1
|
Terracina S, Tarani L, Ceccanti M, Vitali M, Francati S, Lucarelli M, Venditti S, Verdone L, Ferraguti G, Fiore M. The Impact of Oxidative Stress on the Epigenetics of Fetal Alcohol Spectrum Disorders. Antioxidants (Basel) 2024; 13:410. [PMID: 38671857 PMCID: PMC11047541 DOI: 10.3390/antiox13040410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Fetal alcohol spectrum disorders (FASD) represent a continuum of lifelong impairments resulting from prenatal exposure to alcohol, with significant global impact. The "spectrum" of disorders includes a continuum of physical, cognitive, behavioral, and developmental impairments which can have profound and lasting effects on individuals throughout their lives, impacting their health, social interactions, psychological well-being, and every aspect of their lives. This narrative paper explores the intricate relationship between oxidative stress and epigenetics in FASD pathogenesis and its therapeutic implications. Oxidative stress, induced by alcohol metabolism, disrupts cellular components, particularly in the vulnerable fetal brain, leading to aberrant development. Furthermore, oxidative stress is implicated in epigenetic changes, including alterations in DNA methylation, histone modifications, and microRNA expression, which influence gene regulation in FASD patients. Moreover, mitochondrial dysfunction and neuroinflammation contribute to epigenetic changes associated with FASD. Understanding these mechanisms holds promise for targeted therapeutic interventions. This includes antioxidant supplementation and lifestyle modifications to mitigate FASD-related impairments. While preclinical studies show promise, further clinical trials are needed to validate these interventions' efficacy in improving clinical outcomes for individuals affected by FASD. This comprehensive understanding of the role of oxidative stress in epigenetics in FASD underscores the importance of multidisciplinary approaches for diagnosis, management, and prevention strategies. Continued research in this field is crucial for advancing our knowledge and developing effective interventions to address this significant public health concern.
Collapse
Affiliation(s)
- Sergio Terracina
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy (M.L.)
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00185 Roma, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell’Alcolismo e le sue Complicanze, 00185 Rome, Italy;
| | | | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy (M.L.)
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy (M.L.)
- Pasteur Institute Cenci Bolognetti Foundation, Sapienza University of Rome, 00185 Rome, Italy
| | - Sabrina Venditti
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University, 00185 Rome, Italy
| | - Loredana Verdone
- Institute of Molecular Biology and Pathology (IBPM-CNR), 00185 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy (M.L.)
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Roma, Italy
| |
Collapse
|
2
|
Terracina S, Ferraguti G, Tarani L, Messina MP, Lucarelli M, Vitali M, De Persis S, Greco A, Minni A, Polimeni A, Ceccanti M, Petrella C, Fiore M. Transgenerational Abnormalities Induced by Paternal Preconceptual Alcohol Drinking: Findings from Humans and Animal Models. Curr Neuropharmacol 2022; 20:1158-1173. [PMID: 34720083 PMCID: PMC9886817 DOI: 10.2174/1570159x19666211101111430] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/14/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022] Open
Abstract
Alcohol consumption during pregnancy and lactation is a widespread preventable cause of neurodevelopmental impairment in newborns. While the harmful effects of gestational alcohol use have been well documented, only recently, the role of paternal preconceptual alcohol consumption (PPAC) prior to copulating has drawn specific epigenetic considerations. Data from human and animal models have demonstrated that PPAC may affect sperm function, eliciting oxidative stress. In newborns, PPAC may induce changes in behavior, cognitive functions, and emotional responses. Furthermore, PPAC may elicit neurobiological disruptions, visuospatial impairments, hyperactivity disorders, motor skill disruptions, hearing loss, endocrine, and immune alterations, reduced physical growth, placental disruptions, and metabolic alterations. Neurobiological studies on PPAC have also disclosed changes in brain function and structure by disrupting the growth factors pathways. In particular, as shown in animal model studies, PPAC alters brain nerve growth factor (NGF) and brainderived neurotrophic factor (BDNF) synthesis and release. This review shows that the crucial topic of lifelong disabilities induced by PPAC and/or gestational alcohol drinking is quite challenging at the individual, societal, and familial levels. Since a nontoxic drinking behavior before pregnancy (for both men and women), during pregnancy, and lactation cannot be established, the only suggestion for couples planning pregnancies is to completely avoid the consumption of alcoholic beverages.
Collapse
Affiliation(s)
- Sergio Terracina
- Department of Experimental Medicine, Medical Faculty, Sapienza University of Rome, RomeItaly
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Medical Faculty, Sapienza University of Rome, RomeItaly
| | - Luigi Tarani
- Department of Pediatrics, Medical Faculty, “Sapienza” University of Rome, Rome, Italy
| | | | - Marco Lucarelli
- Department of Experimental Medicine, Medical Faculty, Sapienza University of Rome, RomeItaly
| | | | | | - Antonio Greco
- Department of Sense Organs, Sapienza University Hospital of Rome, Rome, Italy
| | - Antonio Minni
- Department of Sense Organs, Sapienza University Hospital of Rome, Rome, Italy
| | - Antonella Polimeni
- Department of Odontostomatological and Maxillofacial Sciences, Sapienza University of Rome, Rome, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell’Alcolismo e le sue Complicanze, Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Rome, Italy,Address correspondence to this author at the Institute of Biochemistry and Cell Biology (IBBC-CNR), Rome, Italy; E-mail:
| |
Collapse
|
3
|
Abstract
At-risk alcohol use is a major contributor to the global health care burden and leads to preventable deaths and diseases including alcohol addiction, alcoholic liver disease, cardiovascular disease, diabetes, traumatic injuries, gastrointestinal diseases, cancers, and fetal alcohol syndrome. Excessive and frequent alcohol consumption has increasingly been linked to alcohol-associated tissue injury and pathophysiology, which have significant adverse effects on multiple organ systems. Extensive research in animal and in vitro models has elucidated the salient mechanisms involved in alcohol-induced tissue and organ injury. In some cases, these pathophysiological mechanisms are shared across organ systems. The major alcohol- and alcohol metabolite-mediated mechanisms include oxidative stress, inflammation and immunometabolic dysregulation, gut leak and dysbiosis, cell death, extracellular matrix remodeling, endoplasmic reticulum stress, mitochondrial dysfunction, and epigenomic modifications. These mechanisms are complex and interrelated, and determining the interplay among them will make it possible to identify how they synergistically or additively interact to cause alcohol-mediated multiorgan injury. In this article, we review the current understanding of pathophysiological mechanisms involved in alcohol-induced tissue injury.
Collapse
Affiliation(s)
- Liz Simon
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA;
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Flavia M Souza-Smith
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Patricia E Molina
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA;
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
4
|
Chung DD, Pinson MR, Bhenderu LS, Lai MS, Patel RA, Miranda RC. Toxic and Teratogenic Effects of Prenatal Alcohol Exposure on Fetal Development, Adolescence, and Adulthood. Int J Mol Sci 2021; 22:ijms22168785. [PMID: 34445488 PMCID: PMC8395909 DOI: 10.3390/ijms22168785] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
Prenatal alcohol exposure (PAE) can have immediate and long-lasting toxic and teratogenic effects on an individual’s development and health. As a toxicant, alcohol can lead to a variety of physical and neurological anomalies in the fetus that can lead to behavioral and other impairments which may last a lifetime. Recent studies have focused on identifying mechanisms that mediate the immediate teratogenic effects of alcohol on fetal development and mechanisms that facilitate the persistent toxic effects of alcohol on health and predisposition to disease later in life. This review focuses on the contribution of epigenetic modifications and intercellular transporters like extracellular vesicles to the toxicity of PAE and to immediate and long-term consequences on an individual’s health and risk of disease.
Collapse
|
5
|
Koabel J, McNivens M, McKee P, Pautassi R, Bordner K, Nizhnikov M. The offspring of alcohol-exposed sires exhibit heightened ethanol intake and behavioral alterations in the elevated plus maze. Alcohol 2021; 92:65-72. [PMID: 33722694 DOI: 10.1016/j.alcohol.2021.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 12/24/2022]
Abstract
Research suggests that addictive traits are indeed heritable, but very few preclinical studies have explored transgenerational effects of paternal alcohol exposure. The present study addressed this gap in knowledge. We explored whether offspring of ethanol-exposed sires would be more likely to accept ethanol than descendants of water-exposed and control sires. We also investigated whether the second generation of ethanol-exposed descendants would accept ethanol more than controls and were more or less likely to experience anxiety-like behavior in behavioral assessments. We exposed male rats to repeated binge doses of alcohol (4 g/kg/day across 8 days), water, or left them untreated and mated them with untreated females. We then bred the offspring of these rats to test transgenerational effects of paternal alcohol exposure. We tested 14-day-old offspring from the first and second filial generation for their acceptance of ethanol and water, and measured anxiety-like behavior in 38-day-old, second-generation offspring using an elevated plus maze. The results indicate that offspring of ethanol-exposed sires increase ethanol acceptance in the first generation compared to untreated controls, whereas in the second-generation increased ethanol acceptance vs. these controls is seen in descendants of both ethanol- and vehicle-treated sires. At adolescence, the second generation of rats derived from alcohol-exposed sires exhibited significantly more time spent in the open arms and significantly more arm entries than any other group. The present study suggests that parental ethanol exposure is associated with lingering effects in the infant and adolescent offspring. The second filial generation was also found to be affected, albeit similarly by grandparental ethanol exposure or by the stress of the vehicle administration.
Collapse
Affiliation(s)
- Jennifer Koabel
- Stony Brook University, 100 Nicolls Road, Stony Brook, New York, 11794, United States.
| | - Meghan McNivens
- Stony Brook University, 100 Nicolls Road, Stony Brook, New York, 11794, United States
| | - Paul McKee
- Southern Connecticut State University, 501 Crescent St., New Haven, CT, 06515, United States
| | - Ricardo Pautassi
- Instituto de Investigaciones Medicas M. y M. Ferreyra (INIMEC-CONICET), Friuli 2434, Córdoba, Cba, 5016, Argentina
| | - Kelly Bordner
- Southern Connecticut State University, 501 Crescent St., New Haven, CT, 06515, United States
| | - Michael Nizhnikov
- Southern Connecticut State University, 501 Crescent St., New Haven, CT, 06515, United States
| |
Collapse
|
6
|
Gangisetty O, Mead EA, Sarkar DK. Sex-Determining Region Y Controls the Effects of Fetal Alcohol Exposure on Proopiomelanocortin Gene Expression. Front Neurosci 2021; 15:608102. [PMID: 33796006 PMCID: PMC8008069 DOI: 10.3389/fnins.2021.608102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 02/17/2021] [Indexed: 12/02/2022] Open
Abstract
Fetal alcohol exposure (FAE) causes various neurodevelopmental deficits in offspring, including reduced expression of the stress regulatory proopiomelanocortin (Pomc) gene and an elevated stress response for multiple generations via the male germline. Male germline-specific effects of FAE on the Pomc gene raises the question if the sex-determining region Y (SRY) may have a role in regulating Pomc gene expression. Using a transgenerational model of FAE in Fischer 344 rats, we determined the role of SRY in the regulation of the Pomc gene. FAEs, like on the Pomc gene, reduced Sry gene expression in sperm and the mediobasal hypothalamus (MBH) in male adult offspring. Fetal alcohol-induced inhibition of Sry gene expression was associated with increased Sry promoter DNA methylation. Additionally, fetal alcohol effects on the Sry gene persisted for three generations in the male germline but not in the female germline. Sry gene knockdown reduced the Pomc gene expression. Sry recruitment onto the Pomc promoter was found to be reduced in the hypothalamus of fetal alcohol-exposed rats compared to control rats. Pomc promoter luciferase activity was increased following Sry overexpression. A site-directed mutagenesis study revealed that SRY binding sites are required for POMC promoter transcription activity. Overall, these findings suggest that SRY plays a stimulatory role in the regulation of Pomc gene expression and may potentially contribute to the fetal alcohol-induced changes in the level of Pomc gene expression for multiple generations.
Collapse
Affiliation(s)
- Omkaram Gangisetty
- Rutgers Endocrine Research Program, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
| | - Edward A Mead
- Rutgers Endocrine Research Program, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
| | - Dipak K Sarkar
- Rutgers Endocrine Research Program, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
| |
Collapse
|
7
|
Toorie AM, Vassoler FM, Qu F, Schonhoff CM, Bradburn S, Murgatroyd CA, Slonim DK, Byrnes EM. A history of opioid exposure in females increases the risk of metabolic disorders in their future male offspring. Addict Biol 2021; 26:e12856. [PMID: 31782234 DOI: 10.1111/adb.12856] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/24/2019] [Accepted: 11/03/2019] [Indexed: 12/20/2022]
Abstract
Worldwide consumption of opioids remains at historic levels. Preclinical studies report intergenerational effects on the endogenous opioid system of future progeny following preconception morphine exposure. Given the role of endogenous opioids in energy homeostasis, such effects could impact metabolism in the next generation. Thus, we examined diet-induced modifications in F1 male progeny of morphine-exposed female rats (MORF1). When fed a high fat-sugar diet (FSD) for 6 weeks, MORF1 males display features of emerging metabolic syndrome; they consume more food, gain more weight, and develop fasting-induced hyperglycemia and hyperinsulinemia. In the hypothalamus, proteins involved in energy homeostasis are modified and RNA sequencing revealed down-regulation of genes associated with neuronal plasticity, coupled with up-regulation of genes associated with immune, inflammatory, and metabolic processes that are specific to FSD-maintained MORF1 males. Thus, limited preconception morphine exposure in female rats increases the risk of metabolic syndrome/type 2 diabetes in the next generation.
Collapse
Affiliation(s)
- Anika M. Toorie
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
- Department of Biology Rhode Island College North Providence Rhode Island
| | - Fair M. Vassoler
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
| | - Fangfang Qu
- Department of Computer Science Tufts University Medford Massachusetts
| | - Christopher M. Schonhoff
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
| | - Steven Bradburn
- Division of Biomedical Sciences Manchester Metropolitan University Manchester UK
| | | | - Donna K. Slonim
- Department of Computer Science Tufts University Medford Massachusetts
| | - Elizabeth M. Byrnes
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
| |
Collapse
|
8
|
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
|
9
|
Alcohol as an early life stressor: Epigenetics, metabolic, neuroendocrine and neurobehavioral implications. Neurosci Biobehav Rev 2020; 118:654-668. [PMID: 32976915 DOI: 10.1016/j.neubiorev.2020.08.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/18/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
Ethanol exposure during gestation is an early life stressor that profoundly dysregulates structure and functions of the embryonal nervous system, altering the cognitive and behavioral development. Such dysregulation is also achieved by epigenetic mechanisms, which, altering the chromatin structure, redraw the entire pattern of gene expression. In parallel, an oxidative stress response at the cellular level and a global upregulation of neuroendocrine stress response, regulated by the HPA axis, exist and persist in adulthood. This neurobehavioral framework matches those observed in other psychiatric diseases such as mood diseases, depression, autism; those early life stressing events, although probably triggered by specific and different epigenetic mechanisms, give rise to largely overlapping neurobehavioral phenotypes. An early diagnosis of prenatal alcohol exposure, using reliable markers of ethanol intake, together with a deeper understanding of the pathogenic mechanisms, some of them reversible by their nature, can offer a temporal "window" of intervention. Supplementing a mother's diet with protective and antioxidant substances in addition to supportive psychological therapies can protect newborns from being affected.
Collapse
|
10
|
Reynolds VL, Butler P, Abernathy MM, Aschenbrenner L, Best DD, Blank J, Crosby M, Custer L, Escobar PA, Kolaja K, Moggs J, Shuey D, Snyder C, Van Vleet T, Zhou J, Hart TK. Nonclinical safety assessment of epigenetic modulatory drugs: Current status and industry perspective. Regul Toxicol Pharmacol 2020; 117:104746. [PMID: 32911461 DOI: 10.1016/j.yrtph.2020.104746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/15/2020] [Accepted: 07/22/2020] [Indexed: 12/30/2022]
Abstract
Pharmaceutic products designed to perturb the function of epigenetic modulators have been approved by regulatory authorities for treatment of advanced cancer. While the predominant effort in epigenetic drug development continues to be in oncology, non-oncology indications are also garnering interest. A survey of pharmaceutical companies was conducted to assess the interest and concerns for developing small molecule direct epigenetic effectors (EEs) as medicines. Survey themes addressed (1) general levels of interest and activity with EEs as therapeutic agents, (2) potential safety concerns, and (3) possible future efforts to develop targeted strategies for nonclinical safety assessment of EEs. Thirteen companies contributed data to the survey. Overall, the survey data indicate the consensus opinion that existing ICH guidelines are effective and appropriate for nonclinical safety assessment activities with EEs. Attention in the framework of study design should, on a case by case basis, be considered for delayed or latent toxicities, carcinogenicity, reproductive toxicity, and the theoretical potential for transgenerational effects. While current guidelines have been appropriate for the nonclinical safety assessments of epigenetic targets, broader experience with a wide range of epigenetic targets will provide information to assess the potential need for new or revised risk assessment strategies for EE drugs.
Collapse
Affiliation(s)
| | | | | | | | - Derek D Best
- Lilly Research Laboratories, Indianapolis, IN, USA
| | - Jim Blank
- Takeda Pharmaceutical, Cambridge, MA, USA
| | - Meredith Crosby
- AbbVie Inc. Global Pharmaceutical Research and Development, Preclinical Safety, 1 North Waukegan Road, North Chicago, IL, USA
| | - Laura Custer
- Bristol-Myers Squibb, Drug Safety Evaluation, 1 Squibb Dr, New Brunswick, NJ, USA
| | | | - Kyle Kolaja
- Bristol-Myers Squibb, Drug Safety Evaluation, 1 Squibb Dr, New Brunswick, NJ, USA
| | - Jonathan Moggs
- Novartis Institutes for BioMedical Research, Translational Medicine, Basel, Switzerland
| | - Dana Shuey
- Incyte Corporation, 1801 Augustine Cutoff, Wilmington, DE, USA
| | | | - Terry Van Vleet
- AbbVie Inc. Global Pharmaceutical Research and Development, Preclinical Safety, 1 North Waukegan Road, North Chicago, IL, USA
| | - Junguo Zhou
- Nonclinical Safety, Janssen Research and Development LLC., Raritan, NJ, USA
| | - Timothy K Hart
- GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, PA, USA
| |
Collapse
|
11
|
Csaba G. Reprogramming of the Immune System by Stress and Faulty Hormonal Imprinting. Clin Ther 2020; 42:983-992. [PMID: 32307123 DOI: 10.1016/j.clinthera.2020.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Hormonal imprinting is taking place perinatally at the first encounter between the developing hormone receptors and their target hormones. However, in this crucial period when the developmental window for physiological imprinting is open, other molecules, such as synthetic hormones and endocrine disruptors can bind to the receptors, leading to faulty imprinting with life-long consequences, especially to the immune system. This review presents the factors of stress and faulty hormonal imprinting that lead to reprogramming of the immune system. METHODS Relevant publications from Pubmed since 1990 were reviewed and synthesized. FINDINGS The developing immune system is rather sensitive to hormonal effects. Faulty hormonal imprinting is able to reprogram the original developmental program present in a given cell, with lifelong consequences, manifested in alteration of hormone binding by receptors, susceptibility to certain (non-infectious) diseases, and triggering of other diseases. As stress mobilizes the hypothalamic-pituitary-adrenal axis if it occurred during gestation or perinatally, it could lead to faulty hormonal imprinting in the immune system, manifested later as allergic and autoimmune diseases or weakness of normal immune defenses. Hormonal imprinting is an epigenetic process and is carried to the offspring without alteration of DNA base sequences. This means that any form of early-life stress alone or in association with hormonal imprinting could be associated with the developmental origin of health and disease (DOHaD). As puberty is also a period of reprogramming, stress or faulty imprinting can change the original (developmental) program, also with life-long consequences. IMPLICATIONS Considering the continuous differentiation of immune cells (from blast-cells) during the whole life, there is a possibility of late-imprinting or stress-activated reprogramming in the immune system at any periods of life, with later pathogenetic consequences.
Collapse
Affiliation(s)
- György Csaba
- Department of Genetics, Cell, and Immunobiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
12
|
Dobs YE, Ali MM. The epigenetic modulation of alcohol/ethanol and cannabis exposure/co-exposure during different stages. Open Biol 2020; 9:180115. [PMID: 30958117 PMCID: PMC6367141 DOI: 10.1098/rsob.180115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Studies have reported the significant economic impact of smoking cannabis and drinking alcohol In the USA. It was estimated that the costs of cannabis-related treatment, hospitalization and loss of work-related pay have amounted to $200 billion. (Andersen AM, Dogan MV, Beach SRH, Philibert RA. 2015 Genes 6, 991-1022. ( doi:10.3390/genes6040991 )). Data from the National Epidemiologic Survey on Alcohol and Related Conditions showed that individuals with general anxiety disorder and substance use disorder (GAD-SUD) have higher psychiatric comorbidity rates than those without substance use disorder (Alegría AA, Hasin DS, Nunes EV, Liu SM, Davies C, Grant BF, Blanco C. 2010 J. Clin. Psychiatry 71, 1187-1195. ( doi:10.4088/JCP.09m05328gry )). Moreover, the criminal justice system is significantly impacted by this cost (Andersen AM, Dogan MV, Beach SRH, Philibert RA. 2015 Genes 6, 991-1022. ( doi:10.3390/genes6040991 )). Despite the increasing use of cannabis, there are still too many obscure facts. One of the new areas that scientific evidence shows is impacted negatively by cannabis use is the epigenome, which is an understudied area that we are still learning about. In addition, over the past few decades, we have seen various social and healthcare changes that have raised critical questions about their ongoing roles in regulating marijuana and alcohol use. This is important because of the increasing popularity and usage across various ages especially young adults and teenagers. More than 97.5 million Americans over 12 years old have used cannabis for non-medical use despite the significant side effects, with 1 in 10 users developing cannabis dependence (Crean RD, Crane NA, Mason BJ. 2011 J. Addict. Med. 5, 1-8. ( doi:10.1097/ADM.0b013e31820c23fa ), Office of Applied Studies. 2006 Substance Abuse and Mental Health Services Administration, USA.). It was reported that 16% of substance abuse admissions in the USA were for cannabis-related symptoms, which is second only to alcohol-related disorders (Agalioti T, Lomvardas S, Parekh B, Yie J, Maniatis T, Thanos D. 2000 Cell 103, 667-678. ( doi:10.1016/S0092-8674(00)00169-0 ), Soutoglou E, Talianidis I. 2002 Science 295, 1901-1904. ( doi:10.1126/science.1068356 )). Today there are thirty-one states and the District of Columbia that currently have legalized marijuana for either medical or recreational use. Data about marijuana use from NIAAA's National Epidemiologic Survey on Alcohol and Related Conditions (NESARC) indicates that 'in total, 79 000 people were interviewed on alcohol and drug use. When examined by age young adults (ages 18-21) were found to be at highest risk for marijuana use and marijuana use disorder, with use increasing from 10.5 to 21.2% and disorder increasing from 4.4 to 7.5%'. 'Given these facts, George Koob, PhD, director of NIAAA stated the importance for the scientific community to convey this information to the public about the potential hazards of marijuana and it's use'. On the other hand, according to the National Institute on Alcohol Abuse and Alcoholism, 16 million adults suffer from alcohol use disorders. To the best of our knowledge, epigenetic mechanisms have been previously studied in alcohol and cannabis abuse separately. Recent studies highlighted the molecular mechanisms that are linked with drug-induced transcriptional regulation, behavioural abnormalities and neurodegeneration, which has emphasized the role of chromatin modification/remodelling in the generation of drug activation of certain genes and the disabling of others, and the effect of that on addiction (Maze I, Nestler EJ. 2011 Ann. N. Y. Acad. Sci. 1216, 99-113. ( doi:10.1111/j.1749-6632.2010.05893.x ); Renthal W, Nestler EJ. 2008 Trends Mol. Med. 14, 341-350. ( doi:10.1016/j.molmed.2008.06.004 )). In this review, we will give an overview of epigenome science relevant to cannabis/the endocannabinoid system and the potential of epigenetic overlap between alcohol and cannabinergic activity at different stages, to aid further investigations that could bring more treatment options to our horizon.
Collapse
Affiliation(s)
- Yasminah Elsaadany Dobs
- 1 Department of Biology and Biomedical Science, North Carolina Central University , Durham, NC , USA
| | - Mohamed Medhat Ali
- 2 Biomedical Sciences Program, Zewail City of Science and Technology , Giza , Egypt.,3 Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University , Egypt
| |
Collapse
|
13
|
Granato A. The Transgenerational Consequences of the Interaction Between Humans and Molecules: Alcohol as a Cultural Artifact. Front Psychol 2020; 11:61. [PMID: 32063877 PMCID: PMC7000371 DOI: 10.3389/fpsyg.2020.00061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/09/2020] [Indexed: 01/19/2023] Open
Affiliation(s)
- Alberto Granato
- Department of Psychology, Catholic University of the Sacred Heart, Milan, Italy
| |
Collapse
|
14
|
Epigenetic regulation of POMC; implications for nutritional programming, obesity and metabolic disease. Front Neuroendocrinol 2019; 54:100773. [PMID: 31344387 DOI: 10.1016/j.yfrne.2019.100773] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 01/07/2023]
Abstract
Proopiomelanocortin (POMC) is a key mediator of satiety. Epigenetic marks such as DNA methylation may modulate POMC expression and provide a biological link between early life exposures and later phenotype. Animal studies suggest epigenetic marks at POMC are influenced by maternal energy excess and restriction, prenatal stress and Triclosan exposure. Postnatal factors including energy excess, folate, vitamin A, conjugated linoleic acid and leptin may also affect POMC methylation. Recent human studies suggest POMC DNA methylation is influenced by maternal nutrition in early pregnancy and associated with childhood and adult obesity. Studies in children propose a link between POMC DNA methylation and elevated lipids and insulin, independent of body habitus. This review brings together evidence from animal and human studies and suggests that POMC is sensitive to nutritional programming and is associated with a wide range of weight-related and metabolic outcomes.
Collapse
|
15
|
Escher J, Robotti S. Pregnancy drugs, fetal germline epigenome, and risks for next-generation pathology: A call to action. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2019; 60:445-454. [PMID: 30891817 DOI: 10.1002/em.22288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/09/2019] [Accepted: 03/16/2019] [Indexed: 06/09/2023]
Abstract
Drugs taken during pregnancy can affect three generations at once: the gestating woman (F0), her exposed fetus (F1), and the fetal germ cells that confer heritable information for the grandchildren (F2). Unfortunately, despite growing evidence for connections between F0 drug exposures and F2 pathology, current approaches to risk assessment overlook this important dimension of risk. In this commentary, we argue that the unique molecular vulnerabilities of the fetal germline, particularly with regard to global epigenomic reprogramming, combined with empirical evidence for F2 effects of F1 in utero drug and other exposures, should change the way we consider potential long-term consequences of pregnancy drugs and alter toxicology's standard somatic paradigm. Specifically, we (1) suggest that pregnancy drugs common in the postwar decades should be investigated as potential contributors to the "missing heritability" of many pathologies now surging in prevalence; (2) call for inclusion of fetal germline risks in pregnancy drug safety assessment; and (3) highlight the need for intensified research to ascertain generational impacts of diethylstilbestrol, a vanguard question of human germline toxicity. Only by fully addressing this important dimension of transplacental exposure can we responsibly evaluate safety of drug exposures during pregnancy and convey the full scope of risks, while also retrospectively comprehending the generational legacy of recent history's unprecedented glut of evolutionarily novel intrauterine exposures. Environ. Mol. Mutagen. 60:445-454, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jill Escher
- Escher Fund for Autism, San Jose, California
| | | |
Collapse
|
16
|
Hollander J, McNivens M, Pautassi RM, Nizhnikov ME. Offspring of male rats exposed to binge alcohol exhibit heightened ethanol intake at infancy and alterations in T-maze performance. Alcohol 2019; 76:65-71. [PMID: 30583252 DOI: 10.1016/j.alcohol.2018.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/28/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Abstract
Alcohol use is associated with a variety of negative consequences, including heightened likelihood of cognitive impairment, proclivity to alcohol use disorders (AUD), and alterations in the drinker's offspring. Children and rodents exposed to alcohol during pregnancy, or those whose fathers consumed alcohol prior to mating, often exhibit neurodevelopmental, physiological, and behavioral deficits. The present study assessed cognitive function and alcohol intake in male and female rats that were offspring of alcohol-exposed fathers. Adult male rats were exposed to alcohol or vehicle (0.0 or 2.0 g/kg, respectively; twice daily for 2 days followed by a rest day, for a total of eight alcohol or vehicle exposure days), or were left untreated and then mated with non-manipulated females. The offspring were assessed for alcohol intake, via intraoral infusion, followed by cognitive assessment via an alternating T-maze task. The results indicated that paternal ethanol exposure, prior to breeding, resulted in offspring that consumed significantly more ethanol than vehicle or untreated controls. Furthermore, the offspring of alcohol-exposed fathers exhibited a significant failure to initiate and complete the T-maze performance tests. Although, when they did engage in the tests they performed at the level of controls (i.e., 80% correct). The present results add to a growing body of literature suggesting that paternal pre-conception alcohol exposure can have deleterious effects on the offspring.
Collapse
|
17
|
Ehrhart F, Roozen S, Verbeek J, Koek G, Kok G, van Kranen H, Evelo CT, Curfs LMG. Review and gap analysis: molecular pathways leading to fetal alcohol spectrum disorders. Mol Psychiatry 2019; 24:10-17. [PMID: 29892052 PMCID: PMC6325721 DOI: 10.1038/s41380-018-0095-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/17/2017] [Accepted: 04/23/2018] [Indexed: 12/30/2022]
Abstract
Alcohol exposure during pregnancy affects the development of the fetus in various ways and may lead to Fetal Alcohol Spectrum Disorders (FASD). FASD is one of the leading preventable forms of neurodevelopmental disorders. In the light of prevention and early intervention, knowledge on how ethanol exposure induces fetal damage is urgently needed. Besides direct ethanol and acetaldehyde toxicity, alcohol increases oxidative stress, and subsequent general effects (e.g., epigenetic imprinting, gene expression, and metabolite levels). The current review provides an overview of the existing knowledge about specific downstream pathways for FASD that affects e.g., the SHH pathway, cholesterol homeostasis, neurotransmitter signaling, and effects on the cytoskeleton. Available human data vary greatly, while animal studies with controlled ethanol exposition are only to a certain limit transferable to humans. The main deficits in knowledge about FASD are the lack of pathophysiological understanding and dose-response relationships, together with the lack of reliable biomarkers for either FASD detection or estimation of susceptibility. In addition to single outcome experiments, omics data should be generated to overcome this problem. Therefore, for future studies we recommend holistic data driven analysis, which allows integrative analyses over multiple levels of genetic variation, transcriptomics and metabolomics data to investigate the whole image of FASD development and to provide insight in potential drug targets for intervention.
Collapse
Affiliation(s)
- Friederike Ehrhart
- Governor Kremers Centre, Maastricht University Medical Centre+, Maastricht, The Netherlands. .,Department of Bioinformatics, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.
| | - Sylvia Roozen
- 0000 0004 0480 1382grid.412966.eGovernor Kremers Centre, Maastricht University Medical Centre+, Maastricht, The Netherlands ,0000 0001 0481 6099grid.5012.6Department of Work and Social Psychology, Maastricht University, Maastricht, The Netherlands
| | - Jef Verbeek
- 0000 0004 0480 1382grid.412966.eDepartment of Internal Medicine, Division of gastroenterology and hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ger Koek
- 0000 0004 0480 1382grid.412966.eGovernor Kremers Centre, Maastricht University Medical Centre+, Maastricht, The Netherlands ,0000 0004 0480 1382grid.412966.eDepartment of Internal Medicine, Division of gastroenterology and hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Gerjo Kok
- 0000 0004 0480 1382grid.412966.eGovernor Kremers Centre, Maastricht University Medical Centre+, Maastricht, The Netherlands ,0000 0001 0481 6099grid.5012.6Department of Work and Social Psychology, Maastricht University, Maastricht, The Netherlands
| | - Henk van Kranen
- 0000 0004 0480 1382grid.412966.eGovernor Kremers Centre, Maastricht University Medical Centre+, Maastricht, The Netherlands ,0000 0001 0481 6099grid.5012.6Institute for Public Health Genomics, Maastricht University, Maastricht, The Netherlands
| | - Chris T. Evelo
- 0000 0004 0480 1382grid.412966.eGovernor Kremers Centre, Maastricht University Medical Centre+, Maastricht, The Netherlands ,0000 0001 0481 6099grid.5012.6Department of Bioinformatics, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Leopold M. G. Curfs
- 0000 0004 0480 1382grid.412966.eGovernor Kremers Centre, Maastricht University Medical Centre+, Maastricht, The Netherlands ,0000 0004 0480 1382grid.412966.eDepartment of Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
18
|
Yuan B, Zhang H, Wang X, Pan Y, Jiang J. Effect of Nano-SiO 2 on Expression and Aberrant Methylation of Imprinted Genes in Lung and Testis. NANOSCALE RESEARCH LETTERS 2018; 13:266. [PMID: 30182198 PMCID: PMC6123335 DOI: 10.1186/s11671-018-2673-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 08/20/2018] [Indexed: 05/11/2023]
Abstract
Nanotechnology has been developing rapidly and is now used in many cutting-edge medical therapeutics. However, there is increasing concern that exposure to nanoparticles (NPs) may induce different systemic diseases as epigenetic mechanisms are associated with more and more disease. The role of NP epigenomic modification is important to disease etiology. Our study aimed to determine the epigenetic mechanisms of damage in lung and testis cells by exposing cells to SiO2 NPs. We used male C57BL/6 mice to characterize the damaging effect of SiO2 NPs on lung and testis cells as well as the resulting methylation state at the imprinted Dlk1/Dio3 domain region. The A549 cells exposed to SiO2 NPs had cell apoptosis, and male mice exposed to SiO2 NPs had altered lung and testis tissues. The genes in the imprinted domains Dlk1/Dio3 region changed in both tissues; Dlk1, Rtl1, and Dio3 are upregulated in testis while Dlk1 and Dio3 are also upregulated in lung tissues. Bisulfite sequencing PCR of male adult lung and testis were mostly hypomethylated, with a few hypermethylated CpGs. These findings indicate that nanoparticles play an important role in DNA methylation of imprinted genes.
Collapse
Affiliation(s)
- Beilei Yuan
- College of Safety Science and Engineering, Nanjing Tech University, 200 Zhongshan North Rd, Nanjing, 210009, China.
| | - Huazhong Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuan Wang
- College of Safety Science and Engineering, Nanjing Tech University, 200 Zhongshan North Rd, Nanjing, 210009, China
| | - Yong Pan
- College of Safety Science and Engineering, Nanjing Tech University, 200 Zhongshan North Rd, Nanjing, 210009, China.
| | - Juncheng Jiang
- College of Safety Science and Engineering, Nanjing Tech University, 200 Zhongshan North Rd, Nanjing, 210009, China.
| |
Collapse
|
19
|
Jabbar S, Reuhl K, Sarkar DK. Prenatal alcohol exposure increases the susceptibility to develop aggressive prolactinomas in the pituitary gland. Sci Rep 2018; 8:7720. [PMID: 29769550 PMCID: PMC5955957 DOI: 10.1038/s41598-018-25785-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/12/2018] [Indexed: 02/07/2023] Open
Abstract
Excess alcohol use is known to promote development of aggressive tumors in various tissues in human patients, but the cause of alcohol promotion of tumor aggressiveness is not clearly understood. We used an animals model of fetal alcohol exposure that is known to promote tumor development and determined if alcohol programs the pituitary to acquire aggressive prolactin-secreting tumors. Our results show that pituitaries of fetal alcohol-exposed rats produced increased levels of intra-pituitary aromatase protein and plasma estrogen, enhanced pituitary tissue growth, and upon estrogen challenge developed prolactin-secreting tumors (prolactinomas) that were hemorrhagic and often penetrated into the surrounding tissue. Pituitary tumors of fetal alcohol-exposed rats produced higher levels of hemorrhage-associated genes and proteins and multipotency genes and proteins. Cells of pituitary tumor of fetal alcohol exposed rat grew into tumor spheres in ultra-low attachment plate, expressed multipotency genes, formed an increased number of colonies, showed enhanced cell migration, and induced solid tumors following inoculation in immunodeficient mice. These data suggest that fetal alcohol exposure programs the pituitary to develop aggressive prolactinoma after estrogen treatment possibly due to increase in stem cell niche within the tumor microenvironment.
Collapse
Affiliation(s)
- Shaima Jabbar
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Lane, New Brunswick, NJ, 08901, USA.,Endocrinology and Animal Biosciences Graduate Program, Rutgers, The State University of New Jersey, 84 Lipman Drive, New Brunswick, NJ, 08901, USA
| | - Kenneth Reuhl
- The Environmental and Occupational Health Sciences Institute, Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 140, Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Dipak K Sarkar
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Lane, New Brunswick, NJ, 08901, USA. .,The Environmental and Occupational Health Sciences Institute, Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 140, Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
20
|
Kou H, Shen L, Luo HW, Chen LB, Wu DF, Wang H. An intergenerational effect of neuroendocrine metabolic programming alteration induced by prenatal ethanol exposure in rats. Reprod Toxicol 2017; 74:85-93. [DOI: 10.1016/j.reprotox.2017.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 08/30/2017] [Accepted: 09/07/2017] [Indexed: 11/17/2022]
|
21
|
Imam MU, Ismail M. The Impact of Traditional Food and Lifestyle Behavior on Epigenetic Burden of Chronic Disease. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1700043. [PMID: 31565292 PMCID: PMC6607231 DOI: 10.1002/gch2.201700043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/12/2017] [Indexed: 05/11/2023]
Abstract
Noncommunicable chronic diseases (NCCDs) are the leading causes of morbidity and mortality globally. The mismatch between present day diets and ancestral genome is suggested to contribute to the NCCDs burden, which is promoted by traditional risk factors like unhealthy diets, physical inactivity, alcohol and tobacco. However, epigenetic evidence now suggests that cumulatively inherited epigenetic modifications may have made humans more prone to the effects of present day lifestyle factors. Perinatal starvation was widespread in the 19th century. This together with more recent events like increasing consumption of western and low fiber diets, smoking, harmful use of alcohol, physical inactivity, and environmental pollutants may have programed the human epigenome for higher NCCDs risk. In this review, on the basis of available epigenetic data it is hypothesized that transgenerational effects of lifestyle factors may be contributing to the current global burden of NCCDs. Thus, there is a need to reconsider prevention strategies so that the subsequent generations will not have to pay for our sins and those of our ancestors.
Collapse
Affiliation(s)
- Mustapha U. Imam
- Precision Nutrition Innovation InstituteCollege of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Maznah Ismail
- Laboratory of Molecular BiomedicineInstitute of BioscienceUniversiti Putra MalaysiaSerdangSelangor43400Malaysia
| |
Collapse
|
22
|
Ornellas F, Carapeto PV, Mandarim‐de‐Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2017. [DOI: 10.1016/j.jpedp.2017.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
23
|
Ornellas F, Carapeto PV, Mandarim-de-Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. J Pediatr (Rio J) 2017; 93:551-559. [PMID: 28822233 DOI: 10.1016/j.jped.2017.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children. SOURCES A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication. SUMMARY OF FINDINGS Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA. CONCLUSIONS Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Priscila V Carapeto
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil.
| | - Marcia B Aguila
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
24
|
Li P, Becker JB, Heitzeg MM, McClellan ML, Reed BG, Zucker RA. Gender differences in the transmission of risk for antisocial behavior problems across generations. PLoS One 2017; 12:e0177288. [PMID: 28505162 PMCID: PMC5432185 DOI: 10.1371/journal.pone.0177288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 04/25/2017] [Indexed: 12/11/2022] Open
Abstract
Previous studies have shown that children of alcohol use disorder (AUD) parents are more likely to develop alcohol problems as well as antisocial and other behavior problems. The purpose of this study was to examine gender discordance in the effect of early maternal and paternal influences on antisocial behaviors of boys and girls, as well as the environmental factors that moderate the parental effects. Specifically, we examined the effects of childhood and adulthood antisocial behavior of the parents on offspring antisocial behavior as young adults. We also examined whether mothers' and fathers' drinking problems when offspring were young children (6-8 years) affected offspring antisocial behavior as young adults (18-21 years). We evaluated 655 children from 339 families in the Michigan Longitudinal Study (MLS), a prospective study of AUD and non-AUD families. Path models were constructed in order to test for the parental contributions to offspring outcomes. We found that both mothers' and fathers' antisocial behavior contributed to the children's young adult antisocial behavior. Only mothers' drinking problems while their children were little had a significant effect on their sons' later drinking, but not on their daughters'. These different parental effects suggest that maternal and paternal influences may be mediated by different mechanisms.
Collapse
Affiliation(s)
- Pin Li
- School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jill B. Becker
- Department of Psychology and the Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mary M. Heitzeg
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michele L. McClellan
- Department of History and the Residential College, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Beth Glover Reed
- School of Social Work and the Department of Women’s Studies, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Robert A. Zucker
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
25
|
Chastain LG, Sarkar DK. Alcohol effects on the epigenome in the germline: Role in the inheritance of alcohol-related pathology. Alcohol 2017; 60:53-66. [PMID: 28431793 DOI: 10.1016/j.alcohol.2016.12.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/04/2016] [Accepted: 12/30/2016] [Indexed: 12/18/2022]
Abstract
Excessive alcohol exposure has severe health consequences, and clinical and animal studies have demonstrated that disruptions in the epigenome of somatic cells, such as those in brain, are an important factor in the development of alcohol-related pathologies, such as alcohol-use disorders (AUDs) and fetal alcohol spectrum disorders (FASDs). It is also well known that alcohol-related health problems are passed down across generations in human populations, but the complete mechanisms for this phenomenon are currently unknown. Recent studies in animal models have suggested that epigenetic factors are also responsible for the transmission of alcohol-related pathologies across generations. Alcohol exposure has been shown to induce changes in the epigenome of sperm of exposed male animals, and these epimutations are inherited in the offspring. This paper reviews evidence for multigenerational and transgenerational epigenetic inheritance of alcohol-related pathology through the germline. We also review the literature on the epigenetic effects of alcohol exposure on somatic cells in brain, and its contribution to AUDs and FASDs. We note gaps in knowledge in this field, such as the lack of clinical studies in human populations and the lack of data on epigenetic inheritance via the female germline, and we suggest future research directions.
Collapse
Affiliation(s)
- Lucy G Chastain
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Lane, New Brunswick, NJ 08901, USA
| | - Dipak K Sarkar
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Lane, New Brunswick, NJ 08901, USA.
| |
Collapse
|
26
|
Laufer BI, Chater-Diehl EJ, Kapalanga J, Singh SM. Long-term alterations to DNA methylation as a biomarker of prenatal alcohol exposure: From mouse models to human children with fetal alcohol spectrum disorders. Alcohol 2017; 60:67-75. [PMID: 28187949 DOI: 10.1016/j.alcohol.2016.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/19/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022]
Abstract
Rodent models of Fetal Alcohol Spectrum Disorders (FASD) have revealed that prenatal alcohol exposure (PAE) results in differential DNA cytosine methylation in the developing brain. The resulting genome-wide methylation changes are enriched in genes with neurodevelopmental functions. The profile of differential methylation is dynamic and present in some form for life. The methylation changes are transmitted across subsequent mitotic divisions, where they are maintained and further modified over time. More recent follow up has identified a profile of the differential methylation in the buccal swabs of young children born with FASD. While distinct from the profile observed in brain tissue from rodent models, there are similarities. These include changes in genes belonging to a number of neurodevelopmental and behavioral pathways. Specifically, there is increased methylation at the clustered protocadherin genes and deregulation of genomically imprinted genes, even though no single gene is affected in all patients studied to date. These novel results suggest further development of a methylation based strategy could enable early and accurate diagnostics and therapeutics, which have remained a challenge in FASD research. There are two aspects of this challenge that must be addressed in the immediate future: First, the long-term differential methylomics observed in rodent models must be functionally confirmed. Second, the similarities in differential methylation must be further established in humans at a methylomic level and overcome a number of technical limitations. While a cure for FASD is challenging, there is an opportunity for the development of early diagnostics and attenuations towards a higher quality of life.
Collapse
|
27
|
Pandey SC, Kyzar EJ, Zhang H. Epigenetic basis of the dark side of alcohol addiction. Neuropharmacology 2017; 122:74-84. [PMID: 28174112 DOI: 10.1016/j.neuropharm.2017.02.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
Abstract
Alcoholism is a complex brain disease characterized by three distinct stages of the addiction cycle that manifest as neuroadaptive changes in the brain. One such stage of the addiction cycle is alcohol withdrawal and the negative affective states that promote drinking and maintain addiction. Repeated alcohol use, genetic predisposition to alcoholism and anxiety, and alcohol exposure during crucial developmental periods all contribute to the development of alcohol-induced withdrawal and negative affective symptoms. Epigenetic modifications within the amygdala have provided a molecular basis of these negative affective symptoms, also known as the dark side of addiction. Here, we propose that allostatic change within the epigenome in the amygdala is a prime mechanism of the biological basis of negative affective states resulting from, and contributing to, alcoholism. Acute alcohol exposure produces an anxiolytic response which is associated with the opening of chromatin due to increased histone acetylation, increased CREB binding protein (CBP) levels, and histone deacetylase (HDAC) inhibition. After chronic ethanol exposure, these changes return to baseline along with anxiety-like behaviors. However, during withdrawal, histone acetylation decreases due to increased HDAC activity and decreased CBP levels in the amygdala circuitry leading to the development of anxiety-like behaviors. Additionally, innately higher expression of the HDAC2 isoform leads to a deficit in global and gene-specific histone acetylation in the amygdala that is associated with a decrease in the expression of several synaptic plasticity-associated genes and maintaining heightened anxiety-like behavior and excessive alcohol intake. Adolescent alcohol exposure also leads to higher expression of HDAC2 and a deficit in histone acetylation leading to decreased expression of synaptic plasticity-associated genes and high anxiety and drinking behavior in adulthood. All these studies indicate that the epigenome can undergo allostatic reprogramming in the amygdaloid circuitry during various stages of alcohol exposure. Furthermore, opening the chromatin by inhibiting HDACs using pharmacological or genetic manipulations can lead to the attenuation of anxiety as well as alcohol intake. Chromatin remodeling provides a clear biological basis for the negative affective states seen during alcohol addiction and presents opportunities for novel drug development and treatment options. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA.
| | - Evan J Kyzar
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA
| |
Collapse
|
28
|
Doehner W, Praße L, Wolpers J, Brückner MK, Ueberham U, Arendt T. Transgenerational transmission of an anticholinergic endophenotype with memory dysfunction. Neurobiol Aging 2016; 51:19-30. [PMID: 28033505 DOI: 10.1016/j.neurobiolaging.2016.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/21/2016] [Accepted: 11/26/2016] [Indexed: 10/20/2022]
Abstract
Impaired cholinergic neurotransmission associated with cognitive dysfunction occurs in various mental disorders of different etiologies including Alzheimer's disease and postalcoholic dementia and others. To address the question whether there exists a common endophenotype with a defined genetic and/or epigenetic signature causing mental dysfunction in these disorders, we investigated 2 generations of offspring born to alcohol-treated mothers. Here, we show that memory impairment and reduced synthesis of acetylcholine occurs in both F1 (exposed to ethanol in utero) and F2 generation (never been exposed to ethanol). Effects in the F2 generation are most likely consequences of transgenerationally transmitted epigenetic modifications in stem cells induced by alcohol. This clearly documents the role of ancestral history of drug abuse on the brain development of subsequent generations. The results further suggest an epigenetic trait for an anticholinergic endophenotype associated with cognitive dysfunction which might be relevant to our understanding of mental impairment in neurodegenerative disorders such as Alzheimer's disease and related disorders.
Collapse
Affiliation(s)
- Wolfram Doehner
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - Lieselotte Praße
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - James Wolpers
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - Martina K Brückner
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - Uwe Ueberham
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - Thomas Arendt
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany.
| |
Collapse
|
29
|
Crews FT, Vetreno RP, Broadwater MA, Robinson DL. Adolescent Alcohol Exposure Persistently Impacts Adult Neurobiology and Behavior. Pharmacol Rev 2016; 68:1074-1109. [PMID: 27677720 PMCID: PMC5050442 DOI: 10.1124/pr.115.012138] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Adolescence is a developmental period when physical and cognitive abilities are optimized, when social skills are consolidated, and when sexuality, adolescent behaviors, and frontal cortical functions mature to adult levels. Adolescents also have unique responses to alcohol compared with adults, being less sensitive to ethanol sedative-motor responses that most likely contribute to binge drinking and blackouts. Population studies find that an early age of drinking onset correlates with increased lifetime risks for the development of alcohol dependence, violence, and injuries. Brain synapses, myelination, and neural circuits mature in adolescence to adult levels in parallel with increased reflection on the consequence of actions and reduced impulsivity and thrill seeking. Alcohol binge drinking could alter human development, but variations in genetics, peer groups, family structure, early life experiences, and the emergence of psychopathology in humans confound studies. As adolescence is common to mammalian species, preclinical models of binge drinking provide insight into the direct impact of alcohol on adolescent development. This review relates human findings to basic science studies, particularly the preclinical studies of the Neurobiology of Adolescent Drinking in Adulthood (NADIA) Consortium. These studies focus on persistent adult changes in neurobiology and behavior following adolescent intermittent ethanol (AIE), a model of underage drinking. NADIA studies and others find that AIE results in the following: increases in adult alcohol drinking, disinhibition, and social anxiety; altered adult synapses, cognition, and sleep; reduced adult neurogenesis, cholinergic, and serotonergic neurons; and increased neuroimmune gene expression and epigenetic modifiers of gene expression. Many of these effects are specific to adolescents and not found in parallel adult studies. AIE can cause a persistence of adolescent-like synaptic physiology, behavior, and sensitivity to alcohol into adulthood. Together, these findings support the hypothesis that adolescent binge drinking leads to long-lasting changes in the adult brain that increase risks of adult psychopathology, particularly for alcohol dependence.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Margaret A Broadwater
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Donita L Robinson
- Bowles Center for Alcohol Studies (F.T.C., R.P.V., M.A.B., D.L.R.), Department of Psychiatry (F.T.C., D.L.R.), and Department of Pharmacology (F.T.C.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
30
|
Agabio R, Campesi I, Pisanu C, Gessa GL, Franconi F. Sex differences in substance use disorders: focus on side effects. Addict Biol 2016; 21:1030-42. [PMID: 27001402 DOI: 10.1111/adb.12395] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 12/19/2022]
Abstract
Although sex differences in several aspects of substance use disorders (SUDs) have been identified, less is known about the importance of possible sex differences in side effects induced by substances of abuse or by medications used to treat SUDs. In the SUD field, the perception of certain subjective effects are actively sought, while all other manifestations might operationally be considered side effects. This article was aimed at reviewing sex differences in side effects induced by alcohol, nicotine, heroin, marijuana and cocaine and by medications approved for alcohol, nicotine and heroin use disorders. A large body of evidence suggests that women are at higher risk of alcohol-induced injury, liver disease, cardiomyopathy, myopathy, brain damages and mortality. The risk of tobacco-induced coronary heart disease, lung disease and health problems is higher for women than for men. Women also experience greater exposure to side effects induced by heroin, marijuana and cocaine. In addition, women appear to be more vulnerable to the side effects induced by medications used to treat SUDs. Patients with SUDs should be advised that the risk of developing health problems may be higher for women than for men after consumption of the same amount of substances of abuse. Doses of medications for SUD women should be adjusted at least according to body weight. The sex differences observed also indicate an urgent need to recruit adequate numbers of female subjects in pre-clinical and clinical studies to improve our knowledge about SUDs in women.
Collapse
Affiliation(s)
- Roberta Agabio
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Cagliari Italy
| | - Ilaria Campesi
- National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems; Osilo, Sassari Italy
- Department of Biomedical Sciences; University of Sassari; Sassari Italy
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Cagliari Italy
| | - Gian Luigi Gessa
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Cagliari Italy
- Neuroscience Institute, Section of Cagliari; National Research Council of Italy; Cagliari Italy
| | - Flavia Franconi
- National Laboratory of Gender Medicine of the National Institute of Biostructures and Biosystems; Osilo, Sassari Italy
- Department of Biomedical Sciences; University of Sassari; Sassari Italy
- Assessorato alle Politiche per la Persona; Regione Basilicata Italy
| |
Collapse
|
31
|
Length of paternal lifespan is manifested in the DNA methylome of their nonagenarian progeny. Oncotarget 2015; 6:30557-67. [PMID: 26436701 PMCID: PMC4741551 DOI: 10.18632/oncotarget.5905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 08/27/2015] [Indexed: 12/19/2022] Open
Abstract
The heritability of lifespan is 20-30%, but only a few genes associated with longevity have been identified. To explain this discrepancy, the inheritance of epigenetic features, such as DNA methylation, have been proposed to contribute to the heritability of lifespan.We investigated whether parental lifespan is associated with DNA methylation profile in nonagenarians. A regression model, adjusted for differences in blood cell proportions, identified 659 CpG sites where the level of methylation was associated with paternal lifespan. However, no association was observed between maternal lifespan and DNA methylation. The 659 CpG sites associated with paternal lifespan were enriched outside of CpG islands and were located in genes associated with development and morphogenesis, as well as cell signaling. The largest difference in the level of methylation between the progeny of the shortest-lived and longest-lived fathers was identified for CpG sites mapping to CXXC5. In addition, the level of methylation in three Notch-genes (NOTCH1, NOTCH3 and NOTCH4) was also associated with paternal lifespan.There are implications for the inheritance of acquired traits via epigenetic mechanisms in mammals. Here we describe DNA methylation features that are associated with paternal lifespan, and we speculate that the identified CpG sites may represent intergenerational epigenetic inheritance.
Collapse
|
32
|
Abstract
Genome-wide association studies of complex physiological traits and diseases consistently found that associated genetic factors, such as allelic polymorphisms or DNA mutations, only explained a minority of the expected heritable fraction. This discrepancy is known as “missing heritability”, and its underlying factors and molecular mechanisms are not established. Epigenetic programs may account for a significant fraction of the “missing heritability.” Epigenetic modifications, such as DNA methylation and chromatin assembly states, reflect the high plasticity of the genome and contribute to stably alter gene expression without modifying genomic DNA sequences. Consistent components of complex traits, such as those linked to human stature/height, fertility, and food metabolism or to hereditary defects, have been shown to respond to environmental or nutritional condition and to be epigenetically inherited. The knowledge acquired from epigenetic genome reprogramming during development, stem cell differentiation/de-differentiation, and model organisms is today shedding light on the mechanisms of (a) mitotic inheritance of epigenetic traits from cell to cell, (b) meiotic epigenetic inheritance from generation to generation, and (c) true transgenerational inheritance. Such mechanisms have been shown to include incomplete erasure of DNA methylation, parental effects, transmission of distinct RNA types (mRNA, non-coding RNA, miRNA, siRNA, piRNA), and persistence of subsets of histone marks.
Collapse
Affiliation(s)
- Marco Trerotola
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy.
| | - Valeria Relli
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy.
| | - Pasquale Simeone
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy.
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy. .,Department of Neuroscience, Imaging and Clinical Sciences, Unit of Physiology and Physiopathology, 'G. d'Annunzio' University, Chieti, Italy.
| |
Collapse
|