1
|
Li X, Luan T, Zhao C, Ling X. Effect of paternal body mass index on maternal and child-health outcomes of singletons after frozen-thawed embryo transfer cycles: a retrospective study. HUM FERTIL 2024; 27:2285343. [PMID: 38205607 DOI: 10.1080/14647273.2023.2285343] [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: 10/20/2022] [Accepted: 10/26/2023] [Indexed: 01/12/2024]
Abstract
The objective was to analyze the effect of paternal body mass index (BMI) on maternal and child-health outcomes of singletons after frozen-thawed embryo transfer (FET) cycles. A retrospective cohort study was conducted between January 2019 and December 2021. Pregnancy, perinatal complications and neonatal outcomes were compared among different paternal BMI. Multivariate logistic regression was performed to evaluate the relationship between different paternal BMI and pregnancy, obstetric and neonatal outcomes. The paternal normal group was more likely to suffer from gestational hypertension than the paternal obesity group (3.59% vs. 2.42%), and paternal underweight group was more likely to suffer from preeclampsia than the other three groups (11.63% vs. 4.43%, 7.57%, 4.03%). Birthweight among infants in the paternal overweight categories was significantly higher than infants in the paternal normal weight categories. The rate of foetal macrosomia was higher among infants in the paternal overweight (12.36%) category, while lower among infants in the paternal underweight categories (2.33%). The incidence of macrosomia in the paternal overweight categories (aOR 1.527, 95% CI 1.078-2.163) was significantly higher than those normal controls after adjustment for known confounding factors. The rates of LGA babies were higher in the paternal overweight category (aOR 1.260, 95% CI 1.001-1.587) compared with those in the paternal normal weight category, before and after adjustment. The results suggest that parental pre-pregnancy overweight or obesity has an adverse effect on the perinatal complications and neonatal outcomes.
Collapse
Affiliation(s)
- Xin Li
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Ting Luan
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Chun Zhao
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiufeng Ling
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| |
Collapse
|
2
|
Sivakumar S, Lama D, Rabhi N. Childhood obesity from the genes to the epigenome. Front Endocrinol (Lausanne) 2024; 15:1393250. [PMID: 39045266 PMCID: PMC11263020 DOI: 10.3389/fendo.2024.1393250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
The prevalence of obesity and its associated comorbidities has surged dramatically in recent decades. Especially concerning is the increased rate of childhood obesity, resulting in diseases traditionally associated only with adulthood. While obesity fundamentally arises from energy imbalance, emerging evidence over the past decade has revealed the involvement of additional factors. Epidemiological and murine studies have provided extensive evidence linking parental obesity to increased offspring weight and subsequent cardiometabolic complications in adulthood. Offspring exposed to an obese environment during conception, pregnancy, and/or lactation often exhibit increased body weight and long-term metabolic health issues, suggesting a transgenerational inheritance of disease susceptibility through epigenetic mechanisms rather than solely classic genetic mutations. In this review, we explore the current understanding of the mechanisms mediating transgenerational and intergenerational transmission of obesity. We delve into recent findings regarding both paternal and maternal obesity, shedding light on the underlying mechanisms and potential sex differences in offspring outcomes. A deeper understanding of the mechanisms behind obesity inheritance holds promise for enhancing clinical management strategies in offspring and breaking the cycle of increased metabolic risk across generations.
Collapse
Affiliation(s)
| | | | - Nabil Rabhi
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
3
|
Freire T, Pulpitel T, Clark X, Mackay F, Raubenheimer D, Simpson SJ, Solon-Biet SM, Crean AJ. The effects of paternal dietary fat versus sugar on offspring body composition and anxiety-related behavior. Physiol Behav 2024; 279:114533. [PMID: 38552707 DOI: 10.1016/j.physbeh.2024.114533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 02/26/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Increasing evidence suggests that the pre-conception parental environment has long-term consequences for offspring health and disease susceptibility. Though much of the work in this field concentrates on maternal influences, there is growing understanding that fathers also play a significant role in affecting offspring phenotypes. In this study, we investigate effects of altering the proportion of dietary fats and carbohydrates on paternal and offspring body composition and anxiety-related behavior in C57Bl/6-JArc mice. We show that in an isocaloric context, greater dietary fat increased body fat and reduced anxiety-like behavior of studs, whereas increased dietary sucrose had no significant effect. These dietary effects were not reflected in offspring traits, rather, we found sex-specific effects that differed between offspring body composition and behavioral traits. This finding is consistent with past paternal effect studies, where transgenerational effects have been shown to be more prominent in one sex over the other. Here, male offspring of fathers fed high-fat diets were heavier at 10 weeks of age due to increased lean body mass, whereas paternal diet had no significant effect on female offspring body fat or lean mass. In contrast, paternal dietary sugar appeared to have the strongest effects on male offspring behavior, with male offspring of high-sucrose fathers spending less time in the closed arms of the elevated plus maze. Both high-fat and high-sugar paternal diets were found to reduce anxiety-like behavior of female offspring, although this effect was only evident when offspring were fed a control diet. This study provides new understanding of the ways in which diet can shape the behavior of fathers and their offspring and contribute to the development of dietary guidelines to improve obesity and mental health conditions, such as anxiety.
Collapse
Affiliation(s)
- Therese Freire
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney NSW, Australia.
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Ximonie Clark
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - Flora Mackay
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Angela J Crean
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| |
Collapse
|
4
|
Pérez Lugo MI, Salas ML, Shrestha A, Ramalingam L. Fish Oil Improves Offspring Metabolic Health of Paternal Obese Mice by Targeting Adipose Tissue. Biomolecules 2024; 14:418. [PMID: 38672435 PMCID: PMC11048145 DOI: 10.3390/biom14040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity is a fast-growing epidemic affecting more than 40% of the US population and leads to co-morbidities such as type 2 diabetes and cancer. More importantly, there is a rapid increase in childhood obesity associated with obesity in parents. Further, offspring are encoded with approximately half of their genetic information from the paternal side. Obesity in fathers at the preconceptional period likely influences the intergenerational development of obesity. This study focuses on the role of fish oil supplementation as a non-pharmacological intervention in fathers and its impact on childhood obesity using animal models. Male mice were fed a low-fat diet or high-fat diet with or without fish oil for 10 weeks and mated with female mice on a chow diet. Offspring were then continued on a chow diet until 8 or 16 weeks. In vivo insulin tolerance was tested to assess the metabolic health of offspring. Further, adipose tissue was harvested upon sacrifice, and genetic markers of inflammation and lipid metabolism in the tissue were analyzed. Offspring of males supplemented with fish oil showed lower body weight, improved insulin tolerance, and altered inflammatory markers. Markers of fatty acid oxidation were higher, while markers of fatty acid synthesis were lower in offspring of fathers fed fish oil. This supports fish oil as an accessible intervention to improve offspring metabolic health.
Collapse
Affiliation(s)
| | | | | | - Latha Ramalingam
- Department of Nutrition and Food Studies, Syracuse University, Syracuse, NY 13244, USA (M.L.S.); (A.S.)
| |
Collapse
|
5
|
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
|
6
|
Mezo-González CE, García-Santillán JA, Kaeffer B, Gourdel M, Croyal M, Bolaños-Jiménez F. Adult rats sired by obese fathers present learning deficits associated with epigenetic and neurochemical alterations linked to impaired brain glutamatergic signaling. Acta Physiol (Oxf) 2024; 240:e14090. [PMID: 38230587 DOI: 10.1111/apha.14090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/10/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
AIM Offspring of obese mothers are at high risk of developing metabolic syndrome and cognitive disabilities. Impaired metabolism has also been reported in the offspring of obese fathers. However, whether brain function can also be affected by paternal obesity has barely been examined. This study aimed to characterize the learning deficits resulting from paternal obesity versus those induced by maternal obesity and to identify the underlying mechanisms. METHODS Founder control and obese female and male Wistar rats were mated to constitute three first-generation (F1) experimental groups: control mother/control father, obese mother/control father, and obese father/control mother. All F1 animals were weaned onto standard chow and underwent a learning test at 4 months of age, after which several markers of glutamate-mediated synaptic plasticity together with the expression of miRNAs targeting glutamate receptors and the concentration of kynurenic and quinolinic acids were quantified in the hippocampus and frontal cortex. RESULTS Maternal obesity induced a severe learning deficit by impairing memory encoding and memory consolidation. The offspring of obese fathers also showed reduced memory encoding but not impaired long-term memory formation. Memory deficits in offspring of obese fathers and obese mothers were associated with a down-regulation of genes encoding NMDA glutamate receptors subunits and several learning-related genes along with impaired expression of miR-296 and miR-146b and increased concentration of kynurenic acid. CONCLUSION Paternal and maternal obesity impair offspring's learning abilities by affecting different processes of memory formation. These cognitive deficits are associated with epigenetic and neurochemical alterations leading to impaired glutamate-mediated synaptic plasticity.
Collapse
Affiliation(s)
| | | | - Bertrad Kaeffer
- UMR Physiologie des Adaptations Nutritionnelles, INRAE - Nantes Université, Nantes, France
| | - Mathilde Gourdel
- CRNH-O Mass Spectrometry Core Facility, Nantes, France
- CNRS, INSERM, L'institut du Thorax, Université de Nantes, Nantes, France
- CHU Nantes, INSERM, CNRS, SFR Santé, INSERM UMS 016, CNRS UMS 3556, Université de Nantes, Nantes, France
| | - Mikaël Croyal
- CRNH-O Mass Spectrometry Core Facility, Nantes, France
- CNRS, INSERM, L'institut du Thorax, Université de Nantes, Nantes, France
- CHU Nantes, INSERM, CNRS, SFR Santé, INSERM UMS 016, CNRS UMS 3556, Université de Nantes, Nantes, France
| | | |
Collapse
|
7
|
Sharma S, Bhonde R. Dilemma of Epigenetic Changes Causing or Reducing Metabolic Disorders in Offsprings of Obese Mothers. Horm Metab Res 2023; 55:665-676. [PMID: 37813098 DOI: 10.1055/a-2159-9128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Maternal obesity is associated with fetal complications predisposing later to the development of metabolic syndrome during childhood and adult stages. High-fat diet seems to influence individuals and their subsequent generations in mediating weight gain, insulin resistance, obesity, high cholesterol, diabetes, and cardiovascular disorder. Research evidence strongly suggests that epigenetic alteration is the major contributor to the development of metabolic syndrome through DNA methylation, histone modifications, and microRNA expression. In this review, we have discussed the outcome of recent studies on the adverse and beneficial effects of nutrients and vitamins through epigenetics during pregnancy. We have further discussed about the miRNAs altered during maternal obesity. Identification of new epigenetic modifiers such as mesenchymal stem cells condition media (MSCs-CM)/exosomes for accelerating the reversal of epigenetic abnormalities for the development of new treatments is yet another aspect of the present review.
Collapse
Affiliation(s)
- Shikha Sharma
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Ramesh Bhonde
- Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Vidyapeeth Pune (Deemed University), Pune, India
| |
Collapse
|
8
|
Mehri K, Hamidian G, Zavvari Oskuye Z, Nayebirad S, Farajdokht F. The role of apelinergic system in metabolism and reproductive system in normal and pathological conditions: an overview. Front Endocrinol (Lausanne) 2023; 14:1193150. [PMID: 37424869 PMCID: PMC10324965 DOI: 10.3389/fendo.2023.1193150] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Lifestyle changes have made metabolic disorders as one of the major threats to life. Growing evidence demonstrates that obesity and diabetes disrupt the reproductive system by affecting the gonads and the hypothalamus-pituitary-gonadal (HPG) axis. Apelin, an adipocytokine, and its receptor (APJ) are broadly expressed in the hypothalamus nuclei, such as paraventricular and supraoptic, where gonadotropin-releasing hormone (GnRH) is released, and all three lobes of the pituitary, indicating that apelin is involved in the control of reproductive function. Moreover, apelin affects food intake, insulin sensitivity, fluid homeostasis, and glucose and lipid metabolisms. This review outlined the physiological effects of the apelinergic system, the relationship between apelin and metabolic disorders such as diabetes and obesity, as well as the effect of apelin on the reproductive system in both gender. The apelin-APJ system can be considered a potential therapeutic target in the management of obesity-associated metabolic dysfunction and reproductive disorders.
Collapse
Affiliation(s)
- Keyvan Mehri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Hamidian
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | | | - Sepehr Nayebirad
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Shi Q, Qi K. Developmental origins of health and disease: Impact of paternal nutrition and lifestyle. Pediatr Investig 2023; 7:111-131. [PMID: 37324600 PMCID: PMC10262906 DOI: 10.1002/ped4.12367] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/29/2023] [Indexed: 09/20/2023] Open
Abstract
Most epidemiological and experimental studies have focused on maternal influences on offspring's health. The impact of maternal undernutrition, overnutrition, hypoxia, and stress is linked to adverse offspring outcomes across a range of systems including cardiometabolic, respiratory, endocrine, and reproduction among others. During the past decade, it has become evident that paternal environmental factors are also linked to the development of diseases in offspring. In this article, we aim to outline the current understanding of the impact of male health and environmental exposure on offspring development, health, and disease and explore the mechanisms underlying the paternal programming of offspring health. The available evidence suggests that poor paternal pre-conceptional nutrition and lifestyle, and advanced age can increase the risk of negative outcomes in offspring, via both direct (genetic/epigenetic) and indirect (maternal uterine environment) effects. Beginning at preconception, and during utero and the early life after birth, cells acquire an epigenetic memory of the early exposure which can be influential across the entire lifespan and program a child's health. Potentially not only mothers but also fathers should be advised that maintaining a healthy diet and lifestyle is important to improve offspring health as well as the parental health status. However, the evidence is mostly based on animal studies, and well-designed human studies are urgently needed to verify findings from animal data.
Collapse
Affiliation(s)
- Qiaoyu Shi
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| | - Kemin Qi
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's HospitalCapital Medical University, National Center for Children's HealthBeijingChina
| |
Collapse
|
10
|
Donato J. Programming of metabolism by adipokines during development. Nat Rev Endocrinol 2023:10.1038/s41574-023-00828-1. [PMID: 37055548 DOI: 10.1038/s41574-023-00828-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 04/15/2023]
Abstract
The intrauterine and early postnatal periods represent key developmental stages in which an organism is highly susceptible to being permanently influenced by maternal factors and nutritional status. Strong evidence indicates that either undernutrition or overnutrition during development can predispose individuals to disease later in life, especially type 2 diabetes mellitus and obesity, a concept known as metabolic programming. Adipose tissue produces important signalling molecules that control energy and glucose homeostasis, including leptin and adiponectin. In addition to their well-characterized metabolic effects in adults, adipokines have been associated with metabolic programming by affecting different aspects of development. Therefore, alterations in the secretion or signalling of adipokines, caused by nutritional insults in early life, might lead to metabolic diseases in adulthood. This Review summarizes and discusses the potential role of several adipokines in inducing metabolic programming through their effects during development. The identification of the endocrine factors that act in early life to permanently influence metabolism represents a key step in understanding the mechanisms behind metabolic programming. Thus, future strategies aiming to prevent and treat these metabolic diseases can be designed, taking into consideration the relationship between adipokines and the developmental origins of health and disease.
Collapse
Affiliation(s)
- Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
11
|
Whittaker J. Dietary trends and the decline in male reproductive health. Hormones (Athens) 2023; 22:165-197. [PMID: 36725796 DOI: 10.1007/s42000-023-00431-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/16/2023] [Indexed: 02/03/2023]
Abstract
Over the twentieth century, male reproductive health has suffered a substantial decline, as evidenced by decreases in sperm counts and testosterone levels and increases in reproductive pathologies. At the same time, the prevalence of chronic diseases such as obesity, diabetes, and metabolic syndrome has risen dramatically. Metabolic and reproductive health are highly interconnected, suggesting that their respective trends are intertwined and, given the timeframe of such trends, environmental and not genetic factors are most likely to be the primary causes. Industrialization, which began in Europe in the mid-eighteenth century, has resulted in profound changes to our diet, lifestyle, and environment, many of which are causal factors in the rise in chronic diseases. Industrialization results in a nutrition transition from an agricultural unprocessed to a modern processed diet, incorporating increases in sugar, vegetable oils, ultra-processed foods, linoleic acid, trans-fats, and total energy. This dietary shift has incurred numerous adverse effects on metabolic and reproductive health, characterized by chronic inflammation, oxidative stress, and insulin resistance. Moreover, these effects appear to multiply across subsequent generations via epigenetic inheritance. Men's fertility is markedly affected by obesity and diabetes, with an increase in total energy via processed food intake arguably being the key factor driving the diabesity pandemic. In contrast, wholefoods rich in micronutrients and phytonutrients support male fertility and a healthy body weight. Therefore, men wanting to maximize their fertility should consider making positive dietary changes, such as replacing processed foods with unprocessed foods that support metabolic and reproductive health.
Collapse
Affiliation(s)
- Joseph Whittaker
- The School of Allied Health and Community, University of Worcester, Henwick Grove, Worcester, WR2 6AJ, UK.
| |
Collapse
|
12
|
Shi Q, Liu X, Fan X, Wang R, Qi K. Paternal dietary ratio of n-6: n-3 polyunsaturated fatty acids programs offspring leptin expression and gene imprinting in mice. Front Nutr 2022; 9:1043876. [PMID: 36618698 PMCID: PMC9816484 DOI: 10.3389/fnut.2022.1043876] [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: 09/15/2022] [Accepted: 12/08/2022] [Indexed: 12/25/2022] Open
Abstract
Background This study determined the effects of the paternal dietary ratio of n-6: n-3 polyunsaturated fatty acids (PUFAs) on leptin expression in the offspring and associated gene imprinting in a mouse model. Methods Three- to four-week-old male C57BL/6J mice (F0) were fed an n-3 PUFA-deficient (n-3 D) diet, a diet with normal n-3 PUFA content (n-3 N; n-6: n-3 = 4.3:1), or a diet with a high n-3 PUFA content (n-3 H; n-6: n-3 = 1.5:1) for 8 weeks. Two subsequent generations were generated by mating F0 and F1 male mice with 10-week-old virgin female C57 BL/6J mice, to produce F1 and F2 offspring. Results Compared to the paternal n-3 D diet, paternal n-3 N and n-3 H diets reduced adipose mRNA expression of leptin (Lep) and its plasma concentrations in juvenile F1 male and female offspring, and adult F1 male and F2 female offspring, with upregulated Lep receptor mRNA expression in the hypothalamus. Meanwhile, paternal n-3 N and n-3 H diets altered the expression of the imprinted genes H19, Igf2, Igf2r, Plagl1, Cdkn1c, Kcnq1ot1, Peg3, and Grb10 in the adipose tissue of juvenile and adult F1 males, with almost no effects on F1 females, while more effects were observed in the adult F2 females than F2 males. Principal component analysis verified that Plagl1, Cdkn1c, and Kcnq1ot1 contributed the most to variation in adipose tissue expression in all offspring. Some of these genes (Plagl1, Cdkn1c, Kcnq1ot1, Peg3, and Grb10) were altered by the paternal n-3 N and n-3 H diets in the F1 and F2 generation testes as well. Furthermore, adipose Lep expression was positively correlated with expressions of H19, Igf2r, Plagl1, and Kcnq1ot1 in juvenile F1 males and females, negatively correlated with the Kcnq1ot1 expression in adult F1 males, and positively correlated with the Plagl1 expression in adult F2 females. Conclusion These data imply that paternal Plagl1, Cdkn1c, and Kcnq1ot1 might be part of the pathways involved in offspring leptin programming. Therefore, a lower ratio of n-6: n-3 PUFAs, with higher intake of n-3 PUFAs in paternal pre-conception, may help maintain the offspring's optimal leptin pattern in a sex-specific manner through multiple generations, and thereby, be beneficial for the offspring's long-term health.
Collapse
|
13
|
Role of Adipose Tissue microRNAs in the Onset of Metabolic Diseases and Implications in the Context of the DOHaD. Cells 2022; 11:cells11233711. [PMID: 36496971 PMCID: PMC9739499 DOI: 10.3390/cells11233711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The worldwide epidemic of obesity is associated with numerous comorbid conditions, including metabolic diseases such as insulin resistance and diabetes, in particular. The situation is likely to worsen, as the increase in obesity rates among children will probably lead to an earlier onset and more severe course for metabolic diseases. The origin of this earlier development of obesity may lie in both behavior (changes in nutrition, physical activity, etc.) and in children's history, as it appears to be at least partly programmed by the fetal/neonatal environment. The concept of the developmental origin of health and diseases (DOHaD), involving both organogenesis and epigenetic mechanisms, encompasses such programming. Epigenetic mechanisms include the action of microRNAs, which seem to play an important role in adipocyte functions. Interestingly, microRNAs seem to play a particular role in propagating local insulin resistance to other key organs, thereby inducing global insulin resistance and type 2 diabetes. This propagation involves the active secretion of exosomes containing microRNAs by adipocytes and adipose tissue-resident macrophages, as well as long-distance communication targeting the muscles and liver, for example. Circulating microRNAs may also be useful as biomarkers for the identification of populations at risk of subsequently developing obesity and metabolic diseases.
Collapse
|
14
|
Sheth VG, Sharma N, Kabeer SW, Tikoo K. Lactobacillus rhamnosus supplementation ameliorates high fat diet-induced epigenetic alterations and prevents its intergenerational inheritance. Life Sci 2022; 311:121151. [DOI: 10.1016/j.lfs.2022.121151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
15
|
Aizawa S, Tochihara A, Yamamuro Y. Paternal high-fat diet alters triglyceride metabolism-related gene expression in liver and white adipose tissue of male mouse offspring. Biochem Biophys Rep 2022; 31:101330. [PMID: 35990577 PMCID: PMC9388883 DOI: 10.1016/j.bbrep.2022.101330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
Obesity is a major public health problem, and its prevalence is progressively increasing worldwide. In addition, accumulating evidence suggests that diverse nutritional and metabolic disturbances including obesity can be transmitted from parents to offspring via transgenerational epigenetic inheritance. The previous reports have shown that paternal obesity has profound impacts on the development and metabolic health of their progeny. However, little information is available concerning the effects of paternal high-fat diet (HFD) exposure on triglyceride metabolism in the offspring. Therefore, we investigated the effects of paternal HFD on triglyceride metabolism and related gene expression in male mouse offspring. We found that paternal HFD exposure significantly increased the body weight, liver and epididymal white adipose tissue (eWAT) weights, and liver triglyceride content in male offspring, despite consuming control diet. In addition, paternal HFD exposure had induced changes in the mRNA expression of genes involved in lipid and triglyceride metabolism in the liver and eWAT. These findings indicate transgenerational inheritance from the paternal metabolic disturbance of triglyceride and support the effects of paternal lifestyle choices on offspring development and health later in life. Paternal HFD exposure increased body weight, liver, and eWAT weights in male offspring. Paternal HFD exposure induced triglyceride metabolism disturbance in male offspring. Triglyceride metabolism-related gene expression in the offspring liver and eWAT was altered by paternal HFD exposure.
Collapse
|
16
|
Prasad M, Rajagopal P, Devarajan N, Veeraraghavan VP, Palanisamy CP, Cui B, Patil S, Jayaraman S. A comprehensive review on high fat diet-induced diabetes mellitus: An epigenetic view. J Nutr Biochem 2022; 107:109037. [PMID: 35533900 DOI: 10.1016/j.jnutbio.2022.109037] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 01/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
Modern lifestyle, genetics, nutritional overload through high-fat diet attributed prevalence and diabetes outcomes with various complications primarily due to obesity in which energy-dense diets frequently affect metabolic health. One possible issue usually associated with elevated chronic fat intake is insulin resistance, and hyperglycaemia constitutes an important function in altering the carbohydrates and lipids metabolism. Similarly, in assessing human susceptibility to weight gain and obesity, genetic variations play a central role, contributing to keen interest in identifying the possible role of epigenetics as a mediator of gene-environmental interactions influencing the production of type 2 diabetes mellitus and its related concerns. Epigenetic modifications associated with the acceptance of a sedentary lifestyle and environmental stress factors in response to energy intake and expenditure imbalances complement genetic alterations and lead to the production and advancement of metabolic disorders such as diabetes and obesity. Methylation of DNA, histone modifications and increases in the expression of non-coding RNAs can result in reduced transcriptional activity of key β-cell genes thus creating insulin resistance. Epigenetics contribute to changes in the expression of the underlying insulin resistance and insufficiency gene networks, along with low-grade obesity-related inflammation, increased ROS generation and DNA damage in multi organs. This review focused on epigenetic mechanisms and metabolic regulations associated with high fat diet (HFD)-induced diabetes mellitus.
Collapse
Affiliation(s)
- Monisha Prasad
- Centre for Molecular Medicine and diagnostic (CoMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
| | - Ponnulakshmi Rajagopal
- Central Research Laboratory, Meenakhsi Ammal Dental College and Hospitals, Academy of Higher Education and Research, Chennai, 600 095, India
| | - Nalini Devarajan
- Central Research Laboratory, Meenakhsi Academy of Higher Education and Research, West K.K. Nagar, Chennai, 600 078, India
| | - Vishnu Priya Veeraraghavan
- State Key Laboratory of Biobased Materials and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Jinan, 250353, China
| | - Chella Perumal Palanisamy
- State Key Laboratory of Biobased Materials and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Jinan, 250353, China
| | - Bo Cui
- State Key Laboratory of Biobased Materials and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Jinan, 250353, China
| | - Shankargouda Patil
- Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Saudi Arabia
| | - Selvaraj Jayaraman
- Centre for Molecular Medicine and diagnostic (CoMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India.
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Zhang X, Hasan AA, Wu H, Gaballa MMS, Zeng S, Liu L, Xie L, Jung T, Grune T, Krämer BK, Kleuser B, Li J, Hocher B. High-fat, sucrose and salt-rich diet during rat spermatogenesis lead to the development of chronic kidney disease in the female offspring of the F2 generation. FASEB J 2022; 36:e22259. [PMID: 35294083 DOI: 10.1096/fj.202101789rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 11/11/2022]
Abstract
Effects of feeding male rats during spermatogenesis a high-fat, high-sucrose and high-salt diet (HFSSD) over two generations (F0 and F1) on renal outcomes are unknown. Male F0 and F1 rats were fed either control diet (F0CD+F1CD) or HFSSD (F0HD+F1HD). The outcomes were glomerular filtration rate and urinary albumin excretion in F1 and F2 offspring. If both outcomes were altered a morphological and molecular assessment was done. F2 offspring of both sexes had a decreased GFR. However, increased urinary albumin excretion was only observed in female F2 F0HD+F1HD offspring compared with controls. F0HD+F1HD female F2 offspring developed glomerulosclerosis (+31%; p < .01) and increased renal interstitial fibrosis (+52%; p < .05). RNA sequencing followed by qRT-PCR validation showed that four genes (Enpp6, Tmem144, Cd300lf, and Actr3b) were differentially regulated in the kidneys of female F2 offspring. lncRNA XR-146683.1 expression decreased in female F0HD+F1HD F2 offspring and its expression was (r = 0.44, p = .027) correlated with the expression of Tmem144. Methylation of CpG islands in the promoter region of the Cd300lf gene was increased (p = .001) in female F2 F0HD+F1HD offspring compared to controls. Promoter CpG island methylation rate of Cd300lf was inversely correlated with Cd300lf mRNA expression in F2 female offspring (r = -0.483, p = .012). Cd300lf mRNA expression was inversely correlated with the urinary albumin-to-creatinine ratio in female F2 offspring (r = -0.588, p = .005). Paternal pre-conceptional unhealthy diet given for two generations predispose female F2 offspring to chronic kidney disease due to epigenetic alterations of renal gene expression. Particularly, Cd300lf gene promotor methylation was inversely associated with Cd300lf mRNA expression and Cd300lf mRNA expression itself was inversely associated with urinary albumin excretion in F2 female offspring whose fathers and grandfathers got a pre-conceptional unhealthy diet.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.,Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany.,Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ahmed A Hasan
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany.,Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Hongwei Wu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Department of Nephrology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Mohamed M S Gaballa
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Suimin Zeng
- The First Hospital of Traditional Chinese Medicine, Yiyang, China
| | - Liping Liu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Li Xie
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany.,Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Jian Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Berthold Hocher
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.,Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China.,Institute of Medical Diagnostics, IMD Berlin, Berlin, Germany
| |
Collapse
|
20
|
Mao Y, Yiran Z, Sisi L, Huixi C, Xia L, Ting W, Guolian D, Xinmei L, Sheng J, Meng Y, Huang H. Advanced paternal age increased metabolic risks in mice offspring. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166355. [DOI: 10.1016/j.bbadis.2022.166355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/07/2022] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
|
21
|
Anwer H, Morris MJ, Noble DWA, Nakagawa S, Lagisz M. Transgenerational effects of obesogenic diets in rodents: A meta-analysis. Obes Rev 2022; 23:e13342. [PMID: 34595817 DOI: 10.1111/obr.13342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
Obesity is a major health condition that affects millions worldwide. There is an increased interest in understanding the adverse outcomes associated with obesogenic diets. A multitude of studies have investigated the transgenerational impacts of maternal and parental obesogenic diets on subsequent generations of offspring, but results have largely been mixed. We conducted a systematic review and meta-analysis on rodent studies to elucidate how obesogenic diets impact the mean and variance of grand-offspring traits. Our study focused on transgenerational effects (i.e., F2 and F3 generations) in one-off and multigenerational exposure studies. From 33 included articles, we obtained 407 effect sizes representing pairwise comparisons of control and treatment grand-offspring groups pertaining to measures of body weight, adiposity, glucose, insulin, leptin, and triglycerides. We found evidence that male and female grand-offspring descended from grandparents exposed to an obesogenic diet displayed phenotypes consistent with metabolic syndrome, especially in cases where the obesogenic diet was continued across generations. Further, we found stronger evidence for the effects of grand-maternal than grand-paternal exposure on grand-offspring traits. A high-fat diet in one-off exposure studies did not seem to impact phenotypic variation, whereas in multigenerational exposure studies it reduced variation in several traits.
Collapse
Affiliation(s)
- Hamza Anwer
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Margaret J Morris
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Daniel W A Noble
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
- Division of Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Shinichi Nakagawa
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Malgorzata Lagisz
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Crisóstomo L, Videira RA, Jarak I, Starčević K, Mašek T, Rato L, Raposo JF, Batterham RL, Oliveira PF, Alves MG. Inherited Metabolic Memory of High-fat Diet Impairs Testicular Fatty Acid Content and Sperm Parameters. Mol Nutr Food Res 2021; 66:e2100680. [PMID: 34939729 DOI: 10.1002/mnfr.202100680] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/29/2021] [Indexed: 11/08/2022]
Abstract
Exposure to a high-fat diet (HFD) from early-life is associated with a testicular metabolic signature linked to abnormal sperm parameters up to two generations after exposure in mice. Hereby, we describe a testicular lipid signature associated with "inherited metabolic memory" of exposure to HFD, persisting up to two generations in mice. Diet-challenged mice (n = 36) were randomly fed after weaning with standard chow (CTRL); HFD for 200 days or transient HFD (HFDt ) (60 days of HFD+140 days of standard chow). Subsequent generations (36 mice per generation) were fed with chow diet. Mice were euthanized 200 days post-weaning. Glucose homeostasis, serum hormones, testicular bioenergetics and antioxidant enzyme activity were evaluated. Testicular lipid-related metabolites and fatty acids were characterized by 1 H-NMR and GC-MS. Sons of HFD display impaired choline metabolism, mitochondrial activity and antioxidant defenses, while grandsons show a shift in testicular ω3/ω6 ratio towards a pro-inflammatory environment. Grandsons of HFDt raise 3-hydroxybutyrate levels with possible implications to testicular insulin resistance. Sperm counts decrease in grandsons of HFD-exposed mice, regardless of the duration of exposure. HFD-induced "inherited metabolic memory" alters testicular fatty acid metabolism with consequences to sperm parameters up to two generations. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Luís Crisóstomo
- Department of Anatomy, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal.,Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Portugal
| | - Romeu A Videira
- REQUINTE/LAQV, Laboratory of Pharmacognosy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Ivana Jarak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Portugal
| | - Kristina Starčević
- Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Tomislav Mašek
- Department of Animal Nutrition and Dietetics, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Luís Rato
- Health School of the Polytechnic Institute of Guarda, Guarda, Portugal
| | - João F Raposo
- NOVA Medical School - New University Lisbon, Lisbon, Portugal.,APDP - Diabetes Portugal, Lisbon, Portugal
| | - Rachel L Batterham
- UCL Centre for Obesity Research, Division of Medicine, University College London, London, UK.,National Institute of Health Research, UCLH Biomedical Research Centre, London, UK
| | - Pedro F Oliveira
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Marco G Alves
- Department of Anatomy, and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal.,Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Portugal
| |
Collapse
|
23
|
Ghai M, Kader F. A Review on Epigenetic Inheritance of Experiences in Humans. Biochem Genet 2021; 60:1107-1140. [PMID: 34792705 DOI: 10.1007/s10528-021-10155-7] [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: 05/04/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022]
Abstract
If genetics defines the inheritance of DNA, epigenetics aims to regulate and make it adaptable. Epigenetic alterations include DNA methylation, chromatin remodelling, post-translational modifications of histone proteins and activity of non-coding RNAs. Several studies, especially in animal models, have reported transgenerational inheritance of epigenetic marks. However, evidence of transgenerational inheritance in humans via germline in the absence of any direct exposure to the driving external stimulus remains controversial. Most of the epimutations exist in relation with genetic variants. The present review looks at intergenerational and transgenerational inheritance in humans, (both father and mother) in response to diet, exposure to chemicals, stress, exercise, and disease status. If not transgenerational, at least intergenerational human studies could help to understand early processes of inheritance. In humans, female and male germline development follow separate paths of epigenetic events and both oocyte and sperm possess their own unique epigenomes. While DNA methylation alterations are reset during epigenetic reprogramming, non-coding RNAs via human sperm provide evidence of being reliable carriers for transgenerational inheritance. Human studies reveal that one mechanism of epigenetic inheritance cannot be applied to the complete human genome. Multiple factors including time, type, and tissue of exposure determine if the modified epigenetic mark could be transmissible and till which generation. Population-specific differences should also be taken into consideration while associating inheritance to an environmental exposure. A longitudinal study targeting one environmental factor, but different population groups should be conducted at a specific geographical location to pinpoint heritable epigenetic changes.
Collapse
Affiliation(s)
- Meenu Ghai
- Discipline of Genetics, School of Life Sciences, Westville Campus, University of KwaZulu-Natal, Private Bag X54001, Durban, KwaZulu Natal, South Africa.
| | - Farzeen Kader
- Discipline of Genetics, School of Life Sciences, Westville Campus, University of KwaZulu-Natal, Private Bag X54001, Durban, KwaZulu Natal, South Africa
| |
Collapse
|
24
|
Hsu CN, Hou CY, Hsu WH, Tain YL. Early-Life Origins of Metabolic Syndrome: Mechanisms and Preventive Aspects. Int J Mol Sci 2021; 22:11872. [PMID: 34769303 PMCID: PMC8584419 DOI: 10.3390/ijms222111872] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
One of the leading global public-health burdens is metabolic syndrome (MetS), despite the many advances in pharmacotherapies. MetS, now known as "developmental origins of health and disease" (DOHaD), can have its origins in early life. Offspring MetS can be programmed by various adverse early-life conditions, such as nutrition imbalance, maternal conditions or diseases, maternal chemical exposure, and medication use. Conversely, early interventions have shown potential to revoke programming processes to prevent MetS of developmental origins, namely reprogramming. In this review, we summarize what is currently known about adverse environmental insults implicated in MetS of developmental origins, including the fundamental underlying mechanisms. We also describe animal models that have been developed to study the developmental programming of MetS. This review extends previous research reviews by addressing implementation of reprogramming strategies to prevent the programming of MetS. These mechanism-targeted strategies include antioxidants, melatonin, resveratrol, probiotics/prebiotics, and amino acids. Much work remains to be accomplished to determine the insults that could induce MetS, to identify the mechanisms behind MetS programming, and to develop potential reprogramming strategies for clinical translation.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan;
| | - Wei-Hsuan Hsu
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Chen Kung University, Tainan 701, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
25
|
César H, Sertorio MN, de Souza EA, Jamar G, Santamarina A, Jucá A, Casagrande BP, Pisani LP. Parental high-fat high-sugar diet programming and hypothalamus adipose tissue axis in male Wistar rats. Eur J Nutr 2021; 61:523-537. [PMID: 34657184 DOI: 10.1007/s00394-021-02690-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 09/28/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE Maternal nutrition during early development and paternal nutrition pre-conception can programme offspring health status. Hypothalamus adipose axis is a target of developmental programming, and paternal and maternal high-fat, high-sugar diet (HFS) may be an important factor that predisposes offspring to develop obesity later in life. This study aims to investigate Wistar rats' maternal and paternal HFS differential contribution on the development, adiposity, and hypothalamic inflammation in male offspring from weaning until adulthood. METHODS Male progenitors were fed a control diet (CD) or HFS for 10 weeks before mating. After mating, dams were fed CD or HFS only during pregnancy and lactation. Forming the following male offspring groups: CD-maternal and paternal CD; MH-maternal HFS and paternal CD; PH-maternal CD and paternal HFS; PMH-maternal and paternal HFS. After weaning, male offspring were fed CD until adulthood. RESULTS Maternal HFS diet increased weight, visceral adiposity, and serum total cholesterol levels, and decreased hypothalamic weight in weanling male rats. In adult male offspring, maternal HFS increased weight, glucose levels, and hypothalamic NFκBp65. Paternal HFS diet lowered hypothalamic insulin receptor levels in weanling offspring and glucose and insulin levels in adult offspring. The combined effects of maternal and paternal HFS diets increased triacylglycerol, leptin levels, and hypothalamic inflammation in weanling rats, and increased visceral adiposity in adulthood. CONCLUSION Male offspring intake of CD diet after weaning reversed part of the effects of parental HFS diet during the perinatal period. However, maternal and paternal HFS diet affected adiposity and hypothalamic inflammation, which remained until adulthood.
Collapse
Affiliation(s)
- Helena César
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo-UNIFESP, Santos, SP, Brazil
| | | | - Esther Alves de Souza
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Giovana Jamar
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Aline Santamarina
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Andrea Jucá
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Breno Picin Casagrande
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Luciana Pellegrini Pisani
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil.
| |
Collapse
|
26
|
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
|
27
|
Crisóstomo L, Jarak I, Rato LP, Raposo JF, Batterham RL, Oliveira PF, Alves MG. Inheritable testicular metabolic memory of high-fat diet causes transgenerational sperm defects in mice. Sci Rep 2021; 11:9444. [PMID: 33941835 PMCID: PMC8093209 DOI: 10.1038/s41598-021-88981-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022] Open
Abstract
The consumption of energy-dense diets has contributed to an increase in the prevalence of obesity and its comorbidities worldwide. The adoption of unhealthy feeding habits often occurs at early age, prompting the early onset of metabolic disease with unknown consequences for reproductive function later in life. Recently, evidence has emerged regarding the intergenerational and transgenerational effects of high-fat diets (HFD) on sperm parameters and testicular metabolism. Hereby, we study the impact of high-fat feeding male mice (F0) on the testicular metabolome and function of their sons (F1) and grandsons (F2). Testicular content of metabolites related to insulin resistance, cell membrane remodeling, nutritional support and antioxidative stress (leucine, acetate, glycine, glutamine, inosine) were altered in sons and grandsons of mice fed with HFD, comparing to descendants of chow-fed mice. Sperm counts were lower in the grandsons of mice fed with HFD, even if transient. Sperm quality was correlated to testicular metabolite content in all generations. Principal Component Analysis of sperm parameters and testicular metabolites revealed an HFD-related phenotype, especially in the diet-challenged generation and their grandsons. Ancestral HFD, even if transient, causes transgenerational "inherited metabolic memory" in the testicular tissue, characterized by changes in testicular metabolome and function.
Collapse
Affiliation(s)
- Luís Crisóstomo
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Ivana Jarak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Luís P Rato
- Health School of the Polytechnic Institute of Guarda, Guarda, Portugal
| | - João F Raposo
- NOVA Medical School - New University Lisbon, Lisbon, Portugal.,APDP - Diabetes Portugal, Lisbon, Portugal
| | - Rachel L Batterham
- UCL Centre for Obesity Research, Division of Medicine, University College London, London, UK.,National Institute of Health Research, UCLH Biomedical Research Centre, London, UK
| | - Pedro F Oliveira
- Department of Chemistry, QOPNA & LAQV, University of Aveiro, Aveiro, Portugal
| | - Marco G Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
28
|
Sun AJ, Li S, Zhang CA, Jensen TK, Lindahl-Jacobsen R, Eisenberg ML. Parental comorbidity and medication use in the USA: a panel study of 785 000 live births. Hum Reprod 2021; 35:669-675. [PMID: 32187368 DOI: 10.1093/humrep/deaa022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/01/2020] [Indexed: 01/24/2023] Open
Abstract
STUDY QUESTION How prevalent is paternal medication use and comorbidity, and are rates of these rising? SUMMARY ANSWER Paternal medication use and comorbidity is common and rising, similar to trends previously described in mothers. WHAT IS KNOWN ALREADY Maternal medication use and comorbidity has been rising for the past few decades. These trends have been linked to potential teratogenicity, maternal morbidity and mortality and poorer fetal outcomes. STUDY DESIGN, SIZE, DURATION This is a Panel (trend) study of 785 809 live births from 2008 to 2016. PARTICIPANTS/MATERIALS, SETTING, METHODS We used the IBM© Marketscan®™ database to gather data on demographic information and International Classification of Diseases codes and Charlson comorbidity index (CCI) during the 12 months prior to the estimated date of conception for mothers and fathers. We similarly examined claims of prescriptions in the 3 months prior to conception. We performed companion analyses of medications used for >90 days in the 12 months prior to conception and of any medication use in the 12 months prior to conception. MAIN RESULTS AND THE ROLE OF CHANCE We confirmed that both maternal medication use and comorbidity (e.g. hypertension, diabetes, hyperlipidemia) rose over the study period, consistent with prior studies. We found a concurrent rise in both paternal medication use 3 months prior to conception (overall use, 31.5-34.9% during the study period; P < 0.0001) and comorbidity (CCI of ≥1 and 10.6-18.0% over study period; P < 0.0001). The most common conditions seen in the CCI were chronic obstructive pulmonary disease for mothers (6.6-11.6%) and hyperlipidemia for fathers (8.6-13.7%). Similar trends for individual medication classes and specific comorbidities such as hypertension, diabetes and hyperlipidemia were also seen. All primary result trends were statistically significant, making the role of chance minimal. LIMITATIONS, REASONS FOR CAUTION As this is a descriptive study, the clinical impact is uncertain and no causal associations may be made. Though the study uses a large and curated database that includes patients from across the USA, our study population is an insured population and our findings may not be generalizable. Mean parental age was seen to slightly increase over the course of the study (<1 year) and may be associated with increased comorbidity and medication use. WIDER IMPLICATIONS OF THE FINDINGS As parental comorbidity and certain medication use may impact fecundability, temporal declines in parental health may impact conception, pregnancy and fetal outcomes. STUDY FUNDING/COMPETING INTEREST(S) None. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Andrew J Sun
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shufeng Li
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chiyuan A Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tina K Jensen
- Department of Environmental Health, University of Southern Denmark, Odense, Denmark
| | - Rune Lindahl-Jacobsen
- Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense, Denmark.,Interdisciplinary Centre on Population Dynamics (CPop), University of Southern Denmark, Odense, Denmark
| | - Michael L Eisenberg
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| |
Collapse
|
29
|
Lecoutre S, Maqdasy S, Breton C. Maternal obesity as a risk factor for developing diabetes in offspring: An epigenetic point of view. World J Diabetes 2021; 12:366-382. [PMID: 33889285 PMCID: PMC8040079 DOI: 10.4239/wjd.v12.i4.366] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 01/30/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023] Open
Abstract
According to the developmental origin of health and disease concept, the risk of many age-related diseases is not only determined by genetic and adult lifestyle factors but also by factors acting during early development. In particular, maternal obesity and neonatal accelerated growth predispose offspring to overweight and type 2 diabetes (T2D) in adulthood. This concept mainly relies on the developmental plasticity of adipose tissue and pancreatic β-cell programming in response to suboptimal milieu during the perinatal period. These changes result in unhealthy hypertrophic adipocytes with decreased capacity to store fat, low-grade inflammation and loss of insulin-producing pancreatic β-cells. Over the past years, many efforts have been made to understand how maternal obesity induces long-lasting adipose tissue and pancreatic β-cell dysfunction in offspring and what are the molecular basis of the transgenerational inheritance of T2D. In particular, rodent studies have shed light on the role of epigenetic mechanisms in linking maternal nutritional manipulations to the risk for T2D in adulthood. In this review, we discuss epigenetic adipocyte and β-cell remodeling during development in the progeny of obese mothers and the persistence of these marks as a basis of obesity and T2D predisposition.
Collapse
Affiliation(s)
- Simon Lecoutre
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141-86, Sweden
- University of Lille, EA4489, Maternal Malnutrition and Programming of Metabolic Diseases, Lille 59000, France
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141-86, Sweden
- Clermont-Ferrand CHU, Department of Endocrinology, Diabetology and Metabolic Diseases, Clermont-Ferrand 63003, France
| | - Christophe Breton
- University of Lille, EA4489, Maternal Malnutrition and Programming of Metabolic Diseases, Lille 59000, France
- U1283-UMR8199-EGID, University of Lille, Institut National de la Santé Et de la Recherche Médicale, Centre National de la Recherche Scientifique, Lille 59000, France
| |
Collapse
|
30
|
Legoff L, D'Cruz SC, Bouchekhchoukha K, Monfort C, Jaulin C, Multigner L, Smagulova F. In utero exposure to chlordecone affects histone modifications and activates LINE-1 in cord blood. Life Sci Alliance 2021; 4:4/6/e202000944. [PMID: 33837044 PMCID: PMC8091598 DOI: 10.26508/lsa.202000944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 01/06/2023] Open
Abstract
In utero exposure to chlordecone affects chromatin and leads to activation of retroelements. This study shows the changes induced by chlordecone in human umbilical cord blood and blood-derived cell line. Environmental factors can induce detrimental consequences into adulthood life. In this study, we examined the epigenetic effects induced by in utero chlordecone (CD) exposure on human male cord blood as well as in blood-derived Ke-37 cell line. Genome-wide analysis of histone H3K4me3 distribution revealed that genes related to chromosome segregation, chromatin organization, and cell cycle have altered occupancy in their promoters. The affected regions were enriched in ESR1, SP family, and IKZF1 binding motifs. We also observed a global reduction in H3K9me3, markedly in repeated sequences of the genome. Decrease in H3K9me3 after CD exposure correlates with decreased methylation in LINE-1 promoters and telomere length extension. These observations on human cord blood were assessed in the Ke-37 human cell line. H3K4me3 and the expression of genes related to immune response, DNA repair, and chromatin organization, which were affected in human cord blood were also altered in CD-exposed Ke-37 cells. Our data suggest that developmental exposure to CD leads to profound changes in histone modification patterns and affects the processes controlled by them in human cord blood.
Collapse
Affiliation(s)
- Louis Legoff
- University of Rennes, EHESP, Inserm, Institut de Recherche en Santé, Environnement et Travail (Irset)-UMR_S 1085, Rennes, France
| | - Shereen Cynthia D'Cruz
- University of Rennes, EHESP, Inserm, Institut de Recherche en Santé, Environnement et Travail (Irset)-UMR_S 1085, Rennes, France
| | - Katia Bouchekhchoukha
- University of Rennes, EHESP, Inserm, Institut de Recherche en Santé, Environnement et Travail (Irset)-UMR_S 1085, Rennes, France
| | - Christine Monfort
- University of Rennes, EHESP, Inserm, Institut de Recherche en Santé, Environnement et Travail (Irset)-UMR_S 1085, Rennes, France
| | - Christian Jaulin
- Institut de Génétique et Développement de Rennes, Epigenetics and Cancer Group, UMR 6290 CNRS, Université Rennes 1, Rennes Cedex, France
| | - Luc Multigner
- University of Rennes, EHESP, Inserm, Institut de Recherche en Santé, Environnement et Travail (Irset)-UMR_S 1085, Rennes, France
| | - Fatima Smagulova
- University of Rennes, EHESP, Inserm, Institut de Recherche en Santé, Environnement et Travail (Irset)-UMR_S 1085, Rennes, France
| |
Collapse
|
31
|
Raad G, Serra F, Martin L, Derieppe MA, Gilleron J, Costa VL, Pisani DF, Amri EZ, Trabucchi M, Grandjean V. Paternal multigenerational exposure to an obesogenic diet drives epigenetic predisposition to metabolic diseases in mice. eLife 2021; 10:61736. [PMID: 33783350 PMCID: PMC8051948 DOI: 10.7554/elife.61736] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 03/28/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is a growing societal scourge. Recent studies have uncovered that paternal excessive weight induced by an unbalanced diet affects the metabolic health of offspring. These reports mainly employed single-generation male exposure. However, the consequences of multigenerational unbalanced diet feeding on the metabolic health of progeny remain largely unknown. Here, we show that maintaining paternal Western diet feeding for five consecutive generations in mice induces an enhancement in fat mass and related metabolic diseases over generations. Strikingly, chow-diet-fed progenies from these multigenerational Western-diet-fed males develop a 'healthy' overweight phenotype characterized by normal glucose metabolism and without fatty liver that persists for four subsequent generations. Mechanistically, sperm RNA microinjection experiments into zygotes suggest that sperm RNAs are sufficient for establishment but not for long-term maintenance of epigenetic inheritance of metabolic pathologies. Progressive and permanent metabolic deregulation induced by successive paternal Western-diet-fed generations may contribute to the worldwide epidemic of metabolic diseases.
Collapse
Affiliation(s)
- Georges Raad
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France.,Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - Fabrizio Serra
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| | - Luc Martin
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | | | - Jérôme Gilleron
- Université Côte d'Azur, Inserm, C3M, Team Cellular and Molecular Pathophysiology of Obesity and Diabetes (7), Nice, France
| | - Vera L Costa
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| | | | | | - Michele Trabucchi
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| | - Valerie Grandjean
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| |
Collapse
|
32
|
Izquierdo V, Palomera-Ávalos V, Pallàs M, Griñán-Ferré C. Resveratrol Supplementation Attenuates Cognitive and Molecular Alterations under Maternal High-Fat Diet Intake: Epigenetic Inheritance over Generations. Int J Mol Sci 2021; 22:1453. [PMID: 33535619 PMCID: PMC7867164 DOI: 10.3390/ijms22031453] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Environmental factors such as maternal high-fat diet (HFD) intake can increase the risk of age-related cognitive decline in adult offspring. Epigenetic mechanisms are a possible link between diet effect and neurodegeneration across generations. Here, we found a significant decrease in triglyceride levels in a high-fat diet with resveratrol (RSV) HFD + RSV group and the offspring. Firstly, we obtained better cognitive performance in HFD+RSV groups and their offspring. Molecularly, a significant increase in DNA methylation (5-mC) levels, as well as increased gene expression of DNA methyltransferase 1 (Dnmt1) and Dnmt3a in HFD + RSV F1 group, were found. Furthermore, a significant increase of N6-Methyladenosine methylation (m6A) levels in HFD+RSV F1, as well as changes in gene expression of its enzymes Methyltransferase like 3 (Mettl3) and FTO alpha-ketoglutarate dependent dioxygenase (Fto) were found. Moreover, we found a decrease in gene expression levels of pro-inflammatory markers such as Interleukin 1β (Il1-β), Interleukin 6 (Il-6), Tumor necrosis factor-α (Tnf-α), C-X-C motif chemokine ligand 10 (Cxcl-10), the pro-inflammatory factors monocyte chemoattractant protein 1 (Mcp-1) and Tumor growth factor-β1 (Tgf-β1) in HFD+RSV and HFD+RSV F1 groups. Moreover, there was increased gene expression of neurotrophins such as Neural growth factor (Ngf), Neurotrophin-3 (Nt3), and its receptors Tropomyosin receptor kinase TrkA and TrkB. Likewise, an increase in protein levels of brain-derived neurotrophic factor (BDNF) and phospho-protein kinase B (p-Akt) in HFD+RSV F1 was found. These results suggest that maternal RSV supplementation under HFD intake prevents cognitive decline in senescence-accelerated mice prone 8 (SAMP8) adult offspring, promoting a reduction in triglycerides and leptin plasma levels, changes in the pro-inflammatory profile, and restoring the epigenetic landscape as well as synaptic plasticity.
Collapse
Affiliation(s)
- Vanesa Izquierdo
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències—Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain; (V.I.); (M.P.)
| | - Verónica Palomera-Ávalos
- Department of Cellular and Molecular Biology, University Center of Biological and Agricultural Sciences, University of Guadalajara, km 15.5 Guadalajara-Nogales highway, 45110 Zapopan, Jalisco, Mexico;
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències—Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain; (V.I.); (M.P.)
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències—Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain; (V.I.); (M.P.)
| |
Collapse
|
33
|
Cruz-Carrillo G, Camacho-Morales A. Metabolic Flexibility Assists Reprograming of Central and Peripheral Innate Immunity During Neurodevelopment. Mol Neurobiol 2021; 58:703-718. [PMID: 33006752 DOI: 10.1007/s12035-020-02154-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/28/2020] [Indexed: 01/03/2023]
Abstract
Central innate immunity assists time-dependent neurodevelopment by recruiting and interacting with peripheral immune cells. Microglia are the major player of central innate immunity integrating peripheral signals arising from the circumventricular regions lacking the blood-brain barrier (BBB), via neural afferent pathways such as the vagal nerve and also by choroid plexus into the brain ventricles. Defective and/or unrestrained activation of central and peripheral immunity during embryonic development might set an aberrant connectome establishment and brain function, leading to major psychiatric disorders in postnatal stages. Molecular candidates leading to central and peripheral innate immune overactivation identified metabolic substrates and lipid species as major contributors of immunological priming, supporting the role of a metabolic flexibility node during trained immunity. Mechanistically, trained immunity is established by an epigenetic program including DNA methylation and histone acetylation, as the major molecular epigenetic signatures to set immune phenotypes. By definition, immunological training sets reprogramming of innate immune cells, enhancing or repressing immune responses towards a second challenge which potentially might contribute to neurodevelopment disorders. Notably, the innate immune training might be set during pregnancy by maternal immune activation stimuli. In this review, we integrate the most valuable scientific evidence supporting the role of metabolic cues assisting metabolic flexibility, leading to innate immune training during development and its effects on aberrant neurological phenotypes in the offspring. We also add reports supporting the role of methylation and histone acetylation signatures as a major epigenetic mechanism regulating immune training.
Collapse
Affiliation(s)
- Gabriela Cruz-Carrillo
- Departamento de Bioquímica. Facultad de Medicina,, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Alberto Camacho-Morales
- Departamento de Bioquímica. Facultad de Medicina,, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico.
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
34
|
Paternal Methyl Donor Supplementation in Rats Improves Fertility, Physiological Outcomes, Gut Microbial Signatures and Epigenetic Markers Altered by High Fat/High Sucrose Diet. Int J Mol Sci 2021; 22:ijms22020689. [PMID: 33445606 PMCID: PMC7826956 DOI: 10.3390/ijms22020689] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 02/07/2023] Open
Abstract
Increased consumption of high fat/sucrose (HF/S) diets has contributed to rising rates of obesity and its co-morbidities globally, while also negatively impacting male reproductive health. Our objective was to examine whether adding a methyl donor cocktail to paternal HF/S diet (HF/S+M) improves health status in fathers and offspring. From 3–12 weeks of age, male Sprague Dawley rats consumed a HF/S or HF/S+M diet. Offspring were followed until 16 weeks of age. Body composition, metabolic markers, gut microbiota, DNA methyltransferase (DNMT) and microRNA expression were measured in fathers and offspring. Compared to HF/S, paternal HF/S+M diet reduced fat mass in offspring (p < 0.005). HF/S+M fathers consumed 16% fewer kcal/day, which persisted in HF/S+M female offspring and was explained in part by changes in serum glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) levels. Compared to HF/S, HF/S+M fathers had a 33% improvement in days until conception and 300% fewer stillbirths. In fathers, adipose tissue DNMT3a and hepatic miR-34a expression were reduced with HF/S+M. Adult male offspring showed upregulated miR-24, -33, -122a and -143 expression while females exhibited downregulated miR-33 expression. Fathers and offspring presented differences in gut microbial signatures. Supplementing a paternal HF/S diet with methyl-donors improved fertility, physiological outcomes, epigenetic and gut microbial signatures intergenerationally.
Collapse
|
35
|
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
|
36
|
Heber MF, Ptak GE. The effects of assisted reproduction technologies on metabolic health and disease†. Biol Reprod 2020; 104:734-744. [PMID: 33330924 PMCID: PMC8023432 DOI: 10.1093/biolre/ioaa224] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/12/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022] Open
Abstract
The increasing prevalence of metabolic diseases places a substantial burden on human health throughout the world. It is believed that predisposition to metabolic disease starts early in life, a period of great susceptibility to epigenetic reprogramming due to environmental insults. Assisted reproductive technologies (ART), i.e., treatments for infertility, may affect embryo development, resulting in multiple adverse health outcomes in postnatal life. The most frequently observed alteration in ART pregnancies is impaired placental nutrient transfer. Moreover, consequent intrauterine growth restriction and low birth weight followed by catch-up growth can all predict future obesity, insulin resistance, and chronic metabolic diseases. In this review, we have focused on evidence of adverse metabolic alterations associated with ART, which can contribute to the development of chronic adult-onset diseases, such as metabolic syndrome, type 2 diabetes, and cardiovascular disease. Due to high phenotypic plasticity, ART pregnancies can produce both offspring with adverse health outcomes, as well as healthy individuals. We further discuss the sex-specific and age-dependent metabolic alterations reflected in ART offspring, and how the degree of interference of a given ART procedure (from mild to more severe manipulation of the egg) affects the occurrence and degree of offspring alterations. Over the last few years, studies have reported signs of cardiometabolic alterations in ART offspring that are detectable at a young age but that do not appear to constitute a high risk of disease and morbidity per se. These abnormal phenotypes could be early indicators of the development of chronic diseases, including metabolic syndrome, in adulthood. The early detection of metabolic alterations could contribute to preventing the onset of disease in adulthood. Such early interventions may counteract the risk factors and improve the long-term health of the individual.
Collapse
Affiliation(s)
| | - Grażyna Ewa Ptak
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.,Faculty of Biosciences, University of Teramo, Teramo, Italy
| |
Collapse
|
37
|
Rutkowska J, Lagisz M, Bonduriansky R, Nakagawa S. Mapping the past, present and future research landscape of paternal effects. BMC Biol 2020; 18:183. [PMID: 33246472 PMCID: PMC7694421 DOI: 10.1186/s12915-020-00892-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although in all sexually reproducing organisms an individual has a mother and a father, non-genetic inheritance has been predominantly studied in mothers. Paternal effects have been far less frequently studied, until recently. In the last 5 years, research on environmentally induced paternal effects has grown rapidly in the number of publications and diversity of topics. Here, we provide an overview of this field using synthesis of evidence (systematic map) and influence (bibliometric analyses). RESULTS We find that motivations for studies into paternal effects are diverse. For example, from the ecological and evolutionary perspective, paternal effects are of interest as facilitators of response to environmental change and mediators of extended heredity. Medical researchers track how paternal pre-fertilization exposures to factors, such as diet or trauma, influence offspring health. Toxicologists look at the effects of toxins. We compare how these three research guilds design experiments in relation to objects of their studies: fathers, mothers and offspring. We highlight examples of research gaps, which, in turn, lead to future avenues of research. CONCLUSIONS The literature on paternal effects is large and disparate. Our study helps in fostering connections between areas of knowledge that develop in parallel, but which could benefit from the lateral transfer of concepts and methods.
Collapse
Affiliation(s)
- Joanna Rutkowska
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Kraków, Poland
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Malgorzata Lagisz
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Russell Bonduriansky
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Shinichi Nakagawa
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| |
Collapse
|
38
|
Ferrari N, Schmitz L, Schmidt N, Mahabir E, Van de Vondel P, Merz WM, Lehmacher W, Stock S, Brockmeier K, Ensenauer R, Fehm T, Joisten C. A lifestyle intervention during pregnancy to reduce obesity in early childhood: the study protocol of ADEBAR - a randomized controlled trial. BMC Sports Sci Med Rehabil 2020; 12:55. [PMID: 32944252 PMCID: PMC7487987 DOI: 10.1186/s13102-020-00198-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/19/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND The prevalence of obesity in childhood is increasing worldwide and may be affected by genetic factors and the lifestyle (exercise, nutrition behavior) of expectant parents. Lifestyle factors affect adipokines, namely leptin, resistin, and adiponectin as well as cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), which are involved in the regulation of maternal metabolic homeostasis, glucose metabolism, and the development of insulin resistance, metabolic syndrome, gestational diabetes mellitus, and hypertension. However, studies focusing on the effect of exercise or a combination of parental exercise and nutrition on the above-mentioned markers in newborns (venous cord blood) and especially on the long-term development of infants' weight gain are lacking. The study will investigate the effects of a multimodal intervention (regular exercise, diet) on parental and childhood adipocytokines (leptin, resistin, adiponectin, TNF-α, IL-6, BDNF). The effect of a lifestyle-related change in "fetal environmental conditions" on the long-term weight development of the child up to the age of two will also be assessed. METHODS/DESIGN A randomized multi-center controlled trial will be conducted in Germany, comparing supervised aerobic and resistance training 2x/week (13th to 36th weeks of gestation) and nutritional counseling (6th to 36th weeks of gestation) during pregnancy with usual care. Thirty women (pre-pregnancy Body Mass Index ≥25 kg/m2, 6th-10th week of gestation) will be included in each group. Maternal anthropometric and physical measurements as well as blood sampling will occur at the 6th-10th, 13th-14th, 21st-24th, and 36th week of gestation, at delivery as well as 8 weeks and 24 months postpartum. Neonatal measurements and umbilical blood sampling will be performed at birth. Maternal and infants' weight development will be assessed every 6 months till 24 months postpartum. A difference in childhood BMI of 1 kg/m2 at the age of two years between both groups will be assumed. A power size of 80% using a significance level of 0.05 and an effect size of 1.0 is presumed. DISCUSSION A better understanding of how lifestyle-related changes in the fetal environment might influence infants' outcome after two years of life could have a profound impact on the prevention and development of infants' obesity. TRIAL REGISTRATION The trial is registered at the German Clinical Trial Register (DRKS00007702); Registered on 10th of August 2016; retrospectively registered https://www.drks.de/drks_web/navigate.do?navigationId=trial.HTML&TRIAL_ID=DRKS00007702.
Collapse
Affiliation(s)
- Nina Ferrari
- Cologne Centre for Prevention in Childhood and Youth/ Heart Centre Cologne, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Department for physical activity in public health, Institute of Movement and Neurosciences, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Laura Schmitz
- Department for physical activity in public health, Institute of Movement and Neurosciences, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Nikola Schmidt
- Department for physical activity in public health, Institute of Movement and Neurosciences, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
- Cologne Center for Musculoskeletal Biomechanics, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 9, 50931 Cologne, Germany
| | | | - Waltraut M. Merz
- Department of Obstetrics and Prenatal Medicine, University Bonn Medical School, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Walter Lehmacher
- Department of Biometry (IMSIE), Faculty of medicine, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Stephanie Stock
- Cologne Institute for Health Economics and Clinical Epidemiology, The University Hospital of Cologne, Gleueler Strasse 176 - 178/II, 50935 Cologne, Germany
| | - Konrad Brockmeier
- Cologne Centre for Prevention in Childhood and Youth/ Heart Centre Cologne, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Department of Paediatric Cardiology, Heart Centre Cologne, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Regina Ensenauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital, University of Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
- Institute of Child Nutrition, Max Rubner-Institut, Haid-und-Neu-Str. 9, 76131 Karlsruhe, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, University of Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Christine Joisten
- Cologne Centre for Prevention in Childhood and Youth/ Heart Centre Cologne, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Department for physical activity in public health, Institute of Movement and Neurosciences, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| |
Collapse
|
39
|
Schellong K, Melchior K, Ziska T, Rancourt RC, Henrich W, Plagemann A. Maternal but Not Paternal High-Fat Diet (HFD) Exposure at Conception Predisposes for 'Diabesity' in Offspring Generations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E4229. [PMID: 32545776 PMCID: PMC7345576 DOI: 10.3390/ijerph17124229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 01/02/2023]
Abstract
While environmental epigenetics mainly focuses on xenobiotic endocrine disruptors, dietary composition might be one of the most important environmental exposures for epigenetic modifications, perhaps even for offspring generations. We performed a large-scale rat study on key phenotypic consequences from parental (F0) high-caloric, high-fat diet (HFD) food intake, precisely and specifically at mating/conception, focusing on 'diabesity' risk in first- (F1) and second- (F2) generation offspring of both sexes. F0 rats (maternal or paternal, respectively) received HFD overfeeding, starting six weeks prior to mating with normally fed control rats. The maternal side F1 offspring of both sexes developed a 'diabesity' predisposition throughout life (obesity, hyperleptinemia, hyperglycemia, insulin resistance), while no respective alterations occurred in the paternal side F1 offspring, neither in males nor in females. Mating the maternal side F1 females with control males under standard feeding conditions led, again, to a 'diabesity' predisposition in the F2 generation, which, however, was less pronounced than in the F1 generation. Our observations speak in favor of the critical impact of maternal but not paternal metabolism around the time frame of reproduction for offspring metabolic health over generations. Such fundamental phenotypic observations should be carefully considered in front of detailed molecular epigenetic approaches on eventual mechanisms.
Collapse
Affiliation(s)
- Karen Schellong
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (K.S.); (K.M.); (T.Z.); (R.C.R.)
| | - Kerstin Melchior
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (K.S.); (K.M.); (T.Z.); (R.C.R.)
| | - Thomas Ziska
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (K.S.); (K.M.); (T.Z.); (R.C.R.)
| | - Rebecca C. Rancourt
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (K.S.); (K.M.); (T.Z.); (R.C.R.)
| | - Wolfgang Henrich
- Clinic of Obstetrics, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany;
| | - Andreas Plagemann
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (K.S.); (K.M.); (T.Z.); (R.C.R.)
| |
Collapse
|
40
|
Lecoutre S, Kwok KHM, Petrus P, Lambert M, Breton C. Epigenetic Programming of Adipose Tissue in the Progeny of Obese Dams. Curr Genomics 2020; 20:428-437. [PMID: 32477000 PMCID: PMC7235387 DOI: 10.2174/1389202920666191118092852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 01/13/2023] Open
Abstract
According to the Developmental Origin of Health and Disease (DOHaD) concept, maternal obesity and the resulting accelerated growth in neonates predispose offspring to obesity and associated metabolic diseases that may persist across generations. In this context, the adipose tissue has emerged as an important player due to its involvement in metabolic health, and its high potential for plasticity and adaptation to environmental cues. Recent years have seen a growing interest in how maternal obesity induces long-lasting adipose tissue remodeling in offspring and how these modifications could be transmitted to subsequent generations in an inter- or transgenerational manner. In particular, epigenetic mechanisms are thought to be key players in the developmental programming of adipose tissue, which may partially mediate parts of the transgenerational inheritance of obesity. This review presents data supporting the role of maternal obesity in the developmental programming of adipose tissue through epigenetic mechanisms. Inter- and transgenerational effects on adipose tissue expansion are also discussed in this review.
Collapse
Affiliation(s)
- Simon Lecoutre
- University of Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F-59000 Lille, France.,Department of Medicine (H7), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Kelvin H M Kwok
- Department of Biosciences and Nutrition, Karolinska Insitutet, 141 86 Stockholm, Sweden
| | - Paul Petrus
- Department of Medicine (H7), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Mélanie Lambert
- Department of Medicine (H7), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Christophe Breton
- University of Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F-59000 Lille, France
| |
Collapse
|
41
|
Guo T, Luo F, Lin Q. You are affected by what your parents eat: Diet, epigenetics, transgeneration and intergeneration. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
42
|
Un Nisa K, Reza MI. Key Relevance of Epigenetic Programming of Adiponectin Gene in Pathogenesis of Metabolic Disorders. Endocr Metab Immune Disord Drug Targets 2020; 20:506-517. [DOI: 10.2174/1871530319666190801142637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022]
Abstract
Background & Objective::
Significant health and social burdens have been created by the
growth of metabolic disorders like type 2 diabetes mellitus (T2DM), atherosclerosis, and non-alcoholic
steatohepatitis, worldwide. The number of the affected population is as yet rising, and it is assessed
that until 2030, 4−5 million individuals will acquire diabetes. A blend of environmental, genetic, epigenetic,
and other factors, such as diet, are accountable for the initiation and progression of metabolic
disorders. Several researches have shown strong relevance of adiponectin gene and metabolic disorders.
In this review, the potential influence of epigenetic mechanisms of adiponectin gene “ADIPOQ”
on increasing the risk of developing metabolic disorders and their potential in treating this major disorder
are discussed.
Results & Conclusion::
Various studies have postulated that a series of factors such as maternal High
fat diet (HFD), oxidative stress, pro-inflammatory mediators, sleep fragmentation throughout lifetime,
from gestation to old age, could accumulate epigenetic marks, including histone remodeling, DNA
methylation, and microRNAs (miRNAs) that, in turn, alter the expression of ADIPOQ gene and result
in hypoadiponectinemia which precipitates insulin resistance (IR) that in turn might induce or accelerate
the onset and development of metabolic disorder. A better understanding of global patterns of epigenetic
modifications and further their alterations in metabolic disorders will bestow better treatment
strategies design.
Collapse
Affiliation(s)
- Kaiser Un Nisa
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| | - Mohammad Irshad Reza
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| |
Collapse
|
43
|
Ma M, Zhang W, Zhang J, Liang Z, Kuang Y, Wang Y. Effect of paternal body mass index on neonatal outcomes of singletons after frozen-thawed embryo transfer cycles: analysis of 7,908 singleton newborns. Fertil Steril 2020; 113:1215-1223.e1. [PMID: 32402450 DOI: 10.1016/j.fertnstert.2020.02.100] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/06/2020] [Accepted: 02/17/2020] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To investigate the effect of paternal body mass index (BMI) on neonatal outcomes of singletons after frozen-thawed embryo transfer (FET) cycles. DESIGN Retrospective cohort study. SETTING Tertiary-care academic medical center. PATIENT(S) A total of 7,908 singleton newborns were divided into four categories based on their paternal BMI: 284 (3.6%) infants were in the paternal underweight category, 4,678 (59.2%) infants were in the paternal normal weight category, 2,585 (32.7%) infants were in the paternal overweight category, and 361 (4.6%) infants were in the paternal obesity category. In addition, we included only infants of women with normal BMI (18.5 kg/m2 ≤ BMI < 25 kg/m2). INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Neonatal outcomes. RESULT(S) The rates of large for gestational age (LGA) infants were significantly higher among those in the paternal overweight and obesity categories than those in the paternal underweight categories. The rates of very LGA were higher among infants in the paternal overweight categories and lower among infants in the paternal underweight categories than the rates in normal controls. The rates of fetal macrosomia were higher among infants in the paternal overweight categories than among infants in the paternal normal weight categories. Compared with normal controls, Z-scores (gestational age- and sex-adjusted birthweight) were significantly higher among the infants in the paternal overweight and paternal obesity categories and significantly lower among the infants in the paternal underweight categories. A positive association was observed in a multiple linear regression model between paternal BMI and newborn birthweights after adjustment for several potential confounders. CONCLUSION(S) Paternal BMI had an independent impact on the birthweight of singletons born after FET cycles. Paternal overweight and paternal obesity were independent risk factors for having LGA infants after FET cycles. Furthermore, paternal overweight was an independent risk factor for fathering infants with macrosomia or very LGA infants after FET cycles.
Collapse
Affiliation(s)
- Meng Ma
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated with JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Wei Zhang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated with JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Jie Zhang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated with JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Zhou Liang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated with JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated with JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Yun Wang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated with JiaoTong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
44
|
Kim JH, Lee S, Cho EJ. The Protective Effects of Acer okamotoanum and Isoquercitrin on Obesity and Amyloidosis in a Mouse Model. Nutrients 2020; 12:nu12051353. [PMID: 32397362 PMCID: PMC7284521 DOI: 10.3390/nu12051353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity increases risk of Alzheimer's Disease (AD). A high fat diet (HFD) can lead to amyloidosis and amyloid beta (Aβ) accumulation, which are hallmarks of AD. In this study, protective effects of the ethyl acetate fraction of Acer okamotoanum (EAO) and isoquercitrin were evaluated on obesity and amyloidosis in the HFD- and Aβ-induced mouse model. To induce obesity and AD by HFD and Aβ, mice were provided with HFD for 10 weeks and were intracerebroventricularly injected with Aβ25-35. For four weeks, 100 and 10 mg/kg/day of EAO and isoquercitrin, respectively, were administered orally. Administration of EAO and isoquercitrin significantly decreased body weight in HFD and Aβ-injected mice. Additionally, EAO- and isoquercitrin-administered groups attenuated abnormal adipokines release via a decrease in leptin and an increase in adiponectin levels compared with the control group. Furthermore, HFD and Aβ-injected mice had damaged liver tissues, but EAO- and isoquercitrin-administered groups attenuated liver damage. Moreover, administration of EAO and isoquercitrin groups down-regulated amyloidosis-related proteins in the brain such as β-secretase, presenilin (PS)-1 and PS-2 compared with HFD and Aβ-injected mice. This study indicated that EAO and isoquercitrin attenuated HFD and Aβ-induced obesity and amyloidosis, suggesting that they could be effective in preventing and treating both obesity and AD.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan 46241, Korea;
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Korea;
| | - Eun Ju Cho
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan 46241, Korea;
- Correspondence: ; Tel.: +82-51-510-2837; Fax: +82-51-583-3648
| |
Collapse
|
45
|
Claycombe-Larson KG, Bundy AN, Roemmich JN. Paternal high-fat diet and exercise regulate sperm miRNA and histone methylation to modify placental inflammation, nutrient transporter mRNA expression and fetal weight in a sex-dependent manner. J Nutr Biochem 2020; 81:108373. [PMID: 32422425 DOI: 10.1016/j.jnutbio.2020.108373] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/10/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
We previously have shown that male offspring (F1) of fathers (F0) fed a high-fat (HF) diet and that exercised had greater skeletal muscle insulin signaling and reduced type 2 diabetes mellitus (T2DM) risk compared to fathers fed HF diet and that remained sedentary. The current study extends this work by hypothesizing that F0 HF diet and exercise regulate F1 T2DM risk by alterations in placental tissue growth via changes in sperm miRNA expression. To test these hypotheses, 3-week-old male C57BL/6 mice were fed a normal-fat diet (16% fat) or an HF diet (45% fat) and assigned to either voluntary wheel running exercise or cage activity for 3 months. Results showed that F0 sperm miRNA 193b expression was decreased while miRNA 204 was increased by paternal exercise. Protein expression of dimethylated histone 3 lysine 9 was decreased with F0 HF diet. Placental and fetal tissue weights were decreased by F0 HF diet in F1 males. Placental interleukin-1β and tumor necrosis factor (TNF)-α mRNA expression was reduced by paternal exercise, while nutrient transporter mRNA expression was decreased by paternal HF diet only in the placentae of F1 females. Treatment of primary placental cell with miRNA 193b inhibited TNF-α mRNA expression, and treatment of TNF-α decreased SLC38a2 mRNA expression. Moreover, paternal exercise increased body weight at weaning in a female offspring. These results demonstrate that placental tissue weight, placental nutrient transporter gene expression and fetal weights are altered by paternal exercise, while placental inflammatory gene expression is influenced by paternal exercise in offspring in a sex-specific manner.
Collapse
Affiliation(s)
- Kate G Claycombe-Larson
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA.
| | - Amy N Bundy
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - James N Roemmich
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| |
Collapse
|
46
|
Harasymowicz NS, Choi YR, Wu CL, Iannucci L, Tang R, Guilak F. Intergenerational Transmission of Diet-Induced Obesity, Metabolic Imbalance, and Osteoarthritis in Mice. Arthritis Rheumatol 2020; 72:632-644. [PMID: 31646754 DOI: 10.1002/art.41147] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 10/17/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Obesity and osteoarthritis (OA) are 2 major public health issues affecting millions of people worldwide. Whereas parental obesity affects the predisposition to diseases such as cancer or diabetes in children, transgenerational influences on musculoskeletal conditions such as OA are poorly understood. This study was undertaken to assess the intergenerational effects of a parental/grandparental high-fat diet on the metabolic and skeletal phenotype, systemic inflammation, and predisposition to OA in 2 generations of offspring in mice. METHODS Metabolic phenotype and predisposition to OA were investigated in the first and second (F1 and F2) generations of offspring (n = 10-16 mice per sex per diet) bred from mice fed a high-fat diet (HFD) or a low-fat control diet. OA was induced by destabilizing the medial meniscus. OA, synovitis, and adipose tissue inflammation were determined histologically, while bone changes were measured using micro-computed tomography. Serum and synovial cytokines were measured by multiplex assay. RESULTS Parental high-fat feeding showed an intergenerational effect, with inheritance of increased weight gain (up to 19% in the F1 generation and 9% in F2), metabolic imbalance, and injury-induced OA in at least 2 generations of mice, despite the fact that the offspring were fed the low-fat diet. Strikingly, both F1 and F2 female mice showed an increased predisposition to injury-induced OA (48% higher predisposition in F1 and 19% in F2 female mice fed the HFD) and developed bone microarchitectural changes that were attributable to parental and grandparental high-fat feeding. CONCLUSION The results of this study reveal a detrimental effect of parental HFD and obesity on the musculoskeletal integrity of 2 generations of offspring, indicating the importance of further investigation of these effects. An improved understanding of the mechanisms involved in the transmissibility of diet-induced changes through multiple generations may help in the development of future therapies that would target the effects of obesity on OA and related conditions.
Collapse
Affiliation(s)
- Natalia S Harasymowicz
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Yun-Rak Choi
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri, and Yonsei University College of Medicine, Seoul, South Korea
| | - Chia-Lung Wu
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Leanne Iannucci
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Ruhang Tang
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| | - Farshid Guilak
- Washington University in St. Louis and Shriners Hospitals for Children, St. Louis, Missouri
| |
Collapse
|
47
|
Zhu Z, Cao F, Li X. Epigenetic Programming and Fetal Metabolic Programming. Front Endocrinol (Lausanne) 2019; 10:764. [PMID: 31849831 PMCID: PMC6901800 DOI: 10.3389/fendo.2019.00764] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/21/2019] [Indexed: 12/30/2022] Open
Abstract
Fetal metabolic programming caused by the adverse intrauterine environment can induce metabolic syndrome in adult offspring. Adverse intrauterine environment introduces fetal long-term relatively irreversible changes in organs and metabolism, and thus causes fetal metabolic programming leading metabolic syndrome in adult offspring. Fetal metabolic programming of obesity and insulin resistance plays a key role in this process. The mechanism of fetal metabolic programming is still not very clear. It is suggested that epigenetic programming, also induced by the adverse intrauterine environment, is a critical underlying mechanism of fetal metabolic programming. Fetal epigenetic programming affects gene expression changes and cellular function through epigenetic modifications without DNA nucleotide sequence changes. Epigenetic modifications can be relatively stably retained and transmitted through mitosis and generations, and thereby induce the development of metabolic syndrome in adult offspring. This manuscript provides an overview of the critical role of epigenetic programming in fetal metabolic programming.
Collapse
Affiliation(s)
- Ziqiang Zhu
- Children's Hospital of Soochow University, Suzhou, China
- Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, China
| | - Fang Cao
- Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, China
| | - Xiaozhong Li
- Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
48
|
Legoff L, D’Cruz SC, Tevosian S, Primig M, Smagulova F. Transgenerational Inheritance of Environmentally Induced Epigenetic Alterations during Mammalian Development. Cells 2019; 8:cells8121559. [PMID: 31816913 PMCID: PMC6953051 DOI: 10.3390/cells8121559] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Genetic studies traditionally focus on DNA as the molecule that passes information on from parents to their offspring. Changes in the DNA code alter heritable information and can more or less severely affect the progeny's phenotype. While the idea that information can be inherited between generations independently of the DNA's nucleotide sequence is not new, the outcome of recent studies provides a mechanistic foundation for the concept. In this review, we attempt to summarize our current knowledge about the transgenerational inheritance of environmentally induced epigenetic changes. We focus primarily on studies using mice but refer to other species to illustrate salient points. Some studies support the notion that there is a somatic component within the phenomenon of epigenetic inheritance. However, here, we will mostly focus on gamete-based processes and the primary molecular mechanisms that are thought to contribute to epigenetic inheritance: DNA methylation, histone modifications, and non-coding RNAs. Most of the rodent studies published in the literature suggest that transgenerational epigenetic inheritance through gametes can be modulated by environmental factors. Modification and redistribution of chromatin proteins in gametes is one of the major routes for transmitting epigenetic information from parents to the offspring. Our recent studies provide additional specific cues for this concept and help better understand environmental exposure influences fitness and fidelity in the germline. In summary, environmental cues can induce parental alterations and affect the phenotypes of offspring through gametic epigenetic inheritance. Consequently, epigenetic factors and their heritability should be considered during disease risk assessment.
Collapse
Affiliation(s)
- Louis Legoff
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
| | - Shereen Cynthia D’Cruz
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
| | - Sergei Tevosian
- University of Florida, Department of Physiological Sciences Box 100144, 1333 Center Drive, Gainesville, FL 32610, USA;
| | - Michael Primig
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
| | - Fatima Smagulova
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, F-35000 Rennes, France; (L.L.); (S.C.D.); (M.P.)
- Correspondence:
| |
Collapse
|
49
|
Andreas E, Reid M, Zhang W, Moley KH. The effect of maternal high-fat/high-sugar diet on offspring oocytes and early embryo development. Mol Hum Reprod 2019; 25:717-728. [PMID: 31588490 PMCID: PMC6884416 DOI: 10.1093/molehr/gaz049] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/02/2019] [Indexed: 01/12/2023] Open
Abstract
Observational human data and several lines of animal experimental data indicate that maternal obesity impairs offspring health. Here, we comprehensively tested the model that maternal obesity causes defects in the next three generations of oocytes and embryos. We exposed female F0 mice to a high-fat/high-sugar (HF/HS) diet for 6 weeks before conception until weaning. Sires, F1 offspring and all subsequent generations were fed control chow diet. Oocytes from F1, F2 and F3 offspring of obese mothers had lower mitochondrial mass and less ATP and citrate than oocytes from offspring of control mothers. F0 blastocysts from HF/HS-exposed mice, but not F1 and F2 blastocysts, had lower mitochondrial mass and membrane potential, less citrate and ATP and smaller total cell number than F0 blastocysts from control mothers. Finally, supplementation of IVF media with the anti-oxidant mito-esculetin partially prevented the oocyte mitochondrial effects caused by maternal HF/HS diet. Our results support the idea that maternal obesity impairs offspring oocyte quality and suggest that antioxidant supplementation should be tested as a means to improve IVF outcomes for obese women.
Collapse
Affiliation(s)
- E Andreas
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - M Reid
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - W Zhang
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - K H Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
50
|
Maternal high-fat diet triggers metabolic syndrome disorders that are transferred to first and second offspring generations. Br J Nutr 2019; 123:59-71. [DOI: 10.1017/s0007114519002708] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractA high-fat (H) diet increases metabolic disorders in offspring. However, there is great variability in the literature regarding the time of exposure, composition of the H diets offered to the genitors and/or offspring and parameters evaluated. Here, we investigated the effect of a H diet subjected to the genitors on different cardio-metabolic parameters on first (F1)- and second (F2)-generation offspring. Female Fischer rats, during mating, gestation and breast-feeding, were subjected to the H diet (G0HF) or control (G0CF) diets. Part of F1 offspring becomes G1 genitors for generating the F2 offspring. After weaning, F1 and F2 rats consumed only the C diet. Nutritional, biometric, biochemical and haemodynamic parameters were evaluated. G0HF genitors had a reduction in food intake but energy intake was similar to the control group. Compared with the control group, the F1H and F2H offspring presented increased plasma leptin, insulin and fasting glucose levels, dietary intake, energy intake, adiposity index, mean arterial pressure, sympathetic drive evidenced by the hexamethonium and insulin resistance. Our data showed that only during mating, gestation and breast-feeding, maternal H diet induced cardio-metabolic disorders characteristic of human metabolic syndrome that were transferred to both females and males of F1 and F2 offspring, even if they were fed control diet after weaning. This process probably occurs due to the disturbance in mechanisms related to leptin that increases energy intake in F1H and F2H offspring. The present data reinforce the importance of balanced diet during pregnancy and breast-feeding for the health of the F1 and F2 offspring.
Collapse
|