1
|
Mutchler AL, Haynes AP, Saleem M, Jamison S, Khan MM, Ertuglu L, Kirabo A. Epigenetic Regulation of Innate and Adaptive Immune Cells in Salt-Sensitive Hypertension. Circ Res 2025; 136:232-254. [PMID: 39819017 DOI: 10.1161/circresaha.124.325439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Access to excess dietary sodium has heightened the risk of cardiovascular diseases, particularly affecting individuals with salt sensitivity of blood pressure. Our research indicates that innate antigen-presenting immune cells contribute to rapid blood pressure increases in response to excess sodium intake. Emerging evidence suggests that epigenetic reprogramming, with subsequent transcriptional and metabolic changes, of innate immune cells allows these cells to have a sustained response to repetitive stimuli. Epigenetic mechanisms also steer T-cell differentiation in response to innate immune signaling. Immune cells respond to environmental and nutritional cues, such as salt, promoting epigenetic regulation changes. This article aims to identify and discuss the role of epigenetic mechanisms in the immune system contributing to salt-sensitive hypertension.
Collapse
Affiliation(s)
- Ashley L Mutchler
- Department of Biomedical Sciences, Meharry Medical College, Nashville TN (A.L.M)
| | - Alexandria Porcia Haynes
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (A.P.H., M.S., M.M.K., A.K.)
| | - Mohammad Saleem
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (A.P.H., M.S., M.M.K., A.K.)
| | | | - Mohd Mabood Khan
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (A.P.H., M.S., M.M.K., A.K.)
| | - Lale Ertuglu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (L.E.)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (A.P.H., M.S., M.M.K., A.K.)
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN (A.K.)
- Vanderbilt Center for Immunobiology, Nashville, TN (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN (A.K.)
- Vanderbilt Institute for Global Health, Nashville, TN (A.K.)
| |
Collapse
|
2
|
Khatib H, Townsend J, Konkel MA, Conidi G, Hasselkus JA. Calling the question: what is mammalian transgenerational epigenetic inheritance? Epigenetics 2024; 19:2333586. [PMID: 38525788 DOI: 10.1080/15592294.2024.2333586] [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: 12/18/2023] [Accepted: 03/17/2024] [Indexed: 03/26/2024] Open
Abstract
While transgenerational epigenetic inheritance has been extensively documented in plants, nematodes, and fruit flies, its existence in mammals remains controversial. Several factors have contributed to this debate, including the lack of a clear distinction between intergenerational and transgenerational epigenetic inheritance (TEI), the inconsistency of some studies, the potential confounding effects of in-utero vs. epigenetic factors, and, most importantly, the biological challenge of epigenetic reprogramming. Two waves of epigenetic reprogramming occur: in the primordial germ cells and the developing embryo after fertilization, characterized by global erasure of DNA methylation and remodelling of histone modifications. Consequently, TEI can only occur if specific genetic regions evade this reprogramming and persist through embryonic development. These challenges have revived the long-standing debate about the possibility of inheriting acquired traits, which has been strongly contested since the Lamarckian and Darwinian eras. As a result, coupled with the absence of universally accepted criteria for transgenerational epigenetic studies, a vast body of literature has emerged claiming evidence of TEI. Therefore, the goal of this study is to advocate for establishing fundamental criteria that must be met for a study to qualify as evidence of TEI. We identified five criteria based on the consensus of studies that critically evaluated TEI. To assess whether published original research papers adhere to these criteria, we examined 80 studies that either claimed or were cited as supporting TEI. The findings of this analysis underscore the widespread confusion in this field and highlight the urgent need for a unified scientific consensus on TEI requirements.
Collapse
Affiliation(s)
- Hasan Khatib
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, WI, USA
| | - Jessica Townsend
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, WI, USA
| | - Melissa A Konkel
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, WI, USA
| | - Gabi Conidi
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, WI, USA
| | - Julia A Hasselkus
- The Department of Animal and Dairy Sciences, The University of Wisconsin, Madison, WI, USA
| |
Collapse
|
3
|
Wu X, Zhang W, Chen H, Weng J. Multifaceted paternal exposures before conception and their epigenetic impact on offspring. J Assist Reprod Genet 2024; 41:2931-2951. [PMID: 39230664 PMCID: PMC11621294 DOI: 10.1007/s10815-024-03243-1] [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: 06/10/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
As scientific research progresses, there is an increasing understanding of the importance of paternal epigenetics in influencing the health and developmental path of offspring. Prior to conception, the environmental exposures and lifestyle choices of fathers can significantly influence the epigenetic state of sperm, including DNA methylation and histone changes, among other factors. These alterations in epigenetic patterns have the potential for transgenerational transmission potential and may exert profound effects on the biological characteristics of descendants. Paternal epigenetic changes not only affect the regulation of gene expression patterns in offspring but also increase the risk to certain diseases. It is crucial to comprehend the conditions that fathers are exposed to before conception and the potential outcomes of these conditions. This understanding is essential for assessing personal reproductive decisions and anticipating health risks for future generations. This review article systematically summarizes and analyzes current research findings regarding how paternal pre-pregnancy exposures influence offspring as well as elucidates underlying mechanisms, aiming to provide a comprehensive perspective for an enhanced understanding of the impact that paternal factors have on offspring health.
Collapse
Affiliation(s)
- Xiaojing Wu
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Weiping Zhang
- The Second People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Huijun Chen
- The Second People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jianfei Weng
- The Second People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| |
Collapse
|
4
|
Luo M, Yi Y, Huang S, Dai S, Xie L, Liu K, Zhang S, Jiang T, Wang T, Yao B, Wang H, Xu D. Gestational dexamethasone exposure impacts hippocampal excitatory synaptic transmission and learning and memory function with transgenerational effects. Acta Pharm Sin B 2023; 13:3708-3727. [PMID: 37719378 PMCID: PMC10501875 DOI: 10.1016/j.apsb.2023.05.013] [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: 02/10/2023] [Revised: 04/20/2023] [Accepted: 05/06/2023] [Indexed: 09/19/2023] Open
Abstract
The formation of learning and memory is regulated by synaptic plasticity in hippocampal neurons. Here we explored how gestational exposure to dexamethasone, a synthetic glucocorticoid commonly used in clinical practice, has lasting effects on offspring's learning and memory. Adult offspring rats of prenatal dexamethasone exposure (PDE) displayed significant impairments in novelty recognition and spatial learning memory, with some phenotypes maintained transgenerationally. PDE impaired synaptic transmission of hippocampal excitatory neurons in offspring of F1 to F3 generations, and abnormalities of neurotransmitters and receptors would impair synaptic plasticity and lead to impaired learning and memory, but these changes failed to carry over to offspring of F5 and F7 generations. Mechanistically, altered hippocampal miR-133a-3p-SIRT1-CDK5-NR2B signaling axis in PDE multigeneration caused inhibition of excitatory synaptic transmission, which might be related to oocyte-specific high expression and transmission of miR-133a-3p. Together, PDE affects hippocampal excitatory synaptic transmission, with lasting consequences across generations, and CDK5 in offspring's peripheral blood might be used as an early-warning marker for fetal-originated learning and memory impairment.
Collapse
Affiliation(s)
- Mingcui Luo
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Yiwen Yi
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Songqiang Huang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Shiyun Dai
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Lulu Xie
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430071, China
| | - Kexin Liu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Shuai Zhang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Tao Jiang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Tingting Wang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Baozhen Yao
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Dan Xu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| |
Collapse
|
5
|
Rivi V, Rigillo G, Toscano Y, Benatti C, Blom JMC. Narrative Review of the Complex Interaction between Pain and Trauma in Children: A Focus on Biological Memory, Preclinical Data, and Epigenetic Processes. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1217. [PMID: 37508714 PMCID: PMC10378710 DOI: 10.3390/children10071217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023]
Abstract
The incidence and collective impact of early adverse experiences, trauma, and pain continue to increase. This underscores the urgent need for translational efforts between clinical and preclinical research to better understand the underlying mechanisms and develop effective therapeutic approaches. As our understanding of these issues improves from studies in children and adolescents, we can create more precise preclinical models and ultimately translate our findings back to clinical practice. A multidisciplinary approach is essential for addressing the complex and wide-ranging effects of these experiences on individuals and society. This narrative review aims to (1) define pain and trauma experiences in childhood and adolescents, (2) discuss the relationship between pain and trauma, (3) consider the role of biological memory, (4) decipher the relationship between pain and trauma using preclinical data, and (5) examine the role of the environment by introducing the importance of epigenetic processes. The ultimate scope is to better understand the wide-ranging effects of trauma, abuse, and chronic pain on children and adolescents, how they occur, and how to prevent or mitigate their effects and develop effective treatment strategies that address both the underlying causes and the associated physiological and psychological effects.
Collapse
Affiliation(s)
- Veronica Rivi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanna Rigillo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Ylenia Toscano
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cristina Benatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Johanna Maria Catharina Blom
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
6
|
Senner CE, Dong Z, Prater M, Branco MR, Watson ED. One-carbon metabolism is required for epigenetic stability in the mouse placenta. Front Cell Dev Biol 2023; 11:1209928. [PMID: 37440923 PMCID: PMC10333575 DOI: 10.3389/fcell.2023.1209928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/02/2023] [Indexed: 07/15/2023] Open
Abstract
One-carbon metabolism, including the folate cycle, has a crucial role in fetal development though its molecular function is complex and unclear. The hypomorphic Mtrr gt allele is known to disrupt one-carbon metabolism, and thus methyl group availability, leading to several developmental phenotypes (e.g., neural tube closure defects, fetal growth anomalies). Remarkably, previous studies showed that some of the phenotypes were transgenerationally inherited. Here, we explored the genome-wide epigenetic impact of one-carbon metabolism in placentas associated with fetal growth phenotypes and determined whether specific DNA methylation changes were inherited. Firstly, methylome analysis of Mtrr gt/gt homozygous placentas revealed genome-wide epigenetic instability. Several differentially methylated regions (DMRs) were identified including at the Cxcl1 gene promoter and at the En2 gene locus, which may have phenotypic implications. Importantly, we discovered hypomethylation and ectopic expression of a subset of ERV elements throughout the genome of Mtrr gt/gt placentas with broad implications for genomic stability. Next, we determined that known spermatozoan DMRs in Mtrr gt/gt males were reprogrammed in the placenta with little evidence of direct or transgenerational germline DMR inheritance. However, some spermatozoan DMRs were associated with placental gene misexpression despite normalisation of DNA methylation, suggesting the inheritance of an alternative epigenetic mechanism. Integration of published wildtype histone ChIP-seq datasets with Mtrr gt/gt spermatozoan methylome and placental transcriptome datasets point towards H3K4me3 deposition at key loci. These data suggest that histone modifications might play a role in epigenetic inheritance in this context. Overall, this study sheds light on the mechanistic complexities of one-carbon metabolism in development and epigenetic inheritance.
Collapse
Affiliation(s)
- Claire E. Senner
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ziqi Dong
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Malwina Prater
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Miguel R. Branco
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Erica D. Watson
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
7
|
Matoba N, Collins JW, Dizon MLV. Fetal Origins of Health Disparities: Transgenerational Consequences of Racism. Dev Neurosci 2023; 46:112-118. [PMID: 37290414 DOI: 10.1159/000531462] [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: 10/14/2022] [Accepted: 05/24/2023] [Indexed: 06/10/2023] Open
Abstract
Despite advances in perinatal medicine, racial disparity in birth outcomes remains a public health problem in the USA. The underlying mechanisms for this long-standing racial disparity are incompletely understood. This review presents transgenerational risk factors for racial disparities in preterm birth, exploring the impact of interpersonal and structural racism, theoretical models of stress, and biological markers of racial disparities.
Collapse
Affiliation(s)
- Nana Matoba
- Department of Pediatrics, Division of Neonatology, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Pediatrics, Division of Neonatology, University of California San Diego, Rady Children's Hospital San Diego, San Diego, California, USA
| | - James W Collins
- Department of Pediatrics, Division of Neonatology, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Maria L V Dizon
- Department of Pediatrics, Division of Neonatology, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
8
|
Verdikt R, Armstrong AA, Allard P. Transgenerational inheritance and its modulation by environmental cues. Curr Top Dev Biol 2022; 152:31-76. [PMID: 36707214 PMCID: PMC9940302 DOI: 10.1016/bs.ctdb.2022.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The epigenome plays an important role in shaping phenotypes. However, whether the environment can alter an organism's phenotype across several generations through epigenetic remodeling in the germline is still a highly debated topic. In this chapter, we briefly review the mechanisms of epigenetic inheritance and their connection with germline development before highlighting specific developmental windows of susceptibility to environmental cues. We further discuss the evidence of transgenerational inheritance to a range of different environmental cues, both epidemiological in humans and experimental in rodent models. Doing so, we pinpoint the current challenges in demonstrating transgenerational inheritance to environmental cues and offer insight in how recent technological advances may help deciphering the epigenetic mechanisms at play. Together, we draw a detailed picture of how our environment can influence our epigenomes, ultimately reshaping our phenotypes, in an extended theory of inheritance.
Collapse
Affiliation(s)
- Roxane Verdikt
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, United States.
| | - Abigail A Armstrong
- Department of Obstetrics/Gynecology and Division of Reproductive Endocrinology and Infertility, University of California, Los Angeles, CA, United States
| | - Patrick Allard
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
9
|
Lee B, Cyrill SL, Lee W, Melchiotti R, Andiappan AK, Poidinger M, Rötzschke O. Analysis of archaic human haplotypes suggests that 5hmC acts as an epigenetic guide for NCO recombination. BMC Biol 2022; 20:173. [PMID: 35927700 PMCID: PMC9354366 DOI: 10.1186/s12915-022-01353-9] [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: 04/01/2021] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
Background Non-crossover (NCO) refers to a mechanism of homologous recombination in which short tracks of DNA are copied between homologue chromatids. The allelic changes are typically restricted to one or few SNPs, which potentially allow for the gradual adaptation and maturation of haplotypes. It is assumed to be a stochastic process but the analysis of archaic and modern human haplotypes revealed a striking variability in local NCO recombination rates. Methods NCO recombination rates of 1.9 million archaic SNPs shared with Denisovan hominids were defined by a linkage study and correlated with functional and genomic annotations as well as ChIP-Seq data from modern humans. Results We detected a strong correlation between NCO recombination rates and the function of the respective region: low NCO rates were evident in introns and quiescent intergenic regions but high rates in splice sites, exons, 5′- and 3′-UTRs, as well as CpG islands. Correlations with ChIP-Seq data from ENCODE and other public sources further identified epigenetic modifications that associated directly with these recombination events. A particularly strong association was observed for 5-hydroxymethylcytosine marks (5hmC), which were enriched in virtually all of the functional regions associated with elevated NCO rates, including CpG islands and ‘poised’ bivalent regions. Conclusion Our results suggest that 5hmC marks may guide the NCO machinery specifically towards functionally relevant regions and, as an intermediate of oxidative demethylation, may open a pathway for environmental influence by specifically targeting recently opened gene loci. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01353-9.
Collapse
Affiliation(s)
- Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science Technology and Research (A*STAR), 8A Biomedical Drive, Singapore, 138648, Singapore.,Present address: Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Samantha Leeanne Cyrill
- Singapore Immunology Network (SIgN), Agency of Science Technology and Research (A*STAR), 8A Biomedical Drive, Singapore, 138648, Singapore.,Present address: Cold Spring Harbor Laboratory, One Bungtown Road, NY, 11724, Cold Spring Harbor, USA
| | - Wendy Lee
- Singapore Immunology Network (SIgN), Agency of Science Technology and Research (A*STAR), 8A Biomedical Drive, Singapore, 138648, Singapore
| | - Rossella Melchiotti
- Singapore Immunology Network (SIgN), Agency of Science Technology and Research (A*STAR), 8A Biomedical Drive, Singapore, 138648, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), Agency of Science Technology and Research (A*STAR), 8A Biomedical Drive, Singapore, 138648, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency of Science Technology and Research (A*STAR), 8A Biomedical Drive, Singapore, 138648, Singapore.,Present address: Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Victoria, 3052, Australia
| | - Olaf Rötzschke
- Singapore Immunology Network (SIgN), Agency of Science Technology and Research (A*STAR), 8A Biomedical Drive, Singapore, 138648, Singapore.
| |
Collapse
|
10
|
Molderings GJ. Systemic mast cell activation disease variants and certain genetically determined comorbidities may be consequences of a common underlying epigenetic disease. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Yin G, Xia L, Hou Y, Li Y, Cao D, Liu Y, Chen J, Liu J, Zhang L, Yang Q, Zhang Q, Tang N. Transgenerational male reproductive effect of prenatal arsenic exposure: abnormal spermatogenesis with Igf2/H19 epigenetic alteration in CD1 mouse. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:1248-1260. [PMID: 33406855 DOI: 10.1080/09603123.2020.1870668] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Developmental exposure to environmental toxicants can induce transgenerational reproductive disease phenotypes through epigenetic mechanisms. We treated pregnant CD-1 (F0) mice with drinking water containing sodium arsenite (85 ppm) from days 8 to 18 of gestation. Male offspring were bred with untreated female mice until the F3 generation was produced. Our results revealed that F0 transient exposure to arsenic can cause decreased sperm quality and histological abnormalities in the F1 and F3. The overall methylation status of Igf2 DMR2 and H19 DMR was significantly lower in the arsenic-exposed group than that of the control group in both F1 and F3. The relative mRNA expression levels of Igf2 and H19 in arsenic-exposed males were significantly increased in both F1 and F3. This study indicates that ancestral exposure to arsenic may result in transgenerational inheritance of an impaired spermatogenesis phenotyping involving both epigenetic alterations and the abnormal expression of Igf2 and H19.
Collapse
Affiliation(s)
- Guoying Yin
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Liting Xia
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yaxing Hou
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yaoyan Li
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Deqing Cao
- Central Laboratory of Preventive Medicine, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yanan Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jingshan Chen
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Juan Liu
- Department of Biomedical Information and Library, Tianjin Medical University, Tianjin, China
| | - Liwen Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qiaoyun Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qiang Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Naijun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, School of Public Health, Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Braz CU, Taylor T, Namous H, Townsend J, Crenshaw T, Khatib H. Paternal diet induces transgenerational epigenetic inheritance of DNA methylation signatures and phenotypes in sheep model. PNAS NEXUS 2022; 1:pgac040. [PMID: 36713326 PMCID: PMC9802161 DOI: 10.1093/pnasnexus/pgac040] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/05/2022] [Accepted: 04/02/2022] [Indexed: 06/18/2023]
Abstract
Transgenerational epigenetic inheritance (TEI) requires transmission of environmentally induced epigenetic changes and associated phenotypes to subsequent generations without continued exposure to the environmental factor that originated the change. TEI is well-established in plants and Caenorhabditis elegans; however, occurrence in mammals is debated and poorly understood. Here, we examined whether paternal diet from weaning to puberty-induced changes in sperm DNA methylation that were transmitted to subsequent generations. Over 100 methylated cytosines, environmentally altered in the F0 generation, were inherited by the F1 and F2 generations. Furthermore, the F0 paternal diet was associated with growth and male fertility phenotypes in subsequent generations. Differentially methylated cytosines were correlated with gene expression. Our results demonstrate that some sperm methylation sites may escape DNA methylation erasure and are transmitted to subsequent generations despite the 2 waves of epigenetic programming: in primordial germ cells and in embryos after fertilization. These results advance our understanding of the complex relationships between nature and nurture.
Collapse
Affiliation(s)
- Camila U Braz
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Todd Taylor
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Hadjer Namous
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica Townsend
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Thomas Crenshaw
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | |
Collapse
|
13
|
Srichaikul K, Hegele RA, Jenkins DJA. Great Chinese Famine and the Effects on Cardiometabolic Health for Future Generations. Hypertension 2022; 79:532-535. [PMID: 35138871 DOI: 10.1161/hypertensionaha.121.18546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Korbua Srichaikul
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Ontario, Canada (K.S., D.J.A.J.).,Clinical Nutrition Risk Factor Modification Centre, St. Michael's Hospital, Toronto, Ontario, Canada (K.S., D.J.A.J.).,Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada (K.S., D.J.A.J.)
| | - Robert A Hegele
- Department of Medicine, Western University, London, Ontario, Canada (R.A.H.)
| | - David J A Jenkins
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Ontario, Canada (K.S., D.J.A.J.).,Clinical Nutrition Risk Factor Modification Centre, St. Michael's Hospital, Toronto, Ontario, Canada (K.S., D.J.A.J.).,Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada (K.S., D.J.A.J.).,Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Ontario, Canada (D.J.A.J.).,Li Ka Shing Knowledge Institute (D.J.A.J.), St. Michael's Hospital, Toronto, Ontario, Canada.,Division of Endocrinology and Metabolism (D.J.A.J.), St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Wilkinson AL, Menelaou K, Rakoczy J, Tan XS, Watson ED. Disruption of Folate Metabolism Causes Poor Alignment and Spacing of Mouse Conceptuses for Multiple Generations. Front Cell Dev Biol 2021; 9:723978. [PMID: 34957089 PMCID: PMC8703036 DOI: 10.3389/fcell.2021.723978] [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: 06/11/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022] Open
Abstract
Abnormal uptake or metabolism of folate increases risk of human pregnancy complications, though the mechanism is unclear. Here, we explore how defective folate metabolism influences early development by analysing mice with the hypomorphic Mtrrgt mutation. MTRR is necessary for methyl group utilisation from folate metabolism, and the Mtrrgt allele disrupts this process. We show that the spectrum of phenotypes previously observed in Mtrrgt/gt conceptuses at embryonic day (E) 10.5 is apparent from E8.5 including developmental delay, congenital malformations, and placental phenotypes. Notably, we report misalignment of some Mtrrgt conceptuses within their implantation sites from E6.5. The degree of misorientation occurs across a continuum, with the most severe form visible upon gross dissection. Additionally, some Mtrrgt/gt conceptuses display twinning. Therefore, we implicate folate metabolism in blastocyst orientation and spacing at implantation. Skewed growth likely influences embryo development since developmental delay and heart malformations (but not defects in neural tube closure or trophoblast differentiation) associate with severe misalignment of Mtrrgt/gt conceptuses. Typically, the uterus is thought to guide conceptus orientation. To investigate a uterine effect of the Mtrrgt allele, we manipulate the maternal Mtrr genotype. Misaligned conceptuses were observed in litters of Mtrr+/+, Mtrr+/gt, and Mtrrgt/gt mothers. While progesterone and/or BMP2 signalling might be disrupted, normal decidual morphology, patterning, and blood perfusion are evident at E6.5 regardless of conceptus orientation. These observations argue against a post-implantation uterine defect as a cause of conceptus misalignment. Since litters of Mtrr+/+ mothers display conceptus misalignment, a grandparental effect is explored. Multigenerational phenotype inheritance is characteristic of the Mtrrgt model, though the mechanism remains unclear. Genetic pedigree analysis reveals that severe conceptus skewing associates with the Mtrr genotype of either maternal grandparent. Moreover, the presence of conceptus skewing after embryo transfer into a control uterus indicates that misalignment is independent of the peri- and/or post-implantation uterus and instead is likely attributed to an embryonic mechanism that is epigenetically inherited. Overall, our data indicates that abnormal folate metabolism influences conceptus orientation over multiple generations with implications for subsequent development. This study casts light on the complex role of folate metabolism during development beyond a direct maternal effect.
Collapse
Affiliation(s)
- Amy L Wilkinson
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Katerina Menelaou
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Joanna Rakoczy
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Xiu S Tan
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Erica D Watson
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Deans C. Biological Prescience: The Role of Anticipation in Organismal Processes. Front Physiol 2021; 12:672457. [PMID: 34975512 PMCID: PMC8719636 DOI: 10.3389/fphys.2021.672457] [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: 02/25/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Anticipation is the act of using information about the past and present to make predictions about future scenarios. As a concept, it is predominantly associated with the psychology of the human mind; however, there is accumulating evidence that diverse taxa without complex neural systems, and even biochemical networks themselves, can respond to perceived future conditions. Although anticipatory processes, such as circadian rhythms, stress priming, and cephalic responses, have been extensively studied over the last three centuries, newer research on anticipatory genetic networks in microbial species shows that anticipatory processes are widespread, evolutionarily old, and not simply reserved for neurological complex organisms. Overall, data suggest that anticipatory responses represent a unique type of biological processes that can be distinguished based on their organizational properties and mechanisms. Unfortunately, an empirically based biologically explicit framework for describing anticipatory processes does not currently exist. This review attempts to fill this void by discussing the existing examples of anticipatory processes in non-cognitive organisms, providing potential criteria for defining anticipatory processes, as well as their putative mechanisms, and drawing attention to the often-overlooked role of anticipation in the evolution of physiological systems. Ultimately, a case is made for incorporating an anticipatory framework into the existing physiological paradigm to advance our understanding of complex biological processes.
Collapse
Affiliation(s)
- Carrie Deans
- Entomology Department, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
16
|
Anastasiadi D, Venney CJ, Bernatchez L, Wellenreuther M. Epigenetic inheritance and reproductive mode in plants and animals. Trends Ecol Evol 2021; 36:1124-1140. [PMID: 34489118 DOI: 10.1016/j.tree.2021.08.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022]
Abstract
Epigenetic inheritance is another piece of the puzzle of nongenetic inheritance, although the prevalence, sources, persistence, and phenotypic consequences of heritable epigenetic marks across taxa remain unclear. We systematically reviewed over 500 studies from the past 5 years to identify trends in the frequency of epigenetic inheritance due to differences in reproductive mode and germline development. Genetic, intrinsic (e.g., disease), and extrinsic (e.g., environmental) factors were identified as sources of epigenetic inheritance, with impacts on phenotype and adaptation depending on environmental predictability. Our review shows that multigenerational persistence of epigenomic patterns is common in both plants and animals, but also highlights many knowledge gaps that remain to be filled. We provide a framework to guide future studies towards understanding the generational persistence and eco-evolutionary significance of epigenomic patterns.
Collapse
Affiliation(s)
- Dafni Anastasiadi
- The New Zealand Institute for Plant and Food Research Ltd, Nelson Research Centre, 293 Akersten St, Nelson 7010, New Zealand
| | - Clare J Venney
- Institut de Biologie Intégrative des Systèmes (IBIS), Département de Biologie, Université Laval, 1030 Avenue de la Médecine, G1V 0A6, Québec, QC, Canada
| | - Louis Bernatchez
- Institut de Biologie Intégrative des Systèmes (IBIS), Département de Biologie, Université Laval, 1030 Avenue de la Médecine, G1V 0A6, Québec, QC, Canada
| | - Maren Wellenreuther
- The New Zealand Institute for Plant and Food Research Ltd, Nelson Research Centre, 293 Akersten St, Nelson 7010, New Zealand; School of Biological Sciences, The University of Auckland, 3A Symonds St, Auckland 1010, New Zealand.
| |
Collapse
|
17
|
Ramakrishna NB, Murison K, Miska EA, Leitch HG. Epigenetic Regulation during Primordial Germ Cell Development and Differentiation. Sex Dev 2021; 15:411-431. [PMID: 34847550 DOI: 10.1159/000520412] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/10/2021] [Indexed: 11/19/2022] Open
Abstract
Germline development varies significantly across metazoans. However, mammalian primordial germ cell (PGC) development has key conserved landmarks, including a critical period of epigenetic reprogramming that precedes sex-specific differentiation and gametogenesis. Epigenetic alterations in the germline are of unique importance due to their potential to impact the next generation. Therefore, regulation of, and by, the non-coding genome is of utmost importance during these epigenomic events. Here, we detail the key chromatin changes that occur during mammalian PGC development and how these interact with the expression of non-coding RNAs alongside broader epitranscriptomic changes. We identify gaps in our current knowledge, in particular regarding epigenetic regulation in the human germline, and we highlight important areas of future research.
Collapse
Affiliation(s)
- Navin B Ramakrishna
- Wellcome/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Genome Institute of Singapore, A*STAR, Biopolis, Singapore, Singapore
| | - Keir Murison
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
| | - Eric A Miska
- Wellcome/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Harry G Leitch
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
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: 2.8] [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
|
19
|
Der Sarkissian A, Sharkey JD. Transgenerational Trauma and Mental Health Needs among Armenian Genocide Descendants. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph181910554. [PMID: 34639853 PMCID: PMC8508035 DOI: 10.3390/ijerph181910554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/01/2021] [Accepted: 10/03/2021] [Indexed: 11/16/2022]
Abstract
The trauma of a genocide can be transmitted to subsequent generations though familial mental health, sociopolitical trauma, and cultural narratives, thereby impacting mental health and well-being. Understanding specific mechanisms that are unique to each ethnic group impacted by genocide illuminates cultural, sociopolitical, and individual factors related to the transmission. For the Armenian community, the unresolved historical loss of the Armenian Genocide of 1915, with the threat of acculturation for such a large diasporic population, a continued denial by the perpetrators, as well as subsequent generations’ refugee experiences, may further exasperate the impact of transgenerational trauma from the genocide. This literature review explores the mental health needs of Armenian youth in the current sociopolitical context and provides implications for how schools and communities may use this knowledge to inform supports that center Armenian community healing. Future directions for research are also discussed.
Collapse
|
20
|
Mukherjee S, Dasgupta S, Mishra PK, Chaudhury K. Air pollution-induced epigenetic changes: disease development and a possible link with hypersensitivity pneumonitis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:55981-56002. [PMID: 34498177 PMCID: PMC8425320 DOI: 10.1007/s11356-021-16056-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/16/2021] [Indexed: 05/16/2023]
Abstract
Air pollution is a serious threat to our health and has become one of the major causes of many diseases including cardiovascular disease, respiratory disease, and cancer. The association between air pollution and various diseases has long been a topic of research interest. However, it remains unclear how air pollution actually impacts health by modulating several important cellular functions. Recently, some evidence has emerged about air pollution-induced epigenetic changes, which are linked with the etiology of various human diseases. Among several epigenetic modifications, DNA methylation represents the most prominent epigenetic alteration underlying the air pollution-induced pathogenic mechanism. Several other types of epigenetic changes, such as histone modifications, miRNA, and non-coding RNA expression, have also been found to have been linked with air pollution. Hypersensitivity pneumonitis (HP), one of the most prevalent forms of interstitial lung diseases (ILDs), is triggered by the inhalation of certain organic and inorganic substances. HP is characterized by inflammation in the tissues around the lungs' airways and may lead to irreversible lung scarring over time. This review, in addition to other diseases, attempts to understand whether certain pollutants influence HP development through such epigenetic modifications.
Collapse
Affiliation(s)
- Suranjana Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.
| | - Sanjukta Dasgupta
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Pradyumna K Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| |
Collapse
|
21
|
Verdikt R, Allard P. Metabolo-epigenetics: the interplay of metabolism and epigenetics during early germ cells development. Biol Reprod 2021; 105:616-624. [PMID: 34132770 PMCID: PMC8444669 DOI: 10.1093/biolre/ioab118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/26/2022] Open
Abstract
Metabolites control epigenetic mechanisms, and conversly, cell metabolism is regulated at the epigenetic level in response to changes in the cellular environment. In recent years, this metabolo-epigenetic control of gene expression has been implicated in the regulation of multiple stages of embryonic development. The developmental potency of stem cells and their embryonic counterparts is directly determined by metabolic rewiring. Here, we review the current knowledge on the interplay between epigenetics and metabolism in the specific context of early germ cell development. We explore the implications of metabolic rewiring in primordial germ cells in light of their epigenetic remodeling during cell fate determination. Finally, we discuss the relevance of concerted metabolic and epigenetic regulation of primordial germ cells in the context of mammalian transgenerational epigenetic inheritance.
Collapse
Affiliation(s)
- Roxane Verdikt
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Patrick Allard
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
22
|
Waterman R, Sultan SE. Transgenerational effects of parent plant competition on offspring development in contrasting conditions. Ecology 2021; 102:e03531. [PMID: 34496058 DOI: 10.1002/ecy.3531] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/13/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022]
Abstract
Conditions during a parent's lifetime can induce phenotypic changes in offspring, providing a potentially important source of variation in natural populations. Yet, to date, biotic factors have seldom been tested as sources of transgenerational effects in plants. In a greenhouse experiment with the generalist annual Polygonum persicaria, we tested for effects of parental competition on offspring by growing isogenic parent plants either individually or in competitive arrays and comparing their seedling progeny in contrasting growth environments. Offspring of competing vs. non-competing parents showed significantly altered development, resulting in greater biomass and total leaf area, but only when growing in neighbor or simulated canopy shade, rather than sunny dry conditions. A follow-up experiment in which parent plants instead competed in dry soil found that offspring in dry soil had slightly reduced growth, both with and without competitors. In neither experiment were effects of parental competition explained by changes in seed provisioning, suggesting a more complex mode of regulatory inheritance. We hypothesize that parental competition in moist soil (i.e., primarily for light) confers specific developmental effects that are beneficial for light-limited offspring, while parental competition in dry soil (i.e., primarily for belowground resources) produces offspring of slightly lower overall quality. Together, these results indicate that competitive conditions during the parental generation can contribute significantly to offspring variation, but these transgenerational effects will depend on the abiotic resources available to both parents and progeny.
Collapse
Affiliation(s)
- Robin Waterman
- Biology Department, Wesleyan University, Middletown, Connecticut, 06459, USA.,Department of Plant Biology, Michigan State University, East Lansing, Michigan, 48823, USA
| | - Sonia E Sultan
- Biology Department, Wesleyan University, Middletown, Connecticut, 06459, USA
| |
Collapse
|
23
|
Gerra MC, Dallabona C, Arendt-Nielsen L. Epigenetic Alterations in Prescription Opioid Misuse: New Strategies for Precision Pain Management. Genes (Basel) 2021; 12:genes12081226. [PMID: 34440400 PMCID: PMC8392465 DOI: 10.3390/genes12081226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 12/11/2022] Open
Abstract
Prescription opioids are used for some chronic pain conditions. However, generally, long-term therapy has unwanted side effects which may trigger addiction, overdose, and eventually cause deaths. Opioid addiction and chronic pain conditions have both been associated with evidence of genetic and epigenetic alterations. Despite intense research interest, many questions about the contribution of epigenetic changes to this typology of addiction vulnerability and development remain unanswered. The aim of this review was to summarize the epigenetic modifications detected in specific tissues or brain areas and associated with opioid prescription and misuse in patients who have initiated prescribed opioid management for chronic non-cancer pain. The review considers the effects of opioid exposure on the epigenome in central and peripheral tissues in animal models and human subjects and highlights the mechanisms in which opioid epigenetics may be involved. This will improve our current understanding, provide the basis for targeted, personalized pain management, and thus balance opioid risks and benefits in managing chronic pain.
Collapse
Affiliation(s)
- Maria Carla Gerra
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark;
- Correspondence:
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, 43123 Parma, Italy;
| | - Lars Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP), SMI, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark;
| |
Collapse
|
24
|
|
25
|
Blanc M, Antczak P, Cousin X, Grunau C, Scherbak N, Rüegg J, Keiter SH. The insecticide permethrin induces transgenerational behavioral changes linked to transcriptomic and epigenetic alterations in zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 779:146404. [PMID: 33752003 DOI: 10.1016/j.scitotenv.2021.146404] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 06/12/2023]
Abstract
The pyrethroid insecticide permethrin is widely used for agricultural and domestic purposes. Previous data indicated that it acts as a developmental neurotoxicant and can induce transgenerational effects in non-target organisms. However, associated underlying mechanisms remain unclear. The aim of this study was to investigate permethrin-related transgenerational effects in the zebrafish model, and to identify possible molecular mechanisms underlying inheritance. Zebrafish (F0) were exposed to permethrin during early-life (2 h post-fertilization up to 28 days). The F1 and F2 offspring generations were obtained by pairing exposed F0 males and females, and were bred unexposed. Locomotor and anxiety behavior were investigated, together with transcriptomic and epigenomic (DNA methylation) changes in brains. Permethrin exposed F0 fish were hypoactive at adulthood, while males from the F1 and F2 generations showed a specific decrease in anxiety-like behavior. In F0, transcriptomic data showed enrichment in pathways related to glutamatergic synapse activity, which may partly underlie the behavioral effects. In F1 and F2 males, dysregulation of similar pathways was observed, including a subset of differentially methylated regions that were inherited from the F0 to the F2 generation and indicated stable dysregulation of glutamatergic signaling. Altogether, the present results provide novel evidence on the transgenerational neurotoxic effects of permethrin, as well as mechanistic insight: a transient exposure induces persistent transcriptional and DNA methylation changes that may translate into transgenerational alteration of glutamatergic signaling and, thus, into behavioral alterations.
Collapse
Affiliation(s)
- Mélanie Blanc
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden; MARBEC, University of Montpellier, CNRS, Ifremer, IRD, Palavas, France; Université Paris-Saclay, AgroParisTech, INRAE, GABI, Domaine de Vilvert, F-78350 Jouy-en-Josas, France.
| | - Philipp Antczak
- Centre for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Xavier Cousin
- MARBEC, University of Montpellier, CNRS, Ifremer, IRD, Palavas, France; Université Paris-Saclay, AgroParisTech, INRAE, GABI, Domaine de Vilvert, F-78350 Jouy-en-Josas, France
| | - Christoph Grunau
- IHPE, Univ. Montpellier, CNRS, Ifremer, Univ. Perpignan Via Domitia, Perpignan, France
| | - Nikolai Scherbak
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden; Örebro Life Science Centre, School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden
| | - Joëlle Rüegg
- Department of Organismal Biology, Uppsala University, Norbyv. 18A, 75236 Uppsala, Sweden
| | - Steffen H Keiter
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden
| |
Collapse
|
26
|
Defective folate metabolism causes germline epigenetic instability and distinguishes Hira as a phenotype inheritance biomarker. Nat Commun 2021; 12:3714. [PMID: 34140513 PMCID: PMC8211854 DOI: 10.1038/s41467-021-24036-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/29/2021] [Indexed: 02/02/2023] Open
Abstract
The mechanism behind transgenerational epigenetic inheritance is unclear, particularly through the maternal grandparental line. We previously showed that disruption of folate metabolism in mice by the Mtrr hypomorphic mutation results in transgenerational epigenetic inheritance of congenital malformations. Either maternal grandparent can initiate this phenomenon, which persists for at least four wildtype generations. Here, we use genome-wide approaches to reveal genetic stability in the Mtrr model and genome-wide differential DNA methylation in the germline of Mtrr mutant maternal grandfathers. We observe that, while epigenetic reprogramming occurs, wildtype grandprogeny and great grandprogeny exhibit transcriptional changes that correlate with germline methylation defects. One region encompasses the Hira gene, which is misexpressed in embryos for at least three wildtype generations in a manner that distinguishes Hira transcript expression as a biomarker of maternal phenotypic inheritance.
Collapse
|
27
|
Mirra P, Desiderio A, Spinelli R, Nigro C, Longo M, Parrillo L, D'Esposito V, Carissimo A, Hedjazifar S, Smith U, Formisano P, Miele C, Raciti GA, Beguinot F. Adipocyte precursor cells from first degree relatives of type 2 diabetic patients feature changes in hsa-mir-23a-5p, -193a-5p, and -193b-5p and insulin-like growth factor 2 expression. FASEB J 2021; 35:e21357. [PMID: 33710685 DOI: 10.1096/fj.202002156rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022]
Abstract
First-degree relatives (FDRs) of type 2 diabetics (T2D) feature dysfunction of subcutaneous adipose tissue (SAT) long before T2D onset. miRNAs have a role in adipocyte precursor cells (APC) differentiation and in adipocyte identity. Thus, impaired miRNA expression may contribute to SAT dysfunction in FDRs. In the present work, we have explored changes in miRNA expression associated with T2D family history which may affect gene expression in SAT APCs from FDRs. Small RNA-seq was performed in APCs from healthy FDRs and matched controls and omics data were validated by qPCR. Integrative analyses of APC miRNome and transcriptome from FDRs revealed down-regulated hsa-miR-23a-5p, -193a-5p and -193b-5p accompanied by up-regulated Insulin-like Growth Factor 2 (IGF2) gene which proved to be their direct target. The expression changes in these marks were associated with SAT adipocyte hypertrophy in FDRs. APCs from FDRs further demonstrated reduced capability to differentiate into adipocytes. Treatment with IGF2 protein decreased APC adipogenesis, while over-expression of hsa-miR-23a-5p, -193a-5p and -193b-5p enhanced adipogenesis by IGF2 targeting. Indeed, IGF2 increased the Wnt Family Member 10B gene expression in APCs. Down-regulation of the three miRNAs and IGF2 up-regulation was also observed in Peripheral Blood Leukocytes (PBLs) from FDRs. In conclusion, APCs from FDRs feature a specific miRNA/gene profile, which associates with SAT adipocyte hypertrophy and appears to contribute to impaired adipogenesis. PBL detection of this profile may help in identifying adipocyte hypertrophy in individuals at high risk of T2D.
Collapse
Affiliation(s)
- Paola Mirra
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Antonella Desiderio
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Rosa Spinelli
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Cecilia Nigro
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Michele Longo
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Luca Parrillo
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Vittoria D'Esposito
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | | | - Shahram Hedjazifar
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pietro Formisano
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Claudia Miele
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Gregory A Raciti
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Francesco Beguinot
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
28
|
Alves N, Neuparth T, Barros S, Santos MM. The anti-lipidemic drug simvastatin modifies epigenetic biomarkers in the amphipod Gammarus locusta. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 209:111849. [PMID: 33387775 DOI: 10.1016/j.ecoenv.2020.111849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/17/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
The adverse effects of certain environmental chemicals have been recently associated with the modulation of the epigenome. Although changes in the epigenetic signature have yet to be integrated into hazard and risk assessment, they are interesting candidates to link environmental exposures and altered phenotypes, since these changes may be passed across multiple non-exposed generations. Here, we addressed the effects of simvastatin (SIM), one of the most prescribed pharmaceuticals in the world, on epigenetic regulation using the amphipod Gammarus locusta as a proxy, to support its integration into hazard and environmental risk assessment. SIM is a known modulator of the epigenome in mammalian cell lines and has been reported to impact G. locusta ecological endpoints at environmentally relevant levels. G. locusta juveniles were exposed to three SIM environmentally relevant concentrations (0.32, 1.6 and 8 µg L-1) for 15 days. Gene transcription levels of selected epigenetic regulators, i.e., dnmt1, dmap1, usp7, kat5 and uhrf1 were assessed, along with the quantification of DNA methylation levels and evaluation of key ecological endpoints: survival and growth. Exposure to 0.32 and 8 µg L-1 SIM induced significant downregulation of DNA methyltransferase 1 (dnmt1), concomitant with global DNA hypomethylation and growth impacts. Overall, this work is the first to validate the basal expression of key epigenetic regulators in a keystone marine crustacean, supporting the integration of epigenetic biomarkers into hazard assessment frameworks.
Collapse
Affiliation(s)
- Nélson Alves
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Endocrine Disruptors and Emerging Contaminants Group, University of Porto, Avenida General Norton de Matos S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre nº 1021/1055, 4169-007 Porto, Portugal
| | - Teresa Neuparth
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Endocrine Disruptors and Emerging Contaminants Group, University of Porto, Avenida General Norton de Matos S/N, 4450-208 Matosinhos, Portugal.
| | - Susana Barros
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Endocrine Disruptors and Emerging Contaminants Group, University of Porto, Avenida General Norton de Matos S/N, 4450-208 Matosinhos, Portugal
| | - Miguel M Santos
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Endocrine Disruptors and Emerging Contaminants Group, University of Porto, Avenida General Norton de Matos S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre nº 1021/1055, 4169-007 Porto, Portugal.
| |
Collapse
|
29
|
miRNA320a-3p/RUNX2 signal programming mediates the transgenerational inheritance of inhibited ovarian estrogen synthesis in female offspring rats induced by prenatal dexamethasone exposure. Pharmacol Res 2021; 165:105435. [PMID: 33485996 DOI: 10.1016/j.phrs.2021.105435] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 12/11/2020] [Accepted: 01/05/2021] [Indexed: 11/22/2022]
Abstract
Our previous studies found that prenatal dexamethasone exposure could cause abnormal follicular development in fetal rats. This study intends to observe the transgenerational inheritance effects of ovarian estrogen inhibition in offspring exposed to dexamethasone (0.2 mg/kg • d) from gestational day 9 (GD9) to GD20 in Wistar rats, and explore the intrauterine programming mechanisms. Prenatal dexamethasone exposure reduced the expression of ovarian cytochrome P450 aromatase (P450arom), the level of serum estradiol (E2) and the number of primordial follicles, while increased the number of atresia follicles before and after birth in F1 offspring rats. At the same time, the expression of miRNA320a-3p in F1 ovaries was down-regulated, and RUNX2 expression increased significantly. These changes were continued to F2 and F3 generations, accompanied by consistently down-regulated miRNA320a-3p expression in oocyte of F1 and F2 adult offspring. In vitro, fetal rat ovaries and KGN human ovarian granulosa cells were treated with dexamethasone. It showed that dexamethasone decreased miRNA320a-3p and P450arom expression, as well as E2 synthesis, and increased RUNX2 expression. All these effects could be reversed by the GR antagonist RU486. The overexpression of miRNA320a-3p in vitro could also reverse the effects of dexamethasone on RUNX2, P450arom, and E2 levels. The dual-luciferase reporter gene experiment further confirmed the direct targeted regulation of miRNA320a-3p on RUNX2. These results indicate that prenatal dexamethasone exposure induces ovarian E2 synthesis inhibition mediated by the GR/miRNA320a-3p/RUNX2/P450arom cascade signal in fetal rat ovary, which has transgenerational inheritance effects and may related to the inhibited miRNA320a-3p expression in oocyte.
Collapse
|
30
|
Christoforou ER, Sferruzzi-Perri AN. Molecular mechanisms governing offspring metabolic programming in rodent models of in utero stress. Cell Mol Life Sci 2020; 77:4861-4898. [PMID: 32494846 PMCID: PMC7658077 DOI: 10.1007/s00018-020-03566-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
The results of different human epidemiological datasets provided the impetus to introduce the now commonly accepted theory coined as 'developmental programming', whereby the presence of a stressor during gestation predisposes the growing fetus to develop diseases, such as metabolic dysfunction in later postnatal life. However, in a clinical setting, human lifespan and inaccessibility to tissue for analysis are major limitations to study the molecular mechanisms governing developmental programming. Subsequently, studies using animal models have proved indispensable to the identification of key molecular pathways and epigenetic mechanisms that are dysregulated in metabolic organs of the fetus and adult programmed due to an adverse gestational environment. Rodents such as mice and rats are the most used experimental animals in the study of developmental programming. This review summarises the molecular pathways and epigenetic mechanisms influencing alterations in metabolic tissues of rodent offspring exposed to in utero stress and subsequently programmed for metabolic dysfunction. By comparing molecular mechanisms in a variety of rodent models of in utero stress, we hope to summarise common themes and pathways governing later metabolic dysfunction in the offspring whilst identifying reasons for incongruencies between models so to inform future work. With the continued use and refinement of such models of developmental programming, the scientific community may gain the knowledge required for the targeted treatment of metabolic diseases that have intrauterine origins.
Collapse
Affiliation(s)
- Efthimia R Christoforou
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge, UK
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge, UK.
| |
Collapse
|
31
|
Adrian-Kalchhauser I, Sultan SE, Shama LNS, Spence-Jones H, Tiso S, Keller Valsecchi CI, Weissing FJ. Understanding 'Non-genetic' Inheritance: Insights from Molecular-Evolutionary Crosstalk. Trends Ecol Evol 2020; 35:1078-1089. [PMID: 33036806 DOI: 10.1016/j.tree.2020.08.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/23/2022]
Abstract
Understanding the evolutionary and ecological roles of 'non-genetic' inheritance (NGI) is daunting due to the complexity and diversity of epigenetic mechanisms. We draw on insights from molecular and evolutionary biology perspectives to identify three general features of 'non-genetic' inheritance systems: (i) they are functionally interdependent with, rather than separate from, DNA sequence; (ii) precise mechanisms vary phylogenetically and operationally; and (iii) epigenetic elements are probabilistic, interactive regulatory factors and not deterministic 'epialleles' with defined genomic locations and effects. We discuss each of these features and offer recommendations for future empirical and theoretical research that implements a unifying inherited gene regulation (IGR) approach to studies of 'non-genetic' inheritance.
Collapse
Affiliation(s)
- Irene Adrian-Kalchhauser
- Centre for Fish and Wildlife Health, Department for Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland.
| | - Sonia E Sultan
- Biology Department, Wesleyan University, Middletown, CT 06459, USA
| | - Lisa N S Shama
- Coastal Ecology Section, Alfred Wegener Institute Helmholtz Centre for Polar and Marine Research, Wadden Sea Station Sylt, Hafenstrasse 43, 25992 List, Germany
| | - Helen Spence-Jones
- Centre for Biological Diversity, School of Biology, University of St Andrews, St. Andrews, UK
| | - Stefano Tiso
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | | | - Franz J Weissing
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747, AG, Groningen, The Netherlands
| |
Collapse
|
32
|
Lowry JL, Ryan ÉB, Esengul YT, Siddique N, Siddique T. Intricacies of aetiology in intrafamilial degenerative disease. Brain Commun 2020; 2:fcaa120. [PMID: 33134917 PMCID: PMC7585693 DOI: 10.1093/braincomms/fcaa120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/23/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023] Open
Abstract
The genetic underpinnings of late-onset degenerative disease have typically been determined by screening families for the segregation of genetic variants with the disease trait in affected, but not unaffected, individuals. However, instances of intrafamilial etiological heterogeneity, where pathogenic variants in a culprit gene are not shared among all affected family members, continue to emerge and confound gene-discovery and genetic counselling efforts. Discordant intrafamilial cases lacking a mutation shared by other affected family members are described as disease phenocopies. This description often results in an over-simplified acceptance of an environmental cause of disease in the phenocopy cases, while the role of intrafamilial genetic heterogeneity, shared de novo mutations or epigenetic aberrations in such families is often ignored. On a related note, it is now evident that the same disease-associated variant can be present in individuals exhibiting clinically distinct phenotypes, thereby genetically uniting seemingly unrelated syndromes to form a spectrum of disease. Herein, we discuss the intricacies of determining complex degenerative disease aetiology and suggest alternative mechanisms of disease transmission that may account for the apparent missing heritability of disease.
Collapse
Affiliation(s)
- Jessica L Lowry
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Éanna B Ryan
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Y Taylan Esengul
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nailah Siddique
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Teepu Siddique
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Pathology Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
33
|
Blake GET, Hall J, Petkovic GE, Watson ED. Analysis of spermatogenesis and fertility in adult mice with a hypomorphic mutation in the Mtrr gene. Reprod Fertil Dev 2020; 31:1730-1741. [PMID: 31537252 DOI: 10.1071/rd19064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/16/2019] [Indexed: 01/22/2023] Open
Abstract
Recent research has focussed on the significance of folate metabolism in male fertility. Knocking down the mouse gene Mtrr impedes the progression of folate and methionine metabolism and results in hyperhomocysteinaemia, dysregulation of DNA methylation and developmental phenotypes (e.g. neural tube, heart and placenta defects). The Mtrrgt mouse line is a model of transgenerational epigenetic inheritance (TEI), the hypothesised cause of which is the inheritance of a yet-to-be determined epigenetic factor via the germline. We investigated Mtrrgt/gt testes and sperm function compared with control C57Bl/6J testes to explore potential defects that might confound our understanding of TEI in the Mtrrgt model. Histological analysis revealed that adult Mtrrgt/gt testes are more spherical in shape than C57Bl/6J testes, though serum testosterone levels were normal and spermatogenesis progressed in a typical manner. Spermatozoa collected from the cauda epididymis showed normal morphology, counts, and viability in Mtrrgt/gt males. Correspondingly, Mtrrgt spermatozoa contributed to normal pregnancy rates. Similar parameters were assessed in Mtrr+/+ and Mtrr+/gt males, which were normal compared with controls. Overall, our data showed that the Mtrrgt allele is unlikely to alter spermatogenesis or male fertility. Therefore, it is improbable that these factors confound the mechanistic study of TEI in Mtrrgt mice.
Collapse
Affiliation(s)
- Georgina E T Blake
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK; and Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Jessica Hall
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Grace E Petkovic
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Erica D Watson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK; and Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK; and Corresponding author.
| |
Collapse
|
34
|
Intrauterine RAS programming alteration-mediated susceptibility and heritability of temporal lobe epilepsy in male offspring rats induced by prenatal dexamethasone exposure. Arch Toxicol 2020; 94:3201-3215. [PMID: 32494933 DOI: 10.1007/s00204-020-02796-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 05/28/2020] [Indexed: 12/22/2022]
Abstract
Partial temporal lobe epilepsy (TLE) has an intrauterine developmental origin. This study was aimed at elucidating the heritable effects and programming mechanism of TLE in offspring rats induced by prenatal dexamethasone exposure (PDE). Pregnant Wistar rats were injected subcutaneously with dexamethasone (0.2 mg/kg day) from gestational day 9 to 20. The F1 and F2 generations of male offspring were administered lithium pilocarpine (LiPC) for electroencephalography and video monitoring in epilepsy or behavioral tests. Results showed that the PDE + LiPC group exhibited TLE susceptibility, which continued throughout F2 generation. Expression of hippocampal glucocorticoid receptor (GR), CCAAT enhancer-binding protein α (C/EBPα), intrauterine renin-angiotensin system (RAS) classical pathway related genes, the H3K27ac level in angiotensin-converting enzyme (ACE) promoter, as well as high mobility group box 1 (HMGB1) and toll-like receptor 4 (TLR4) were increased, but glutamate dehydrogenase (GLUD) 1/2 expression were decreased, accompanied by increased glutamate levels in PDE fetal and adult rats, as well as in F1 and F2 offspring of the PDE + LiPC group. These consistent changes were also observed by treating the H19-7 fetal hippocampal cell line with dexamethasone and were reversed by GR inhibitor (RU486) and ACE inhibitor (enalaprilat). Our results confirmed that PDE-induced H3K27ac enrichment in the ACE promoter and enhanced the RAS classic pathway via activating GR-C/EBPα-p300 in utero, which caused changes of the HMGB1 pathway and glutamate excitatory damage. Intrauterine programming mediated by abnormal histone modification of hippocampal ACE could continue to adulthood and even F2 generation, which induced the heritability of TLE in male offspring rats.
Collapse
|
35
|
Patel NJ, Hogan KJ, Rizk E, Stewart K, Madrid A, Vadakkadath Meethal S, Alisch R, Borth L, Papale LA, Ondoma S, Gorges LR, Weber K, Lake W, Bauer A, Hariharan N, Kuehn T, Cook T, Keles S, Newton MA, Iskandar BJ. Ancestral Folate Promotes Neuronal Regeneration in Serial Generations of Progeny. Mol Neurobiol 2020; 57:2048-2071. [PMID: 31919777 PMCID: PMC7125003 DOI: 10.1007/s12035-019-01812-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
Folate supplementation in F0 mating rodents increases regeneration of injured spinal axons in vivo in 4 or more generations of progeny (F1-F4) in the absence of interval folate administration to the progeny. Transmission of the enhanced regeneration phenotype to untreated progeny parallels axonal growth in neuron culture after in vivo folate administration to the F0 ancestors alone, in correlation with differential patterns of genomic DNA methylation and RNA transcription in treated lineages. Enhanced axonal regeneration phenotypes are observed with diverse folate preparations and routes of administration, in outbred and inbred rodent strains, and in two rodent genera comprising rats and mice, and are reversed in F4-F5 progeny by pretreatment with DNA demethylating agents prior to phenotyping. Uniform transmission of the enhanced regeneration phenotype to progeny together with differential patterns of DNA methylation and RNA expression is consistent with a non-Mendelian mechanism. The capacity of an essential nutritional co-factor to induce a beneficial transgenerational phenotype in untreated offspring carries broad implications for the diagnosis, prevention, and treatment of inborn and acquired disorders.
Collapse
Affiliation(s)
- Nirav J Patel
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Kirk J Hogan
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA
| | - Elias Rizk
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Krista Stewart
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Andy Madrid
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Sivan Vadakkadath Meethal
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Reid Alisch
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Laura Borth
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Ligia A Papale
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Solomon Ondoma
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Logan R Gorges
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Kara Weber
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Wendell Lake
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Andrew Bauer
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Nithya Hariharan
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Thomas Kuehn
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA
| | - Thomas Cook
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Sunduz Keles
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
- Department of Statistics, University of Wisconsin, Madison, WI, USA
| | - Michael A Newton
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
- Department of Statistics, University of Wisconsin, Madison, WI, USA
| | - Bermans J Iskandar
- Department of Neurological Surgery, University of Wisconsin, 600 Highland Avenue, K4/832, Madison, WI, 53792, USA.
| |
Collapse
|
36
|
Jeremias G, Gonçalves FJM, Pereira JL, Asselman J. Prospects for incorporation of epigenetic biomarkers in human health and environmental risk assessment of chemicals. Biol Rev Camb Philos Soc 2020; 95:822-846. [PMID: 32045110 DOI: 10.1111/brv.12589] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
Abstract
Epigenetic mechanisms have gained relevance in human health and environmental studies, due to their pivotal role in disease, gene × environment interactions and adaptation to environmental change and/or contamination. Epigenetic mechanisms are highly responsive to external stimuli and a wide range of chemicals has been shown to determine specific epigenetic patterns in several organisms. Furthermore, the mitotic/meiotic inheritance of such epigenetic marks as well as the resulting changes in gene expression and cell/organismal phenotypes has now been demonstrated. Therefore, epigenetic signatures are interesting candidates for linking environmental exposures to disease as well as informing on past exposures to stressors. Accordingly, epigenetic biomarkers could be useful tools in both prospective and retrospective risk assessment but epigenetic endpoints are currently not yet incorporated into risk assessments. Achieving a better understanding on this apparent impasse, as well as identifying routes to promote the application of epigenetic biomarkers within environmental risk assessment frameworks are the objectives of this review. We first compile evidence from human health studies supporting the use of epigenetic exposure-associated changes as reliable biomarkers of exposure. Then, specifically focusing on environmental science, we examine the potential and challenges of developing epigenetic biomarkers for environmental fields, and discuss useful organisms and appropriate sequencing techniques to foster their development in this context. Finally, we discuss the practical incorporation of epigenetic biomarkers in the environmental risk assessment of chemicals, highlighting critical data gaps and making key recommendations for future research within a regulatory context.
Collapse
Affiliation(s)
- Guilherme Jeremias
- Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.,CESAM - Centre for Environmental and Marine Studies, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Fernando J M Gonçalves
- Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.,CESAM - Centre for Environmental and Marine Studies, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Joana L Pereira
- Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.,CESAM - Centre for Environmental and Marine Studies, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Jana Asselman
- Laboratory of Environmental Toxicology and Aquatic Ecology, Environmental Toxicology Unit - GhEnToxLab, Ghent University, 9000, Gent, Belgium
| |
Collapse
|
37
|
Ly L, Chan D, Landry M, Angle C, Martel J, Trasler J. Impact of mothers' early life exposure to low or high folate on progeny outcome and DNA methylation patterns. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa018. [PMID: 33240529 PMCID: PMC7673481 DOI: 10.1093/eep/dvaa018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/03/2020] [Accepted: 09/19/2020] [Indexed: 05/14/2023]
Abstract
The dynamic patterning of DNA and histone methylation during oocyte development presents a potentially susceptible time for epigenetic disruption due to early life environmental exposure of future mothers. We investigated whether maternal exposure to folic acid deficient and supplemented diets starting in utero could affect oocytes and cause adverse developmental and epigenetic effects in next generation progeny. Female BALB/c mice (F0) were placed on one of four amino acid defined diets for 4 weeks before pregnancy and throughout gestation and lactation: folic acid control (rodent recommended daily intake; Ctrl), 7-fold folic acid deficient, 10-fold folic acid supplemented or 20-fold folic acid supplemented diets. F1 female pups were weaned onto Ctrl diets, mated to produce the F2 generation and the F2 offspring were examined at E18.5 for developmental and epigenetic abnormalities. Resorption rates were increased and litter sizes decreased amongst F2 E18.5-day litters in the 20-fold folic acid supplemented group. Increases in abnormal embryo outcomes were observed in all three folic acid deficient and supplemented groups. Subtle genome-wide DNA methylation alterations were found in the placentas and brains of F2 offspring in the 7-fold folic acid deficient , 10-fold folic acid supplemented and 20-fold folic acid supplemented groups; in contrast, global and imprinted gene methylation were not affected. The findings show that early life female environmental exposures to both low and high folate prior to oocyte maturation can compromise oocyte quality, adversely affecting offspring of the next generation, in part by altering DNA methylation patterns.
Collapse
Affiliation(s)
- Lundi Ly
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Donovan Chan
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Mylène Landry
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Camille Angle
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Josée Martel
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jacquetta Trasler
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- Correspondence address. Research Institute of the McGill University Health Centre, 1001 Boulevard Décarie, Block E.M.0.3211, Montreal, QC, Canada H4A 3J1. Tel: +1-514-934-1934 (ext. 25235); Fax: +1-514-933-9673; E-mail:
| |
Collapse
|
38
|
Stoica V, Gardan DA, Constantinescu I, Gardan IP, Calenic B, Diculescu M. Transgenerational Effects of Traumatic Historical Events on the Incidence of Metabolic Syndrome/ Nonalcoholic Fatty Liver Disease in the Romanian Population. J Med Life 2020; 13:475-483. [PMID: 33456595 PMCID: PMC7803300 DOI: 10.25122/jml-2020-0156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Concerns for successful public health management are integrated into the core business of government-responsible institutions. Diseases associated with metabolic syndrome are very common in the Romanian population. In our study, we focused on the cardiovascular and non-alcoholic fatty liver disease (NAFLD). The article starts from the hypothesis that the increased incidence of such diseases is determined today by the cumulative effect of traumatic historical events such as the famine of 1946-47 and the communist political regime specific to the 80s and 90s. This study aims to present the arguments that indicate the correlation of economic variables whose variation can be easily determined by traumatic events that affected the economy, with variables able to measure the incidence of various diseases usually associated with metabolic syndrome or NAFLD. A series of statistical data were analyzed from the official sources available in the form of consecutive value data for the 1995-2018 period. The results highlighted a direct and strong link between the variable gross domestic product (GDP) per capita in USD, 2011 purchasing power parity (PPP) and specific incidence of circulatory, nutritional endocrine and metabolic diseases, as well as a strong and inverse link between GDP and infant's deaths per 1000 live births. Conclusions highlight that the effects of traumatic historical events must be made aware through medical education of the population, supporting the idea according to which the incidence of various metabolic diseases is greater for the offspring of those who have actively suffered during such events.
Collapse
Affiliation(s)
- Victor Stoica
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy”, Bucharest, Romania,Department of Gastroenterology, Fundeni Clinical Institute, Bucharest, Romania
| | - Daniel Adrian Gardan
- Faculty of Economic Sciences, Spiru Haret University, Bucharest, Romania,* Corresponding Author: Daniel Adrian Gardan,Faculty of Economic Sciences,Spiru Haret University Phone/fax: +40721108979 E-mail:
| | - Ileana Constantinescu
- Department of Immunology and Transplant Immunology, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania,Center of Imunogenetics and Virusology, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Bogdan Calenic
- Department of Biochemistry, Faculty of Dentistry, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Mircea Diculescu
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy”, Bucharest, Romania,Department of Gastroenterology, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
39
|
Roy S, Kumar V, Bossier P, Norouzitallab P, Vanrompay D. Phloroglucinol Treatment Induces Transgenerational Epigenetic Inherited Resistance Against Vibrio Infections and Thermal Stress in a Brine Shrimp ( Artemia franciscana) Model. Front Immunol 2019; 10:2745. [PMID: 31827471 PMCID: PMC6890837 DOI: 10.3389/fimmu.2019.02745] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/08/2019] [Indexed: 01/05/2023] Open
Abstract
Emerging, infectious diseases in shrimp like acute hepatopancreatic necrosis disease (AHPND) caused by Vibrio parahaemolyticus and mortality caused by other Vibrio species such as Vibrio harveyi are worldwide related to huge economic losses in industrial shrimp production. As a strategy to prevent disease outbreaks, a plant-based phenolic compound could be used as a biocontrol agent. Here, using the brine shrimp (Artemia franciscana) as a model system, we showed that phloroglucinol treatment of the parental animals at early life stages resulted in transgenerational inherited increased resistance in their progeny against biotic stress, i.e., bacteria (V. parahaemolyticus AHPND strain and V. harveyi) and abiotic stress, i.e., lethal heat shock. Increased resistance was recorded in three subsequent generations. Innate immune-related gene expression profiles and potential epigenetic mechanisms were studied to discover the underlying protective mechanisms. Our results showed that phloroglucinol treatment of the brine shrimp parents significantly (P < 0.05) enhanced the expression of a core set of innate immune genes (DSCAM, proPO, PXN, HSP90, HSP70, and LGBP) in subsequent generations. We also demonstrated that epigenetic mechanisms such as DNA methylation, m6A RNA methylation, and histone acetylation and methylation (active chromatin marker i.e., H3K4Me3, H3K4me1, H3K27me1, H3 hyperacetylation, H3K14ac and repression marker, i.e., H3K27me3, H4 hypoacetylation) might play a role in regulation of gene expression leading toward the observed transgenerational inheritance of the resistant brine shrimp progenies. To our knowledge, this is the first report on transgenerational inheritance of a compound-induced robust protected phenotype in brine shrimp, particularly protected against AHPND caused by V. parahaemolyticus and vibriosis caused by V. harveyi. Results showed that epigenetic reprogramming is likely to play a role in the underlying mechanism.
Collapse
Affiliation(s)
- Suvra Roy
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Laboratory of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- ICAR-Central Inland Fisheries Research Institute, Barrackpore, India
| | - Vikash Kumar
- Laboratory of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- ICAR-Central Inland Fisheries Research Institute, Barrackpore, India
| | - Peter Bossier
- Laboratory of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Parisa Norouzitallab
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Laboratory of Aquaculture & Artemia Reference Center, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Daisy Vanrompay
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
40
|
Wylde Z, Spagopoulou F, Hooper AK, Maklakov AA, Bonduriansky R. Parental breeding age effects on descendants' longevity interact over 2 generations in matrilines and patrilines. PLoS Biol 2019; 17:e3000556. [PMID: 31765371 PMCID: PMC6901263 DOI: 10.1371/journal.pbio.3000556] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/09/2019] [Accepted: 11/07/2019] [Indexed: 01/09/2023] Open
Abstract
Individuals within populations vary enormously in mortality risk and longevity, but the causes of this variation remain poorly understood. A potentially important and phylogenetically widespread source of such variation is maternal age at breeding, which typically has negative effects on offspring longevity. Here, we show that paternal age can affect offspring longevity as strongly as maternal age does and that breeding age effects can interact over 2 generations in both matrilines and patrilines. We manipulated maternal and paternal ages at breeding over 2 generations in the neriid fly Telostylinus angusticollis. To determine whether breeding age effects can be modulated by the environment, we also manipulated larval diet and male competitive environment in the first generation. We found separate and interactive effects of parental and grand-parental ages at breeding on descendants' mortality rate and life span in both matrilines and patrilines. These breeding age effects were not modulated by grand-parental larval diet quality or competitive environment. Our findings suggest that variation in maternal and paternal ages at breeding could contribute substantially to intrapopulation variation in mortality and longevity.
Collapse
Affiliation(s)
- Zachariah Wylde
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Foteini Spagopoulou
- Uppsala Centre for Evolution and Genomics, Uppsala University, Uppsala, Sweden
| | - Amy K. Hooper
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Alexei A. Maklakov
- Uppsala Centre for Evolution and Genomics, Uppsala University, Uppsala, Sweden
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Russell Bonduriansky
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
41
|
Post CM, Boule LA, Burke CG, O'Dell CT, Winans B, Lawrence BP. The Ancestral Environment Shapes Antiviral CD8 + T cell Responses across Generations. iScience 2019; 20:168-183. [PMID: 31569050 PMCID: PMC6817732 DOI: 10.1016/j.isci.2019.09.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/05/2019] [Accepted: 09/11/2019] [Indexed: 11/18/2022] Open
Abstract
Recent studies have linked health fates of children to environmental exposures of their great grandparents. However, few studies have considered whether ancestral exposures influence immune function across generations. Here, we report transgenerational inheritance of altered T cell responses resulting from maternal (F0) exposure to the aryl hydrocarbon receptor ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Since F0 exposure to TCDD has been linked to transgenerational transmission of reproductive problems, we asked whether maternal TCDD exposure also caused transgenerational changes in immune function. F0 exposure caused transgenerational effects on the CD8+ T cell response to influenza virus infection in females but not in males. Outcrosses showed changes were passed through both parental lineages. These data demonstrate that F0 exposure to an aryl hydrocarbon receptor (AHR) agonist causes durable changes to immune responses that can affect subsequent generations. This has broad implications for understanding how the environment of prior generations shapes susceptibility to pathogens and antiviral immunity in later generations.
Collapse
Affiliation(s)
- Christina M Post
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA
| | - Lisbeth A Boule
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA; Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA
| | - Catherine G Burke
- Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA
| | - Colleen T O'Dell
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA
| | - Bethany Winans
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA; Department of Microbiology & Immunology, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
42
|
Santangeli S, Consales C, Pacchierotti F, Habibi HR, Carnevali O. Transgenerational effects of BPA on female reproduction. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 685:1294-1305. [PMID: 31272786 DOI: 10.1016/j.scitotenv.2019.06.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/31/2019] [Accepted: 06/02/2019] [Indexed: 06/09/2023]
Abstract
Bisphenol A (BPA) is an abundant environmental contaminant and studies have shown the presence of BPA in the urine of over 90% of population tested in Canada and USA. In addition to its reported harmful effects, there is concern for its transgenerational effects. For a compound to induce transgenerational effect, an epigenetic mark should be mitotically and meiotically stable without reprogramming in primordial germ cells and post fertilization embryos. In the present study, female zebrafish were treated with an environmental dose (20 μg/L) of BPA and then crossed with untreated males. To assess epigenetic effects, transcript levels of several genes involved in female reproduction were measured in adult and in 24 hpf embryos up to F3 generation. Exposure to BPA affected adult female fertility up to F2 generation. In F0, F1 and F2 ovaries transcript levels for several genes involved in reproduction, including esr, star, lhcgr and fshr were affected. To investigate epigenetic mechanisms of gene expression modulation, we studied promoter DNA methylation. Among genes involved in gonadal differentiation, amh transcript level was reduced in 24 hpf embryos, up to the F3 generation. Variation in amh transcript level was associated with hyper-methylation of its promoter and changes in H3K4me3/H3K27me3 enrichment, coherent with gene silencing. The findings provide evidence for transgenerational effects of BPA in zebrafish and demonstrate that amh is susceptible to stable epigenetic alterations. CAPSULE: Transgenerational effects of BPA on female reproductive physiology.
Collapse
Affiliation(s)
- Stefania Santangeli
- Dipartimento Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy.
| | - Claudia Consales
- Laboratory of Biosafety and Risk Assessment, Division of Health Protection Technologies, ENEA Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy.
| | - Francesca Pacchierotti
- Laboratory of Biosafety and Risk Assessment, Division of Health Protection Technologies, ENEA Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy.
| | - Hamid R Habibi
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada.
| | - Oliana Carnevali
- Dipartimento Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy; INBB Consorzio Interuniversitario di Biosistemi e Biostrutture, 00136 Roma, Italy.
| |
Collapse
|
43
|
Belleau P, Deschênes A, Scott-Boyer MP, Lambrot R, Dalvai M, Kimmins S, Bailey J, Droit A. Inferring and modeling inheritance of differentially methylated changes across multiple generations. Nucleic Acids Res 2019; 46:e85. [PMID: 29750268 PMCID: PMC6101575 DOI: 10.1093/nar/gky362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/24/2018] [Indexed: 01/01/2023] Open
Abstract
High-throughput methylation sequencing enables genome-wide detection of differentially methylated sites (DMS) or regions (DMR). Increasing evidence suggests that treatment-induced DMS can be transmitted across generations, but the analysis of induced methylation changes across multiple generations is complicated by the lack of sound statistical methods to evaluate significance levels. Due to software design, DMS detection was usually made on each generation separately, thus disregarding stochastic effects expected when a large number of DMS is detected in each generation. Here, we present a novel method based on Monte Carlo sampling, methylInheritance, to evaluate that the number of conserved DMS between several generations is associated to an effect inherited from a treatment and not randomness. Moreover, we developed an inheritance simulation package, methInheritSim, to demonstrate the performance of the methylInheritance method and to evaluate the power of different experimental designs. Finally, we applied methylInheritance to a DNA methylation dataset obtained from early-life persistent organic pollutants (POPs) exposed Sprague-Dawley female rats and their descendants through a paternal transmission. The results show that metylInheritance can efficiently identify treatment-induced inherited methylation changes. Specifically, we identified two intergenerationally conserved DMS at transcription start site (TSS); one of those persisted transgenerationally. Three transgenerationally conserved DMR were found at intra or integenic regions.
Collapse
Affiliation(s)
- Pascal Belleau
- Département de Médecine Moléculaire - Université Laval, Faculté de médecine, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, bureau 4633, Québec, QC G1V 0A6, Canada
| | - Astrid Deschênes
- Centre de Recherche du CHU de Québec - Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Marie-Pier Scott-Boyer
- Centre de Recherche du CHU de Québec - Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Romain Lambrot
- Department of Animal Sciences, McGill University, Ste. Anne de Bellevue, Quebec, H9 × 3V9 Canada and Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Mathieu Dalvai
- Centre de recherche en reproduction, développement et santé intergénérationnelle - Université Laval, Faculté des sciences de l'agriculture et de l'alimentation, Pavillon Paul-Comtois, 2425 rue de l'Agriculture, Québec, QC G1V 0A6, Canada
| | - Sarah Kimmins
- Department of Animal Sciences, McGill University, Ste. Anne de Bellevue, Quebec, H9 × 3V9 Canada and Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Janice Bailey
- Centre de recherche en reproduction, développement et santé intergénérationnelle - Université Laval, Faculté des sciences de l'agriculture et de l'alimentation, Pavillon Paul-Comtois, 2425 rue de l'Agriculture, Québec, QC G1V 0A6, Canada
| | - Arnaud Droit
- Département de Médecine Moléculaire - Université Laval, Faculté de médecine, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, bureau 4633, Québec, QC G1V 0A6, Canada.,Centre de Recherche du CHU de Québec - Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| |
Collapse
|
44
|
Marin P, Genitoni J, Barloy D, Maury S, Gibert P, Ghalambor CK, Vieira C. Biological invasion: The influence of the hidden side of the (epi)genome. Funct Ecol 2019. [DOI: 10.1111/1365-2435.13317] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pierre Marin
- Laboratoire de Biométrie et Biologie Evolutive UMR 5558, CNRS, Université Lyon 1 Université de Lyon Villeurbanne France
| | - Julien Genitoni
- ESE, Ecology and Ecosystem Health, Agrocampus Ouest INRA Rennes France
- LBLGC EA 1207 INRA, Université d'Orléans, USC 1328 Orléans France
| | - Dominique Barloy
- ESE, Ecology and Ecosystem Health, Agrocampus Ouest INRA Rennes France
| | - Stéphane Maury
- LBLGC EA 1207 INRA, Université d'Orléans, USC 1328 Orléans France
| | - Patricia Gibert
- Laboratoire de Biométrie et Biologie Evolutive UMR 5558, CNRS, Université Lyon 1 Université de Lyon Villeurbanne France
| | - Cameron K. Ghalambor
- Department of Biology and Graduate Degree Program in Ecology Colorado State University Fort Collins Colorado
| | - Cristina Vieira
- Laboratoire de Biométrie et Biologie Evolutive UMR 5558, CNRS, Université Lyon 1 Université de Lyon Villeurbanne France
| |
Collapse
|
45
|
Guida MC, Birse RT, Dall'Agnese A, Toto PC, Diop SB, Mai A, Adams PD, Puri PL, Bodmer R. Intergenerational inheritance of high fat diet-induced cardiac lipotoxicity in Drosophila. Nat Commun 2019; 10:193. [PMID: 30643137 PMCID: PMC6331650 DOI: 10.1038/s41467-018-08128-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/17/2018] [Indexed: 12/26/2022] Open
Abstract
Obesity is strongly correlated with lipotoxic cardiomyopathy, heart failure and thus mortality. The incidence of obesity has reached alarming proportions worldwide, and increasing evidence suggests that the parents' nutritional status may predispose their offspring to lipotoxic cardiomyopathy. However, to date, mechanisms underlying intergenerational heart disease risks have yet to be elucidated. Here we report that cardiac dysfunction induced by high-fat-diet (HFD) persists for two subsequent generations in Drosophila and is associated with reduced expression of two key metabolic regulators, adipose triglyceride lipase (ATGL/bmm) and transcriptional cofactor PGC-1. We provide evidence that targeted expression of ATGL/bmm in the offspring of HFD-fed parents protects them, and the subsequent generation, from cardio-lipotoxicity. Furthermore, we find that intergenerational inheritance of lipotoxic cardiomyopathy correlates with elevated systemic H3K27 trimethylation. Lowering H3K27 trimethylation genetically or pharmacologically in the offspring of HFD-fed parents prevents cardiac pathology. This suggests that metabolic homeostasis is epigenetically regulated across generations.
Collapse
Affiliation(s)
- Maria Clara Guida
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Ryan Tyge Birse
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- Biocompatibles Inc., 300 Four Falls Corporate Center, 300 Conshohocken State Road, West Conshohocken, PA, 19428-2998, USA
| | - Alessandra Dall'Agnese
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA, 02142, USA
| | - Paula Coutinho Toto
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Soda Balla Diop
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | | | - Peter D Adams
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- IRCCS Fondazione Santa Lucia, 00142, Rome, Italy
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
46
|
Sato A. Chaperones, Canalization, and Evolution of Animal Forms. Int J Mol Sci 2018; 19:E3029. [PMID: 30287767 PMCID: PMC6213012 DOI: 10.3390/ijms19103029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/18/2022] Open
Abstract
Over half a century ago, British developmental biologist Conrad Hal Waddington proposed the idea of canalization, that is, homeostasis in development. Since the breakthrough that was made by Rutherford and Lindquist (1998), who proposed a role of Hsp90 in developmental buffering, chaperones have gained much attention in the study of canalization. However, recent studies have revealed that a number of other molecules are also potentially involved in canalization. Here, I introduce the emerging role of DnaJ chaperones in canalization. I also discuss how the expression levels of such buffering molecules can be altered, thereby altering organismal development. Since developmental robustness is maternally inherited in various organisms, I propose that dynamic bet hedging, an increase in within-clutch variation in offspring phenotypes that is caused by unpredictable environmental challenges to the mothers, plays a key role in altering the expression levels of buffering molecules. Investigating dynamic bet hedging at the molecular level and how it impacts upon morphological phenotypes will help our understanding of the molecular mechanisms of canalization and evolutionary processes.
Collapse
Affiliation(s)
- Atsuko Sato
- Department of Biology, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-0012, Japan.
- Marine Biological Association of the UK, The Laboratory, Plymouth PL1 2PB, UK.
| |
Collapse
|
47
|
Potabattula R, Dittrich M, Böck J, Haertle L, Müller T, Hahn T, Schorsch M, Hajj NE, Haaf T. Allele-specific methylation of imprinted genes in fetal cord blood is influenced by cis-acting genetic variants and parental factors. Epigenomics 2018; 10:1315-1326. [PMID: 30238782 PMCID: PMC6240887 DOI: 10.2217/epi-2018-0059] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To examine the effects of genetic variation, parental age and BMI on parental allele-specific methylation of imprinted genes in fetal cord blood samples. Methodology: We have developed SNP genotyping and deep bisulphite sequencing assays for six imprinted genes to determine parental allele-specific methylation patterns in diploid somatic tissues. Results: Multivariate linear regression analyses revealed a negative correlation of paternal age with paternal MEG3 allele methylation in fetal cord blood. Methylation of the maternal PEG3 allele showed a positive correlation with maternal age. Paternal BMI was positively correlated with paternal MEST allele methylation. In addition to parental origin, allele-specific methylation of most imprinted genes was largely dependent on the underlying SNP haplotype. Conclusion: Our study supports the idea that parental factors can have an impact, although of small effect size, on the epigenome of the next generation, providing an additional layer of complexity to phenotypic diversity.
Collapse
Affiliation(s)
- Ramya Potabattula
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Marcus Dittrich
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany.,Department of Bioinformatics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Julia Böck
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Larissa Haertle
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Tobias Müller
- Department of Bioinformatics, Julius Maximilians University, 97074 Würzburg, Germany
| | | | | | - Nady El Hajj
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, 97074 Würzburg, Germany
| |
Collapse
|
48
|
Chiapperino L, Panese F. Gendered imaginaries: situating knowledge of epigenetic programming of health. SOCIOLOGY OF HEALTH & ILLNESS 2018; 40:1233-1249. [PMID: 30066339 DOI: 10.1111/1467-9566.12779] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Our paper explores the value-laden and epistemic resources that scientists working in epigenetics and developmental programming of health and disease (DOHaD) mobilise to produce scientific representations of pregnancy and parenthood, which in turn imagine norms, values, and responsibilities for the protection of future generations. In order to do so, we first describe the place of questions regarding the relative weight of paternal and maternal influences on the health of the offspring in the discursive formalisation of this research in scientific publications. This enables us to identify the mutual constitution of 'prototypes' (i.e. experimental designs, settings, techniques) and 'stereotypes' (i.e. social meanings, beliefs, norms and values) of parental roles in DOHaD and epigenetic biomedical sciences, by means of a specific gendered figuration of paternal influences: the 'father-as-sperm'. Second, and drawing from a set of interviews (N = 15), we describe a tension between this dominant, objectifying molecular discourse and the perspective of individual scientists. The situated perspective of individual researchers provides in fact evidence for a conflictual (moral and epistemic) economy of gendered engagements with parental figurations in DOHaD and epigenetic research, and consequently suggests a more fine-grained, as well as conflictual web of socio-political positioning of this 'knowledge' in its societal circulation.
Collapse
Affiliation(s)
- Luca Chiapperino
- Faculty of Social and Political Sciences, University of Lausanne, Switzerland
| | - Francesco Panese
- Faculty of Social and Political Sciences, University of Lausanne, Switzerland
- Institute of the Humanities in Medicine (IHM), Lausanne University Hospital (CHUV), Switzerland
| |
Collapse
|
49
|
Padmanabhan N, Menelaou K, Gao J, Anderson A, Blake GET, Li T, Daw BN, Watson ED. Abnormal folate metabolism causes age-, sex- and parent-of-origin-specific haematological defects in mice. J Physiol 2018; 596:4341-4360. [PMID: 30024025 PMCID: PMC6138292 DOI: 10.1113/jp276419] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/27/2018] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS Folate (folic acid) deficiency and mutations in folate-related genes in humans result in megaloblastic anaemia. Folate metabolism, which requires the enzyme methionine synthase reductase (MTRR), is necessary for DNA synthesis and the transmission of one-carbon methyl groups for cellular methylation. In this study, we show that the hypomorphic Mtrrgt/gt mutation in mice results in late-onset and sex-specific blood defects, including macrocytic anaemia, extramedullary haematopoiesis and lymphopenia. Notably, when either parent carries an Mtrrgt allele, blood phenotypes result in their genetically wildtype adult daughters, the effects of which are parent specific. Our data establish a new model for studying the mechanism of folate metabolism in macrocytic anaemia aetiology and suggest that assessing parental folate status might be important when diagnosing adult patients with unexplained anaemia. ABSTRACT The importance of the vitamin folate (also known as folic acid) in erythrocyte formation, maturation and/or longevity is apparent since folate deficiency in humans causes megaloblastic anaemia. Megaloblastic anaemia is a type of macrocytic anaemia whereby erythrocytes are enlarged and fewer in number. Folate metabolism is required for thymidine synthesis and one-carbon metabolism, though its specific role in erythropoiesis is not well understood. Methionine synthase reductase (MTRR) is a key enzyme necessary for the progression of folate metabolism since knocking down the Mtrr gene in mice results in hyperhomocysteinaemia and global DNA hypomethylation. We demonstrate here that abnormal folate metabolism in mice caused by Mtrrgt/gt homozygosity leads to haematopoietic phenotypes that are sex and age dependent. Specifically, Mtrrgt/gt female mice displayed macrocytic anaemia, which might be due to defective erythroid differentiation at the exclusion of haemolysis. This was associated with increased renal Epo mRNA expression, hypercellular bone marrow, and splenic extramedullary haematopoiesis. In contrast, the male response differed since Mtrrgt/gt male mice were not anaemic but did display erythrocytic macrocytosis and lymphopenia. Regardless of sex, these phenotypes were late onset. Remarkably, we also show that when either parent carries an Mtrrgt allele, a haematological defect results in their adult wildtype daughters. However, the specific phenotype was dependent upon the sex of the parent. For instance, wildtype daughters of Mtrr+/gt females displayed normocytic anaemia. In contrast, wildtype daughters of Mtrr+/gt males exhibited erythrocytic microcytosis not associated with anaemia. Therefore, abnormal folate metabolism affects adult haematopoiesis in an age-, sex- and parent-specific manner.
Collapse
Affiliation(s)
- Nisha Padmanabhan
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Katerina Menelaou
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Jiali Gao
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Alexander Anderson
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Georgina E. T. Blake
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Tanya Li
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - B. Nuala Daw
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Erica D. Watson
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| |
Collapse
|
50
|
Moggs J, Terranova R. Chromatin dynamics underlying latent responses to xenobiotics. Toxicol Res (Camb) 2018; 7:606-617. [PMID: 30090610 PMCID: PMC6062062 DOI: 10.1039/c7tx00317j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/26/2018] [Indexed: 11/21/2022] Open
Abstract
Pleiotropic xenobiotics can trigger dynamic alterations in mammalian chromatin structure and function but many of these are likely non-adverse and simply reflect short-term changes in DNA transactions underlying normal homeostatic, adaptive and protective cellular responses. However, it is plausible that a subset of xenobiotic-induced perturbations of somatic tissue or germline epigenomes result in delayed-onset and long-lasting adverse effects, in particular if they occur during critical stages of growth and development. These could include reprogramming, dedifferentiation, uncontrolled growth, and cumulative toxicity effects through molecular memory of prior xenobiotic exposures or altered susceptibility to subsequent xenobiotic exposures. Here we discuss the current evidence for epigenetic mechanisms underlying latent responses to xenobiotics, and the potential for identifying molecular epigenetic changes that are prodromal to overt morphologic or functional toxicity phenotypes.
Collapse
Affiliation(s)
- Jonathan Moggs
- Preclinical Safety , Translational Medicine , Novartis Institutes for BioMedical Research , Basel , Switzerland
| | - Rémi Terranova
- Preclinical Safety , Translational Medicine , Novartis Institutes for BioMedical Research , Basel , Switzerland
| |
Collapse
|