1
|
Qin H, Yu M, Han N, Zhu M, Li X, Zhou J. Antidepressant effects of esketamine via the BDNF/AKT/mTOR pathway in mice with postpartum depression and their offspring. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110992. [PMID: 38484929 DOI: 10.1016/j.pnpbp.2024.110992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Postpartum depression (PPD) is a serious mental health problem that can negatively affect future generations. BDNF/AKT/mTOR signaling in the frontal lobe and hippocampus in mice is associated with depression, but its role in mice with PPD and their offspring is unknown. This study was aimed at investigating the effects of esketamine (ESK), a drug approved for treatment of refractory depression, on the BDNF/AKT/mTOR pathway in mice with PPD and their offspring. A model of chronic unpredictable mild stress with pregnancy was used. ESK was injected into postpartum mice, and behavioral tests were conducted to predict the severity of symptoms at the end of lactation and in the offspring after adulthood. Both mice with PPD and their offspring showed significant anxiety- and depression-like behaviors that were ameliorated with the ESK intervention. ESK enhanced exploratory behavior in unfamiliar environments, increased the preference for sucrose, and ameliorated the impaired BDNF/AKT/mTOR signaling in the frontal and hippocampal regions in mice. Thus, ESK may have great potential in treating PPD and decreasing the incidence of depression in offspring.
Collapse
Affiliation(s)
- Han Qin
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Miao Yu
- Department of Science Experiment Center, China Medical University, Shenyang, China
| | - Nianjiao Han
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Meilin Zhu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xia Li
- Department of Gynecology, The First Hospital, China Medical University, Shenyang, China.
| | - Jing Zhou
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Vázquez-Sánchez A, Rodríguez-Ríos D, Colín-Castelán D, Molina-Torres J, Ramírez-Chávez E, Romo-Morales GDC, Zaina S, Lund G. Effects of paternal arachidonic acid supplementation on offspring behavior and hypothalamus inflammation markers in the mouse. PLoS One 2024; 19:e0300141. [PMID: 38512839 PMCID: PMC10956830 DOI: 10.1371/journal.pone.0300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Arachidonic acid (AA) is involved in inflammation and plays a role in growth and brain development in infants. We previously showed that exposure of mouse sires to AA for three consecutive generations induces a cumulative change in fatty acid (FA) involved in inflammation and an increase in body and liver weight in the offspring. Here, we tested the hypothesis that paternal AA exposure changes the progeny's behavioral response to a proinflammatory insult, and asked whether tissue-specific FA are associated with that response. Male BALB/c mice were supplemented daily with three doses of AA for 10 days and crossed to non-supplemented females (n = 3/dose). Two-month-old unsupplemented male and female offspring (n = 6/paternal AA dose) were exposed to Gram-negative bacteria-derived lipopolysaccharides (LPS) or saline control two hours prior to open field test (OFT) behavioral analysis and subsequent sacrifice. We probed for significant effects of paternal AA exposure on: OFT behaviors; individual FA content of blood, hypothalamus and hypothalamus-free brain; hypothalamic expression profile of genes related to inflammation (Tnfa, Il1b, Cox1, Cox2) and FA synthesis (Scd1, Elovl6). All parameters were affected by paternal AA supplementation in a sex-specific manner. Paternal AA primed the progeny for behavior associated with increased anxiety, with a marked sex dimorphism: high AA doses acted as surrogate of LPS in males, realigning a number of OFT behaviors that in females were differential between saline and LPS groups. Progeny hypothalamic Scd1, a FA metabolism enzyme with documented pro-inflammatory activity, showed a similar pattern of differential expression between saline and LPS groups at high paternal AA dose in females, that was blunted in males. Progeny FA generally were not affected by LPS, but displayed non-linear associations with paternal AA doses. In conclusion, we document that paternal exposure to AA exerts long-term behavioral and biochemical effects in the progeny in a sex-specific manner.
Collapse
Affiliation(s)
| | | | - Dannia Colín-Castelán
- Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, Leon Campus, Leon, Gto., Mexico
| | - Jorge Molina-Torres
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Irapuato, Mexico
| | | | | | - Silvio Zaina
- Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, Leon Campus, Leon, Gto., Mexico
| | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Irapuato, Mexico
| |
Collapse
|
3
|
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
|
4
|
Cincotta SA, Richardson N, Foecke MH, Laird DJ. Differential susceptibility of male and female germ cells to glucocorticoid-mediated signaling. eLife 2024; 12:RP90164. [PMID: 38226689 PMCID: PMC10945581 DOI: 10.7554/elife.90164] [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] [Indexed: 01/17/2024] Open
Abstract
While physiologic stress has long been known to impair mammalian reproductive capacity through hormonal dysregulation, mounting evidence now suggests that stress experienced prior to or during gestation may also negatively impact the health of future offspring. Rodent models of gestational physiologic stress can induce neurologic and behavioral changes that persist for up to three generations, suggesting that stress signals can induce lasting epigenetic changes in the germline. Treatment with glucocorticoid stress hormones is sufficient to recapitulate the transgenerational changes seen in physiologic stress models. These hormones are known to bind and activate the glucocorticoid receptor (GR), a ligand-inducible transcription factor, thus implicating GR-mediated signaling as a potential contributor to the transgenerational inheritance of stress-induced phenotypes. Here, we demonstrate dynamic spatiotemporal regulation of GR expression in the mouse germline, showing expression in the fetal oocyte as well as the perinatal and adult spermatogonia. Functionally, we find that fetal oocytes are intrinsically buffered against changes in GR signaling, as neither genetic deletion of GR nor GR agonism with dexamethasone altered the transcriptional landscape or the progression of fetal oocytes through meiosis. In contrast, our studies revealed that the male germline is susceptible to glucocorticoid-mediated signaling, specifically by regulating RNA splicing within the spermatogonia, although this does not abrogate fertility. Together, our work suggests a sexually dimorphic function for GR in the germline, and represents an important step towards understanding the mechanisms by which stress can modulate the transmission of genetic information through the germline.
Collapse
Affiliation(s)
- Steven A Cincotta
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San FranciscoSan FranciscoUnited States
| | - Nainoa Richardson
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San FranciscoSan FranciscoUnited States
| | - Mariko H Foecke
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San FranciscoSan FranciscoUnited States
| | - Diana J Laird
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
5
|
Kretschmer M, Fischer V, Gapp K. When Dad's Stress Gets under Kid's Skin-Impacts of Stress on Germline Cargo and Embryonic Development. Biomolecules 2023; 13:1750. [PMID: 38136621 PMCID: PMC10742275 DOI: 10.3390/biom13121750] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Multiple lines of evidence suggest that paternal psychological stress contributes to an increased prevalence of neuropsychiatric and metabolic diseases in the progeny. While altered paternal care certainly plays a role in such transmitted disease risk, molecular factors in the germline might additionally be at play in humans. This is supported by findings on changes to the molecular make up of germ cells and suggests an epigenetic component in transmission. Several rodent studies demonstrate the correlation between paternal stress induced changes in epigenetic modifications and offspring phenotypic alterations, yet some intriguing cases also start to show mechanistic links in between sperm and the early embryo. In this review, we summarise efforts to understand the mechanism of intergenerational transmission from sperm to the early embryo. In particular, we highlight how stress alters epigenetic modifications in sperm and discuss the potential for these modifications to propagate modified molecular trajectories in the early embryo to give rise to aberrant phenotypes in adult offspring.
Collapse
Affiliation(s)
- Miriam Kretschmer
- Laboratory of Epigenetics and Neuroendocrinology, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, 8057 Zürich, Switzerland; (M.K.); (V.F.)
- Neuroscience Center Zurich, ETH Zürich and University of Zürich, 8057 Zürich, Switzerland
| | - Vincent Fischer
- Laboratory of Epigenetics and Neuroendocrinology, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, 8057 Zürich, Switzerland; (M.K.); (V.F.)
- Neuroscience Center Zurich, ETH Zürich and University of Zürich, 8057 Zürich, Switzerland
| | - Katharina Gapp
- Laboratory of Epigenetics and Neuroendocrinology, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, 8057 Zürich, Switzerland; (M.K.); (V.F.)
- Neuroscience Center Zurich, ETH Zürich and University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
6
|
LaDage LD, McCormick GL, Robbins TR, Longwell AS, Langkilde T. The effects of early-life and intergenerational stress on the brain. Proc Biol Sci 2023; 290:20231356. [PMID: 38018110 PMCID: PMC10685117 DOI: 10.1098/rspb.2023.1356] [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: 06/16/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023] Open
Abstract
Stress experienced during ontogeny can have profound effects on the adult phenotype. However, stress can also be experienced intergenerationally, where an offspring's phenotype can be moulded by stress experienced by the parents. Although early-life and intergenerational stress can alter anatomy, physiology, and behaviour, nothing is known about how these stress contexts interact to affect the neural phenotype. Here, we examined how early-life and intergenerational stress affect the brain in eastern fence lizards (Sceloporus undulatus). Some lizard populations co-occur with predatory fire ants, and stress from fire ant attacks exerts intergenerational physiological and behavioural changes in lizards. However, it is unclear if intergenerational stress, or the interaction between intergenerational and early-life stress, modulates the brain. To test this, we captured gravid females from fire ant invaded and uninvaded populations, and subjected offspring to three early-life stress treatments: (1) fire ant attack, (2) corticosterone, or (3) a control. Corticosterone and fire ant attack decreased some aspects of the neural phenotype while population of origin and the interaction of early-life stress and population had no effects on the brain. These results suggest that early-life stressors may better predict adult brain variation than intergenerational stress in this species.
Collapse
Affiliation(s)
- Lara D. LaDage
- Division of Mathematics & Natural Sciences, Penn State Altoona, 3000 Ivyside Dr., Altoona, PA 16601, USA
| | - Gail L. McCormick
- Eberly College of Science, Pennsylvania State University, University Park, PA 16802, USA
| | - Travis R. Robbins
- Department of Biology, University of Nebraska Omaha, 6001 Dodge St., Omaha, NE 68182, USA
| | - Anna S. Longwell
- Division of Mathematics & Natural Sciences, Penn State Altoona, 3000 Ivyside Dr., Altoona, PA 16601, USA
| | - Tracy Langkilde
- Eberly College of Science, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
7
|
Cincotta SA, Richardson N, Foecke MH, Laird DJ. Differential susceptibility of male and female germ cells to glucocorticoid-mediated signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.547215. [PMID: 37425891 PMCID: PMC10327205 DOI: 10.1101/2023.06.30.547215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
While physiologic stress has long been known to impair mammalian reproductive capacity through hormonal dysregulation, mounting evidence now suggests that stress experienced prior to or during gestation may also negatively impact the health of future offspring. Rodent models of gestational physiologic stress can induce neurologic and behavioral changes that persist for up to three generations, suggesting that stress signals can induce lasting epigenetic changes in the germline. Treatment with glucocorticoid stress hormones is sufficient to recapitulate the transgenerational changes seen in physiologic stress models. These hormones are known to bind and activate the glucocorticoid receptor (GR), a ligand-inducible transcription factor, thus implicating GR-mediated signaling as a potential contributor to the transgenerational inheritance of stress-induced phenotypes. Here we demonstrate dynamic spatiotemporal regulation of GR expression in the mouse germline, showing expression in the fetal oocyte as well as the perinatal and adult spermatogonia. Functionally, we find that fetal oocytes are intrinsically buffered against changes in GR signaling, as neither genetic deletion of GR nor GR agonism with dexamethasone altered the transcriptional landscape or the progression of fetal oocytes through meiosis. In contrast, our studies revealed that the male germline is susceptible to glucocorticoid-mediated signaling, specifically by regulating RNA splicing within the spermatogonia, although this does not abrogate fertility. Together, our work suggests a sexually dimorphic function for GR in the germline, and represents an important step towards understanding the mechanisms by which stress can modulate the transmission of genetic information through the germline.
Collapse
Affiliation(s)
- Steven A. Cincotta
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Nainoa Richardson
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Mariko H. Foecke
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Diana J. Laird
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Yeramilli V, Cheddadi R, Benjamin H, Martin C. The Impact of Stress, Microbial Dysbiosis, and Inflammation on Necrotizing Enterocolitis. Microorganisms 2023; 11:2206. [PMID: 37764050 PMCID: PMC10534571 DOI: 10.3390/microorganisms11092206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of intestinal morbidity and mortality in neonates. A large body of work exists; however, the pathogenesis of NEC remains poorly understood. Numerous predictors have been implicated in the development of NEC, with relatively less emphasis on maternal factors. Utilizing human tissue plays a crucial role in enhancing our comprehension of the underlying mechanisms accountable for this devastating disease. In this review, we will discuss how maternal stress affects the pathogenesis of NEC and how changes in the intestinal microbiome can influence the development of NEC. We will also discuss the results of transcriptomics-based studies and analyze the gene expression changes in NEC tissues and other molecular targets associated with the pathogenesis of NEC.
Collapse
Affiliation(s)
| | | | | | - Colin Martin
- Division of Pediatric, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave. S., Lowder Building Suite 300, Birmingham, AL 35233, USA
| |
Collapse
|
9
|
Jyothi AK, Thotakura B, Priyadarshini C S, Subramanian M, Rajila HS. Evidence of alterations in the learning and memory in offspring of stress-induced male rats. J Basic Clin Physiol Pharmacol 2023; 34:473-487. [PMID: 34428362 DOI: 10.1515/jbcpp-2020-0183] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES There is extensive data pointing to offspring outcomes related to maternal life incidents, but there is less research concerning the association between paternal life events and progeny brain development and behaviour. As male gametogenesis is a continuous process, the incidences happening in life can modify the epigenetic regulation, altering the offspring's development and behaviour. The present study evaluates the effects of paternal stress during different life periods on their offspring's learning ability, memory, morphological and biochemical changes in the prefrontal cortex and hippocampus in the rat model. METHODS Four weeks' old male rats were subjected to five variable stressors at the rate of one per day. Stress received male rats were bred with naive female rats for 1 to 3 nights. The offspring's learning and memory were assessed by the Morris water maze test and automated Y maze. Following behavioural studies, offspring were euthanized to examine global DNA methylation, neurotransmitter levels, namely acetylcholine, glutamate in the hippocampus and frontal cortex. RESULTS The offspring of stress-induced animals exhibited a delay in acquiring learning and defect in memory and altered global DNA methylation in the hippocampus (p=0.000124). There was significant reduction of acetylcholine and glutamate levels in hippocampus (p=0.000018, p=0.00001, respectively) and in prefrontal cortex (p=0.00001, p=0.00001, respectively). HPA axis of offspring was altered considerably (p=0.00001). The histomorphometry of the prefrontal cortex and different hippocampal regions revealed a statistically significant (p<0.05) reduction in neuronal numbers in the offspring of stressed animals compared to that of control. These impacts were markedly high in the offspring of fathers who received stress during both pubertal and adult periods. CONCLUSIONS The findings of this study demonstrate that paternal stress can impact offspring learning and memory.
Collapse
Affiliation(s)
- Ashok Kumar Jyothi
- Department of Anatomy, Basaveshwara Medical College and Hospital, Chitradurga, Karnataka, India
- Department of Anatomy, Tagore Medical College & Hospital, Chennai, Tamil Nadu, India
| | - Balaji Thotakura
- Department of Anatomy, Chettinad Academy of Research and Education, Chennai, Chennai, Tamil Nadu, India
| | | | - Manickam Subramanian
- Department of Anatomy, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | | |
Collapse
|
10
|
Raymann S, Schalbetter SM, Schaer R, Bernhardt AC, Mueller FS, Meyer U, Weber-Stadlbauer U. Late prenatal immune activation in mice induces transgenerational effects via the maternal and paternal lineages. Cereb Cortex 2023; 33:2273-2286. [PMID: 36857721 DOI: 10.1093/cercor/bhac207] [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: 12/23/2021] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 11/14/2022] Open
Abstract
Prenatal exposure to infectious or noninfectious immune activation is an environmental risk factor for neurodevelopmental disorders and mental illnesses. Recent research using animal models suggests that maternal immune activation (MIA) during early to middle stages of pregnancy can induce transgenerational effects on brain and behavior, likely via inducing stable epigenetic modifications across generations. Using a mouse model of viral-like MIA, which is based on gestational treatment with poly(I:C), the present study explored whether transgenerational effects can also emerge when MIA occurs in late pregnancy. Our findings demonstrate that the direct descendants born to poly(I:C)-treated mothers display deficits in temporal order memory, which are similarly present in second- and third-generation offspring. These transgenerational effects were mediated via both the maternal and paternal lineages and were accompanied by transient changes in maternal care. In addition to the cognitive effects, late prenatal immune activation induced generation-spanning effects on the prefrontal expression of gamma-aminobutyric acid (GABA)ergic genes, including parvalbumin and distinct alpha-subunits of the GABAA receptor. Together, our results suggest that MIA in late pregnancy has the potential to affect cognitive functions and prefrontal gene expression patterns in multiple generations, highlighting its role in shaping disease risk across generations.
Collapse
Affiliation(s)
- Stephanie Raymann
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Ron Schaer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Alexandra C Bernhardt
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
11
|
Preconception paternal mental disorders and child health: Mechanisms and interventions. Neurosci Biobehav Rev 2023; 144:104976. [PMID: 36435393 DOI: 10.1016/j.neubiorev.2022.104976] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Mental illness is a significant global health issue with a steady prevalence. High heritability is suspected, but genome-wide association studies only identified a small number of risk genes associated with mental disorders. This 'missing inheritance' can be partially explained by epigenetic heredity. Evidence from numerous animal models and human studies supports the possibility that preconception paternal mental health influences their offspring's mental health via nongenetic means. Here, we review two potential pathways, including sperm epigenetics and seminal plasma components. The current review highlights the role of sperm epigenetics and explores epigenetic message origination and susceptibility to chronic stress. Meanwhile, possible spatiotemporal windows and events that induce sexually dimorphic modes and effects of paternal stress transmission are inferred in this review. Additionally, we discuss emerging interventions that could potentially block the intergenerational transmission of paternal psychiatric disorders and reduce the incidence of mental illness. Understanding the underlying mechanisms by which preconception paternal stress impacts offspring health is critical for identifying strategies supporting healthy development and successfully controlling the prevalence of mental illness.
Collapse
|
12
|
Švorcová J. Transgenerational Epigenetic Inheritance of Traumatic Experience in Mammals. Genes (Basel) 2023; 14:120. [PMID: 36672861 PMCID: PMC9859285 DOI: 10.3390/genes14010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, we have seen an increasing amount of evidence pointing to the existence of a non-genetic heredity of the effects of events such as separation from parents, threat to life, or other traumatising experiences such as famine. This heredity is often mediated by epigenetic regulations of gene expression and may be transferred even across several generations. In this review, we focus on studies which involve transgenerational epigenetic inheritance (TEI), with a short detour to intergenerational studies focused on the inheritance of trauma or stressful experiences. The reviewed studies show a plethora of universal changes which stress exposure initiates on multiple levels of organisation ranging from hormonal production and the hypothalamic-pituitary-adrenal (HPA) axis modulation all the way to cognition, behaviour, or propensity to certain psychiatric or metabolic disorders. This review will also provide an overview of relevant methodology and difficulties linked to implementation of epigenetic studies. A better understanding of these processes may help us elucidate the evolutionary pathways which are at work in the course of emergence of the diseases and disorders associated with exposure to trauma, either direct or in a previous generation.
Collapse
Affiliation(s)
- Jana Švorcová
- Department of Philosophy and History of Science, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| |
Collapse
|
13
|
Vinogradov S, Chafee MV, Lee E, Morishita H. Psychosis spectrum illnesses as disorders of prefrontal critical period plasticity. Neuropsychopharmacology 2023; 48:168-185. [PMID: 36180784 PMCID: PMC9700720 DOI: 10.1038/s41386-022-01451-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 01/05/2023]
Abstract
Emerging research on neuroplasticity processes in psychosis spectrum illnesses-from the synaptic to the macrocircuit levels-fill key gaps in our models of pathophysiology and open up important treatment considerations. In this selective narrative review, we focus on three themes, emphasizing alterations in spike-timing dependent and Hebbian plasticity that occur during adolescence, the critical period for prefrontal system development: (1) Experience-dependent dysplasticity in psychosis emerges from activity decorrelation within neuronal ensembles. (2) Plasticity processes operate bidirectionally: deleterious environmental and experiential inputs shape microcircuits. (3) Dysregulated plasticity processes interact across levels of scale and time and include compensatory mechanisms that have pathogenic importance. We present evidence that-given the centrality of progressive dysplastic changes, especially in prefrontal cortex-pharmacologic or neuromodulatory interventions will need to be supplemented by corrective learning experiences for the brain if we are to help people living with these illnesses to fully thrive.
Collapse
Affiliation(s)
- Sophia Vinogradov
- Department of Psychiatry & Behavioral Science, University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Matthew V Chafee
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Erik Lee
- Masonic Institute for the Developing Brain, University of Minnesota Medical School, Minneapolis, MN, USA
- University of Minnesota Informatics Institute, University of Minnesota, Minneapolis, MN, USA
| | - Hirofumi Morishita
- Department of Psychiatry, Neuroscience, & Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
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
|
15
|
Korgan AC, Foxx CL, Hashmi H, Sago SA, Stamper CE, Heinze JD, O'Leary E, King JL, Perrot TS, Lowry CA, Weaver ICG. Effects of paternal high-fat diet and maternal rearing environment on the gut microbiota and behavior. Sci Rep 2022; 12:10179. [PMID: 35715467 PMCID: PMC9205913 DOI: 10.1038/s41598-022-14095-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Exposing a male rat to an obesogenic high-fat diet (HFD) influences attractiveness to potential female mates, the subsequent interaction of female mates with infant offspring, and the development of stress-related behavioral and neural responses in offspring. To examine the stomach and fecal microbiome's potential roles, fecal samples from 44 offspring and stomach samples from offspring and their fathers were collected and bacterial community composition was studied by 16 small subunit ribosomal RNA (16S rRNA) gene sequencing. Paternal diet (control, high-fat), maternal housing conditions (standard or semi-naturalistic housing), and maternal care (quality of nursing and other maternal behaviors) affected the within-subjects alpha-diversity of the offspring stomach and fecal microbiomes. We provide evidence from beta-diversity analyses that paternal diet and maternal behavior induced community-wide shifts to the adult offspring gut microbiome. Additionally, we show that paternal HFD significantly altered the adult offspring Firmicutes to Bacteroidetes ratio, an indicator of obesogenic potential in the gut microbiome. Additional machine-learning analyses indicated that microbial species driving these differences converged on Bifidobacterium pseudolongum. These results suggest that differences in early-life care induced by paternal diet and maternal care significantly influence the microbiota composition of offspring through the microbiota-gut-brain axis, having implications for adult stress reactivity.
Collapse
Affiliation(s)
- Austin C Korgan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Christine L Foxx
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
- Oak Ridge Institute for Science and Education Research Participation Program, Oak Ridge, TN, 37830, USA
- U.S. Department of Agriculture (USDA), National Animal Health Laboratory Network (NAHLN), Animal and Plant Health Inspection Service (APHIS), Ames, IA, 50010, USA
| | - Heraa Hashmi
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Saydie A Sago
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Christopher E Stamper
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
- Rocky Mountain MIRECC for Veteran Suicide Prevention, 1700 N Wheeling St, G-3-116M, Aurora, CO, 80045, USA
| | - Jared D Heinze
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Elizabeth O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Jillian L King
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Tara S Perrot
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, 80309, USA
- Department of Psychology and Neuroscience and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), The Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, 80045, USA
- Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Aurora, CO, 80045, USA
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Department of Psychiatry, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
16
|
Effects of early life adversities upon memory processes and cognition in rodent models. Neuroscience 2022; 497:282-307. [PMID: 35525496 DOI: 10.1016/j.neuroscience.2022.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 01/14/2023]
Abstract
Exposure to stressors in early postnatal life induces long-lasting modifications in brainfunction.Thisplasticity,an essential characteristic of the brain that enables adaptation to the environment, may also induce impairments in some psychophysiological functions, including learning and memory. Early life stress (ELS) has long-term effects on thehypothalamic-pituitary-adrenal axisresponse to stressors, and has been reported to lead toneuroinflammation,altered levelsof neurotrophic factors, modifications inneurogenesis andsynaptic plasticity,with changes in neurotransmitter systems and network functioning. In this review, we focus on early postnatal stress in animal models and their effects on learning and memory.Many studies have reported ELS-induced impairments in different types of memories, including spatial memory, fear memory, recognition (both for objects and social) memory, working memory and reversal learning. Studies are not always in agreement, however, no effects, or sometimes facilitation, being reported, depending on the nature and intensity of the early intervention, as well as the age when the outcome was evaluated and the sex of the animals. When considering processes occurring after consolidation, related with memory maintenance or modification, there are a very reduced number of reports. Future studies addressing the mechanisms underlying memory changes for ELS should shed some light on the understanding of the different effects induced by stressors of different types and intensities on cognitive functions.
Collapse
|
17
|
Varela RB, Cararo JH, Tye SJ, Carvalho AF, Valvassori SS, Fries GR, Quevedo J. Contributions of epigenetic inheritance to the predisposition of major psychiatric disorders: theoretical framework, evidence, and implications. Neurosci Biobehav Rev 2022; 135:104579. [DOI: 10.1016/j.neubiorev.2022.104579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/10/2022] [Accepted: 02/11/2022] [Indexed: 02/08/2023]
|
18
|
Paternal environmental exposure-induced spermatozoal small noncoding RNA alteration meditates the intergenerational epigenetic inheritance of multiple diseases. Front Med 2021; 16:176-184. [PMID: 34515940 DOI: 10.1007/s11684-021-0885-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022]
Abstract
Studies of human and mammalian have revealed that environmental exposure can affect paternal health conditions as well as those of the offspring. However, studies that explore the mechanisms that meditate this transmission are rare. Recently, small noncoding RNAs (sncRNAs) in sperm have seemed crucial to this transmission due to their alteration in sperm in response to environmental exposure, and the methodology of microinjection of isolated total RNA or sncRNAs or synthetically identified sncRNAs gradually lifted the veil of sncRNA regulation during intergenerational inheritance along the male line. Hence, by reviewing relevant literature, this study intends to answer the following research concepts: (1) paternal environmental factors that can be passed on to offspring and are attributed to spermatozoal sncRNAs, (2) potential role of paternal spermatozoal sncRNAs during the intergenerational inheritance process, and (3) the potential mechanism by which spermatozoal sncRNAs meditate intergenerational inheritance. In summary, increased attention highlights the hidden wonder of spermatozoal sncRNAs during intergenerational inheritance. Therefore, in the future, more studies should focus on the origin of RNA alteration, the target of RNA regulation, and how sncRNA regulation during embryonic development can be sustained even in adult offspring.
Collapse
|
19
|
Duffy KA, Bale TL, Epperson CN. Germ Cell Drivers: Transmission of Preconception Stress Across Generations. Front Hum Neurosci 2021; 15:642762. [PMID: 34322003 PMCID: PMC8311293 DOI: 10.3389/fnhum.2021.642762] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Abstract
Exposure to stress can accelerate maturation and hasten reproduction. Although potentially adaptive, the trade-off is higher risk for morbidity and mortality. In humans, the intergenerational effects of stress have been demonstrated, but the precise mechanisms are unknown. Strikingly, even if parental stress occurs prior to conception, as adults, their offspring show worse mental and physical health. Emerging evidence primarily from preclinical models suggests that epigenetic programming may encode preconception stress exposures in germ cells, potentially impacting the phenotype of the offspring. In this narrative review, we evaluate the strength of the evidence for this mechanism across animals and humans in both males and females. The strongest evidence comes from studies of male mice, in which paternal preconception stress is associated with a host of phenotypic changes in the offspring and stress-induced changes in the small non-coding RNA content in sperm have been implicated. Two recent studies in men provide evidence that some small non-coding RNAs in sperm are responsive to past and current stress, including some of the same ones identified in mice. Although preliminary evidence suggests that findings from mice may map onto men, the next steps will be (1) considering whether stress type, severity, duration, and developmental timing affect germ cell epigenetic markers, (2) determining whether germ cell epigenetic markers contribute to disease risk in the offspring of stress-exposed parents, and (3) overcoming methodological challenges in order to extend this research to females.
Collapse
Affiliation(s)
- Korrina A. Duffy
- Colorado Center for Women’s Behavioral Health and Wellness, Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, United States
| | - Tracy L. Bale
- Center for Epigenetic Research in Child Health and Brain Development, Department of Pharmacology and Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - C. Neill Epperson
- Colorado Center for Women’s Behavioral Health and Wellness, Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Family Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Center for Women’s Health Research, University of Colorado School of Medicine, Aurora, CO, United States
- Helen and Arthur E. Johnson Depression Center, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
20
|
Kringel D, Malkusch S, Lötsch J. Drugs and Epigenetic Molecular Functions. A Pharmacological Data Scientometric Analysis. Int J Mol Sci 2021; 22:7250. [PMID: 34298869 PMCID: PMC8311652 DOI: 10.3390/ijms22147250] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Interactions of drugs with the classical epigenetic mechanism of DNA methylation or histone modification are increasingly being elucidated mechanistically and used to develop novel classes of epigenetic therapeutics. A data science approach is used to synthesize current knowledge on the pharmacological implications of epigenetic regulation of gene expression. Computer-aided knowledge discovery for epigenetic implications of current approved or investigational drugs was performed by querying information from multiple publicly available gold-standard sources to (i) identify enzymes involved in classical epigenetic processes, (ii) screen original biomedical scientific publications including bibliometric analyses, (iii) identify drugs that interact with epigenetic enzymes, including their additional non-epigenetic targets, and (iv) analyze computational functional genomics of drugs with epigenetic interactions. PubMed database search yielded 3051 hits on epigenetics and drugs, starting in 1992 and peaking in 2016. Annual citations increased to a plateau in 2000 and show a downward trend since 2008. Approved and investigational drugs in the DrugBank database included 122 compounds that interacted with 68 unique epigenetic enzymes. Additional molecular functions modulated by these drugs included other enzyme interactions, whereas modulation of ion channels or G-protein-coupled receptors were underrepresented. Epigenetic interactions included (i) drug-induced modulation of DNA methylation, (ii) drug-induced modulation of histone conformations, and (iii) epigenetic modulation of drug effects by interference with pharmacokinetics or pharmacodynamics. Interactions of epigenetic molecular functions and drugs are mutual. Recent research activities on the discovery and development of novel epigenetic therapeutics have passed successfully, whereas epigenetic effects of non-epigenetic drugs or epigenetically induced changes in the targets of common drugs have not yet received the necessary systematic attention in the context of pharmacological plasticity.
Collapse
Affiliation(s)
- Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Sebastian Malkusch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
21
|
Breton CV, Landon R, Kahn LG, Enlow MB, Peterson AK, Bastain T, Braun J, Comstock SS, Duarte CS, Hipwell A, Ji H, LaSalle JM, Miller RL, Musci R, Posner J, Schmidt R, Suglia SF, Tung I, Weisenberger D, Zhu Y, Fry R. Exploring the evidence for epigenetic regulation of environmental influences on child health across generations. Commun Biol 2021; 4:769. [PMID: 34158610 PMCID: PMC8219763 DOI: 10.1038/s42003-021-02316-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 06/03/2021] [Indexed: 02/08/2023] Open
Abstract
Environmental exposures, psychosocial stressors and nutrition are all potentially important influences that may impact health outcomes directly or via interactions with the genome or epigenome over generations. While there have been clear successes in large-scale human genetic studies in recent decades, there is still a substantial amount of missing heritability to be elucidated for complex childhood disorders. Mounting evidence, primarily in animals, suggests environmental exposures may generate or perpetuate altered health outcomes across one or more generations. One putative mechanism for these environmental health effects is via altered epigenetic regulation. This review highlights the current epidemiologic literature and supporting animal studies that describe intergenerational and transgenerational health effects of environmental exposures. Both maternal and paternal exposures and transmission patterns are considered, with attention paid to the attendant ethical, legal and social implications.
Collapse
Affiliation(s)
- Carrie V Breton
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Remy Landon
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Linda G Kahn
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Michelle Bosquet Enlow
- Department of Psychiatry, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alicia K Peterson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Theresa Bastain
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Joseph Braun
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Sarah S Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Cristiane S Duarte
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - Alison Hipwell
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hong Ji
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, Davis, CA, USA
| | | | - Rashelle Musci
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jonathan Posner
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - Rebecca Schmidt
- Department of Public Health Sciences, UC Davis School of Medicine, Davis, CA, USA
| | | | - Irene Tung
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Weisenberger
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California and Department of Epidemiology and Biostatistics, University of California, San Francisco, Oakland, CA, USA
| | - Rebecca Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, UNC Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
22
|
Epigenetic Mechanisms of Paternal Stress in Offspring Development and Diseases. Int J Genomics 2021; 2021:6632719. [PMID: 33532485 PMCID: PMC7837765 DOI: 10.1155/2021/6632719] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 01/10/2023] Open
Abstract
The major biological function of the sperm cell is to transmit the paternal genetic and epigenetic information to the embryo as well as the following offspring. Sperm has a unique epigenome. An increasing body of epidemiological study supports that paternal stress induced by environmental exposures and lifestyle can modulate the sperm epigenome (including histone modification, DNA methylation, and noncoding RNA expression), sperm-egg fusion, embryo development, and offspring health. Based on the existing literature, we have summarized the paternal exposure on sperm epigenome along with the representative phenotypes of offspring and the possible mechanism involved.
Collapse
|
23
|
Xu S, Zhang Y, Xu Z, Song L. Effect of the cPKCγ-Ng Signaling System on Rapid Eye Movement Sleep Deprivation-Induced Learning and Memory Impairment in Rats. Front Psychiatry 2021; 12:763032. [PMID: 34777065 PMCID: PMC8586205 DOI: 10.3389/fpsyt.2021.763032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Rapid eye movement sleep deprivation (REM-SD) can cause a decline in learning and memory and lead to changes in behavior. Therefore, REM sleep plays a key role in processes that govern learning and memory. However, the mechanism underlying REM-SD-induced learning and memory impairment is unclear and the underlying molecular signaling still needs to be identified. In the present study, we investigated the role of the cPKCγ-Ng signaling pathway in REM-SD-induced learning and memory impairment. Method: Sixty male rats were divided into Control, REM-SD, REM-SD+cPKCγ activator PMA, REM-SD+cPKCγ inhibitor H-7, and sleep revival (SR) groups. The Morris water maze was used to assess spatial learning and memory. Western blot analysis was used to detect cPKCγ total protein expression and membrane translocation levels, and Ng total protein expression and phosphorylation levels. Results: The REM-SD group performed worse on the Morris water maze test than the control group. Western blot analysis showed that cPKCγ membrane translocation and Ng phosphorylation levels were significantly lower in the REM-SD group. SR following REM-SD restored learning and memory ability, cPKCγ transmembrane translocation, and Ng phosphorylation levels, but not to levels observed before REM-SD. PMA and H-7 significantly improved/disrupted task ability as well as cPKCγ transmembrane translocation and Ng phosphorylation levels in REM-SD rats. Conclusion: The REM-SD induced learning and memory impairment in rats and may be associated with the cPKCγ-Ng signaling pathway. Specifically, activation of the cPKCγ-Ng signaling pathway may protect against REM-SD.
Collapse
Affiliation(s)
- Shu Xu
- Neurorehabilitation Center, Beijing Boai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Yanbo Zhang
- Department of Neurobiology, The Second Affiliation Hospital of Shandong First Medical University, Tai'an, China
| | - Zhiqing Xu
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Luping Song
- Neurorehabilitation Center, Beijing Boai Hospital, China Rehabilitation Research Center, Beijing, China.,Department of Rehabilitation Medicine, Shenzhen University General Hospital, Shenzhen, China
| |
Collapse
|
24
|
van Steenwyk G, Gapp K, Jawaid A, Germain P, Manuella F, Tanwar DK, Zamboni N, Gaur N, Efimova A, Thumfart KM, Miska EA, Mansuy IM. Involvement of circulating factors in the transmission of paternal experiences through the germline. EMBO J 2020; 39:e104579. [PMID: 33034389 PMCID: PMC7705452 DOI: 10.15252/embj.2020104579] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Environmental factors can change phenotypes in exposed individuals and offspring and involve the germline, likely via biological signals in the periphery that communicate with germ cells. Here, using a mouse model of paternal exposure to traumatic stress, we identify circulating factors involving peroxisome proliferator-activated receptor (PPAR) pathways in the effects of exposure to the germline. We show that exposure alters metabolic functions and pathways, particularly lipid-derived metabolites, in exposed fathers and their offspring. We collected data in a human cohort exposed to childhood trauma and observed similar metabolic alterations in circulation, suggesting conserved effects. Chronic injection of serum from trauma-exposed males into controls recapitulates metabolic phenotypes in the offspring. We identify lipid-activated nuclear receptors PPARs as potential mediators of the effects from father to offspring. Pharmacological PPAR activation in vivo reproduces metabolic dysfunctions in the offspring and grand-offspring of injected males and affects the sperm transcriptome in fathers and sons. In germ-like cells in vitro, both serum and PPAR agonist induce PPAR activation. Together, these results highlight the role of circulating factors as potential communication vectors between the periphery and the germline.
Collapse
Affiliation(s)
- Gretchen van Steenwyk
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
| | - Katharina Gapp
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
- Laboratory of Molecular and Behavioral NeuroscienceETH ZurichZurichSwitzerland
- Gurdon InstituteUniversity of CambridgeCambridgeUK
- Wellcome Trust Sanger InstituteHinxtonUK
- Department of GeneticsUniversity of CambridgeCambridgeUK
| | - Ali Jawaid
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
- Laboratory of Translational Research in Neuropsychiatric DisordersBRAINCITY Nencki‐EMBL Center of Excellence for Neural Plasticity and Brain DisordersWarsawPoland
| | - Pierre‐Luc Germain
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Statistical Bioinformatics GroupSwiss Institute of BioinformaticsZürichSwitzerland
| | - Francesca Manuella
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
| | - Deepak K Tanwar
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
- Statistical Bioinformatics GroupSwiss Institute of BioinformaticsZürichSwitzerland
| | - Nicola Zamboni
- Institute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Niharika Gaur
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
| | - Anastasiia Efimova
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
| | - Kristina M Thumfart
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
| | - Eric A Miska
- Gurdon InstituteUniversity of CambridgeCambridgeUK
- Wellcome Trust Sanger InstituteHinxtonUK
- Department of GeneticsUniversity of CambridgeCambridgeUK
| | - Isabelle M Mansuy
- Laboratory of NeuroepigeneticsBrain Research InstituteMedical Faculty of the University of ZurichZurichSwitzerland
- Institute for NeuroscienceDepartment of Health Sciences and TechnologyETH ZurichZurichSwitzerland
- Zurich Neuroscience CenterETH Zurich and University of ZurichZurichSwitzerland
| |
Collapse
|
25
|
Rutkowska J, Lagisz M, Bonduriansky R, Nakagawa S. Mapping the past, present and future research landscape of paternal effects. BMC Biol 2020; 18:183. [PMID: 33246472 PMCID: PMC7694421 DOI: 10.1186/s12915-020-00892-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although in all sexually reproducing organisms an individual has a mother and a father, non-genetic inheritance has been predominantly studied in mothers. Paternal effects have been far less frequently studied, until recently. In the last 5 years, research on environmentally induced paternal effects has grown rapidly in the number of publications and diversity of topics. Here, we provide an overview of this field using synthesis of evidence (systematic map) and influence (bibliometric analyses). RESULTS We find that motivations for studies into paternal effects are diverse. For example, from the ecological and evolutionary perspective, paternal effects are of interest as facilitators of response to environmental change and mediators of extended heredity. Medical researchers track how paternal pre-fertilization exposures to factors, such as diet or trauma, influence offspring health. Toxicologists look at the effects of toxins. We compare how these three research guilds design experiments in relation to objects of their studies: fathers, mothers and offspring. We highlight examples of research gaps, which, in turn, lead to future avenues of research. CONCLUSIONS The literature on paternal effects is large and disparate. Our study helps in fostering connections between areas of knowledge that develop in parallel, but which could benefit from the lateral transfer of concepts and methods.
Collapse
Affiliation(s)
- Joanna Rutkowska
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Kraków, Poland
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Malgorzata Lagisz
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Russell Bonduriansky
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Shinichi Nakagawa
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| |
Collapse
|
26
|
Karlsson H, Merisaari H, Karlsson L, Scheinin NM, Parkkola R, Saunavaara J, Lähdesmäki T, Lehtola SJ, Keskinen M, Pelto J, Lewis JD, Tuulari JJ. Association of Cumulative Paternal Early Life Stress With White Matter Maturation in Newborns. JAMA Netw Open 2020; 3:e2024832. [PMID: 33231637 PMCID: PMC7686861 DOI: 10.1001/jamanetworkopen.2020.24832] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
IMPORTANCE Early life stress (ELS) has been shown to affect brain development and health outcomes. Recent animal studies have linked paternal early stress exposures with next-generation outcomes. Epigenetic inheritance through the male germline has been suggested to be one of the mechanisms. OBJECTIVES To test whether paternal ELS, as measured using the Trauma and Distress Scale, is associated with neonate brain development. DESIGN, SETTING, AND PARTICIPANTS This cohort study included data from participants from the prospective 2-generation FinnBrain Birth Cohort, which was collected from 2011 to 2015. Pregnant women and the fathers were consecutively recruited at gestational week 12 from maternity clinics in Finland. Magnetic resonance imaging data were analyzed in 2019. Participants in this study were 72 families (infant, father, mother). EXPOSURE Paternal exposure to ELS. MAIN OUTCOMES AND MEASURES Fractional anisotropy (FA) values in the major white-matter tracts of the newborn brain. RESULTS A total of 72 trios (infant, mother, and father) were analyzed. At the time of delivery, the mean (SD) age was 31.0 (4.4) years for fathers and 30.3 (4.5) years for mothers. Forty-one infants (57%) were boys; mean (SD) child age at inclusion was 26.9 (7.2) days from birth and 205 (8) days from estimated conception. Increasing levels of paternal ELS were associated with higher FA values in the newborn brain in the body of the corpus callosum, right superior corona radiata, and retrolenticular parts of the internal capsule. This association persisted after controlling for maternal ELS, maternal socioeconomic status (SES), maternal body mass index, maternal depressive symptoms during pregnancy, child sex, and child age from birth and gestation corrected age when imaged. In additional region-of-interest analyses, the association between FA values and paternal Trauma and Distress Scale sum scores remained statistically significant in the earliest maturing regions of the brain, eg, the genu of the corpus callosum (in the regression models, β = 0.00096; 95% CI, 0.00034-0.00158; P = .003) and the splenium (β = 0.00090; 95% CI, 0.00000-0.00180; P = .049). CONCLUSIONS AND RELEVANCE This cohort study found a statistically significant association between paternal ELS and offspring brain development. This finding may have far-reaching implications in pediatrics, as it suggests the possibility of a novel route of intergenerational inheritance of ELS on next-generation brain development.
Collapse
Affiliation(s)
- Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Harri Merisaari
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
- Department of Child Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Noora M. Scheinin
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Jani Saunavaara
- Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Tuire Lähdesmäki
- Department of Pediatric Neurology, Turku University Hospital and University of Turku, Turku, Finland
| | - Satu J. Lehtola
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Maria Keskinen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Juho Pelto
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - John D. Lewis
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jetro J. Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
- Turku Collegium for Science and Medicine, University of Turku, Turku, Finland
| |
Collapse
|
27
|
Reh RK, Dias BG, Nelson CA, Kaufer D, Werker JF, Kolb B, Levine JD, Hensch TK. Critical period regulation across multiple timescales. Proc Natl Acad Sci U S A 2020; 117:23242-23251. [PMID: 32503914 PMCID: PMC7519216 DOI: 10.1073/pnas.1820836117] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Brain plasticity is dynamically regulated across the life span, peaking during windows of early life. Typically assessed in the physiological range of milliseconds (real time), these trajectories are also influenced on the longer timescales of developmental time (nurture) and evolutionary time (nature), which shape neural architectures that support plasticity. Properly sequenced critical periods of circuit refinement build up complex cognitive functions, such as language, from more primary modalities. Here, we consider recent progress in the biological basis of critical periods as a unifying rubric for understanding plasticity across multiple timescales. Notably, the maturation of parvalbumin-positive (PV) inhibitory neurons is pivotal. These fast-spiking cells generate gamma oscillations associated with critical period plasticity, are sensitive to circadian gene manipulation, emerge at different rates across brain regions, acquire perineuronal nets with age, and may be influenced by epigenetic factors over generations. These features provide further novel insight into the impact of early adversity and neurodevelopmental risk factors for mental disorders.
Collapse
Affiliation(s)
- Rebecca K Reh
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Brian G Dias
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA 30322
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329
| | - Charles A Nelson
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Graduate School of Education, Harvard University, Cambridge, MA 02138
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Janet F Werker
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Bryan Kolb
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Joel D Levine
- Department of Biology, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Takao K Hensch
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115;
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, Cambridge, MA 02138
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Tokyo 113-0033, Japan
| |
Collapse
|
28
|
Gapp K, van Steenwyk G, Germain PL, Matsushima W, Rudolph KLM, Manuella F, Roszkowski M, Vernaz G, Ghosh T, Pelczar P, Mansuy IM, Miska EA. Alterations in sperm long RNA contribute to the epigenetic inheritance of the effects of postnatal trauma. Mol Psychiatry 2020; 25:2162-2174. [PMID: 30374190 PMCID: PMC7473836 DOI: 10.1038/s41380-018-0271-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/11/2018] [Indexed: 12/22/2022]
Abstract
Psychiatric diseases have a strong heritable component known to not be restricted to DNA sequence-based genetic inheritance alone but to also involve epigenetic factors in germ cells. Initial evidence suggested that sperm RNA is causally linked to the transmission of symptoms induced by traumatic experiences. Here, we show that alterations in long RNA in sperm contribute to the inheritance of specific trauma symptoms. Injection of long RNA fraction from sperm of males exposed to postnatal trauma recapitulates the effects on food intake, glucose response to insulin and risk-taking in adulthood whereas the small RNA fraction alters body weight and behavioural despair. Alterations in long RNA are maintained after fertilization, suggesting a direct link between sperm and embryo RNA.
Collapse
Affiliation(s)
- K Gapp
- Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QN, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - G van Steenwyk
- Laboratory of Neuroepigenetics, University of Zürich and Swiss Federal Institute of Technology, Brain Research Institute, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - P L Germain
- Laboratory of Neuroepigenetics, University of Zürich and Swiss Federal Institute of Technology, Brain Research Institute, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - W Matsushima
- Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QN, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - K L M Rudolph
- Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QN, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - F Manuella
- Laboratory of Neuroepigenetics, University of Zürich and Swiss Federal Institute of Technology, Brain Research Institute, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - M Roszkowski
- Laboratory of Neuroepigenetics, University of Zürich and Swiss Federal Institute of Technology, Brain Research Institute, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - G Vernaz
- Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QN, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - T Ghosh
- Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QN, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - P Pelczar
- Center for Transgenic Models, University of Basel, Mattenstrasse 22, CH-4002, Basel, Switzerland
| | - I M Mansuy
- Laboratory of Neuroepigenetics, University of Zürich and Swiss Federal Institute of Technology, Brain Research Institute, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| | - E A Miska
- Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QN, UK.
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK.
| |
Collapse
|
29
|
Zhang Y, Fu T, Han S, Ding Y, Wang J, Zheng J, Li J. Monocular Deprivation Affects Visual Cortex Plasticity Through cPKCγ-Modulated GluR1 Phosphorylation in Mice. Invest Ophthalmol Vis Sci 2020; 61:44. [PMID: 32343785 PMCID: PMC7401946 DOI: 10.1167/iovs.61.4.44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose To determine how visual cortex plasticity changes after monocular deprivation (MD) in mice and whether conventional protein kinase C gamma (cPKCγ) plays a role in visual cortex plasticity. Methods cPKCγ membrane translocation levels were quantified by using immunoblotting to explore the effects of MD on cPKCγ activation. Electrophysiology was used to record field excitatory postsynaptic potential (fEPSP) amplitude with the goal of observing changes in visual cortex plasticity after MD. Immunoblotting was also used to determine the phosphorylation levels of GluR1 at Ser831. Light transmission was analyzed using electroretinography to examine the effects of MD and cPKCγ on mouse retinal function. Results Membrane translocation levels of cPKCγ significantly increased in the contralateral visual cortex of MD mice compared to wild-type (WT) mice (P < 0.001). In the contralateral visual cortex, long-term potentiation (LTP) and the phosphorylation levels of GluR1 at Ser 831 were increased in cPKCγ+/+ mice after MD. Interestingly, these levels could be downregulated by cPKCγ knockout compared to cPKCγ+/++MD mice (P < 0.001). Compared to the right eyes of WT mice, the amplitudes of a-waves and b-waves declined in deprived right eyes of mice after MD (P < 0.001). There were no significant differences when comparing cPKCγ+/+ and cPKCγ−/− mice with MD. Conclusions cPKCγ participates in the plasticity of the visual cortex after MD, which is characterized by increased LTP in the contralateral visual cortex, which may be a result of cPKCγ-mediated phosphorylation of GluR1 at Ser 831.
Collapse
|
30
|
Galan C, Krykbaeva M, Rando OJ. Early life lessons: The lasting effects of germline epigenetic information on organismal development. Mol Metab 2020; 38:100924. [PMID: 31974037 PMCID: PMC7300385 DOI: 10.1016/j.molmet.2019.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND An organism's metabolic phenotype is primarily affected by its genotype, its lifestyle, and the nutritional composition of its food supply. In addition, it is now clear from studies in many different species that ancestral environments can also modulate metabolism in at least one to two generations of offspring. SCOPE OF REVIEW We limit ourselves here to paternal effects in mammals, primarily focusing on studies performed in inbred rodent models. Although hundreds of studies link paternal diets and offspring metabolism, the mechanistic basis by which epigenetic information in sperm programs nutrient handling in the next generation remains mysterious. Our goal in this review is to provide a brief overview of paternal effect paradigms and the germline epigenome. We then pivot to exploring one key mystery in this literature: how do epigenetic changes in sperm, most of which are likely to act transiently in the early embryo, ultimately direct a long-lasting physiological response in offspring? MAJOR CONCLUSIONS Several potential mechanisms exist by which transient epigenetic modifications, such as small RNAs or methylation states erased shortly after fertilization, could be transferred to more durable heritable information. A detailed mechanistic understanding of this process will provide deep insights into early development, and could be of great relevance for human health and disease.
Collapse
Affiliation(s)
- Carolina Galan
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Marina Krykbaeva
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
31
|
Long-Term Impact of Early-Life Stress on Hippocampal Plasticity: Spotlight on Astrocytes. Int J Mol Sci 2020; 21:ijms21144999. [PMID: 32679826 PMCID: PMC7404101 DOI: 10.3390/ijms21144999] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
Adverse experiences during childhood are among the most prominent risk factors for developing mood and anxiety disorders later in life. Early-life stress interventions have been established as suitable models to study the neurobiological basis of childhood adversity in rodents. Different models such as maternal separation, impaired maternal care and juvenile stress during the postweaning/prepubertal life phase are utilized. Especially within the limbic system, they induce lasting alterations in neuronal circuits, neurotransmitter systems, neuronal architecture and plasticity that are further associated with emotional and cognitive information processing. Recent studies found that astrocytes, a special group of glial cells, have altered functions following early-life stress as well. As part of the tripartite synapse, astrocytes interact with neurons in multiple ways by affecting neurotransmitter uptake and metabolism, by providing gliotransmitters and by providing energy to neurons within local circuits. Thus, astrocytes comprise powerful modulators of neuronal plasticity and are well suited to mediate the long-term effects of early-life stress on neuronal circuits. In this review, we will summarize current findings on altered astrocyte function and hippocampal plasticity following early-life stress. Highlighting studies for astrocyte-related plasticity modulation as well as open questions, we will elucidate the potential of astrocytes as new targets for interventions against stress-induced neuropsychiatric disorders.
Collapse
|
32
|
Bound Together: How Psychoanalysis Diminishes Inter-generational DNA Trauma. Am J Psychoanal 2020; 80:196-218. [PMID: 32488025 DOI: 10.1057/s11231-020-09247-x] [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: 11/21/2022]
Abstract
The concept of intergenerational transmission of trauma plays a fundamental role in psychoanalysis. While it is known that intergenerational trauma can be transmitted through attachment relationships, a new branch of genetics (epigenetics) has emerged to study the interaction between human behavior and changes in DNA expression. Therefore, psychoanalysis, which has proven to reduce the intergenerational transmission of trauma from a behavioral perspective, can play a positive role in regulating DNA changes caused by environmental stress. The present paper focuses on recent research suggesting a direct correlation between psychological trauma and DNA modifications. In particular, DNA changes caused by psychological trauma can be transmitted from generation to generation, validating the psychoanalytic concept of intergenerational transmission of trauma. This evidence not only supports the essential role psychoanalysis has in influencing human behavior, but also suggests that it affects not only the individuals who undergo it but their offspring, as well, via the epigenetic passage of DNA.
Collapse
|
33
|
Paternal restraint stress affects offspring metabolism via ATF-2 dependent mechanisms in Drosophila melanogaster germ cells. Commun Biol 2020; 3:208. [PMID: 32367035 PMCID: PMC7198565 DOI: 10.1038/s42003-020-0935-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/07/2020] [Indexed: 12/22/2022] Open
Abstract
Paternal environmental factors can epigenetically influence gene expressions in offspring. We demonstrate that restraint stress, an experimental model for strong psychological stress, to fathers affects the epigenome, transcriptome, and metabolome of offspring in a MEKK1-dATF2 pathway-dependent manner in Drosophila melanogaster. Genes involved in amino acid metabolism are upregulated by paternal restraint stress, while genes involved in glycolysis and the tricarboxylic acid (TCA) cycle are downregulated. The effects of paternal restraint stress are also confirmed by metabolome analysis. dATF-2 is highly expressed in testicular germ cells, and restraint stress also induces p38 activation in the testes. Restraint stress induces Unpaired 3 (Upd3), a Drosophila homolog of Interleukin 6 (IL-6). Moreover, paternal overexpression of upd3 in somatic cells disrupts heterochromatin in offspring but not in offspring from dATF-2 mutant fathers. These results indicate that paternal restraint stress affects metabolism in offspring via inheritance of dATF-2-dependent epigenetic changes. Ki-Hyeon Seong et al. report that paternal environmental stress affects the metabolism of their offspring in Drosophila melanogaster. They exposed male flies to stress by preventing them from moving for 10 hours at a time and then measured gene expression and metabolite levels in their offspring, who showed increased expression of amino acid and one-carbon metabolism-related genes and downregulation of glycolysis and the TCA cycle.
Collapse
|
34
|
Tyebji S, Hannan AJ, Tonkin CJ. Pathogenic Infection in Male Mice Changes Sperm Small RNA Profiles and Transgenerationally Alters Offspring Behavior. Cell Rep 2020; 31:107573. [DOI: 10.1016/j.celrep.2020.107573] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/01/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022] Open
|
35
|
Baxter FA, Drake AJ. Non-genetic inheritance via the male germline in mammals. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180118. [PMID: 30966887 PMCID: PMC6460076 DOI: 10.1098/rstb.2018.0118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Numerous studies in humans and in animal models have demonstrated that exposure to adverse environmental conditions in early life results in long-term structural and functional changes in an organism, increasing the risk of cardiometabolic, neurobehavioural and reproductive disorders in later life. Such effects are not limited to the first generation offspring but may be transmitted to a second or a number of subsequent generations, through non-genomic mechanisms. While the transmission of ‘programmed’ effects through the maternal line could occur as a consequence of multiple influences, for example, altered maternal physiology, the inheritance of effects through the male line is more difficult to explain and there is much interest in a potential role for transgenerational epigenetic inheritance. In this review, we will discuss the mechanisms by which induced effects may be transmitted through the paternal lineage, with a particular focus on the role of epigenetic inheritance. This article is part of the theme issue ‘Developing differences: early-life effects and evolutionary medicine’.
Collapse
Affiliation(s)
- Faye A Baxter
- 1 Royal Hospital for Sick Children , 9 Sciennes Road, Edinburgh EH9 1LF , UK
| | - Amanda J Drake
- 1 Royal Hospital for Sick Children , 9 Sciennes Road, Edinburgh EH9 1LF , UK.,2 University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh , 47 Little France Crescent, Edinburgh EH16 4TJ , UK
| |
Collapse
|
36
|
Aoued HS, Sannigrahi S, Hunter SC, Doshi N, Sathi ZS, Chan AWS, Walum H, Dias BG. Proximate causes and consequences of intergenerational influences of salient sensory experience. GENES BRAIN AND BEHAVIOR 2020; 19:e12638. [PMID: 31943801 DOI: 10.1111/gbb.12638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/06/2020] [Accepted: 01/06/2020] [Indexed: 12/20/2022]
Abstract
Salient sensory environments experienced by a parental generation can exert intergenerational influences on offspring. While these data provide an exciting new perspective on biological inheritance, questions remain about causes and consequences of intergenerational influences of salient sensory experience. We previously showed that exposing male mice to a salient olfactory experience, like olfactory fear conditioning, resulted in offspring demonstrating a sensitivity to the odor used to condition the paternal generation and possessing enhanced neuroanatomical representation for that odor. In this study, we first injected RNA extracted from sperm of male mice that underwent olfactory fear conditioning into naïve single-cell zygotes and found that adults that developed from these embryos had increased sensitivity and enhanced neuroanatomical representation for the odor (Odor A) with which the paternal male had been conditioned. Next, we found that female, but not male offspring sired by males conditioned with Odor A show enhanced consolidation of a weak single-trial Odor A + shock fear conditioning protocol. Our data provide evidence that RNA found in the paternal germline after exposure to salient sensory experiences can contribute to intergenerational influences of such experiences, and that such intergenerational influences confer an element of adaptation to the offspring. In so doing, our study of intergenerational influences of parental sensory experience adds to existing literature on intergenerational influences of parental exposures to stress and dietary manipulations and suggests that some causes (sperm RNA) and consequences (behavioral flexibility) of intergenerational influences of parental experiences may be conserved across a variety of parental experiences.
Collapse
Affiliation(s)
- Hadj S Aoued
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia
| | - Soma Sannigrahi
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia
| | - Sarah C Hunter
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia
| | - Nandini Doshi
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia
| | - Zakia S Sathi
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia
| | - Anthony W S Chan
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia.,Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | - Hasse Walum
- Division of Autism and Related Disabilities, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Silvio O. Conte Center for Oxytocin and Social Cognition, Center for Translational Social Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Brian G Dias
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
37
|
Sperm RNA: Quo vadis? Semin Cell Dev Biol 2020; 97:123-130. [DOI: 10.1016/j.semcdb.2019.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 06/26/2019] [Accepted: 07/08/2019] [Indexed: 12/27/2022]
|
38
|
Champagne FA. Interplay between paternal germline and maternal effects in shaping development: The overlooked importance of behavioural ecology. Funct Ecol 2019. [DOI: 10.1111/1365-2435.13411] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
39
|
Hippocampal Protein Kinase C Gamma Signaling Mediates the Impairment of Spatial Learning and Memory in Prenatally Stressed Offspring Rats. Neuroscience 2019; 414:186-199. [DOI: 10.1016/j.neuroscience.2019.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/06/2019] [Accepted: 06/23/2019] [Indexed: 12/13/2022]
|
40
|
Pollak DD, Weber-Stadlbauer U. Transgenerational consequences of maternal immune activation. Semin Cell Dev Biol 2019; 97:181-188. [PMID: 31233834 DOI: 10.1016/j.semcdb.2019.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/12/2019] [Accepted: 06/20/2019] [Indexed: 01/10/2023]
Abstract
Prenatal exposure to infectious or inflammatory insults is increasingly recognized in the etiology of neuropsychiatric diseases, including schizophrenia, autism, depression and bipolar disorder. New discoveries highlight that maternal immune activation can lead to pathological effects on brain and behavior in multiple generations. This review describes the transgenerational consequences of maternal immune activation in shaping brain and behavior anomalies and disease risk across generations. We discuss potential underlying mechanisms of transmission, by which prenatal immune activation can mediate generation-spanning changes in brain development and functions and how external influences could further determine the specificity of the phenotype across generations. The identification of the underlying mechanisms appears relevant to infection-related neuropsychiatric illnesses independently of existing diagnostic classifications and may help identifying complex patterns of generation-spanning transmission beyond genetic inheritance. The herein described principles emphasize the importance of considering ancestral infectious histories in clinical research aiming at developing new preventive treatment strategies against infection-related neurodevelopmental disorders and mental illnesses.
Collapse
Affiliation(s)
- Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.
| |
Collapse
|
41
|
Sun W, von Meyenn F, Peleg‐Raibstein D, Wolfrum C. Environmental and Nutritional Effects Regulating Adipose Tissue Function and Metabolism Across Generations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900275. [PMID: 31179229 PMCID: PMC6548959 DOI: 10.1002/advs.201900275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/21/2019] [Indexed: 05/12/2023]
Abstract
The unabated rise in obesity prevalence during the last 40 years has spurred substantial interest in understanding the reasons for this epidemic. Studies in mice and humans have demonstrated that obesity is a highly heritable disease; however genetic variations within specific populations have so far not been able to explain this phenomenon to its full extent. Recent work has demonstrated that environmental cues can be sensed by an organism to elicit lasting changes, which in turn can affect systemic energy metabolism by different epigenetic mechanisms such as changes in small noncoding RNA expression, DNA methylation patterns, as well as histone modifications. These changes can directly modulate cellular function in response to environmental cues, however research during the last decade has demonstrated that some of these modifications might be transmitted to subsequent generations, thus modulating energy metabolism of the progeny in an inter- as well as transgenerational manner. In this context, adipose tissue has become a focus of research due to its plasticity, which allows the formation of energy storing (white) as well as energy wasting (brown/brite/beige) cells within the same depot. In this Review, the effects of environmental induced obesity with a particular focus on adipose tissue are discussed.
Collapse
Affiliation(s)
- Wenfei Sun
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| | - Ferdinand von Meyenn
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| | - Daria Peleg‐Raibstein
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| | - Christian Wolfrum
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| |
Collapse
|
42
|
Yeshurun S, Hannan AJ. Transgenerational epigenetic influences of paternal environmental exposures on brain function and predisposition to psychiatric disorders. Mol Psychiatry 2019. [PMID: 29520039 DOI: 10.1038/s41380-018-0039-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, striking new evidence has demonstrated non-genetic inheritance of acquired traits associated with parental environmental exposures. In particular, this transgenerational modulation of phenotypic traits is of direct relevance to psychiatric disorders, including depression, post-traumatic stress disorder, and other anxiety disorders. Here we review the recent progress in this field, with an emphasis on acquired traits of psychiatric illnesses transmitted epigenetically via the male lineage. We discuss the transgenerational effects of paternal exposure to stress vs. positive stimuli, such as exercise, and discuss their impact on the behavioral, affective and cognitive characteristics of their progeny. Furthermore, we review the recent evidence suggesting that these transgenerational effects are mediated by epigenetic mechanisms, including changes in DNA methylation and small non-coding RNAs in the sperm. We discuss the urgent need for more research exploring transgenerational epigenetic effects in animal models and human populations. These future studies may identify epigenetic mechanisms as potential contributors to the 'missing heritability' observed in genome-wide association studies of psychiatric illnesses and other human disorders. This exciting new field of transgenerational epigenomics will facilitate the development of novel strategies to predict, prevent and treat negative epigenetic consequences on offspring health, and psychiatric disorders in particular.
Collapse
Affiliation(s)
- Shlomo Yeshurun
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, 3010, Australia. .,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
43
|
|
44
|
Floriou-Servou A, von Ziegler L, Stalder L, Sturman O, Privitera M, Rassi A, Cremonesi A, Thöny B, Bohacek J. Distinct Proteomic, Transcriptomic, and Epigenetic Stress Responses in Dorsal and Ventral Hippocampus. Biol Psychiatry 2018; 84:531-541. [PMID: 29605177 DOI: 10.1016/j.biopsych.2018.02.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/24/2018] [Accepted: 02/05/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Acutely stressful experiences can trigger neuropsychiatric disorders and impair cognitive processes by altering hippocampal function. Although the intrinsic organization of the hippocampus is highly conserved throughout its long dorsal-ventral axis, the dorsal (anterior) hippocampus mediates spatial navigation and memory formation, whereas the ventral (posterior) hippocampus is involved in emotion regulation. To understand the molecular consequences of stress, detailed genome-wide screens are necessary and need to distinguish between dorsal and ventral hippocampal regions. While transcriptomic screens have become a mainstay in basic and clinical research, proteomic methods are rapidly evolving and hold even greater promise to reveal biologically and clinically relevant biomarkers. METHODS Here, we provide the first combined transcriptomic (RNA sequencing) and proteomic (sequential window acquisition of all theoretical mass spectra [SWATH-MS]) profiling of dorsal and ventral hippocampus in mice. We used three different acute stressors (novelty, swim, and restraint) to assess the impact of stress on both regions. RESULTS We demonstrated that both hippocampal regions display radically distinct molecular responses and that the ventral hippocampus is particularly sensitive to the effects of stress. Separately analyzing these structures greatly increased the sensitivity to detect stress-induced changes. For example, protein interaction cluster analyses revealed a stress-responsive epigenetic network around histone demethylase Kdm6b restricted to the ventral hippocampus, and acute stress reduced methylation of its enzymatic target H3K27me3. Selective Kdm6b knockdown in the ventral hippocampus led to behavioral hyperactivity/hyperresponsiveness. CONCLUSIONS These findings underscore the importance of considering dorsal and ventral hippocampus separately when conducting high-throughput molecular analyses, which has important implications for fundamental research as well as clinical studies.
Collapse
Affiliation(s)
- Amalia Floriou-Servou
- Laboratory of Molecular and Behavioral Neuroscience, Department of Health Science and Technology of ETH Zurich, Institute for Neuroscience, Neuroscience Center Zurich, Zurich, Switzerland
| | - Lukas von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Department of Health Science and Technology of ETH Zurich, Institute for Neuroscience, Neuroscience Center Zurich, Zurich, Switzerland; Laboratory of Neuroepigenetics, Medical Faculty of the University Zürich and Department of Health Science and Technology of the ETH Zürich, Zurich, Switzerland
| | - Luzia Stalder
- Laboratory of Neuroepigenetics, Medical Faculty of the University Zürich and Department of Health Science and Technology of the ETH Zürich, Zurich, Switzerland
| | - Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Department of Health Science and Technology of ETH Zurich, Institute for Neuroscience, Neuroscience Center Zurich, Zurich, Switzerland
| | - Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Department of Health Science and Technology of ETH Zurich, Institute for Neuroscience, Neuroscience Center Zurich, Zurich, Switzerland
| | - Anahita Rassi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Beat Thöny
- Division of Metabolism, University Children's Hospital Zurich, Zurich, Switzerland
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Department of Health Science and Technology of ETH Zurich, Institute for Neuroscience, Neuroscience Center Zurich, Zurich, Switzerland.
| |
Collapse
|
45
|
Stressing the Seminal Role of Paternal Experience in Transgenerational ‘Epigenopathy’ Affecting Offspring Health and Disease Susceptibility. Neuroscience 2018; 388:472-473. [DOI: 10.1016/j.neuroscience.2018.07.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 07/08/2018] [Indexed: 01/06/2023]
|
46
|
Ju LS, Yang JJ, Morey TE, Gravenstein N, Seubert CN, Resnick JL, Zhang JQ, Martynyuk AE. Role of epigenetic mechanisms in transmitting the effects of neonatal sevoflurane exposure to the next generation of male, but not female, rats. Br J Anaesth 2018; 121:406-416. [PMID: 30032879 PMCID: PMC6200111 DOI: 10.1016/j.bja.2018.04.034] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/03/2018] [Accepted: 05/02/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Clinical studies report learning disabilities and attention-deficit/hyperactivity disorders in those exposed to general anaesthesia early in life. Rats, primarily males, exposed to GABAergic anaesthetics as neonates exhibit behavioural abnormalities, exacerbated responses to stress, and reduced expression of hypothalamic K+-2Cl- Cl- exporter (Kcc2). The latter is implicated in development of psychiatric disorders, including male predominant autism spectrum disorders. We tested whether parental early life exposure to sevoflurane, the most frequently used anaesthetic in paediatrics, affects the next generation of unexposed rats. METHODS Offspring (F1) of unexposed or exposed to sevoflurane on postnatal day 5 Sprague-Dawley rats (F0) were subjected to behavioural and brain gene expression evaluations. RESULTS Male, but not female, progeny of sevoflurane-exposed parents exhibited abnormalities in behavioural testing and Kcc2 expression. Male F1 rats of both exposed parents exhibited impaired spatial memory and expression of hippocampal and hypothalamic Kcc2. Offspring of only exposed sires had abnormalities in elevated plus maze and prepulse inhibition of startle, but normal spatial memory and impaired expression of hypothalamic, but not hippocampal, Kcc2. In contrast to exposed F0, their progeny exhibited normal corticosterone responses to stress. Bisulphite sequencing revealed increased CpG site methylation in the Kcc2 promoter in F0 sperm and F1 male hippocampus and hypothalamus that was in concordance with the changes in Kcc2 expression in specific F1 groups. CONCLUSIONS Neonatal exposure to sevoflurane can affect the next generation of males through epigenetic modification of Kcc2 expression, while F1 females are at diminished risk.
Collapse
Affiliation(s)
- L-S Ju
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - J-J Yang
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - T E Morey
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - N Gravenstein
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA; The McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA
| | - C N Seubert
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - J L Resnick
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - J-Q Zhang
- Department of Anesthesiology, Zhengzhou University, Zhengzhou, China
| | - A E Martynyuk
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA; The McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
47
|
Gilardi F, Augsburger M, Thomas A. Will Widespread Synthetic Opioid Consumption Induce Epigenetic Consequences in Future Generations? Front Pharmacol 2018; 9:702. [PMID: 30018553 PMCID: PMC6037745 DOI: 10.3389/fphar.2018.00702] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/11/2018] [Indexed: 11/13/2022] Open
Abstract
A growing number of evidence demonstrates that ancestral exposure to xenobiotics (pollutants, drugs of abuse, etc.) can perturb the physiology and behavior of descendants. Both maternal and paternal transmission of phenotype across generations has been proved, demonstrating that parental drug history may have significant implications for subsequent generations. In the last years, the burden of novel synthetic opioid (NSO) consumption, due to increased medical prescription of pain medications and to easier accessibility of these substances on illegal market, is raising new questions first in term of public health, but also about the consequences of the parental use of these drugs on future generations. Besides being associated to the neonatal abstinence syndrome, in utero exposure to opioids has an impact on neuronal development with long-term repercussions that are potentially transmitted to subsequent generations. In addition, recent reports suggest that opioid use even before conception influences the reactivity to opioids of the progeny and the following generations, likely through epigenetic mechanisms. This review describes the current knowledge about the transgenerational effects of opioid consumption. We summarize the preclinical and clinical findings showing the implications for the subsequent generations of parental exposure to opioids earlier in life. Limitations of the existing data on NSOs and new perspectives of the research are also discussed, as well as clinical and forensic consequences.
Collapse
Affiliation(s)
- Federica Gilardi
- Forensic Toxicology and Chemistry Unit, University Center of Legal Medicine, Lausanne University Hospital - Geneva University Hospitals, Geneva, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Marc Augsburger
- Forensic Toxicology and Chemistry Unit, University Center of Legal Medicine, Lausanne University Hospital - Geneva University Hospitals, Geneva, Switzerland
| | - Aurelien Thomas
- Forensic Toxicology and Chemistry Unit, University Center of Legal Medicine, Lausanne University Hospital - Geneva University Hospitals, Geneva, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
48
|
Harker A, Carroll C, Raza S, Kolb B, Gibb R. Preconception Paternal Stress in Rats Alters Brain and Behavior in Offspring. Neuroscience 2018; 388:474-485. [PMID: 29964157 DOI: 10.1016/j.neuroscience.2018.06.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022]
Abstract
Whereas environmental challenges during gestation have been repeatedly shown to alter offspring brain architecture and behavior, exploration examining the consequences of paternal preconception experience on offspring outcome is limited. The goal of this study was to examine the effects of preconception paternal stress (PPS) on cerebral plasticity and behavior in the offspring. Several behavioral assays were performed on offspring between postnatal days 33 (P33) and 101 (P101). Following behavioral testing, the brains were harvested and dendritic morphology (dendritic complexity, length, and spine density) were examined on cortical pyramidal cells in medial prefrontal cortex (mPFC), orbital frontal cortex (OFC), parietal cortex (Par1), and the CA1 area of the hippocampus. As anticipated, behavior was altered on both the activity box assay and elevated plus maze and performance was impaired in the Whishaw tray reaching task. Neuroanatomical measures revealed a heavier brain in stressed animals and dendritic changes in all regions measured, the precise effect varying with the measure and cerebral region. Thus, PPS impacted both behavior and neuronal morphology of offspring. These effects likely have an epigenetic basis given that in a parallel study of littermates of the current animals we found extensive epigenetic changes at P21.
Collapse
Affiliation(s)
- Allonna Harker
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada.
| | - Cathy Carroll
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada.
| | - Sarah Raza
- Canadian Institute for Advanced Research, Toronto, ON, Canada.
| | - Bryan Kolb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada; Canadian Institute for Advanced Research, Toronto, ON, Canada.
| | - Robbin Gibb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada.
| |
Collapse
|
49
|
Jawaid A, Roszkowski M, Mansuy IM. Transgenerational Epigenetics of Traumatic Stress. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 158:273-298. [PMID: 30072057 DOI: 10.1016/bs.pmbts.2018.03.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Traumatic stress is a type of environmental experience that can modify behavior, cognition and physiological functions such as metabolism, in mammals. Many of the effects of traumatic stress can be transmitted to subsequent generations even when individuals from these generations are not exposed to any traumatic stressor. This book chapter discusses the concept of epigenetic/non-genomic inheritance of such traits involving the germline in mammals. It includes a comprehensive review of animal and human studies on inter- and transgenerational inheritance of the effects of traumatic stress, some of the epigenetic changes in the germline currently known to be associated with traumatic stress, and possible mechanisms for their induction and maintenance during development and adulthood. We also describe some experimental interventions that attempted to prevent the transmission of these effects, and consider the evolutionary importance of transgenerational inheritance and future outlook of the field.
Collapse
Affiliation(s)
- Ali Jawaid
- Laboratory of Neuroepigenetics, Medical Faculty of the University of Zurich and Department of Health Science and Technology of the Swiss Federal Institute of Technology, Neuroscience Center Zurich, Zurich, Switzerland
| | - Martin Roszkowski
- Laboratory of Neuroepigenetics, Medical Faculty of the University of Zurich and Department of Health Science and Technology of the Swiss Federal Institute of Technology, Neuroscience Center Zurich, Zurich, Switzerland
| | - Isabelle M Mansuy
- Laboratory of Neuroepigenetics, Medical Faculty of the University of Zurich and Department of Health Science and Technology of the Swiss Federal Institute of Technology, Neuroscience Center Zurich, Zurich, Switzerland.
| |
Collapse
|
50
|
Selectively Impaired Endocannabinoid-Dependent Long-Term Depression in the Lateral Habenula in an Animal Model of Depression. Cell Rep 2018; 20:289-296. [PMID: 28700932 DOI: 10.1016/j.celrep.2017.06.049] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/22/2017] [Accepted: 06/19/2017] [Indexed: 12/18/2022] Open
Abstract
Abnormal potentiation in the lateral habenula (LHb) has been suggested to mediate depression-like behaviors. However, the underlying mechanisms of the synaptic efficacy regulation of LHb synapses and the potential for their modulation are only poorly understood. Here, we report that long-term synaptic depression (LTD) occurs in the LHb upon both low-frequency stimulation (LFS) and moderate-frequency stimulation (MFS). LFS-induced LTD (LFS-LTD) is accompanied by a reduction in presynaptic release probability, which is endocannabinoid (eCB) signaling dependent. Surprisingly, exposure to an acute stressor completely masks the induction of LFS-LTD in the LHb while leaving the MFS-induced LTD intact. Pharmacological activation of cannabinoid receptor 1 (CB1R) or blockade of αCaMKII successfully restored LTD in the LHb in an animal model of depression. Thus, our findings reveal a form of synaptic strength regulation and a stress-induced shift of synaptic plasticity in the LHb.
Collapse
|