypothesized that age-induced changes in the sperm epigenome are profound, physiological and dynamic over years, yet stable over days and months, and likely irreversible. WIDER IMPLICATIONS This review raises a concern about delayed fatherhood and age-associated changes in the sperm epigenome that may compromise reproductive health of fathers and transfer altered epigenetic information to subsequent generations. Prospective studies using healthy males that consider confounders are recommended. We suggest a broader discussion focused on regulation of the father's age in natural and ART conceptions is needed. The professional community should be informed and should raise awareness in the population and when counseling older men.
Collapse
Affiliation(s)
| | | | - J Richard Pilsner
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Stephen A Krawetz
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Oleg Sergeyev
- Correspondence address. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskye Gory, House 1, Building 40, Room 322, Moscow 119992, Russia. E-mail: https://orcid.org/0000-0002-5745-3348
| |
Collapse
|
32
|
Aich U, Chowdhury S, Jennions MD. Separating the effects of paternal age and mating history: Evidence for sex-specific paternal effect in eastern mosquitofish. Evolution 2022; 76:1565-1577. [PMID: 35544673 PMCID: PMC9543789 DOI: 10.1111/evo.14498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 01/22/2023]
Abstract
Paternal age and past mating effort by males are often confounded, which can affect our understanding of a father's age effects. To our knowledge, only a few studies have standardized mating history when testing for effects of paternal age, and none has simultaneously disentangled how paternal age and mating history might jointly influence offspring traits. Here, we experimentally manipulated male mating history to tease apart its effects from those of paternal age on female fertility and offspring traits in the eastern mosquitofish (Gambusia holbrooki). Male age did not affect female fertility. However, males with greater past mating effort produced significantly larger broods. Paternal age and mating history interacted to affect sons' body size: sons sired by old-virgin males were larger than those sired by old-mated males, but this was not the case for younger fathers. Intriguingly, however, sons sired by old-virgin males tended to produce fewer sperms than those sired by old-mated males, indicating a potential trade-off in beneficial paternal effects. Finally, neither paternal age nor mating history affected daughter's fitness. Our results highlight that variation in offspring traits attributed to paternal age effect could partly arise due to a father's mating history, and not simply to his chronological age.
Collapse
Affiliation(s)
- Upama Aich
- Division of Ecology & EvolutionResearch School of BiologyThe Australian National UniversityCanberraAustralia,School of Biological SciencesMonash UniversityClaytonVictoriaAustralia
| | - Shawan Chowdhury
- School of Biological SciencesUniversity of QueenslandSaint LuciaQueenslandAustralia
| | - Michael D. Jennions
- Division of Ecology & EvolutionResearch School of BiologyThe Australian National UniversityCanberraAustralia
| |
Collapse
|
33
|
Shenoi VN, Brengdahl MI, Grace JL, Eriksson B, Rydén P, Friberg U. A genome-wide test for paternal indirect genetic effects on lifespan in Drosophila melanogaster. Proc Biol Sci 2022; 289:20212707. [PMID: 35538781 PMCID: PMC9091837 DOI: 10.1098/rspb.2021.2707] [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] [Indexed: 01/04/2023] Open
Abstract
Exposing sires to various environmental manipulations has demonstrated that paternal effects can be non-trivial also in species where male investment in offspring is almost exclusively limited to sperm. Whether paternal effects also have a genetic component (i.e. paternal indirect genetic effects (PIGEs)) in such species is however largely unknown, primarily because of methodological difficulties separating indirect from direct effects of genes. PIGEs may nevertheless be important since they have the capacity to contribute to evolutionary change. Here we use Drosophila genetics to construct a breeding design that allows testing nearly complete haploid genomes (more than 99%) for PIGEs. Using this technique, we estimate the variance in male lifespan due to PIGEs among four populations and compare this to the total paternal genetic variance (the sum of paternal indirect and direct genetic effects). Our results indicate that a substantial part of the total paternal genetic variance results from PIGEs. A screen of 38 haploid genomes, randomly sampled from a single population, suggests that PIGEs also influence variation in lifespan within populations. Collectively, our results demonstrate that PIGEs may constitute an underappreciated source of phenotypic variation.
Collapse
Affiliation(s)
| | | | - Jaime L. Grace
- Department of Biology, Loyola University Chicago, 1032 W. Sheridan Rd., Chicago, IL 60660, USA
| | - Björn Eriksson
- Unit of Chemical Ecology, Department of Plant Protection Biology, Swedish University of Agricultural Sciences, Sundsvägen 14, Box 102, 230 53 Alnarp, Sweden
| | - Patrik Rydén
- Department of Mathematics and Mathematical Statistics, Umeå University, 901 87 Umeå, Sweden,Computational Life Science Cluster (CLiC), Umeå University, 901 87 Umeå, Sweden
| | - Urban Friberg
- IFM Biology, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
34
|
Costes V, Chaulot-Talmon A, Sellem E, Perrier JP, Aubert-Frambourg A, Jouneau L, Pontlevoy C, Hozé C, Fritz S, Boussaha M, Le Danvic C, Sanchez MP, Boichard D, Schibler L, Jammes H, Jaffrézic F, Kiefer H. Predicting male fertility from the sperm methylome: application to 120 bulls with hundreds of artificial insemination records. Clin Epigenetics 2022; 14:54. [PMID: 35477426 PMCID: PMC9047354 DOI: 10.1186/s13148-022-01275-x] [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: 12/23/2021] [Accepted: 04/08/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Conflicting results regarding alterations to sperm DNA methylation in cases of spermatogenesis defects, male infertility and poor developmental outcomes have been reported in humans. Bulls used for artificial insemination represent a relevant model in this field, as the broad dissemination of bull semen considerably alleviates confounding factors and enables the precise assessment of male fertility. This study was therefore designed to assess the potential for sperm DNA methylation to predict bull fertility. RESULTS A unique collection of 100 sperm samples was constituted by pooling 2-5 ejaculates per bull from 100 Montbéliarde bulls of comparable ages, assessed as fertile (n = 57) or subfertile (n = 43) based on non-return rates 56 days after insemination. The DNA methylation profiles of these samples were obtained using reduced representation bisulfite sequencing. After excluding putative sequence polymorphisms, 490 fertility-related differentially methylated cytosines (DMCs) were identified, most of which were hypermethylated in subfertile bulls. Interestingly, 46 genes targeted by DMCs are involved in embryonic and fetal development, sperm function and maturation, or have been related to fertility in genome-wide association studies; five of these were further analyzed by pyrosequencing. In order to evaluate the prognostic value of fertility-related DMCs, the sperm samples were split between training (n = 67) and testing (n = 33) sets. Using a Random Forest approach, a predictive model was built from the methylation values obtained on the training set. The predictive accuracy of this model was 72% on the testing set and 72% on individual ejaculates collected from an independent cohort of 20 bulls. CONCLUSION This study, conducted on the largest set of bull sperm samples so far examined in epigenetic analyses, demonstrated that the sperm methylome is a valuable source of male fertility biomarkers. The next challenge is to combine these results with other data on the same sperm samples in order to improve the quality of the model and better understand the interplay between DNA methylation and other molecular features in the regulation of fertility. This research may have potential applications in human medicine, where infertility affects the interaction between a male and a female, thus making it difficult to isolate the male factor.
Collapse
Affiliation(s)
- Valentin Costes
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France.,R&D Department, ALLICE, 149 rue de Bercy, 75012, Paris, France.,Université Paris-Saclay, AgroParisTech, INRAE, GABI, 78350, Jouy-en-Josas, France
| | - Aurélie Chaulot-Talmon
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Eli Sellem
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France.,R&D Department, ALLICE, 149 rue de Bercy, 75012, Paris, France
| | - Jean-Philippe Perrier
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Anne Aubert-Frambourg
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Luc Jouneau
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Charline Pontlevoy
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Chris Hozé
- R&D Department, ALLICE, 149 rue de Bercy, 75012, Paris, France.,Université Paris-Saclay, AgroParisTech, INRAE, GABI, 78350, Jouy-en-Josas, France
| | - Sébastien Fritz
- R&D Department, ALLICE, 149 rue de Bercy, 75012, Paris, France.,Université Paris-Saclay, AgroParisTech, INRAE, GABI, 78350, Jouy-en-Josas, France
| | - Mekki Boussaha
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, 78350, Jouy-en-Josas, France
| | | | - Marie-Pierre Sanchez
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, 78350, Jouy-en-Josas, France
| | - Didier Boichard
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, 78350, Jouy-en-Josas, France
| | | | - Hélène Jammes
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Florence Jaffrézic
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, 78350, Jouy-en-Josas, France
| | - Hélène Kiefer
- INRAE, BREED, Université Paris-Saclay, UVSQ, 78350, Jouy-en-Josas, France. .,Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France.
| |
Collapse
|
35
|
Bufi R, Korstanje R. The impact of genetic background on mouse models of kidney disease. Kidney Int 2022; 102:38-44. [DOI: 10.1016/j.kint.2022.03.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022]
|
36
|
Prell A, Sen MO, Potabattula R, Bernhardt L, Dittrich M, Hahn T, Schorsch M, Zacchini F, Ptak GE, Niemann H, Haaf T. Species-Specific Paternal Age Effects and Sperm Methylation Levels of Developmentally Important Genes. Cells 2022; 11:cells11040731. [PMID: 35203380 PMCID: PMC8870257 DOI: 10.3390/cells11040731] [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: 12/15/2021] [Revised: 01/11/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
A growing number of sperm methylome analyses have identified genomic loci that are susceptible to paternal age effects in a variety of mammalian species, including human, bovine, and mouse. However, there is little overlap between different data sets. Here, we studied whether or not paternal age effects on the sperm epigenome have been conserved in mammalian evolution and compared methylation patterns of orthologous regulatory regions (mainly gene promoters) containing both conserved and non-conserved CpG sites in 94 human, 36 bovine, and 94 mouse sperm samples, using bisulfite pyrosequencing. We discovered three (NFKB2, RASGEF1C, and RPL6) age-related differentially methylated regions (ageDMRs) in humans, four (CHD7, HDAC11, PAK1, and PTK2B) in bovines, and three (Def6, Nrxn2, and Tbx19) in mice. Remarkably, the identified sperm ageDMRs were all species-specific. Most ageDMRs were in genomic regions with medium methylation levels and large methylation variation. Orthologous regions in species not showing this age effect were either hypermethylated (>80%) or hypomethylated (<20%). In humans and mice, ageDMRs lost methylation, whereas bovine ageDMRs gained methylation with age. Our results are in line with the hypothesis that sperm ageDMRs are in regions under epigenomic evolution and may be part of an epigenetic mechanism(s) for lineage-specific environmental adaptations and provide a solid basis for studies on downstream effects in the genes analyzed here.
Collapse
Affiliation(s)
- Andreas Prell
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany; (A.P.); (M.O.S.); (R.P.); (L.B.); (M.D.)
| | - Mustafa Orkun Sen
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany; (A.P.); (M.O.S.); (R.P.); (L.B.); (M.D.)
| | - Ramya Potabattula
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany; (A.P.); (M.O.S.); (R.P.); (L.B.); (M.D.)
| | - Laura Bernhardt
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany; (A.P.); (M.O.S.); (R.P.); (L.B.); (M.D.)
| | - Marcus Dittrich
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany; (A.P.); (M.O.S.); (R.P.); (L.B.); (M.D.)
- Department of Bioinformatics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Thomas Hahn
- Fertility Center, 65189 Wiesbaden, Germany; (T.H.); (M.S.)
| | | | - Federica Zacchini
- PERCUROS BV, 2333 CL Leiden, The Netherlands;
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
- Wolfson Centre for Age-Related Diseases, King’s College London, London SE1 1UL, UK
| | - Grazyna Ewa Ptak
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Heiner Niemann
- Clinic for Gastroenterology, Hepatology and Endocrinology, Medical University Hannover, 30625 Hannover, Germany;
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany; (A.P.); (M.O.S.); (R.P.); (L.B.); (M.D.)
- Correspondence: ; Tel.: +49-931-3188738
| |
Collapse
|
37
|
Mao Y, Yiran Z, Sisi L, Huixi C, Xia L, Ting W, Guolian D, Xinmei L, Sheng J, Meng Y, Huang H. Advanced paternal age increased metabolic risks in mice offspring. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166355. [DOI: 10.1016/j.bbadis.2022.166355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/07/2022] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
|
38
|
D Palmer R, Papa V, Vaccarezza M. The Ability of Nutrition to Mitigate Epigenetic Drift: A Novel Look at Regulating Gene Expression. J Nutr Sci Vitaminol (Tokyo) 2022; 67:359-365. [PMID: 34980713 DOI: 10.3177/jnsv.67.359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Epigenetic drift causes modification in gene expression during aging and a myriad of physiological changes that are mostly undesirable, remove youthful phenotype and are related to biological decay and disease onset. The epigenome is considered a stable regulator of genetic expression. Moreover, evidence is now accumulating that commonly available compounds found in foods can influence the epigenome to embrace a more youthful and therefore, more disease resistant state. Here we explore the correlation between nutriment and the epigenetic regulation through various types of alimentation. The aim is not to discuss specific chemicals involved in disease onset. Instead, we offer a brief glance at pathogens and offer a practical pathway into epigenetic regulation, hypothesizing that epigenetic drift might be attenuated by several foods able to drive a more youthful and disease resistant phenotype.
Collapse
Affiliation(s)
| | - Veronica Papa
- Department of Motor Sciences and Wellness, University of Naples "Parthenope".,FABAP Research Center
| | - Mauro Vaccarezza
- Curtin Medical School, Faculty of Health Sciences, Curtin Health Innovation Research Institute (CHIRI), Curtin University.,Department of Translational Medicine, University of Ferrara
| |
Collapse
|
39
|
Liu X, Lou H, Zhang J, Du M, Du Y, Wu S, Guan Y, Liu J. Clinical outcome analysis of frozen-thawed embryo transfer on Day 7. Front Endocrinol (Lausanne) 2022; 13:1082597. [PMID: 36568064 PMCID: PMC9780262 DOI: 10.3389/fendo.2022.1082597] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To investigate the clinical outcomes of Day 7 (D7) frozen-thawed embryo transfer (FET) and to provide a reference value for clinical work. METHODS This was a retrospective cohort study. Patients undergoing FET cycles in the Reproductive Medicine Center of the Third Affiliated Hospital of Zhengzhou University between December 2015 and January 2021 were included. According to the developmental stage of the embryos at transfer, the embryos were divided into three groups: Day (D) 5, D6 and D7 blastocysts. Group D7 was compared with Groups D5 and D6. Simultaneously, the preimplantation genetic testing (PGT) and non-PGT cycles in Group D7 were analyzed and compared. The main outcomes were the clinical pregnancy, live birth and miscarriage rates. The secondary outcomes were the implantation and euploidy rates. RESULTS In total, 5945, 4094 and 137 FET cycles were included in the D5, D6 and D7 groups, respectively. The clinical pregnancy rate was significantly lower in Group D7 than in Groups D5 (13.9% vs 62.9%, P <0.001) and D6 (13.9% vs 51.4%, P <0.001). Additionally, the live birth rate was significantly lower in Group D7 than in Groups D5 (7.3% vs 50.7%, P <0.001) and D6 (7.3% vs 40.5%, P <0.001). However, the miscarriage rate was significantly higher in Group D7 than in Groups D5 (47.4% vs 18.2%, P =0.001) and D6 (47.4% vs 20.6%, P =0.004). The clinical pregnancy and live birth rates for D7 blastocysts were significantly higher in the PGT group than in the non-PGT group (41.7% vs 13.9%, P=0.012; 33.3% vs 7.3%, P =0.003). CONCLUSIONS D7 blastocyst transfer can yield a live birth rate that is lower than that for D5 and D6 blastocysts but has value for transfer. PGT for D7 blastocysts may reduce the number of ineffective transfers and improve the outcome of D7 blastocyst transfer, which can be performed according to a patient's situation.
Collapse
|
40
|
Kemoun P, Ader I, Planat-Benard V, Dray C, Fazilleau N, Monsarrat P, Cousin B, Paupert J, Ousset M, Lorsignol A, Raymond-Letron I, Vellas B, Valet P, Kirkwood T, Beard J, Pénicaud L, Casteilla L. A gerophysiology perspective on healthy ageing. Ageing Res Rev 2022; 73:101537. [PMID: 34883201 DOI: 10.1016/j.arr.2021.101537] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/23/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022]
Abstract
Improvements in public health and health care have resulted in significant increases in lifespan globally, but also in a significant increase in chronic disease prevalence. This has led to a focus on healthy ageing bringing a shift from a pathology-centered to an intrinsic capacity and function-centered view. In parallel, the emerging field of geroscience has promoted the exploration of the biomolecular drivers of ageing towards a transverse vision by proposing an integrated set of molecular hallmarks. In this review, we propose to take a step further in this direction, highlighting a gerophysiological perspective that considers the notion of homeostasis/allostasis relating to robustness/fragility respectively. While robustness is associated with homeostasis achieved by an optimal structure/function relationship in all organs, successive repair processes occurring after daily injuries and infections result in accumulation of scar healing leading to progressive tissue degeneration, allostasis and frailty. Considering biological ageing as the accumulation of scarring at the level of the whole organism emphasizes three transverse and shared elements in the body - mesenchymal stroma cells/immunity/metabolism (SIM). This SIM tryptich drives tissue and organ fate to regulate the age-related evolution of body functions. It provides the basis of a gerophysiology perspective, possibly representing a better way to decipher healthy ageing, not only by defining a composite biomarker(s) but also by developing new preventive/curative strategies.
Collapse
|
41
|
Affiliation(s)
- Ying Zhang
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, Québec, Canada
| | - Marc-André Sirard
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, Québec, Canada
| |
Collapse
|
42
|
Zacchini F, Sampino S, Zietek M, Chan A. Delayed parenthood and its influence on offspring health: What have we learned from the mouse model. Biol Reprod 2021; 106:58-65. [PMID: 34725675 DOI: 10.1093/biolre/ioab202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/18/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
Delayed parenthood is constantly increasing worldwide due to various socio-economic factors. In the last decade, a growing number of epidemiological studies have suggested a link between advanced parental age and an increased risk of diseases in the offspring. Also, poor reproductive outcome has been described in pregnancies conceived by aged parents. Similarly, animal studies showed that aging negatively affects gametes, early embryonic development, pregnancy progression and the postnatal phenotype of resulting offspring. However, how and to what extent parental age is a risk factor for the health of future generations is still subject to debate. Notwithstanding the limitation of an animal model, the mouse model represents a useful tool to understand not only the influence of parental age on offspring phenotype but also the biological mechanisms underlying the poor reproductive outcome and the occurrence of diseases in the descendants. The present review aims at i) providing an overview of the current knowledge from mouse model about the risks associated with conception at advanced age (e.g. neurodevelopmental and metabolic disorders), ii) highlighting the candidate biological mechanisms underlying this phenomenon, and iii) discussing on how murine-derived data can be relevant to humans.
Collapse
Affiliation(s)
- Federica Zacchini
- Percuros BV, Leiden, The Netherlands.,Wolfson Center for Age Related Diseases, King's College London, London, United Kingdom
| | - Silvestre Sampino
- Department of Experimental Embryology, IGAB PAS, Jastrzebiec, Poland
| | - Marta Zietek
- Department of Experimental Embryology, IGAB PAS, Jastrzebiec, Poland
| | - Alan Chan
- Percuros BV, Leiden, The Netherlands
| |
Collapse
|
43
|
Van Opstal J, Fieuws S, Spiessens C, Soubry A. Male age interferes with embryo growth in IVF treatment. Hum Reprod 2021; 36:107-115. [PMID: 33164068 DOI: 10.1093/humrep/deaa256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 09/02/2020] [Indexed: 12/23/2022] Open
Abstract
STUDY QUESTION Does male age affect embryo growth or quality in couples undergoing IVF treatment? SUMMARY ANSWER Advanced paternal age (APA) is negatively associated with the chance of an optimal eight-cell embryo on the third day of development. WHAT IS KNOWN ALREADY Literature shows that APA is associated with decreased sperm quality and fecundity. However, the effect of male age on embryo growth in an IVF setting remains inconclusive. Literature concerning male influences on IVF success is scarce and approaches used to analyse embryo outcomes differ by study. STUDY DESIGN, SIZE, DURATION This study was part of the longitudinal Epigenetic Legacy of Paternal Obesity (ELPO) study for which fathers and mothers were followed from pre-pregnancy until the birth of their child. Couples were recruited from April 2015 to September 2017. A total of 1057 embryos from 87 couples were studied. PARTICIPANTS/MATERIALS, SETTING, METHODS Dutch-speaking couples planning to undergo an IVF treatment were recruited at the Leuven University Fertility Center in Flanders, Belgium. Anthropometrics were documented and compared to the general Flemish population. Semen characteristics, pregnancy rates and the following embryo characteristics were recorded: number of blastomeres, symmetry and percentage fragmentation. Statistical modelling was applied taking into account correlation of within-cycle outcomes and use of multiple cycles per couple. MAIN RESULTS AND THE ROLE OF CHANCE We observed a significant inverse association between APA and a key determinant for scoring of embryo quality: older men were less likely to produce an embryo of eight blastomeres at Day 3, compared to younger fathers; odds ratio for the effect of 1 year equals 0.960 (95% CI: 0.930-0.991; P = 0.011). Our finding remained significant after adjusting for female age and male and female BMI. Degree of fragmentation and symmetry were not significantly related to male age. LIMITATIONS, REASONS FOR CAUTION Because of the study's small sample size and its monocentric nature, a larger study is warranted to confirm our results. In addition, distribution of BMI and level of education were not representative of the general Flemish population. Although we corrected for BMI status, we do not exclude that obesity may be one of the determinants of infertility in our study population. Furthermore, it is known from other European countries that a higher education eases access to fertility treatment. Hence, caution should be taken when interpreting our findings from a fertility setting to the general population. WIDER IMPLICATIONS OF THE FINDINGS We suggest a heightened need for future research into male age and its potential effects on embryo growth, embryo quality and ART outcomes. Clinical decision-making and preventative public health programmes would benefit from a better understanding of the role of men, carried forward by the Paternal Origins of Health and Disease (POHaD) paradigm. We hope the current finding will encourage others to examine the role of the sperm epigenome in embryo development according to paternal age. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by a research grant from KU Leuven University (OT/14/109). The authors declare no competing financial, professional or personal interests. TRIAL REGISTRATION NUMBER KU Leuven S57378 (ML11309), B322201523225.
Collapse
Affiliation(s)
- Jolien Van Opstal
- Epidemiology Research Center, Department of Public Health and Primary Care, Faculty of Medicine, KU Leuven - University of Leuven, Leuven 3000, Belgium
| | - Steffen Fieuws
- L-Biostat, Department of Public Health and Primary Care, Faculty of Medicine, KU Leuven - University of Leuven, Leuven 3000, Belgium
| | - Carl Spiessens
- Leuven University Fertility Clinic, KU Leuven - University of Leuven, Leuven 3000, Belgium
| | - Adelheid Soubry
- Epidemiology Research Center, Department of Public Health and Primary Care, Faculty of Medicine, KU Leuven - University of Leuven, Leuven 3000, Belgium
| |
Collapse
|
44
|
Vuarin P, Lesobre L, Levêque G, Saint Jalme M, Lacroix F, Hingrat Y, Sorci G. Paternal age negatively affects sperm production of the progeny. Ecol Lett 2021; 24:719-727. [PMID: 33565248 DOI: 10.1111/ele.13696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 01/02/2023]
Abstract
Parental age has profound consequences for offspring's phenotype. However, whether patrilineal age affects offspring sperm production remains unknown, despite the importance of sperm production for male reproductive success in species facing post-copulatory sexual selection. Using a longitudinal dataset on ejaculate attributes of the houbara bustard, we showed that offspring sired by old fathers had different age-dependent trajectories of sperm production compared to offspring sired by young fathers. Specifically, they produced less sperm (-48%) in their first year of life, and 14% less during their lifetime. Paternal age had the strongest effect, with weak evidence for grandpaternal or great grandpaternal age effects. These results show that paternal age can affect offspring reproductive success by reducing sperm production, establishing an intergenerational link between ageing and sexual selection.
Collapse
Affiliation(s)
- Pauline Vuarin
- Reneco International Wildlife Consultants LLC, Abu Dhabi, PoBox 61741, United Arab Emirates.,Biogéosciences, UMR 6282 CNRS, Université de Bourgogne Franche-Comté, 6 boulevard Gabriel, Dijon, 21000, France
| | - Loïc Lesobre
- Reneco International Wildlife Consultants LLC, Abu Dhabi, PoBox 61741, United Arab Emirates
| | - Gwènaëlle Levêque
- Emirates Center for Wildlife Propagation, BP 47, route de Midelt, Missour, 33250, Morocco
| | - Michel Saint Jalme
- Centre d'Ecologie et des Sciences de la Conservation, UMR 7204 MNHN CNRS-UPMC, Museum National d'Histoire Naturelle, 43 et 61 rue Buffon, Paris, 75005, France
| | - Frédéric Lacroix
- Reneco International Wildlife Consultants LLC, Abu Dhabi, PoBox 61741, United Arab Emirates
| | - Yves Hingrat
- Reneco International Wildlife Consultants LLC, Abu Dhabi, PoBox 61741, United Arab Emirates
| | - Gabriele Sorci
- Biogéosciences, UMR 6282 CNRS, Université de Bourgogne Franche-Comté, 6 boulevard Gabriel, Dijon, 21000, France
| |
Collapse
|
45
|
Li S, Li J, Pan R, Cheng J, Cui Q, Chen J, Yuan Z. Sodium rutin extends lifespan and health span in mice including positive impacts on liver health. Br J Pharmacol 2021; 179:1825-1838. [PMID: 33555034 DOI: 10.1111/bph.15410] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/15/2021] [Accepted: 02/01/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Ageing is associated with progressive metabolic dysregulation. Rutin is a metabolic regulator with a poor solubility. Using soluble sodium rutin we investigating the effect and mechanisms of rutin in ageing process. EXPERIMENTAL APPROACH Wild type male mice were treated with or without sodium rutin ( 0.2 mg·ml-1 in drinking water from 8-month-old until end of life. Kaplan-Meier survival curve was used for lifespan assay, ageing-related histopathology analysis and metabolic analysis were performed to determine the effects of chronic sodium rutin on the longevity. Serological test, liver tissue metabolomics and transcriptomics were used for liver function assay. SiRNA knockdown Angptl8 and autophagy flux assay in HepG2 cell lines explored the mechanism through which sodium rutin might impact the function of hepatocyte. KEY RESULTS Sodium rutin treatment extends the lifespan of mice by 10%. Sodium rutin supplementation alleviates ageing-related pathological changes and promotes behaviour performance in ageing mice. Sodium rutin supplementation altered the whole-body metabolism in mice, which exhibited increased energy expenditure and lower respiratory quotient. Transcriptomics analysis showed that Sodium rutin affected the expression of metabolic genes. Metabolomics analysis showed that Sodium rutin reduced liver steatosis through increased lipid β-oxidation. Sodium rutin treatment increased the autophagy level both in vivo and in vitro. The inhibition of autophagy partially abolished the sodium rutin-mediated effect on lipolysis in HepG2 cells. CONCLUSION AND IMPLICATIONS Sodium rutin treatment extends the lifespan and health span of mice with beneficial effects on metabolism, which were achieved by enhancing the autophagy activity in hepatocytes.
Collapse
Affiliation(s)
- Shuoshuo Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jun Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Ruiyuan Pan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jinbo Cheng
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China.,Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
| | - Qinghua Cui
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jianxin Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
| |
Collapse
|
46
|
Oluwayiose OA, Wu H, Saddiki H, Whitcomb BW, Balzer LB, Brandon N, Suvorov A, Tayyab R, Sites CK, Hill L, Marcho C, Pilsner JR. Sperm DNA methylation mediates the association of male age on reproductive outcomes among couples undergoing infertility treatment. Sci Rep 2021; 11:3216. [PMID: 33547328 PMCID: PMC7864951 DOI: 10.1038/s41598-020-80857-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Parental age at time of offspring conception is increasing in developed countries. Advanced male age is associated with decreased reproductive success and increased risk of adverse neurodevelopmental outcomes in offspring. Mechanisms for these male age effects remain unclear, but changes in sperm DNA methylation over time is one potential explanation. We assessed genome-wide methylation of sperm DNA from 47 semen samples collected from male participants of couples seeking infertility treatment. We report that higher male age was associated with lower likelihood of fertilization and live birth, and poor embryo development (p < 0.05). Furthermore, our multivariable linear models showed male age was associated with alterations in sperm methylation at 1698 CpGs and 1146 regions (q < 0.05), which were associated with > 750 genes enriched in embryonic development, behavior and neurodevelopment among others. High dimensional mediation analyses identified four genes (DEFB126, TPI1P3, PLCH2 and DLGAP2) with age-related sperm differential methylation that accounted for 64% (95% CI 0.42-0.86%; p < 0.05) of the effect of male age on lower fertilization rate. Our findings from this modest IVF population provide evidence for sperm methylation as a mechanism of age-induced poor reproductive outcomes and identifies possible candidate genes for mediating these effects.
Collapse
Affiliation(s)
- Oladele A Oluwayiose
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, 173A Goessmann, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - Haotian Wu
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, 722 W 168th St, New York, NY, 10032, USA
| | - Hachem Saddiki
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, 715 North Pleasant Street, Amherst, MA, USA
| | - Brian W Whitcomb
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, 715 North Pleasant Street, Amherst, MA, USA
| | - Laura B Balzer
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, 715 North Pleasant Street, Amherst, MA, USA
| | - Nicole Brandon
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, 173A Goessmann, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - Alexander Suvorov
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, 173A Goessmann, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - Rahil Tayyab
- Division of Reproductive Endocrinology and Infertility, Baystate Medical Center, 759 Chestnut Street, Springfield, MA, USA
| | - Cynthia K Sites
- Division of Reproductive Endocrinology and Infertility, Baystate Medical Center, 759 Chestnut Street, Springfield, MA, USA
| | - Lisa Hill
- Division of Reproductive Endocrinology and Infertility, Baystate Medical Center, 759 Chestnut Street, Springfield, MA, USA
| | - Chelsea Marcho
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, 173A Goessmann, 686 North Pleasant Street, Amherst, MA, 01003, USA
| | - J Richard Pilsner
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, 173A Goessmann, 686 North Pleasant Street, Amherst, MA, 01003, USA.
| |
Collapse
|
47
|
Pilsner JR, Shershebnev A, Wu H, Marcho C, Dribnokhodova O, Shtratnikova V, Sergeyev O, Suvorov A. Aging-induced changes in sperm DNA methylation are modified by low dose of perinatal flame retardants. Epigenomics 2021; 13:285-297. [PMID: 33401928 DOI: 10.2217/epi-2020-0404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aims: Paternal age is increasing in developed countries. Understanding of aging-related epigenetic changes in sperm is needed as well as factors that modify such changes. Materials & methods: Young pubertal and mature rats were exposed perinatally to vehicle or environmental xenobiotic 2,2',4,4'-tetrabromodiphenyl ether. Epididymal sperm was reduced representation bisulfite sequenced. Differentially methylated regions (DMRs) were identified via MethPipe. Results: In control animals, 5319 age-dependent DMRs were identified. Age-related DMRs were enriched for embryonic development. In exposed rats, DNA methylation was higher in young and lower in mature animals then in controls. Conclusions: Sperm methylome undergoes significant age-dependent changes, which may represent a causal link between paternal age and offspring phenotype. Environmental xenobiotics can interfere with the natural process of epigenetic aging.
Collapse
Affiliation(s)
- J Richard Pilsner
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA 01003, USA
| | - Alex Shershebnev
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA 01003, USA
| | - Haotian Wu
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA 01003, USA
| | - Chelsea Marcho
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA 01003, USA
| | | | | | | | - Alexander Suvorov
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA 01003, USA.,Federal Budget Institution of Science Central Research Institute of Epidemiology of The Federal Service on Customers' Rights Protection & Human Well-being Surveillance, Novogireevskaya .3a, Moscow 111123, Russia
| |
Collapse
|
48
|
Yoshizaki K, Kimura R, Kobayashi H, Oki S, Kikkawa T, Mai L, Koike K, Mochizuki K, Inada H, Matsui Y, Kono T, Osumi N. Paternal age affects offspring via an epigenetic mechanism involving REST/NRSF. EMBO Rep 2021; 22:e51524. [PMID: 33399271 PMCID: PMC7857438 DOI: 10.15252/embr.202051524] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Advanced paternal age can have deleterious effects on various traits in the next generation. Here, we establish a paternal‐aging model in mice to understand the molecular mechanisms of transgenerational epigenetics. Whole‐genome target DNA methylome analyses of sperm from aged mice reveal more hypo‐methylated genomic regions enriched in REST/NRSF binding motifs. Gene set enrichment analyses also reveal the upregulation of REST/NRSF target genes in the forebrain of embryos from aged fathers. Offspring derived from young mice administrated with a DNA de‐methylation drug phenocopy the abnormal vocal communication of pups derived from aged fathers. In conclusion, hypo‐methylation of sperm DNA can be a key molecular feature modulating neurodevelopmental programs in offspring by causing fluctuations in the expression of REST/NRSF target genes.
Collapse
Affiliation(s)
- Kaichi Yoshizaki
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Disease Model, Aichi Developmental Disability Center, Aichi, Japan
| | - Ryuichi Kimura
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisato Kobayashi
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo, Japan.,Department of Embryology, Nara Medical University, Nara, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Shinya Oki
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takako Kikkawa
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Lingling Mai
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kohei Koike
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Physiology, Center for Integrative Physiology and Molecular Medicine, Saarland University School of Medicine, Homburg, Germany
| | - Kentaro Mochizuki
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.,Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hitoshi Inada
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.,Laboratory of Health and Sports Science, Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Yasuhisa Matsui
- The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.,Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Tomohiro Kono
- The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.,Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
49
|
Vasilyev SA, Tolmacheva EN, Vasilyeva OY, Markov AV, Zhigalina DI, Zatula LA, Lee VA, Serdyukova ES, Sazhenova EA, Nikitina TV, Kashevarova AA, Lebedev IN. LINE-1 retrotransposon methylation in chorionic villi of first trimester miscarriages with aneuploidy. J Assist Reprod Genet 2021; 38:139-149. [PMID: 33170392 PMCID: PMC7823001 DOI: 10.1007/s10815-020-02003-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/02/2020] [Indexed: 01/15/2023] Open
Abstract
PURPOSE High frequency of aneuploidy in meiosis and cleavage stage coincides with waves of epigenetic genome reprogramming that may indicate a possible association between epigenetic mechanisms and aneuploidy occurrence. This study aimed to assess the methylation level of the long interspersed repeat element 1 (LINE-1) retrotransposon in chorionic villi of first trimester miscarriages with a normal karyotype and aneuploidy. METHODS The methylation level was assessed at 19 LINE-1 promoter CpG sites in chorionic villi of 141 miscarriages with trisomy of chromosomes 2, 6, 8-10, 13-15, 16, 18, 20-22, and monosomy X using massive parallel sequencing. RESULTS The LINE-1 methylation level was elevated statistically significant in chorionic villi of miscarriages with both trisomy (45.2 ± 4.3%) and monosomy X (46.9 ± 4.2%) compared with that in induced abortions (40.0 ± 2.4%) (p < 0.00001). The LINE-1 methylation levels were specific for miscarriages with different aneuploidies and significantly increased in miscarriages with trisomies 8, 14, and 18 and monosomy X (p < 0.05). The LINE-1 methylation level increased with gestational age both for group of miscarriages regardless of karyotype (R = 0.21, p = 0.012) and specifically for miscarriages with trisomy 16 (R = 0.48, p = 0.007). LINE-1 methylation decreased with maternal age in miscarriages with a normal karyotype (R = - 0.31, p = 0.029) and with trisomy 21 (R = - 0.64, p = 0.024) and increased with paternal age for miscarriages with trisomy 16 (R = 0.38, p = 0.048) and monosomy X (R = 0.73, p = 0.003). CONCLUSION Our results indicate that the pathogenic effects of aneuploidy in human embryogenesis can be supplemented with significant epigenetic changes in the repetitive sequences.
Collapse
Affiliation(s)
- Stanislav A Vasilyev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia.
- Biological Institute, National Research Tomsk State University, Lenina, 36, Tomsk, Russia.
| | - Ekaterina N Tolmacheva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
| | - Oksana Yu Vasilyeva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
| | - Anton V Markov
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
| | - Daria I Zhigalina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
| | - Lada A Zatula
- Department of Medical Genetics, Siberian State Medical University, Moskovskiy Trakt, 2, Tomsk, Russia
| | - Vasilissa A Lee
- Department of Medical Genetics, Siberian State Medical University, Moskovskiy Trakt, 2, Tomsk, Russia
| | - Ekaterina S Serdyukova
- Biological Institute, National Research Tomsk State University, Lenina, 36, Tomsk, Russia
| | - Elena A Sazhenova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
| | - Tatyana V Nikitina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
| | - Anna A Kashevarova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
| | - Igor N Lebedev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Nab. R. Ushaiki, 10, Tomsk, Russia
- Department of Medical Genetics, Siberian State Medical University, Moskovskiy Trakt, 2, Tomsk, Russia
| |
Collapse
|
50
|
Mao Y, Zhang C, Wang Y, Meng Y, Chen L, Dennis CL, Sheng J, Wu Y, Huang H. Association Between Paternal Age and Birth Weight in Preterm and Full-Term Birth: A Retrospective Study. Front Endocrinol (Lausanne) 2021; 12:706369. [PMID: 34367073 PMCID: PMC8341720 DOI: 10.3389/fendo.2021.706369] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/29/2021] [Indexed: 01/24/2023] Open
Abstract
PURPOSE While it is well documented that maternal adverse exposures contribute to a series defects on offspring health according to the Developmental Origins of Health and Disease (DOHaD) theory, paternal evidence is still insufficient. Advanced paternal age is associated with multiple metabolism and psychiatric disorders. Birth weight is the most direct marker to evaluate fetal growth. Therefore, we designed this study to explore the association between paternal age and birth weight among infants born at term and preterm (<37 weeks gestation). METHODS A large retrospective study was conducted using population-based hospital data from January 2015 to December 2019 that included 69,964 cases of singleton infant births with complete paternal age data. The primary outcome was infant birth weight stratified by sex and gestational age including small for gestational age (SGA, 10th percentile) and large for gestational age (LGA, 90th percentile). Birth weight percentiles by gestational age were based on those published in the INTERGROWTH-21st neonatal weight-for gestational-age standard. Logistic regression analysis and linear regression model were used to estimate the association between paternal age and infant birth weight. RESULTS Advanced paternal age was associated with a higher risk for a preterm birth [35-44 years: adjusted odds ratio (OR) = 1.13, 95%CI (1.03 to 1.24); >44 years: OR = 1.36, 95%CI (1.09 to 1.70)]. Paternal age exerted an opposite effect on birth weight with an increased risk of SGA among preterm infants (35-44years: OR = 1.85, 95%CI (1.18 to 2.89) and a decreased risk among term infant (35-44years: OR = 0.81, 95%CI (0.68 to 0.98); >44 years: OR = 0.50, 95%CI (0.26 to 0.94). U-shaped associations were found in that LGA risk among term infants was higher in both younger (<25 years) (OR = 1.32; 95%CI, 1.07 to 1.62) and older (35-44 years) (OR = 1.07; 95% CI, 1.01 to 1.14) fathers in comparison to those who were 25 to 34 years old at the time of delivery. CONCLUSIONS Our study found advanced paternal age increased the risk of SGA among preterm infants and for LGA among term infants. These findings likely reflect a pathophysiology etiology and have important preconception care implications and suggest the need for antenatal monitoring.
Collapse
Affiliation(s)
- Yiting Mao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Chen Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yinyu Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yicong Meng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Lei Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cindy-Lee Dennis
- Bloomberg Faculty of Nursing, University of Toronto, Toronto, ON, Canada
| | - Jianzhong Sheng
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanting Wu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Hefeng Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|