1
|
Capobianco E, Pirrone I. The POHaD paradigm: role of the placenta in paternal programming. Placenta 2025:S0143-4004(25)00138-9. [PMID: 40350327 DOI: 10.1016/j.placenta.2025.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/31/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
The Paternal Origins of Health and Disease (POHaD) paradigm emerges from the well-known Developmental Origins of Health and Disease (DOHaD) concept. Research into the programming of metabolic diseases originating from the paternal germline began over 20 years ago, focusing on the father's pre-conceptional exposure, such as metabolic disorders and lifestyle habits (kind of diet, exercise, smoking, drugs consumption, etc.). This exposure can lead to epigenetic marks not only in his germ cells but also in other components of the semen that impact the health of future generations. The significant role of the paternal genome in the fetal component of the placenta and the recognition of the placenta's involvement in postnatal disease programming underscore the importance of studying the placenta in paternal programming research. The aim of this work is to review what we know so far about paternal programming highlighting the variations in the phenotype of the placenta and the influence of it in the programming of metabolic pathologies in the offspring of fathers exposed to metabolic disorders such as obesity and diabetes.
Collapse
Affiliation(s)
- Evangelina Capobianco
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina.
| | - Irune Pirrone
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| |
Collapse
|
2
|
Li Y, Liu L, Zhang Y, Bai S, Jiang Y, Lai C, Li X, Bai W. Paternal Cyanidin-3-O-Glucoside Diet Improved High-Fat, High-Fructose Diet-Induced Intergenerational Inheritance in Male Offspring's Susceptibility to High-Fat Diet-Induced Testicular and Sperm Damage. Reprod Sci 2025; 32:1102-1114. [PMID: 39836315 DOI: 10.1007/s43032-024-01780-9] [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: 05/24/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
High-fructose and high-fat diet (HFHFD) has been associated with impaired spermatogenesis, leading to decreased sperm quality and increased male infertility, with similar effects observed in offspring. Cyanidin-3-O-glucoside (C3G), a recognized food antioxidant, has shown promise in protecting in male reproduction and modulating epigenetic modifications. However, its potential role in ameliorating intergenerational inheritance induced by HFHFD remains underexplored. In this study, we investigated the effects of paternal HFHFD on reproductive injury of offspring and the protective effect of C3G. Paternal mice were subjected to 12 weeks of HFHFD induction and C3G treatment was conducted for 8 weeks. Offspring obtained via in vitro fertilization were fed either a normal diet (ND) or high-fat diet (HFD). Our findings indicate that while the paternal HFHFD did not result in observable reproductive impairments in paternal mice, it did affect offspring testicular function through intergenerational inheritance, rendering them more susceptible to testicular damage and reduced sperm counts when exposed to an HFD. Notably, C3G intervention significantly mitigated these effects, suggesting its potential as a therapeutic compound for alleviating the impact of paternal intergenerational inheritance on male fertility resulting from HFHFD. These results underscore the importance of further exploring the mechanisms underlying intergenerational inheritance and the potential of interventions such as C3G in mitigating its effects, with implications for both basic research and clinical practice.
Collapse
Affiliation(s)
- Yuxi Li
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, 601 Huangpu Rd, Guangzhou, 510632, PR China
| | - Liwang Liu
- The First Clinical Medical College of Jinan University, Guangzhou, 510632, PR China
| | - Yulin Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, 601 Huangpu Rd, Guangzhou, 510632, PR China
| | - Shun Bai
- Center for Reproduction and Genetics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, PR China
| | - Yan Jiang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, 601 Huangpu Rd, Guangzhou, 510632, PR China
| | - Caiyong Lai
- The Sixth Affiliated Hospital of Jinan University, Dongguan, 523576, PR China
| | - Xusheng Li
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, 601 Huangpu Rd, Guangzhou, 510632, PR China.
- The Sixth Affiliated Hospital of Jinan University, Dongguan, 523576, PR China.
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, 601 Huangpu Rd, Guangzhou, 510632, PR China.
| |
Collapse
|
3
|
Godden AM, Silva WTAF, Kiehl B, Jolly C, Folkes L, Alavioon G, Immler S. Environmentally induced variation in sperm sRNAs is linked to gene expression and transposable elements in zebrafish offspring. Heredity (Edinb) 2025; 134:234-246. [PMID: 40121340 PMCID: PMC11977266 DOI: 10.1038/s41437-025-00752-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
Environmental factors affect not only paternal condition but may translate into the following generations where sperm-mediated small RNAs (sRNAs) can contribute to the transmission of paternal effects. sRNAs play a key role in the male germ line in genome maintenance and repair, and particularly in response to environmental stress and the resulting increase in transposable element (TE) activity. Here, we investigated how the social environment (high competition, low competition) of male zebrafish Danio rerio affects sRNAs in sperm and how these are linked to gene expression and TE activity in their offspring. In a first experiment, we collected sperm samples after exposing males to each social environment for 2 weeks to test for differentially expressed sperm micro- (miRNA) and piwi-interacting RNAs (piRNA). In a separate experiment, we performed in vitro fertilisations after one 2-week period using a split-clutch design to control for maternal effects and collected embryos at 24 h to test for differentially expressed genes and TEs. We developed new computational prediction tools to link sperm sRNAs with differentially expressed TEs and genes in the embryos. Our results support the idea that the molecular stress response in the male germ line has significant down-stream effects on the molecular pathways, and we provide a direct link between sRNAs, TEs and gene expression.
Collapse
Affiliation(s)
- Alice M Godden
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Willian T A F Silva
- Uppsala University, Department of Evolutionary Biology, Norbyvägen 18D, 75310, Uppsala, Sweden
- Department of Physics, Chemistry and Biology, Linköping University, 58183, Linköping, Sweden
| | - Berrit Kiehl
- Uppsala University, Department of Evolutionary Biology, Norbyvägen 18D, 75310, Uppsala, Sweden
| | - Cécile Jolly
- Uppsala University, Department of Evolutionary Biology, Norbyvägen 18D, 75310, Uppsala, Sweden
| | - Leighton Folkes
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Ghazal Alavioon
- Uppsala University, Department of Evolutionary Biology, Norbyvägen 18D, 75310, Uppsala, Sweden
| | - Simone Immler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
- Uppsala University, Department of Evolutionary Biology, Norbyvägen 18D, 75310, Uppsala, Sweden.
| |
Collapse
|
4
|
Inoue Y, Yokoyama M, Inoue S, Imai M, Onji H, Yano A, Uchikura Y, Matsubara Y, Matsubara K, Hamada H, Tomita H, Iwama N, Watanabe Z, Ishikuro M, Obara T, Metoki H, Ota C, Kuriyama S, Arima T, Yaegashi N, Saito M, Sugiyama T. Association between paternal physique and obesity in children at the age of 3 years: the Japan Environment and Children's Study. J Dev Orig Health Dis 2025; 16:e17. [PMID: 40116036 DOI: 10.1017/s2040174424000473] [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: 03/23/2025]
Abstract
Obesity during development has been reported to be a determinant factor in the future development of non-communicable diseases (NCDs). Parental obesity is suggested to be a predictor of children's obesity, and it is important to consider parental factors to prevent NCDs in the progeny. Previously, we showed that paternal height had a stronger association with infant birth weight than paternal body mass index (BMI) in the Japanese population. However, only a few studies have examined the association between paternal physique and postnatal obesity. This study aimed to investigate the association between parental physique and obesity in children at the age of 3. This study used fixed data on 33,291 parent-child pairs from the Japan Environment and Children's Study, an ongoing national birth cohort study. The association between paternal physique (BMI and height) and children's obesity at the age of 3 was examined using multivariate logistic regression analysis. The higher the paternal BMI quartiles, the higher the odds ratio for obesity in male and female children at 3 years of age (P < 0.0001). However, paternal height quartiles were not associated with male or female obesity. These results differ from the association between paternal physique and infant birth weight, and it is possible that prenatal epigenetic and environmental factors of paternal origin were responsible for the differences between these two studies. The association between paternal BMI and obesity in children at the age of 3 suggests that paternal factors may be involved in the development of NCDs in future progeny.
Collapse
Affiliation(s)
- Yui Inoue
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Maki Yokoyama
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shota Inoue
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Matome Imai
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Hiroshi Onji
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Akiko Yano
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yuka Uchikura
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yuko Matsubara
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Keiichi Matsubara
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Hirotaka Hamada
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hasumi Tomita
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Noriyuki Iwama
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Zen Watanabe
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Mami Ishikuro
- Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Division of Molecular Epidemiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Taku Obara
- Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Division of Molecular Epidemiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hirohito Metoki
- Division of Public Health, Hygiene and Epidemiology, Tohoku Medical Pharmaceutical University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Chiharu Ota
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shinichi Kuriyama
- Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Division of Molecular Epidemiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Miyagi, Japan
| | - Takahiro Arima
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Nobuo Yaegashi
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masatoshi Saito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Takashi Sugiyama
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
5
|
Nohara K, Suzuki T, Okamura K, Kawai T, Nakabayashi K. Acquired sperm hypomethylation by gestational arsenic exposure is re-established in both the paternal and maternal genomes of post-epigenetic reprogramming embryos. Epigenetics Chromatin 2025; 18:4. [PMID: 39815295 PMCID: PMC11737231 DOI: 10.1186/s13072-025-00569-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND DNA methylation plays a crucial role in mammalian development. While methylome changes acquired in the parental genomes are believed to be erased by epigenetic reprogramming, accumulating evidence suggests that methylome changes in sperm caused by environmental factors are involved in the disease phenotypes of the offspring. These findings imply that acquired sperm methylome changes are transferred to the embryo after epigenetic reprogramming. However, our understanding of this process remains incomplete. Our previous study showed that arsenic exposure of F0 pregnant mice paternally increased tumor incidence in F2 offspring. The sperm methylome of arsenic-exposed F1 males exhibited characteristic features, including enrichment of hypomethylated cytosines at the promoters of retrotransposons LINEs and LTRs. Hypomethylation of retrotransposons is potentially detrimental. Determining whether these hypomethylation changes in sperm are transferred to the embryo is important in confirming the molecular pathway of intergenerational transmission of paternal effects of arsenic exposure. RESULTS We investigated the methylome of F2 male embryos after epigenetic reprogramming by reduced representation bisulfite sequencing (RRBS) and allele-specific analysis. To do so, embryos were obtained by crossing control or gestationally arsenic-exposed F1 males (C3H/HeN strain) with control females (C57BL/6 strain). The results revealed that the methylome of F2 embryos in the arsenic group was globally hypomethylated and enriched for hypomethylated cytosines in certain genomic regions, including LTR and LINE, as observed in F1 sperm of the arsenic group. Unexpectedly, the characteristic methylome features were detected not only in the paternal genome but also in the maternal genome of embryos. Furthermore, these methylation changes were found to rarely occur at the same positions between F1 sperm and F2 embryos. CONCLUSIONS The results of this study revealed that the characteristics of arsenic-induced methylome changes in F1 sperm are reproduced in both the paternal and maternal genomes of post-epigenetic reprogramming embryos. Furthermore, the results suggest that this re-establishment is achieved in collaboration with other factors that mediate region-specific methylation changes. These results also highlight the possibility that arsenic-induced sperm methylome changes could contribute to the development of disease predisposition in offspring.
Collapse
Affiliation(s)
- Keiko Nohara
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, 305‑ 8506, Japan.
| | - Takehiro Suzuki
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, 305‑ 8506, Japan
| | - Kazuyuki Okamura
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, 305‑ 8506, Japan
| | - Tomoko Kawai
- Department of Maternal‑Fetal Biology, National Center for Child Health and Development, Tokyo, 157‑8535, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal‑Fetal Biology, National Center for Child Health and Development, Tokyo, 157‑8535, Japan
| |
Collapse
|
6
|
Hug E, Renaud Y, Guiton R, Ben Sassi M, Marcaillou C, Moazamian A, Gharagozloo P, Drevet JR, Saez F. Exploring the Epigenetic Landscape of Spermatozoa: Impact of Oxidative Stress and Antioxidant Supplementation on DNA Methylation and Hydroxymethylation. Antioxidants (Basel) 2024; 13:1520. [PMID: 39765848 PMCID: PMC11726892 DOI: 10.3390/antiox13121520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/15/2025] Open
Abstract
Reproductive success is dependent on gamete integrity, and oxidative stress alters male nuclei, meaning that no DNA repair is possible due to chromatin compaction. The composition of sperm makes it highly sensitive to reactive oxygen species (ROS) but, at the same time, ROS are needed for sperm physiology. Over the past 30 years, much attention has been paid to the consequences of oxidative stress on sperm properties and the protective effects of antioxidant formulations to help fertility. Spermatozoa also carry epigenetic marks, critical for embryo development and the health of offspring. As DNA oxidative damage may disturb the sperm epigenome, we used an established mouse model of post-testicular sperm DNA oxidation to investigate sperm DNA methylation and hydroxymethylation. We also analyzed the potential corrective effect of oral antioxidant supplementation, proven to reduce sperm DNA oxidative damage, on sperm DNA methyl/hydroxymethyl marks. We show that sperm DNA oxidation is associated with a significant increase in overall hydroxymethylation. Oral antioxidant supplementation led to unexpected mild epigenetic changes. Antioxidant supplementation should not be proposed without proper clinical evaluation as it may alter sperm epigenetic marks, leading to a risk of paternally inherited epigenetic alterations.
Collapse
Affiliation(s)
- Elisa Hug
- GReD Institute, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France (A.M.)
- EVALSEM, Clermont Auvergne Innovation, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Yoan Renaud
- GReD Institute, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France (A.M.)
| | - Rachel Guiton
- GReD Institute, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France (A.M.)
| | - Mehdi Ben Sassi
- IntegraGen SA, Génopole Campus 1—Bât. 8, 5 Rue de Henri Desbruères, 91000 Evry, France
- Centre de Recherche des Cordeliers, INSERM UMRS1138, CNRS SNC 5096, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Charles Marcaillou
- IntegraGen SA, Génopole Campus 1—Bât. 8, 5 Rue de Henri Desbruères, 91000 Evry, France
| | - Aron Moazamian
- GReD Institute, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France (A.M.)
- EVALSEM, Clermont Auvergne Innovation, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France
- CellOxess LLC, Ewing, NJ 08638, USA
| | | | - Joël R. Drevet
- GReD Institute, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France (A.M.)
- EVALSEM, Clermont Auvergne Innovation, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Fabrice Saez
- GReD Institute, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France (A.M.)
- EVALSEM, Clermont Auvergne Innovation, Université Clermont Auvergne, Faculté de Médecine, CRBC, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France
| |
Collapse
|
7
|
Weigel Muñoz M, Cohen DJ, Da Ros VG, González SN, Rebagliati Cid A, Sulzyk V, Cuasnicu PS. Physiological and pathological aspects of epididymal sperm maturation. Mol Aspects Med 2024; 100:101321. [PMID: 39340983 DOI: 10.1016/j.mam.2024.101321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
In mammals, sperm that leave the testes are nonfunctional and require a complex post-testicular maturation process to acquire their ability to recognize and fertilize the egg. The crucial maturation changes that provide sperm their fertilizing capability occur while passing through the epididymis. Due to the widespread use of assisted reproductive technologies to address male infertility, there has been a significant decrease in research focusing on the mechanisms underlying the maturation process over the past decades. Considering that up to 40% of male infertility is idiopathic and could be reflecting sperm maturation defects, the study of post-testicular sperm maturation will clearly contribute to a better understanding of the causes of male infertility and to the development of both new approaches to maturing sperm in vitro and safer male contraceptive methods. Based on this, the present review focuses on the physiopathology of the epididymis as well as on current approaches under investigation to improve research in sperm maturation and as potential therapeutic options for male infertility.
Collapse
Affiliation(s)
- Mariana Weigel Muñoz
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Débora J Cohen
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Vanina G Da Ros
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Soledad N González
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Abril Rebagliati Cid
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Valeria Sulzyk
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Patricia S Cuasnicu
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Han C. Gene expression programs in mammalian spermatogenesis. Development 2024; 151:dev202033. [PMID: 38691389 DOI: 10.1242/dev.202033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Mammalian spermatogenesis, probably the most complex of all cellular developmental processes, is an ideal model both for studying the specific mechanism of gametogenesis and for understanding the basic rules governing all developmental processes, as it entails both cell type-specific and housekeeping molecular processes. Spermatogenesis can be viewed as a mission with many tasks to accomplish, and its success is genetically programmed and ensured by the collaboration of a large number of genes. Here, I present an overview of mammalian spermatogenesis and the mechanisms underlying each step in the process, covering the cellular and molecular activities that occur at each developmental stage and emphasizing their gene regulation in light of recent studies.
Collapse
Affiliation(s)
- Chunsheng Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100101 Beijing, China
| |
Collapse
|
11
|
Jimenez-Gonzalez A, Ansaloni F, Nebendahl C, Alavioon G, Murray D, Robak W, Sanges R, Müller F, Immler S. Paternal starvation affects metabolic gene expression during zebrafish offspring development and lifelong fitness. Mol Ecol 2024; 33:e17296. [PMID: 38361456 DOI: 10.1111/mec.17296] [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: 09/19/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 02/17/2024]
Abstract
Dietary restriction in the form of fasting is a putative key to a healthier and longer life, but these benefits may come at a trade-off with reproductive fitness and may affect the following generation(s). The potential inter- and transgenerational effects of long-term fasting and starvation are particularly poorly understood in vertebrates when they originate from the paternal line. We utilised the externally fertilising zebrafish amenable to a split-egg clutch design to explore the male-specific effects of fasting/starvation on fertility and fitness of offspring independently of maternal contribution. Eighteen days of fasting resulted in reduced fertility in exposed males. While average offspring survival was not affected, we detected increased larval growth rate in F1 offspring from starved males and more malformed embryos at 24 h post-fertilisation in F2 offspring produced by F1 offspring from starved males. Comparing the transcriptomes of F1 embryos sired by starved and fed fathers revealed robust and reproducible increased expression of muscle composition genes but lower expression of lipid metabolism and lysosome genes in embryos from starved fathers. A large proportion of these genes showed enrichment in the yolk syncytial layer suggesting gene regulatory responses associated with metabolism of nutrients through paternal effects on extra-embryonic tissues which are loaded with maternal factors. We compared the embryo transcriptomes to published adult transcriptome datasets and found comparable repressive effects of starvation on metabolism-associated genes. These similarities suggest a physiologically relevant, directed and potentially adaptive response transmitted by the father, independently from the offspring's nutritional state, which was defined by the mother.
Collapse
Affiliation(s)
- Ada Jimenez-Gonzalez
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Federico Ansaloni
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Genova, Italy
- Area of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | | | - Ghazal Alavioon
- Department of Evolutionary Biology, Uppsala University, Uppsala, Sweden
| | - David Murray
- School of Biological Sciences, University of East Anglia, Norwich, UK
- Centre for Environment, Fisheries, and Aquaculture Science, Lowestoft, UK
| | - Weronika Robak
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Remo Sanges
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Genova, Italy
- Area of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Ferenc Müller
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Simone Immler
- Department of Evolutionary Biology, Uppsala University, Uppsala, Sweden
- School of Biological Sciences, University of East Anglia, Norwich, UK
| |
Collapse
|
12
|
Murashov AK, Pak ES, Mar J, O’Brien K, Fisher-Wellman K, Bhat KM. Paternal Western diet causes transgenerational increase in food consumption in Drosophila with parallel alterations in the offspring brain proteome and microRNAs. FASEB J 2023; 37:e22966. [PMID: 37227156 PMCID: PMC10234493 DOI: 10.1096/fj.202300239rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/26/2023]
Abstract
Several lines of evidence indicate that ancestral diet might play an important role in determining offspring's metabolic traits. However, it is not yet clear whether ancestral diet can affect offspring's food choices and feeding behavior. In the current study, taking advantage of Drosophila model system, we demonstrate that paternal Western diet (WD) increases offspring food consumption up to the fourth generation. Paternal WD also induced alterations in F1 offspring brain proteome. Using enrichment analyses of pathways for upregulated and downregulated proteins, we found that upregulated proteins had significant enrichments in terms related to translation and translation factors, whereas downregulated proteins displayed enrichments in small molecule metabolic processes, TCA cycles, and electron transport chain (ETC). Using MIENTURNET miRNA prediction tool, dme-miR-10-3p was identified as the top conserved miRNA predicted to target proteins regulated by ancestral diet. RNAi-based knockdown of miR-10 in the brain significantly increased food consumption, implicating miR-10 as a potential factor in programming feeding behavior. Together, these findings suggest that ancestral nutrition may influence offspring feeding behavior through alterations in miRNAs.
Collapse
Affiliation(s)
- Alexander K. Murashov
- Department of Physiology & East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Elena S. Pak
- Department of Physiology & East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Jordan Mar
- Department of Molecular Medicine, University of South Florida, Tampa, FL
| | - Kevin O’Brien
- Department of Biostatistics, College of Allied Health Sciences, East Carolina University, Greenville, NC
| | - Kelsey Fisher-Wellman
- Department of Physiology & East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Krishna M. Bhat
- Department of Molecular Medicine, University of South Florida, Tampa, FL
| |
Collapse
|
13
|
Hernandez A, Martinez ME, Chaves C, Anselmo J. Epigenetic developmental programming and intergenerational effects of thyroid hormones. VITAMINS AND HORMONES 2023; 122:23-49. [PMID: 36863795 PMCID: PMC10938172 DOI: 10.1016/bs.vh.2023.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Mounting evidence is showing that altered signaling through the nuclear hormone receptor superfamily can cause abnormal, long-term epigenetic changes which translate into pathological modifications and susceptibility to disease. These effects seem to be more prominent if the exposure occurs early in life, when transcriptomic profiles are rapidly changing. At this time, the coordination of the complex coordinated processes of cell proliferation and differentiation that characterize mammalian development. Such exposures may also alter the epigenetic information of the germ line, potentially leading to developmental changes and abnormal outcomes in subsequent generations. Thyroid hormone (TH) signaling is mediated by specific nuclear receptors, which have the ability to markedly change chromatin structure and gene transcription, and can also regulate other determinants of epigenetic marks. TH exhibits pleiotropic effects in mammals, and during development, its action is regulated in a highly dynamic manner to suit the rapidly evolving needs of multiple tissues. Their molecular mechanisms of action, timely developmental regulation and broad biological effects place THs in a central position to play a role in the developmental epigenetic programming of adult pathophysiology and, through effects on the germ line, in inter- and trans-generational epigenetic phenomena. These areas of epigenetic research are in their infancy, and studies regarding THs are limited. In the context of their characteristics as epigenetic modifiers and their finely tuned developmental action, here we review some of the observations underscoring the role that altered TH action may play in the developmental programming of adult traits and in the phenotypes of subsequent generations via germ line transmission of altered epigenetic information. Considering the relatively high prevalence of thyroid disease and the ability of some environmental chemicals to disrupt TH action, the epigenetic effects of abnormal levels of TH action may be important contributors to the non-genetic etiology of human disease.
Collapse
Affiliation(s)
- Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, United States; Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States; Department of Medicine, Tufts University School of Medicine, Boston, MA, United States.
| | - M Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, United States
| | - Carolina Chaves
- Serviço de Endocrinologia e Nutrição, Hospital Divino Espírito Santo, Ponta Delgada, Açores, Portugal
| | - Joao Anselmo
- Serviço de Endocrinologia e Nutrição, Hospital Divino Espírito Santo, Ponta Delgada, Açores, Portugal
| |
Collapse
|
14
|
Jung YH, Wang HLV, Ruiz D, Bixler BJ, Linsenbaum H, Xiang JF, Forestier S, Shafik AM, Jin P, Corces VG. Recruitment of CTCF to an Fto enhancer is responsible for transgenerational inheritance of BPA-induced obesity. Proc Natl Acad Sci U S A 2022; 119:e2214988119. [PMID: 36469784 PMCID: PMC9897486 DOI: 10.1073/pnas.2214988119] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
The mechanisms by which environmentally-induced epiphenotypes are transmitted transgenerationally in mammals are poorly understood. Here we show that exposure of pregnant mouse females to bisphenol A (BPA) results in obesity in the F2 progeny due to increased food intake. This epiphenotype can be transmitted up to the F6 generation. Analysis of chromatin accessibility in sperm of the F1-F6 generations reveals alterations at sites containing binding motifs for CCCTC-binding factor (CTCF) at two cis-regulatory elements (CREs) of the Fto gene that correlate with transmission of obesity. These CREs show increased interactions in sperm of obese mice with the Irx3 and Irx5 genes, which are involved in the differentiation of appetite-controlling neurons. Deletion of the CTCF site in Fto results in mice that have normal food intake and fail to become obese when ancestrally exposed to BPA. The results suggest that epigenetic alterations of Fto can lead to the same phenotypes as genetic variants.
Collapse
Affiliation(s)
- Yoon Hee Jung
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Hsiao-Lin V. Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Daniel Ruiz
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Brianna J. Bixler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Hannah Linsenbaum
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Jian-Feng Xiang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Samantha Forestier
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Andrew M. Shafik
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| | - Victor G. Corces
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA30322
| |
Collapse
|
15
|
Food abundance in men before puberty predicts a range of cancers in grandsons. Nat Commun 2022; 13:7507. [PMID: 36473854 PMCID: PMC9726939 DOI: 10.1038/s41467-022-35217-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Nutritional conditions early in human life may influence phenotypic characteristics in later generations. A male-line transgenerational pathway, triggered by the early environment, has been postulated with support from animal and a small number of human studies. Here we analyse individuals born in Uppsala Sweden 1915-29 with linked data from their children and parents, which enables us to explore the hypothesis that pre-pubertal food abundance may trigger a transgenerational effect on cancer events. We used cancer registry and cause-of-death data to analyse 3422 cancer events in grandchildren (G2) by grandparental (G0) food access. We show that variation in harvests and food access in G0 predicts cancer occurrence in G2 in a specific way: abundance among paternal grandfathers, but not any other grandparent, predicts cancer occurrence in grandsons but not in granddaughters. This male-line response is observed for several groups of cancers, suggesting a general susceptibility, possibly acquired in early embryonic development. We observed no transgenerational influence in the middle generation.
Collapse
|
16
|
Patlar B. On the Role of Seminal Fluid Protein and Nucleic Acid Content in Paternal Epigenetic Inheritance. Int J Mol Sci 2022; 23:ijms232314533. [PMID: 36498858 PMCID: PMC9739459 DOI: 10.3390/ijms232314533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The evidence supports the occurrence of environmentally-induced paternal epigenetic inheritance that shapes the offspring phenotype in the absence of direct or indirect paternal care and clearly demonstrates that sperm epigenetics is one of the major actors mediating these paternal effects. However, in most animals, while sperm makes up only a small portion of the seminal fluid, males also have a complex mixture of proteins, peptides, different types of small noncoding RNAs, and cell-free DNA fragments in their ejaculate. These seminal fluid contents (Sfcs) are in close contact with the reproductive cells, tissues, organs, and other molecules of both males and females during reproduction. Moreover, their production and use are adjusted in response to environmental conditions, making them potential markers of environmentally- and developmentally-induced paternal effects on the next generation(s). Although there is some intriguing evidence for Sfc-mediated paternal effects, the underlying molecular mechanisms remain poorly defined. In this review, the current evidence regarding the links between seminal fluid and environmental paternal effects and the potential pathways and mechanisms that seminal fluid may follow in mediating paternal epigenetic inheritance are discussed.
Collapse
Affiliation(s)
- Bahar Patlar
- Animal Ecology, Department of Zoology, Martin-Luther University Halle-Wittenberg, 06099 Halle (Saale), Germany
| |
Collapse
|
17
|
Indriastuti R, Pardede BP, Gunawan A, Ulum MF, Arifiantini RI, Purwantara B. Sperm Transcriptome Analysis Accurately Reveals Male Fertility Potential in Livestock. Animals (Basel) 2022; 12:2955. [PMID: 36359078 PMCID: PMC9657999 DOI: 10.3390/ani12212955] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 08/13/2023] Open
Abstract
Nowadays, selection of superior male candidates in livestock as a source of frozen semen based on sperm quality at the cellular level is not considered accurate enough for predicting the potential of male fertility. Sperm transcriptome analysis approaches, such as messenger RNA levels, have been shown to correlate with fertility rates. Using this technology in livestock growth has become the principal method, which can be widely applied to predict male fertility potential in the livestock industry through the analysis of the sperm transcriptome. It provides the gene expression to validate the function of sperm in spermatogenesis, fertilization, and embryo development, as the parameters of male fertility. This review proposes a transcriptomic analysis approach as a high-throughput method to predict the fertility potential of livestock more accurately in the future.
Collapse
Affiliation(s)
- Rhesti Indriastuti
- Reproductive Biology Study Program, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor 16680, Indonesia
- Tuah Sakato Technology and Resource Development Center, Department of Animal Husbandry and Animal Health of West Sumatra, Payakumbuh 26229, Indonesia
| | - Berlin Pandapotan Pardede
- Department of Veterinary Clinic, Reproduction, and Pathology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor 16680, Indonesia
| | - Asep Gunawan
- Department of Animal Production and Technology, Faculty of Animal Science, IPB University, Bogor 16680, Indonesia
| | - Mokhamad Fakhrul Ulum
- Department of Veterinary Clinic, Reproduction, and Pathology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor 16680, Indonesia
| | - Raden Iis Arifiantini
- Department of Veterinary Clinic, Reproduction, and Pathology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor 16680, Indonesia
| | - Bambang Purwantara
- Department of Veterinary Clinic, Reproduction, and Pathology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor 16680, Indonesia
| |
Collapse
|
18
|
Mayes C, Lawson-Boyd E, Meloni M. Situating the Father: Strengthening Interdisciplinary Collaborations between Sociology, History and the Emerging POHaD Paradigm. Nutrients 2022; 14:3884. [PMID: 36235537 PMCID: PMC9572680 DOI: 10.3390/nu14193884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
(1) Background: Albeit the main focus remains largely on mothers, in recent years Developmental Origins of Health and Disease (DOHaD) scientists, including epigeneticists, have started to examine how a father's environment affects disease risk in children and argued that more attention needs to be given to father's health-related behaviors for their influence on offspring at preconception (i.e., sperm health) as well as paternal lifestyle influences over the first 1000 days. This research ushers in a new paternal origins of health and disease (POHaD) paradigm and is considered a welcome equalization to the overemphasis on maternal influences. Epigeneticists are excited by the possibilities of the POHaD paradigm but are also cautious about how to interpret data and avoid biased impression of socio-biological reality. (2) Methods: We review sociological and historical literatures on the intersection of gender, food and diet across different social and historical contexts to enrich our understanding of the father; (3) Results: Sociological and historical research on family food practices and diet show that there are no "fathers" in the abstract or vacuum, but they are differently classed, racialized and exist in socially stratified situations where choices may be constrained or unavailable. This confirms that epigeneticists researching POHaD need to be cautious in interpreting paternal and maternal dietary influences on offspring health; (4) Conclusions: We suggest that interdisciplinary approach to this new paradigm, which draws on sociology, history and public health, can help provide the social and historical context for interpreting and critically understanding paternal lifestyles and influences on offspring health.
Collapse
Affiliation(s)
- Christopher Mayes
- Alfred Deakin Institute for Citizenship and Globalisation, Deakin University, 75 Pigdons Rd, Geelong 3216, Australia
| | | | | |
Collapse
|
19
|
Gurunathan S, Kang MH, Song H, Kim NH, Kim JH. The role of extracellular vesicles in animal reproduction and diseases. J Anim Sci Biotechnol 2022; 13:62. [PMID: 35681164 PMCID: PMC9185900 DOI: 10.1186/s40104-022-00715-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/05/2022] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are nanosized membrane-enclosed compartments that serve as messengers in cell-to-cell communication, both in normal physiology and in pathological conditions. EVs can transfer functional proteins and genetic information to alter the phenotype and function of recipient cells, which undergo different changes that positively affect their structural and functional integrity. Biological fluids are enriched with several subpopulations of EVs, including exosomes, microvesicles (MVs), and apoptotic bodies carrying several cargoes, such as lipids, proteins, and nucleic acids. EVs associated with the reproductive system are actively involved in the regulation of different physiological events, including gamete maturation, fertilization, and embryo and fetal development. EVs can influence follicle development, oocyte maturation, embryo production, and endometrial-conceptus communication. EVs loaded with cargoes are used to diagnose various diseases, including pregnancy disorders; however, these are dependent on the type of cell of origin and pathological characteristics. EV-derived microRNAs (miRNAs) and proteins in the placenta regulate inflammatory responses and trophoblast invasion through intercellular delivery in the placental microenvironment. This review presents evidence regarding the types of extracellular vesicles, and general aspects of isolation, purification, and characterization of EVs, particularly from various types of embryos. Further, we discuss EVs as mediators and messengers in reproductive biology, the effects of EVs on placentation and pregnancy disorders, the role of EVs in animal reproduction, in the male reproductive system, and mother and embryo cross-communication. In addition, we emphasize the role of microRNAs in embryo implantation and the role of EVs in reproductive and therapeutic medicine. Finally, we discuss the future perspectives of EVs in reproductive biology.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Nam Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
20
|
Transgenerational epigenetic impacts of parental infection on offspring health and disease susceptibility. Trends Genet 2022; 38:662-675. [PMID: 35410793 PMCID: PMC8992946 DOI: 10.1016/j.tig.2022.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022]
Abstract
Maternal immune activation (MIA) and infection during pregnancy are known to reprogramme offspring phenotypes. However, the epigenetic effects of preconceptual paternal infection and paternal immune activation (PIA) are not currently well understood. Recent reports show that paternal infection and immune activation can affect offspring phenotypes, particularly brain function, behaviour, and immune system functioning, across multiple generations without re-exposure to infection. Evidence from other environmental exposures indicates that epigenetic inheritance also occurs in humans. Given the growing impact of the coronavirus disease 2019 (COVID-19) pandemic, it is imperative that we investigate all of the potential epigenetic mechanisms and multigenerational phenotypes that may arise from both maternal and paternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as well as associated MIA, PIA, and inflammation. This will allow us to understand and, if necessary, mitigate any potential changes in disease susceptibility in the children, and grandchildren, of affected parents.
Collapse
|
21
|
Gallo A. Reprotoxic Impact of Environment, Diet, and Behavior. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:1303. [PMID: 35162326 PMCID: PMC8834893 DOI: 10.3390/ijerph19031303] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/14/2022] [Accepted: 01/22/2022] [Indexed: 02/01/2023]
Abstract
Reproductive health is progressively declining due to multiples endogenous and exogenous factors, such as environmental contaminants, diet and behavior. Accumulated evidences confirm that fertility and reproductive function have been adversely affected by exposure to chemical contaminants released in the environment. Today, the impact of diet and behavior on reproductive processes is also receiving special attention from the scientific community. Indeed, a close relationship between diet and fertility has been proven. Furthermore, a combination of unhealthy behavior, such as exposure to hazardous compounds and stress factors, poses living organisms at higher risk of reprotoxic effects. In particular, it has been described that poor life behaviors are associated with reduced male and female fertility due to decreased gamete quality and function. Most of the erroneous behaviors are, furthermore, a source of oxidative stress that, leading to epigenetic alterations, results in an impaired reproductive fitness. This review reports the detrimental impact of the most common environmental chemical stressors, diet, and behavior on reproductive functionality and success. Although clear evidences are still scarce, reassuring data are provided that a healthy diet and reverting unhealthy lifestyles may be of help to recover physiological reproductive conditions.
Collapse
Affiliation(s)
- Alessandra Gallo
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy
| |
Collapse
|
22
|
Chromatin alterations during the epididymal maturation of mouse sperm refine the paternally inherited epigenome. Epigenetics Chromatin 2022; 15:2. [PMID: 34991687 PMCID: PMC8734183 DOI: 10.1186/s13072-021-00433-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Paternal lifestyle choices and male exposure history have a critical influence on the health and fitness of the next generation. Accordingly, defining the processes of germline programming is essential to resolving how the epigenetic memory of paternal experiences transmits to their offspring. Established dogma holds that all facets of chromatin organization and histone posttranslational modification are complete before sperm exits the testes. However, recent clinical and animal studies suggest that patterns of DNA methylation change during epididymal maturation. In this study, we used complementary proteomic and deep-sequencing approaches to test the hypothesis that sperm posttranslational histone modifications change during epididymal transit. RESULTS Using proteomic analysis to contrast immature spermatozoa and mature sperm isolated from the mouse epididymis, we find progressive changes in multiple histone posttranslational modifications, including H3K4me1, H3K27ac, H3K79me2, H3K64ac, H3K122ac, H4K16ac, H3K9me2, and H4K20me3. Interestingly, some of these changes only occurred on histone variant H3.3, and most involve chromatin modifications associated with gene enhancer activity. In contrast, the bivalent chromatin modifications, H3K4me3, and H3K27me3 remained constant. Using chromatin immunoprecipitation coupled with deep sequencing, we find that changes in histone h3, lysine 27 acetylation (H3K27ac) involve sharpening broad diffuse regions into narrow peaks centered on the promoter regions of genes driving embryonic development. Significantly, many of these regions overlap with broad domains of H3K4me3 in oocytes and ATAC-seq signatures of open chromatin identified in MII oocytes and sperm. In contrast, histone h3, lysine 9 dimethylation (H3K9me2) becomes enriched within the promoters of genes driving meiosis and in the distal enhancer regions of tissue-specific genes sequestered at the nuclear lamina. Maturing sperm contain the histone deacetylase enzymes HDAC1 and HDAC3, suggesting the NuRD complex may drive some of these changes. Finally, using Western blotting, we detected changes in chromatin modifications between caput and caudal sperm isolated from rams (Ovis aries), inferring changes in histone modifications are a shared feature of mammalian epididymal maturation. CONCLUSIONS These data extend our understanding of germline programming and reveal that, in addition to trafficking noncoding RNAs, changes in histone posttranslational modifications are a core feature of epididymal maturation.
Collapse
|
23
|
Hotzy C, Fowler E, Kiehl B, Francis R, Mason J, Moxon S, Rostant W, Chapman T, Immler S. Evolutionary history of sexual selection affects microRNA profiles in Drosophila sperm. Evolution 2021; 76:310-319. [PMID: 34874067 DOI: 10.1111/evo.14411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/15/2021] [Accepted: 10/22/2021] [Indexed: 11/28/2022]
Abstract
The presence of small RNAs in sperm is a relatively recent discovery and little is currently known about their importance and functions. Environmental changes including social conditions and dietary manipulations are known to affect the composition and expression of some small RNAs in sperm and may elicit a physiological stress response resulting in an associated change in gamete miRNA profiles. Here, we tested how microRNA profiles in sperm are affected by variation in both sexual selection and dietary regimes in Drosophila melanogaster selection lines. The selection lines were exposed to standard versus low yeast diet treatments and three different population sex ratios (male-biased, female-biased or equal sex) in a full-factorial design. After 38 generations of selection, all males were maintained on their selected diet and in a common garden male-only environment prior to sperm sampling. We performed transcriptome analyses on miRNAs in purified sperm samples. We found 11 differentially expressed miRNAs with the majority showing differences between male- and female-biased lines. Dietary treatment only had a significant effect on miRNA expression levels in interaction with sex ratio. Our findings suggest that long-term adaptation may affect miRNA profiles in sperm and that these may show varied interactions with short-term environmental changes. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Cosima Hotzy
- Department of Evolutionary Biology, Uppsala University, Norbyvägen 18D, Uppsala, 752 36, Sweden
| | - Emily Fowler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Berrit Kiehl
- Department of Evolutionary Biology, Uppsala University, Norbyvägen 18D, Uppsala, 752 36, Sweden
| | - Roy Francis
- Department of Evolutionary Biology, Uppsala University, Norbyvägen 18D, Uppsala, 752 36, Sweden
| | - Janet Mason
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Simon Moxon
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Wayne Rostant
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Tracey Chapman
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Simone Immler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
24
|
Neyroud AS, Chiechio R, Yefimova M, Lo Faro MJ, Dejucq-Rainsford N, Jaillard S, Even-Hernandez P, Marchi V, Ravel C. Extra-cellular vesicles of the male genital tract: new actors in male fertility? Basic Clin Androl 2021; 31:25. [PMID: 34645388 PMCID: PMC8515699 DOI: 10.1186/s12610-021-00141-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular Vesicles (EVs) are membrane-limited particles containing proteins, lipids, metabolites and nucleic acids that are secreted by healthy and cancerous cells. These vesicles are very heterogeneous in size and content and mediate a variety of biological functions. Three subtypes of EV have been described in the male genital tract: microvesicles, myelinosomes and exosomes. Each type of EVs depends on the location of secretion such as the testis, prostate or epididymis. It has been shown that EVs can fuse together and deliver information to recipient cells, for example spermatozoa in the male genital tract. Cryo-electron microscopy remains the reference technique for determining EV morphology, but quantifying the absolute concentration of these EVs in biological fluids remains a challenge from a clinical point of view. The field of bio detection has considerably increased with the introduction of nanomaterials in biosensors and will provide a better understanding of the impact of these EVs. However, functional modifications of male gametes result from interactions with the components of the intraluminal fluid all along the genital tract and depend on the secretion and absorption of proteins and lipids from the local microenvironment. We cannot therefore exclude the possibility of epigenetic modulation of the information that will be transmitted to the embryo and therefore to the next generation via EVs.
Collapse
Affiliation(s)
- Anne-Sophie Neyroud
- CHU Rennes, Service de Biologie de la Reproduction-CECOS, 35000, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Régina Chiechio
- Physics and Astronomy Department "E. Majorana", University of Catania, Via S. Sofia 64, 95123, Catania, Italy
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Marina Yefimova
- CHU Rennes, Service de Biologie de la Reproduction-CECOS, 35000, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, St-Petersburg, 194223, Russia
| | - Maria Josè Lo Faro
- Physics and Astronomy Department "E. Majorana", University of Catania, Via S. Sofia 64, 95123, Catania, Italy
| | - Nathalie Dejucq-Rainsford
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Sylvie Jaillard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Pascale Even-Hernandez
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Valérie Marchi
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Célia Ravel
- CHU Rennes, Service de Biologie de la Reproduction-CECOS, 35000, Rennes, France.
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
25
|
Ben Maamar M, Nilsson EE, Skinner MK. Epigenetic transgenerational inheritance, gametogenesis and germline development†. Biol Reprod 2021; 105:570-592. [PMID: 33929020 PMCID: PMC8444706 DOI: 10.1093/biolre/ioab085] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
One of the most important developing cell types in any biological system is the gamete (sperm and egg). The transmission of phenotypes and optimally adapted physiology to subsequent generations is in large part controlled by gametogenesis. In contrast to genetics, the environment actively regulates epigenetics to impact the physiology and phenotype of cellular and biological systems. The integration of epigenetics and genetics is critical for all developmental biology systems at the cellular and organism level. The current review is focused on the role of epigenetics during gametogenesis for both the spermatogenesis system in the male and oogenesis system in the female. The developmental stages from the initial primordial germ cell through gametogenesis to the mature sperm and egg are presented. How environmental factors can influence the epigenetics of gametogenesis to impact the epigenetic transgenerational inheritance of phenotypic and physiological change in subsequent generations is reviewed.
Collapse
Affiliation(s)
- Millissia Ben Maamar
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Eric E Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
26
|
Paternal environmental exposure-induced spermatozoal small noncoding RNA alteration meditates the intergenerational epigenetic inheritance of multiple diseases. Front Med 2021; 16:176-184. [PMID: 34515940 DOI: 10.1007/s11684-021-0885-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022]
Abstract
Studies of human and mammalian have revealed that environmental exposure can affect paternal health conditions as well as those of the offspring. However, studies that explore the mechanisms that meditate this transmission are rare. Recently, small noncoding RNAs (sncRNAs) in sperm have seemed crucial to this transmission due to their alteration in sperm in response to environmental exposure, and the methodology of microinjection of isolated total RNA or sncRNAs or synthetically identified sncRNAs gradually lifted the veil of sncRNA regulation during intergenerational inheritance along the male line. Hence, by reviewing relevant literature, this study intends to answer the following research concepts: (1) paternal environmental factors that can be passed on to offspring and are attributed to spermatozoal sncRNAs, (2) potential role of paternal spermatozoal sncRNAs during the intergenerational inheritance process, and (3) the potential mechanism by which spermatozoal sncRNAs meditate intergenerational inheritance. In summary, increased attention highlights the hidden wonder of spermatozoal sncRNAs during intergenerational inheritance. Therefore, in the future, more studies should focus on the origin of RNA alteration, the target of RNA regulation, and how sncRNA regulation during embryonic development can be sustained even in adult offspring.
Collapse
|
27
|
Ozturk N, Dansranjavin T, Gies S, Calay D, Shiplu S, Creppe C, Hendrickx J, Schagdarsurengin U. H4K20me3 marks distal intergenic and repetitive regions in human mature spermatozoa. Development 2021; 148:271169. [PMID: 34345914 DOI: 10.1242/dev.196477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 06/30/2021] [Indexed: 01/05/2023]
Abstract
Sperm histones represent an essential part of the paternally transmitted epigenome, but uncertainty exists about the role of those remaining in non-coding and repetitive DNA. We therefore analyzed the genome-wide distribution of the heterochromatic marker H4K20me3 in human sperm and somatic (K562) cells. To specify the function of sperm histones, we compared all H4K20me3-containing and -free loci in the sperm genome. Sperm and somatic cells possessed a very similar H4K20me3 distribution: H4K20me3 peaks occurred mostly in distal intergenic regions and repetitive gene clusters (in particular genes encoding odorant-binding factors and zinc-finger antiviral proteins). In both cell types, H4K20me3 peaks were enriched in LINEs, ERVs, satellite DNA and low complexity repeats. In contrast, H4K20me3-free nucleosomes occurred more frequently in genic regions (in particular promoters, exons, 5'-UTR and 3'-UTR) and were enriched in genes encoding developmental factors (in particular transcription activators and repressors). H4K20me3-free nucleosomes were also detected in substantial quantities in distal intergenic regions and were enriched in SINEs. Thus, evidence suggests that paternally transmitted histones may have a dual purpose: maintenance and regulation of heterochromatin and guidance towards transcription of euchromatin.
Collapse
Affiliation(s)
- Nihan Ozturk
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany.,Working Group Epigenetics of the Urogenital System, Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany
| | - Temuujin Dansranjavin
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany
| | - Sabrina Gies
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany.,Working Group Epigenetics of the Urogenital System, Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany
| | - Damien Calay
- Epigenetics and Bioinformatics Services Team, Diagenode SA, 4102 Liège, Belgium
| | - Shanjid Shiplu
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany.,Working Group Epigenetics of the Urogenital System, Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany
| | - Catherine Creppe
- Epigenetics and Bioinformatics Services Team, Diagenode SA, 4102 Liège, Belgium
| | - Jan Hendrickx
- Epigenetics and Bioinformatics Services Team, Diagenode SA, 4102 Liège, Belgium
| | - Undraga Schagdarsurengin
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany.,Working Group Epigenetics of the Urogenital System, Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
28
|
Merrill SM, Moore SR, Gladish N, Giesbrecht GF, Dewey D, Konwar C, MacIssac JL, Kobor MS, Letourneau NL. Paternal adverse childhood experiences: Associations with infant DNA methylation. Dev Psychobiol 2021; 63:e22174. [PMID: 34333774 DOI: 10.1002/dev.22174] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022]
Abstract
Adverse childhood experiences (ACEs), or cumulative childhood stress exposures, such as abuse, neglect, and household dysfunction, predict later health problems in both the exposed individuals and their offspring. One potential explanation suggests exposure to early adversity predicts epigenetic modification, especially DNA methylation (DNAm), linked to later health. Stress experienced preconception by mothers may associate with DNAm in the next generation. We hypothesized that fathers' exposure to ACEs also associates with their offspring DNAm, which, to our knowledge, has not been previously explored. An epigenome-wide association study (EWAS) of blood DNAm (n = 45) from 3-month-old infants was regressed onto fathers' retrospective ACEs at multiple Cytosine-phosphate-Guanosine (CpG) sites to discover associations. This accounted for infants' sex, age, ethnicity, cell type proportion, and genetic variability. Higher ACE scores associated with methylation values at eight CpGs. Post-hoc analysis found no contribution of paternal education, income, marital status, and parental postpartum depression, but did with paternal smoking and BMI along with infant sleep latency. These same CpGs also contributed to the association between paternal ACEs and offspring attention problems at 3 years. Collectively, these findings suggested there were biological associations with paternal early life adversity and offspring DNAm in infancy, potentially affecting offspring later childhood outcomes.
Collapse
Affiliation(s)
- Sarah M Merrill
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Sarah R Moore
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Nicole Gladish
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada.,Owerko Centre at the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah Dewey
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada.,Owerko Centre at the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Chaini Konwar
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Julia L MacIssac
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Michael S Kobor
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada.,Program in Child and Brain Development, CIFAR, Toronto, Ontario, Canada
| | - Nicole L Letourneau
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada.,Owerko Centre at the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada.,Faculty of Nursing, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
29
|
Lawson-Boyd E, Meloni M. Gender Beneath the Skull: Agency, Trauma and Persisting Stereotypes in Neuroepigenetics. Front Hum Neurosci 2021; 15:667896. [PMID: 34211381 PMCID: PMC8239152 DOI: 10.3389/fnhum.2021.667896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/04/2021] [Indexed: 11/24/2022] Open
Abstract
Epigenetics stands in a complex relationship to issues of sex and gender. As a scientific field, it has been heavily criticized for disproportionately targeting the maternal body and reproducing deterministic views of biological sex (Kenney and Müller, 2017; Lappé, 2018; Richardson et al., 2014). And yet, it also represents the culmination of a long tradition of engaging with developmental biology as a feminist cause, because of the dispersal of the supposed 'master code' of DNA among wider cellular, organismic and ecological contexts (Keller, 1988). In this paper, we explore a number of tensions at the intersection of sex, gender and trauma that are playing out in the emerging area of neuroepigenetics - a relatively new subfield of epigenetics specifically interested in environment-brain relations through epigenetic modifications in neurons. Using qualitative interviews with leading scientists, we explore how trauma is conceptualized in neuroepigenetics, paying attention to its gendered dimensions. We address a number of concerns raised by feminist STS researchers in regard to epigenetics, and illustrate why we believe close engagement with neuroepigenetic claims, and neuroepigenetic researchers themselves, is a crucial step for social scientists interested in questions of embodiment and trauma. We argue this for three reasons: (1) Neuroepigenetic studies are recognizing the agential capacities of biological materials such as genes, neurotransmitters and methyl groups, and how they influence memory formation; (2) Neuroepigenetic conceptions of trauma are yet to be robustly coupled with social and anthropological theories of violence (Eliot, 2021; Nelson, 2021; Walby, 2013); (3) In spite of the gendered assumptions we find in neuroepigenetics, there are fruitful spaces - through collaboration - to be conceptualizing gender beyond culture-biology and nature-nurture binaries (Lock and Nguyen, 2010). To borrow Gravlee's (2009: 51) phrase, we find reason for social scientists to consider how gender is not only constructed, but how it may "become biology" via epigenetic and other biological pathways. Ultimately, we argue that a robust epigenetic methodology is one which values the integrity of expertise outside its own field, and can have an open, not empty mind to cross-disciplinary dialogue.
Collapse
Affiliation(s)
- Elsher Lawson-Boyd
- Alfred Deakin Institute for Citizenship and Globalisation, Deakin University, Burwood, VIC, Australia
| | - Maurizio Meloni
- Alfred Deakin Institute for Citizenship and Globalisation, Deakin University, Burwood, VIC, Australia
| |
Collapse
|
30
|
Prakash MA, Kumaresan A, Ebenezer Samuel King JP, Nag P, Sharma A, Sinha MK, Kamaraj E, Datta TK. Comparative Transcriptomic Analysis of Spermatozoa From High- and Low-Fertile Crossbred Bulls: Implications for Fertility Prediction. Front Cell Dev Biol 2021; 9:647717. [PMID: 34041237 PMCID: PMC8141864 DOI: 10.3389/fcell.2021.647717] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Crossbred bulls produced by crossing Bos taurus and Bos indicus suffer with high incidence of infertility/subfertility problems; however, the etiology remains poorly understood. The uncertain predictability and the inability of semen evaluation techniques to maintain constant correlation with fertility demand for alternate methods for bull fertility prediction. Therefore, in this study, the global differential gene expression between high- and low-fertile crossbred bull sperm was assessed using a high-throughput RNA sequencing technique with the aim to identify transcripts associated with crossbred bull fertility. Crossbred bull sperm contained transcripts for 13,563 genes, in which 2,093 were unique to high-fertile and 5,454 were unique to low-fertile bulls. After normalization of data, a total of 776 transcripts were detected, in which 84 and 168 transcripts were unique to high-fertile and low-fertile bulls, respectively. A total of 176 transcripts were upregulated (fold change > 1) and 209 were downregulated (<1) in low-fertile bulls. Gene ontology analysis identified that the sperm transcripts involved in the oxidative phosphorylation pathway and biological process such as multicellular organism development, spermatogenesis, and in utero embryonic development were downregulated in low-fertile crossbred bull sperm. Sperm transcripts upregulated and unique to low-fertile bulls were majorly involved in translation (biological process) and ribosomal pathway. With the use of RT-qPCR, selected sperm transcripts (n = 12) were validated in crossbred bulls (n = 12) with different fertility ratings and found that the transcriptional abundance of ZNF706, CRISP2, TNP2, and TNP1 genes was significantly (p < 0.05) lower in low-fertile bulls than high-fertile bulls and was positively (p < 0.05) correlated with conception rate. It is inferred that impaired oxidative phosphorylation could be the predominant reason for low fertility in crossbred bulls and that transcriptional abundance of ZNF706, CRISP2, TNP2, and TNP1 genes could serve as potential biomarkers for fertility in crossbred bulls.
Collapse
Affiliation(s)
- Mani Arul Prakash
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of Indian Council of Agricultural Research (ICAR)-National Dairy Research Institute, Bengaluru, India
| | - Arumugam Kumaresan
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of Indian Council of Agricultural Research (ICAR)-National Dairy Research Institute, Bengaluru, India
| | - John Peter Ebenezer Samuel King
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of Indian Council of Agricultural Research (ICAR)-National Dairy Research Institute, Bengaluru, India
| | - Pradeep Nag
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of Indian Council of Agricultural Research (ICAR)-National Dairy Research Institute, Bengaluru, India
| | - Ankur Sharma
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of Indian Council of Agricultural Research (ICAR)-National Dairy Research Institute, Bengaluru, India
| | - Manish Kumar Sinha
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of Indian Council of Agricultural Research (ICAR)-National Dairy Research Institute, Bengaluru, India
| | - Elango Kamaraj
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of Indian Council of Agricultural Research (ICAR)-National Dairy Research Institute, Bengaluru, India
| | - Tirtha Kumar Datta
- Animal Genomics Laboratory, Indian Council of Agricultural Research (ICAR), National Dairy Research Institute, Karnal, India
| |
Collapse
|
31
|
Tanga BM, Qamar AY, Raza S, Bang S, Fang X, Yoon K, Cho J. Semen evaluation: methodological advancements in sperm quality-specific fertility assessment - A review. Anim Biosci 2021; 34:1253-1270. [PMID: 33902175 PMCID: PMC8255896 DOI: 10.5713/ab.21.0072] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/30/2021] [Indexed: 11/27/2022] Open
Abstract
Assessment of male fertility is based on the evaluation of sperm. Semen evaluation measures various sperm quality parameters as fertility indicators. However, semen evaluation has limitations, and it requires the advancement and application of strict quality control methods to interpret the results. This article reviews the recent advances in evaluating various sperm-specific quality characteristics and methodologies, with the help of different assays to assess sperm-fertility status. Sperm evaluation methods that include conventional microscopic methods, computer-assisted sperm analyzers (CASA), and flow cytometric analysis, provide precise information related to sperm morphology and function. Moreover, profiling fertility-related biomarkers in sperm or seminal plasma can be helpful in predicting fertility. Identification of different sperm proteins and diagnosis of DNA damage has positively contributed to the existing pool of knowledge about sperm physiology and molecular anomalies associated with different infertility issues in males. Advances in methods and sperm-specific evaluation has subsequently resulted in a better understanding of sperm biology that has improved the diagnosis and clinical management of male factor infertility. Accurate sperm evaluation is of paramount importance in the application of artificial insemination and assisted reproductive technology. However, no single test can precisely determine fertility; the selection of an appropriate test or a set of tests and parameters is required to accurately determine the fertility of specific animal species. Therefore, a need to further calibrate the CASA and advance the gene expression tests is recommended for faster and field-level applications.
Collapse
Affiliation(s)
- Bereket Molla Tanga
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea.,Faculty of Veterinary Medicine, Hawassa University, 05, Hawassa, Ethiopia
| | - Ahmad Yar Qamar
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea.,Department of Clinical Sciences, College of Veterinary and Animal Sciences, Jhang 35200, Sub-campus University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Sanan Raza
- Department of Clinical Sciences, College of Veterinary and Animal Sciences, Jhang 35200, Sub-campus University of Veterinary and Animal Sciences, Lahore 54000, Pakistan.,Department of Clinical Sciences, College of Veterinary and Animal Sciences, Jhang 35200, Sub-campus University of Veterinary and Animal Sciences, Lahore 54000, PakistanDepartment of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Aydin Adnan Menderes University, Aydin 09016, Turkey
| | - Seonggyu Bang
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Xun Fang
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Kiyoung Yoon
- Department of Companion Animal, Shingu College, Seongnam 13174, Korea
| | - Jongki Cho
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
32
|
Crean AJ, Immler S. Evolutionary consequences of environmental effects on gamete performance. Philos Trans R Soc Lond B Biol Sci 2021; 376:20200122. [PMID: 33866815 DOI: 10.1098/rstb.2020.0122] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Variation in pre- and post-release gamete environments can influence evolutionary processes by altering fertilization outcomes and offspring traits. It is now widely accepted that offspring inherit epigenetic information from both their mothers and fathers. Genetic and epigenetic alterations to eggs and sperm-acquired post-release may also persist post-fertilization with consequences for offspring developmental success and later-life fitness. In externally fertilizing species, gametes are directly exposed to anthropogenically induced environmental impacts including pollution, ocean acidification and climate change. When fertilization occurs within the female reproductive tract, although gametes are at least partially protected from external environmental variation, the selective environment is likely to vary among females. In both scenarios, gamete traits and selection on gametes can be influenced by environmental conditions such as temperature and pollution as well as intrinsic factors such as male and female reproductive fluids, which may be altered by changes in male and female health and physiology. Here, we highlight some of the pathways through which changes in gamete environments can affect fertilization dynamics, gamete interactions and ultimately offspring fitness. We hope that by drawing attention to this important yet often overlooked source of variation, we will inspire future research into the evolutionary implications of anthropogenic interference of gamete environments including the use of assisted reproductive technologies. This article is part of the theme issue 'How does epigenetics influence the course of evolution?'
Collapse
Affiliation(s)
- Angela J Crean
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Simone Immler
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| |
Collapse
|
33
|
Rokade S, Upadhya M, Bhat DS, Subhedar N, Yajnik CS, Ghose A, Rath S, Bal V. Transient systemic inflammation in adult male mice results in underweight progeny. Am J Reprod Immunol 2021; 86:e13401. [PMID: 33576153 DOI: 10.1111/aji.13401] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
PROBLEM While the testes represent an immune-privileged organ, there is evidence that systemic inflammation is accompanied by local inflammatory responses. We therefore examined whether transient systemic inflammation caused any inflammatory and functional consequences in murine testes. METHOD OF STUDY Using a single systemic administration of Toll-like receptor (TLR) agonists [lipopolysaccharide (LPS) or peptidoglycan (PG) or polyinosinic-polycytidylic acid (polyIC)] in young adult male mice, we assessed testicular immune-inflammatory landscape and reproductive functionality. RESULTS Our findings demonstrated a significant induction of testicular TNF-α, IL-1β and IL-6 transcripts within 24 h of TLR agonist injection. By day 6, these cytokine levels returned to baseline. While there was no change in caudal sperm counts at early time points, eight weeks later, twofold decrease in sperm count and reduced testicular testosterone levels were evident. When these mice were subjected to mating studies, no differences in mating efficiencies or litter sizes were observed compared with controls. Nonetheless, the neonatal weights of progeny from LPS/PG/polyIC-treated sires were significantly lower than controls. Postnatal weight gain up to three weeks was also slower in the progeny of LPS/polyIC-treated sires. Placental weights at 17.5 days post-coitum were significantly lower in females mated to LPS- and polyIC-treated males. Given this likelihood of an epigenetic effect, we found lower testicular levels of histone methyltransferase enzyme, mixed-lineage leukaemia-1, in mice given LPS/PG/polyIC 8 weeks earlier. CONCLUSION Exposure to transient systemic inflammation leads to transient local inflammation in the testes, with persistent sperm-mediated consequences for foetal development.
Collapse
Affiliation(s)
- Sushama Rokade
- Indian Institute of Science Education and Research (IISER), Pune, India
| | - Manoj Upadhya
- Indian Institute of Science Education and Research (IISER), Pune, India
| | | | | | | | - Aurnab Ghose
- Indian Institute of Science Education and Research (IISER), Pune, India
| | - Satyajit Rath
- Indian Institute of Science Education and Research (IISER), Pune, India.,KEM Hospital Research Centre, Pune, India
| | - Vineeta Bal
- Indian Institute of Science Education and Research (IISER), Pune, India
| |
Collapse
|
34
|
Bernhardt L, Dittrich M, El-Merahbi R, Saliba AE, Müller T, Sumara G, Vogel J, Nichols-Burns S, Mitchell M, Haaf T, El Hajj N. A genome-wide transcriptomic analysis of embryos fathered by obese males in a murine model of diet-induced obesity. Sci Rep 2021; 11:1979. [PMID: 33479343 PMCID: PMC7820458 DOI: 10.1038/s41598-021-81226-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/04/2021] [Indexed: 12/18/2022] Open
Abstract
Paternal obesity is known to have a negative impact on the male’s reproductive health as well as the health of his offspring. Although epigenetic mechanisms have been implicated in the non-genetic transmission of acquired traits, the effect of paternal obesity on gene expression in the preimplantation embryo has not been fully studied. To this end, we investigated whether paternal obesity is associated with gene expression changes in eight-cell stage embryos fathered by males on a high-fat diet. We used single embryo RNA-seq to compare the gene expression profile of embryos generated by males on a high fat (HFD) versus control (CD) diet. This analysis revealed significant upregulation of the Samd4b and Gata6 gene in embryos in response to a paternal HFD. Furthermore, we could show a significant increase in expression of both Gata6 and Samd4b during differentiation of stromal vascular cells into mature adipocytes. These findings suggest that paternal obesity may induce changes in the male germ cells which are associated with the gene expression changes in the resulting preimplantation embryos.
Collapse
Affiliation(s)
- Laura Bernhardt
- Institute of Human Genetics, Julius Maximilians University, 97074, Würzburg, Germany
| | - Marcus Dittrich
- Institute of Human Genetics, Julius Maximilians University, 97074, Würzburg, Germany.,Department of Bioinformatics, Julius Maximilians University, 97074, Würzburg, Germany
| | - Rabih El-Merahbi
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Josef-Schneider-Str. 2, Haus D15, 97080, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97080, Würzburg, Germany
| | - Tobias Müller
- Department of Bioinformatics, Julius Maximilians University, 97074, Würzburg, Germany
| | - Grzegorz Sumara
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Josef-Schneider-Str. 2, Haus D15, 97080, Würzburg, Germany.,Nencki Institute of Experimental Biology, PAS, 02-093, Warsaw, Poland
| | - Jörg Vogel
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97080, Würzburg, Germany.,Institute of Molecular Infection Biology, University of Würzburg, Josef-Schneider-Straße 2, 97080, Würzburg, Germany
| | - Stefanie Nichols-Burns
- Laboratory for Molecular Medicine, Department of Obstetrics and Gynaecology, Erlangen University Hospital, Universitaetsstrasse, Erlangen, Germany.,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Megan Mitchell
- Laboratory for Molecular Medicine, Department of Obstetrics and Gynaecology, Erlangen University Hospital, Universitaetsstrasse, Erlangen, Germany.,School of Paediatrics and Reproductive Health, The Robinson Institute, University of Adelaide, Adelaide, SA, Australia
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, 97074, Würzburg, Germany
| | - Nady El Hajj
- Institute of Human Genetics, Julius Maximilians University, 97074, Würzburg, Germany. .,College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Education City, Doha, Qatar.
| |
Collapse
|
35
|
Nohara K, Nakabayashi K, Okamura K, Suzuki T, Suzuki S, Hata K. Gestational arsenic exposure induces site-specific DNA hypomethylation in active retrotransposon subfamilies in offspring sperm in mice. Epigenetics Chromatin 2020; 13:53. [PMID: 33267854 PMCID: PMC7709384 DOI: 10.1186/s13072-020-00375-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 11/12/2020] [Indexed: 01/26/2023] Open
Abstract
Background Environmental impacts on a fetus can disrupt germ cell development leading to epimutations in mature germ cells. Paternal inheritance of adverse health effects through sperm epigenomes, including DNA methylomes, has been recognized in human and animal studies. However, the impacts of gestational exposure to a variety of environmental factors on the germ cell epigenomes are not fully investigated. Arsenic, a naturally occurring contaminant, is one of the most concerning environmental chemicals, that is causing serious health problems, including an increase in cancer, in highly contaminated areas worldwide. We previously showed that gestational arsenic exposure of pregnant C3H mice paternally induces hepatic tumor increase in the second generation (F2). In the present study, we have investigated the F1 sperm DNA methylomes genome-widely by one-base resolution analysis using a reduced representation bisulfite sequencing (RRBS) method. Results We have clarified that gestational arsenic exposure increases hypomethylated cytosines in all the chromosomes and they are significantly overrepresented in the retrotransposon LINEs and LTRs, predominantly in the intergenic regions. Closer analyses of detailed annotated DNA sequences showed that hypomethylated cytosines are especially accumulated in the promoter regions of the active full-length L1MdA subfamily in LINEs, and 5′LTRs of the active IAPE subfamily in LTRs. This is the first report that has identified the specific positions of methylomes altered in the retrotransposon elements by environmental exposure, by genome-wide methylome analysis. Conclusion Lowered DNA methylation potentially enhances L1MdA retrotransposition and cryptic promoter activity of 5′LTR for coding genes and non-coding RNAs. The present study has illuminated the environmental impacts on sperm DNA methylome establishment that can lead to augmented retrotransposon activities in germ cells and can cause harmful effects in the following generation.
Collapse
Affiliation(s)
- Keiko Nohara
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, 305-8506, Japan.
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Kazuyuki Okamura
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, 305-8506, Japan
| | - Takehiro Suzuki
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, 305-8506, Japan
| | - Shigekatsu Suzuki
- Center for Environmental Biology and Ecosystem Studies, National Institute for Environmental Studies, Tsukuba, 305-8506, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| |
Collapse
|
36
|
Wu C, Sirard MA. Parental Effects on Epigenetic Programming in Gametes and Embryos of Dairy Cows. Front Genet 2020; 11:557846. [PMID: 33173533 PMCID: PMC7591718 DOI: 10.3389/fgene.2020.557846] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
The bovine represents an important agriculture species and dairy breeds have experienced intense genetic selection over the last decades. The selection of breeders focused initially on milk production, but now includes feed efficiency, health, and fertility, although these traits show lower heritability. The non-genetic paternal and maternal effects on the next generation represent a new research topic that is part of epigenetics. The evidence for embryo programming from both parents is increasing. Both oocytes and spermatozoa carry methylation marks, histones modifications, small RNAs, and chromatin state variations. These epigenetic modifications may remain active in the early zygote and influence the embryonic period and beyond. In this paper, we review parental non-genetic effects retained in gametes on early embryo development of dairy cows, with emphasis on parental age (around puberty), the metabolism of the mother at the time of conception and in vitro culture (IVC) conditions. In our recent findings, transcriptomic signatures and DNA methylation patterns of blastocysts and gametes originating from various parental and IVC conditions revealed surprisingly similar results. Embryos from all these experiments displayed a metabolic signature that could be described as an "economy" mode where protein synthesis is reduced, mitochondria are considered less functional. In the absence of any significant phenotype, these results indicated a possible similar adaptation of the embryo to younger parental age, post-partum metabolic status and IVC conditions mediated by epigenetic factors.
Collapse
Affiliation(s)
| | - Marc-André Sirard
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Département des Sciences Animales, Faculté des Sciences de l’Agriculture et de l’Alimentation, Université Laval, Québec City, QC, Canada
| |
Collapse
|
37
|
Prakash MA, Kumaresan A, Sinha MK, Kamaraj E, Mohanty TK, Datta TK, Morrell JM. RNA-Seq analysis reveals functionally relevant coding and non-coding RNAs in crossbred bull spermatozoa. Anim Reprod Sci 2020; 222:106621. [PMID: 33069132 PMCID: PMC7607363 DOI: 10.1016/j.anireprosci.2020.106621] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022]
Abstract
RNA-Seq analysis was done to characterize the transcriptome of crossbred bull spermatozoa. Among the 13,814 transcripts detected, 431 had FPKM > 1 and 13,673 had FPKM > 0 or < 1. Coding and non-coding RNAs account for 13,145 (95.15%) and 152 (1.1%), respectively. Sperm transcripts were mainly related to ribosome, oxidative phosphorylation and spliceosome pathways. qPCR analysis showed individual variations in transcriptional abundance of selected genes.
Sperm, which are believed to be transcriptionally and translationally inactive, synthesize RNA and proteins before there is gradual disappearance of the ribosome during chromatin compaction. Sperm transfer several functionally relevant transcripts to the oocyte, controlling maternal-zygotic transition and embryonic development. The present study was undertaken to profile and analyze sperm transcripts comprehensively using Next Generation Ribonucleic acid sequencing technology in Holstein Friesian x Tharparkar crossbred bulls. The results from global transcriptomic profiling revealed transcripts for 13,814 genes; of which 431 transcripts were expressed with >1 FPKM and 13,383 transcripts were expressed with >0 or <1 FPKM. The abundant mRNA transcripts of crossbred bull sperm were PRM1 and HMGB4. Gene ontology of transcripts with>1 FPKM revealed there was a major involvement in the structural constituent of ribosomes and translation. Results from pathway enrichment indicated the connection between ribosome, oxidative phosphorylation and spliceosome pathways and the transcripts of crossbred bull spermatozoa. The transcriptional abundance of selected genes, validated using RT-qPCR, indicated significant variations between bulls. Collectively, it may be inferred that the transcripts in crossbred bull sperm were heavily implicated in functions such as the structural constituent of ribosomes and translation, and pathways such as ribosome, oxidative phosphorylation and spliceosome. Further studies using larger sample sizes are required to understand the possible implications of transcriptomic variations on semen quality and fertility.
Collapse
Affiliation(s)
- Mani Arul Prakash
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, 560030 Karnataka, India
| | - Arumugam Kumaresan
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, 560030 Karnataka, India.
| | - Manish Kumar Sinha
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, 560030 Karnataka, India
| | - Elango Kamaraj
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, 560030 Karnataka, India
| | - Tushar Kumar Mohanty
- Animal Reproduction, Gynaecology and Obstetrics, National Dairy Research Institute, Karnal, 132001 Haryana, India
| | - Tirtha Kumar Datta
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, 132001 Haryana, India
| | - Jane M Morrell
- Clinical Sciences, Swedish University of Agricultural Sciences, 750 07 Uppsala, Sweden
| |
Collapse
|
38
|
Environmental Impact on Male (In)Fertility via Epigenetic Route. J Clin Med 2020; 9:jcm9082520. [PMID: 32764255 PMCID: PMC7463911 DOI: 10.3390/jcm9082520] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/21/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
In the last 40 years, male reproductive health-which is very sensitive to both environmental exposure and metabolic status-has deteriorated and the poor sperm quality observed has been suggested to affect offspring development and its health in adult life. In this scenario, evidence now suggests that epigenetics shapes endocrine functions, linking genetics and environment. During fertilization, spermatozoa share with the oocyte their epigenome, along with their haploid genome, in order to orchestrate embryo development. The epigenetic signature of spermatozoa is the result of a dynamic modulation of the epigenetic marks occurring, firstly, in the testis-during germ cell progression-then, along the epididymis, where spermatozoa still receive molecules, conveyed by epididymosomes. Paternal lifestyle, including nutrition and exposure to hazardous substances, alters the phenotype of the next generations, through the remodeling of a sperm epigenetic blueprint that dynamically reacts to a wide range of environmental and lifestyle stressors. With that in mind, this review will summarize and discuss insights into germline epigenetic plasticity caused by environmental stimuli and diet and how spermatozoa may be carriers of induced epimutations across generations through a mechanism known as paternal transgenerational epigenetic inheritance.
Collapse
|
39
|
Kaspar D, Hastreiter S, Irmler M, Hrabé de Angelis M, Beckers J. Nutrition and its role in epigenetic inheritance of obesity and diabetes across generations. Mamm Genome 2020; 31:119-133. [PMID: 32350605 PMCID: PMC7368866 DOI: 10.1007/s00335-020-09839-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Nutritional constraints including not only caloric restriction or protein deficiency, but also energy-dense diets affect metabolic health and frequently lead to obesity and insulin resistance, as well as glucose intolerance and type 2 diabetes. The effects of these environmental factors are often mediated via epigenetic modifiers that target the expression of metabolic genes. More recently, it was discovered that such parentally acquired metabolic changes can alter the metabolic health of the filial and grand-filial generations. In mammals, this epigenetic inheritance can either follow an intergenerational or transgenerational mode of inheritance. In the case of intergenerational inheritance, epimutations established in gametes persist through the first round of epigenetic reprogramming occurring during preimplantation development. For transgenerational inheritance, epimutations persist additionally throughout the reprogramming that occurs during germ cell development later in embryogenesis. Differentially expressed transcripts, genomic cytosine methylations, and several chemical modifications of histones are prime candidates for tangible marks which may serve as epimutations in inter- and transgenerational inheritance and which are currently being investigated experimentally. We review, here, the current literature in support of epigenetic inheritance of metabolic traits caused by nutritional constraints and potential mechanisms in man and in rodent model systems.
Collapse
Affiliation(s)
- Daniela Kaspar
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Sieglinde Hastreiter
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Martin Hrabé de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany
- Chair of Experimental Genetics, Technische Universität München, Weihenstephan, Germany
- Deutsches Zentrum für Diabetesforschung E.V. (DZD), Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany.
- Chair of Experimental Genetics, Technische Universität München, Weihenstephan, Germany.
- Deutsches Zentrum für Diabetesforschung E.V. (DZD), Neuherberg, Germany.
| |
Collapse
|
40
|
Lamb SD, Chia JHZ, Johnson SL. Paternal exposure to a common herbicide alters the behavior and serotonergic system of zebrafish offspring. PLoS One 2020; 15:e0228357. [PMID: 32275662 PMCID: PMC7147785 DOI: 10.1371/journal.pone.0228357] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/13/2020] [Indexed: 12/18/2022] Open
Abstract
Increasingly, studies are revealing that endocrine disrupting chemicals (EDCs) can alter animal behavior. Early life exposure to EDCs may permanently alter phenotypes through to adulthood. In addition, the effects of EDCs may not be isolated to a single generation − offspring may indirectly be impacted, via non-genetic processes. Here, we analyzed the effects of paternal atrazine exposure on behavioral traits (distance moved, exploration, bottom-dwelling time, latency to enter the top zone, and interaction with a mirror) and whole-brain mRNA of genes involved in the serotonergic system regulation (slc6a4a, slc6a4b, htr1Aa, htr1B, htr2B) of zebrafish (Danio rerio). F0 male zebraFIsh were exposed to atrazine at 0.3, 3 or 30 part per billion (ppb) during early juvenile development, the behavior of F1 progeny was tested at adulthood, and the effect of 0.3 ppb atrazine treatment on mRNA transcription was quantified. Paternal exposure to atrazine significantly reduced interactions with a mirror (a proxy for aggression) and altered the latency to enter the top zone of a tank in unexposed F1 offspring. Bottom-dwelling time (a proxy for anxiety) also appeared to be somewhat affected, and activity (distance moved) was reduced in the context of aggression. slc6a4a and htr1Aa mRNA transcript levels were found to correlate positively with anxiety levels in controls, but we found that this relationship was disrupted in the 0.3 ppb atrazine treatment group. Overall, paternal atrazine exposure resulted in alterations across a variety of behavioral traits and showed signs of serotonergic system dysregulation, demonstrating intergenerational effects. Further research is needed to explore transgenerational effects on behavior and possible mechanisms underpinning behavioral effects.
Collapse
Affiliation(s)
- Simon D. Lamb
- Department of Zoology, University of Otago, Dunedin, Otago, New Zealand
- * E-mail: (SDL); (SLJ)
| | - Jolyn H. Z. Chia
- Department of Zoology, University of Otago, Dunedin, Otago, New Zealand
| | - Sheri L. Johnson
- Department of Zoology, University of Otago, Dunedin, Otago, New Zealand
- * E-mail: (SDL); (SLJ)
| |
Collapse
|
41
|
Ryan CP, Kuzawa CW. Germline epigenetic inheritance: Challenges and opportunities for linking human paternal experience with offspring biology and health. Evol Anthropol 2020; 29:180-200. [DOI: 10.1002/evan.21828] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/30/2019] [Accepted: 02/21/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Calen P. Ryan
- Department of AnthropologyNorthwestern University Evanston Illinois USA
| | - Christopher W. Kuzawa
- Department of AnthropologyNorthwestern University Evanston Illinois USA
- Institute for Policy Research Northwestern University Evanston Illinois USA
| |
Collapse
|
42
|
Jeremias G, Gonçalves FJM, Pereira JL, Asselman J. Prospects for incorporation of epigenetic biomarkers in human health and environmental risk assessment of chemicals. Biol Rev Camb Philos Soc 2020; 95:822-846. [PMID: 32045110 DOI: 10.1111/brv.12589] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
Abstract
Epigenetic mechanisms have gained relevance in human health and environmental studies, due to their pivotal role in disease, gene × environment interactions and adaptation to environmental change and/or contamination. Epigenetic mechanisms are highly responsive to external stimuli and a wide range of chemicals has been shown to determine specific epigenetic patterns in several organisms. Furthermore, the mitotic/meiotic inheritance of such epigenetic marks as well as the resulting changes in gene expression and cell/organismal phenotypes has now been demonstrated. Therefore, epigenetic signatures are interesting candidates for linking environmental exposures to disease as well as informing on past exposures to stressors. Accordingly, epigenetic biomarkers could be useful tools in both prospective and retrospective risk assessment but epigenetic endpoints are currently not yet incorporated into risk assessments. Achieving a better understanding on this apparent impasse, as well as identifying routes to promote the application of epigenetic biomarkers within environmental risk assessment frameworks are the objectives of this review. We first compile evidence from human health studies supporting the use of epigenetic exposure-associated changes as reliable biomarkers of exposure. Then, specifically focusing on environmental science, we examine the potential and challenges of developing epigenetic biomarkers for environmental fields, and discuss useful organisms and appropriate sequencing techniques to foster their development in this context. Finally, we discuss the practical incorporation of epigenetic biomarkers in the environmental risk assessment of chemicals, highlighting critical data gaps and making key recommendations for future research within a regulatory context.
Collapse
Affiliation(s)
- Guilherme Jeremias
- Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.,CESAM - Centre for Environmental and Marine Studies, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Fernando J M Gonçalves
- Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.,CESAM - Centre for Environmental and Marine Studies, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Joana L Pereira
- Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.,CESAM - Centre for Environmental and Marine Studies, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Jana Asselman
- Laboratory of Environmental Toxicology and Aquatic Ecology, Environmental Toxicology Unit - GhEnToxLab, Ghent University, 9000, Gent, Belgium
| |
Collapse
|
43
|
Swanson GM, Estill M, Diamond MP, Legro RS, Coutifaris C, Barnhart KT, Huang H, Hansen KR, Trussell JC, Coward RM, Zhang H, Goodrich R, Krawetz SA. Human chromatin remodeler cofactor, RNA interactor, eraser and writer sperm RNAs responding to obesity. Epigenetics 2020; 15:32-46. [PMID: 31354029 PMCID: PMC6961666 DOI: 10.1080/15592294.2019.1644880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/05/2019] [Accepted: 07/09/2019] [Indexed: 12/23/2022] Open
Abstract
In the United States almost 33% of adults are considered obese (BMI > 30 kg/m2). Both animal models and to a lesser extent human studies, have associated BMI, a measure of obesity, with alterations in sperm DNA methylation and RNAs. Sperm RNAs from the Assessment of Multiple Gestations from Ovarian Stimulation trial, were isolated and sequenced. A Generalized Linear Model identified 487 BMI associated human sperm RNA elements (short exon-sized sequences). They partitioned into four patterns; a continual increase with BMI, increase once obese (BMI>30 kg/m2); a steady decrease with BMI; and decrease once overweight (BMI 25 - 30 kg/m2). Gene Ontology revealed a unique relationship between BMI and transcripts associated with chromosome organization, adipogenesis, cellular stress and obesity-related inflammation. Coregulatory networks linked by Chromatin remodeler cofactors, RNA interactors, Erasers and Writers (CREWs) were uncovered to reveal a hierarchical epigenetic response pathway.
Collapse
Affiliation(s)
- Grace M. Swanson
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, USA
| | - Molly Estill
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, USA
| | - Michael P. Diamond
- Department of Obstetrics and Gynecology, Augusta University, Augusta, USA
| | - Richard S. Legro
- Department of Obstetrics and Gynecology, Pennsylvania State University, Hershey, USA
| | - Christos Coutifaris
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, USA
| | - Kurt T. Barnhart
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, USA
| | - Hao Huang
- Department of Biostatistics, Yale University School of Public Health, New Haven, USA
| | - Karl R. Hansen
- Department of Obstetrics and Gynecology, University of Oklahoma College of Medicine, Oklahoma City, USA
| | - J. C. Trussell
- Urology Department, Upstate Medical University, Syracuse, USA
| | - R. Matthew Coward
- Department of Urology, University of North Carolina School of Medicine, Chapel Hill, USA
| | - Heping Zhang
- Department of Biostatistics, Yale University School of Public Health, New Haven, USA
| | - Robert Goodrich
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, USA
| | - Stephen A. Krawetz
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, USA
| |
Collapse
|
44
|
Paternal inheritance of diet induced metabolic traits correlates with germline regulation of diet induced coding gene expression. Genomics 2020; 112:567-573. [DOI: 10.1016/j.ygeno.2019.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/27/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
|
45
|
Nätt D, Kugelberg U, Casas E, Nedstrand E, Zalavary S, Henriksson P, Nijm C, Jäderquist J, Sandborg J, Flinke E, Ramesh R, Örkenby L, Appelkvist F, Lingg T, Guzzi N, Bellodi C, Löf M, Vavouri T, Öst A. Human sperm displays rapid responses to diet. PLoS Biol 2019; 17:e3000559. [PMID: 31877125 PMCID: PMC6932762 DOI: 10.1371/journal.pbio.3000559] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/18/2019] [Indexed: 11/25/2022] Open
Abstract
The global rise in obesity and steady decline in sperm quality are two alarming trends that have emerged during recent decades. In parallel, evidence from model organisms shows that paternal diet can affect offspring metabolic health in a process involving sperm tRNA-derived small RNA (tsRNA). Here, we report that human sperm are acutely sensitive to nutrient flux, both in terms of sperm motility and changes in sperm tsRNA. Over the course of a 2-week diet intervention, in which we first introduced a healthy diet followed by a diet rich in sugar, sperm motility increased and stabilized at high levels. Small RNA-seq on repeatedly sampled sperm from the same individuals revealed that tsRNAs were up-regulated by eating a high-sugar diet for just 1 week. Unsupervised clustering identified two independent pathways for the biogenesis of these tsRNAs: one involving a novel class of fragments with specific cleavage in the T-loop of mature nuclear tRNAs and the other exclusively involving mitochondrial tsRNAs. Mitochondrial involvement was further supported by a similar up-regulation of mitochondrial rRNA-derived small RNA (rsRNA). Notably, the changes in sugar-sensitive tsRNA were positively associated with simultaneous changes in sperm motility and negatively associated with obesity in an independent clinical cohort. This rapid response to a dietary intervention on tsRNA in human sperm is attuned with the paternal intergenerational metabolic responses found in model organisms. More importantly, our findings suggest shared diet-sensitive mechanisms between sperm motility and the biogenesis of tsRNA, which provide novel insights about the interplay between nutrition and male reproductive health.
Collapse
Affiliation(s)
- Daniel Nätt
- Linköping University, Department of Clinical and Experimental Medicine, Division of Neurobiology, Linkoping, Sweden
| | - Unn Kugelberg
- Linköping University, Department of Clinical and Experimental Medicine, Division of Neurobiology, Linkoping, Sweden
| | - Eduard Casas
- Josep Carreras Leukaemia Research Institute (IJC), Program for Predictive and Personalized Medicine of Cancer (PMPPC-IGTP), Barcelona, Spain
| | - Elizabeth Nedstrand
- Linköping University, Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, Linköping, Sweden
| | - Stefan Zalavary
- Linköping University, Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, Linköping, Sweden
| | - Pontus Henriksson
- Karolinska Institute, Department of Biosciences and Nutrition, Huddinge, Sweden
- Linköping University, Department of Medical and Health Sciences, Division of Community Medicine, Linköping, Sweden
| | - Carola Nijm
- Linköping University, Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, Linköping, Sweden
| | - Julia Jäderquist
- Linköping University, Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, Linköping, Sweden
| | - Johanna Sandborg
- Karolinska Institute, Department of Biosciences and Nutrition, Huddinge, Sweden
- Linköping University, Department of Medical and Health Sciences, Division of Community Medicine, Linköping, Sweden
| | - Eva Flinke
- Linköping University, Department of Medical and Health Sciences, Division of Community Medicine, Linköping, Sweden
| | - Rashmi Ramesh
- Linköping University, Department of Clinical and Experimental Medicine, Division of Neurobiology, Linkoping, Sweden
| | - Lovisa Örkenby
- Linköping University, Department of Clinical and Experimental Medicine, Division of Neurobiology, Linkoping, Sweden
| | - Filip Appelkvist
- Linköping University, Department of Clinical and Experimental Medicine, Division of Neurobiology, Linkoping, Sweden
| | - Thomas Lingg
- Linköping University, Department of Clinical and Experimental Medicine, Division of Neurobiology, Linkoping, Sweden
| | - Nicola Guzzi
- Lund University, Stem Cell Center, Department of Laboratory Medicine, Division of Molecular Hematology, Lund, Sweden
| | - Cristian Bellodi
- Lund University, Stem Cell Center, Department of Laboratory Medicine, Division of Molecular Hematology, Lund, Sweden
| | - Marie Löf
- Karolinska Institute, Department of Biosciences and Nutrition, Huddinge, Sweden
- Linköping University, Department of Medical and Health Sciences, Division of Community Medicine, Linköping, Sweden
| | - Tanya Vavouri
- Josep Carreras Leukaemia Research Institute (IJC), Program for Predictive and Personalized Medicine of Cancer (PMPPC-IGTP), Barcelona, Spain
| | - Anita Öst
- Linköping University, Department of Clinical and Experimental Medicine, Division of Neurobiology, Linkoping, Sweden
| |
Collapse
|
46
|
David I, Canario L, Combes S, Demars J. Intergenerational Transmission of Characters Through Genetics, Epigenetics, Microbiota, and Learning in Livestock. Front Genet 2019; 10:1058. [PMID: 31737041 PMCID: PMC6834772 DOI: 10.3389/fgene.2019.01058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Evolutionary biologists studying wild species have demonstrated that genetic and non-genetic sources of information are inherited across generations and are therefore responsible for phenotypic resemblance between relatives. Although it has been postulated that non-genetic sources of inheritance are important in natural selection, they are not taken into account for livestock selection that is based on genetic inheritance only. According to the natural selection theory, the contribution of non-genetic inheritance may be significant for the transmission of characters. If this theory is confirmed in livestock, not considering non-genetic means of transmission in selection schemes might prevent achieving maximum progress in the livestock populations being selected. The present discussion paper reviews the different mechanisms of genetic and non-genetic inheritance reported in the literature as occurring in livestock species. Non-genetic sources of inheritance comprise information transmitted via physical means, such as epigenetic and microbiota inheritance, and those transmitted via learning mechanisms: behavioral, cultural and ecological inheritance. In the first part of this paper we review the evidence that suggests that both genetic and non-genetic information contribute to inheritance in livestock (i.e. transmitted from one generation to the next and causing phenotypic differences between individuals) and discuss how the environment may influence non-genetic inherited factors. Then, in a second step, we consider methods for favoring the transmission of non-genetic inherited factors by estimating and selecting animals on their extended transmissible value and/or introducing favorable non-genetic factors via the animals’ environment.
Collapse
Affiliation(s)
- Ingrid David
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| | - Laurianne Canario
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| | - Sylvie Combes
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| | - Julie Demars
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet Tolosan, France
| |
Collapse
|
47
|
Liberman N, Wang SY, Greer EL. Transgenerational epigenetic inheritance: from phenomena to molecular mechanisms. Curr Opin Neurobiol 2019; 59:189-206. [PMID: 31634674 DOI: 10.1016/j.conb.2019.09.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
Inherited information not encoded in the DNA sequence can regulate a variety of complex phenotypes. However, how this epigenetic information escapes the typical epigenetic erasure that occurs upon fertilization and how it regulates behavior is still unclear. Here we review recent examples of brain related transgenerational epigenetic inheritance and delineate potential molecular mechanisms that could regulate how non-genetic information could be transmitted.
Collapse
Affiliation(s)
- Noa Liberman
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston MA 02115, USA
| | - Simon Yuan Wang
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston MA 02115, USA
| | - Eric Lieberman Greer
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston MA 02115, USA.
| |
Collapse
|
48
|
Zhang Z, Luo X, Lv Y, Yan L, Xu S, Wang Y, Zhong Y, Hang C, Jyotsnav J, Lai D, Shen Z, Xu X, Ma X, Chen Z, Pan Y, Du L. Intrauterine Growth Restriction Programs Intergenerational Transmission of Pulmonary Arterial Hypertension and Endothelial Dysfunction via Sperm Epigenetic Modifications. Hypertension 2019; 74:1160-1171. [PMID: 31596625 DOI: 10.1161/hypertensionaha.119.13634] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intrauterine life represents a window of phenotypic plasticity which carries consequences for later health in adulthood as well as health of subsequent generations. Intrauterine growth-restricted fetuses (intrauterine growth restriction [IUGR]) have a higher risk of pulmonary arterial hypertension in adulthood. Endothelial dysfunction, characterized by hyperproliferation, invasive migration, and disordered angiogenesis, is a hallmark of pulmonary arterial hypertension pathogenesis. Growing evidence suggests that intergenerational transmission of disease, including metabolic syndrome, can be induced by IUGR. Epigenetic modification of the paternal germline is implicated in this transmission. However, it is unclear whether offspring of individuals born with IUGR are also at risk of developing pulmonary arterial hypertension and endothelial dysfunction. Using a model of maternal caloric restriction to induce IUGR, we found that first and second generations of IUGR exhibited elevated pulmonary arterial pressure, myocardial, and vascular remodeling after prolonged exposure to hypoxia. Primary pulmonary vascular endothelial cells (PVECs) from both first and second generations of IUGR exhibited greater proliferation, migration, and angiogenesis. Moreover, in 2 generations, PVECs-derived ET-1 (endothelin-1) was activated by IUGR and hypoxia, and its knockdown mitigated PVECs dysregulation. Most interestingly, within ET-1 first intron, reduced DNA methylation and enhanced tri-methylation of lysine 4 on histone H3 were observed in PVECs and sperm of first generation of IUGR, with DNA demethylation in PVECs of second generation of IUGR. These results suggest that IUGR permanently altered epigenetic signatures of ET-1 from the sperm and PVECs in the first generation, which was subsequently transferred to PVECs of offspring. This mechanism would yield 2 generations with endothelial dysfunction and pulmonary arterial hypertension-like pathophysiological features in adulthood.
Collapse
Affiliation(s)
- Ziming Zhang
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xiaofei Luo
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Ying Lv
- Department of Pediatric Health Care (Y.L.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Lingling Yan
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Shanshan Xu
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yu Wang
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Ying Zhong
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Chengcheng Hang
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Joynauth Jyotsnav
- From the Department of Pediatrics (Z.Z., X.L., L.Y., S.X., Y.W., Y.Z., C.H., J.J.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Dengming Lai
- Department of Neonatal Surgery (D.L.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Zheng Shen
- Laboratory Test Center (Z.S.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xuefeng Xu
- Department of Respiratory Medicine (X.X.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xiaolu Ma
- Department of Neonatology (X.M., Z.C., L.D.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Zheng Chen
- Department of Neonatology (X.M., Z.C., L.D.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yun Pan
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China (Y.P.)
| | - Lizhong Du
- Department of Neonatology (X.M., Z.C., L.D.), the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
49
|
Sharma U. Paternal Contributions to Offspring Health: Role of Sperm Small RNAs in Intergenerational Transmission of Epigenetic Information. Front Cell Dev Biol 2019; 7:215. [PMID: 31681757 PMCID: PMC6803970 DOI: 10.3389/fcell.2019.00215] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/18/2019] [Indexed: 12/21/2022] Open
Abstract
The most fundamental process for the perpetuation of a species is the transfer of information from parent to offspring. Although genomic DNA contributes to the majority of the inheritance, it is now clear that epigenetic information −information beyond the underlying DNA sequence − is also passed on to future generations. However, the mechanism and extent of such inheritance are not well-understood. Here, I review some of the concepts, evidence, and mechanisms of intergenerational epigenetic inheritance via sperm small RNAs. Recent studies provide evidence that mature sperm are highly abundant in small non-coding RNAs. These RNAs are modulated by paternal environmental conditions and potentially delivered to the zygote at fertilization, where they can regulate early embryonic development. Intriguingly, sperm small RNA payload undergoes dramatic changes during testicular and post-testicular maturation, making the mature sperm epigenome highly unique and distinct from testicular germ cells. I explore the mechanism of sperm small RNA remodeling during post-testicular maturation in the epididymis, and the potential role of this reprograming in intergenerational epigenetic inheritance.
Collapse
Affiliation(s)
- Upasna Sharma
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
50
|
Sharp GC, Lawlor DA. Paternal impact on the life course development of obesity and type 2 diabetes in the offspring. Diabetologia 2019; 62:1802-1810. [PMID: 31451867 PMCID: PMC6731203 DOI: 10.1007/s00125-019-4919-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022]
Abstract
The aetiologies of obesity and type 2 diabetes are incredibly complex, but the potential role of paternal influences remains relatively understudied. A better understanding of paternal influences on offspring risk of obesity and type 2 diabetes could have profound implications for public health, clinical practice and society. In this review, we outline potential biological and social mechanisms through which fathers might exert an impact on the health of their offspring. We also present a systematically compiled overview of the current evidence linking paternal factors to offspring development of obesity and type 2 diabetes throughout the life course. Although evidence is accumulating to support paternal associations with offspring outcomes, more high-quality research is needed to overcome specific methodological challenges and provide stronger causal evidence.
Collapse
Affiliation(s)
- Gemma C Sharp
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
- Bristol Dental School, University of Bristol, Bristol, UK.
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and the University of Bristol, Bristol, UK
| |
Collapse
|