1
|
Richards-Steed R, Wan N, Bakian A, Medina RM, Brewer SC, Smith KR, VanDerslice JA. Observational methods for human studies of transgenerational effects. Epigenetics 2024; 19:2366065. [PMID: 38870389 PMCID: PMC11178273 DOI: 10.1080/15592294.2024.2366065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024] Open
Abstract
There are substantial challenges in studying human transgenerational epigenetic outcomes resulting from environmental conditions. The task requires specialized methods and tools that incorporate specific knowledge of multigenerational relationship combinations of probands and their ancestors, phenotype data for individuals, environmental information of ancestors and their descendants, which can span historical to present datasets, and informative environmental data that chronologically aligns with ancestors and descendants over space and time. As a result, there are few epidemiologic studies of potential transgenerational effects in human populations, thus limiting the knowledge of ancestral environmental conditions and the potential impacts we face with modern human health outcomes. In an effort to overcome some of the challenges in studying human transgenerational effects, we present two transgenerational study designs: transgenerational space-time cluster detection and transgenerational case-control study design. Like other epidemiological methods, these methods determine whether there are statistical associations between phenotypic outcomes (e.g., adverse health outcomes) among probands and the shared environments and environmental factors facing their ancestors. When the ancestor is a paternal grandparent, a statistically significant association provides some evidence that a transgenerational inheritable factor may be involved. Such results may generate useful hypotheses that can be explored using epigenomic data to establish conclusive evidence of transgenerational heritable effects. Both methods are proband-centric: They are designed around the phenotype of interest in the proband generation for case selection and family pedigree creation. In the examples provided, we incorporate at least three generations of paternal lineage in both methods to observe a potential transgenerational effect.
Collapse
Affiliation(s)
| | - Neng Wan
- Geography, University of Utah Department of Geography, Salt Lake City, UT, USA
| | - Amanda Bakian
- Psychiatry, University of Utah Health, Salt Lake City, UT, USA
| | - Richard M. Medina
- Geography, University of Utah Department of Geography, Salt Lake City, UT, USA
| | - Simon C. Brewer
- Geography, University of Utah Department of Geography, Salt Lake City, UT, USA
| | - Ken R. Smith
- Child and Consumer Studies, University of Utah Health, Salt Lake City, UT, USA
| | | |
Collapse
|
2
|
Champroux A, Tang Y, Dickson DA, Meng A, Harrington A, Liaw L, Marzi M, Nicassio F, Schlaeger TM, Feig LA. Transmission of reduced levels of miR-34/449 from sperm to preimplantation embryos is a key step in the transgenerational epigenetic inheritance of the effects of paternal chronic social instability stress. Epigenetics 2024; 19:2346694. [PMID: 38739481 PMCID: PMC11093028 DOI: 10.1080/15592294.2024.2346694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
The transgenerational effects of exposing male mice to chronic social instability (CSI) stress are associated with decreased sperm levels of multiple members of the miR-34/449 family that persist after their mating through preimplantation embryo (PIE) development. Here we demonstrate the importance of these miRNA changes by showing that restoring miR-34c levels in PIEs derived from CSI stressed males prevents elevated anxiety and defective sociability normally found specifically in their adult female offspring. It also restores, at least partially, levels of sperm miR-34/449 normally reduced in their male offspring who transmit these sex-specific traits to their offspring. Strikingly, these experiments also revealed that inducing miR-34c levels in PIEs enhances the expression of its own gene and that of miR-449 in these cells. The same induction of embryo miR-34/449 gene expression likely occurs after sperm-derived miR-34c is introduced into oocytes upon fertilization. Thus, suppression of this miRNA amplification system when sperm miR-34c levels are reduced in CSI stressed mice can explain how a comparable fold-suppression of miR-34/449 levels can be found in PIEs derived from them, despite sperm containing ~50-fold lower levels of these miRNAs than those already present in PIEs. We previously found that men exposed to early life trauma also display reduced sperm levels of miR-34/449. And here we show that miR-34c can also increase the expression of its own gene, and that of miR-449 in human embryonic stem cells, suggesting that human PIEs derived from men with low sperm miR-34/449 levels may also contain this potentially harmful defect.
Collapse
Affiliation(s)
- Alexandre Champroux
- Development, Molecular & Chemical Biology/Medical, Tufts University, Boston, MA, USA
| | - Yang Tang
- Stem Cell Program, Boston Children’s Hospital, Boston, MA, USA
| | - David A. Dickson
- Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Alice Meng
- Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Anne Harrington
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | - Lucy Liaw
- Center for Genomic Studies, Instituto Italiano di Tecnologia Institution, Milan, Italy
| | - Matteo Marzi
- Center for Genomic Studies, Instituto Italiano di Tecnologia Institution, Milan, Italy
| | - Francesco Nicassio
- Center for Genomic Studies, Instituto Italiano di Tecnologia Institution, Milan, Italy
| | | | - Larry A. Feig
- Development, Molecular & Chemical Biology/Medical, Tufts University, Boston, MA, USA
- Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
3
|
Khalid S, Kearney M, McReynolds DE. Can social adversity alter the epigenome, trigger oral disease, and affect future generations? Ir J Med Sci 2024; 193:2597-2606. [PMID: 38740675 PMCID: PMC11450135 DOI: 10.1007/s11845-024-03697-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024]
Abstract
The nature versus nurture debate has intrigued scientific circles for decades. Although extensive research has established a clear relationship between genetics and disease development, recent evidence has highlighted the insufficiency of attributing adverse health outcomes to genetic factors alone. In fact, it has been suggested that environmental influences, such as socioeconomic position (SEP), may play a much larger role in the development of disease than previously thought, with extensive research suggesting that low SEP is associated with adverse health conditions. In relation to oral health, a higher prevalence of caries (tooth decay) exists among those of low SEP. Although little is known about the biological mechanisms underlying this relationship, epigenetic modifications resulting from environmental influences have been suggested to play an important role. This review explores the intersection of health inequalities and epigenetics, the role of early-life social adversity and its long-term epigenetic impacts, and how those living within the lower hierarchies of the socioeconomic pyramid are indeed at higher risk of developing diseases, particularly in relation to oral health. A deeper understanding of these mechanisms could lead to the development of targeted interventions for individuals of low SEP to improve oral health or identify those who are at higher risk of developing oral disease.
Collapse
Affiliation(s)
- Sakr Khalid
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Michaela Kearney
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - David E McReynolds
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
4
|
Champroux A, Sadat-Shirazi M, Chen X, Hacker J, Yang Y, Feig LA. Astrocyte-Derived Exosomes Regulate Sperm miR-34c Levels to Mediate the Transgenerational Effects of Paternal Chronic Social Instability Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.21.537854. [PMID: 37786715 PMCID: PMC10541588 DOI: 10.1101/2023.04.21.537854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
The effects of chronically stressing male mice can be transmitted across generations by stress-specific changes in their sperm miRNA content that induce stress-specific phenotypes in their offspring. But how each stress paradigm alters the levels of distinct sets of sperm miRNAs is not known. We showed previously that exposure of male mice to chronic social instability (CSI) stress results in elevated anxiety and reduced sociability specifically in their female offspring across multiple generations because it reduces miR-34c levels in sperm of stressed males and their unstressed male offspring. Here we describe evidence that a strocyte-derived exos omes ( A-Exos ) carrying miR-34c mediate how CSI stress has this transgenerational effect on sperm. We found that CSI stress decreases miR-34c carried by A-Exos in the prefrontal cortex and amygdala, as well as in the blood of males. Importantly, miR-34c A-Exos levels are also reduced in these tissues in their F1 male offspring, who despite not being exposed to stress exhibit reduced sperm miR-34c levels and transmit the same stress-associated traits to their male and female offspring. Furthermore, restoring A-Exos miR-34c content in the blood of CSI-stressed males by intravenous injection of miR-34c-containing A-Exos restores miR-34c levels in their sperm. These findings reveal an unexpected role for A-Exos in maintaining sperm miR-34c levels by a process that when suppressed by CSI stress mediates this example of transgenerational epigenetic inheritance.
Collapse
|
5
|
Freire T, Pulpitel T, Clark X, Mackay F, Raubenheimer D, Simpson SJ, Solon-Biet SM, Crean AJ. The effects of paternal dietary fat versus sugar on offspring body composition and anxiety-related behavior. Physiol Behav 2024; 279:114533. [PMID: 38552707 DOI: 10.1016/j.physbeh.2024.114533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 02/26/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Increasing evidence suggests that the pre-conception parental environment has long-term consequences for offspring health and disease susceptibility. Though much of the work in this field concentrates on maternal influences, there is growing understanding that fathers also play a significant role in affecting offspring phenotypes. In this study, we investigate effects of altering the proportion of dietary fats and carbohydrates on paternal and offspring body composition and anxiety-related behavior in C57Bl/6-JArc mice. We show that in an isocaloric context, greater dietary fat increased body fat and reduced anxiety-like behavior of studs, whereas increased dietary sucrose had no significant effect. These dietary effects were not reflected in offspring traits, rather, we found sex-specific effects that differed between offspring body composition and behavioral traits. This finding is consistent with past paternal effect studies, where transgenerational effects have been shown to be more prominent in one sex over the other. Here, male offspring of fathers fed high-fat diets were heavier at 10 weeks of age due to increased lean body mass, whereas paternal diet had no significant effect on female offspring body fat or lean mass. In contrast, paternal dietary sugar appeared to have the strongest effects on male offspring behavior, with male offspring of high-sucrose fathers spending less time in the closed arms of the elevated plus maze. Both high-fat and high-sugar paternal diets were found to reduce anxiety-like behavior of female offspring, although this effect was only evident when offspring were fed a control diet. This study provides new understanding of the ways in which diet can shape the behavior of fathers and their offspring and contribute to the development of dietary guidelines to improve obesity and mental health conditions, such as anxiety.
Collapse
Affiliation(s)
- Therese Freire
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney NSW, Australia.
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Ximonie Clark
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - Flora Mackay
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| | - Angela J Crean
- Charles Perkins Centre, The University of Sydney NSW, Australia; School of Life and Environmental Sciences, Faculty of Science, The University of Sydney NSW, Australia
| |
Collapse
|
6
|
Costa DL. Grandchildren's Longevity and Their Grandfathers' POW Trauma in the U.S. Civil War. Demography 2024; 61:337-361. [PMID: 38393987 DOI: 10.1215/00703370-11191183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
I document the transmission of a grandfather's net nutritional deprivation and psychosocial stress in young adulthood across multiple generations using the grandfather's ex-prisoner of war (ex-POW) status in the U.S. Civil War (1861-1865). Using a newly created dataset, I uncover an association between a grandfather's ex-POW status and the longevity after age 45 of his sons and male-line grandsons but not of his daughters, granddaughters, female-line grandsons, children-in-law, or grandchildren-in-law. Male-line grandsons lost roughly a year of life at age 45 (4% of remaining life expectancy) if descended from ex-POWs who suffered severe captivity conditions than if descended from non-POWs. If their grandfathers faced a less harsh captivity, male-line grandsons lost less than a year of life compared with those descended from non-POWs. I find that the grandfather's age at exposure and the grandson's education, as well as the son's and the grandson's poor late gestational conditions (proxied by season of birth), mediate this relationship. I rule out socioeconomic status, marriage and mortality selection, and cultural or psychological transmission from grandfathers to grandsons as explanations. I cannot rule out an epigenetic explanation.
Collapse
Affiliation(s)
- Dora L Costa
- Department of Economics, University of California, Los Angeles, Los Angeles, CA, USA
- National Bureau of Economic Research, Cambridge, MA, USA
| |
Collapse
|
7
|
Yaremenko AV, Pechnikova NA, Porpodis K, Damdoumis S, Aggeli A, Theodora P, Domvri K. Association of Fetal Lung Development Disorders with Adult Diseases: A Comprehensive Review. J Pers Med 2024; 14:368. [PMID: 38672994 PMCID: PMC11051200 DOI: 10.3390/jpm14040368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Fetal lung development is a crucial and complex process that lays the groundwork for postnatal respiratory health. However, disruptions in this delicate developmental journey can lead to fetal lung development disorders, impacting neonatal outcomes and potentially influencing health outcomes well into adulthood. Recent research has shed light on the intriguing association between fetal lung development disorders and the development of adult diseases. Understanding these links can provide valuable insights into the developmental origins of health and disease, paving the way for targeted preventive measures and clinical interventions. This review article aims to comprehensively explore the association of fetal lung development disorders with adult diseases. We delve into the stages of fetal lung development, examining key factors influencing fetal lung maturation. Subsequently, we investigate specific fetal lung development disorders, such as respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), congenital diaphragmatic hernia (CDH), and other abnormalities. Furthermore, we explore the potential mechanisms underlying these associations, considering the role of epigenetic modifications, transgenerational effects, and intrauterine environmental factors. Additionally, we examine the epidemiological evidence and clinical findings linking fetal lung development disorders to adult respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), and other respiratory ailments. This review provides valuable insights for healthcare professionals and researchers, guiding future investigations and shaping strategies for preventive interventions and long-term care.
Collapse
Affiliation(s)
- Alexey V. Yaremenko
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
| | - Nadezhda A. Pechnikova
- Laboratory of Chemical Engineering A’, School of Chemical Engineering, Faculty of Engineering, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (N.A.P.); (A.A.)
- Saint Petersburg Pasteur Institute, Saint Petersburg 197101, Russia
| | - Konstantinos Porpodis
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
| | - Savvas Damdoumis
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
| | - Amalia Aggeli
- Laboratory of Chemical Engineering A’, School of Chemical Engineering, Faculty of Engineering, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (N.A.P.); (A.A.)
| | - Papamitsou Theodora
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Kalliopi Domvri
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
- Pathology Department, George Papanikolaou Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| |
Collapse
|
8
|
Rice RC, Gil DV, Baratta AM, Frawley RR, Hill SY, Farris SP, Homanics GE. Inter- and transgenerational heritability of preconception chronic stress or alcohol exposure: Translational outcomes in brain and behavior. Neurobiol Stress 2024; 29:100603. [PMID: 38234394 PMCID: PMC10792982 DOI: 10.1016/j.ynstr.2023.100603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/19/2024] Open
Abstract
Chronic stress and alcohol (ethanol) use are highly interrelated and can change an individual's behavior through molecular adaptations that do not change the DNA sequence, but instead change gene expression. A recent wealth of research has found that these nongenomic changes can be transmitted across generations, which could partially account for the "missing heritability" observed in genome-wide association studies of alcohol use disorder and other stress-related neuropsychiatric disorders. In this review, we summarize the molecular and behavioral outcomes of nongenomic inheritance of chronic stress and ethanol exposure and the germline mechanisms that could give rise to this heritability. In doing so, we outline the need for further research to: (1) Investigate individual germline mechanisms of paternal, maternal, and biparental nongenomic chronic stress- and ethanol-related inheritance; (2) Synthesize and dissect cross-generational chronic stress and ethanol exposure; (3) Determine cross-generational molecular outcomes of preconception ethanol exposure that contribute to alcohol-related disease risk, using cancer as an example. A detailed understanding of the cross-generational nongenomic effects of stress and/or ethanol will yield novel insight into the impact of ancestral perturbations on disease risk across generations and uncover actionable targets to improve human health.
Collapse
Affiliation(s)
- Rachel C. Rice
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniela V. Gil
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Annalisa M. Baratta
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Remy R. Frawley
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shirley Y. Hill
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sean P. Farris
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gregg E. Homanics
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Costa DL, Lewis C, Yetter N. Children and Grandchildren of Union Army Veterans: New Data Collections to Study the Persistence of Longevity and Socioeconomic Status Across Generations. HISTORICAL METHODS 2024; 56:223-239. [PMID: 38742179 PMCID: PMC11090407 DOI: 10.1080/01615440.2023.2301578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
This paper introduces four new intergenerational and multigenerational datasets which follow both sons and daughters and which can be used to study the persistence of longevity, socioeconomic status, family structure, and geographic mobility across generations. The data follow the children of Black and White Union Army (US Civil War, 1861-5) veterans from birth to death, linking them to the available censuses. The White samples include an over-sample of children of ex-POWs. A separate collection links grandchildren of White Union Army veterans to their death records. The data were created with high quality manual linkage procedures utilizing a wide variety of records to establish links.
Collapse
|
10
|
Kimmins S, Anderson RA, Barratt CLR, Behre HM, Catford SR, De Jonge CJ, Delbes G, Eisenberg ML, Garrido N, Houston BJ, Jørgensen N, Krausz C, Lismer A, McLachlan RI, Minhas S, Moss T, Pacey A, Priskorn L, Schlatt S, Trasler J, Trasande L, Tüttelmann F, Vazquez-Levin MH, Veltman JA, Zhang F, O'Bryan MK. Frequency, morbidity and equity - the case for increased research on male fertility. Nat Rev Urol 2024; 21:102-124. [PMID: 37828407 DOI: 10.1038/s41585-023-00820-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/14/2023]
Abstract
Currently, most men with infertility cannot be given an aetiology, which reflects a lack of knowledge around gamete production and how it is affected by genetics and the environment. A failure to recognize the burden of male infertility and its potential as a biomarker for systemic illness exists. The absence of such knowledge results in patients generally being treated as a uniform group, for whom the strategy is to bypass the causality using medically assisted reproduction (MAR) techniques. In doing so, opportunities to prevent co-morbidity are missed and the burden of MAR is shifted to the woman. To advance understanding of men's reproductive health, longitudinal and multi-national centres for data and sample collection are essential. Such programmes must enable an integrated view of the consequences of genetics, epigenetics and environmental factors on fertility and offspring health. Definition and possible amelioration of the consequences of MAR for conceived children are needed. Inherent in this statement is the necessity to promote fertility restoration and/or use the least invasive MAR strategy available. To achieve this aim, protocols must be rigorously tested and the move towards personalized medicine encouraged. Equally, education of the public, governments and clinicians on the frequency and consequences of infertility is needed. Health options, including male contraceptives, must be expanded, and the opportunities encompassed in such investment understood. The pressing questions related to male reproductive health, spanning the spectrum of andrology are identified in the Expert Recommendation.
Collapse
Affiliation(s)
- Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- The Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- The Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, Quebec, Canada
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Christopher L R Barratt
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Hermann M Behre
- Center for Reproductive Medicine and Andrology, University Hospital, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sarah R Catford
- Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, The Royal Women's Hospital, Melbourne, Victoria, Australia
| | | | - Geraldine Delbes
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Sante Biotechnologie, Laval, Quebec, Canada
| | - Michael L Eisenberg
- Department of Urology and Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - Nicolas Garrido
- IVI Foundation, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Brendan J Houston
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia
| | - Niels Jørgensen
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Csilla Krausz
- Department of Experimental and Clinical Biomedical Sciences, 'Mario Serio', University of Florence, University Hospital of Careggi Florence, Florence, Italy
| | - Ariane Lismer
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Robert I McLachlan
- Hudson Institute of Medical Research and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
- Monash IVF Group, Richmond, Victoria, Australia
| | - Suks Minhas
- Department of Surgery and Cancer Imperial, London, UK
| | - Tim Moss
- Healthy Male and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Allan Pacey
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Lærke Priskorn
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Stefan Schlatt
- Centre for Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Jacquetta Trasler
- Departments of Paediatrics, Human Genetics and Pharmacology & Therapeutics, McGill University and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Leonardo Trasande
- Center for the Investigation of Environmental Hazards, Department of Paediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Mónica Hebe Vazquez-Levin
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Fundación IBYME, Buenos Aires, Argentina
| | - Joris A Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
11
|
Wang R, Feng W, Wang Y, Jiang Y, Lin Y, Chen X. Maternal obstructive sleep apnea aggravates metabolic dysfunction-associated fatty liver disease via HMGB1-TLR4 signaling-mediated endoplasmic reticulum stress in male offspring rats. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166889. [PMID: 37730152 DOI: 10.1016/j.bbadis.2023.166889] [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: 07/03/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023]
Abstract
AIMS/HYPOTHESIS Maternal obstructive sleep apnea (MOSA) may inflict long-term metabolic effects on offspring. We hypothesize that MOSA increases the propensity for metabolic dysregulation in offspring and thus facilitates the development of metabolic dysfunction-associated fatty liver disease (MAFLD). This study aims to test the hypothesis and explore the underlying mechanism. METHODS The MOSA rat model of upper airway obstruction was established and fecundated. The postweaning male offspring (n = 171) from both the control group and MOSA group were randomly fed the normal chow diet (NCD, n = 89) or high-fat diet (HFD, n = 82) for the next 5 months. Liver function, lipid profile, glucose, and insulin levels were measured. Expression levels of fibrosis-related proteins and endoplasmic reticulum (ER) stress-related proteins in liver tissues were assessed using immunohistochemistry and western blotting. RESULTS MOSA increased body and liver weight in male offspring, along with augmented liver organ coefficient. Serum levels of aminotransferases, low-density lipoprotein, high-density lipoprotein, triglycerides, total cholesterol, total bile acid, fasting glucose, and insulin increased significantly. MOSA exacerbated HFD-induced hepatic steatosis and fibrosis. These effects were driven by the overactivated double-stranded RNA-activated protein kinase (PKR)-like eukaryotic initiation factor 2(PERK)-activating transcription factor (ATF)4-C/EBP homologous protein (CHOP) signaling pathway-induced ER stress, and hyperacetylation and release of high mobility group box-1(HMGB1) elicited above signaling in a TLR4-dependent manner. CONCLUSIONS These findings indicate that MOSA can exert prolonged adverse effects manifested as metabolic dysfunction in male offspring. Therefore, surveillance and management of OSA during pregnancy may be necessary to prevent and alleviate MAFLD in offspring.
Collapse
Affiliation(s)
- Ruhua Wang
- Department of Gastroenterology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Wei Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yonghong Jiang
- Department of Gastroenterology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Yiguang Lin
- Central Laboratory, Fist Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China..
| | - Xueqing Chen
- Department of Gastroenterology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
12
|
Dias BG. Legacies of salient environmental experiences-insights from chemosensation. Chem Senses 2024; 49:bjae002. [PMID: 38219073 PMCID: PMC10825851 DOI: 10.1093/chemse/bjae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Indexed: 01/15/2024] Open
Abstract
Evidence for parental environments profoundly influencing the physiology, biology, and neurobiology of future generations has been accumulating in the literature. Recent efforts to understand this phenomenon and its underlying mechanisms have sought to use species like rodents and insects to model multi-generational legacies of parental experiences like stress and nutritional exposures. From these studies, we have come to appreciate that parental exposure to salient environmental experiences impacts the cadence of brain development, hormonal responses to stress, and the expression of genes that govern cellular responses to stress in offspring. Recent studies using chemosensory exposure have emerged as a powerful tool to shed new light on how future generations come to be influenced by environments to which parents are exposed. With a specific focus on studies that have leveraged such use of salient chemosensory experiences, this review synthesizes our current understanding of the concept, causes, and consequences of the inheritance of chemosensory legacies by future generations and how this field of inquiry informs the larger picture of how parental experiences can influence offspring biology.
Collapse
Affiliation(s)
- Brian G Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, United States
- Division of Endocrinology, Diabetes and Metabolism, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA, United States
| |
Collapse
|
13
|
Stratakis CA. Genes and environment: An old pair in a new era. Maturitas 2023; 178:107851. [PMID: 37806009 DOI: 10.1016/j.maturitas.2023.107851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/16/2023] [Indexed: 10/10/2023]
Abstract
What is the relationship between our genes and the environment we live in with regard to health? Like the debate about nature or nurture in the determination of our personality and behavior, the issue of genes and environment has been discussed intensely in the last two centuries. Is it Darwin or Lamarck who is right about the basic determinants of our health, especially as we age in a rapidly changing environment? Evolutionary biology as proposed by Darwin with natural selection at its core may not be able to explain almost instant adjustments of phenotypic traits to the pressures of the environment. Epigenesis, a concept that dates from Aristotle, provides a mechanism for the environment to affect variation in genetic traits that may become heritable. Indeed, Lamarck first described the inheritance of acquired characteristics. Thus, it appears that in contemporary genetics, both Darwin and Lamarck are right: environmental pressures may affect our genes through epigenetics, in ways that allow for inheritance of the changes, a Lamarckian concept; however, evolution through natural selection is the basis for incorporation (or rejection) of new traits and their sustained inheritance, a Darwinian concept. In this review, we present the synthesis of Darwin's and Lamarck's theories, the only way to understand how our health, and that of our progeny, responds to challenging and fast-changing environmental cues. In addition, we present other examples of environment-driven changes in disease frequency or expression.
Collapse
Affiliation(s)
- Constantine A Stratakis
- NIH Clinical Center, NICHD, NIH, Bethesda, MD, USA; Research, Human Genetics & Precision Medicine, IMBB, FORTH, Heraklion, Greece; Medical Genetics, H. Dunant Hospital, Athens, Greece; Science Board, ELPEN Research Institute, Athens, Greece; European University of Cyprus, Medical School, Nicosia, Cyprus.
| |
Collapse
|
14
|
Golding MC. Teratogenesis and the epigenetic programming of congenital defects: Why paternal exposures matter. Birth Defects Res 2023; 115:1825-1834. [PMID: 37424262 PMCID: PMC10774456 DOI: 10.1002/bdr2.2215] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/11/2023]
Abstract
Until recently, clinicians and researchers did not realize paternal exposures could impact child developmental outcomes. Indeed, although there is growing recognition that sperm carry a large amount of non-genomic information and that paternal stressors influence the health of the next generation, toxicologists are only now beginning to explore the role paternal exposures have in dysgenesis and the incidence of congenital malformations. In this commentary, I will briefly summarize the few studies describing congenital malformations resulting from preconception paternal stressors, argue for the theoretical expansion of teratogenic perspectives into the male preconception period, and discuss some of the challenges in this newly emerging branch of toxicology. I argue that we must consider gametes the same as any other malleable precursor cell type and recognize that environmentally-induced epigenetic changes acquired during the formation of the sperm and oocyte hold equal teratogenic potential as exposures during early development. Here, I propose the term epiteratogen to reference agents acting outside of pregnancy that, through epigenetic mechanisms, induce congenital malformations. Understanding the interactions between the environment, the essential epigenetic processes intrinsic to spermatogenesis, and their cumulative influences on embryo patterning is essential to addressing a significant blind spot in the field of developmental toxicology.
Collapse
Affiliation(s)
- Michael C. Golding
- Department of Veterinary Physiology & Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA, 77843
| |
Collapse
|
15
|
Wibisono P, Sun J. Pathogen infection induces specific transgenerational modifications to gene expression and fitness in Caenorhabditis elegans. Front Physiol 2023; 14:1225858. [PMID: 37811492 PMCID: PMC10556243 DOI: 10.3389/fphys.2023.1225858] [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: 05/20/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
How pathogen infection in a parental generation affects response in future generations to the same pathogen via epigenetic modifications has been the topic of recent studies. These studies focused on changes attributed to transgenerational epigenetic inheritance and how these changes cause an observable difference in behavior or immune response in a population. However, we questioned if pathogen infection causes hidden epigenetic changes to fitness that are not observable at the population level. Using the nematode Caenorhabditis elegans as a model organism, we examined the generation-to-generation differences in survival of both an unexposed and primed lineage of animals against a human opportunistic pathogen Salmonella enterica. We discovered that training a lineage of C. elegans against a specific pathogen does not cause a significant change to overall survival, but rather narrows survival variability between generations. Quantification of gene expression revealed reduced variation of a specific member of the TFEB lipophagic pathway. We also provided the first report of a repeating pattern of survival times over the course of 12 generations in the control lineage of C. elegans. This repeating pattern indicates that the variability in survival between generations of the control lineage is not random but may be regulated by unknown mechanisms. Overall, our study indicates that pathogen infection can cause specific phenotypic changes due to epigenetic modifications, and a possible system of epigenetic regulation between generations.
Collapse
Affiliation(s)
- Phillip Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Jingru Sun
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
16
|
Costa DL. Overweight grandsons and grandfathers' starvation exposure. JOURNAL OF HEALTH ECONOMICS 2023; 91:102796. [PMID: 37541079 PMCID: PMC10593129 DOI: 10.1016/j.jhealeco.2023.102796] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 08/06/2023]
Abstract
Much of the increase in the prevalence of overweight and obesity has been in developing countries with a history of famines and malnutrition. This paper is the first to examine overweight among adult grandsons of grandfathers exposed to starvation during developmental ages. I study grandsons born to grandfathers who served in the Union Army during the US Civil War (1861-5) where some grandfathers experienced severe net malnutrition because they suffered a harsh POW experience. I find that male-line but not female-line grandsons of grandfathers who survived a severe captivity during their growing years faced a 21% increase in mean overweight and a 2% increase in mean BMI compared to grandsons of non-POWs. Male-line grandsons descended from grandfathers who experienced a harsh captivity faced a 22%-28% greater risk of dying every year after age 45 relative to grandsons descended from non-POWs, with overweight accounting for 9%-14% of the excess risk.
Collapse
Affiliation(s)
- Dora L Costa
- University of California, Los Angeles, United States of America; National Bureau of Economic Research, United States of America.
| |
Collapse
|
17
|
Bounds D, Woo MA, Nyamathi A, Kehoe P, Roy B, Yadav K, Cabrera-Mino C, Kumar R. Brain Changes Linked to Cognitive Symptomatology in Homeless Youth. Clin Nurs Res 2023:10547738231168465. [PMID: 37157815 DOI: 10.1177/10547738231168465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Youth impacted by homelessness experience diminished cognition due to a variety of reasons including mental health symptoms, alcohol and substance use, and adverse childhood experiences. However, the status of specific brain regions which could impact important cognitive functions in homeless youth remains unclear. In this pilot comparative and correlational study, a series of demographic, psychological, cognitive assessments, and brain magnetic resonance imaging were performed in 10 male youth experiencing homelessness and 9 age-matched healthy male controls (age range: 18-25 years). Participants experiencing homelessness had significantly decreased regional brain gray matter tissue in comparison to the controls. Moreover, there were strong inverse correlations between the brain regions classically associated with executive decision-making (prefrontal cortices), depression (insular lobes), and conflict resolution (anterior cingulate), and the level of the symptoms detected by their questionnaires.
Collapse
Affiliation(s)
| | - Mary A Woo
- University of California Los Angeles, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Svanes C, Holloway JW, Krauss-Etschmann S. Preconception origins of asthma, allergies and lung function: The influence of previous generations on the respiratory health of our children. J Intern Med 2023; 293:531-549. [PMID: 36861185 DOI: 10.1111/joim.13611] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Emerging research suggests that exposures occurring years before conception are important determinants of the health of future offspring and subsequent generations. Environmental exposures of both the father and mother, or exposure to disease processes such as obesity or infections, may influence germline cells and thereby cause a cascade of health outcomes in multiple subsequent generations. There is now increasing evidence that respiratory health is influenced by parental exposures that occur long before conception. The strongest evidence relates adolescent tobacco smoking and overweight in future fathers to increased asthma and lower lung function in their offspring, supported by evidence on parental preconception occupational exposures and air pollution. Although this literature is still sparse, the epidemiological analyses reveal strong effects that are consistent across studies with different designs and methodologies. The results are strengthened by mechanistic research from animal models and (scarce) human studies that have identified molecular mechanisms that can explain the epidemiological findings, suggesting transfer of epigenetic signals through germline cells, with susceptibility windows in utero (both male and female line) and prepuberty (male line). The concept that our lifestyles and behaviours may influence the health of our future children represents a new paradigm. This raises concerns for future health in decades to come with respect to harmful exposures but may also open for radical rethinking of preventive strategies that may improve health in multiple generations, reverse the imprint of our parents and forefathers, and underpin strategies that can break the vicious circle of propagation of health inequalities across generations.
Collapse
Affiliation(s)
- Cecilie Svanes
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway.,Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
19
|
Ribas-Aulinas F, Ribo S, Casas E, Mourin-Fernandez M, Ramon-Krauel M, Diaz R, Lerin C, Kalko SG, Vavouri T, Jimenez-Chillaron JC. Intergenerational Inheritance of Hepatic Steatosis in a Mouse Model of Childhood Obesity: Potential Involvement of Germ-Line microRNAs. Nutrients 2023; 15:nu15051241. [PMID: 36904241 PMCID: PMC10005268 DOI: 10.3390/nu15051241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Childhood obesity increases the risk of developing metabolic syndrome later in life. Moreover, metabolic dysfunction may be inherited into the following generation through non-genomic mechanisms, with epigenetics as a plausible candidate. The pathways involved in the development of metabolic dysfunction across generations in the context of childhood obesity remain largely unexplored. We have developed a mouse model of early adiposity by reducing litter size at birth (small litter group, SL: 4 pups/dam; control group, C: 8 pups/dam). Mice raised in small litters (SL) developed obesity, insulin resistance and hepatic steatosis with aging. Strikingly, the offspring of SL males (SL-F1) also developed hepatic steatosis. Paternal transmission of an environmentally induced phenotype strongly suggests epigenetic inheritance. We analyzed the hepatic transcriptome in C-F1 and SL-F1 mice to identify pathways involved in the development of hepatic steatosis. We found that the circadian rhythm and lipid metabolic process were the ontologies with highest significance in the liver of SL-F1 mice. We explored whether DNA methylation and small non-coding RNAs might be involved in mediating intergenerational effects. Sperm DNA methylation was largely altered in SL mice. However, these changes did not correlate with the hepatic transcriptome. Next, we analyzed small non-coding RNA content in the testes of mice from the parental generation. Two miRNAs (miR-457 and miR-201) appeared differentially expressed in the testes of SL-F0 mice. They are known to be expressed in mature spermatozoa, but not in oocytes nor early embryos, and they may regulate the transcription of lipogenic genes, but not clock genes, in hepatocytes. Hence, they are strong candidates to mediate the inheritance of adult hepatic steatosis in our murine model. In conclusion, litter size reduction leads to intergenerational effects through non-genomic mechanisms. In our model, DNA methylation does not seem to play a role on the circadian rhythm nor lipid genes. However, at least two paternal miRNAs might influence the expression of a few lipid-related genes in the first-generation offspring, F1.
Collapse
Affiliation(s)
| | - Sílvia Ribo
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Eduard Casas
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | | | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Susana G. Kalko
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Tanya Vavouri
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | - Josep C. Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- School of Medicine, University of Barcelona, L’Hospitalet, 08907 Barcelona, Spain
- Correspondence: or ; Tel.: +34-934024267
| |
Collapse
|
20
|
Madrid A, Alisch RS, Rizk E, Papale LA, Hogan KJ, Iskandar BJ. Transgenerational epigenetic inheritance of axonal regeneration after spinal cord injury. ENVIRONMENTAL EPIGENETICS 2023; 9:dvad002. [PMID: 36843857 PMCID: PMC9949995 DOI: 10.1093/eep/dvad002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/01/2023] [Accepted: 01/14/2023] [Indexed: 05/14/2023]
Abstract
Human epidemiological studies reveal that dietary and environmental alterations influence the health of the offspring and that the effect is not limited to the F1 or F2 generations. Non-Mendelian transgenerational inheritance of traits in response to environmental stimuli has been confirmed in non-mammalian organisms including plants and worms and are shown to be epigenetically mediated. However, transgenerational inheritance beyond the F2 generation remains controversial in mammals. Our lab previously discovered that the treatment of rodents (rats and mice) with folic acid significantly enhances the regeneration of injured axons following spinal cord injury in vivo and in vitro, and the effect is mediated by DNA methylation. The potential heritability of DNA methylation prompted us to investigate the following question: Is the enhanced axonal regeneration phenotype inherited transgenerationally without exposure to folic acid supplementation in the intervening generations? In the present review, we condense our findings showing that a beneficial trait (i.e., enhanced axonal regeneration after spinal cord injury) and accompanying molecular alterations (i.e., DNA methylation), triggered by an environmental exposure (i.e., folic acid supplementation) to F0 animals only, are inherited transgenerationally and beyond the F3 generation.
Collapse
Affiliation(s)
- Andy Madrid
- Department of Neurological Surgery, University of Wisconsin—Madison, Madison, WI 53719, USA
| | - Reid S Alisch
- Department of Neurological Surgery, University of Wisconsin—Madison, Madison, WI 53719, USA
| | - Elias Rizk
- Department of Neurosurgery, Penn State Children’s Hospital, Hershey, PA 17033, USA
| | - Ligia A Papale
- Department of Neurological Surgery, University of Wisconsin—Madison, Madison, WI 53719, USA
| | - Kirk J Hogan
- Department of Anesthesiology, University of Wisconsin—Madison, Madison, WI 53719, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin—Madison, Madison, WI 53719, USA
| | - Bermans J Iskandar
- Department of Neurological Surgery, University of Wisconsin—Madison, Madison, WI 53719, USA
| |
Collapse
|
21
|
Duhuze Karera MG, Wentzel A, Ishimwe MCS, Gatete JDD, Jagannathan R, Horlyck-Romanovsky MF, Sumner AE. A Scoping Review of Trials Designed to Achieve Remission of Type 2 Diabetes with Lifestyle Intervention Alone: Implications for Sub-Saharan Africa. Diabetes Metab Syndr Obes 2023; 16:677-692. [PMID: 36923683 PMCID: PMC10010137 DOI: 10.2147/dmso.s403054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
According to the International Diabetes Federation, sub-Saharan Africa is experiencing the highest anticipate increase in the prevalence of type 2 diabetes (T2D) in the world and has the highest percent of people living with T2D who are undiagnosed. Therefore, diagnosis and treatment need prioritization. However, pharmacological hypoglycemics are often unavailable and bariatric surgery is not an option. Therefore, the ability to induce T2D remission through lifestyle intervention alone (LSI-alone) needs assessment. This scoping review evaluated trials designed to induce T2D remission by LSI-alone. PubMed, Embase, Cochrane, and CINAHL databases were searched for trials designed to induce T2D remission through LSI-alone. Of the 928 identified, 63 duplicates were removed. With abstract review, 727 irrelevant articles were excluded. After full-text review, 112 inappropriate articles were removed. The remaining 26 articles described 16 trials. These trials were published between 1984 and 2021 and were conducted in 10 countries, none of which were in Africa. Remission rates varied across trials. Predictors of remission were 10% weight loss and higher BMI, lower A1C and shorter T2D duration at enrollment. However, LSI-alone regimens for newly diagnosed and established T2D were very different. In newly diagnosed T2D, LSI-alone were relatively low-cost and focused on exercise and dietary counseling with or without calorie restriction (~1500 kcal/d). Presumably due to differences in cost, LSI-alone trials in newly diagnosed T2D had higher enrollments and longer duration. For established T2D trials, the focus was on arduous phased dietary interventions; phase 1: low-calorie meal replacement (<1000 kcal/day); phase 2: food re-introduction; phase 3: weight maintenance. In short, LSI-alone can induce remission in both newly diagnosed and established T2D. To demonstrate efficacy in Africa, initial trials could focus on newly diagnosed T2D. Insight gained could provide proof of concept and a foundation in Africa on which successful studies of LSI-alone in established T2D could be built.
Collapse
Affiliation(s)
- M Grace Duhuze Karera
- Section on Ethnicity and Health, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Institute of Global Health Equity Research, University of Global Health Equity, Kigali, Rwanda
| | - Annemarie Wentzel
- Hypertension in Africa Research Team, North-West University, Potchefstroom, South Africa
- South African Medical Research Council, Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - M C Sage Ishimwe
- Institute of Global Health Equity Research, University of Global Health Equity, Kigali, Rwanda
| | - Jean de Dieu Gatete
- Section on Ethnicity and Health, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ram Jagannathan
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Margrethe F Horlyck-Romanovsky
- Section on Ethnicity and Health, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, New York, NY, USA
| | - Anne E Sumner
- Section on Ethnicity and Health, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- National Institute of Minority Health and Health Disparities, NIH, Bethesda, MD, USA
- Correspondence: Anne E Sumner, Building 10, CRC-Rm 6940, Bethesda, MD, 20892-1612, USA, Tel +1 301-402-4240, Email
| |
Collapse
|
22
|
Food abundance in men before puberty predicts a range of cancers in grandsons. Nat Commun 2022; 13:7507. [PMID: 36473854 PMCID: PMC9726939 DOI: 10.1038/s41467-022-35217-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Nutritional conditions early in human life may influence phenotypic characteristics in later generations. A male-line transgenerational pathway, triggered by the early environment, has been postulated with support from animal and a small number of human studies. Here we analyse individuals born in Uppsala Sweden 1915-29 with linked data from their children and parents, which enables us to explore the hypothesis that pre-pubertal food abundance may trigger a transgenerational effect on cancer events. We used cancer registry and cause-of-death data to analyse 3422 cancer events in grandchildren (G2) by grandparental (G0) food access. We show that variation in harvests and food access in G0 predicts cancer occurrence in G2 in a specific way: abundance among paternal grandfathers, but not any other grandparent, predicts cancer occurrence in grandsons but not in granddaughters. This male-line response is observed for several groups of cancers, suggesting a general susceptibility, possibly acquired in early embryonic development. We observed no transgenerational influence in the middle generation.
Collapse
|
23
|
Boscardin C, Manuella F, Mansuy IM. Paternal transmission of behavioural and metabolic traits induced by postnatal stress to the 5th generation in mice. ENVIRONMENTAL EPIGENETICS 2022; 8:dvac024. [PMID: 36518875 PMCID: PMC9730319 DOI: 10.1093/eep/dvac024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/16/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Life experiences and environmental conditions in childhood can change the physiology and behaviour of exposed individuals and, in some cases, of their offspring. In rodent models, stress/trauma, poor diet, and endocrine disruptors in a parent have been shown to cause phenotypes in the direct progeny, suggesting intergenerational inheritance. A few models also examined transmission to further offspring and suggested transgenerational inheritance, but such multigenerational inheritance is not well characterized. Our previous work on a mouse model of early postnatal stress showed that behaviour and metabolism are altered in the offspring of exposed males up to the 4th generation in the patriline and up to the 2nd generation in the matriline. The present study examined if symptoms can be transmitted beyond the 4th generation in the patriline. Analyses of the 5th and 6th generations of mice revealed that altered risk-taking and glucose regulation caused by postnatal stress are still manifested in the 5th generation but are attenuated in the 6th generation. Some of the symptoms are expressed in both males and females, but some are sex-dependent and sometimes opposite. These results indicate that postnatal trauma can affect behaviour and metabolism over many generations, suggesting epigenetic mechanisms of transmission.
Collapse
Affiliation(s)
- Chiara Boscardin
- Laboratory of Neuroepigenetics, Brain Research Institute, Faculty of Medicine of the University Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
- Institute for Neuroscience, Department of Health Science and Technology of ETH Zürich, Centre for Neuroscience Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Francesca Manuella
- Laboratory of Neuroepigenetics, Brain Research Institute, Faculty of Medicine of the University Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
- Institute for Neuroscience, Department of Health Science and Technology of ETH Zürich, Centre for Neuroscience Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Isabelle M Mansuy
- *Correspondence address. Laboratory of Neuroepigenetics, University of Zürich and ETH Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland. Tel: +41 44 6353360; Fax: +41 44 635 33 03; E-mail:
| |
Collapse
|
24
|
Sperm-inherited H3K27me3 epialleles are transmitted transgenerationally in cis. Proc Natl Acad Sci U S A 2022; 119:e2209471119. [PMID: 36161922 PMCID: PMC9546627 DOI: 10.1073/pnas.2209471119] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The transmission of chromatin states from parent cells to daughter cells preserves cell-specific transcriptional states and thus cell identity through cell division. The mechanism that underpins this process is not fully understood. The role that chromatin states serve in transmitting gene expression information across generations via sperm and oocytes is even less understood. Here, we utilized a model in which Caenorhabditis elegans sperm and oocyte alleles were inherited in different states of the repressive mark H3K27me3. This resulted in the alleles achieving different transcriptional states within the nuclei of offspring. Using this model, we showed that sperm alleles inherited without H3K27me3 were sensitive to up-regulation in offspring somatic and germline tissues, and tissue context determined which genes were up-regulated. We found that the subset of sperm alleles that were up-regulated in offspring germlines retained the H3K27me3(-) state and were transmitted to grandoffspring as H3K27me3(-) and up-regulated epialleles, demonstrating that H3K27me3 can serve as a transgenerational epigenetic carrier in C. elegans.
Collapse
|
25
|
Evidence of transgenerational effects on autism spectrum disorder using multigenerational space-time cluster detection. Int J Health Geogr 2022; 21:13. [PMID: 36192740 PMCID: PMC9531495 DOI: 10.1186/s12942-022-00313-4] [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: 07/13/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022] Open
Abstract
Background Transgenerational epigenetic risks associated with complex health outcomes, such as autism spectrum disorder (ASD), have attracted increasing attention. Transgenerational environmental risk exposures with potential for epigenetic effects can be effectively identified using space-time clustering. Specifically applied to ancestors of individuals with disease outcomes, space-time clustering characterized for vulnerable developmental stages of growth can provide a measure of relative risk for disease outcomes in descendants. Objectives (1) Identify space-time clusters of ancestors with a descendent with a clinical ASD diagnosis and matched controls. (2) Identify developmental windows of ancestors with the highest relative risk for ASD in descendants. (3) Identify how the relative risk may vary through the maternal or paternal line. Methods Family pedigrees linked to residential locations of ASD cases in Utah have been used to identify space-time clusters of ancestors. Control family pedigrees of none-cases based on age and sex have been matched to cases 2:1. The data have been categorized by maternal or paternal lineage at birth, childhood, and adolescence. A total of 3957 children, both parents, and maternal and paternal grandparents were identified. Bernoulli space-time binomial relative risk (RR) scan statistic was used to identify clusters. Monte Carlo simulation was used for statistical significance testing. Results Twenty statistically significant clusters were identified. Thirteen increased RR (> 1.0) space-time clusters were identified from the maternal and paternal lines at a p-value < 0.05. The paternal grandparents carry the greatest RR (2.86–2.96) during birth and childhood in the 1950’s–1960, which represent the smallest size clusters, and occur in urban areas. Additionally, seven statistically significant clusters with RR < 1 were relatively large in area, covering more rural areas of the state. Conclusion This study has identified statistically significant space-time clusters during critical developmental windows that are associated with ASD risk in descendants. The geographic space and time clusters family pedigrees with over 3 + generations, which we refer to as a person’s geographic legacy, is a powerful tool for studying transgenerational effects that may be epigenetic in nature. Our novel use of space-time clustering can be applied to any disease where family pedigree data is available. Supplementary Information The online version contains supplementary material available at 10.1186/s12942-022-00313-4.
Collapse
|
26
|
Belmonte KCD, Holmgren EB, Wills TA, Gidday JM. Epigenetic conditioning induces intergenerational resilience to dementia in a mouse model of vascular cognitive impairment. Alzheimers Dement 2022; 18:1711-1720. [PMID: 35170835 PMCID: PMC9790554 DOI: 10.1002/alz.12616] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Epigenetic stimuli induce beneficial or detrimental changes in gene expression, and consequently, phenotype. Some of these phenotypes can manifest across the lifespan-and even in subsequent generations. Here, we used a mouse model of vascular cognitive impairment and dementia (VCID) to determine whether epigenetically induced resilience to specific dementia-related phenotypes is heritable by first-generation progeny. METHODS Our systemic epigenetic therapy consisted of 2 months of repetitive hypoxic "conditioning" (RHC) prior to chronic cerebral hypoperfusion in adult C57BL/6J mice. Resultant changes in object recognition memory and hippocampal long-term potentiation (LTP) were assessed 3 and 4 months later, respectively. RESULTS Hypoperfusion-induced memory/plasticity deficits were abrogated by RHC. Moreover, similarly robust dementia resilience was documented in untreated cerebral hypoperfused animals derived from RHC-treated parents. CONCLUSIONS Our results in experimental VCID underscore the efficacy of epigenetics-based treatments to prevent memory loss, and demonstrate for the first time the heritability of an induced resilience to dementia.
Collapse
Affiliation(s)
- Krystal Courtney D. Belmonte
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Eleanor B. Holmgren
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Tiffany A. Wills
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Jeff M. Gidday
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of Biochemistry and Molecular BiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| |
Collapse
|
27
|
Mbiydzenyuy NE, Hemmings SMJ, Qulu L. Prenatal maternal stress and offspring aggressive behavior: Intergenerational and transgenerational inheritance. Front Behav Neurosci 2022; 16:977416. [PMID: 36212196 PMCID: PMC9539686 DOI: 10.3389/fnbeh.2022.977416] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Even though studies have shown that prenatal maternal stress is associated with increased reactivity of the HPA axis, the association between prenatal maternal stress and fetal glucocorticoid exposure is complex and most likely dependent on unidentified and poorly understood variables including nature and timing of prenatal insults. The precise mechanisms in which prenatal maternal stress influence neuroendocrine signaling between the maternal-placental-fetal interface are still unclear. The aim of this review article is to bring comprehensive basic concepts about prenatal maternal stress and mechanisms of transmission of maternal stress to the fetus. This review covers recent studies showing associations between maternal stress and alterations in offspring aggressive behavior, as well as the possible pathways for the “transmission” of maternal stress to the fetus: (1) maternal-fetal HPA axis dysregulation; (2) intrauterine environment disruption due to variations in uterine artery flow; (3) epigenetic modifications of genes implicated in aggressive behavior. Here, we present evidence for the phenomenon of intergenerational and transgenerational transmission, to better understands the mechanism(s) of transmission from parent to offspring. We discuss studies showing associations between maternal stress and alterations in offspring taking note of neuroendocrine, brain architecture and epigenetic changes that may suggest risk for aggressive behavior. We highlight animal and human studies that focus on intergenerational transmission following exposure to stress from a biological mechanistic point of view, and maternal stress-induced epigenetic modifications that have potential to impact on aggressive behavior in later generations.
Collapse
Affiliation(s)
- Ngala Elvis Mbiydzenyuy
- Department of Basic Science, School of Medicine, Copperbelt University, Ndola, Zambia
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Cape Town, South Africa
- *Correspondence: Ngala Elvis Mbiydzenyuy,
| | - Sian Megan Joanna Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lihle Qulu
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
28
|
Salinas-Roca B, Rubió-Piqué L, Montull-López A. Polyphenol Intake in Pregnant Women on Gestational Diabetes Risk and Neurodevelopmental Disorders in Offspring: A Systematic Review. Nutrients 2022; 14:3753. [PMID: 36145129 PMCID: PMC9502213 DOI: 10.3390/nu14183753] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/03/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
The intake of foods containing polyphenols can have a protective role to avoid comorbidities during pregnancy and, at the same time, promote transgenerational health. This review aims to describe the effect of polyphenol intake through supplements or polyphenol-rich foods during pregnancy on the incidence and evolution of gestational diabetes mellitus (GDM), as well as the link with the neurodevelopment of the fetus. Using PRISMA procedures, a systematic review was conducted by searching in biomedical databases (PubMed, Cinahl and Scopus) from January to June 2022. Full articles were screened (n = 419) and critically appraised. Fourteen studies were selected and were divided into two different thematic blocks considering (i) the effect of polyphenols in GDM and (ii) the effect of GDM to mental disorders in the offspring. A positive relationship was observed between the intake of polyphenols and the prevention and control of cardiometabolic complications during pregnancy, such as GDM, which could be related to thwarted inflammatory and oxidative processes, as well as neuronal factors. GDM is related to a greater risk of suffering from diseases related to neurodevelopment, such as attention deficit hyperactivity disorder, autism spectrum disorder and learning disorder. Further clinical research on the molecule protective mechanism of polyphenols on pregnant women is required to understand the transgenerational impact on fetal neurodevelopment.
Collapse
Affiliation(s)
- Blanca Salinas-Roca
- Global Research on Wellbeing (GRoW) Research Group, Blanquerna School of Health Science, Ramon Llull University, Padilla, 326-332, 08025 Barcelona, Spain
- Department of Nursing and Physiotherapy, University of Lleida, Montserrat Roig 2, 25198 Lleida, Spain
| | - Laura Rubió-Piqué
- Antioxidants Research Group, Food Technology Department, AGROTECNIO-CERCA Center, University of Lleida, Av/Alcalde Rovira Roure 191, 25198 Lleida, Spain
| | - Anna Montull-López
- Department of Nursing and Physiotherapy, University of Lleida, Montserrat Roig 2, 25198 Lleida, Spain
| |
Collapse
|
29
|
Birmingham K, Iles-Caven Y, Golding J. ALSPAC parents' descriptions of childhood stresses in their parents and grandparents. Wellcome Open Res 2022; 6:115. [PMID: 35693334 PMCID: PMC9156897 DOI: 10.12688/wellcomeopenres.16732.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
Background: There is evidence that childhood stresses or traumas influence individuals' descendants' health and wellbeing through epigenetic mechanisms. However, few longitudinal studies have details of such ancestral data. Methods: Nearly 7,000 parents of the original Avon Longitudinal Study of Parents and Children (ALSPAC) cohort completed questionnaires concerning their parents' and grandparents' childhoods. As part of a questionnaire validation exercise we conducted recorded interviews with 100 of these parents. Here we describe some of the vivid accounts from these interviews of stresses encountered by the parents' ancestors. Results: The interviews provided insights into the childhoods of two previous generations of this cohort, most of whom had lived through one, if not two, World Wars. Many children were brought up, not by their parents but by relatives or acquaintances and/or left home very young to 'go into service' or start work. A few interviewees had wealthy relatives with nannies and governesses and attended expensive boarding schools but by far the most frequent accounts were of poverty, often severe, with related lack of education and illiteracy, alcoholism and violence, alongside devastating effects of the World Wars. Conclusions: Although the interviews focussed on stresses in childhood and therefore the accounts seemed somewhat negative, many interviewees described their relatives as having secure, stable childhoods. Of the many struggling families though, the predominant impression was their remarkable resilience; all went on to have children or grandchildren who are stable enough to participate for three decades, entirely altruistically, in ALSPAC.
Collapse
Affiliation(s)
- Karen Birmingham
- Centre for Academic Child Health, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Yasmin Iles-Caven
- Centre for Academic Child Health, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Jean Golding
- Centre for Academic Child Health, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| |
Collapse
|
30
|
Svanes C, Johannessen A, Bertelsen RJ, Dharmage S, Benediktsdottir B, Bråbäck L, Gislason T, Holm M, Jõgi O, Lodge CJ, Malinovschi A, Martinez-Moratalla J, Oudin A, Sánchez-Ramos JL, Timm S, Janson C, Real FG, Schlünssen V. Cohort profile: the multigeneration Respiratory Health in Northern Europe, Spain and Australia (RHINESSA) cohort. BMJ Open 2022; 12:e059434. [PMID: 35654464 PMCID: PMC9163543 DOI: 10.1136/bmjopen-2021-059434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/07/2022] [Indexed: 11/30/2022] Open
Abstract
PURPOSE The Respiratory Health in Northern Europe, Spain and Australia (RHINESSA) cohort was established to (1) investigate how exposures before conception and in previous generations influence health and disease, particularly allergies and respiratory health, (2) identify susceptible time windows and (3) explore underlying mechanisms. The ultimate aim is to facilitate efficient intervention strategies targeting multiple generations. PARTICIPANTS RHINESSA includes study participants of multiple generations from ten study centres in Norway (1), Denmark (1), Sweden (3), Iceland (1), Estonia (1), Spain (2) and Australia (1). The RHINESSA core cohort, adult offspring generation 3 (G3), was first investigated in 2014-17 in a questionnaire study (N=8818, age 18-53 years) and a clinical study (subsample, n=1405). Their G2 parents participated in the population-based cohorts, European Community Respiratory Heath Survey and Respiratory Health In Northern Europe, followed since the early 1990s when they were 20-44 years old, at 8-10 years intervals. Study protocols are harmonised across generations. FINDINGS TO DATE Collected data include spirometry, skin prick tests, exhaled nitric oxide, anthropometrics, bioimpedance, blood pressure; questionnaire/interview data on respiratory/general/reproductive health, indoor/outdoor environment, smoking, occupation, general characteristics and lifestyle; biobanked blood, urine, gingival fluid, skin swabs; measured specific and total IgE, DNA methylation, sex hormones and oral microbiome. Research results suggest that parental environment years before conception, in particular, father's exposures such as smoking and overweight, may be of key importance for asthma and lung function, and that there is an important susceptibility window in male prepuberty. Statistical analyses developed to approach causal inference suggest that these associations may be causal. DNA methylation studies suggest a mechanism for transfer of father's exposures to offspring health and disease through impact on offspring DNA methylation. FUTURE PLANS Follow-up is planned at 5-8 years intervals, first in 2021-2023. Linkage with health registries contributes to follow-up of the cohort.
Collapse
Affiliation(s)
- Cecilie Svanes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
- Centre for International Health, University of Bergen Department of Global Public Health and Primary Care, Bergen, Norway
| | - Ane Johannessen
- Centre for International Health, University of Bergen Department of Global Public Health and Primary Care, Bergen, Norway
| | - Randi Jacobsen Bertelsen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Oral Helath Centre of Expertise Western Norway, Bergen, Norway
| | - Shyamali Dharmage
- Allergy and Health Unit, School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Bryndis Benediktsdottir
- Medical Faculty, University of Iceland, Reykjavik, Iceland
- Department of Sleep, Landspitali University Hospital Reykjavík, Reykjavik, Iceland
| | - Lennart Bråbäck
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå Universitet, Umeå, Sweden
| | - Thorarinn Gislason
- Department of Sleep, Landspitali University Hospital Reykjavík, Reykjavik, Iceland
| | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Goteborg, Sweden
| | - Oskar Jõgi
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Lung Clinic, Tartu University Hospital, Tartu, Estonia
| | - Caroline J Lodge
- Allergy and Lung Health Unit, School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Jesus Martinez-Moratalla
- Servicio de Neumología, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha - Campus de Albacete, Albacete, Spain
| | - Anna Oudin
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå Universitet, Umeå, Sweden
| | | | - Signe Timm
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Research Unit, Kolding Hospital, University Hospital of Southern Denmark, Kolding, Denmark
| | - Christer Janson
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences: Respiratory, Allergy, Sleep Research, Uppsala University, Uppsala, Sweden
| | - Francisco Gomez Real
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Vivi Schlünssen
- Department of Public Health - Work, Environment and Health, Danish Ramazzini Centre, Aarhus Universitet, Aarhus, Denmark
- National Research Centre for the Working Environment, Kobenhavn, Denmark
| |
Collapse
|
31
|
Van de Pette M, Dimond A, Galvão AM, Millership SJ, To W, Prodani C, McNamara G, Bruno L, Sardini A, Webster Z, McGinty J, French PMW, Uren AG, Castillo-Fernandez J, Watkinson W, Ferguson-Smith AC, Merkenschlager M, John RM, Kelsey G, Fisher AG. Epigenetic changes induced by in utero dietary challenge result in phenotypic variability in successive generations of mice. Nat Commun 2022; 13:2464. [PMID: 35513363 PMCID: PMC9072353 DOI: 10.1038/s41467-022-30022-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 04/13/2022] [Indexed: 11/22/2022] Open
Abstract
Transmission of epigenetic information between generations occurs in nematodes, flies and plants, mediated by specialised small RNA pathways, modified histones and DNA methylation. Similar processes in mammals can also affect phenotype through intergenerational or trans-generational mechanisms. Here we generate a luciferase knock-in reporter mouse for the imprinted Dlk1 locus to visualise and track epigenetic fidelity across generations. Exposure to high-fat diet in pregnancy provokes sustained re-expression of the normally silent maternal Dlk1 in offspring (loss of imprinting) and increased DNA methylation at the somatic differentially methylated region (sDMR). In the next generation heterogeneous Dlk1 mis-expression is seen exclusively among animals born to F1-exposed females. Oocytes from these females show altered gene and microRNA expression without changes in DNA methylation, and correct imprinting is restored in subsequent generations. Our results illustrate how diet impacts the foetal epigenome, disturbing canonical and non-canonical imprinting mechanisms to modulate the properties of successive generations of offspring.
Collapse
Affiliation(s)
- Mathew Van de Pette
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Andrew Dimond
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - António M Galvão
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Institute of Animal Reproduction and Food Research of PAS, Department of Reproductive Immunology and Pathology, Olsztyn, Poland
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Steven J Millership
- Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Wilson To
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Chiara Prodani
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Gráinne McNamara
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Ludovica Bruno
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Alessandro Sardini
- Whole Animal Physiology and Imaging, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Zoe Webster
- Transgenics and Embryonic Stem Cell Laboratory, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - James McGinty
- Photonics Group, Department of Physics, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Paul M W French
- Photonics Group, Department of Physics, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Anthony G Uren
- Cancer Genomics Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | | | - William Watkinson
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Matthias Merkenschlager
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Rosalind M John
- Cardiff School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge, CB2 0QQ, UK
| | - Amanda G Fisher
- Lymphocyte Development & Epigenetic Memory Groups, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
32
|
Clarke AJ, van El CG. Genomics and justice: mitigating the potential harms and inequities that arise from the implementation of genomics in medicine. Hum Genet 2022; 141:1099-1107. [PMID: 35412078 PMCID: PMC9160156 DOI: 10.1007/s00439-022-02453-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/04/2022]
Abstract
Advances in human genetics raise many social and ethical issues. The application of genomic technologies to healthcare has raised many questions at the level of the individual and the family, about conflicts of interest among professionals, and about the limitations of genomic testing. In this paper, we attend to broader questions of social justice, such as how the implementation of genomics within healthcare could exacerbate pre-existing inequities or the discrimination against social groups. By anticipating these potential problems, we hope to minimise their impact. We group the issues to address into six categories: (i) access to healthcare in general, not specific to genetics. This ranges from healthcare insurance to personal behaviours. (ii) data management and societal discrimination against groups on the basis of genetics. (iii) epigenetics research recognises how early life exposure to stress, including malnutrition and social deprivation, can lead to ill health in adult life and further social disadvantage. (iv) psychiatric genomics and the genetics of IQ may address important questions of therapeutics but could also be used to disadvantage specific social or ethnic groups. (v) complex diseases are influenced by many factors, including genetic polymorphisms of individually small effect. A focus on these polygenic influences distracts from environmental factors that are more open to effective interventions. (vi) population genomic screening aims to support couples making decisions about reproduction. However, this remains a highly contentious area. We need to maintain a careful balance of the competing social and ethical tensions as the technology continues to develop.
Collapse
Affiliation(s)
- A J Clarke
- Division of Cancer & Genetics, School of Medicine, Cardiff University, Wales, UK.
| | - C G van El
- Department of Human Genetics and Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Transgenerational epigenetic impacts of parental infection on offspring health and disease susceptibility. Trends Genet 2022; 38:662-675. [PMID: 35410793 PMCID: PMC8992946 DOI: 10.1016/j.tig.2022.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022]
Abstract
Maternal immune activation (MIA) and infection during pregnancy are known to reprogramme offspring phenotypes. However, the epigenetic effects of preconceptual paternal infection and paternal immune activation (PIA) are not currently well understood. Recent reports show that paternal infection and immune activation can affect offspring phenotypes, particularly brain function, behaviour, and immune system functioning, across multiple generations without re-exposure to infection. Evidence from other environmental exposures indicates that epigenetic inheritance also occurs in humans. Given the growing impact of the coronavirus disease 2019 (COVID-19) pandemic, it is imperative that we investigate all of the potential epigenetic mechanisms and multigenerational phenotypes that may arise from both maternal and paternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as well as associated MIA, PIA, and inflammation. This will allow us to understand and, if necessary, mitigate any potential changes in disease susceptibility in the children, and grandchildren, of affected parents.
Collapse
|
34
|
Malecki KMC, Andersen JK, Geller AM, Harry GJ, Jackson CL, James KA, Miller GW, Ottinger MA. Integrating Environment and Aging Research: Opportunities for Synergy and Acceleration. Front Aging Neurosci 2022; 14:824921. [PMID: 35264945 PMCID: PMC8901047 DOI: 10.3389/fnagi.2022.824921] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/12/2022] [Indexed: 12/25/2022] Open
Abstract
Despite significant overlaps in mission, the fields of environmental health sciences and aging biology are just beginning to intersect. It is increasingly clear that genetics alone does not predict an individual’s neurological aging and sensitivity to disease. Accordingly, aging neuroscience is a growing area of mutual interest within environmental health sciences. The impetus for this review came from a workshop hosted by the National Academies of Sciences, Engineering, and Medicine in June of 2020, which focused on integrating the science of aging and environmental health research. It is critical to bridge disciplines with multidisciplinary collaborations across toxicology, comparative biology, epidemiology to understand the impacts of environmental toxicant exposures and age-related outcomes. This scoping review aims to highlight overlaps and gaps in existing knowledge and identify essential research initiatives. It begins with an overview of aging biology and biomarkers, followed by examples of synergy with environmental health sciences. New areas for synergistic research and policy development are also discussed. Technological advances including next-generation sequencing and other-omics tools now offer new opportunities, including exposomic research, to integrate aging biomarkers into environmental health assessments and bridge disciplinary gaps. This is necessary to advance a more complete mechanistic understanding of how life-time exposures to toxicants and other physical and social stressors alter biological aging. New cumulative risk frameworks in environmental health sciences acknowledge that exposures and other external stressors can accumulate across the life course and the advancement of new biomarkers of exposure and response grounded in aging biology can support increased understanding of population vulnerability. Identifying the role of environmental stressors, broadly defined, on aging biology and neuroscience can similarly advance opportunities for intervention and translational research. Several areas of growing research interest include expanding exposomics and use of multi-omics, the microbiome as a mediator of environmental stressors, toxicant mixtures and neurobiology, and the role of structural and historical marginalization and racism in shaping persistent disparities in population aging and outcomes. Integrated foundational and translational aging biology research in environmental health sciences is needed to improve policy, reduce disparities, and enhance the quality of life for older individuals.
Collapse
Affiliation(s)
- Kristen M. C. Malecki
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- *Correspondence: Kristen M. C. Malecki,
| | | | - Andrew M. Geller
- United States Environmental Protection Agency, Office of Research and Development, Durham, NC, United States
| | - G. Jean Harry
- Division of National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Chandra L. Jackson
- Division of Intramural Research, Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, United States
- Department of Health and Human Services, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, United States
| | - Katherine A. James
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Denver, CO, United States
| | - Gary W. Miller
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Mary Ann Ottinger
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| |
Collapse
|
35
|
Intergenerational effects of early-life health shocks during the Chinese 1959–1961 famine. AGEING & SOCIETY 2022. [DOI: 10.1017/s0144686x22000113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Abstract
A large literature has examined early-life insult and later-life health outcomes. However, whether early-life exposure might persist into the outcomes of future generations remains unclear. Using data from the China Family Panel Studies, this study examines the intergenerational effects of early-life health shocks during the great famine in China, distinguishes the intergenerational effects of in utero and early-life famine exposure, and estimates whether there is a sex-specific transgenerational response. Difference-in-difference results show that first-generation male in utero famine exposure (1959–1961) is associated with a series of health and economic disadvantages in the second generation, compared with the unexposed post-famine-born cohort (1964–1965) in China. The effect persists in the third generation but attenuates, and there is no same-sex transgenerational response. These findings may suggest a novel source of multigenerational persistence in health and economic poverty and may point to a need to consider evidence of transgenerational mechanisms.
Collapse
|
36
|
Qi L, Liu YP, Wang SM, Shi H, Chen XL, Wang NN, Su YC. Abnormal BMI in Male and/or Female Partners Are Deleterious for Embryonic Development and Pregnancy Outcome During ART Process: A Retrospective Study. Front Endocrinol (Lausanne) 2022; 13:856667. [PMID: 35528007 PMCID: PMC9068983 DOI: 10.3389/fendo.2022.856667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To investigate the effect of BMI in male and/or female partners on embryo development and clinical pregnancy outcome during ART. METHODS Data of 11,130 cycles between January 2018 and December 2020 were retrospectively analyzed. They were divided into Group A, B, C, and D based on couples' BMI values, also were divided into Group I, II, III and IV in IVF cycles and Group i, ii, iii, and iv in ICSI cycles. After grouping, inter-group indicators comparisons and logistic regression analysis were performed. RESULTS In IVF cycles, CPR in Group I and Group III were higher than Group IV. In Group III, it was higher than Group II. The AR in Group IV was higher, but the LBR was lower than Group I, Group II, and Group III. Logistic regression analysis results suggested that AR in Group IV was higher than that in Group I in IVF cycles, whereas LBR was lower.In ICSI cycles, high-quality embryo rate in Group i and Group ii were both higher than that in Group iii and Group iv. The CPR in Group i was higher than Group ii and Group iv, and in Group iii was higher than Group ii and Group iv. The AR in Group i was lower than Group iii and Group iv, and AR in Group ii was lower than Group iv. LBR, in Group I it was higher than Group ii, Group iii, and Group iv. Logistic regression analysis results suggested CPR in Group ii was significantly lower than that in Group i. AR in Group iii was considerably higher than that in Group i. LBR in Group ii and Group iv were significantly lower than that in Group i. CONCLUSION Female higher BMI was not conducive to the formation of high-quality embryos in ICSI cycle. Female and/or male BMI affected AR and LBR more than CPR not only in IVF cycles, but also in ICSI cycles.
Collapse
|
37
|
Moura FH, Macias-Franco A, Pena-Bello CA, Archilia EC, Batalha IM, Silva AEM, Moreira GM, Norris AB, Schütz LF, Fonseca MA. Sperm DNA 5-methyl cytosine and RNA N6-methyladenosine methylation are differently affected during periods of body weight losses and body weight gain of young and mature breeding bulls. J Anim Sci 2021; 100:6460477. [PMID: 34902028 PMCID: PMC8849232 DOI: 10.1093/jas/skab362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022] Open
Abstract
Aiming to characterize the effects of nutritional status on epigenetic markers, such as DNA 5-methyl cytosine (mC) methylation and RNA N6-methyladenosine (m6A) methylation, of bovine sperm, 12 Angus × Hereford crossbred breeding bulls were submitted to nutritional changes for a period of 180 d: no change in body weight (BW) (phase 1 = 12 d), BW loss (phase 2 = 78 d), and BW gain (phase 3 = 90 d) in a repeated measures design. Animals were fed Beardless wheat (Triticum aestivum) hay and mineral mix. Statistical analyses were performed using SAS 9.4 (SAS Inst., Cary, NC). Higher levels of RNA m6A (P = 0.004) and DNA methylation (P = 0.007) of spermatic cells were observed at phase 2 compared with phase 1. In phase 3, sperm RNA m6A methylation levels continued to be higher (P = 0.004), whereas the DNA of sperm cells was similar (P = 0.426) compared with phase 1. Growing bulls had a tendency (P = 0.109) of higher RNA m6A methylation levels than mature bulls. Phase 2 altered scrotal circumference (P < 0.001), sperm volume (P = 0.007), sperm total motility (P = 0.004), sperm progressive motility (P = 0.004), total sperm count (P = 0.049), normal sperm (P < 0.001), abnormal sperm (P < 0.001), primary sperm defects (P = 0.039), and secondary sperm defects (P < 0.001). In phase 3, bulls had scrotal circumference, sperm volume, sperm motility, sperm progressive motility, total sperm count, normal and abnormal spermatozoa, and primary and secondary spermatozoa defects similar to phase 1 (P > 0.05). Serum concentrations of insulin-like growth factor-1 and leptin decreased during phase 2 (P = 0.010), while no differences (P > 0.05) were detected between phases 3 and 1; growing bulls tended (P = 0.102) to present higher leptin levels than mature bulls. Specific for mature bulls, DNA methylation was positively correlated with leptin concentration (0.569, P = 0.021), whereas for young bulls, DNA methylation was positively correlated with abnormal spermatozoa (0.824, P = 0.006), primary spermatozoa defect (0.711, P = 0.032), and secondary spermatozoa defect (0.661, P = 0.052) and negatively correlated with normal spermatozoa (-0.824, P = 0.006), total sperm count (-0.702, P = 0.035), and sperm concentration (-0.846, P = 0.004). There was no significant correlation (P > 0.05) between RNA m6A and hormones and semen traits. In conclusion, the nutritional status of breeding bulls alters epigenetic markers, such as DNA methylation and RNA m6A methylation, in sperm, and the impact of change seems to be age dependent. These markers may serve as biomarkers of sperm quality and fertility of bulls in the future. Detrimental effects on sperm production and seminal quality are observed at periods and places when and where environmental and nutritional limitations are a year-round reality and may carry hidden players that may influence a lifetime of underperformance.
Collapse
Affiliation(s)
- Felipe H Moura
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Arturo Macias-Franco
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Camilo A Pena-Bello
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Evandro C Archilia
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Isadora M Batalha
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Aghata E M Silva
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Gabriel M Moreira
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Aaron B Norris
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA,Department of Natural Resources Management, Texas Tech University, Lubbock, TX 79430, USA
| | - Luis F Schütz
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA
| | - Mozart A Fonseca
- Department of Animal, Veterinary and Rangeland Sciences, University of Nevada, Reno, Reno, NV 89557, USA,Corresponding author:
| |
Collapse
|
38
|
Jaakkola JM, Rovio SP, Pahkala K, Viikari J, Rönnemaa T, Jula A, Niinikoski H, Mykkänen J, Juonala M, Hutri-Kähönen N, Kähönen M, Lehtimäki T, Raitakari OT. Childhood exposure to parental smoking and life-course overweight and central obesity. Ann Med 2021; 53:208-216. [PMID: 33305629 PMCID: PMC7901689 DOI: 10.1080/07853890.2020.1853215] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/14/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To evaluate the association between childhood parental smoking exposure and the risk of overweight/obesity from childhood to adulthood. METHODS This study leverages the data from two longitudinal population based cohort studies, the Cardiovascular Risk in Young Finns Study between years 1980-2011/2012 (YFS; N = 2,303; baseline age 3-18 years) and the Special Turku Coronary Risk Factor Intervention Project between years 1989-2009/2010 (STRIP; N = 632; baseline age 7 months). Weight, height and waist circumference were measured from childhood to adulthood. Overweight/obesity was defined as body mass index ≥25 kg/m2 in adults and using the Cole criteria in children. Central obesity was defined as waist circumference > 100/90 cm in men/women and as a waist-to-height ratio > 0.50 in children. Statistical analyses were adjusted for age, sex, socioeconomic status, smoking, birth weight, parental ages, diet and physical activity. RESULTS Childhood parental smoking exposure was associated with increased risk for life-course overweight/obesity (YFS: RR1.13, 95%CI 1.02-1.24; STRIP: RR1.57, 95%CI 1.10-2.26) and central obesity (YFS: RR1.18, 95%CI 1.01-1.38; STRIP: RR1.45, 95%CI 0.98-2.15). CONCLUSIONS Childhood exposure to parental smoking is associated with increased risk of overweight/obesity over the life-course. KEY MESSAGES Exposure to parental smoking in childhood was associated with increased risk of overweight/obesity, central obesity and adiposity measured by skinfold thickness from childhood to adulthood.
Collapse
Affiliation(s)
- Johanna M. Jaakkola
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Suvi P. Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Paavo Nurmi Centre, Sports and Exercise Medicine Unit, Department of Health and Physical activity, University of Turku, Turku, Finland
| | - Jorma Viikari
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Tapani Rönnemaa
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Antti Jula
- Department of Chronic Disease Prevention, Institute for Health and Welfare, Turku, Finland
| | - Harri Niinikoski
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Juha Mykkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Markus Juonala
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli T. Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
39
|
López-Cervantes JP, Lønnebotn M, Jogi NO, Calciano L, Kuiper IN, Darby MG, Dharmage SC, Gómez-Real F, Hammer B, Bertelsen RJ, Johannessen A, Würtz AML, Mørkve Knudsen T, Koplin J, Pape K, Skulstad SM, Timm S, Tjalvin G, Krauss-Etschmann S, Accordini S, Schlünssen V, Kirkeleit J, Svanes C. The Exposome Approach in Allergies and Lung Diseases: Is It Time to Define a Preconception Exposome? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:12684. [PMID: 34886409 PMCID: PMC8657011 DOI: 10.3390/ijerph182312684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/30/2022]
Abstract
Emerging research suggests environmental exposures before conception may adversely affect allergies and lung diseases in future generations. Most studies are limited as they have focused on single exposures, not considering that these diseases have a multifactorial origin in which environmental and lifestyle factors are likely to interact. Traditional exposure assessment methods fail to capture the interactions among environmental exposures and their impact on fundamental biological processes, as well as individual and temporal factors. A valid estimation of exposure preconception is difficult since the human reproductive cycle spans decades and the access to germ cells is limited. The exposome is defined as the cumulative measure of external exposures on an organism (external exposome), and the associated biological responses (endogenous exposome) throughout the lifespan, from conception and onwards. An exposome approach implies a targeted or agnostic analysis of the concurrent and temporal multiple exposures, and may, together with recent technological advances, improve the assessment of the environmental contributors to health and disease. This review describes the current knowledge on preconception environmental exposures as related to respiratory health outcomes in offspring. We discuss the usefulness and feasibility of using an exposome approach in this research, advocating for the preconception exposure window to become included in the exposome concept.
Collapse
Affiliation(s)
- Juan Pablo López-Cervantes
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Marianne Lønnebotn
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Nils Oskar Jogi
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
| | - Lucia Calciano
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (L.C.); (S.A.)
| | | | - Matthew G. Darby
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa;
| | - Shyamali C. Dharmage
- School of Population and Global Health, University of Melbourne, Melbourne, VIC 3010, Australia; (S.C.D.); (J.K.)
| | - Francisco Gómez-Real
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5053 Bergen, Norway
| | - Barbara Hammer
- Department of Pulmonology, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Ane Johannessen
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
| | - Anne Mette Lund Würtz
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
| | - Toril Mørkve Knudsen
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
| | - Jennifer Koplin
- School of Population and Global Health, University of Melbourne, Melbourne, VIC 3010, Australia; (S.C.D.); (J.K.)
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Kathrine Pape
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
| | - Svein Magne Skulstad
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Signe Timm
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark;
- Research Unit, Kolding Hospital, University Hospital of Southern Denmark, 6000 Kolding, Denmark
| | - Gro Tjalvin
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | | | - Simone Accordini
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (L.C.); (S.A.)
| | - Vivi Schlünssen
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
- National Research Centre for the Working Environment, 2100 Copenhagen, Denmark
| | - Jorunn Kirkeleit
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Cecilie Svanes
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| |
Collapse
|
40
|
Leckman JF, Ponguta LA, Pavarini G, Hein SD, McCarthy MF, Staiti H, Hanöz-Penney S, Rubinstein J, Pruett KD, Yazgan MY, Fallon NS, Hartl FJ, Ziv M, Salah R, Britto PR, Fitzpatrick S, Panter-Brick C. Love and peace across generations: Biobehavioral systems and global partnerships. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2021; 8:100092. [PMID: 35757671 PMCID: PMC9216554 DOI: 10.1016/j.cpnec.2021.100092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 01/02/2023] Open
Abstract
Children's environments - especially relationships with caregivers - sculpt not only developing brains but also multiple bio-behavioral systems that influence long-term cognitive and socioemotional outcomes, including the ability to empathize with others and interact in prosocial and peaceful ways. This speaks to the importance of investing resources in effective and timely programs that work to enhance early childhood development (ECD) and, by extension, reach communities at-scale. Given the limited resources currently devoted to ECD services, and the devastating impact of COVID-19 on children and communities, there is a clear need to spur government leaders and policymakers to further invest in ECD and related issues including gender and racial equity. This essay offers concrete examples of scholarly paradigms and leadership efforts that focus on child development to build a peaceful, equitable, just, and sustainable world. As scholars and practitioners, we need to continue to design, implement, assess, and revise high-quality child development programs that generate much-needed evidence for policy and programmatic changes. We must also invest in global partnerships to foster the next generation of scholars, practitioners, and advocates dedicated to advance our understanding of the bio-behavioral systems that underlie love, sociality, and peace across generations. Especially where supported by structural interventions, ECD programs can help create more peaceful, just, and socially equitable societies.
Collapse
Affiliation(s)
- James F. Leckman
- Child Study Center, Yale University, New Haven, CT, USA
- Departments of Psychiatry, Psychology and Pediatrics, Yale University, New Haven, CT, USA
- Early Childhood Peace Consortium, New York, NY, USA
- Ethox Centre, Wellcome Centre for Ethics and Humanities, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liliana Angelica Ponguta
- Child Study Center, Yale University, New Haven, CT, USA
- Early Childhood Peace Consortium, New York, NY, USA
| | - Gabriela Pavarini
- Early Childhood Peace Consortium, New York, NY, USA
- Ethox Centre, Wellcome Centre for Ethics and Humanities, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Sascha D. Hein
- Early Childhood Peace Consortium, New York, NY, USA
- Freie Universität, Berlin, Germany
| | - Michael F. McCarthy
- Child Study Center, Yale University, New Haven, CT, USA
- Early Childhood Peace Consortium, New York, NY, USA
- Warner School of Education and Human Development, University of Rochester, Rochester, NY, USA
| | | | - Suna Hanöz-Penney
- Early Childhood Peace Consortium, New York, NY, USA
- Mother Child Education Foundation (AÇEV), Istanbul, Turkey
| | - Joanna Rubinstein
- Early Childhood Peace Consortium, New York, NY, USA
- United Nations Sustainable Development Solutions Network, Paris, France
| | - Kyle D. Pruett
- Child Study Center, Yale University, New Haven, CT, USA
- Early Childhood Peace Consortium, New York, NY, USA
| | - M. Yanki Yazgan
- Child Study Center, Yale University, New Haven, CT, USA
- Early Childhood Peace Consortium, New York, NY, USA
- Güzel Günler Clinic, Istanbul, Turkey
| | - N. Shemrah Fallon
- Child Study Center, Yale University, New Haven, CT, USA
- Early Childhood Peace Consortium, New York, NY, USA
| | - Franz J. Hartl
- Early Childhood Peace Consortium, New York, NY, USA
- University Web Operations, Yale University, New Haven, CT, USA
| | - Margalit Ziv
- The International Networking Group on Peacebuilding with Young Children, Belfast, Northern Ireland, UK
| | - Rima Salah
- Child Study Center, Yale University, New Haven, CT, USA
- Early Childhood Peace Consortium, New York, NY, USA
| | - Pia Rebello Britto
- Early Childhood Peace Consortium, New York, NY, USA
- Country Representative, UNICEF, Lao’s People Democratic Republic (Lao PDR)
| | - Siobhán Fitzpatrick
- Early Childhood Peace Consortium, New York, NY, USA
- The International Networking Group on Peacebuilding with Young Children, Belfast, Northern Ireland, UK
- The Early Years – the Organization for Young Children, Belfast, Northern Ireland, UK
| | - Catherine Panter-Brick
- Early Childhood Peace Consortium, New York, NY, USA
- Jackson Institute for Global Affairs and Department of Anthropology, Yale University, New Haven, CT, USA
| |
Collapse
|
41
|
Hime GR, Stonehouse SLA, Pang TY. Alternative models for transgenerational epigenetic inheritance: Molecular psychiatry beyond mice and man. World J Psychiatry 2021; 11:711-735. [PMID: 34733638 PMCID: PMC8546770 DOI: 10.5498/wjp.v11.i10.711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/19/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Mental illness remains the greatest chronic health burden globally with few in-roads having been made despite significant advances in genomic knowledge in recent decades. The field of psychiatry is constantly challenged to bring new approaches and tools to address and treat the needs of vulnerable individuals and subpopulations, and that has to be supported by a continuous growth in knowledge. The majority of neuropsychiatric symptoms reflect complex gene-environment interactions, with epigenetics bridging the gap between genetic susceptibility and environmental stressors that trigger disease onset and drive the advancement of symptoms. It has more recently been demonstrated in preclinical models that epigenetics underpins the transgenerational inheritance of stress-related behavioural phenotypes in both paternal and maternal lineages, providing further supporting evidence for heritability in humans. However, unbiased prospective studies of this nature are practically impossible to conduct in humans so preclinical models remain our best option for researching the molecular pathophysiologies underlying many neuropsychiatric conditions. While rodents will remain the dominant model system for preclinical studies (especially for addressing complex behavioural phenotypes), there is scope to expand current research of the molecular and epigenetic pathologies by using invertebrate models. Here, we will discuss the utility and advantages of two alternative model organisms-Caenorhabditis elegans and Drosophila melanogaster-and summarise the compelling insights of the epigenetic regulation of transgenerational inheritance that are potentially relevant to human psychiatry.
Collapse
Affiliation(s)
- Gary R Hime
- Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, VIC, Australia
| | - Sophie LA Stonehouse
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville 3052, VIC, Australia
| | - Terence Y Pang
- Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, VIC, Australia
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville 3052, VIC, Australia
| |
Collapse
|
42
|
Tjalvin G, Svanes Ø, Igland J, Bertelsen RJ, Benediktsdóttir B, Dharmage S, Forsberg B, Holm M, Janson C, Jõgi NO, Johannessen A, Malinovschi A, Pape K, Real FG, Sigsgaard T, Torén K, Vindenes HK, Zock JP, Schlünssen V, Svanes C. Maternal preconception occupational exposure to cleaning products and disinfectants and offspring asthma. J Allergy Clin Immunol 2021; 149:422-431.e5. [PMID: 34674855 DOI: 10.1016/j.jaci.2021.08.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/08/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Emerging research suggests health effects in offspring after parental chemical exposures before conception. Many future mothers are exposed to potent chemicals at work, but potential offspring health effects are hardly investigated. OBJECTIVE We sought to investigate childhood asthma in relation to mother's occupational exposure to cleaning products and disinfectants before conception. METHODS The multicenter Respiratory Health In Northern Europe/Respiratory Health In Northern Europe, Spain and Australia generation study investigated asthma and wheeze starting at age less than 10 years in 3318 mother-offspring pairs. From an asthma-specific Job-Exposure Matrix and mothers' occupational history, we defined maternal occupational exposure to indoor cleaning agents (cleaning products/detergents and disinfectants) starting before conception, in the 2-year period around conception and pregnancy, or after birth. Never-employed mothers were excluded. Exposed groups include cleaners, health care workers, cooks, and so forth. Associations were analyzed using mixed-effects logistic regression and ordinary logistic regression with clustered robust SEs and adjustment for maternal education. RESULTS Maternal occupational exposure to indoor cleaning starting preconception and continuing (n = 610) was associated with offspring's childhood asthma: odds ratio 1.56 (95% CI, 1.05-2.31), childhood asthma with nasal allergies: 1.77 (1.13-2.77), and childhood wheeze and/or asthma: 1.71 (95% CI, 1.19-2.44). Exposure starting around conception and pregnancy (n = 77) was associated with increased childhood wheeze and/or asthma: 2.25 (95% CI, 1.03-4.91). Exposure starting after birth was not associated with asthma outcomes (1.13 [95% CI, 0.71-1.80], 1.15 [95% CI, 0.67-1.97], 1.08 [95% CI, 0.69-1.67]). CONCLUSIONS Mother's occupational exposure to indoor cleaning agents starting before conception, or around conception and pregnancy, was associated with more childhood asthma and wheeze in offspring. Considering potential implications for vast numbers of women in childbearing age using cleaning agents, and their children, further research is imperative.
Collapse
Affiliation(s)
- Gro Tjalvin
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway; Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway.
| | - Øistein Svanes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jannicke Igland
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway; Department of Health and Caring Sciences, Faculty of Health and Social Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Randi Jacobsen Bertelsen
- Department of Clinical Science, University of Bergen, Bergen, Norway; Oral Health Center of Expertise in Western Norway, Bergen, Norway
| | - Bryndís Benediktsdóttir
- Medical Faculty, University of Iceland, Reykjavík, Iceland; Department of Sleep, Landspitali University Hospital Reykjavík, Reykjavík, Iceland
| | - Shyamali Dharmage
- Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia
| | - Bertil Forsberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Nils Oskar Jõgi
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway; Tartu University Lung Clinic, Tartu, Estonia
| | - Ane Johannessen
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Kathrine Pape
- National Research Centre for the Working Environment, Aarhus, Denmark; Department of Public Health, Aarhus University, Environment, Work and Health, Danish Ramazzini Centre, Aarhus, Denmark
| | - Francisco Gomez Real
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Torben Sigsgaard
- Department of Public Health, Aarhus University, Environment, Work and Health, Danish Ramazzini Centre, Aarhus, Denmark
| | - Kjell Torén
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hilde Kristin Vindenes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jan-Paul Zock
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Vivi Schlünssen
- National Research Centre for the Working Environment, Aarhus, Denmark; Department of Public Health, Aarhus University, Environment, Work and Health, Danish Ramazzini Centre, Aarhus, Denmark
| | - Cecilie Svanes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| |
Collapse
|
43
|
Stener-Victorin E, Deng Q. Epigenetic inheritance of polycystic ovary syndrome - challenges and opportunities for treatment. Nat Rev Endocrinol 2021; 17:521-533. [PMID: 34234312 DOI: 10.1038/s41574-021-00517-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the main cause of female infertility worldwide and is associated with a substantially increased lifetime risk of comorbidities, including type 2 diabetes mellitus, psychiatric disorders and gynaecological cancers. Despite its high prevalence (~15%) and substantial economic burden, the aetiology of PCOS remains elusive. The genetic loci linked to PCOS so far account for only ~10% of its heritability, which is estimated at 70%. However, growing evidence suggests that altered epigenetic and developmental programming resulting from hormonal dysregulation of the maternal uterine environment contributes to the pathogenesis of PCOS. Male as well as female relatives of women with PCOS are also at an increased risk of developing PCOS-associated reproductive and metabolic disorders. Although PCOS phenotypes are highly heterogenous, hyperandrogenism is thought to be the principal driver of this condition. Current treatments for PCOS are suboptimal as they can only alleviate some of the symptoms; preventative and targeted treatments are sorely needed. This Review presents an overview of the current understanding of the aetiology of PCOS and focuses on the developmental origin and epigenetic inheritance of this syndrome.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
44
|
Ishimwe MCS, Wentzel A, Shoup EM, Osei-Tutu NH, Hormenu T, Patterson AC, Bagheri H, DuBose CW, Mabundo LS, Ha J, Sherman A, Sumner AE. Beta-cell failure rather than insulin resistance is the major cause of abnormal glucose tolerance in Africans: insight from the Africans in America study. BMJ Open Diabetes Res Care 2021; 9:9/1/e002447. [PMID: 34531244 PMCID: PMC8449936 DOI: 10.1136/bmjdrc-2021-002447] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Uncertainties exist on whether the main determinant of abnormal glucose tolerance (Abnl-GT) in Africans is β-cell failure or insulin resistance (IR). Therefore, we determined the prevalence, phenotype and characteristics of Abnl-GT due to β-cell failure versus IR in 486 African-born blacks (male: 64%, age: 38±10 years (mean±SD)) living in America. RESEARCH DESIGN AND METHODS Oral glucose tolerance test were performed. Abnl-GT is a term which includes both diabetes and prediabetes and was defined as fasting plasma glucose (FPG) ≥5.6 mmol/L and/or 2-hour glucose ≥7.8 mmol/L. IR was defined by the lowest quartile of the Matsuda Index (≤2.98) and retested using the upper quartile of homeostatic model assessment of insulin resistance (HOMA-IR) (≥2.07). Abnl-GT-IR required both Abnl-GT and IR. Abnl-GT-β-cell failure was defined as Abnl-GT without IR. Beta-cell compensation was assessed by the Disposition Index (DI). Fasting lipids were measured. Visceral adipose tissue (VAT) volume was obtained with abdominal CT scan. RESULTS The prevalence of Abnl-GT was 37% (182/486). For participants with Abnl-GT, IR occurred in 38% (69/182) and β-cell failure in 62% (113/182). Compared with Africans with Abnl-GT-IR, Africans with Abnl-GT-β-cell failure had lower body mass index (BMI) (30.8±4.3 vs 27.4±4.0 kg/m2), a lower prevalence of obesity (52% vs 19%), less VAT (163±72 vs 107±63 cm2), lower triglyceride (1.21±0.60 vs 0.85±0.42 mmol/L) and lower FPG (5.9±1.4 vs 5.3±0.6 mmol/L) and 2-hour glucose concentrations (10.0±3.1 vs 9.0±1.9 mmol/L) (all p<0.001) and higher DI, high-density lipoprotein (HDL), low-density lipoprotein particle size and HDL particle size (all p<0.01). Analyses with Matsuda Index and HOMA-IR yielded similar results. Potential confounders such as income, education, alcohol and fiber intake did not differ by group. CONCLUSIONS Beta-cell failure occurred in two-thirds of participants with Abnl-GT and may be a more frequent determinant of Abnl-GT in Africans than IR. As BMI category, degree of glycemia and lipid profile appeared more favorable when Abnl-GT was due to β-cell failure rather than IR, the clinical course and optimal interventions may differ. TRIAL REGISTRATION NUMBER NCT00001853.
Collapse
Affiliation(s)
| | | | - Elyssa M Shoup
- NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Thomas Hormenu
- NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Hadi Bagheri
- Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Joon Ha
- NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | - Arthur Sherman
- NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | - Anne E Sumner
- NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
45
|
Hardy I, Lloyd A, Morisset AS, Camirand Lemyre F, Baillargeon JP, Fraser WD. Healthy for My Baby Research Protocol- a Randomized Controlled Trial Assessing a Preconception Intervention to Improve the Lifestyle of Overweight Women and Their Partners. Front Public Health 2021; 9:670304. [PMID: 34414154 PMCID: PMC8369366 DOI: 10.3389/fpubh.2021.670304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/05/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Preconception lifestyle interventions appear promising to reduce pregnancy complications, prevent adult cardiometabolic diseases, and prevent childhood obesity. These interventions have almost exclusively been studied in populations of obese infertile women. The development of preconception lifestyle interventions targeting a broader population of overweight and obese women without a history infertility and their partners is needed. Methods: This study is a multicenter open label parallel group randomized controlled trial. Sixty-eight non-infertile women with overweight or obesity in the preconception period and their partners will be recruited from the Sherbrooke and Quebec City regions. The couples will be randomized in a 1:1 ratio to receive the Healthy for my Baby intervention or standard care in the preconception period and pregnancy. Women and their partners will be invited to take part in this lifestyle intervention which includes motivational interviews and daily self-monitoring of lifestyle goals through a mobile phone application. The primary endpoint of this study is the diet quality of women during the preconception period, which will be evaluated using the C-HEI 2007 score at baseline, 2, 4- and 6-months following study enrolment. Women's dietary quality will also be evaluated through the measure of urinary biomarkers of habitual dietary intake at baseline and 2 months in preconception, and 24–26 weeks in pregnancy. Additional indicators of women's lifestyle as well as anthropometric measures will be documented in preconception and pregnancy. For the pregnancy period, the main secondary endpoint is the pattern of gestational weight gain. Pregnancy and neonatal complications will also be evaluated. For partners, diet quality, other lifestyle habits, and anthropometric measures will be documented in the preconception and pregnancy periods. Discussion: This study will evaluate the effectiveness of a low-cost intervention designed to improve diet and other lifestyle characteristics of women in the preconception period who are overweight or obese. If the Healthy for my Baby intervention is efficacious regarding dietary measures, larger trials will be needed to evaluate the impact of this intervention on the rates of pregnancy complications, childhood obesity, and adult cardiometabolic disease. Clinical Trial Registration:clinicaltrials.gov (NCT04242069).
Collapse
Affiliation(s)
- Isabelle Hardy
- Department of Obstetrics and Gynecology, University of Sherbrooke and Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Amanda Lloyd
- Institute of Biological, Environmental, and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Anne-Sophie Morisset
- School of Nutrition, Faculty of Agricultural and Food Science, Laval University, Laval, QC, Canada
| | - Felix Camirand Lemyre
- Department of Mathematics, University of Sherbrooke and CRCHUS, Sherbrooke, QC, Canada
| | - Jean-Patrice Baillargeon
- Endocrine Division, Department of Medicine, University of Sherbrooke and CRCHUS, Sherbrooke, QC, Canada
| | - William D Fraser
- Department of Obstetrics and Gynecology, University of Sherbrooke and Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| |
Collapse
|
46
|
Birmingham K, Iles-Caven Y, Golding J. ALSPAC parents' descriptions of childhood stresses in their parents and grandparents. Wellcome Open Res 2021; 6:115. [PMID: 35693334 PMCID: PMC9156897 DOI: 10.12688/wellcomeopenres.16732.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2021] [Indexed: 08/29/2023] Open
Abstract
Background: There is evidence that childhood stresses or traumas influence individuals' descendants' health and wellbeing through epigenetic mechanisms. However, few longitudinal studies have details of such ancestral data. Methods: Nearly 7,000 parents of the original Avon Longitudinal Study of Parents and Children (ALSPAC) cohort completed questionnaires concerning their parents' and grandparents' childhoods. Validation of these questionnaires involved conducting recorded interviews with 100 of these parents. Results: The interviews provided insights into the childhoods of two previous generations of this cohort, most of whom had lived through one, if not two, World Wars. Many children were brought up, not by their parents but by relatives or acquaintances and/or left home very young to 'go into service' or start work. A few interviewees had wealthy relatives with nannies and governesses and attended expensive boarding schools but by far the most frequent accounts were of poverty, often severe, with related lack of education and illiteracy, alcoholism and violence, alongside devastating effects of the World Wars. Conclusions: Although the interviews focussed on stresses in childhood and therefore the accounts seemed somewhat negative, many interviewees described their relatives as having secure, stable childhoods. Of the many struggling families though, the predominant impression was their remarkable resilience; all went on to have children or grandchildren who are stable enough to participate for three decades, entirely altruistically, in ALSPAC.
Collapse
Affiliation(s)
- Karen Birmingham
- Centre for Academic Child Health, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Yasmin Iles-Caven
- Centre for Academic Child Health, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Jean Golding
- Centre for Academic Child Health, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| |
Collapse
|
47
|
Mallinson PAC, Kulkarni B, Bhogadi S, Kinra S. Association between parents' socioeconomic conditions and nutritional status during childhood and the risk of cardiovascular disease in their adult offspring: an intergenerational study in south India. J Epidemiol Community Health 2021; 75:1091-1097. [PMID: 33980719 PMCID: PMC8515111 DOI: 10.1136/jech-2020-216261] [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: 12/13/2020] [Revised: 04/09/2021] [Accepted: 04/22/2021] [Indexed: 11/11/2022]
Abstract
Background Some researchers have suggested that parents’ exposure to poor socioeconomic conditions during childhood can increase their offspring’s risk of cardiovascular disease, primarily through poor maternal nutrition and growth. However, epidemiological data on this association are limited. In an intergenerational cohort from rural India, we examined the association of parental childhood socioeconomic conditions and stature with offspring’s cardiovascular risk, hypothesising an inverse association between the two. Methods We analysed data on 3175 adult offspring (aged 18–35 years, 58% men) and their parents from the third wave of the Andhra Pradesh Children and Parents’ Study (2010–12). We used multilevel linear regression to estimate the association of parents’ Standard of Living Index (SLI, an asset-based measure of socioeconomic conditions) in childhood, height and leg length with subclinical atherosclerosis and cardiovascular risk factors in their offspring. Results In multivariable models adjusted for offspring’s socioeconomic conditions in childhood and adulthood, associations (beta coefficients and 95% CIs) of mother’s and father’s childhood SLI (per SD) were −0.00 mm (−0.01, 0.01) and 0.01 mm (−0.00, 0.02) for carotid intima media thickness, −0.17 mm Hg (−0.61, 0.27) and −0.30 mm Hg (−0.78, 0.20) for systolic blood pressure, −0.43 mg/dL (−2.00, 1.15) and −1.07 mg/dL (−2.79, 0.65) for total cholesterol and −0.00mU/L (−0.04, 0.03) and 0.01mU/L (−0.03, 0.04) for log fasting insulin. Results were of similar magnitude for parental height and leg length. Conclusions Our findings do not support an inverse association between parental childhood socioeconomic conditions or stature and offspring’s risk of cardiovascular disease. Intergenerational socioeconomic influences on cardiovascular risk may be of limited public health significance for this setting.
Collapse
Affiliation(s)
| | - Bharati Kulkarni
- Clinical Division, National Institute of Nutrition, Hyderabad, India
| | - Santhi Bhogadi
- Indian Institute of Public Health Hyderabad, Public Health Foundation of India, Hyderabad, India
| | - Sanjay Kinra
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
48
|
Arshad SH, Patil V, Mitchell F, Potter S, Zhang H, Ewart S, Mansfield L, Venter C, Holloway JW, Karmaus WJ. Cohort Profile Update: The Isle of Wight Whole Population Birth Cohort (IOWBC). Int J Epidemiol 2021; 49:1083-1084. [PMID: 32637984 DOI: 10.1093/ije/dyaa068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 03/31/2020] [Indexed: 01/09/2023] Open
Affiliation(s)
- S Hasan Arshad
- David Hide Asthma and Allergy Research Centre, Isle of Wight, St. Mary's Hospital, Newport, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Veeresh Patil
- David Hide Asthma and Allergy Research Centre, Isle of Wight, St. Mary's Hospital, Newport, UK
| | - Frances Mitchell
- David Hide Asthma and Allergy Research Centre, Isle of Wight, St. Mary's Hospital, Newport, UK
| | - Stephen Potter
- David Hide Asthma and Allergy Research Centre, Isle of Wight, St. Mary's Hospital, Newport, UK
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Susan Ewart
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Linda Mansfield
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Carina Venter
- Section of Allergy and Immunology, University of Colorado, Children Hospital Colorado, Denver, CO, USA
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Wilfried J Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| |
Collapse
|
49
|
Čater M, Majdič G. How early maternal deprivation changes the brain and behavior? Eur J Neurosci 2021; 55:2058-2075. [PMID: 33870558 DOI: 10.1111/ejn.15238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 01/30/2023]
Abstract
Early life stress can adversely influence brain development and reprogram brain function and consequently behavior in adult life. Adequate maternal care in early childhood is therefore particularly important for the normal brain development, and adverse early life experiences can lead to altered emotional, behavioral, and neuroendocrine stress responses in the adulthood. As a form of neonatal stress, maternal deprivation/separation is often used in behavioral studies to examine the effects of early life stress and for modeling the development of certain psychiatric disorders and brain pathologies in animal models. The temporary loss of maternal care during the critical postpartum periods remodels the offspring's brain and provokes long-term effects on learning and cognition, the development of mental disorders, aggression, and an increased tendency for the drug abuse. Early life stress through maternal deprivation affects neuroendocrine responses to stress in adolescence and adulthood by dysregulating the hypothalamic-pituitary-adrenal axis and permanently disrupts stress resilience. In this review, we focused on how improper maternal care during early postnatal life affects brain development resulting in modified behavior later in life.
Collapse
Affiliation(s)
- Maša Čater
- Veterinary Faculty, Laboratory for Animal Genomics, Institute for Preclinical Studies, University of Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Gregor Majdič
- Veterinary Faculty, Laboratory for Animal Genomics, Institute for Preclinical Studies, University of Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| |
Collapse
|
50
|
A next-generation sequencing study on mechanisms by which restraint and social instability stresses of male mice alter offspring anxiety-like behavior. Sci Rep 2021; 11:7952. [PMID: 33846458 PMCID: PMC8042048 DOI: 10.1038/s41598-021-87060-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/23/2021] [Indexed: 11/08/2022] Open
Abstract
Pathophysiological mechanisms for depression/anxiety are largely unknown. Evidence for transgenerational transmission of acquired epigenetic marks remains limited. We bred unstressed (US) female mice with adolescently restraint-stressed (RS), social instability-stressed (SI) or US males to produce RS, SI and control F1 offspring, respectively. Compared to controls, while paternal RS decreased anxiety-like behavior (ALB) in both female and male offspring, paternal SI increased ALB only in female offspring. Next-generation sequencing and bioinformatics using RS and SI female offspring identified 5 candidate anxiety-transmitting (CAT) genes; each showed a consistent pattern of DNA methylation from F0 spermatozoa through F1 blastocysts to fetal and adult hippocampi. Further analyses validated 4 CAT genes, demonstrated that paternal SI caused ALB differences between male and female offspring through modifying the CAT genes, and indicated a strong correlation between inflammation and ALB pathogenesis and an important function for intronic DNA methylation in regulating ALB-related genes. In conclusion, this study identified important CAT genes and suggested the possibility that stresses on males might alter offspring's ALB by modifying sperm DNA methylation.
Collapse
|