1
|
Shi Y, Li W, Yu X, Zhao Y, Zhu D, Song Y, Zhao Z, Gu Y, Wei B, Li L, Yu D, Zhang P, Gao Q, Sun M. Paternal Obesity-Induced H3K27me3 Elevation Leads to MANF-Mediated Transgenerational Metabolic Dysfunction in Female Offspring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415956. [PMID: 40041941 DOI: 10.1002/advs.202415956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/20/2025] [Indexed: 04/26/2025]
Abstract
Paternal lifestyle and environmental exposures can alter epigenetic changes in sperm and play a critical role in the offspring's future health, yet the underlying mechanisms remain elusive. The present study established a model of paternal obesity and found that the increased levels of H3K27me3 in sperm persist into the 8-cell embryo stage, resulting in a transgenerational decrease of Manf, which causes endoplasmic reticulum stress and activates the GRP78-PERK-EIF2α-ATF4-CHOP axis. This consequently leads to impaired glucose metabolism and apoptosis in the liver of female offspring. Based on these findings, the F0 mice are treated with 3-deazaneplanocin A, an EZH2 inhibitor, which successfully prevented metabolic dysfunction in F0 mice of the high-fat diet (HFD) group. Meanwhile, intravenous injection of recombinant human MANF in F1 female offspring can successfully rescue the metabolic dysfunction in the HFD-F1 group. These results demonstrate that paternal obesity triggers transgenerational metabolic dysfunction through sperm H3K27me3-dependent epigenetic regulation. The present study also identifies the H3K27me3-MANF pathway as a potentially preventive and therapeutic strategy for diabetes, although further studies are needed to validate its clinical applicability.
Collapse
Affiliation(s)
- Yajun Shi
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Weisheng Li
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
- Department of Gynecology, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), Shandong province, Qingdao, 266000, China
| | - Xi Yu
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Yan Zhao
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Dan Zhu
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Yueyang Song
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Zejun Zhao
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Yannan Gu
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Bin Wei
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Lingjun Li
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Dongyi Yu
- Center for Medical Genetics and Prenatal Diagnosis, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Shandong province, Jinan, 250000, China
| | - Pengjie Zhang
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Qinqin Gao
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
| | - Miao Sun
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, 215031, China
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| |
Collapse
|
2
|
Narula K, Kenkre JS, Loh WJ, Tan T. Obesity, insulin resistance and fertility: unresolved questions and emerging insights. Curr Opin Endocrinol Diabetes Obes 2025:01266029-990000000-00124. [PMID: 40125660 DOI: 10.1097/med.0000000000000907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
PURPOSE OF REVIEW Obesity significantly impacts fertility in women, contributing to hormonal imbalances, ovulatory dysfunction, and poor reproductive outcomes. This is especially pronounced in polycystic ovary syndrome (PCOS), where obesity and insulin resistance exacerbate fertility challenges. Moreover, obesity is a risk factor for type 2 diabetes (T2D) and gestational diabetes (GDM), further complicating reproductive health. Effective weight loss interventions before conception are essential to improve fertility and reduce the risks of adverse perinatal outcomes, such as GDM, hypertensive disorders, and neonatal complications. RECENT FINDINGS Lifestyle modifications, including modest calorie restriction and exercise, improve ovulatory function and pregnancy rates but have limited impact on live-birth rates during fertility treatments. Very low-calorie diets (VLCDs) achieve rapid weight loss but raise concerns about maternal nutrition. Pharmacotherapy offers modest benefits for weight loss and fertility, though teratogenic risks persist. Bariatric surgery often results in significant weight loss and enhanced fertility, yet requires careful timing and management of potential nutrient deficiencies. SUMMARY Weight-loss interventions show promise in addressing obesity-related fertility issues, but long-term outcomes and optimal strategies remain unclear. Further research is needed to bridge these gaps and improve reproductive outcomes following weight reduction.
Collapse
Affiliation(s)
| | - Julia S Kenkre
- Imperial College London Division of Experimental Medicine: Imperial College London Faculty of Medicine, London, UK
| | | | - Tricia Tan
- Imperial College London Division of Experimental Medicine: Imperial College London Faculty of Medicine, London, UK
| |
Collapse
|
3
|
Su L, Dreyfuss JM, Ferraz Bannitz R, Wolfs D, Hansbury G, Richardson L, Charmant C, Patel J, Ginsburg ES, Racowsky C, Fore R, Efthymiou V, Desmond J, Goldfine A, Ferguson-Smith A, Pan H, Hivert MF, Isganaitis E, Patti ME. Type 2 diabetes impacts DNA methylation in human sperm. Clin Epigenetics 2025; 17:49. [PMID: 40108650 PMCID: PMC11924665 DOI: 10.1186/s13148-025-01853-9] [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: 05/05/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
AIMS/HYPOTHESIS Disorders of the reproductive system, including hypogonadism and reduced fertility, are an under-recognized complication of diabetes. Based on experimental data in mice, hyperglycemia and obesity may modify epigenetic marks in sperm and impact health and development of offspring, but data are more limited in humans. Thus, we sought to study the impact of type 2 diabetes and glycemic control on sperm quality and DNA methylation. METHODS In this prospective cohort study, we recruited 40 men with BMI greater than 25 kg/m2 including 18 with type 2 diabetes, 6 with prediabetes, and 16 normoglycemic controls. Assessments were repeated after 3 months in 9 men with type 2 diabetes and 7 controls. We analyzed reproductive hormones, sperm concentration and motility, and sperm DNA methylation (MethylationEPIC BeadChip). RESULTS Men with type 2 diabetes had higher levels of follicle-stimulating hormone (FSH), but similar testosterone levels and sperm quality as controls. Sperm DNA methylation was stable with repeat sampling at 3 months in men with and without type 2 diabetes. We identified differential methylation at 655 of 745,804 CpG sites in men with type 2 diabetes versus controls (FDR < 0.05). Of these, 96.5% showed higher methylation in type 2 diabetes, with a mean difference in DNA methylation (beta value, β) of 0.16 ± 0.004 (16 ± 0.4%). Ontology analysis of differentially methylated loci revealed annotation to genes regulating synaptic signaling, actin, cAMP-dependent pathways, and G protein-coupled receptor pathways. 24% of probes differentially regulated in men with type 2 diabetes versus control overlapped with probes associated with HbA1c, suggesting additional factors beyond glycemic control contributed to diabetes-associated differences in DNA methylation. CONCLUSIONS/INTERPRETATION Men with type 2 diabetes showed higher DNA methylation levels in sperm relative to normoglycemic controls with similar BMI. Whether these differences are reversible with glucose-lowering treatment or may contribute to post-fertilization transcriptional regulation warrants further investigation. TRIAL REGISTRATION NCT03860558.
Collapse
Affiliation(s)
- Lei Su
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Research Division, Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
| | - Rafael Ferraz Bannitz
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
| | - Danielle Wolfs
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
| | - Georgia Hansbury
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
| | - Lauren Richardson
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
| | - Charnice Charmant
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
| | - Jay Patel
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Elizabeth S Ginsburg
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Catherine Racowsky
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Ruby Fore
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School , Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Vissarion Efthymiou
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
| | - Jessica Desmond
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
| | - Allison Goldfine
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA
- Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Hui Pan
- Bioinformatics and Biostatistics Core, Research Division, Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School , Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Elvira Isganaitis
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA.
| | - Mary Elizabeth Patti
- Research Division, Harvard Medical School, Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA.
| |
Collapse
|
4
|
Sutovsky P, Zigo M, Tirpak F, Oko R. Paternal contributions to mammalian zygote - Beyond sperm-oocyte fusion. Curr Top Dev Biol 2025; 162:387-446. [PMID: 40180516 DOI: 10.1016/bs.ctdb.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Contrary to a common misconception that the fertilizing spermatozoon acts solely as a vehicle for paternal genome delivery to the zygote, this chapter aims to illustrate how the male gamete makes other essential contributions , including the sperm borne-oocyte activation factors, centrosome components, and components of the sperm proteome and transcriptome that help to lay the foundation for pregnancy establishment and maintenance to term, and the newborn and adult health. Our inquiry starts immediately after sperm plasma membrane fusion with its oocyte counterpart, the oolemma. Parallel to and following sperm incorporation in the egg cytoplasm, some of the sperm structures (perinuclear theca) are dissolved and spent to induce development, others (nucleus, centriole) are transformed into zygotic structures enabling it, and yet others (mitochondrial and fibrous sheath, axonemal microtubules and outer dense fibers) are recycled as to not stand in its way. Noteworthy advances in this research include the identification of several sperm-borne oocyte activating factor candidates, the role of autophagy in the post-fertilization sperm mitochondrion degradation, new insight into zygotic centrosome origins and function, and the contributions of sperm-delivered RNA cargos to early embryo development. In concluding remarks, the unresolved issues, and clinical and biotechnological implications of sperm-vectored paternal inheritance are discussed.
Collapse
Affiliation(s)
- Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, United States.
| | - Michal Zigo
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| | - Filip Tirpak
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| | - Richard Oko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
5
|
Mehta P, Singh R. The composition of human sperm sncRNAome: a cross-country small RNA profiling. Reprod Biol Endocrinol 2025; 23:36. [PMID: 40050854 PMCID: PMC11883963 DOI: 10.1186/s12958-025-01358-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/07/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Over the last decade, numerous studies have implicated sperm-borne small non-coding RNAs (sncRNAs) in fertility and transgenerational inheritance. Spermatozoa contain a variety of small RNAs; however, inter-individual and inter-population variations in the human sperm sncRNA content (sncRNAome) have not yet been ascertained. METHODS We performed sncRNA sequencing in 54 normozoospermic proven fertile Indian donors. We also obtained a second semen sample from 13 donors and a third semen sample from eight donors and repeated sncRNA sequencing. To better understand sperm sncRNAome similarities and variations, sncRNA sequencing data for eligible Chinese (n = 87), US (n = 14), and Spanish (n = 2) normozoospermic (fertile or presumptive fertile) samples were downloaded and analyzed in a uniform manner. sncRNA data were compared within and across populations to identify similarities and differences. RESULTS In Indian samples, rsRNAs (13.71-78.76%), YsRNAs (0.64-76.53%) and tsRNAs (5.63-35.16%) constituted the major fraction and miRNAs, piRNAs, mt-tsRNAs, and other sncRNAs constituted the minor fraction. Across three other populations, rsRNAs (11-80%) and tsRNAs (10-60%) constituted the major fraction, and YsRNAs (0.62-4.28%), miRNAs (0.41-7.37%), piRNAs (1.37-4.36%), mt-tsRNAs (0.14-4.33%), and other sncRNAs constituted the minor fraction. Only 47 miRNAs were consistent across the Indian samples, and only 17 miRNAs were consistent across the four populations. Interestingly, all piRNAs detected in Indian samples were derived from the chromosome 15 piRNA cluster, which were also predominantly present in other populations. tRNA-Gly-GCC contributed approximately 50% of the tsRNA pool across all populations. The mt-tsRNAs also originated majorly from one mt-tRNA that differed across populations. Among the rsRNAs, the maximum number of reads belonged to 28S, followed by 18S, 5S, 5.8S, and 45S in decreasing order. Y4sRNAs were the most abundant YsRNAs, while the second most common contributor differed across populations. CONCLUSIONS The human sperm sncRNAome has a 'core component' that shows small variations and a 'peripheral component' that shows significant variations across individuals and populations. The availability of the normal human sperm sncRNAome would help delineate biologically meaningful variations from sample-to-sample natural/random variations.
Collapse
Affiliation(s)
- Poonam Mehta
- CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajender Singh
- CSIR-Central Drug Research Institute, Lucknow, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
6
|
Latham KE. Paternal Effects in Mammalian Reproduction: Functional, Environmental, and Clinical Relevance of Sperm Components in Early Embryos and Beyond. Mol Reprod Dev 2025; 92:e70020. [PMID: 40123230 PMCID: PMC11931271 DOI: 10.1002/mrd.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
In addition to widely recognized contributions of the paternal genome, centriole, and oocyte-activation factors, sperm deliver a wide range of macromolecules to the fertilized embryo. The impacts of these factors on the embryo, progeny, and even subsequent generations have become increasingly apparent, along with an understanding of an extensive potential for male health and environmental exposures to exert both immediate and long-term impacts on mammalian reproduction. Available data reveal that sperm factors interact with and regulate the actions of oocyte factors as well as exerting additional direct effects on the early embryo. This review provides a summary of the nature and mechanisms of paternal effects in early mammalian embryos, long-term effects in progeny, susceptibility of sperm components to diverse environmental factors, and potential approaches to mitigate adverse effects of such exposures.
Collapse
Affiliation(s)
- Keith E. Latham
- Department of Animal ScienceMichigan State UniversityEast LansingMichiganUSA
- Department of Obstetrics, Gynecology and Reproductive BiologyMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
7
|
Soubry A. Introducing artificial intelligence and sperm epigenetics in the fertility clinic: a novel foundation for diagnostics and prediction modelling. FRONTIERS IN REPRODUCTIVE HEALTH 2025; 7:1506312. [PMID: 40083331 PMCID: PMC11903727 DOI: 10.3389/frph.2025.1506312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/30/2025] [Indexed: 03/16/2025] Open
Abstract
Worldwide, infertility is a rising problem. A couple's lifestyle, age and environmental exposures can interfere with reproductive health. The scientific field tries to understand the various processes how male and female factors may affect fertility, but translation to the clinic is limited. I here emphasize potential reasons for failure in optimal treatment planning and especially why current prediction modelling falls short. First, Assisted Reproductive Technology (ART) has become a mainstream solution for couples experiencing infertility, while potential causes of infertility remain unexplored or undetermined. For instance, the role of men is generally left out of preconceptional testing and care. Second, regularly used statistical or computational methods to estimate pregnancy outcomes miss important biological and environmental factors, including features from the male side (e.g., age, smoking, obesity status, alcohol use and occupation), as well as genetic and epigenetic characteristics. I suggest using an integrated approach of biostatistics and machine learning methods to improve diagnostics and prediction modelling in the fertility clinic. The novelty of this concept includes the use of empirically collected information on the sperm epigenome combined with readily available data from medical records from both partners and lifestyle factors. As the reproductive field needs well-designed models at different levels, derivatives are needed. The objectives of patients, clinicians, and embryologists differ slightly, and mathematical models need to be adapted accordingly. A multidisciplinary approach where patients are seen by both, clinicians and biomedically skilled counsellors, could help provide evidence-based assistance to improve pregnancy success. Next, when it concerns factors that may change the ability to produce optimal embryos in ART, the embryologist would benefit from a personalized prediction model, including medical history of the patient as well as genetic and epigenetic data from easily accessible germ cells, such as sperm.
Collapse
Affiliation(s)
- Adelheid Soubry
- Epigenetic Epidemiology Lab, Department of Human Genetics, Faculty of Medicine, KU Leuven—University of Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Leggio L, Paternò G, Cavallaro F, Falcone M, Vivarelli S, Manna C, Calogero AE, Cannarella R, Iraci N. Sperm epigenetics and sperm RNAs as drivers of male infertility: truth or myth? Mol Cell Biochem 2025; 480:659-682. [PMID: 38717684 PMCID: PMC11835981 DOI: 10.1007/s11010-024-04962-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/08/2024] [Indexed: 02/19/2025]
Abstract
Male infertility represents a complex clinical condition that often challenges the ability of reproductive specialists to find its etiology and then propose an adequate treatment. The unexplained decline in sperm count, as well as the association between male infertility and mortality, morbidity, and cancer, has prompted researchers toward an urgent need to better understand the causes of male infertility. Therefore, molecular biologists are increasingly trying to study whether sperm epigenetic alterations may be involved in male infertility and embryo developmental abnormalities. In this context, research is also trying to uncover the hidden role of sperm RNAs, both coding and non-coding. This narrative review aims to thoroughly and comprehensively present the relationship between sperm epigenetics, sperm RNAs, and human fertility. We first focused on the technological aspects of studying sperm epigenetics and RNAs, relating to the complex role(s) played in sperm maturation, fertilization, and embryo development. Then, we examined the intricate connections between epigenetics and RNAs with fertility measures, namely sperm concentration, embryo growth and development, and live birth rate, in both animal and human studies. A better understanding of the molecular mechanisms involved in sperm epigenetic regulation, as well as the impact of RNA players, will help to tackle infertility.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | - Greta Paternò
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | - Fabrizio Cavallaro
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | - Marco Falcone
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | - Silvia Vivarelli
- Department of Biomedical and Dental Sciences, Morphological and Functional Imaging, Section of Occupational Medicine, University of Messina, 98125, Messina, Italy
| | - Claudio Manna
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
- Biofertility IVF and Infertility Center, Rome, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy.
| |
Collapse
|
9
|
Calcaterra V, Tiranini L, Magenes VC, Rossi V, Cucinella L, Nappi RE, Zuccotti G. Impact of Obesity on Pubertal Timing and Male Fertility. J Clin Med 2025; 14:783. [PMID: 39941454 PMCID: PMC11818283 DOI: 10.3390/jcm14030783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Childhood obesity has profound effects on puberty in boys and girls, altering its timing, progression, and associated hormonal changes. Also, later male fertility could be impaired by childhood and pubertal obesity in light of the impact of inflammatory markers on semen quality. The aim of this narrative review is to explore the intricate relationship between childhood obesity and its impact on pubertal development and fertility, with a specific focus on boys. Such a relationship between obesity and pubertal timing in males is highly influenced by metabolic, hormonal, genetic, epigenetic, and environmental factors. While many studies suggest that obesity accelerates pubertal onset in boys, some studies do not confirm these findings, especially in cases of severe obesity. In fact, delayed puberty has also been reported in certain instances. Obesity influences fertility through different central and peripheral processes, including an altered endocrine milieu, inflammatory environment, and epigenetic modifications that alter semen quality and vitality, leading to subfertility or infertility. The early identification and management of potential issues associated with obesity are crucial for ensuring optimal reproductive health in adulthood. Further research is essential to clarify these associations and to develop targeted interventions aimed at preventing the negative health outcomes associated with obesity-related disruptions in puberty and fertility.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (V.R.); (G.Z.)
| | - Lara Tiranini
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.T.); (L.C.); (R.E.N.)
| | | | - Virginia Rossi
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (V.R.); (G.Z.)
| | - Laura Cucinella
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.T.); (L.C.); (R.E.N.)
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Rossella Elena Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.T.); (L.C.); (R.E.N.)
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (V.R.); (G.Z.)
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milano, Italy
| |
Collapse
|
10
|
Akhatova A, Jones C, Coward K, Yeste M. How do lifestyle and environmental factors influence the sperm epigenome? Effects on sperm fertilising ability, embryo development, and offspring health. Clin Epigenetics 2025; 17:7. [PMID: 39819375 PMCID: PMC11740528 DOI: 10.1186/s13148-025-01815-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
Recent studies support the influence of paternal lifestyle and diet before conception on the health of the offspring via epigenetic inheritance through sperm DNA methylation, histone modification, and small non-coding RNA (sncRNA) expression and regulation. Smoking may induce DNA hypermethylation in genes related to anti-oxidation and insulin resistance. Paternal diet and obesity are associated with greater risks of metabolic dysfunction in offspring via epigenetic alterations in the sperm. Metabolic changes, such as high blood glucose levels and increased body weight, are commonly observed in the offspring of fathers subjected to chronic stress, in addition to an enhanced risk of depressive-like behaviour and increased sensitivity to stress in both the F0 and F1 generations. DNA methylation is correlated with alterations in sperm quality and the ability to fertilise oocytes, possibly via a differentially regulated MAKP81IP3 signalling pathway. Paternal exposure to toxic endocrine-disrupting chemicals (EDCs) is also linked to the transgenerational transmission of increased predisposition to disease, infertility, testicular disorders, obesity, and polycystic ovarian syndrome (PCOS) in females through epigenetic changes during gametogenesis. As the success of assisted reproductive technology (ART) is also affected by paternal diet, BMI, and alcohol consumption, its outcomes could be improved by modifying factors that are dependent on male lifestyle choices and environmental factors. This review discusses the importance of epigenetic signatures in sperm-including DNA methylation, histone retention, and sncRNA-for sperm functionality, early embryo development, and offspring health. We also discuss the mechanisms by which paternal lifestyle and environmental factors (obesity, smoking, EDCs, and stress) may impact the sperm epigenome.
Collapse
Affiliation(s)
- Ayazhan Akhatova
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- School of Medicine, Nazarbayev University, Zhanybek-Kerey Khan Street 5/1, 010000, Astana, Kazakhstan
| | - Celine Jones
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Kevin Coward
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, 17003, Girona, Spain.
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, 17003, Girona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain.
| |
Collapse
|
11
|
Ku CW, Pek JW, Cheung YB, Tharmalingam Durgahshree MD, Chan M, Lee YH, Godfrey K, Yap F, Chan JKY, Loy SL. Investigating male factors and their relationships with reproductive health outcomes: a case-control study protocol for Towards Optimal Fertility, Fathering, and Fatherhood studY (TOFFFY) in Singapore. BMJ Open 2025; 15:e088143. [PMID: 39819913 PMCID: PMC11751997 DOI: 10.1136/bmjopen-2024-088143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025] Open
Abstract
INTRODUCTION Despite the global prevalence of low fertility rates, male contributions to fertility and reproductive health outcomes have been understudied. This study aims to investigate the male contribution to fertility and explore the underlying biological mechanisms. Specifically, we aim to (1) identify male factors associated with successful pregnancy, (2) develop a fertility index incorporating modifiable factors for both males and females to predict pregnancy rate and (3) explore the relationship of male modifiable factors with semen parameters and molecular characteristics. METHODS AND ANALYSIS We will conduct an unmatched case-control study involving 240 couples with impaired male fertility (cases) and 240 couples with normal male fertility (controls). Between July 2024 and June 2026, we will recruit 480 eligible couples from KK Women's and Children's Hospital, Singapore. Male and female participants will complete questionnaires on sociodemographics, general health and lifestyle factors, and their anthropometry and body fat composition will be measured. Blood and semen samples from the male participants will be collected for biochemical, molecular and semen analyses. Predictive male factors will be identified using the least absolute shrinkage and selection operator method, accounting for female factors. We will construct a logistic regression model incorporating both male and female factors to derive a fertility index, which will be evaluated using cross-validation on subsets of the study population. Multivariable linear regression will be used to explore relationships between male modifiable exposures and semen parameters. ETHICS AND DISSEMINATION The study protocol has received approval from the Centralised Institutional Review Board of SingHealth (2024/2120), Singapore. Participants will provide written informed consent. Study results will be disseminated through conferences and peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER NCT06293235.
Collapse
Affiliation(s)
- Chee Wai Ku
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | - Jun Wei Pek
- Temasek Life Sciences Laboratory, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Yin Bun Cheung
- Program in Health Services and Systems Research and Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
- Tampere Centre for Child, Adolescent and Maternal Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | - Melinda Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
| | - Yie Hou Lee
- Duke-NUS Medical School, Singapore
- Singapore-MIT Alliance for Research and Technology, Singapore
| | - Keith Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton and Southampton University Hospitals NHS Trust, Southampton, UK
- University of Southampton and University Hospital Southampton National Health Service Foundation Trust, National Institute for Health Research Southampton Biomedical Research Centre, Southampton, UK
| | - Fabian Yap
- Duke-NUS Medical School, Singapore
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | - See Ling Loy
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
- Duke-NUS Medical School, Singapore
| |
Collapse
|
12
|
Adthapanyawanich K, Aitsarangkun Na Ayutthaya K, Kreungnium S, Mark PJ, Nakata H, Chen W, Chinda K, Amatyakul P, Tongpob Y. Molecular Mechanisms and Therapeutic Potential of Mulberry Fruit Extract in High-Fat Diet-Induced Male Reproductive Dysfunction: A Comprehensive Review. Nutrients 2025; 17:273. [PMID: 39861403 PMCID: PMC11767445 DOI: 10.3390/nu17020273] [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: 09/25/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
High-fat diet (HFD)-induced obesity represents a significant challenge to male reproductive health, affecting approximately 13% of the global adult population. This comprehensive review synthesizes current evidence regarding mulberry (Morus alba L.) fruit extract's therapeutic potential for HFD-induced male reproductive dysfunction. Through comprehensive analysis of the peer-reviewed literature from multiple databases (PubMed, Web of Science, Scopus, and Google Scholar; 2005-2024), we evaluated mulberry extract's effects on testicular morphology, spermatogenesis, sperm parameters, and the underlying molecular mechanisms. Mechanistic studies reveal that standardized mulberry extract mediates protective effects through multiple pathways: enhanced antioxidant enzyme activities (SOD: +45%, Catalase: +38%, GPx: +35%), reduced inflammatory markers (TNF-α: -64%, IL-6: -58%), and modulated NF-κB signaling (-42.3%). These effects are facilitated by mulberry's rich phytochemical profile, particularly anthocyanins (2.92-5.35 mg/g dry weight) and polyphenols (4.23-6.38 mg/g). The extract demonstrates particular efficacy in preserving seminiferous tubule integrity and maintaining blood-testis barrier function, with treated groups maintaining up to 85% of normal tubular architecture compared to HFD controls. Key molecular mechanisms include AMPK/SIRT1 pathway activation (2.3-fold increase), enhanced mitochondrial function (67% increase in mtDNA copy number), and epigenetic regulation of metabolic pathways. Temporal analysis indicates optimal therapeutic effects after 28 days of treatment, with initial improvements observable within 14 days. While current evidence is promising, limitations include predominant reliance on rodent models and lack of standardized extraction protocols. Future research priorities include well-designed human clinical trials, standardization of preparation methods, and investigation of potential synergistic effects with other therapeutic agents. This comprehensive review indicates that mulberry extract is a promising therapeutic candidate for obesity-related male infertility, warranting further clinical investigation.
Collapse
Affiliation(s)
- Kannika Adthapanyawanich
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (K.A.); (K.A.N.A.); (S.K.)
- Centre of Excellence in Medical Biotechnology (CEMB), Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand;
| | | | - Siriporn Kreungnium
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (K.A.); (K.A.N.A.); (S.K.)
| | - Peter J. Mark
- School of Human Sciences, The University of Western Australia, Perth 6009, Australia
| | - Hiroki Nakata
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu 923-8511, Ishikawa, Japan
| | - Wai Chen
- Curtin Medical School, and Curtin enAble Institute, Curtin University, Perth 6102, Australia
- Fiona Stanley Hospital, Perth 6150, Australia
| | - Kroekkiat Chinda
- Centre of Excellence in Medical Biotechnology (CEMB), Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand;
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Patcharada Amatyakul
- Department of Obstetrics and Gynecology, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand;
| | - Yutthapong Tongpob
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (K.A.); (K.A.N.A.); (S.K.)
- Centre of Excellence in Medical Biotechnology (CEMB), Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand;
| |
Collapse
|
13
|
Achkar ME, Atieh O, Ghadban C, Awad T, Ghadban E, Grandjean V, Yarkiner Z, Raad G, Khalife MF. Preconceptional paternal obesity may increase the risk of congenital urogenital anomalies in offspring: A case-control study. Andrology 2025; 13:45-54. [PMID: 38837622 PMCID: PMC11635552 DOI: 10.1111/andr.13673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Congenital urogenital anomalies affect 4-60 per 10,000 births. Maternal obesity, along with other risk factors, is well documented as a contributing factor. However, the impact of paternal obesity on risk is unclear. Obesity is prevalent among men of reproductive age, highlighting the need for further research into the potential association between paternal obesity and offspring congenital urogenital anomalies. OBJECTIVES This study aims to determine the association between paternal obesity and the risk of congenital urogenital malformations in offspring. METHODS Case-control study conducted on 179 newborns (91 cases, 88 controls) selected from the Notre Dame des Secours-university hospital database. Cases were identified as newborns presenting at least one congenital urogenital abnormality, defined as developmental anomalies that can result in a variety of malformations affecting the kidneys, ureters, bladder, and urethra. Controls were identified as newborns without any congenital abnormalities. The exclusion criteria were maternal obesity, infections during pregnancy, chronic diseases, prematurity, growth retardation, assisted reproductive technologies for conception, substance abuse, down syndrome, and other malformations. Data were collected through phone interviews, medical records, and questionnaires. In this study, the exposure was the preconceptional paternal body mass index (BMI), which was calculated based on self-reported height and weight. According to guidelines from the US Centers for Disease Control and Prevention (CDC), individuals are considered to be in the healthy weight range if their BMI (kg/m2) is between 18.5 and < 25. They are classified as overweight if their BMI is ≥ 25, obese class I if their BMI is between 30 and < 35, obese class II if their BMI is between 35 and < 40, and obese class III if their BMI is 40 or higher. Logistic regression analysis was employed to quantify the association between paternal obesity and urogenital conditions in offspring. RESULTS Significant differences in median (minimum-maximum) paternal BMI values were noted between the cases and controls at the time of conception (cases: 27.7 (43-20.1), controls: 24.8 (40.7-19.6); p < 0.0001). Logistic regression analysis confirmed that at the time of conception, compared to normal-weight fathers, overweight fathers displayed a heightened risk of offspring congenital malformations, with an odds ratio (OR) of 4.44 (95% CI = 2.1-9.1). Similarly, fathers categorized as obese Class I at conception had approximately eight times higher odds (OR = 8.62, 95% CI = 2.91-25.52) of having offspring with urogenital conditions compared to normal-weight fathers. Additionally, fathers classified as obese Class II at conception exhibited 5.75 times higher odds (OR = 5.75, 95% CI = 0.96-34.44) of having offspring with urogenital conditions in comparison to normal-weight fathers. DISCUSSION AND CONCLUSION We found that the risk of urogenital malformations increased with paternal BMI during the preconceptional period. The findings suggest the importance of addressing paternal obesity in efforts to reduce the risk of urogenital congenital malformations in offspring.
Collapse
Affiliation(s)
- Mariella El Achkar
- School of Medicine and Medical SciencesHoly Spirit University of KaslikJouniehLebanon
| | - Ornina Atieh
- School of Medicine and Medical SciencesHoly Spirit University of KaslikJouniehLebanon
| | - Carole Ghadban
- School of Medicine and Medical SciencesHoly Spirit University of KaslikJouniehLebanon
| | - Toufic Awad
- School of Medicine and Medical SciencesHoly Spirit University of KaslikJouniehLebanon
| | - Elie Ghadban
- School of Medicine and Medical SciencesHoly Spirit University of KaslikJouniehLebanon
| | - Valérie Grandjean
- Université Côte d'AzurInsermC3M, Team Control of Gene Expression (10)NiceFrance
| | - Zalihe Yarkiner
- Department of Basic Sciences and HumanitiesFaculty of Arts and SciencesCyprus International University, North NicosiaNorthern Cyprus via MersinNicosiaTurkey
| | - Georges Raad
- School of Medicine and Medical SciencesHoly Spirit University of KaslikJouniehLebanon
| | - Marie‐Claude Fadous Khalife
- School of Medicine and Medical SciencesHoly Spirit University of KaslikJouniehLebanon
- Notre Dame des Secours University Hospital CenterByblosLebanon
| |
Collapse
|
14
|
Dahlen CR, Ramírez-Zamudio GD, Bochantin-Winders KA, Hurlbert JL, Crouse MS, McLean KJ, Diniz WJS, Amat S, Snider AP, Caton JS, Reynolds LP. International Symposium on Ruminant Physiology: Paternal Nutrient Supply: Impacts on Physiological and Whole Animal Outcomes in Offspring. J Dairy Sci 2024:S0022-0302(24)01425-5. [PMID: 39710267 DOI: 10.3168/jds.2024-25800] [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: 10/01/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024]
Abstract
Recent evidence suggests that environmental factors experienced by sires can be transmitted through the ejaculate (seminal plasma + sperm) into the female reproductive tract, influencing fertilization, embryo development, and postnatal offspring outcomes. This concept is termed paternal programming. In rodents, sire nutrition was shown to directly alter offspring outcomes through sperm epigenetic signatures, DNA damage/oxidative stress, cytokine profiles, and/or the seminal microbiome. Response variables impacted in rodent models, including adiposity, muscle mass, metabolic responses, and reproductive performance, could have major productivity and financial implications for producers if these paternal programming responses are also present in ruminant species. However, a paucity of data exist regarding paternal programming in ruminants. The limited data in the literature mainly point to alterations in sperm epigenome as a result of sire diet or environment. Global nutrition has been implicated in ruminant models to alter seminal cytokine profiles, which could subsequently alter the uterine environment and immune response to mating. Several reports indicate that embryo development and epigenetic signatures can be impacted by sire plane of nutrition and inclusion of specific feed ingredients into diets (polyunsaturated fatty acids, folic acid, and rumen protected methionine). Models of sheep nutrition indicate that addition of rumen protected methionine can impact DNA methylation and offspring performance characteristics extending to the F3 generation, and that divergent planes of sire nutrition can cause altered hormone profiles and insulin/glucose metabolism in offspring. There are almost unlimited opportunities for discovery in this area, but researchers are encouraged to target critical questions such as whether and the extent to which paternal programming effects are present in common management scenarios, the mechanisms by which paternal programming is inherited in ruminants, and whether the effects of paternal nutrition interact with those of maternal nutrition to influence offspring physiology, whole animal outcomes, and herd or flock productivity.
Collapse
Affiliation(s)
| | - Germán D Ramírez-Zamudio
- North Dakota State University, Fargo, ND, USA; University of São Paulo, Pirassununga, SP, Brazil
| | | | | | | | | | | | - Samat Amat
- North Dakota State University, Fargo, ND, USA
| | | | | | | |
Collapse
|
15
|
Kampmann U, Suder LB, Nygaard M, Geiker NRW, Nielsen HS, Almstrup K, Bruun JM, Magkos F, Ovesen P, Catalano P. Prepregnancy and Gestational Interventions to Prevent Childhood Obesity. J Clin Endocrinol Metab 2024; 110:e8-e18. [PMID: 39401333 DOI: 10.1210/clinem/dgae724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Indexed: 12/19/2024]
Abstract
Childhood obesity is a significant global health issue with complex and multifactorial origins, often beginning before conception and influenced by both maternal and paternal health. The increased prevalence of prepregnancy obesity and gestational diabetes mellitus in women of reproductive age contributes to a heightened risk of metabolic dysfunction in offspring. Current clinical practices often implement lifestyle interventions after the first trimester and have limited success, implying that they miss a critical window for effective metabolic adjustments. This review examines the limitations of lifestyle interventions during pregnancy in improving perinatal outcomes and highlights the importance of initiating such interventions before conception to positively impact parental health and fetal development. A re-evaluation of strategies is needed to enhance the metabolic health of prospective parents as a preventive measure against childhood obesity.
Collapse
Affiliation(s)
- Ulla Kampmann
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, DK-8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, DK-8200, Denmark
| | - Louise Birk Suder
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, DK-8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, DK-8200, Denmark
| | - Malene Nygaard
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, DK-1958, Denmark
| | | | - Henriette Svarre Nielsen
- Department of Gynecology and Obstetrics, Copenhagen University Hospital Hvidovre, Hvidovre, DK 2650, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen N, DK-2200, Denmark
| | - Kristian Almstrup
- Department of Growth and reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, DK-2100, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Jens Meldgaard Bruun
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, DK-8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, DK-8200, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, DK-1958, Denmark
| | - Per Ovesen
- Department of Clinical Medicine, Aarhus University, Aarhus N, DK-8200, Denmark
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus N, DK-8200, Denmark
| | - Patrick Catalano
- Division of Reproductive Endocrinology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
16
|
Zhao Z, Yang T, Li F. Sperm RNA code in spermatogenesis and male infertility. Reprod Biomed Online 2024; 49:104375. [PMID: 39481211 DOI: 10.1016/j.rbmo.2024.104375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/22/2024] [Accepted: 07/05/2024] [Indexed: 11/02/2024]
Abstract
Spermatozoa are traditionally thought to be transcriptionally inert, but recent studies have revealed the presence of sperm RNA, some of which is derived from the residues of spermatocyte transcription and some from epididymosomes. Paternal sperm RNA can be affected by external factors and further modified at the post-transcriptional level, for example N6-methyladenosine (m6A), thus shaping spermatogenesis and reproductive outcome. This review briefly introduces the origin of sperm RNA and, on this basis, summarizes the current knowledge on RNA modifications and their functional role in spermatogenesis and male infertility. The bottlenecks and knowledge gaps in the current research on RNA modification in male reproduction have also been indicated. Further investigations are needed to elucidate the functional consequences of these modifications, providing new therapeutic and preventive strategies for reproductive health and genetic inheritance.
Collapse
Affiliation(s)
- Zhongyi Zhao
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Tingting Yang
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| | - Fuping Li
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| |
Collapse
|
17
|
Saez Lancellotti TE, Avena MV, Funes AK, Bernal-López MR, Gómez-Huelgas R, Fornes MW. Exploring the impact of lipid stress on sperm cytoskeleton: insights and prospects. Nat Rev Urol 2024:10.1038/s41585-024-00952-1. [PMID: 39528754 DOI: 10.1038/s41585-024-00952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The decline in male fertility correlates with the global rise in obesity and dyslipidaemia, representing significant public health challenges. High-fat diets induce metabolic alterations, including hypercholesterolaemia, hepatic steatosis and atherosclerosis, with detrimental effects on testicular function. Testicular tissue, critically dependent on lipids for steroidogenesis, is particularly vulnerable to these metabolic disruptions. Excessive lipid accumulation within the testes, including cholesterol, triglycerides and specific fatty acids, disrupts essential sperm production processes such as membrane formation, maturation, energy metabolism and cell signalling. This leads to apoptosis, impaired spermatogenesis, and abnormal sperm morphology and function, ultimately compromising male fertility. During spermiogenesis, round spermatids undergo extensive reorganization, including the formation of the acrosome, manchette and specialized filamentous structures, which are essential for defining the final sperm cell shape. In this Perspective, we examine the impact of high-fat diets on the cytoskeleton of spermatogenic cells and its consequences to identify the mechanisms underlying male infertility associated with dyslipidaemia. Understanding these processes may facilitate the development of therapeutic strategies, such as dietary interventions or natural product supplementation, that aim to address infertility in men with obesity and hypercholesterolaemia. The investigation of cytoskeleton response to lipid stress extends beyond male reproduction, offering insights with broader implications.
Collapse
Affiliation(s)
- Tania E Saez Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina.
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina.
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.
| | - María V Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Abi K Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María-Rosa Bernal-López
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Gómez-Huelgas
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel W Fornes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
18
|
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
|
19
|
Vozdova M, Kubickova S, Kopecka V, Sipek J, Rubes J. Effect of body mass index on semen quality, sperm chromatin integrity and sperm DNA methylation. Obes Res Clin Pract 2024; 18:380-387. [PMID: 39358131 DOI: 10.1016/j.orcp.2024.09.276] [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: 05/13/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Obesity represents a growing problem due to its impacts on human health and reproduction. In this study, we analysed semen quality, sperm DNA integrity and gene-specific CpG methylation in 116 healthy men from normal population. The men were divided into three groups according to their body mass index (BMI), and their ejaculates were analysed using standard methods, sperm chromatin structure assay (SCSA), methylation next generation sequencing (NGS) and amplicon sequencing. The sperm methylation NGS revealed six significantly differentially methylated regions (DMRs). Using subsequent targeted amplicon sequencing in 116 men, two of the DMRs were proved as differentially methylated in sperm of men with normal BMI vs. BMI ≥ 25. The DMRs were located in the EPHA8 and ANKRD11 gene. Also, we detected a significant decline in the EPHA8, ANKRD11 and CFAP46 gene methylation in association with increasing BMI values. The genes EPHA8 and ANKRD11 are involved in the nervous system and brain development; the CFAP46 gene plays a role in a flagellar assembly and is associated with sperm motility. Significantly lower rates of motile and progressive motile sperm were observed in men with BMI ≥ 30. Our results show that excess body weight can modify CpG methylation of specific genes, affect sperm motility, and compromise sperm chromatin integrity. These factors can stand behind the observed reduced fertility in men with obesity. The methylation changes might be transmitted to their offspring through sperm, and become a basis for possible developmental and reproductive issues in the next generation.
Collapse
Affiliation(s)
- Miluse Vozdova
- Department of Genetics and Reproductive Biotechnologies, Central European Institute of Technology, Veterinary Research Institute, Brno, Czech Republic.
| | - Svatava Kubickova
- Department of Genetics and Reproductive Biotechnologies, Central European Institute of Technology, Veterinary Research Institute, Brno, Czech Republic
| | - Vera Kopecka
- Department of Genetics and Reproductive Biotechnologies, Central European Institute of Technology, Veterinary Research Institute, Brno, Czech Republic
| | - Jaroslav Sipek
- Department of Genetics and Reproductive Biotechnologies, Central European Institute of Technology, Veterinary Research Institute, Brno, Czech Republic
| | - Jiri Rubes
- Department of Genetics and Reproductive Biotechnologies, Central European Institute of Technology, Veterinary Research Institute, Brno, Czech Republic
| |
Collapse
|
20
|
Liu S, Holmes AD, Katzman S, Sharma U. A sperm-enriched 5'fragment of tRNA-Valine regulates preimplantation embryonic transcriptome and development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607197. [PMID: 39211093 PMCID: PMC11361008 DOI: 10.1101/2024.08.08.607197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sperm small RNAs have been implicated in intergenerational epigenetic inheritance of paternal environmental effects; however, their biogenesis and functions remain poorly understood. We previously identified a 5' fragment of tRNA-Valine-CAC-2 (tRFValCAC) as one of the most abundant small RNA in mature sperm. tRFValCAC is specifically enriched in sperm during post-testicular maturation in the epididymis, and we found that it is delivered to sperm from epididymis epithelial cells via extracellular vesicles. Here, we investigated the mechanistic basis of tRFValCAC delivery to sperm and its functions in the early embryo. We show that tRFValCAC interacts with an RNA binding protein, heterogeneous nuclear ribonucleoprotein A/B (hnRNPAB), in the epididymis, and this interaction regulates the sorting and packing of tRFValCAC into extracellular vesicles. In the embryo, we found that tRFValCAC regulates early embryonic mRNA processing and splicing. Inhibition of tRFValCAC in preimplantation embryos altered the transcript abundance of genes involved in RNA splicing and mRNA processing. Importantly, tRFValCAC-inhibited embryos showed altered mRNA splicing, including alternative splicing of various splicing factors and genes important for proper preimplantation embryonic development. Finally, we find that inhibition of tRFValCAC in zygotes delayed preimplantation embryonic development. Together, our results reveal a novel function of a sperm-enriched tRF in regulating alternating splicing and preimplantation embryonic development and shed light on the mechanism of sperm small RNA-mediated epigenetic inheritance.
Collapse
Affiliation(s)
- Simeiyun Liu
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Andrew D. Holmes
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Sol Katzman
- Genomics Institute, University of California, Santa Cruz, California, 95064
| | - Upasna Sharma
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| |
Collapse
|
21
|
Carrageta DF, Pereira SC, Ferreira R, Monteiro MP, Oliveira PF, Alves MG. Signatures of metabolic diseases on spermatogenesis and testicular metabolism. Nat Rev Urol 2024; 21:477-494. [PMID: 38528255 DOI: 10.1038/s41585-024-00866-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 03/27/2024]
Abstract
Diets leading to caloric overload are linked to metabolic disorders and reproductive function impairment. Metabolic and hormonal abnormalities stand out as defining features of metabolic disorders, and substantially affect the functionality of the testis. Metabolic disorders induce testicular metabolic dysfunction, chronic inflammation and oxidative stress. The disruption of gastrointestinal, pancreatic, adipose tissue and testicular hormonal regulation induced by metabolic disorders can also contribute to a state of compromised fertility. In this Review, we will delve into the effects of high-fat diets and metabolic disorders on testicular metabolism and spermatogenesis, which are crucial elements for male reproductive function. Moreover, metabolic disorders have been shown to influence the epigenome of male gametes and might have a potential role in transmitting phenotype traits across generations. However, the existing evidence strongly underscores the unmet need to understand the mechanisms responsible for transgenerational paternal inheritance of male reproductive function impairment related to metabolic disorders. This knowledge could be useful for developing targeted interventions to prevent, counteract, and most of all break the perpetuation chain of male reproductive dysfunction associated with metabolic disorders across generations.
Collapse
Affiliation(s)
- David F Carrageta
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Sara C Pereira
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Mariana P Monteiro
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Marco G Alves
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Campus de Santiago Agra do Crasto, Aveiro, Portugal.
| |
Collapse
|
22
|
Tahiri I, Llana SR, Fos-Domènech J, Milà-Guash M, Toledo M, Haddad-Tóvolli R, Claret M, Obri A. Paternal obesity induces changes in sperm chromatin accessibility and has a mild effect on offspring metabolic health. Heliyon 2024; 10:e34043. [PMID: 39100496 PMCID: PMC11296027 DOI: 10.1016/j.heliyon.2024.e34043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024] Open
Abstract
The increasing global burden of metabolic disorders including obesity and diabetes necessitates a comprehensive understanding of their etiology, which not only encompasses genetic and environmental factors but also parental influence. Recent evidence has unveiled paternal obesity as a contributing factor to offspring's metabolic health via sperm epigenetic modifications. In this study, we investigated the impact of a Western diet-induced obesity in C57BL/6 male mice on sperm chromatin accessibility and the subsequent metabolic health of their progeny. Utilizing Assay for Transposase-Accessible Chromatin with sequencing, we discovered 450 regions with differential accessibility in sperm from obese fathers, implicating key developmental and metabolic pathways. Contrary to expectations, these epigenetic alterations in sperm were not predictive of long-term metabolic disorders in offspring, who exhibited only mild transient metabolic changes early in life. Both male and female F1 progeny showed no enduring predisposition to obesity or diabetes. These results underscore the biological resilience of offspring to paternal epigenetic inheritance, suggesting a complex interplay between inherited epigenetic modifications and the offspring's own developmental compensatory mechanisms. This study calls for further research into the biological processes that confer this resilience, which could inform interventional strategies to combat the heritability of metabolic diseases.
Collapse
Affiliation(s)
- Iasim Tahiri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Sergio R. Llana
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Júlia Fos-Domènech
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Maria Milà-Guash
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Miriam Toledo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
23
|
Crafa A, Cannarella R, Calogero AE, Gunes S, Agarwal A. Behind the Genetics: The Role of Epigenetics in Infertility-Related Testicular Dysfunction. Life (Basel) 2024; 14:803. [PMID: 39063558 PMCID: PMC11277947 DOI: 10.3390/life14070803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
In recent decades, we have witnessed a progressive decline in male fertility. This is partly related to the increased prevalence of chronic diseases (e.g., obesity and diabetes mellitus) and risky lifestyle behaviors. These conditions alter male fertility through various non-genetic mechanisms. However, there is increasing evidence that they are also capable of causing sperm epigenetic alterations, which, in turn, can cause infertility. Furthermore, these modifications could be transmitted to offspring, altering their general and reproductive health. Therefore, these epigenetic modifications could represent one of the causes of the progressive decline in sperm count recorded in recent decades. This review focuses on highlighting epigenetic modifications at the sperm level induced by non-genetic causes of infertility. In detail, the effects on DNA methylation, histone modifications, and the expression profiles of non-coding RNAs are evaluated. Finally, a focus on the risk of transgenerational inheritance is presented. Our narrative review aims to demonstrate how certain conditions can alter gene expression, potentially leading to the transmission of anomalies to future generations. It emphasizes the importance of the early detection and treatment of reversible conditions (such as obesity and varicocele) and the modification of risky lifestyle behaviors. Addressing these issues is crucial for individual health, in preserving fertility, and in ensuring the well-being of future generations.
Collapse
Affiliation(s)
- Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (A.C.); (R.C.); (A.E.C.)
- Global Andrology Forum, Moreland Hills, OH 44022, USA
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (A.C.); (R.C.); (A.E.C.)
- Global Andrology Forum, Moreland Hills, OH 44022, USA
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44106, USA
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (A.C.); (R.C.); (A.E.C.)
- Global Andrology Forum, Moreland Hills, OH 44022, USA
| | - Sezgin Gunes
- Global Andrology Forum, Moreland Hills, OH 44022, USA
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, 55280 Samsun, Türkiye
| | - Ashok Agarwal
- Global Andrology Forum, Moreland Hills, OH 44022, USA
- Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| |
Collapse
|
24
|
Cannarella R, Crafa A, Curto R, Condorelli RA, La Vignera S, Calogero AE. Obesity and male fertility disorders. Mol Aspects Med 2024; 97:101273. [PMID: 38593513 DOI: 10.1016/j.mam.2024.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Often associated with obesity, male infertility represents a widespread condition that challenges the wellbeing of the couple. In this article, we provide a comprehensive and critical analysis of studies exploring the association between obesity and male reproductive function, to evaluate the frequency of this association, and establish the effects of increased body weight on conventional and biofunctional sperm parameters and infertility. In an attempt to find possible molecular markers of infertility in obese male patients, the numerous mechanisms responsible for infertility in overweight/obese patients are reviewed in depth. These include obesity-related functional hypogonadism, insulin resistance, hyperinsulinemia, chronic inflammation, adipokines, irisin, gut hormones, gut microbiome, and sperm transcriptome. According to meta-analytic evidence, excessive body weight negatively influences male reproductive health. This can occurr through a broad array of molecular mechanisms. Some of these are not yet fully understood and need to be further elucidated in the future. A better understanding of the effects of metabolic disorders on spermatogenesis and sperm fertilizing capacity is very useful for identifying new diagnostic markers and designing therapeutic strategies for better clinical management of male infertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
25
|
Tomar A, Gomez-Velazquez M, Gerlini R, Comas-Armangué G, Makharadze L, Kolbe T, Boersma A, Dahlhoff M, Burgstaller JP, Lassi M, Darr J, Toppari J, Virtanen H, Kühnapfel A, Scholz M, Landgraf K, Kiess W, Vogel M, Gailus-Durner V, Fuchs H, Marschall S, Hrabě de Angelis M, Kotaja N, Körner A, Teperino R. Epigenetic inheritance of diet-induced and sperm-borne mitochondrial RNAs. Nature 2024; 630:720-727. [PMID: 38839949 PMCID: PMC11186758 DOI: 10.1038/s41586-024-07472-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/26/2024] [Indexed: 06/07/2024]
Abstract
Spermatozoa harbour a complex and environment-sensitive pool of small non-coding RNAs (sncRNAs)1, which influences offspring development and adult phenotypes1-7. Whether spermatozoa in the epididymis are directly susceptible to environmental cues is not fully understood8. Here we used two distinct paradigms of preconception acute high-fat diet to dissect epididymal versus testicular contributions to the sperm sncRNA pool and offspring health. We show that epididymal spermatozoa, but not developing germ cells, are sensitive to the environment and identify mitochondrial tRNAs (mt-tRNAs) and their fragments (mt-tsRNAs) as sperm-borne factors. In humans, mt-tsRNAs in spermatozoa correlate with body mass index, and paternal overweight at conception doubles offspring obesity risk and compromises metabolic health. Sperm sncRNA sequencing of mice mutant for genes involved in mitochondrial function, and metabolic phenotyping of their wild-type offspring, suggest that the upregulation of mt-tsRNAs is downstream of mitochondrial dysfunction. Single-embryo transcriptomics of genetically hybrid two-cell embryos demonstrated sperm-to-oocyte transfer of mt-tRNAs at fertilization and suggested their involvement in the control of early-embryo transcription. Our study supports the importance of paternal health at conception for offspring metabolism, shows that mt-tRNAs are diet-induced and sperm-borne and demonstrates, in a physiological setting, father-to-offspring transfer of sperm mitochondrial RNAs at fertilization.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Body Mass Index
- Diet, High-Fat/adverse effects
- Embryo, Mammalian/cytology
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Epididymis/cytology
- Epigenesis, Genetic/genetics
- Fertilization/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Mice, Inbred C57BL
- Mitochondria/genetics
- Mitochondria/metabolism
- Mitochondria/pathology
- Obesity/genetics
- Obesity/metabolism
- Obesity/etiology
- Oocytes/metabolism
- Overweight/genetics
- Overweight/metabolism
- Paternal Inheritance/genetics
- RNA, Mitochondrial/genetics
- RNA, Mitochondrial/metabolism
- RNA, Small Untranslated/genetics
- RNA, Small Untranslated/metabolism
- RNA, Transfer/genetics
- RNA, Transfer/metabolism
- Spermatozoa/metabolism
- Testis/cytology
- Transcription, Genetic
Collapse
Affiliation(s)
- A Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - M Gomez-Velazquez
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - R Gerlini
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - G Comas-Armangué
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - L Makharadze
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - T Kolbe
- Unit of in vivo and in vitro Models, Center for Biological Sciences, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- IFA-Tulln, University of Natural Resources and Life Sciences, Vienna, Austria
| | - A Boersma
- Unit of in vivo and in vitro Models, Center for Biological Sciences, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - M Dahlhoff
- Unit of in vivo and in vitro Models, Center for Biological Sciences, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - J P Burgstaller
- Institute of Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Group Molecular Reproduction, IFA-Tulln, Tulln, Austria
| | - M Lassi
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - J Darr
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - J Toppari
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Turku, Finland
- Center for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - H Virtanen
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Turku, Finland
- Center for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - A Kühnapfel
- University of Leipzig, Medical Faculty, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
| | - M Scholz
- University of Leipzig, Medical Faculty, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
| | - K Landgraf
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| | - W Kiess
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - M Vogel
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - V Gailus-Durner
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - H Fuchs
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - S Marschall
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - M Hrabě de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - N Kotaja
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Turku, Finland
| | - A Körner
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - R Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
26
|
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
|
27
|
Cánepa ET, Berardino BG. Epigenetic mechanisms linking early-life adversities and mental health. Biochem J 2024; 481:615-642. [PMID: 38722301 DOI: 10.1042/bcj20230306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/15/2024]
Abstract
Early-life adversities, whether prenatal or postnatal exposure, have been linked to adverse mental health outcomes later in life increasing the risk of several psychiatric disorders. Research on its neurobiological consequences demonstrated an association between exposure to adversities and persistent alterations in the structure, function, and connectivity of the brain. Consistent evidence supports the idea that regulation of gene expression through epigenetic mechanisms are involved in embedding the impact of early-life experiences in the genome and mediate between social environments and later behavioral phenotypes. In addition, studies from rodent models and humans suggest that these experiences and the acquired risk factors can be transmitted through epigenetic mechanisms to offspring and the following generations potentially contributing to a cycle of disease or disease risk. However, one of the important aspects of epigenetic mechanisms, unlike genetic sequences that are fixed and unchangeable, is that although the epigenetic markings are long-lasting, they are nevertheless potentially reversible. In this review, we summarize our current understanding of the epigenetic mechanisms involved in the mental health consequences derived from early-life exposure to malnutrition, maltreatment and poverty, adversities with huge and pervasive impact on mental health. We also discuss the evidence about transgenerational epigenetic inheritance in mammals and experimental data suggesting that suitable social and pharmacological interventions could reverse adverse epigenetic modifications induced by early-life negative social experiences. In this regard, these studies must be accompanied by efforts to determine the causes that promote these adversities and that result in health inequity in the population.
Collapse
Affiliation(s)
- Eduardo T Cánepa
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| | - Bruno G Berardino
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| |
Collapse
|
28
|
Pan X, Dai W, Wang Z, Li S, Sun T, Miao N. PIWI-Interacting RNAs: A Pivotal Regulator in Neurological Development and Disease. Genes (Basel) 2024; 15:653. [PMID: 38927589 PMCID: PMC11202748 DOI: 10.3390/genes15060653] [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: 04/13/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
PIWI-interacting RNAs (piRNAs), a class of small non-coding RNAs (sncRNAs) with 24-32 nucleotides (nt), were initially identified in the reproductive system. Unlike microRNAs (miRNAs) or small interfering RNAs (siRNAs), piRNAs normally guide P-element-induced wimpy testis protein (PIWI) families to slice extensively complementary transposon transcripts without the seed pairing. Numerous studies have shown that piRNAs are abundantly expressed in the brain, and many of them are aberrantly regulated in central neural system (CNS) disorders. However, the role of piRNAs in the related developmental and pathological processes is unclear. The elucidation of piRNAs/PIWI would greatly improve the understanding of CNS development and ultimately lead to novel strategies to treat neural diseases. In this review, we summarized the relevant structure, properties, and databases of piRNAs and their functional roles in neural development and degenerative disorders. We hope that future studies of these piRNAs will facilitate the development of RNA-based therapeutics for CNS disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Nan Miao
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China; (X.P.); (W.D.); (Z.W.); (S.L.); (T.S.)
| |
Collapse
|
29
|
Aitken RJ. What is driving the global decline of human fertility? Need for a multidisciplinary approach to the underlying mechanisms. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 6:1364352. [PMID: 38726051 PMCID: PMC11079147 DOI: 10.3389/frph.2024.1364352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
An intense period of human population expansion over the past 250 years is about to cease. Total fertility rates are falling dramatically all over the world such that highly industrialized nations, including China and the tiger economies of SE Asia, will see their populations decline significantly in the coming decades. The socioeconomic, geopolitical and environmental ramifications of this change are considerable and invite a multidisciplinary consideration of the underlying mechanisms. In the short-term, socioeconomic factors, particularly urbanization and delayed childbearing are powerful drivers of reduced fertility. In parallel, lifestyle factors such as obesity and the presence of numerous reproductive toxicants in the environment, including air-borne pollutants, nanoplastics and electromagnetic radiation, are seriously compromising reproductive health. In the longer term, it is hypothesized that the reduction in family size that accompanies the demographic transition will decrease selection pressure on high fertility genes leading to a progressive loss of human fecundity. Paradoxically, the uptake of assisted reproductive technologies at scale, may also contribute to such fecundity loss by encouraging the retention of poor fertility genotypes within the population. Since the decline in fertility rate that accompanies the demographic transition appears to be ubiquitous, the public health implications for our species are potentially devastating.
Collapse
Affiliation(s)
- Robert John Aitken
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, School of Environmental and Life Sciences, College of Engineering Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
30
|
Ali N, Amelkina O, Santymire RM, Koepfli KP, Comizzoli P, Vazquez JM. Semen proteome and transcriptome of the endangered black-footed ferret (Mustela nigripes) show association with the environment and fertility outcome. Sci Rep 2024; 14:7063. [PMID: 38528039 PMCID: PMC10963785 DOI: 10.1038/s41598-024-57096-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/14/2024] [Indexed: 03/27/2024] Open
Abstract
The ex situ population of the endangered black-footed ferret (Mustela nigripes) has been experiencing declines in reproductive success over the past 30 years of human-managed care. A potential cause may be environmental-dependent inbreeding depression with diet being one of the contributing factors since ferrets are not fed their natural diet of prairie dogs. Here, we generated and analyzed semen proteome and transcriptome data from both wild and ex situ ferrets maintained on various diets. We identified 1757 proteins across all samples, with 149 proteins unique to the semen of wild ferrets and forming a ribosomal predicted protein-protein interaction cluster. Wild ferrets also differed from ex situ ferrets in their transcriptomic profile, showing enrichment in ribosomal RNA processing and potassium ion transport. Successful fertility outcomes documented for ex situ ferrets showed the strongest association with the semen transcriptome, with enrichment in genes involved in translation initiation and focal adhesion. Fertility also synergized with the effect of diet on differentially expressed transcriptomes, mainly affecting genes enriched in mitochondrial function. Our data and functional networks are important for understanding the causes and mechanisms of declining fertility in the ex situ ferret population and can be used as a resource for future conservation efforts.
Collapse
Affiliation(s)
- Nadya Ali
- Committee on Evolutionary Biology, University of Chicago, Chicago, IL, USA.
| | - Olga Amelkina
- Smithsonian's National Zoo and Conservation Biology Institute, Washington D.C., USA.
| | | | - Klaus-Peter Koepfli
- Smithsonian's National Zoo and Conservation Biology Institute, Washington D.C., USA.
- Smithsonian-Mason School of Conservation, George Mason University, Front Royal, VA, USA.
| | - Pierre Comizzoli
- Smithsonian's National Zoo and Conservation Biology Institute, Washington D.C., USA
| | - Juan M Vazquez
- Department of Integrative Biology, University of California, Berkeley, USA.
| |
Collapse
|
31
|
Zhao Y, Wang K, Zhao C, Liu N, Wang Z, Yang W, Cheng Z, Zhou L, Wang K. The function of tRNA-derived small RNAs in cardiovascular diseases. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102114. [PMID: 38314096 PMCID: PMC10835008 DOI: 10.1016/j.omtn.2024.102114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
tRNA-derived small RNAs (tsRNAs) constitute a subgroup of small noncoding RNAs (ncRNAs) originating from tRNA molecules. Their rich content, evolutionary conservatism, high stability, and widespread existence makes them significant in disease research. These characteristics have positioned tsRNAs as key players in various physiological and pathological processes. tsRNA actively participates in regulating many cellular processes, such as cell death, proliferation, and metabolism. tsRNAs could be promising diagnostic markers for cardiovascular diseases (CVDs). tsRNAs have been identified in serums, suggesting their utility as early indicators for the diagnosis of CVDs. Moreover, the regulatory roles of tsRNAs in CVDs make them promising targets for therapeutic intervention. This review provides a succinct overview of the characteristics, classification, and regulatory functions of tsRNAs in the context of CVDs. By shedding light on the intricate roles of tsRNAs, this knowledge could pave the way for the development of innovative diagnostic tools and therapeutic strategies for CVDs.
Collapse
Affiliation(s)
- Yan Zhao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, P.R. China
| | - Kai Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, P.R. China
| | - Chun Zhao
- College of Biology, Hunan University, Changsha 410082, P.R. China
| | - Ning Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, P.R. China
| | - Zhihong Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, P.R. China
| | - Wenting Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, P.R. China
| | - Zewei Cheng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, P.R. China
| | - Luyu Zhou
- College of Biology, Hunan University, Changsha 410082, P.R. China
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, P.R. China
| |
Collapse
|
32
|
Ferreira SRG, Macotela Y, Velloso LA, Mori MA. Determinants of obesity in Latin America. Nat Metab 2024; 6:409-432. [PMID: 38438626 DOI: 10.1038/s42255-024-00977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024]
Abstract
Obesity rates are increasing almost everywhere in the world, although the pace and timing for this increase differ when populations from developed and developing countries are compared. The sharp and more recent increase in obesity rates in many Latin American countries is an example of that and results from regional characteristics that emerge from interactions between multiple factors. Aware of the complexity of enumerating these factors, we highlight eight main determinants (the physical environment, food exposure, economic and political interest, social inequity, limited access to scientific knowledge, culture, contextual behaviour and genetics) and discuss how they impact obesity rates in Latin American countries. We propose that initiatives aimed at understanding obesity and hampering obesity growth in Latin America should involve multidisciplinary, global approaches that consider these determinants to build more effective public policy and strategies, accounting for regional differences and disease complexity at the individual and systemic levels.
Collapse
Affiliation(s)
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM Campus-Juriquilla, Querétaro, Mexico
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Faculty of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo A Mori
- Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil.
| |
Collapse
|
33
|
Donnelly JM, Walsh JM, Horan MK, Mehegan J, Molloy EJ, Byrne DF, McAuliffe FM. Parental Height and Weight Influence Offspring Adiposity at 2 Years; Findings from the ROLO Kids Birth Cohort Study. Am J Perinatol 2024; 41:422-428. [PMID: 34965588 DOI: 10.1055/s-0041-1740299] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE The perinatal period and in utero environment are important for fetal growth, development, and fetal programming. This study aimed to determine the effect of parental anthropometry and the maternal metabolic milieu on offspring adiposity at 2 years of age. STUDY DESIGN This longitudinal birth cohort includes analysis of maternal (n = 337) and paternal (n = 219) anthropometry and maternal and fetal metabolic markers (n = 337), including glucose, homeostatic model of assessment (HOMA), C-peptide, and leptin from participants of the ROLO (the Randomized Control Trial of Low) pregnancy study, and their partners, to determine an association with offspring anthropometry at two years of age. RESULTS Linear regression, when adjusted for confounders, indicated maternal and paternal anthropometry and was associated with offspring weight and length at 2 years of age. Maternal height was negatively associated with general adiposity in the total cohort of children (p = 0.002) and in female children (p = 0.006) and central adiposity in the total child cohort (p < 0.001). Paternal height was also negatively associated with general adiposity in all children (p = 0.002) and central adiposity in total (p = 0.023) and female children (p = 0.008). Maternal glucose, insulin resistance, and fetal C-peptide positively correlated with anthropometry in total, male, and female children. CONCLUSION Parental anthropometry in the perinatal period has a long-lasting effect on offspring anthropometry beyond the neonatal period. Maternal and fetal metabolic factors influence adiposity, and this extends beyond the perinatal period. Parental adiposity may play a significant role in early childhood adiposity and may be a target for interventions to decrease the risk of early childhood obesity. KEY POINTS · Parental height and weight were associated with offspring anthropometry and measures of offspring adiposity at 2 years of age.. · Maternal glucose, insulin resistance, and fetal C-peptide correlated with offspring anthropometry.. · Parental anthropometry has long-term effect on offspring adiposity and is seen at 2 years of age..
Collapse
Affiliation(s)
- Jean M Donnelly
- University College Dublin, Perinatal Research Centre, School of Medicine, Department of Obstetrics and Gynecology, University College Dublin, National Maternity Hospital, Dublin, Ireland
- Department of Neonatology Our Lady's Children's Hospital Crumlin, Ireland
| | - Jennifer M Walsh
- University College Dublin, Perinatal Research Centre, School of Medicine, Department of Obstetrics and Gynecology, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Mary K Horan
- University College Dublin, Perinatal Research Centre, School of Medicine, Department of Obstetrics and Gynecology, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - John Mehegan
- University College Dublin, School of Public Health, Physiotherapy and Sports Science, Dublin, Ireland
| | - Eleanor J Molloy
- Department of Neonatology Our Lady's Children's Hospital Crumlin, Ireland
- Department of Paediatrics, University of Dublin, Dublin, Ireland
- Department of Neonatology, Coombe Women and Infants Hospital, Dublin, Ireland
| | - David F Byrne
- University College Dublin, Perinatal Research Centre, School of Medicine, Department of Obstetrics and Gynecology, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Fionnuala M McAuliffe
- University College Dublin, Perinatal Research Centre, School of Medicine, Department of Obstetrics and Gynecology, University College Dublin, National Maternity Hospital, Dublin, Ireland
| |
Collapse
|
34
|
Schrott R, Feinberg JI, Newschaffer CJ, Hertz-Picciotto I, Croen LA, Fallin MD, Volk HE, Ladd-Acosta C, Feinberg AP. Exposure to air pollution is associated with DNA methylation changes in sperm. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae003. [PMID: 38559770 PMCID: PMC10980975 DOI: 10.1093/eep/dvae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/10/2024] [Accepted: 02/02/2024] [Indexed: 04/04/2024]
Abstract
Exposure to air pollutants has been associated with adverse health outcomes in adults and children who were prenatally exposed. In addition to reducing exposure to air pollutants, it is important to identify their biologic targets in order to mitigate the health consequences of exposure. One molecular change associated with prenatal exposure to air pollutants is DNA methylation (DNAm), which has been associated with changes in placenta and cord blood tissues at birth. However, little is known about how air pollution exposure impacts the sperm epigenome, which could provide important insights into the mechanism of transmission to offspring. In the present study, we explored whether exposure to particulate matter less than 2.5 microns in diameter, particulate matter less than 10 microns in diameter, nitrogen dioxide (NO2), or ozone (O3) was associated with DNAm in sperm contributed by participants in the Early Autism Risk Longitudinal Investigation prospective pregnancy cohort. Air pollution exposure measurements were calculated as the average exposure for each pollutant measured within 4 weeks prior to the date of sample collection. Using array-based genome-scale methylation analyses, we identified 80, 96, 35, and 67 differentially methylated regions (DMRs) significantly associated with particulate matter less than 2.5 microns in diameter, particulate matter less than 10 microns in diameter, NO2, and O3, respectively. While no DMRs were associated with exposure to all four pollutants, we found that genes overlapping exposure-related DMRs had a shared enrichment for gene ontology biological processes related to neurodevelopment. Together, these data provide compelling support for the hypothesis that paternal exposure to air pollution impacts DNAm in sperm, particularly in regions implicated in neurodevelopment.
Collapse
Affiliation(s)
- Rose Schrott
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Jason I Feinberg
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Craig J Newschaffer
- Department of Biobehavioral Health, College of Health and Human Development, Pennsylvania State University, State College, PA 16802, USA
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, MIND (Medical Investigations of Neurodevelopmental Disorders) Institute, University of California, Davis, CA 95616, USA
| | - Lisa A Croen
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| | - M Daniele Fallin
- Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Heather E Volk
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Christine Ladd-Acosta
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Andrew P Feinberg
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Center for Epigenetics, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
35
|
Jansen E, Marceau K, Sellers R, Chen T, Garfield CF, Leve LD, Neiderhiser JM, Spotts EL, Roary M. The role of fathers in child development from preconception to postnatal influences: Opportunities for the National Institutes of Health Environmental influences on Child Health Outcomes (ECHO) program. Dev Psychobiol 2024; 66:e22451. [PMID: 38388196 PMCID: PMC10902630 DOI: 10.1002/dev.22451] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/21/2023] [Accepted: 12/04/2023] [Indexed: 02/24/2024]
Abstract
A growing body of literature highlights the important role of paternal health and socioemotional characteristics in child development, from preconception through adolescence. Much of this research addresses the indirect effects of fathers, for instance, their influence on maternal behaviors during the prenatal period or via the relationship with their partner. However, emerging evidence also recognizes the direct role of paternal health and behavior for child health and adjustment across development. This critical review presents evidence of biological and sociocultural influences of fathers on preconception, prenatal, and postnatal contributions to child development. The National Institutes of Health Environmental influences on Child Health Outcomes (ECHO) program incorporates in its central conceptualization the impact of fathers on family and child outcomes. This critical synthesis of the literature focuses on three specific child outcomes in the ECHO program: health outcomes (e.g., obesity), neurodevelopmental outcomes (e.g., emotional, behavioral, psychopathological development), and positive health. We highlight the unique insights gained from the literature to date and provide next steps for future studies on paternal influences.
Collapse
Affiliation(s)
- Elena Jansen
- Division of Child & Adolescent Psychiatry, Department
of Psychiatry & Behavioral Sciences, Johns Hopkins University School of
Medicine, Baltimore, Maryland, USA
| | - Kristine Marceau
- Department of Human Development and Family Studies, Purdue
University, West Lafayette, Indiana, USA
| | - Ruth Sellers
- Faculty of Education, University of Cambridge, Cambridge,
UK
| | - Tong Chen
- Department of Psychology, The Pennsylvania State
University, University Park, Pennsylvania, USA
| | - Craig F. Garfield
- Lurie Children’s Hospital of Chicago, Chicago,
Illinois, USA
- Northwestern University, Chicago, Illinois, USA
| | - Leslie D. Leve
- Prevention Science Institute, University of Oregon, Eugene,
Oregon, USA
| | - Jenae M. Neiderhiser
- Department of Psychology, The Pennsylvania State
University, University Park, Pennsylvania, USA
| | - Erica L. Spotts
- Office of Behavioral and Social Sciences Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Mary Roary
- Substance Abuse and Mental Health Service Administration,
United States Department of Health & Human Services, Rockville, Maryland,
USA
| | | |
Collapse
|
36
|
Kimmins S, Anderson RA, Barratt CLR, Behre HM, Catford SR, De Jonge CJ, Delbes G, Eisenberg ML, Garrido N, Houston BJ, Jørgensen N, Krausz C, Lismer A, McLachlan RI, Minhas S, Moss T, Pacey A, Priskorn L, Schlatt S, Trasler J, Trasande L, Tüttelmann F, Vazquez-Levin MH, Veltman JA, Zhang F, O'Bryan MK. Frequency, morbidity and equity - the case for increased research on male fertility. Nat Rev Urol 2024; 21:102-124. [PMID: 37828407 DOI: 10.1038/s41585-023-00820-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/14/2023]
Abstract
Currently, most men with infertility cannot be given an aetiology, which reflects a lack of knowledge around gamete production and how it is affected by genetics and the environment. A failure to recognize the burden of male infertility and its potential as a biomarker for systemic illness exists. The absence of such knowledge results in patients generally being treated as a uniform group, for whom the strategy is to bypass the causality using medically assisted reproduction (MAR) techniques. In doing so, opportunities to prevent co-morbidity are missed and the burden of MAR is shifted to the woman. To advance understanding of men's reproductive health, longitudinal and multi-national centres for data and sample collection are essential. Such programmes must enable an integrated view of the consequences of genetics, epigenetics and environmental factors on fertility and offspring health. Definition and possible amelioration of the consequences of MAR for conceived children are needed. Inherent in this statement is the necessity to promote fertility restoration and/or use the least invasive MAR strategy available. To achieve this aim, protocols must be rigorously tested and the move towards personalized medicine encouraged. Equally, education of the public, governments and clinicians on the frequency and consequences of infertility is needed. Health options, including male contraceptives, must be expanded, and the opportunities encompassed in such investment understood. The pressing questions related to male reproductive health, spanning the spectrum of andrology are identified in the Expert Recommendation.
Collapse
Affiliation(s)
- Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- The Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- The Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, Quebec, Canada
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Christopher L R Barratt
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Hermann M Behre
- Center for Reproductive Medicine and Andrology, University Hospital, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sarah R Catford
- Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, The Royal Women's Hospital, Melbourne, Victoria, Australia
| | | | - Geraldine Delbes
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Sante Biotechnologie, Laval, Quebec, Canada
| | - Michael L Eisenberg
- Department of Urology and Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - Nicolas Garrido
- IVI Foundation, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Brendan J Houston
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia
| | - Niels Jørgensen
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Csilla Krausz
- Department of Experimental and Clinical Biomedical Sciences, 'Mario Serio', University of Florence, University Hospital of Careggi Florence, Florence, Italy
| | - Ariane Lismer
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Robert I McLachlan
- Hudson Institute of Medical Research and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
- Monash IVF Group, Richmond, Victoria, Australia
| | - Suks Minhas
- Department of Surgery and Cancer Imperial, London, UK
| | - Tim Moss
- Healthy Male and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Allan Pacey
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Lærke Priskorn
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Stefan Schlatt
- Centre for Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Jacquetta Trasler
- Departments of Paediatrics, Human Genetics and Pharmacology & Therapeutics, McGill University and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Leonardo Trasande
- Center for the Investigation of Environmental Hazards, Department of Paediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Mónica Hebe Vazquez-Levin
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Fundación IBYME, Buenos Aires, Argentina
| | - Joris A Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
37
|
Feinberg JI, Schrott R, Ladd-Acosta C, Newschaffer CJ, Hertz-Picciotto I, Croen LA, Daniele Fallin M, Feinberg AP, Volk HE. Epigenetic changes in sperm are associated with paternal and child quantitative autistic traits in an autism-enriched cohort. Mol Psychiatry 2024; 29:43-53. [PMID: 37100868 DOI: 10.1038/s41380-023-02046-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/28/2023]
Abstract
There is a need to consider paternal contributions to autism spectrum disorder (ASD) more strongly. Autism etiology is complex, and heritability is not explained by genetics alone. Understanding paternal gametic epigenetic contributions to autism could help fill this knowledge gap. In the present study, we explored whether paternal autistic traits, and the sperm epigenome, were associated with autistic traits in children at 36 months enrolled in the Early Autism Risk Longitudinal Investigation (EARLI) cohort. EARLI is a pregnancy cohort that recruited and enrolled pregnant women in the first half of pregnancy who already had a child with ASD. After maternal enrollment, EARLI fathers were approached and asked to provide a semen specimen. Participants were included in the present study if they had genotyping, sperm methylation data, and Social Responsiveness Scale (SRS) score data available. Using the CHARM array, we performed genome-scale methylation analyses on DNA from semen samples contributed by EARLI fathers. The SRS-a 65-item questionnaire measuring social communication deficits on a quantitative scale-was used to evaluate autistic traits in EARLI fathers (n = 45) and children (n = 31). We identified 94 significant child SRS-associated differentially methylated regions (DMRs), and 14 significant paternal SRS-associated DMRs (fwer p < 0.05). Many child SRS-associated DMRs were annotated to genes implicated in ASD and neurodevelopment. Six DMRs overlapped across the two outcomes (fwer p < 0.1), and, 16 DMRs overlapped with previous child autistic trait findings at 12 months of age (fwer p < 0.05). Child SRS-associated DMRs contained CpG sites independently found to be differentially methylated in postmortem brains of individuals with and without autism. These findings suggest paternal germline methylation is associated with autistic traits in 3-year-old offspring. These prospective results for autism-associated traits, in a cohort with a family history of ASD, highlight the potential importance of sperm epigenetic mechanisms in autism.
Collapse
Affiliation(s)
- Jason I Feinberg
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rose Schrott
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Ladd-Acosta
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Craig J Newschaffer
- Department of Biobehavioral Health, College of Health and Human Development, Pennsylvania State University, State College, PA, USA
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, MIND (Medical Investigations of Neurodevelopmental Disorders) Institute, University of California, Davis, CA, USA
| | - Lisa A Croen
- Autism Research Program, Division of Research, Kaiser Permanente, Oakland, CA, USA
| | - M Daniele Fallin
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Andrew P Feinberg
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Center for Epigenetics, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Heather E Volk
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
38
|
Karahan G, Martel J, Rahimi S, Farag M, Matias F, MacFarlane AJ, Chan D, Trasler J. Higher incidence of embryonic defects in mouse offspring conceived with assisted reproduction from fathers with sperm epimutations. Hum Mol Genet 2023; 33:48-63. [PMID: 37740387 PMCID: PMC10729866 DOI: 10.1093/hmg/ddad160] [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: 05/31/2023] [Revised: 08/30/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023] Open
Abstract
Assisted reproductive technologies (ART) account for 1-6% of births in developed countries. While most children conceived are healthy, increases in birth and genomic imprinting defects have been reported; such abnormal outcomes have been attributed to underlying parental infertility and/or the ART used. Here, we assessed whether paternal genetic and lifestyle factors, that are associated with male infertility and affect the sperm epigenome, can influence ART outcomes. We examined how paternal factors, haploinsufficiency for Dnmt3L, an important co-factor for DNA methylation reactions, and/or diet-induced obesity, in combination with ART (superovulation, in vitro fertilization, embryo culture and embryo transfer), could adversely influence embryo development and DNA methylation patterning in mice. While male mice fed high-fat diets (HFD) gained weight and showed perturbed metabolic health, their sperm DNA methylation was minimally affected by the diet. In contrast, Dnmt3L haploinsufficiency induced a marked loss of DNA methylation in sperm; notably, regions affected were associated with neurodevelopmental pathways and enriched in young retrotransposons, sequences that can have functional consequences in the next generation. Following ART, placental imprinted gene methylation and growth parameters were impacted by one or both paternal factors. For embryos conceived by natural conception, abnormality rates were similar for WT and Dnmt3L+/- fathers. In contrast, paternal Dnmt3L+/- genotype, as compared to WT fathers, resulted in a 3-fold increase in the incidence of morphological abnormalities in embryos generated by ART. Together, the results indicate that embryonic morphological and epigenetic defects associated with ART may be exacerbated in offspring conceived by fathers with sperm epimutations.
Collapse
Affiliation(s)
- Gurbet Karahan
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Josée Martel
- Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Sophia Rahimi
- Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Mena Farag
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Fernando Matias
- Nutrition Research Division, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | | | - Donovan Chan
- Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Jacquetta Trasler
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
- Department of Pediatrics, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| |
Collapse
|
39
|
Stratakis CA. Genes and environment: An old pair in a new era. Maturitas 2023; 178:107851. [PMID: 37806009 DOI: 10.1016/j.maturitas.2023.107851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/16/2023] [Indexed: 10/10/2023]
Abstract
What is the relationship between our genes and the environment we live in with regard to health? Like the debate about nature or nurture in the determination of our personality and behavior, the issue of genes and environment has been discussed intensely in the last two centuries. Is it Darwin or Lamarck who is right about the basic determinants of our health, especially as we age in a rapidly changing environment? Evolutionary biology as proposed by Darwin with natural selection at its core may not be able to explain almost instant adjustments of phenotypic traits to the pressures of the environment. Epigenesis, a concept that dates from Aristotle, provides a mechanism for the environment to affect variation in genetic traits that may become heritable. Indeed, Lamarck first described the inheritance of acquired characteristics. Thus, it appears that in contemporary genetics, both Darwin and Lamarck are right: environmental pressures may affect our genes through epigenetics, in ways that allow for inheritance of the changes, a Lamarckian concept; however, evolution through natural selection is the basis for incorporation (or rejection) of new traits and their sustained inheritance, a Darwinian concept. In this review, we present the synthesis of Darwin's and Lamarck's theories, the only way to understand how our health, and that of our progeny, responds to challenging and fast-changing environmental cues. In addition, we present other examples of environment-driven changes in disease frequency or expression.
Collapse
Affiliation(s)
- Constantine A Stratakis
- NIH Clinical Center, NICHD, NIH, Bethesda, MD, USA; Research, Human Genetics & Precision Medicine, IMBB, FORTH, Heraklion, Greece; Medical Genetics, H. Dunant Hospital, Athens, Greece; Science Board, ELPEN Research Institute, Athens, Greece; European University of Cyprus, Medical School, Nicosia, Cyprus.
| |
Collapse
|
40
|
Santos-Pereira M, Pereira SC, Rebelo I, Spadella MA, Oliveira PF, Alves MG. Decoding the Influence of Obesity on Prostate Cancer and Its Transgenerational Impact. Nutrients 2023; 15:4858. [PMID: 38068717 PMCID: PMC10707940 DOI: 10.3390/nu15234858] [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: 10/18/2023] [Revised: 11/12/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
In recent decades, the escalating prevalence of metabolic disorders, notably obesity and being overweight, has emerged as a pressing concern in public health. Projections for the future indicate a continual upward trajectory in obesity rates, primarily attributable to unhealthy dietary patterns and sedentary lifestyles. The ramifications of obesity extend beyond its visible manifestations, intricately weaving a web of hormonal dysregulation, chronic inflammation, and oxidative stress. This nexus of factors holds particular significance in the context of carcinogenesis, notably in the case of prostate cancer (PCa), which is a pervasive malignancy and a leading cause of mortality among men. A compelling hypothesis arises from the perspective of transgenerational inheritance, wherein genetic and epigenetic imprints associated with obesity may wield influence over the development of PCa. This review proposes a comprehensive exploration of the nuanced mechanisms through which obesity disrupts prostate homeostasis and serves as a catalyst for PCa initiation. Additionally, it delves into the intriguing interplay between the transgenerational transmission of both obesity-related traits and the predisposition to PCa. Drawing insights from a spectrum of sources, ranging from in vitro and animal model research to human studies, this review endeavors to discuss the intricate connections between obesity and PCa. However, the landscape remains partially obscured as the current state of knowledge unveils only fragments of the complex mechanisms linking these phenomena. As research advances, unraveling the associated factors and underlying mechanisms promises to unveil novel avenues for understanding and potentially mitigating the nexus between obesity and the development of PCa.
Collapse
Affiliation(s)
- Mariana Santos-Pereira
- iBiMED-Institute of Biomedicine and Department of Medical Science, University of Aveiro, 3810-193 Aveiro, Portugal;
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal;
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal
| | - Sara C. Pereira
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal;
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal
- LAQV-REQUIMTE and Department of Chemistry, Campus Universitario de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Irene Rebelo
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biologic Sciences, Pharmaceutical Faculty, University of Porto, 4050-313 Porto, Portugal;
| | - Maria A. Spadella
- Human Embryology Laboratory, Marília Medical School, Marília 17519-030, SP, Brazil;
| | - Pedro F. Oliveira
- LAQV-REQUIMTE and Department of Chemistry, Campus Universitario de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Marco G. Alves
- iBiMED-Institute of Biomedicine and Department of Medical Science, University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
41
|
Olotu O, Ahmedani A, Kotaja N. Small Non-Coding RNAs in Male Reproduction. Semin Reprod Med 2023; 41:213-225. [PMID: 38346711 DOI: 10.1055/s-0044-1779726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Male reproductive functions are strictly regulated in order to maintain sperm production and fertility. All processes are controlled by precise regulation of gene expression, which creates specific gene expression programs for different developmental stages and cell types, and forms the functional basis for the reproductive system. Small non-coding RNAs (sncRNAs) are involved in gene regulation by targeting mRNAs for translational repression and degradation through complementary base pairing to recognize their targets. This review article summarizes the current knowledge on the function of different classes of sncRNAs, in particular microRNAs (miRNAs) and PIWI-interacting RNAs (piRNAs), during male germ cell differentiation, with the focus on sncRNAs expressed in the germline. Although transcriptionally inactive, mature spermatozoa contain a complex population of sncRNAs, and we also discuss the recently identified role of sperm sncRNAs in the intergenerational transmission of epigenetic information on father's environmental and lifestyle exposures to offspring. Finally, we summarize the current information on the utility of sncRNAs as potential biomarkers of infertility that may aid in the diagnosis and prediction of outcomes of medically assisted reproduction.
Collapse
Affiliation(s)
- Opeyemi Olotu
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ammar Ahmedani
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Noora Kotaja
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
42
|
Horne BD. Neighborhood Deprivation Measures and DNA Methylation Clocks-Understanding the Real Needs of Each Person. JAMA Netw Open 2023; 6:e2344688. [PMID: 38019521 DOI: 10.1001/jamanetworkopen.2023.44688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Affiliation(s)
- Benjamin D Horne
- Intermountain Heart Institute, Intermountain Medical Center, Salt Lake City, Utah
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| |
Collapse
|
43
|
Abstract
Obesity is a common complex trait that elevates the risk for various diseases, including type 2 diabetes and cardiovascular disease. A combination of environmental and genetic factors influences the pathogenesis of obesity. Advances in genomic technologies have driven the identification of multiple genetic loci associated with this disease, ranging from studying severe onset cases to investigating common multifactorial polygenic forms. Additionally, findings from epigenetic analyses of modifications to the genome that do not involve changes to the underlying DNA sequence have emerged as key signatures in the development of obesity. Such modifications can mediate the effects of environmental factors, including diet and lifestyle, on gene expression and clinical presentation. This review outlines what is known about the genetic and epigenetic contributors to obesity susceptibility, along with the albeit limited therapeutic options currently available. Furthermore, we delineate the potential mechanisms of actions through which epigenetic changes can mediate environmental influences and the related opportunities they present for future interventions in the management of obesity.
Collapse
Affiliation(s)
- Khanh Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Diabetes and Endocrinology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
44
|
Cimadomo D, Rienzi L, Conforti A, Forman E, Canosa S, Innocenti F, Poli M, Hynes J, Gemmell L, Vaiarelli A, Alviggi C, Ubaldi FM, Capalbo A. Opening the black box: why do euploid blastocysts fail to implant? A systematic review and meta-analysis. Hum Reprod Update 2023; 29:570-633. [PMID: 37192834 DOI: 10.1093/humupd/dmad010] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/22/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND A normal chromosomal constitution defined through PGT-A assessing all chromosomes on trophectoderm (TE) biopsies represents the strongest predictor of embryo implantation. Yet, its positive predictive value is not higher than 50-60%. This gap of knowledge on the causes of euploid blastocysts' reproductive failure is known as 'the black box of implantation'. OBJECTIVE AND RATIONALE Several embryonic, maternal, paternal, clinical, and IVF laboratory features were scrutinized for their putative association with reproductive success or implantation failure of euploid blastocysts. SEARCH METHODS A systematic bibliographical search was conducted without temporal limits up to August 2021. The keywords were '(blastocyst OR day5 embryo OR day6 embryo OR day7 embryo) AND (euploid OR chromosomally normal OR preimplantation genetic testing) AND (implantation OR implantation failure OR miscarriage OR abortion OR live birth OR biochemical pregnancy OR recurrent implantation failure)'. Overall, 1608 items were identified and screened. We included all prospective or retrospective clinical studies and randomized-controlled-trials (RCTs) that assessed any feature associated with live-birth rates (LBR) and/or miscarriage rates (MR) among non-mosaic euploid blastocyst transfer after TE biopsy and PGT-A. In total, 41 reviews and 372 papers were selected, clustered according to a common focus, and thoroughly reviewed. The PRISMA guideline was followed, the PICO model was adopted, and ROBINS-I and ROB 2.0 scoring were used to assess putative bias. Bias across studies regarding the LBR was also assessed using visual inspection of funnel plots and the trim and fill method. Categorical data were combined with a pooled-OR. The random-effect model was used to conduct the meta-analysis. Between-study heterogeneity was addressed using I2. Whenever not suitable for the meta-analysis, the included studies were simply described for their results. The study protocol was registered at http://www.crd.york.ac.uk/PROSPERO/ (registration number CRD42021275329). OUTCOMES We included 372 original papers (335 retrospective studies, 30 prospective studies and 7 RCTs) and 41 reviews. However, most of the studies were retrospective, or characterized by small sample sizes, thus prone to bias, which reduces the quality of the evidence to low or very low. Reduced inner cell mass (7 studies, OR: 0.37, 95% CI: 0.27-0.52, I2 = 53%), or TE quality (9 studies, OR: 0.53, 95% CI: 0.43-0.67, I2 = 70%), overall blastocyst quality worse than Gardner's BB-grade (8 studies, OR: 0.40, 95% CI: 0.24-0.67, I2 = 83%), developmental delay (18 studies, OR: 0.56, 95% CI: 0.49-0.63, I2 = 47%), and (by qualitative analysis) some morphodynamic abnormalities pinpointed through time-lapse microscopy (abnormal cleavage patterns, spontaneous blastocyst collapse, longer time of morula formation I, time of blastulation (tB), and duration of blastulation) were all associated with poorer reproductive outcomes. Slightly lower LBR, even in the context of PGT-A, was reported among women ≥38 years (7 studies, OR: 0.87, 95% CI: 0.75-1.00, I2 = 31%), while obesity was associated with both lower LBR (2 studies, OR: 0.66, 95% CI: 0.55-0.79, I2 = 0%) and higher MR (2 studies, OR: 1.8, 95% CI: 1.08-2.99, I2 = 52%). The experience of previous repeated implantation failures (RIF) was also associated with lower LBR (3 studies, OR: 0.72, 95% CI: 0.55-0.93, I2 = 0%). By qualitative analysis, among hormonal assessments, only abnormal progesterone levels prior to transfer were associated with LBR and MR after PGT-A. Among the clinical protocols used, vitrified-warmed embryo transfer was more effective than fresh transfer (2 studies, OR: 1.56, 95% CI: 1.05-2.33, I2 = 23%) after PGT-A. Lastly, multiple vitrification-warming cycles (2 studies, OR: 0.41, 95% CI: 0.22-0.77, I2 = 50%) or (by qualitative analysis) a high number of cells biopsied may slightly reduce the LBR, while simultaneous zona-pellucida opening and TE biopsy allowed better results than the Day 3 hatching-based protocol (3 studies, OR: 1.41, 95% CI: 1.18-1.69, I2 = 0%). WIDER IMPLICATIONS Embryo selection aims at shortening the time-to-pregnancy, while minimizing the reproductive risks. Knowing which features are associated with the reproductive competence of euploid blastocysts is therefore critical to define, implement, and validate safer and more efficient clinical workflows. Future research should be directed towards: (i) systematic investigations of the mechanisms involved in reproductive aging beyond de novo chromosomal abnormalities, and how lifestyle and nutrition may accelerate or exacerbate their consequences; (ii) improved evaluation of the uterine and blastocyst-endometrial dialogue, both of which represent black boxes themselves; (iii) standardization/automation of embryo assessment and IVF protocols; (iv) additional invasive or preferably non-invasive tools for embryo selection. Only by filling these gaps we may finally crack the riddle behind 'the black box of implantation'.
Collapse
Affiliation(s)
- Danilo Cimadomo
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Laura Rienzi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University, Naples, Italy
| | - Eric Forman
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Columbia University Irving Medical Centre, New York, NY, USA
| | | | - Federica Innocenti
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Maurizio Poli
- Centrum voor Kinderwens, Dijklander Hospital, Purmerend, The Netherlands
- Juno Genetics, Rome, Italy
| | - Jenna Hynes
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Columbia University Irving Medical Centre, New York, NY, USA
| | - Laura Gemmell
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Columbia University Irving Medical Centre, New York, NY, USA
| | - Alberto Vaiarelli
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Carlo Alviggi
- Department of Public Health, Federico II University, Naples, Italy
| | | | | |
Collapse
|
45
|
Oluwayiose OA, Houle E, Whitcomb BW, Suvorov A, Rahil T, Sites CK, Krawetz SA, Visconti PE, Pilsner JR. Urinary phthalate metabolites and small non-coding RNAs from seminal plasma extracellular vesicles among men undergoing infertility treatment. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121529. [PMID: 37003585 PMCID: PMC11881107 DOI: 10.1016/j.envpol.2023.121529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 02/24/2023] [Accepted: 03/27/2023] [Indexed: 05/21/2023]
Abstract
Non-coding RNA (ncRNA) cargo of extracellular vesicles (EVs) in the male reproductive tract play critical roles in semen quality and emerging evidence suggests their susceptibility to environmental factors. Male phthalate exposures have been linked to poor semen quality, sperm DNA methylation profiles and embryo development; however, there is limited evidence on their potential impact on EV ncRNAs profiles. We evaluated the association between urinary phthalate metabolites and small ncRNAs (sncRNAs) of seminal plasma EVs (spEV) among men receiving clinical infertility care. We conducted sncRNA sequencing of EVs in 96 seminal plasma samples collected from the Sperm Environmental Epigenetics and Development Study (SEEDS). Sequencing reads were mapped to human transcriptome databases using STAR. Urinary metabolite concentrations of thirteen phthalates and two DiNCH, a phthalate alternative, were measured via tandem mass spectrometry. Associations with normalized counts were assessed using EdgeR (FDR<0.05) adjusting for urinary dilution via specific gravity, age, BMI, batch, and biotype-specific total counts. Select metabolites, MEOHP, MECPP, ∑DEHP, MCPP, MCNP, MCOP, were negatively (p < 0.05) correlated with miRNA relative abundance. Similarly, nine metabolites including MEOHP, MECPP, MEHP, MCPP, MHBP, MHiNCH, MiBP, MEHHP, MCOP and ∑DEHP were associated (q < 0.05) with normalized counts from 23 unique ncRNA transcripts (7 miRNAs (pre & mature); 6 tRFs; and 10 piRNAs), most (78%) of which displayed increased expression patterns. miRNA and tRFs gene targets were enriched in vesicle-mediated transport and developmental-related ontology terms, such as tyrosine kinase, head development, and cell morphogenesis. Six genes (MAPK1, BMPR1A/2, PTEN, TGFBR2, TP53 and APP) were present in all the ontology terms and predicted to form protein association networks. piRNAs were annotated to pseudogenes of genes important in EV cargo transfer and embryonic development. This is the first study to associate phthalate exposures to altered spEV sncRNA profiles. Future studies are needed to determine their impact on reproductive outcomes.
Collapse
Affiliation(s)
- Oladele A Oluwayiose
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Emily Houle
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, 48201, 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
| | - Alexander Suvorov
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, 686 North Pleasant Street, Amherst, MA, USA
| | - Tayyab Rahil
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, UMass Chan-Baystate, 759 Chestnut Street, Springfield, MA, USA
| | - Cynthia K Sites
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, UMass Chan-Baystate, 759 Chestnut Street, Springfield, MA, USA
| | - Stephen A Krawetz
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA; Center for Molecular Medicine and Genetics, Wayne State School of Medicine, USA
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N. Pleasant St, Amherst, MA, 01003, USA
| | - J Richard Pilsner
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48201, USA.
| |
Collapse
|
46
|
Liu S, Sharma U. Sperm RNA Payload: Implications for Intergenerational Epigenetic Inheritance. Int J Mol Sci 2023; 24:5889. [PMID: 36982962 PMCID: PMC10052761 DOI: 10.3390/ijms24065889] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
There is mounting evidence that ancestral life experiences and environment can influence phenotypes in descendants. The parental environment regulates offspring phenotypes potentially via modulating epigenetic marks in the gametes. Here, we review examples of across-generational inheritance of paternal environmental effects and the current understanding of the role of small RNAs in such inheritance. We discuss recent advances in revealing the small RNA payload of sperm and how environmental conditions modulate sperm small RNAs. Further, we discuss the potential mechanism of inheritance of paternal environmental effects by focusing on sperm small RNA-mediated regulation of early embryonic gene expression and its role in influencing offspring phenotypes.
Collapse
Affiliation(s)
| | - Upasna Sharma
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| |
Collapse
|
47
|
Jyothi AK, Thotakura B, Priyadarshini SC, Patil S, Poojari MS, Subramanian M. Paternal stress alters synaptic density and expression of GAP-43, GRIN1, M1 and SYP genes in the hippocampus and cortex of offspring of stress-induced male rats. Morphologie 2023; 107:67-79. [PMID: 35715368 DOI: 10.1016/j.morpho.2022.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/16/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023]
Abstract
Adverse experiences during pregnancy have a negative impact on the neuronal structure and behavior of offspring, but the effects of a father's life events on the outcome of progeny are scarce. The present study is intended to investigate whether paternal stress affects the offspring brain structure, especially those regions concerned with learning and formation of memory, namely the hippocampus (HC) and prefrontal cortex (PFC), and also the expression of certain genes linked to learning and memory in the offspring. Induced stress to male rats by five stressors, one per day followed by allowing them to mate with the normal, unstressed female. Synaptophysin immunoreactivity was assessed in the tissue sections of the HC and PFC as well as expression of genes concerned with learning and memory was evaluated by RT-PCR in the progeny of stress-received males. The progeny of stressed rats had reduced antisynaptophysin immunoreactivity in the HC and PFC. The synaptic density in HC was less in the A-S (Offspring of male rats who received stress during adulthood) and PA-S (offspring of male rats who received stress during both adolescence and adulthood) than in P-S (offspring of male rats who received stress during adolescence) and C-C (offspring of control) groups. Similar results were observed even in the PFC. The results of post hoc tests proved that the HC and PFC of the progeny of stress-exposed rats exhibited considerably less synaptic density than control (P<0.05), and the levels of expression of GAP-43, GRIN1, M1, and SYP genes in HC and PFC were down-regulated. This study concludes that paternal adverse experiences can affect the offspring's synaptic plasticity and also the genes, which can regulate learning and formation of memory.
Collapse
Affiliation(s)
- A K Jyothi
- Department of Anatomy, Basaveshwara Medical College and Hospital, 577502 Chitradurga, Karnataka, India
| | - B Thotakura
- Department of Anatomy, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, 603103 Kanchipuram, Tamil Nadu, India.
| | - S C Priyadarshini
- Department of Anatomy, Tagore Medical College & Hospital, 600127 Chennai, Tamil Nadu, India
| | - S Patil
- Department of Anatomy, Basaveshwara Medical College and Hospital, 577502 Chitradurga, Karnataka, India
| | - M S Poojari
- Department of Anatomy, Basaveshwara Medical College and Hospital, 577502 Chitradurga, Karnataka, India
| | - M Subramanian
- Department of Anatomy, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, 603103 Kanchipuram, Tamil Nadu, India
| |
Collapse
|
48
|
Svanes C, Holloway JW, Krauss-Etschmann S. Preconception origins of asthma, allergies and lung function: The influence of previous generations on the respiratory health of our children. J Intern Med 2023; 293:531-549. [PMID: 36861185 DOI: 10.1111/joim.13611] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Emerging research suggests that exposures occurring years before conception are important determinants of the health of future offspring and subsequent generations. Environmental exposures of both the father and mother, or exposure to disease processes such as obesity or infections, may influence germline cells and thereby cause a cascade of health outcomes in multiple subsequent generations. There is now increasing evidence that respiratory health is influenced by parental exposures that occur long before conception. The strongest evidence relates adolescent tobacco smoking and overweight in future fathers to increased asthma and lower lung function in their offspring, supported by evidence on parental preconception occupational exposures and air pollution. Although this literature is still sparse, the epidemiological analyses reveal strong effects that are consistent across studies with different designs and methodologies. The results are strengthened by mechanistic research from animal models and (scarce) human studies that have identified molecular mechanisms that can explain the epidemiological findings, suggesting transfer of epigenetic signals through germline cells, with susceptibility windows in utero (both male and female line) and prepuberty (male line). The concept that our lifestyles and behaviours may influence the health of our future children represents a new paradigm. This raises concerns for future health in decades to come with respect to harmful exposures but may also open for radical rethinking of preventive strategies that may improve health in multiple generations, reverse the imprint of our parents and forefathers, and underpin strategies that can break the vicious circle of propagation of health inequalities across generations.
Collapse
Affiliation(s)
- Cecilie Svanes
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway.,Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
49
|
Ribas-Aulinas F, Ribo S, Casas E, Mourin-Fernandez M, Ramon-Krauel M, Diaz R, Lerin C, Kalko SG, Vavouri T, Jimenez-Chillaron JC. Intergenerational Inheritance of Hepatic Steatosis in a Mouse Model of Childhood Obesity: Potential Involvement of Germ-Line microRNAs. Nutrients 2023; 15:nu15051241. [PMID: 36904241 PMCID: PMC10005268 DOI: 10.3390/nu15051241] [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: 01/10/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Childhood obesity increases the risk of developing metabolic syndrome later in life. Moreover, metabolic dysfunction may be inherited into the following generation through non-genomic mechanisms, with epigenetics as a plausible candidate. The pathways involved in the development of metabolic dysfunction across generations in the context of childhood obesity remain largely unexplored. We have developed a mouse model of early adiposity by reducing litter size at birth (small litter group, SL: 4 pups/dam; control group, C: 8 pups/dam). Mice raised in small litters (SL) developed obesity, insulin resistance and hepatic steatosis with aging. Strikingly, the offspring of SL males (SL-F1) also developed hepatic steatosis. Paternal transmission of an environmentally induced phenotype strongly suggests epigenetic inheritance. We analyzed the hepatic transcriptome in C-F1 and SL-F1 mice to identify pathways involved in the development of hepatic steatosis. We found that the circadian rhythm and lipid metabolic process were the ontologies with highest significance in the liver of SL-F1 mice. We explored whether DNA methylation and small non-coding RNAs might be involved in mediating intergenerational effects. Sperm DNA methylation was largely altered in SL mice. However, these changes did not correlate with the hepatic transcriptome. Next, we analyzed small non-coding RNA content in the testes of mice from the parental generation. Two miRNAs (miR-457 and miR-201) appeared differentially expressed in the testes of SL-F0 mice. They are known to be expressed in mature spermatozoa, but not in oocytes nor early embryos, and they may regulate the transcription of lipogenic genes, but not clock genes, in hepatocytes. Hence, they are strong candidates to mediate the inheritance of adult hepatic steatosis in our murine model. In conclusion, litter size reduction leads to intergenerational effects through non-genomic mechanisms. In our model, DNA methylation does not seem to play a role on the circadian rhythm nor lipid genes. However, at least two paternal miRNAs might influence the expression of a few lipid-related genes in the first-generation offspring, F1.
Collapse
Affiliation(s)
| | - Sílvia Ribo
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Eduard Casas
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | | | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Susana G. Kalko
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Tanya Vavouri
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | - Josep C. Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- School of Medicine, University of Barcelona, L’Hospitalet, 08907 Barcelona, Spain
- Correspondence: or ; Tel.: +34-934024267
| |
Collapse
|
50
|
Zhang Y, Qi J, Zhao J, Li M, Zhang Y, Hu H, Wei L, Zhou K, Qin H, Qu P, Cao W, Liu E. Effect of Dietetic Obesity on Testicular Transcriptome in Cynomolgus Monkeys. Genes (Basel) 2023; 14:genes14030557. [PMID: 36980830 PMCID: PMC10048326 DOI: 10.3390/genes14030557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Obesity is a metabolic disorder resulting from behavioral, environmental and heritable causes, and can have a negative impact on male reproduction. There have been few experiments in mice, rats, and rabbits on the effects of obesity on reproduction, which has inhibited the development of better treatments for male subfertility caused by obesity. Nonhuman primates are most similar to human beings in anatomy, physiology, metabolism, and biochemistry and are appropriate subjects for obesity studies. In this investigation, we conducted a transcriptome analysis of the testes of cynomolgus monkeys on high-fat, high-fructose, and cholesterol-rich diets to determine the effect of obesity on gene expression in testes. The results showed that the testes of obese monkeys had abnormal morphology, and their testes transcriptome was significantly different from that of non-obese animals. We identified 507 differentially abundant genes (adjusted p value < 0.01, log2 [FC] > 2) including 163 up-regulated and 344 down-regulated genes. Among the differentially abundant genes were ten regulatory genes, including IRF1, IRF6, HERC5, HERC6, IFIH1, IFIT2, IFIT5, IFI35, RSAD2, and UBQLNL. Gene ontology (GO) and KEGG pathway analysis was conducted, and we found that processes and pathways associated with the blood testes barrier (BTB), immunity, inflammation, and DNA methylation in gametes were preferentially enriched. We also found abnormal expression of genes related to infertility (TDRD5, CLCN2, MORC1, RFX8, SOHLH1, IL2RB, MCIDAS, ZPBP, NFIA, PTPN11, TSC22D3, MAPK6, PLCB1, DCUN1D1, LPIN1, and GATM) and down-regulation of testosterone in monkeys with dietetic obesity. This work not only provides an important reference for research and treatment on male infertility caused by obesity, but also valuable insights into the effects of diet on gene expression in testes.
Collapse
Affiliation(s)
- Yanru Zhang
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
| | - Jia Qi
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
| | - Juan Zhao
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
- Department of Hematology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Miaojing Li
- Department of Hematology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yulin Zhang
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
| | - Huizhong Hu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
| | - Liangliang Wei
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
| | - Kai Zhou
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
| | - Hongyu Qin
- Precision Medicine Center, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Pengxiang Qu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
| | - Wenbin Cao
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an 710049, China
- Correspondence: (W.C.); (E.L.)
| | - Enqi Liu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
- Correspondence: (W.C.); (E.L.)
| |
Collapse
|