1
|
Ban M, Feng W, Hou M, Zhang Z, Cui L. IVF exposure induced intergenerational effects on metabolic phenotype in mice. Reprod Biomed Online 2024; 49:103992. [PMID: 38889592 DOI: 10.1016/j.rbmo.2024.103992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/20/2024] [Accepted: 04/09/2024] [Indexed: 06/20/2024]
Abstract
RESEARCH QUESTION What is the potential transmission of metabolic phenotype from IVF offspring to the subsequent generation? DESIGN An IVF mouse model was established. The F1 generation mice were produced though IVF or natural mating and the F2 generation was obtained through the mating of F1 generation males with normal females. Their metabolic phenotype, including systemic and hepatic glucolipid metabolism, was examined. RESULTS It was found that IVF F1 males exhibited metabolic changes. Compared with the control group, the IVF F1 generation showed increased body weight, elevated fasting glucose and insulin, and increased serum triglyceride concentrations. IVF F1 mice also showed an increased expression of hepatic lipogenesis and autophagy genes. Moreover, IVF F1 males transmitted some metabolic changes to their own male progeny (IVF F2) in the absence of a dietary challenge. IVF F2 mice had increased peri-epididymal and subcutaneous fat and decreased insulin sensitivity. Under the 'second hit' of a high-fat diet, IVF F2 mice further showed increased hepatic lipid deposition with unaltered autophagy levels. CONCLUSION This research demonstrates the impact of IVF on hepatic glucose-lipid metabolism in two successive generations of offspring, highlighting the need for additional investigation. Enhanced understanding of the mechanisms underlying the transmission of multigenerational effects induced by IVF could potentially lead to the advancement of therapeutic interventions for individuals experiencing infertility.
Collapse
Affiliation(s)
- Miaomiao Ban
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Wanbing Feng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Min Hou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Zhirong Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Linlin Cui
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China.; State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, The Second Hospital, Shandong University, Shandong, China..
| |
Collapse
|
2
|
Anuradha R, Srinivas M, Satyavani M, Suresh K, Muralidhar MN, Rajender Rao K. Preconceptional paternal caloric restriction of high-fat diet-induced obesity in Wistar rats dysregulates the metabolism of their offspring via AMPK/SIRT1 pathway. Lipids Health Dis 2024; 23:174. [PMID: 38851752 PMCID: PMC11162063 DOI: 10.1186/s12944-024-02161-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Obesity is a metabolic syndrome where allelic and environmental variations together determine the susceptibility of an individual to the disease. Caloric restriction (CR) is a nutritional dietary strategy recognized to be beneficial as a weight loss regime in obese individuals. Preconceptional parental CR is proven to have detrimental effects on the health and development of their offspring. As yet studies on maternal CR effect on their offspring are well established but paternal CR studies are not progressing. In current study, the impact of different paternal CR regimes in diet-induced obese male Wistar rats (WNIN), on their offspring concerning metabolic syndrome are addressed. METHODS High-fat diet-induced obese male Wistar rats were subjected to caloric restriction of 50% (HFCR-I) and 40% (HFCR-II) and then they were mated with normal females. The male parent's reproductive function was assessed by sperm parameters and their DNMT's mRNA expression levels were also examined. The offspring's metabolic function was assessed by physiological, biochemical and molecular parameters. RESULTS The HFCR-I male parents have shown reduced body weights, compromised male fertility and reduced DNA methylation activity. Further, the HFCR-I offspring showed attenuation of the AMPK/SIRT1 pathway, which is associated with the progression of proinflammatory status and oxidative stress. In line, the HFCR-I offspring also developed altered glucose and lipid homeostasis by exhibiting impaired glucose tolerance & insulin sensitivity, dyslipidemia and steatosis. However, these effects were largely mitigated in HFCR-II offspring. Regarding the obesogenic effects, female offspring exhibited greater susceptibility than male offspring, suggesting that females are more prone to the influences of the paternal diet. CONCLUSION The findings highlight that HFCR-I resulted in paternal undernutrition, impacting the health of offspring, whereas HFCR-II largely restored the effects of a high-fat diet on their offspring. As a result, moderate caloric restriction has emerged as an effective weight loss strategy with minimal implications on future generations. This underscores the shared responsibility of fathers in contributing to sperm-specific epigenetic imprints that influence the health of adult offspring.
Collapse
Affiliation(s)
- Rachakatla Anuradha
- Animal Facility, ICMR-National Institute of Nutrition, Tarnaka, Hyderabad, 500007, Telangana, India
| | - M Srinivas
- Animal Facility, ICMR-National Institute of Nutrition, Tarnaka, Hyderabad, 500007, Telangana, India
| | - M Satyavani
- Animal Facility, ICMR-National Institute of Nutrition, Tarnaka, Hyderabad, 500007, Telangana, India
| | - K Suresh
- Animal Facility, ICMR-National Institute of Nutrition, Tarnaka, Hyderabad, 500007, Telangana, India
| | - M N Muralidhar
- ICMR-Centre for Research, Management and Control of Haemoglobinopathies (ICMR- CRMCH), Chandrapur, 442406, Maharashtra, India
| | - Kalashikam Rajender Rao
- Animal Facility, ICMR-National Institute of Nutrition, Tarnaka, Hyderabad, 500007, Telangana, India.
| |
Collapse
|
3
|
Abdulghani MF, Al-Fayyadh S. Natural products for managing metabolic syndrome: a scoping review. Front Pharmacol 2024; 15:1366946. [PMID: 38746011 PMCID: PMC11091304 DOI: 10.3389/fphar.2024.1366946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction Metabolic syndrome comprises a collection of metabolic disorders stemming from factors like genetic predisposition, inadequate nutrition, stress, decreased physical activity, aging, and ethnicity. Although traditional pharmaceutical treatments exist for metabolic syndrome, their limited popularity is attributed to high costs and adverse effects. Consequently, natural products with fewer side effects have been explored for managing this condition. This literature review aims to explore the role of natural products including herbs, botanicals, vitamins, minerals, probiotics, and dietary supplements in managing metabolic syndrome. Methods This scoping review was conducted in five steps, involving the formulation of a research question, the retrieval and extraction of relevant studies, the selection of pertinent studies, the organization of information into tables, and the reporting of results. Data was collected from various databases including Embase, Science Direct, PubMed, Google Scholar, Scopus, and Web of Science, with a focus on studies published from 2010 to the present, available in English and with full-text accessibility. Results We identified 1,259 articles, screened their titles, abstracts, and full texts, ultimately incorporating 169 pertinent articles into this review (comprising 90 review articles, 32 trial articles, 6 in vitro articles, 38 in vivo articles, 1 experimental article and 2 observational articles). The study's outcomes revealed that natural products, encompassing plants and their derivatives, vitamins and supplements, as well as probiotics, can exert a beneficial influence on metabolic syndrome by regulating blood sugar, blood pressure, lipid profiles, obesity, and abnormal cholesterol and triglyceride levels. Conclusion The current study underscores the significance of natural products in addressing metabolic syndrome. Consequently, it is advisable to conduct further extensive research to assess the efficacy of these products, potentially integrating them into treatment regimens for individuals with metabolic syndrome.
Collapse
|
4
|
Abdulghani MF, Al-Fayyadh S. Natural products for managing metabolic syndrome: a scoping review. Front Pharmacol 2024; 15. [DOI: https:/doi.org/10.3389/fphar.2024.1366946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024] Open
Abstract
IntroductionMetabolic syndrome comprises a collection of metabolic disorders stemming from factors like genetic predisposition, inadequate nutrition, stress, decreased physical activity, aging, and ethnicity. Although traditional pharmaceutical treatments exist for metabolic syndrome, their limited popularity is attributed to high costs and adverse effects. Consequently, natural products with fewer side effects have been explored for managing this condition. This literature review aims to explore the role of natural products including herbs, botanicals, vitamins, minerals, probiotics, and dietary supplements in managing metabolic syndrome.MethodsThis scoping review was conducted in five steps, involving the formulation of a research question, the retrieval and extraction of relevant studies, the selection of pertinent studies, the organization of information into tables, and the reporting of results. Data was collected from various databases including Embase, Science Direct, PubMed, Google Scholar, Scopus, and Web of Science, with a focus on studies published from 2010 to the present, available in English and with full-text accessibility.ResultsWe identified 1,259 articles, screened their titles, abstracts, and full texts, ultimately incorporating 169 pertinent articles into this review (comprising 90 review articles, 32 trial articles, 6 in vitro articles, 38 in vivo articles, 1 experimental article and 2 observational articles). The study’s outcomes revealed that natural products, encompassing plants and their derivatives, vitamins and supplements, as well as probiotics, can exert a beneficial influence on metabolic syndrome by regulating blood sugar, blood pressure, lipid profiles, obesity, and abnormal cholesterol and triglyceride levels.ConclusionThe current study underscores the significance of natural products in addressing metabolic syndrome. Consequently, it is advisable to conduct further extensive research to assess the efficacy of these products, potentially integrating them into treatment regimens for individuals with metabolic syndrome.
Collapse
|
5
|
Kok DE, Saunders R, Nelson A, Smith D, Ford D, Mathers JC, McKay JA. Influence of maternal folate depletion on Art3 DNA methylation in the murine adult brain; potential consequences for brain and neurocognitive health. Mutagenesis 2024; 39:196-204. [PMID: 38417824 PMCID: PMC11040152 DOI: 10.1093/mutage/geae007] [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: 10/04/2023] [Accepted: 02/27/2024] [Indexed: 03/01/2024] Open
Abstract
The developmental origins of health and disease hypothesis suggest early-life environment impacts health outcomes throughout the life course. In particular, epigenetic marks, including DNA methylation, are thought to be key mechanisms through which environmental exposures programme later-life health. Adequate maternal folate status before and during pregnancy is essential in the protection against neural tube defects, but data are emerging that suggest early-life folate exposures may also influence neurocognitive outcomes in childhood and, potentially, thereafter. Since folate is key to the supply of methyl donors for DNA methylation, we hypothesize that DNA methylation may be a mediating mechanism through which maternal folate influences neurocognitive outcomes. Using bisulphite sequencing, we measured DNA methylation of five genes (Art3, Rsp16, Tspo, Wnt16, and Pcdhb6) in the brain tissue of adult offspring of dams who were depleted of folate (n = 5, 0.4 mg folic acid/kg diet) during pregnancy (~19-21 days) and lactation (mean 22 days) compared with controls (n = 6, 2 mg folic acid/kg diet). Genes were selected as methylation of their promoters had previously been found to be altered by maternal folate intake in mice and humans across the life course, and because they have potential associations with neurocognitive outcomes. Maternal folate depletion was significantly associated with Art3 gene hypomethylation in subcortical brain tissue of adult mice at 28 weeks of age (mean decrease 6.2%, P = .03). For the other genes, no statistically significant differences were found between folate depleted and control groups. Given its association with neurocognitive outcomes, we suggest Art3 warrants further study in the context of lifecourse brain health. We have uncovered a potential biomarker that, once validated in accessible biospecimens and human context, may be useful to track the impact of early-life folate exposure on later-life neurocognitive health, and potentially be used to develop and monitor the effects of interventions.
Collapse
Affiliation(s)
- Dieuwertje E Kok
- Division of Human Nutrition and Health, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen Stippeneng 4, 6708 WE Wageningen Wageningen Campus l Building 124 (Helix), Wageningen, The Netherlands
| | - Rachael Saunders
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Northumberland Building, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Andrew Nelson
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Northumberland Building, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Darren Smith
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Northumberland Building, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Dianne Ford
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Northumberland Building, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - John C Mathers
- Human Nutrition & Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Room M2.060, 2nd floor William Leech Building, Framlington Place, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Jill A McKay
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Northumberland Building, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| |
Collapse
|
6
|
Crean AJ, Senior AM, Freire T, Clark TD, Mackay F, Austin G, Pulpitel TJ, Nobrega MA, Barrès R, Simpson SJ. Paternal dietary macronutrient balance and energy intake drive metabolic and behavioral differences among offspring. Nat Commun 2024; 15:2982. [PMID: 38582785 PMCID: PMC10998877 DOI: 10.1038/s41467-024-46782-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 03/11/2024] [Indexed: 04/08/2024] Open
Abstract
Paternal diet can influence the phenotype of the next generation, yet, the dietary components inducing specific responses in the offspring are not identified. Here, we use the Nutritional Geometry Framework to determine the effects of pre-conception paternal dietary macronutrient balance on offspring metabolic and behavioral traits in mice. Ten isocaloric diets varying in the relative proportion of protein, fats, and carbohydrates are fed to male mice prior to mating. Dams and offspring are fed standard chow and never exposed to treatment diets. Body fat in female offspring is positively associated with the paternal consumption of fat, while in male offspring, an anxiety-like phenotype is associated to paternal diets low in protein and high in carbohydrates. Our study uncovers that the nature and the magnitude of paternal effects are driven by interactions between macronutrient balance and energy intake and are not solely the result of over- or undernutrition.
Collapse
Affiliation(s)
- Angela Jane Crean
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Alistair McNair Senior
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Therese Freire
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Thomas Daniel Clark
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Flora Mackay
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Gracie Austin
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Tamara Jayne Pulpitel
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | | | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK, 2200, Denmark.
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur & Centre National pour la Recherche Scientifique (CNRS), Valbonne, 06560, France.
| | - Stephen James Simpson
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
7
|
McPherson NO, Nottle M, McIlfatrick S, Saini A, Hamilton H, Bowman E, Tully CA, Pacella-Ince L, Zander-Fox D, Bakos HW. Clinical use of progesterone in human sperm preparation media for increasing IVF success. Reprod Biomed Online 2024; 48:103625. [PMID: 38402675 DOI: 10.1016/j.rbmo.2023.103625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 02/27/2024]
Abstract
RESEARCH QUESTION Can the addition of progesterone and neurotensin, molecular agents found in the female reproductive tract, after sperm washing increase the fertilization potential of human spermatozoa? DESIGN (i) Cohort study of 24 men. Spermatozoa selected by swim-up were incubated in either progesterone or neurotensin (0.1-100 µM) for 1-4 h, and hyperactive motility and binding to hyaluronan (0.1-100 µM) were assessed. The effect of progesterone 10 µM on sperm function was assessed in a blinded manner, including: hyperactive motility, binding to hyaluronan, tyrosine phosphorylation, acrosome reaction and oxidative DNA damage. (i) Embryo safety testing [one-cell mouse embryo assay (MEA), endotoxin and sterility counts (n = 3)] in preclinical embryo models of IVF (murine and porcine, n = 7 each model) and a small preliminary human study (n = 4) of couples undergoing standard IVF with oocytes inseminated with spermatozoa ± 10 µM progesterone. RESULTS Progesterone 10 µM increased sperm binding to hyaluronan, hyperactive motility and tyrosine phosphorylation (all P < 0.05). Neurotensin had no effect (P > 0.05). Progesterone 10 µM in human embryo culture media passed embryo safety testing (MEA, endotoxin concentration and sterility plate count). In preclinical models of IVF, the exposure of spermatozoa to progesterone 10 µM and oocytes to progesterone 1 µM was not detrimental, and increased the fertilization rate in mice and the blastocyst cell number in mice and pigs (all P ≤ 0.03). In humans, every transferred blastocyst that had been produced from spermatozoa exposed to progesterone resulted in a live birth. CONCLUSION The addition of progesterone to sperm preparation media shows promise as an adjunct to current methods for increasing fertilization potential. Randomized controlled trials are required to determine the clinical utility of progesterone for improving IVF outcomes.
Collapse
Affiliation(s)
- Nicole O McPherson
- Discipline of Reproduction and Development, School of Biomedicine, University of Adelaide, Adelaide, Australia; Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, Australia; Robinson Research Institute, University of Adelaide, Adelaide, Australia; Monash IVF Group, Clayton, Australia.
| | - Mark Nottle
- Discipline of Reproduction and Development, School of Biomedicine, University of Adelaide, Adelaide, Australia; Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Stephan McIlfatrick
- Discipline of Reproduction and Development, School of Biomedicine, University of Adelaide, Adelaide, Australia; Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Anmol Saini
- Discipline of Reproduction and Development, School of Biomedicine, University of Adelaide, Adelaide, Australia; Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | | | | | - Cathryn A Tully
- Discipline of Reproduction and Development, School of Biomedicine, University of Adelaide, Adelaide, Australia; Robinson Research Institute, University of Adelaide, Adelaide, Australia; Repromed, Dulwich, Australia
| | - Leanne Pacella-Ince
- Discipline of Reproduction and Development, School of Biomedicine, University of Adelaide, Adelaide, Australia; Robinson Research Institute, University of Adelaide, Adelaide, Australia; Repromed, Dulwich, Australia
| | - Deirdre Zander-Fox
- Discipline of Reproduction and Development, School of Biomedicine, University of Adelaide, Adelaide, Australia; Monash IVF Group, Clayton, Australia; Monash University, Clayton, Australia
| | - Hassan W Bakos
- Monash IVF Group, Clayton, Australia; University of Newcastle, Newcastle, Australia; Memphasys Ltd, Homebush, Australia
| |
Collapse
|
8
|
Giannubilo SR, Marzioni D, Tossetta G, Montironi R, Meccariello ML, Ciavattini A. The "Bad Father": Paternal Role in Biology of Pregnancy and in Birth Outcome. BIOLOGY 2024; 13:165. [PMID: 38534435 DOI: 10.3390/biology13030165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024]
Abstract
Pregnancy is generally studied as a biological interaction between a mother and a fetus; however, the father, with his characteristics, lifestyle, genetics, and living environment, is by no means unrelated to the outcome of pregnancy. The half of the fetal genetic heritage of paternal derivation can be decisive in cases of inherited chromosomal disorders, and can be the result of de novo genetic alterations. In addition to the strictly pathological aspects, paternal genetics may transmit thrombophilic traits that affect the implantation and vascular construction of the feto-placental unit, lead to placenta-mediated diseases such as pre-eclampsia and fetal growth retardation, and contribute to the multifactorial genesis of preterm delivery. Biological aspects of immunological tolerance to paternal antigens also appear to be crucial for these pathologies. Finally, this review describes the biological findings by which the environment, exposure to pathogens, lifestyle, and nutritional style of the father affect fetal pathophysiological and epigenetic definition.
Collapse
Affiliation(s)
- Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica Delle Marche, 60123 Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, 60126 Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, 60126 Ancona, Italy
| | - Ramona Montironi
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica Delle Marche, 60123 Ancona, Italy
| | - Maria Liberata Meccariello
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica Delle Marche, 60123 Ancona, Italy
| | - Andrea Ciavattini
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica Delle Marche, 60123 Ancona, Italy
| |
Collapse
|
9
|
Bochantin-Winders KA, Baumgaertner F, Hurlbert JL, Menezes ACB, Kirsch JD, Dorsam ST, Schauer CS, Dahlen CR. Divergent planes of nutrition in mature rams influences body composition, hormone and metabolite concentrations, and offspring birth measurements, but not semen characteristics or offspring growth. J Anim Sci 2024; 102:skae207. [PMID: 39044680 PMCID: PMC11347781 DOI: 10.1093/jas/skae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024] Open
Abstract
Objectives of this experiment were to characterize the effects of ram plane of nutrition on body composition, concentrations of hormones and metabolites, sperm characteristics, and offspring outcomes. Mature Rambouillet rams (n = 24, BW = 82.9 ± 2.63 kg) were individually housed and randomly assigned to either a positive (POS; n = 8), maintenance (MAINT; n = 8), or negative (NEG; n = 8) plane of nutrition for an 84-day feeding period. Rams were fed a common diet, with daily feed allocations adjusted weekly based on body weight (BW) to achieve the targeted weight gain or loss (approximately 12% of initial BW). On 0, 28, 56, and 84-d, body condition score (BCS) and scrotal circumference (SC) were recorded, and blood and semen were collected. Following the feeding period, rams were placed in pens with 10 ewes each for a 28-d breeding period. Ewes were managed similarly throughout gestation and body weight and measurements were recorded at birth and weaning. Data were analyzed as repeated measures in time where appropriate with the mixed procedure of SAS, and individual ram was the experimental unit for all analysis. Ram BW was influenced by a treatment × day interaction (P < 0.001), with POS (0.12 ± 0.01 kg) having greater daily weight change than MAINT (0.1 ± 0.01 kg), which was greater than NEG (-0.12 ± 0.01 kg). Ram BCS and SC were influenced by treatment × day interactions (P ≤ 0.01), being similar on day 0 but POS being greater than NEG by day 56. Concentrations of triiodothyronine (T3) and T3:T4 ratio exhibited treatment × day interactions (P ≤ 0.02), as POS had greater values than NEG by day 84 (P ≤ 0.02). Concentration of insulin-like growth factor-1 was greater in POS than MAINT and NEG (P ≤ 0.02), and non-esterified fatty acids and thyroxine (T4) were influenced by a day effect (P ≤ 0.01), but testosterone was unaffected (P ≥ 0.09). Minimal differences in semen volume, sperm concentration, motility, or morphology were observed among treatments (P ≥ 0.31). A similar proportion of ewes bred by rams in the respective treatments lambed and weaned lambs (P ≥ 0.54). Birth weight, chest circumference, and shoulder-hip length were greater (P ≤ 0.05) in NEG lambs compared with POS and MAINT; however, no differences were detected in weaning weight and weaning body measurements (P ≥ 0.40). Findings suggest paternal nutrition during the period of sperm development may influence offspring outcomes, potentially as a result of in-utero programming of paternal origin.
Collapse
Affiliation(s)
| | | | - Jennifer L Hurlbert
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| | - Ana Clara B Menezes
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - James D Kirsch
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| | - Sheri T Dorsam
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| | - Christopher S Schauer
- Hettinger Research Extension Center, North Dakota State University, Hettinger, ND 58639, USA
| | - Carl R Dahlen
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| |
Collapse
|
10
|
Peel A, Saini A, Deluao JC, McPherson NO. Sperm DNA damage: The possible link between obesity and male infertility, an update of the current literature. Andrology 2023; 11:1635-1652. [PMID: 36789664 DOI: 10.1111/andr.13409] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/13/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
Obesity prevalence worldwide is increasing significantly. Whilst maternal obesity has clear detrimental impacts on fertility, pregnancy and foetal outcomes, more recently there has been an increasing focus on the role of paternal obesity in human fertility. Recent meta-analyses have indicated that obesity in men negatively affects basic sperm parameters such as sperm count, concentration and motility, increases the incidence of infertility and reduces the chances of conception. Sperm DNA damage, typically characterised by DNA strand breaks and oxidation of DNA nucleotides, is a specialised marker of sperm quality that has been independently associated with recurrent miscarriage, reduced assisted reproduction success and increased mutational loads in subsequent offspring. Whilst, there are still conflicting data in humans as to the association of obesity in men with sperm DNA damage, evidence from rodent models is clear, indicating that male obesity increases sperm DNA damage. Human data are often conflicting because of the large heterogeneity amongst studies, the use of body mass index as the indicator of obesity and the methods used for detection of sperm DNA damage. Furthermore, comorbidities of obesity (i.e., heat stress, adipokines, insulin resistance, changes in lipids, hypogonadism and obstructive sleep apnoea) are also independently associated with increased sperm DNA damage that is not always modified in men with obesity, and as such may provide a causative link to the discrepancies amongst human studies. In this review, we provide an update on the literature regarding the associations between obesity in men and fertility, basic sperm parameters and sperm DNA damage. We further discuss potential reasons for the discrepancies in the literature and outline possible direct and indirect mechanisms of increased sperm DNA damage resulting from obesity. Finally, we summarise intergenerational obesity through the paternal linage and how sperm DNA damage may contribute to the transmission.
Collapse
Affiliation(s)
- Andrew Peel
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anmol Saini
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Joshua C Deluao
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nicole O McPherson
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
- Repromed IVF Adelaide, Dulwich, South Australia, Australia
| |
Collapse
|
11
|
Liu S, Sharma U. Sperm RNA Payload: Implications for Intergenerational Epigenetic Inheritance. Int J Mol Sci 2023; 24:5889. [PMID: 36982962 PMCID: PMC10052761 DOI: 10.3390/ijms24065889] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
There is mounting evidence that ancestral life experiences and environment can influence phenotypes in descendants. The parental environment regulates offspring phenotypes potentially via modulating epigenetic marks in the gametes. Here, we review examples of across-generational inheritance of paternal environmental effects and the current understanding of the role of small RNAs in such inheritance. We discuss recent advances in revealing the small RNA payload of sperm and how environmental conditions modulate sperm small RNAs. Further, we discuss the potential mechanism of inheritance of paternal environmental effects by focusing on sperm small RNA-mediated regulation of early embryonic gene expression and its role in influencing offspring phenotypes.
Collapse
Affiliation(s)
| | - Upasna Sharma
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| |
Collapse
|
12
|
Townsend J, Braz CU, Taylor T, Khatib H. Effects of paternal methionine supplementation on sperm DNA methylation and embryo transcriptome in sheep. ENVIRONMENTAL EPIGENETICS 2022; 9:dvac029. [PMID: 36727109 PMCID: PMC9885981 DOI: 10.1093/eep/dvac029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 06/18/2023]
Abstract
Environmental effects on gene expression and offspring development can be mediated by epigenetic modifications. It is well established that maternal diet influences DNA methylation patterns and phenotypes in the offspring; however, the epigenetic effects of paternal diet on developing offspring warrants further investigation. Here, we examined how a prepubertal methionine-enriched paternal diet affected sperm DNA methylation and its subsequent effects on embryo gene expression. Three treatment and three control rams were bred to seven ewes, and blastocysts were flushed for RNA extraction. Semen was collected from all rams and submitted for reduced representation bisulfite sequencing analysis. In total, 166 differentially methylated cytosines were identified in the sperm from treatment versus control rams. Nine genes were found to be differentially expressed in embryos produced from treatment versus control rams, and seven differentially methylated cytosines in the sperm were found to be highly correlated with gene expression in the embryos. Our results demonstrate that sperm methylation differences induced by diet may influence fetal programming.
Collapse
Affiliation(s)
- Jessica Townsend
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, 1675 Observatory Dr., Madison, WI 53706, USA
| | - Camila U Braz
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, 1675 Observatory Dr., Madison, WI 53706, USA
| | - Todd Taylor
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, 1675 Observatory Dr., Madison, WI 53706, USA
| | - Hasan Khatib
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, 1675 Observatory Dr., Madison, WI 53706, USA
| |
Collapse
|
13
|
Verdikt R, Armstrong AA, Allard P. Transgenerational inheritance and its modulation by environmental cues. Curr Top Dev Biol 2022; 152:31-76. [PMID: 36707214 PMCID: PMC9940302 DOI: 10.1016/bs.ctdb.2022.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The epigenome plays an important role in shaping phenotypes. However, whether the environment can alter an organism's phenotype across several generations through epigenetic remodeling in the germline is still a highly debated topic. In this chapter, we briefly review the mechanisms of epigenetic inheritance and their connection with germline development before highlighting specific developmental windows of susceptibility to environmental cues. We further discuss the evidence of transgenerational inheritance to a range of different environmental cues, both epidemiological in humans and experimental in rodent models. Doing so, we pinpoint the current challenges in demonstrating transgenerational inheritance to environmental cues and offer insight in how recent technological advances may help deciphering the epigenetic mechanisms at play. Together, we draw a detailed picture of how our environment can influence our epigenomes, ultimately reshaping our phenotypes, in an extended theory of inheritance.
Collapse
Affiliation(s)
- Roxane Verdikt
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, United States.
| | - Abigail A Armstrong
- Department of Obstetrics/Gynecology and Division of Reproductive Endocrinology and Infertility, University of California, Los Angeles, CA, United States
| | - Patrick Allard
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
14
|
Effect of Paternal Diet on Spermatogenesis and Offspring Health: Focus on Epigenetics and Interventions with Food Bioactive Compounds. Nutrients 2022; 14:nu14102150. [PMID: 35631291 PMCID: PMC9143121 DOI: 10.3390/nu14102150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
Infertility is a growing public health problem. Consumption of antioxidant bioactive food compounds (BFCs) that include micronutrients and non-nutrients has been highlighted as a potential strategy to protect against oxidative and inflammatory damage in the male reproductive system induced by obesity, alcohol, and toxicants and, thus, improve spermatogenesis and the fertility parameters. Paternal consumption of such dietary compounds could not only benefit the fathers but their offspring as well. Studies in the new field of paternal origins of health and disease show that paternal malnutrition can alter sperm epigenome, and this can alter fetal development and program an increased risk of metabolic diseases and breast cancer in adulthood. BFCs, such as ascorbic acid, α-tocopherol, polyunsaturated fatty acids, trace elements, carnitines, N-acetylcysteine, and coenzyme Q10, have been shown to improve male gametogenesis, modulate epigenetics of germ cells, and the epigenetic signature of the offspring, restoring offspring metabolic health induced by stressors during early life. This indicates that, from a father’s perspective, preconception is a valuable window of opportunity to start potential nutritional interventions with these BFCs to maximize sperm epigenetic integrity and promote adequate fetal growth and development, thus preventing chronic disease in adulthood.
Collapse
|
15
|
Sánchez-Garrido MA, García-Galiano D, Tena-Sempere M. Early programming of reproductive health and fertility: novel neuroendocrine mechanisms and implications in reproductive medicine. Hum Reprod Update 2022; 28:346-375. [PMID: 35187579 PMCID: PMC9071071 DOI: 10.1093/humupd/dmac005] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/29/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND According to the Developmental Origins of Health and Disease (DOHaD) hypothesis, environmental changes taking place during early maturational periods may alter normal development and predispose to the occurrence of diverse pathologies later in life. Indeed, adverse conditions during these critical developmental windows of high plasticity have been reported to alter the offspring developmental trajectory, causing permanent functional and structural perturbations that in the long term may enhance disease susceptibility. However, while solid evidence has documented that fluctuations in environmental factors, ranging from nutrient availability to chemicals, in early developmental stages (including the peri-conceptional period) have discernible programming effects that increase vulnerability to develop metabolic perturbations, the impact and eventual mechanisms involved, of such developmental alterations on the reproductive phenotype of offspring have received less attention. OBJECTIVE AND RATIONALE This review will summarize recent advances in basic and clinical research that support the concept of DOHaD in the context of the impact of nutritional and hormonal perturbations, occurring during the periconceptional, fetal and early postnatal stages, on different aspects of reproductive function in both sexes. Special emphasis will be given to the effects of early nutritional stress on the timing of puberty and adult gonadotropic function, and to address the underlying neuroendocrine pathways, with particular attention to involvement of the Kiss1 system in these reproductive perturbations. The implications of such phenomena in terms of reproductive medicine will also be considered. SEARCH METHODS A comprehensive MEDLINE search, using PubMed as main interface, of research articles and reviews, published mainly between 2006 and 2021, has been carried out. Search was implemented using multiple terms, focusing on clinical and preclinical data from DOHaD studies, addressing periconceptional, gestational and perinatal programming of reproduction. Selected studies addressing early programming of metabolic function have also been considered, when relevant. OUTCOMES A solid body of evidence, from clinical and preclinical studies, has documented the impact of nutritional and hormonal fluctuations during the periconceptional, prenatal and early postnatal periods on pubertal maturation, as well as adult gonadotropic function and fertility. Furthermore, exposure to environmental chemicals, such as bisphenol A, and maternal stress has been shown to negatively influence pubertal development and gonadotropic function in adulthood. The underlying neuroendocrine pathways and mechanisms involved have been also addressed, mainly by preclinical studies, which have identified an, as yet incomplete, array of molecular and neurohormonal effectors. These include, prominently, epigenetic regulatory mechanisms and the hypothalamic Kiss1 system, which likely contribute to the generation of reproductive alterations in conditions of early nutritional and/or metabolic stress. In addition to the Kiss1 system, other major hypothalamic regulators of GnRH neurosecretion, such as γ-aminobutyric acid and glutamate, may be targets of developmental programming. WIDER IMPLICATIONS This review addresses an underdeveloped area of reproductive biology and medicine that may help to improve our understanding of human reproductive disorders and stresses the importance, and eventual pathogenic impact, of early determinants of puberty, adult reproductive function and fertility.
Collapse
Affiliation(s)
- Miguel Angel Sánchez-Garrido
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - David García-Galiano
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
16
|
Billah MM, Khatiwada S, Morris MJ, Maloney CA. Effects of paternal overnutrition and interventions on future generations. Int J Obes (Lond) 2022; 46:901-917. [PMID: 35022547 PMCID: PMC9050512 DOI: 10.1038/s41366-021-01042-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023]
Abstract
In the last two decades, evidence from human and animal studies suggests that paternal obesity around the time of conception can have adverse effects on offspring health through developmental programming. This may make significant contributions to the current epidemic of obesity and related metabolic and reproductive complications like diabetes, cardiovascular disease, and subfertility/infertility. To date, changes in seminal fluid composition, sperm DNA methylation, histone composition, small non-coding RNAs, and sperm DNA damage have been proposed as potential underpinning mechanism to program offspring health. In this review, we discuss current human and rodent evidence on the impact of paternal obesity/overnutrition on offspring health, followed by the proposed mechanisms, with a focus on sperm DNA damage underpinning paternal programming. We also summarize the different intervention strategies implemented to minimize effects of paternal obesity. Upon critical review of literature, we find that obesity-induced altered sperm quality in father is linked with compromised offspring health. Paternal exercise intervention before conception has been shown to improve metabolic health. Further work to explore the mechanisms underlying benefits of paternal exercise on offspring are warranted. Conversion to healthy diets and micronutrient supplementation during pre-conception have shown some positive impacts towards minimizing the impact of paternal obesity on offspring. Pharmacological approaches e.g., metformin are also being applied. Thus, interventions in the obese father may ameliorate the potential detrimental impacts of paternal obesity on offspring.
Collapse
Affiliation(s)
| | - Saroj Khatiwada
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Margaret J Morris
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | |
Collapse
|
17
|
Paternal periconception metabolic health and offspring programming. Proc Nutr Soc 2022; 81:119-125. [DOI: 10.1017/s0029665121003736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The association between maternal metabolic status at the time of conception and subsequent embryogenesis and offspring development has been studied in detail. However, less attention has been given to the significance of paternal nutrition and metabolism in directing offspring health. Despite this disparity, emerging evidence has begun to highlight an important connection between paternal metabolic well-being, semen quality, embryonic development and ultimately adult offspring health. This has established a new component within the Developmental Origins of Health and Disease hypothesis. Building on the decades of understanding and insight derived from the numerous models of maternal programming, attention is now becoming focused on defining the mechanisms underlying the links between paternal well-being, post-fertilisation development and offspring health. Understanding how the health and fitness of the father impact on semen quality is of fundamental importance for providing better information to intending fathers. Furthermore, assisted reproductive practices such as in vitro fertilisation rely on our ability to select the best quality sperm from a diverse and heterogeneous population. With considerable advances in sequencing capabilities, our understanding of the molecular and epigenetic composition of the sperm and seminal plasma, and their association with male metabolic health, has developed dramatically over recent years. This review will summarise our current understanding of how a father's metabolic status at the time of conception can affect sperm quality, post-fertilisation embryonic and fetal development and offspring health.
Collapse
|
18
|
Bauer KC, York EM, Cirstea MS, Radisavljevic N, Petersen C, Huus KE, Brown EM, Bozorgmehr T, Berdún R, Bernier L, Lee AHY, Woodward SE, Krekhno Z, Han J, Hancock REW, Ayala V, MacVicar BA, Finlay BB. Gut microbes shape microglia and cognitive function during malnutrition. Glia 2022; 70:820-841. [PMID: 35019164 PMCID: PMC9305450 DOI: 10.1002/glia.24139] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/17/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022]
Abstract
Fecal-oral contamination promotes malnutrition pathology. Lasting consequences of early life malnutrition include cognitive impairment, but the underlying pathology and influence of gut microbes remain largely unknown. Here, we utilize an established murine model combining malnutrition and iterative exposure to fecal commensals (MAL-BG). The MAL-BG model was analyzed in comparison to malnourished (MAL mice) and healthy (CON mice) controls. Malnourished mice display poor spatial memory and learning plasticity, as well as altered microglia, non-neuronal CNS cells that regulate neuroimmune responses and brain plasticity. Chronic fecal-oral exposures shaped microglial morphology and transcriptional profile, promoting phagocytic features in MAL-BG mice. Unexpectedly, these changes occurred independently from significant cytokine-induced inflammation or blood-brain barrier (BBB) disruption, key gut-brain pathways. Metabolomic profiling of the MAL-BG cortex revealed altered polyunsaturated fatty acid (PUFA) profiles and systemic lipoxidative stress. In contrast, supplementation with an ω3 PUFA/antioxidant-associated diet (PAO) mitigated cognitive deficits within the MAL-BG model. These findings provide valued insight into the malnourished gut microbiota-brain axis, highlighting PUFA metabolism as a potential therapeutic target.
Collapse
Affiliation(s)
- Kylynda C. Bauer
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Elisa M. York
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Mihai S. Cirstea
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Nina Radisavljevic
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Biochemistry and Molecular Biology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Charisse Petersen
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
| | - Kelsey E. Huus
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Eric M. Brown
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | | | - Rebeca Berdún
- Institut de Recerca Biomèdica de Lleida (IRB‐Lleida)LleidaSpain
- Department of Experimental MedicineUniversitat de Lleida (UdL)LleidaSpain
| | - Louis‐Philippe Bernier
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Amy H. Y. Lee
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Sarah E. Woodward
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Zakhar Krekhno
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
| | - Jun Han
- The Metabolomics Innovation CentreUniversity of VictoriaVictoriaCanada
| | - Robert E. W. Hancock
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Victoria Ayala
- Institut de Recerca Biomèdica de Lleida (IRB‐Lleida)LleidaSpain
- Department of Experimental MedicineUniversitat de Lleida (UdL)LleidaSpain
| | - Brian A. MacVicar
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Barton Brett Finlay
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
- Biochemistry and Molecular Biology DepartmentUniversity of British ColumbiaVancouverCanada
| |
Collapse
|
19
|
Moura FH, Fonseca MA, Macias-Franco A, Archilia EC, Batalha IM, Pena-Bello CA, Silva AEM, Moreira GM, Schütz LF, Norris AB. Characterization of body composition and liver epigenetic markers during periods of negative energy balance and subsequent compensatory growth in postpubertal beef bulls. J Anim Sci 2022; 100:6532621. [PMID: 35184171 PMCID: PMC9036401 DOI: 10.1093/jas/skac047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/16/2022] [Indexed: 11/14/2022] Open
Abstract
This study aimed to characterize the effects of dietary restriction and subsequent re-alimentation on body composition and hepatic gene expression of epigenetic markers of DNA methylation, RNA m6A methylation, and histone acetylation in the liver of postpubertal beef bulls. Twelve Angus × Hereford crossbred bulls (n = 6, 23 ± 0.55 mo [young bulls], 558 ± 6.1 kg; and n = 6, 47 ± 1.2 mo [mature bulls], 740 ± 30.5 kg) were submitted to two dietary regimes per offering of the same hay: low plane of nutrition (90 d) and compensatory growth (90 d). Each animal acted as its own control and were fed Beardless wheat (Triticum aestivum) hay and mineral mix during the trial. Statistical analyses were performed using SAS 9.4 following a pre-post repeated measures design. Bulls in negative energy balance (NEB) decreased (P < 0.001) empty body weight (EBW; 23.1% [-139.1 kg]), empty body fat (EBF; 39.8% [-85.4 kg]), and empty body protein (EBP; 14.9% [-13.5 kg]) and fully recovered at the end of the trial. Body fat accounted for 77.1% of daily changes in body energy status, whereas body protein accounted for only 22.9% (P < 0.001). Relative abundance of epigenetic markers transcripts was analyzed via qPCR. Bulls at NEB tended (P ≤ 0.097) to increase gene expression of epigenetic markers of RNA m6A methylation (METTL14, VIRMA, and WTAP) and increased (P ≤ 0.050) the gene expression of epigenetic markers of DNA methylation (DNMT3A) and histone-acetylation (SIRT3 and SIRT7). Young bulls had a tendency (P ≤ 0.072) of higher RNA m6A methylation, VIRMA, and WTAP than mature bulls. Effect of diet × age interaction was not detected (P ≥ 0.137) for METTL14, VIRMA, WTAP, DNMT3A, SIRT3, or SIRT7. Younger bulls tended to have greater RNA m6A methylation levels than mature bulls, indicating that, while contemporaneously fed the same diet during periods of undernourishment followed by compensatory growth, age has an impact on this epigenetic mechanism. In conclusion, metabolic status seems to carry a greater impact on regulating bovine hepatic epigenetic mechanisms that modulate gene transcription, such as DNA methylation and histone acetylation, than on epigenetic mechanisms that regulate gene translation, such as RNA m6A methylation. During periods of undernourishment followed by compensatory growth, body fat pools appear to change more dynamically and are easily detected having a greater impact on epigenetic markers that modulate hepatic gene transcription rather than translation.
Collapse
Affiliation(s)
- Felipe H Moura
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Mozart A Fonseca
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA,Corresponding author:
| | - Arturo Macias-Franco
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Evandro C Archilia
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Isadora M Batalha
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Camilo A Pena-Bello
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Aghata E M Silva
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Gabriel M Moreira
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Luis F Schütz
- Department of Agriculture, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Aaron B Norris
- Department of Natural Resources Management, Texas Tech University, Lubbock, TX 79430, USA
| |
Collapse
|
20
|
Furse S, Watkins AJ, Williams HEL, Snowden SG, Chiarugi D, Koulman A. Paternal nutritional programming of lipid metabolism is propagated through sperm and seminal plasma. Metabolomics 2022; 18:13. [PMID: 35141784 PMCID: PMC8828597 DOI: 10.1007/s11306-022-01869-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The paternal diet affects lipid metabolism in offspring for at least two generations through nutritional programming. However, we do not know how this is propagated to the offspring. OBJECTIVES We tested the hypothesis that the changes in lipid metabolism that are driven by paternal diet are propagated through spermatozoa and not seminal plasma. METHODS We applied an updated, purpose-built computational network analysis tool to characterise control of lipid metabolism systemically (Lipid Traffic Analysis v2.3) on a known mouse model of paternal nutritional programming. RESULTS The analysis showed that the two possible routes for programming effects, the sperm (genes) and seminal plasma (influence on the uterine environment), both have a distinct effect on the offspring's lipid metabolism. Further, the programming effects in offspring suggest that changes in lipid distribution are more important than alterations in lipid biosynthesis. CONCLUSIONS These results show how the uterine environment and genes both affect lipid metabolism in offspring, enhancing our understanding of the link between parental diet and metabolism in offspring.
Collapse
Affiliation(s)
- Samuel Furse
- Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
- Metabolic Disease Unit, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
- Biological Chemistry Group, Jodrell Laboratory, Royal Botanic Gardens Kew, Richmond, UK.
| | - Adam J Watkins
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Huw E L Williams
- Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Stuart G Snowden
- Department of Biological Sciences, Royal Holloway College, University of London, Egham, TW20 0EX, Surrey, UK
| | - Davide Chiarugi
- Bioinformatics and Biostatistics Core, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
- Metabolic Disease Unit, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
21
|
Belmonte-Tebar A, San Martin Perez E, Nam Cha S, Soler Valls AJ, Singh ND, de la Casa-Esperon E. Diet effects on mouse meiotic recombination: a warning for recombination studies. Genetics 2022; 220:iyab190. [PMID: 34791205 PMCID: PMC8733447 DOI: 10.1093/genetics/iyab190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Meiotic recombination is a critical process for sexually reproducing organisms. This exchange of genetic information between homologous chromosomes during meiosis is important not only because it generates genetic diversity, but also because it is often required for proper chromosome segregation. Consequently, the frequency and distribution of crossovers are tightly controlled to ensure fertility and offspring viability. However, in many systems, it has been shown that environmental factors can alter the frequency of crossover events. Two studies in flies and yeast point to nutritional status affecting the frequency of crossing over. However, this question remains unexplored in mammals. Here, we test how crossover frequency varies in response to diet in Mus musculus males. We use immunohistochemistry to estimate crossover frequency in multiple genotypes under two diet treatments. Our results indicate that while crossover frequency was unaffected by diet in some strains, other strains were sensitive even to small composition changes between two common laboratory chows. Therefore, recombination is both resistant and sensitive to certain dietary changes in a strain-dependent manner and, hence, this response is genetically determined. Our study is the first to report a nutrition effect on genome-wide levels of recombination. Moreover, our work highlights the importance of controlling diet in recombination studies and may point to diet as a potential source of variability among studies, which is relevant for reproducibility.
Collapse
Affiliation(s)
- Angela Belmonte-Tebar
- Regional Center for Biomedical Research (C.R.I.B.), University of Castilla-La Mancha, Albacete 02008, Spain
| | - Estefania San Martin Perez
- Regional Center for Biomedical Research (C.R.I.B.), University of Castilla-La Mancha, Albacete 02008, Spain
| | - Syonghyun Nam Cha
- Pathology Department and Biobank of Albacete, University Hospital Complex of Albacete, Albacete 02006, Spain
| | | | - Nadia D Singh
- Department of Biology, Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Elena de la Casa-Esperon
- Regional Center for Biomedical Research (C.R.I.B.), University of Castilla-La Mancha, Albacete 02008, Spain
- Department of Inorganic and Organic Chemistry and Biochemistry, School of Pharmacy, University of Castilla-La Mancha, Albacete 02071, Spain
| |
Collapse
|
22
|
Billah MM, Khatiwada S, Lecomte V, Morris MJ, Maloney CA. Ameliorating high-fat diet-induced sperm and testicular oxidative damage by micronutrient-based antioxidant intervention in rats. Eur J Nutr 2022; 61:3741-3753. [PMID: 35708759 PMCID: PMC9464124 DOI: 10.1007/s00394-022-02917-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/19/2022] [Indexed: 12/18/2022]
Abstract
PURPOSE Emerging evidence from rodent studies suggests that high-fat-diet (HFD)-induced obesity is characterized by increased oxidative damage in sperm and testis. However, interventions using micronutrient supplementation to mitigate oxidative damage in obesity have not been extensively studied. This study aimed to investigate the effect of an antioxidant-based micronutrient supplement (added folate, vitamin B6, choline, betaine, and zinc) on sperm and testicular oxidative damage in HFD-fed male Sprague Dawley rats. METHODS Rats (3-weeks-old, 12/group) were weaned onto control (C) or HFD (H) or these diets with micronutrient supplement (CS; HS); sperm and testis were harvested at 30.5 weeks. To assess oxidative stress and antioxidant capacity in testis, levels of malondialdehyde (MDA), glutathione (GSH), folate and susceptibility index (SI) of pro-oxidative damage, mRNA expression of Nrf2, NFκB-p65, IL-6, IL-10 and TNF-α, in addition to superoxide-dismutase (SOD), catalase and glutathione-peroxidase (GPx) activities were measured. 8-hydroxy-2-deoxyguanosine (8-OHdG) were assessed in both sperm and testis. RESULTS HFD-fed rats had significantly increased 8-OHdG content in sperm and testis, increased testicular SI, decreased testicular weight, SOD and GPx activity compared to control. Strikingly, supplementation of HFD appeared to significantly reduce 8-OHdG in sperm and testis (22% and 24.3%, respectively), reduce testicular SI and MDA content (28% and 40%, respectively), increase testicular weight (24%), SOD and GPX activity (30% and 70%, respectively) and GSH content (19%). Moreover, supplementation had significant impact to increase testicular folate content regardless of diet. Furthermore, an overall effect of supplementation to increase testicular mRNA expression of Nrf2 was observed across groups. Interestingly, testicular SI was positively correlated with sperm and testicular 8-OHdG and MDA content, suggesting a critical role of testicular antioxidant activity to combat oxidative damage in sperm and testis. CONCLUSION Our findings suggest that antioxidant-based micronutrient supplement has the potential to interrupt HFD-induced sperm and testicular oxidative damage by improving testicular antioxidant capacity.
Collapse
Affiliation(s)
| | - Saroj Khatiwada
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Virginie Lecomte
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Margaret J Morris
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | |
Collapse
|
23
|
Hieronimus B, Ensenauer R. Influence of maternal and paternal pre-conception overweight/obesity on offspring outcomes and strategies for prevention. Eur J Clin Nutr 2021; 75:1735-1744. [PMID: 34131301 PMCID: PMC8636250 DOI: 10.1038/s41430-021-00920-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023]
Abstract
Overweight, obesity, and their comorbidities remain global health challenges. When established early in life, overweight is often sustained into adulthood and contributes to the early onset of non-communicable diseases. Parental pre-conception overweight and obesity is a risk factor for overweight and obesity in childhood and beyond. This increased risk likely is based on an interplay of genetic alterations and environmental exposures already at the beginning of life, although mechanisms are still poorly defined. In this narrative review, potential routes of transmission of pre-conceptional overweight/obesity from mothers and fathers to their offspring as well as prevention strategies are discussed. Observational evidence suggests that metabolic changes due to parental overweight/obesity affect epigenetic markers in oocytes and sperms alike and may influence epigenetic programming and reprogramming processes during embryogenesis. While weight reduction in overweight/obese men and women, who plan to become pregnant, seems advisable to improve undesirable outcomes in offspring, caution might be warranted. Limited evidence suggests that weight loss in men and women in close proximity to conception might increase undesirable offspring outcomes at birth due to nutritional deficits and/or metabolic disturbances in the parent also affecting gamete quality. A change in the dietary pattern might be more advisable. The data reviewed here suggest that pre-conception intervention strategies should shift from women to couples, and future studies should address possible interactions between maternal and paternal contribution to longitudinal childhood outcomes. Randomized controlled trials focusing on effects of pre-conceptional diet quality on long-term offspring health are warranted.
Collapse
Affiliation(s)
- Bettina Hieronimus
- Institute of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
| | - Regina Ensenauer
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany.
| |
Collapse
|
24
|
Dolati P, Zamiri MJ, Akhlaghi A, Khodabandeh Z, Mehrabani D, Atashi H, Jamhiri I. Reproductive and embryological toxicity of lead acetate in male mice and their offspring and mitigation effects of quercetin. J Trace Elem Med Biol 2021; 67:126793. [PMID: 34049200 DOI: 10.1016/j.jtemb.2021.126793] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 10/24/2022]
Abstract
Exposure to heavy metals not only impacts on fertility in males, it may also affect the offspring. The aim of the present study was to examine the toxic effects of lead acetate on fertility in male mice and their offspring, and the potential effect of quercetin on mitigating the likely effects. Experimental mice were randomly divided into three groups and administered with (i) distilled water (control); (ii) lead acetate (150 mg/kg BW/day); (iii) lead acetate (150 mg/kg BW/day) with quercetin (75 mg/kg BW/day). Lead acetate administration in male mice adversely affected their fertility through changes in sperm motility, viability, morphology, maturity, membrane integrity, and intracellular reactive oxygen species (P < 0.05). Similar findings were observed in the offspring of the lead-treated male mice. Early embryonic development and implantation rate were also adversely influenced in both the sires and offspring when male mice were treated with lead acetate (P < 0.05). The data demonstrated that down-regulation of Cks2 (CDC28 protein kinase regulatory subunit-2) in sperm had an association with early embryonic development in lead acetate treated group. In conclusion, lead acetate administration adversely impacted on the fertility of the male mice and their male offspring fertility; on the other hand, paternal quercetin co-administration somewhat ameliorated the adverse effects of lead on male mice and their offspring.
Collapse
Affiliation(s)
- Parisa Dolati
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran.
| | - Mohammad Javad Zamiri
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran.
| | - Amir Akhlaghi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran.
| | - Zahra Khodabandeh
- Stem Cells Technology Research Center, Shiraz University of Medical Science, Shiraz, Iran.
| | - Davood Mehrabani
- Stem Cells Technology Research Center, Shiraz University of Medical Science, Shiraz, Iran; Li Ka Shing Centre for Health Research and Innovation, University of Alberta, Edmonton, AB, Canada.
| | - Hadi Atashi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran.
| | - Iman Jamhiri
- Stem Cells Technology Research Center, Shiraz University of Medical Science, Shiraz, Iran.
| |
Collapse
|
25
|
Consequences of Paternal Nutrition on Offspring Health and Disease. Nutrients 2021; 13:nu13082818. [PMID: 34444978 PMCID: PMC8400857 DOI: 10.3390/nu13082818] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/30/2022] Open
Abstract
It is well established that the maternal diet during the periconceptional period affects the progeny’s health. A growing body of evidence suggests that the paternal diet also influences disease onset in offspring. For many years, sperm was considered only to contribute half of the progeny’s genome. It now appears that it also plays a crucial role in health and disease in offspring’s adult life. The nutritional status and environmental exposure of fathers during their childhood and/or the periconceptional period have significant transgenerational consequences. This review aims to describe the effects of various human and rodent paternal feeding patterns on progeny’s metabolism and health, including fasting or intermittent fasting, low-protein and folic acid deficient food, and overnutrition in high-fat and high-sugar diets. The impact on pregnancy outcome, metabolic pathways, and chronic disease onset will be described. The biological and epigenetic mechanisms underlying the transmission from fathers to their progeny will be discussed. All these data provide evidence of the impact of paternal nutrition on progeny health which could lead to preventive diet recommendations for future fathers.
Collapse
|
26
|
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: 11] [Impact Index Per Article: 3.7] [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
|
27
|
Eberle C, Kirchner MF, Herden R, Stichling S. Paternal metabolic and cardiovascular programming of their offspring: A systematic scoping review. PLoS One 2020; 15:e0244826. [PMID: 33382823 PMCID: PMC7775047 DOI: 10.1371/journal.pone.0244826] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There is lots of evidence that maternal peri-gestational metabolic, genomic and environmental conditions are closely linked to metabolic and cardiovascular outcomes in their offspring later in life. Moreover, there is also lotsof evidence that underlining mechanisms, such as molecular as well as epigenetic changes may alter the intrauterine environment leading to cardio-metabolic diseases in their offspring postnatal. But, there is also increasing evidence that cardio-metabolic diseases may be closely linked to their paternal metabolic risk factors, such as obesity, Type 2 Diabetes and other risk factors. OBJECTIVE To analyse the evidence as well as specific risk factors of paternal trans-generational programming of cardio-metabolic diseases in their offspring. METHODS Within a systematic scoping review, we performed a literature search in MEDLINE (PubMed) and EMBASE databases in August 2020 considering original research articles (2000-2020) that examined the impact of paternal programming on metabolic and cardiovascular offspring health. Epidemiological, clinical and experimental studies as well as human and animal model studies were included. RESULTS From n = 3.199 citations, n = 66 eligible studies were included. We selected n = 45 epidemiological as well as clinical studies and n = 21 experimental studies. In brief, pre-conceptional paternal risk factors, such as obesity, own birth weight, high-fat and low-protein diet, undernutrition, diabetes mellitus, hyperglycaemia, advanced age, smoking as well as environmental chemical exposure affect clearly metabolic and cardiovascular health of their offspring later in life. CONCLUSIONS There is emerging evidence that paternal risk factors, such as paternal obesity, diabetes mellitus, nutritional habits, advanced age and exposure to environmental chemicals or cigarette smoke, are clearly associated with adverse effects in metabolic and cardiovascular health in their offspring. Compared to maternal programming, pre-conceptional paternal factors might also have also a substantial effect in the sense of trans-generational programming of their offspring and need further research.
Collapse
Affiliation(s)
- Claudia Eberle
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda–University of Applied Sciences, Fulda, Germany
- * E-mail:
| | - Michaela F. Kirchner
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda–University of Applied Sciences, Fulda, Germany
| | - Raphaela Herden
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda–University of Applied Sciences, Fulda, Germany
| | - Stefanie Stichling
- Medicine with Specialization in Internal Medicine and General Medicine, Hochschule Fulda–University of Applied Sciences, Fulda, Germany
| |
Collapse
|
28
|
Gross N, Taylor T, Crenshaw T, Khatib H. The Intergenerational Impacts of Paternal Diet on DNA Methylation and Offspring Phenotypes in Sheep. Front Genet 2020; 11:597943. [PMID: 33250925 PMCID: PMC7674940 DOI: 10.3389/fgene.2020.597943] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/14/2020] [Indexed: 11/13/2022] Open
Abstract
Knowledge of non-genomic inheritance of traits is currently limited. Although it is well established that maternal diet influences offspring inheritance of traits through DNA methylation, studies on the impact of prepubertal paternal diet on DNA methylation are rare. This study aimed to evaluate the impact of prepubertal diet in Polypay rams on complex traits, DNA methylation, and transmission of traits to offspring. A total of 10 littermate pairs of F0 rams were divided so that one ram was fed a control diet, and the other was fed the control diet with supplemental methionine. Diet was associated with earlier age at puberty in treatment vs. control F0 rams. F0 treatment rams tended to show decreased pubertal weight compared to control rams; however, no differences were detected in overall growth. A total of ten F0 rams were bred, and the entire F1 generation was fed a control diet. Diet of F0 rams had a significant association with scrotal circumference (SC) and weight at puberty of F1 offspring. The paternal diet was not significantly associated with F1 ram growth or age at puberty. The DNA methylation of F0 ram sperm was assessed, and genes related to both sexual development (e.g., DAZAP1, CHD7, TAB1, MTMR2, CELSR1, MGAT1) and body weight (e.g., DUOX2, DUOXA2) were prevalent in the data. These results provide novel information about the mechanisms through which the prepubertal paternal diet may alter body weight at puberty and sexual development.
Collapse
Affiliation(s)
- Nicole Gross
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Todd Taylor
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Thomas Crenshaw
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Hasan Khatib
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
29
|
Male Factors: the Role of Sperm in Preimplantation Embryo Quality. Reprod Sci 2020; 28:1788-1811. [DOI: 10.1007/s43032-020-00334-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
|
30
|
Morgan HL, Aljumah A, Rouillon C, Watkins AJ. Paternal low protein diet and the supplementation of methyl-donors impact fetal growth and placental development in mice. Placenta 2020; 103:124-133. [PMID: 33120048 PMCID: PMC7907633 DOI: 10.1016/j.placenta.2020.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022]
Abstract
Introduction Paternal low-protein diet can alter sperm methylation status, fetal growth and program offspring ill-health, however its impact on the placenta remains poorly defined. Here we examine the influence paternal low-protein diet has on fetal and placental development and the additional impact of supplementary methyl-donors on fetoplacental physiology. Methods Male C57BL/6J mice were fed a control normal protein diet (NPD; 18% protein), a low-protein diet (LPD; 9% protein) or LPD with methyl-donor supplementation (MD-LPD; choline chloride, betaine, methionine, folic acid, vitamin B12) for a minimum of 8 weeks. Males were mated with 8–11 week old female C57BL/6J mice and fetal and placental tissue collected on embryonic day 17.5. Results Paternal LPD was associated with increased fetal weights compared to NPD and MD-LPD with 22% fetuses being above the 90th centile for fetal weight. However, LPD and MD-LPD placental weights were reduced when compared to NPD. Placentas from LPD fathers demonstrated a reduced junctional zone area and reduced free-fatty acid content. MD-LPD placentas did not mirror these finding, demonstrating an increased chorion area, a reduction in junctional-specific glycogen staining and reduced placental Dnmt3bexpression, none of which were apparent in either NPD or LPD placentas. Discussion A sub-optimal paternal diet can influence fetal growth and placental development, and dietary methyl-donor supplementation alters placental morphology and gene expression differentially to that observed with LPD alone. Understanding how paternal diet and micro-nutrient supplementation influence placental development is crucial for determining connections between paternal well-being and future offspring health. Paternal low protein diet (LPD) increased late gestation fetal weight. Supplementing the LPD with methyl donors (MD-LPD) normalised fetal weight. Placental weight and morphology are altered by both LPD and MD-LPD. Placental metabolite content and gene expression were perturbed by paternal LPD and MD-LPD.
Collapse
Affiliation(s)
- Hannah L Morgan
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK.
| | - Arwa Aljumah
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Charlène Rouillon
- INRAE, Fish Physiology and Genomics, Bat 16A, Campus de Beaulieu, Rennes, France
| | - Adam J Watkins
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK; Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK(1)
| |
Collapse
|
31
|
Janbek J, Kriegbaum M, Grand MK, Specht IO, Lind BS, Andersen CL, Heitmann BL. The Copenhagen Primary Care Laboratory Pregnancy (CopPreg) database. BMJ Open 2020; 10:e034318. [PMID: 32448791 PMCID: PMC7252999 DOI: 10.1136/bmjopen-2019-034318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
PURPOSE The Copenhagen Primary Care Laboratory Pregnancy (CopPreg) database was established based on data from The Danish Medical Birth Register and the Copenhagen Primary Care Laboratory (CopLab) database. The aim was to provide a biomedical and epidemiological data resource for research in early disease programming (eg, parental clinical biomarker levels and pregnancy/ birth outcomes or long-term health in the offspring). PARTICIPANTS The cohort consisted in total of 203 608 women (with 340 891 pregnancies) who gave birth to 348 248 children and with 200 590 related fathers. In this paper, we focused on women and fathers who had clinical test requisitions prior to and during pregnancy, and on all children. Thus, the cohort in focus consisted of 203 054 pregnancies with requisitions on 147 045 pregnant women, 39 815 fathers with requisitions during periconception and 65 315 children with requisitions. FINDINGS TO DATE In addition to information on pregnancy and birth health status and general socio-demographic data, over 2.2 million clinically relevant test results were available for pregnancies with requisitions, over 1.5 million for children and over 600 000 test results were available for the fathers with requisitions during periconception. These were ordered by general practitioners in the primary care setting only and included general blood tests, nutritional biomarkers (macronutrients and micronutrients) and hormone tests. Information on tests related to infections, allergies, heart and lung function and sperm analyses (fathers) were also available. FUTURE PLANS The CopPreg database provides ready to use and valid data from already collected, objectively measured and analysed clinical tests. With several research projects planned, we further invite national and international researchers to use this vast data resource. In a coming paper, we will explore and discuss the indication bias in our cohort.
Collapse
Affiliation(s)
- Janet Janbek
- Danish Dementia Research Centre, Department of Neurology, The Neuroscience Centre, Rigshospitalet, Copenhagen, Denmark
- Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Margit Kriegbaum
- Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Mia Klinten Grand
- Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Ina Olmer Specht
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| | - Bent Struer Lind
- Department of Clinical Biochemistry, Copenhagen University Hospital, Hvidovre, Denmark
| | - Christen Lykkegaard Andersen
- Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Berit Lilienthal Heitmann
- Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| |
Collapse
|
32
|
Thayer ZM, Rutherford J, Kuzawa CW. The Maternal Nutritional Buffering Model: an evolutionary framework for pregnancy nutritional intervention. EVOLUTION MEDICINE AND PUBLIC HEALTH 2020; 2020:14-27. [PMID: 32015877 PMCID: PMC6990448 DOI: 10.1093/emph/eoz037] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/17/2020] [Indexed: 02/07/2023]
Abstract
Evidence that fetal nutrition influences adult health has heightened interest in nutritional interventions targeting pregnancy. However, as is true for other placental mammals, human females have evolved mechanisms that help buffer the fetus against short-term fluctuations in maternal diet and energy status. In this review, we first discuss the evolution of increasingly elaborate vertebrate strategies of buffering offspring from environmental fluctuations during development, including the important innovation of the eutherian placenta. We then present the Maternal Nutritional Buffering Model, which argues that, in contrast to many micronutrients that must be derived from dietary sources, the effects of short-term changes in maternal macronutrient intake during pregnancy, whether due to a deficit or supplementation, will be minimized by internal buffering mechanisms that work to ensure a stable supply of essential resources. In contrast to the minimal effects of brief macronutrient supplementation, there is growing evidence that sustained improvements in early life and adult pre-pregnancy nutrition could improve birth outcomes in offspring. Building on these and other observations, we propose that strategies to improve fetal macronutrient delivery will be most effective if they modify the pregnancy metabolism of mothers by targeting nutrition prior to conception and even during early development, as a complement to the conventional focus on bolstering macronutrient intake during pregnancy itself. Our model leads to the prediction that birth weight will be more strongly influenced by the mother’s chronic pre-pregnancy nutrition than by pregnancy diet, and highlights the need for policy solutions aimed at optimizing future, intergenerational health outcomes. Lay summary: We propose that strategies to improve fetal macronutrient delivery will be most effective if they modify the pregnancy metabolism of mothers by targeting nutrition prior to conception and even during early development, as a complement to the conventional focus on bolstering macronutrient intake during pregnancy itself.
Collapse
Affiliation(s)
- Zaneta M Thayer
- Department of Anthropology, Dartmouth College, Hinman Box 6047, Hanover, NH 03755, USA
| | - Julienne Rutherford
- Department of Women, Children and Family Health Science, University of Illinois Chicago, 845 S. Damen Ave., MC 802, Chicago, IL 60612, USA
| | - Christopher W Kuzawa
- Department of Anthropology and Institute for Policy Research, Northwestern University, 1810 Hinman Ave, Evanston, IL 60208, USA
| |
Collapse
|
33
|
McPherson NO, Lane M. Metformin treatment of high-fat diet-fed obese male mice restores sperm function and fetal growth, without requiring weight loss. Asian J Androl 2020; 22:560-568. [PMID: 32098932 PMCID: PMC7705976 DOI: 10.4103/aja.aja_141_19] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Male obesity is associated with subfertility and increased disease risk of offspring. It is unknown if effects can be reversed through pharmacological interventions. Five- to 6-week-old C57BL6 male mice were fed control diet (n = 10, CD) or high-fat diet (n = 20, HFD) for 16 weeks. Animals fed with a HFD were then allocated to continuation of HFD (n = 8) or HFD with metformin 28 mg kg−1 day−1 (n = 8) for 6 weeks. Animals fed with CD continued on a CD (n = 9). Males were mated with fertile C57BL6 females for the assessment of pregnancy and fetal growth. Sperm motility, spermatozoa and testicular morphology, sperm-zona pellucida binding, sperm reactive oxygen species (ROS) (intracellular [DCFDA], superoxide [MSR], and oxidative DNA lesions [8OHdG]), and mitochondrial membrane potential (JC1) were assessed. Metformin treatment of HFD males improved glucose tolerance (+12%, P < 0.05) and reduced Homeostatic Model Assessment of Insulin Resistance (HOMA-IR; −36%, P < 0.05). This occurred in the absence of a change in body weight or adiposity. Metformin treatment of HFD-fed males restored testicular morphology (+33%, P < 0.05), sperm motility (+66%, P < 0.05), sperm–zona pellucida binding (+25%, P < 0.05), sperm intracellular ROS concentrations (−32%, P < 0.05), and oxidative DNA lesions (−45%, P < 0.05) to the levels of the CD males. Metformin treatment of HFD fathers increased fetal weights and lengths compared with those born to HFD fathers (+8%, P < 0.05), with fetal lengths restored to those of fetuses of CD males. Short-term metformin treatment in men who are obese could be a potential intervention for the treatment of subfertility, without the need for a reduction in body weight/adiposity.
Collapse
Affiliation(s)
- Nicole O McPherson
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide 5005, Australia.,Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide 5005, Australia.,Repromed, Dulwich 5065, Australia
| | - Michelle Lane
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide 5005, Australia.,Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide 5005, Australia.,Monash IVF Group, Melbourne 3000, Australia
| |
Collapse
|
34
|
Silva LBAR, Pinheiro-Castro N, Novaes GM, Pascoal GDFL, Ong TP. Bioactive food compounds, epigenetics and chronic disease prevention: Focus on early-life interventions with polyphenols. Food Res Int 2019; 125:108646. [DOI: 10.1016/j.foodres.2019.108646] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
|
35
|
Morgan HL, Paganopoulou P, Akhtar S, Urquhart N, Philomin R, Dickinson Y, Watkins AJ. Paternal diet impairs F1 and F2 offspring vascular function through sperm and seminal plasma specific mechanisms in mice. J Physiol 2019; 598:699-715. [PMID: 31617219 DOI: 10.1113/jp278270] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS A low protein diet had minimal effects on paternal cardiovascular function or renin-angiotensin system activity. Paternal low protein diet modified F1 neonatal and adult offspring renin-angiotensin system activity and cardiovascular function in a sperm and/or seminal plasma specific manner. Paternal low protein diet modified F1 male offspring testicular expression of central epigenetic regulators. Significant changes in F2 neonatal offspring growth and tissue angiotensin-converting enzyme activity were programmed by paternal low protein diet in a sperm and/or seminal plasma specific manner. ABSTRACT Although the impact of maternal diet on adult offspring health is well characterized, the role that a father's diet has on his offspring's health remains poorly defined. We establish the significance of a sup-optimal paternal low protein diet for offspring vascular homeostasis and define the sperm and seminal plasma specific programming effects on cardiovascular health. Male C57BL6 mice were fed either a control normal protein diet (NPD; 18% protein) or an isocaloric low protein diet (LPD; 9% protein) for a minimum of 7 weeks. Using artificial insemination, in combination with vasectomized male mating, we generated offspring derived from either NPD or LPD sperm (devoid of seminal plasma) but in the presence of NPD or LPD seminal plasma (devoid of sperm). We observed that either LPD sperm or seminal fluid at conception impaired adult offspring vascular function in response to both vasoconstrictors and dilators. Underlying these changes in vascular function were significant changes in serum, lung and kidney angiotensin-converting enzyme (ACE) activity, established in F1 offspring from 3 weeks of age, maintained into adulthood and present also within juvenile F2 offspring. Furthermore, we observed differential expression of multiple central renin-angiotensin system regulators in adult offspring kidneys. Finally, paternal diet modified the expression profiles of central epigenetic regulators of DNA methylation, histone modifications and RNA methylation in adult F1 male testes. These novel data reveal the impact of sub-optimal paternal nutrition on offspring cardiovascular well-being, programming offspring cardiovascular function through both sperm and seminal plasma specific mechanisms over successive generations.
Collapse
Affiliation(s)
- Hannah L Morgan
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, UK
| | - Panaigota Paganopoulou
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, UK
| | - Sofia Akhtar
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Natalie Urquhart
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Ranmini Philomin
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Yasmin Dickinson
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Adam J Watkins
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, UK.,School of Life and Health Sciences, Aston University, Birmingham, UK
| |
Collapse
|
36
|
Dietary Micronutrient Supplementation for 12 Days in Obese Male Mice Restores Sperm Oxidative Stress. Nutrients 2019; 11:nu11092196. [PMID: 31547309 PMCID: PMC6770166 DOI: 10.3390/nu11092196] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022] Open
Abstract
Male obesity, which often co-presents with micronutrient deficiencies, is associated with sub-fertility. Here we investigate whether short-term dietary supplementation of micronutrients (zinc, selenium, lycopene, vitamins E and C, folic acid, and green tea extract) to obese mice for 12 days (designed to span the epididymal transit) could improve sperm quality and fetal outcomes. Five-week-old C57BL6 males were fed a control diet (CD, n = 24) or high fat diet (HFD, n = 24) for 10 weeks before allocation to the 12-day intervention of maintaining their original diets (CD, n = 12, HFD n = 12) or with micronutrient supplementation (CD + S, n = 12, HFD + S, n = 12). Measures of sperm quality (motility, morphology, capacitation, binding), sperm oxidative stress (DCFDA, MSR, and 8OHdG), early embryo development (2-cell cleavage, 8OHdG), and fetal outcomes were assessed. HFD + S males had reduced sperm intracellular reactive oxygen species (ROS) concentrations and 8OHdG lesions, which resulted in reduced 8OHdG lesions in the male pronucleus, increased 2-cell cleavage rates, and partial restoration of fetal weight similar to controls. Sub-fertility associated with male obesity may be restored with very short-term micronutrient supplementation that targets the timing of the transit of sperm through the epididymis, which is the developmental window where sperm are the most susceptible to oxidative damage.
Collapse
|
37
|
Nixon B, De Iuliis GN, Dun MD, Zhou W, Trigg NA, Eamens AL. Profiling of epididymal small non-protein-coding RNAs. Andrology 2019; 7:669-680. [PMID: 31020794 DOI: 10.1111/andr.12640] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/04/2019] [Accepted: 03/30/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Our understanding of epididymal physiology and function has been transformed over the three decades in which the International Meeting Series on the Epididymis has been hosted. This transformation has occurred along many fronts, but among the most significant advances has been the unexpected discovery of the diversity of small non-protein-coding RNAs (sRNAs) expressed in the epididymal epithelium and differentially accumulated in the luminal population of spermatozoa. OBJECTIVES Here we survey recent literature pertaining to profiling the sRNA landscape of the mammalian epididymis with the goal of demonstrating the contribution that these key regulatory elements, and their associated pathways, make to epididymal physiology and sperm maturation. RESULTS AND DISCUSSION High throughput sequencing strategies have fueled an unprecedented advance in our understanding of RNA biology. In the last decade, such high throughput profiling tools have been increasingly applied to study the mammalian epididymis, presaging the discovery of diverse classes of sRNA expressed along the length of the tract. Among the best studied sRNA classes are the microRNAs (miRNA), a sRNA species shown to act in concert with endocrine signals to fine-tune the segmental patterning of epididymal gene expression. In addition to performing this homeostatic role, epithelial cell-derived sRNAs also selectively accumulate into the epididymosomes and spermatozoa that occupy the duct lumen. This exciting discovery alludes to a novel form of intracellular communication that contributes to the establishment of the sperm epigenome and its modification under conditions of paternal stress. CONCLUSION Compelling literature has identified sRNAs as a crucial regulatory tier that allows the epididymis to fulfill its combined roles of sperm transport, maturation, and storage. Continued research in this emerging field will contribute to our growing understanding of the etiology of male factor infertility and potentially allow for the future design of rational therapeutic options for these individuals.
Collapse
Affiliation(s)
- B Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia.,Reproduction and Pregnancy Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - G N De Iuliis
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia.,Reproduction and Pregnancy Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - M D Dun
- Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,Cancer Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - W Zhou
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia.,Reproduction and Pregnancy Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - N A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia.,Reproduction and Pregnancy Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - A L Eamens
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
38
|
Dupont C, Kappeler L, Saget S, Grandjean V, Lévy R. Role of miRNA in the Transmission of Metabolic Diseases Associated With Paternal Diet-Induced Obesity. Front Genet 2019; 10:337. [PMID: 31057600 PMCID: PMC6482346 DOI: 10.3389/fgene.2019.00337] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/29/2019] [Indexed: 12/25/2022] Open
Abstract
The concept of Developmental Origins of Health and Diseases (DOHaD) recognizes that an unfavorable maternal environment alters the developmental trajectory of the fetus and can lead to long-term risk of developing chronic noncommunicable diseases. More recently, the concept of a paternal transmission [Paternal Origins of Health and Diseases (POHaD)] has emerged stressing the impact of paternal overweight or obesity on offspring's health and development. While very few examples of paternal epigenetic inheritance of metabolic disorders have been evidenced in human, many experimental mouse models based on high-fat diet (HFD)-induced paternal obesity have been developed to breakdown molecular mechanisms involved in the process. Besides DNA methylation and chromatin structure, sperm short noncoding RNAs have been considered as the main epigenetic vector of inheritance of paternally environmentally induced changes. Among them, sperm miRNAs are one particular subspecies sensitive to environmental changes and obesity can modify the sperm miRNA profile. Once delivered into the zygote, these molecules might induce epigenetic modifications in the embryo, thereby leading to consequences for fetus development and offspring physical and metabolic health later on in life. Furthermore, some data also suggest that metabolic pathologies may be intergenerationally or transgenerationally transmitted.
Collapse
Affiliation(s)
- Charlotte Dupont
- Sorbonne Université, Inserm, Centre de Recherche St-Antoine, CRSA, AP-HP, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| | - Laurent Kappeler
- Sorbonne Université, INSERM, Institute of Cardiometabolism, Centre de Recherche St-Antoine, CRSA, Paris, France
| | - Sarah Saget
- Sorbonne Université, INSERM, Institute of Cardiometabolism, Centre de Recherche St-Antoine, CRSA, Paris, France
| | - Valérie Grandjean
- Inserm U1065, Team Control of Gene Expression (10), Université Cote d’Azur, Nice, France
| | - Rachel Lévy
- Sorbonne Université, Inserm, Centre de Recherche St-Antoine, CRSA, AP-HP, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| |
Collapse
|
39
|
Velazquez MA, Fleming TP, Watkins AJ. Periconceptional environment and the developmental origins of disease. J Endocrinol 2019; 242:T33-T49. [PMID: 30707679 DOI: 10.1530/joe-18-0676] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/01/2019] [Indexed: 01/08/2023]
Abstract
The concept emerging from Professor David Barker’s seminal research on the developmental origins of later-life disease has progressed in many directions since it was first published. One critical question being when during gestation might environment alter the developmental programme with such enduring consequences. Here, we review the growing consensus from clinical and animal research that the period around conception, embracing gamete maturation and early embryogenesis might be the most vulnerable period. We focus on four types of environmental exposure shown to modify periconceptional reproduction and offspring development and health: maternal overnutrition and obesity; maternal undernutrition; paternal diet and health; and assisted reproductive technology. These conditions may act through diverse epigenetic, cellular and physiological mechanisms to alter gene expression and cellular signalling and function in the conceptus affecting offspring growth and metabolism leading to increased risk for cardiometabolic and neurological disease in later life.
Collapse
Affiliation(s)
- Miguel A Velazquez
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom of Great Britain and Northern Ireland
| | - Tom P Fleming
- Biological Sciences, University of Southampton, Southampton, United Kingdom of Great Britain and Northern Ireland
| | - Adam J Watkins
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
40
|
Cheng PJ, Pastuszak AW, Hotaling JM. Is it time to start folate supplementation in men? The effect of paternal folate status on embryonic growth. Fertil Steril 2019; 111:251-252. [DOI: 10.1016/j.fertnstert.2018.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 11/27/2022]
|
41
|
Morgan HL, Watkins AJ. Transgenerational Impact of Environmental Change. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1200:71-89. [PMID: 31471795 DOI: 10.1007/978-3-030-23633-5_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ability to adapt to changing environmental conditions is critical for any species to survive. Many environmental changes occur too rapidly for an organism's genome to adapt in time. Accordingly, being able to modify either its own phenotype, or the phenotype of its offspring to better suit future anticipated environmental conditions could afford an organism a significant advantage. However, a range of animal models and human epidemiological data sets are now showing that environmental factors such as changes in the quality or quantity of an individual's diet, temperature, stress or exposure to pollutants can all adversely affect the quality of parental gametes, the development of the preimplantation embryo and the health and wellbeing of offspring over multiple generations. This chapter will examine transgenerational effects of both maternal and paternal environmental factors on offspring development and wellbeing in both human and animal model studies. Changes in the epigenetic status of either parental or grand-parental gametes provide one candidate mechanism through which the impacts of environmental experience can be passed from one generation to another. This chapter will therefore also focus on the impact of parental and grand-parental diet on epigenetic transgenerational inheritance and offspring phenotype.
Collapse
Affiliation(s)
- Hannah L Morgan
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, UK
| | - Adam J Watkins
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
42
|
Antioxidants modulation of sperm genome and epigenome damage: Fact or fad? Converging evidence from animal and human studies. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2018. [DOI: 10.1016/j.mefs.2018.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
43
|
Fleming TP, Watkins AJ, Velazquez MA, Mathers JC, Prentice AM, Stephenson J, Barker M, Saffery R, Yajnik CS, Eckert JJ, Hanson MA, Forrester T, Gluckman PD, Godfrey KM. Origins of lifetime health around the time of conception: causes and consequences. Lancet 2018; 391:1842-1852. [PMID: 29673874 PMCID: PMC5975952 DOI: 10.1016/s0140-6736(18)30312-x] [Citation(s) in RCA: 651] [Impact Index Per Article: 108.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 12/03/2017] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
Abstract
Parental environmental factors, including diet, body composition, metabolism, and stress, affect the health and chronic disease risk of people throughout their lives, as captured in the Developmental Origins of Health and Disease concept. Research across the epidemiological, clinical, and basic science fields has identified the period around conception as being crucial for the processes mediating parental influences on the health of the next generation. During this time, from the maturation of gametes through to early embryonic development, parental lifestyle can adversely influence long-term risks of offspring cardiovascular, metabolic, immune, and neurological morbidities, often termed developmental programming. We review periconceptional induction of disease risk from four broad exposures: maternal overnutrition and obesity; maternal undernutrition; related paternal factors; and the use of assisted reproductive treatment. Studies in both humans and animal models have demonstrated the underlying biological mechanisms, including epigenetic, cellular, physiological, and metabolic processes. We also present a meta-analysis of mouse paternal and maternal protein undernutrition that suggests distinct parental periconceptional contributions to postnatal outcomes. We propose that the evidence for periconceptional effects on lifetime health is now so compelling that it calls for new guidance on parental preparation for pregnancy, beginning before conception, to protect the health of offspring.
Collapse
Affiliation(s)
- Tom P Fleming
- Biological Sciences, University of Southampton, Southampton, UK
| | - Adam J Watkins
- School of Medicine, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham, UK
| | - Miguel A Velazquez
- School of Natural and Environmental Sciences, Newcastle University, Newcastle, UK
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine and Newcastle University Institute for Ageing, Newcastle University, Newcastle, UK
| | - Andrew M Prentice
- MRC Unit, The Gambia and MRC International Nutrition Group, London School of Hygiene & Tropical Medicine, London, UK
| | - Judith Stephenson
- UCL EGA Institute for Women's Health, Faculty of Population Health Sciences, University College London, London, UK
| | - Mary Barker
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Richard Saffery
- Cancer & Disease Epigenetics, Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | | | - Judith J Eckert
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Mark A Hanson
- Institute of Developmental Sciences, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Terrence Forrester
- University of the West Indies Solutions for Developing Countries, The University of the West Indies, Mona, Jamaica
| | - Peter D Gluckman
- Liggins Institute, University of Auckland, Auckland, New Zealand; Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK; Institute of Developmental Sciences, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| |
Collapse
|
44
|
Kwan STC, King JH, Grenier JK, Yan J, Jiang X, Roberson MS, Caudill MA. Maternal Choline Supplementation during Normal Murine Pregnancy Alters the Placental Epigenome: Results of an Exploratory Study. Nutrients 2018; 10:nu10040417. [PMID: 29597262 PMCID: PMC5946202 DOI: 10.3390/nu10040417] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022] Open
Abstract
The placental epigenome regulates processes that affect placental and fetal development, and could be mediating some of the reported effects of maternal choline supplementation (MCS) on placental vascular development and nutrient delivery. As an extension of work previously conducted in pregnant mice, the current study sought to explore the effects of MCS on various epigenetic markers in the placenta. RNA and DNA were extracted from placentas collected on embryonic day 15.5 from pregnant mice fed a 1X or 4X choline diet, and were subjected to genome-wide sequencing procedures or mass-spectrometry-based assays to examine placental imprinted gene expression, DNA methylation patterns, and microRNA (miRNA) abundance. MCS yielded a higher (fold change = 1.63-2.25) expression of four imprinted genes (Ampd3, Tfpi2, Gatm and Aqp1) in the female placentas and a lower (fold change = 0.46-0.62) expression of three imprinted genes (Dcn, Qpct and Tnfrsf23) in the male placentas (false discovery rate (FDR) ≤ 0.05 for both sexes). Methylation in the promoter regions of these genes and global placental DNA methylation were also affected (p ≤ 0.05). Additionally, a lower (fold change = 0.3; Punadjusted = 2.05 × 10-4; FDR = 0.13) abundance of miR-2137 and a higher (fold change = 1.25-3.92; p < 0.05) expression of its target genes were detected in the 4X choline placentas. These data demonstrate that the placental epigenome is responsive to maternal choline intake during murine pregnancy and likely mediates some of the previously described choline-induced effects on placental and fetal outcomes.
Collapse
Affiliation(s)
| | - Julia H King
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA.
| | - Jennifer K Grenier
- RNA Sequencing Core, Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA.
| | - Jian Yan
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA.
| | - Xinyin Jiang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA.
- Department of Health and Nutrition Sciences, Brooklyn College, Brooklyn, NY 11210, USA.
| | - Mark S Roberson
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA.
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
45
|
Illum LRH, Bak ST, Lund S, Nielsen AL. DNA methylation in epigenetic inheritance of metabolic diseases through the male germ line. J Mol Endocrinol 2018; 60:R39-R56. [PMID: 29203518 DOI: 10.1530/jme-17-0189] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 12/26/2022]
Abstract
The global rise in metabolic diseases can be attributed to a complex interplay between biology, behavior and environmental factors. This article reviews the current literature concerning DNA methylation-based epigenetic inheritance (intergenerational and transgenerational) of metabolic diseases through the male germ line. Included are a presentation of the basic principles for DNA methylation in developmental programming, and a description of windows of susceptibility for the inheritance of environmentally induced aberrations in DNA methylation and their associated metabolic disease phenotypes. To this end, escapees, genomic regions with the intrinsic potential to transmit acquired paternal epigenetic information across generations by escaping the extensive programmed DNA demethylation that occurs during gametogenesis and in the zygote, are described. The ongoing descriptive and functional examinations of DNA methylation in the relevant biological samples, in conjugation with analyses of non-coding RNA and histone modifications, hold promise for improved delineation of the effect size and mechanistic background for epigenetic inheritance of metabolic diseases.
Collapse
Affiliation(s)
| | - Stine Thorhauge Bak
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Endocrinology and Diabetes, Aarhus University Hospital, Aarhus, Denmark
| | - Sten Lund
- Department of Clinical Medicine, Endocrinology and Diabetes, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
46
|
Fontelles CC, da Cruz RS, Hilakivi-Clarke L, de Assis S, Ong TP. Developmental Origins of Breast Cancer: A Paternal Perspective. Methods Mol Biol 2018; 1735:91-103. [PMID: 29380308 DOI: 10.1007/978-1-4939-7614-0_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The developmental origins of breast cancer have been considered predominantly from a maternal perspective. Although accumulating evidence suggests a paternal programming effect on metabolic diseases, the potential impact of fathers' experiences on their daughters' breast cancer risk has received less attention. In this chapter, we focus on the developmental origins of breast cancer and examine the emerging evidence for a role of fathers' experiences.
Collapse
Affiliation(s)
- Camile Castilho Fontelles
- Department of Food and Experimental Nutrition, Food Research Center (FoRC), Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Georgetown University Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | | | | | - Sonia de Assis
- Georgetown University Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Thomas Prates Ong
- Department of Food and Experimental Nutrition, Food Research Center (FoRC), Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
47
|
Paternal low protein diet programs preimplantation embryo gene expression, fetal growth and skeletal development in mice. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1371-1381. [DOI: 10.1016/j.bbadis.2017.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 12/25/2022]
|
48
|
Sales VM, Ferguson-Smith AC, Patti ME. Epigenetic Mechanisms of Transmission of Metabolic Disease across Generations. Cell Metab 2017; 25:559-571. [PMID: 28273478 PMCID: PMC5404272 DOI: 10.1016/j.cmet.2017.02.016] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Both human and animal studies indicate that environmental exposures experienced during early life can robustly influence risk for adult disease. Moreover, environmental exposures experienced by parents during either intrauterine or postnatal life can also influence the health of their offspring, thus initiating a cycle of disease risk across generations. In this Perspective, we focus on epigenetic mechanisms in germ cells, including DNA methylation, histone modification, and non-coding RNAs, which collectively may provide a non-genetic molecular legacy of prior environmental exposures and influence transcriptional regulation, developmental trajectories, and adult disease risk in offspring.
Collapse
Affiliation(s)
- Vicencia Micheline Sales
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center and Harvard Medical School, One Joslin Place, Sixth Floor, Boston, MA 02215, USA
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Mary-Elizabeth Patti
- Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center and Harvard Medical School, One Joslin Place, Sixth Floor, Boston, MA 02215, USA.
| |
Collapse
|
49
|
Lucas ES, Watkins AJ. The Long-Term Effects of the Periconceptional Period on Embryo Epigenetic Profile and Phenotype; The Paternal Role and His Contribution, and How Males Can Affect Offspring's Phenotype/Epigenetic Profile. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1014:137-154. [PMID: 28864989 DOI: 10.1007/978-3-319-62414-3_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The number of adults afflicted with heart disease, obesity and diabetes, central components of metabolic disorder, has grown rapidly in recent decades, affecting up to one quarter of the world's population. Typically, these diseases are attributed to lifestyle factors such as poor diet, lack of exercise and smoking. However, studies have now identified strong associations between patterns of growth during foetal and neonatal life and an increase predisposition towards developing heart disease, obesity and diabetes in adult life. While the connection between a mother's diet and the long-term health of her offspring has been studied in great detail, our understanding of whether offspring health might be affected by a father's diet remains limited. Greater insight into the impact that paternal nutrition has on sperm quality, epigenetic status and potential offspring programming mechanisms is needed to redress this parental-programming knowledge imbalance. Disturbances in paternal reproductive epigenetic status represents one key mechanism linking paternal diet with the programing of offspring development and adult health, as many enzymatic processes involved in epigenetic regulation use metabolic intermediates to modify DNA and histones. Here, poor paternal nutrition could result in perturbed sperm and testicular epigenetic status, impacting on post-fertilisation gene transcriptional regulation and protein expression in offspring tissues, resulting in increased incidences of metabolic disorder in adult life.
Collapse
Affiliation(s)
- Emma S Lucas
- Division of Reproductive Health, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
| | - Adam J Watkins
- Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|