1
|
Patterson RA, Cho NA, Fernandes TS, Tuplin EWN, Lowry DE, Silva GAV, Reimer RA. Effects of a paternal diet high in animal protein (casein) versus plant protein (pea protein with added methionine) on offspring metabolic and gut microbiota outcomes in rats. Appl Physiol Nutr Metab 2025; 50:1-15. [PMID: 39689296 DOI: 10.1139/apnm-2024-0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Evidence suggests that paternal diet can influence offspring metabolic health intergenerationally but whether dietary animal and plant proteins differ in their impact on fathers and their offspring is not known. Our objective was to examine the effects of a paternal diet high in casein versus pea protein on fathers and their offspring. Five-week-old male rats were fed: (1) control, (2) high animal protein (AP, 36.1% of energy as casein), or (3) high plant protein (PP, 36.1% of energy as pea protein with added methionine) diets for 8-11 weeks before being mated. Offspring were challenged with a high fat/sucrose diet (HFD) from 10 to 16 weeks of age. Metabolic and microbial outcomes were assessed in both generations. In fathers fed PP diet, enhanced insulin sensitivity and lower liver triglycerides were seen alongside altered hepatic microRNA expression and gut microbial profiles. Few changes were seen in their offspring. In contrast, the paternal AP diet influenced adult offspring hepatic microRNA expression and programmed a latent increase in adiposity, dysregulated satiety hormones, and modified gut microbial composition in their adult offspring that occurred following the HFD. Overall, a diet high in pea protein with added methionine demonstrated protective effects on biomarkers of metabolic health in the fathers but led to minimal effects on the offspring while a paternal diet high in casein led to evidence of an increase in characteristics of metabolic dysfunction in their adult offspring when unmasked by exposure to a HFD for 6 weeks.
Collapse
Affiliation(s)
- Riley A Patterson
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Nicole A Cho
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Tyra S Fernandes
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Erin W Noye Tuplin
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Dana E Lowry
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Gabriel A Venegas Silva
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
2
|
Correa-Navarro VA, Romo-Morales GDC, Sánchez-Palafox JE, Rodríguez-Ríos D, Molina-Torres J, Ramírez-Chávez E, Zaina S, Lund G. A Survey of Fatty Acid Content of the Male Reproductive System in Mice Supplemented With Arachidonic Acid. J Lipids 2024; 2024:3351340. [PMID: 39734583 PMCID: PMC11671656 DOI: 10.1155/jl/3351340] [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: 07/29/2024] [Accepted: 11/15/2024] [Indexed: 12/31/2024] Open
Abstract
Paternal exposure to high-fat diets or individual fatty acids (FAs) including arachidonic acid (AA) modifies progeny traits by poorly understood mechanisms. Specific male reproductive system FAs may be involved in paternal inheritance, as they can modify a range of cellular components, including the epigenome. Our objective was to determine FAs in compartments of the male reproductive system that potentially affect ejaculate composition-right and left testicular interstitial fluid (TIF), vesicular gland fluid (VGF), and epididymal adipose tissue (EAT)-in mice exposed to AA or vehicle daily for 10 days (n = 9-10/group). Whole blood (WB) and interscapular brown adipose tissue (IBAT) FA profiles were used as reference. AA significantly affected only VGF FAs relative to vehicle, that is, increased and decreased levels of arachidic and docosahexaenoic acid, respectively, versus vehicle (0.28% ± 0.01% and 0.23% ± 0.03%, respectively, p = 0.049, and 2.42% ± 0.47% and 3.00% ± 0.58%, respectively, p = 0.041). AA affected distinct FAs in WB. Additionally, we uncovered AA-dependent and AA-independent FA laterality. Myristic acid was higher in AA-exposed left versus right TIF (0.68% ± 0.35% and 0.60% ± 0.11%, respectively, p = 0.004). Right TIF contained higher oleic and linoleic acid and lower stearic acid than left TIF (29.01% ± 3.07% and 24.00% ± 2.18%, respectively, p = 0.005; 9.14% ± 1.88% and 7.05% ± 1.36%, respectively, p = 0.005; and 21.90% ± 2.92% and 26.01% ± 2.46%, respectively, p = 0.036), irrespective of exposure to AA. The TIF oleic/stearic acid ratio suggested higher Stearoyl-CoA Desaturase 1 activity in the right versus the left testis (1.35 ± 0.32 and 1.00 ± 0.17, respectively, p = 1.0 × 10-4). Multitissue comparisons revealed that TIF and VGF FA profiles were distinct from WB, EAT, or IBAT counterparts, suggesting tissue-specific FA fingerprints. In conclusion, AA modulated selected VGF long-chain FAs that may impact on uterine inflammation and subsequent embryonic development. AA altered local FA synthesis or selective uptake, rather than eliciting passive uptake from WB. Additionally, we uncover a significant laterality of testis FAs that may result in asymmetric sperm cell phenotypes.
Collapse
Affiliation(s)
- Viridiana Abigail Correa-Navarro
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Gloria del Carmen Romo-Morales
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Jaime Eduardo Sánchez-Palafox
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Dalia Rodríguez-Ríos
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Jorge Molina-Torres
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Enrique Ramírez-Chávez
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| | - Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, 20 de Enero 929, Leon, Guanajuato, Mexico
| | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Km 9.6 Libramiento Norte, Carretera Irapuato-León, Irapuato, Guanajuato 36824, Mexico
| |
Collapse
|
3
|
Mao Y, Meng Y, Zou K, Qin N, Wang Y, Yan J, Chen P, Cheng Y, Shi W, Zhou C, Chen H, Sheng J, Liu X, Pan J, Huang H. Advanced paternal age exacerbates neuroinflammation in offspring via m6A modification-mediated intergenerational inheritance. J Neuroinflammation 2024; 21:249. [PMID: 39367406 PMCID: PMC11453047 DOI: 10.1186/s12974-024-03248-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND The trend of postponing childbearing age is prevalent worldwide. Advanced paternal age (APA) is associated with adverse pregnancy outcomes and offspring health. However, the underlying mechanism by which paternal aging affects the risk of offspring neuropsychiatric disorders is unclear. Our study aims to explore the behavioral phenotypes and the pathologic epigenetic alterations of APA offspring inherited from aging sperm. METHODS Behavioral tests, ELISA assay, immunofluorescence and western blotting were performed on offspring mice. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA immunoprecipitation sequencing (RIP-seq) were used to investigate the modified N6-methyladenosine (m6A) profiles of paternal sperm and offspring hippocampus. Intervention of gene expression by lentivirus and adeno-associated virus in both vivo and vitro examined the potential therapeutic targets of intergenerational inherited neuroinflammation. RESULTS In our study, APA offspring exhibit cognitive impairment and autism-like behavior. An increase in neuroinflammation in APA offspring is associated with microglial overactivation, which manifests as abnormal morphology and augmented engulfment. MeRIP-seq of F0 sperm and F1 hippocampus reveal that Nr4a2 is hypermethylated with decreased expression in APA offspring involving in synaptic plasticity and microglial function. In addition, Ythdc1, an m6A reader protein, is markedly elevated in aging sperm and remains elevated in adult hippocampus of APA group. Enhanced Ythdc1 recognizes and suppresses the hypermethylated Nr4a2, thereby contributing to the abnormal phenotype in offspring. The overexpression of Ythdc1 triggers microglial activation in vitro and its suppression in the hippocampus of APA progeny alleviates behavioral aberrations and attenuates neuroinflammation. CONCLUSION Our study provides additional evidence of the abnormal behavioral phenotypes of APA offspring and reveals potential epigenetic inheritance signatures and targeted genes for future research.
Collapse
Affiliation(s)
- Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China
| | - Yicong Meng
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kexin Zou
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ningxin Qin
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinyu Wang
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Yan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - PinJia Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Weihui Shi
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Chengliang Zhou
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huixi Chen
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianzhong Sheng
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Xinmei Liu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China.
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China.
| |
Collapse
|
4
|
Jiang Y, Zhang H, Chen S, Ewart S, Holloway JW, Arshad H, Karmaus W. Intergenerational association of DNA methylation between parents and offspring. Sci Rep 2024; 14:19812. [PMID: 39191877 DOI: 10.1038/s41598-024-69317-3] [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: 09/14/2023] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Early patterning of DNA methylation (DNAm) may play an important role in later disease development. To better understand intergenerational epigenetic inheritance, we investigated the correlation between DNAm in blood in mother-newborn and in father-newborn pairs in the Isle of Wight (IoW) birth cohort. For parent-newborn pairs (n = 48), offspring DNAm was measured in cord blood and the parent's DNAm in whole blood. Mothers' DNAm was analyzed at birth (Guthrie card), age 18, early and late pregnancy respectively, and fathers' DNAm was measured during the mother's pregnancy. Linear regressions were applied to assess the intergenerational correlation of parental DNAm with that of offspring. Among various pairs of mother-newborn and father-newborn DNAm, the pairs where the mothers' DNAm was measured at age 18 years exhibited the highest number of CpGs with significant intergenerational correlation in DNAm, with 1829 CpGs (0.54%) of the 338,526 CpGs studied (FDR < 0.05). Amongst these 1829 CpGs, 986 (54%) are known quantitative trait loci (QTL) for CpG methylation (methQTL). When the mother's DNAm was assessed at early pregnancy, the number of CpGs showing intergenerational correlation was the smallest (384 CpGs, 0.11%). The second smallest number of such CpGs (559 CpGs, 0.17%) was found when investigating DNAm in offspring cord blood and father pairs. The low proportions of intergenerationally correlated CpGs suggest that epigenetic inheritance is limited.
Collapse
Affiliation(s)
- Yu Jiang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA.
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Su Chen
- Department of Mathematical Science, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susan Ewart
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA.
| |
Collapse
|