1
|
He Y, Zhang Z, Zheng Q, Su H, Liu M, Chen X, Qi L, Zhang Y, Xu Z, Tang J. Paternal alcohol exposure affected offspring mesenteric artery via ROS-Cacna1c and DNA hypomethylation. J Hypertens 2025; 43:631-641. [PMID: 39791443 DOI: 10.1097/hjh.0000000000003947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Paternal preconception alcohol exposure affects fetal development; however, it is largely unknown about the influences on offspring vasculature and mechanisms. METHODS Offspring born form paternal rats treated with alcohol or water before pregnant was raised until 3 months of age. Vessel tone of mesenteric arteries was detected using myograph system; whole-cell calcium channel current in smooth muscle cells was tested using patch-clamp; molecule expressions were detected with real-time PCR, western blotting, and Dihydroethidium (DHE); DNA methylations were determined using targeted bisulfate sequencing assay. Following 5-aza-2'-deoxycytidine incubation, vessel tone in offspring mesenteric artery and Cacna1c expression in A7r5 was tested. RESULTS When comparing with the control, stress-strain curve was left-shifted in alcohol. There was lower incremental distensibility and endothelium-dependent dilation associated with endothelial nitric oxide synthase. Agonists-induced constrictions were greater in alcohol offspring than that in control, associated with higher expression of AT1R, Cacna1c, and reactive oxygen species (ROS). Baseline and Ang II-stimulated calcium channel currents were higher in alcohol group. Tempol and apocynin could restore Ang II-increased constriction and calcium channel current in alcohol offspring. When comparing with the control, there was lower DNA methylation of Cacna1c promotor in alcohol offspring mesenteric artery and in paternal sperm. 5-aza-2'-deoxycytidine increased contraction in control offspring mesenteric artery and Cacna1c expression in A7r5. CONCLUSION Paternal preconception alcohol exposure-affected offspring mesenteric artery was via ROS-Cacna1c. Abnormal offspring vascular functions might be inherited via DNA hypomethylation of Cacna1c promotor from paternal sperm exposed to alcohol. These data gained provided important clues for cardiovascular disorders at germ cell origin.
Collapse
Affiliation(s)
- Yun He
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
- Taixing People's Hospital
| | - Ze Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
- Taixing People's Hospital
| | - Qiutong Zheng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
| | - Hongyu Su
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
| | - Mingxing Liu
- Infection Management Department, The First Affiliated Hospital of Soochow University, Jiangsu
| | - Xueyi Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
| | - Linglu Qi
- Women's Hospital School of Medicine Zhejiang University, Zhejiang
| | - Yumeng Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
- Wuxi Maternity and Child Health Care Hospital, Jiangsu, China
| | - Jiaqi Tang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Jiangsu
| |
Collapse
|
2
|
Hellmann JK, Rogers MM. The transgenerational consequences of paternal social isolation and predation exposure in threespined sticklebacks. J Anim Ecol 2024; 93:1328-1337. [PMID: 39049666 DOI: 10.1111/1365-2656.14151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/22/2024] [Indexed: 07/27/2024]
Abstract
Parents routinely encounter stress in the ecological environment that can affect offspring development (transgenerational plasticity: TGP); however, parents' interactions with conspecifics may alter how parents respond to ecological stressors. During social buffering, the presence of conspecifics can reduce the response to or increase the speed of recovery from a stressor. This may have cascading effects on offspring if conspecifics can mitigate parental responses to ecological stress in ways that blunt the transmission of stress-induced transgenerational effects. Here, we simultaneously manipulated both paternal social isolation and experience with predation risk prior to fertilisation in threespined stickleback (Gasterosteus aculeatus). We generated offspring via in-vitro fertilisation to allow us to isolate paternal effects mediated via sperm alone (i.e. in the absence of paternal care). If social buffering mitigates TGP induced by paternal exposure to predation risk, then we expect the transgenerational effects of predation exposure to be weaker when a conspecific is present compared to when the father is isolated. Offspring of predator-exposed fathers showed reduced anxiety-like behaviour and tended to be captured faster by the predator. Fathers who were socially isolated also had offspring that were captured faster by a live predator, suggesting that paternal social isolation may have maladaptive effects on how offspring respond to ecological stressors. Despite additive effects of paternal social isolation and paternal predation risk, we found no evidence of an interaction between these paternal treatments, suggesting that the presence of a conspecific did not buffer fathers and/or offspring from the effects of predation risk. Our results suggest that socially induced stress is an important, yet underappreciated, mediator of TGP and can elicit transgenerational effects even in species that do not form permanent social groups. Future studies should therefore consider how the parental social environment can affect both within and trans-generational responses to ecological stressors.
Collapse
Affiliation(s)
- Jennifer K Hellmann
- Department of Biology, University of Dayton, Dayton, Ohio, USA
- Department of Evolution, Ecology, and Organismal Biology, The Ohio State University, Columbus, Ohio, USA
| | - Michaela M Rogers
- Department of Biology, University of Dayton, Dayton, Ohio, USA
- Department of Evolution, Ecology, and Organismal Biology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
3
|
Basel A, Bhadsavle SS, Scaturro KZ, Parkey GK, Gaytan MN, Patel JJ, Thomas KN, Golding MC. Parental Alcohol Exposures Associate with Lasting Mitochondrial Dysfunction and Accelerated Aging in a Mouse Model. Aging Dis 2024:AD.2024.0722. [PMID: 39122451 DOI: 10.14336/ad.2024.0722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Although detrimental changes in mitochondrial morphology and function are widely described symptoms of fetal alcohol exposure, no studies have followed these mitochondrial deficits into adult life or determined if they predispose individuals with fetal alcohol spectrum disorders (FASDs) to accelerated biological aging. Here, we used a multiplex preclinical mouse model to compare markers of cellular senescence and age-related outcomes induced by maternal, paternal, and dual-parental alcohol exposures. We find that even in middle life (postnatal day 300), the adult offspring of alcohol-exposed parents exhibited significant increases in markers of stress-induced premature cellular senescence in the brain and liver, including an upregulation of cell cycle inhibitory proteins and increased senescence-associated β-galactosidase activity. Strikingly, in the male offspring, we observe an interaction between maternal and paternal alcohol use, with histological indicators of accelerated age-related liver disease in the dual-parental offspring exceeding those induced by either maternal or paternal alcohol use alone. Our studies indicate that chronic parental alcohol use causes enduring mitochondrial dysfunction in offspring, resulting in a reduced NAD+/NAHD ratio and altered expression of the NAD+-dependent deacetylases SIRT1 and SIRT3. These observations suggest that some aspects of FASDs may be linked to accelerated aging due to programmed changes in the regulation of mitochondrial function and cellular bioenergetics.
Collapse
|
4
|
Liao H, Lu D, Reisinger SN, Kleeman EA, van de Garde N, Gubert C, Hannan AJ. Mimicking bacterial infection in male mice changes sperm small RNA profiles and multigenerationally alters offspring behavior and physiology. Brain Behav Immun 2024; 119:520-538. [PMID: 38636562 DOI: 10.1016/j.bbi.2024.04.017] [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: 12/21/2023] [Revised: 03/22/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Paternal pre-conceptual exposures, including stress, diet, substance abuse, parasite infection, and viral immune activation via Poly I:C, have been reported to influence the brains and behavior of offspring through sperm epigenetic changes. However, the effects of paternal (F0) pre-conceptual exposure to bacterial-induced immune activation on the behavior and physiology of F1 and F2 generations remain unexplored. We examined this using C57BL/6J mice. Eight-week-old males (F0) received a single intraperitoneal injection of the bacterial mimetic lipopolysaccharide (LPS: 5 mg/kg) or 0.9 % saline (vehicle control) before mating with naïve females at four weeks post-injection. Comprehensive behavioral assessments were conducted to investigate anxiety, social behaviors, depressive-like behaviors and cognition in both the F1 and F2 generations within the age range of 8 to 14 weeks. Results demonstrated that only female offspring of LPS-exposed fathers exhibited reduced anxiety levels in the light/dark box, large open field, and novelty-suppressed feeding test. These F1 female offspring also exhibited heightened sociability in the 3-chambered social interaction test and a reduced preference for saccharin in the saccharin preference test. Additionally, the F1 male offspring of LPS-challenged males demonstrated an increased total distance traveled in the light/dark box and a longer distance covered in the light zone. They also exhibited diminished preference for social novelty in the 3-chambered social interaction test and an elevated novel arm preference index in the Y-maze. In the F2 generation, male descendants of LPS-treated fathers showed reduced latency to feed in the novelty-suppressed feeding test. Additionally, the F2 generation of LPS-challenged fathers, but not the F1 generation, displayed enhanced immune response in both sexes after an acute LPS immune challenge (5 mg/kg). Analysis of sperm small noncoding RNA profiles from LPS-treated F0 mice revealed significant changes at 4 weeks after administration of LPS. These changes included three microRNAs, eight PIWI-interacting RNAs, and two transfer RNAs, exhibiting significant upregulation (mmu-miR-146a-5p, mmu-piR-27082 and mmu-piR-29102) or downregulation (mmu-miR-5110, mmu-miR-467e-3p, mmu-piR-22583, mmu-piR-23548, mmu-piR-36341, mmu-piR-50293, mmu-piR-16583, mmu-piR-36507, Mus_musculus_tRNA-Ile-AAT-2-1 and Mus_musculus_tRNA-Tyr-GTA-1-1). Additionally, we detected 52 upregulated small noncoding RNAs (including 9 miRNAs, 41 piRNAs, and 2 tRNAs) and 7 downregulated small noncoding RNAs (3 miRNAs, 3 piRNAs, and 1 tRNA) in the sperm of F1 offspring from LPS-treated males. These findings provide compelling evidence for the involvement of epigenetic mechanisms in the modulation of brain function and immunity, and associated behavioral and immunological traits, across generations, in response to bacterial infection.
Collapse
Affiliation(s)
- Huan Liao
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Da Lu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Nicholas van de Garde
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
5
|
Goolsby BC, Smith EJ, Muratore IB, Coto ZN, Muscedere ML, Traniello JFA. Differential Neuroanatomical, Neurochemical, and Behavioral Impacts of Early-Age Isolation in a Eusocial Insect. BRAIN, BEHAVIOR AND EVOLUTION 2024; 99:171-183. [PMID: 38857586 DOI: 10.1159/000539546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 05/21/2024] [Indexed: 06/12/2024]
Abstract
INTRODUCTION Social experience early in life appears to be necessary for the development of species-typical behavior. Although isolation during critical periods of maturation has been shown to impact behavior by altering gene expression and brain development in invertebrates and vertebrates, workers of some ant species appear resilient to social deprivation and other neurobiological challenges that occur during senescence or due to loss of sensory input. It is unclear if and to what degree neuroanatomy, neurochemistry, and behavior will show deficiencies if social experience in the early adult life of worker ants is compromised. METHODS We reared newly eclosed adult workers of Camponotus floridanus under conditions of social isolation for 2-53 days, quantified brain compartment volumes, recorded biogenic amine levels in individual brains, and evaluated movement and behavioral performance to compare the neuroanatomy, neurochemistry, brood-care behavior, and foraging (predatory behavior) of isolated workers with that of workers experiencing natural social contact after adult eclosion. RESULTS We found that the volume of the antennal lobe, which processes olfactory inputs, was significantly reduced in workers isolated for an average of 40 days, whereas the size of the mushroom bodies, centers of higher-order sensory processing, increased after eclosion and was not significantly different from controls. Titers of the neuromodulators serotonin, dopamine, and octopamine remained stable and were not significantly different in isolation treatments and controls. Brood care, predation, and overall movement were reduced in workers lacking social contact early in life. CONCLUSION These results suggest that the behavioral development of isolated workers of C. floridanus is specifically impacted by a reduction in the size of the antennal lobe. Task performance and locomotor ability therefore appear to be sensitive to a loss of social contact through a reduction of olfactory processing ability rather than change in the size of the mushroom bodies, which serve important functions in learning and memory, or the central complex, which controls movement.
Collapse
Affiliation(s)
- Billie C Goolsby
- Department of Biology, Boston University, Boston, Massachusetts, USA
- Department of Biology, Stanford University, Stanford, California, USA
| | - E Jordan Smith
- Department of Biology, Boston University, Boston, Massachusetts, USA
| | - Isabella B Muratore
- Department of Biology, Boston University, Boston, Massachusetts, USA
- Chemistry Department, United States Naval Academy, Annapolis, Maryland, USA
| | - Zach N Coto
- Department of Biology, Boston University, Boston, Massachusetts, USA
| | - Mario L Muscedere
- Department of Biology, Boston University, Boston, Massachusetts, USA
| | | |
Collapse
|
6
|
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
|
7
|
Goolsby BC, Smith EJ, Muratore IB, Coto ZN, Muscedere ML, Traniello JFA. Differential Neuroanatomical, Neurochemical, and Behavioral Impacts of Early-Age Isolation in a Eusocial Insect. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.546928. [PMID: 37425857 PMCID: PMC10326991 DOI: 10.1101/2023.06.29.546928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Social experience early in life appears to be necessary for the development of species-typical behavior. Although isolation during critical periods of maturation has been shown to impact behavior by altering gene expression and brain development in invertebrates and vertebrates, workers of some ant species appear resilient to social deprivation and other neurobiological challenges that occur during senescence or due to loss of sensory input. It is unclear if and to what degree neuroanatomy, neurochemistry, and behavior will show deficiencies if social experience in the early adult life of worker ants is compromised. We reared newly-eclosed adult workers of Camponotus floridanus under conditions of social isolation for 2 to 53 days, quantified brain compartment volumes, recorded biogenic amine levels in individual brains, and evaluated movement and behavioral performance to compare the neuroanatomy, neurochemistry, brood-care behavior, and foraging (predatory behavior) of isolated workers with that of workers experiencing natural social contact after adult eclosion. We found that the volume of the antennal lobe, which processes olfactory inputs, was significantly reduced in workers isolated for an average of 40 days, whereas the size of the mushroom bodies, centers of higher-order sensory processing, increased after eclosion and was not significantly different from controls. Titers of the neuromodulators serotonin, dopamine, and octopamine remained stable and were not significantly different in isolation treatments and controls. Brood care, predation, and overall movement were reduced in workers lacking social contact early in life. These results suggest that the behavioral development of isolated workers of C. floridanus is specifically impacted by a reduction in the size of the antennal lobe. Task performance and locomotor ability therefore appear to be sensitive to a loss of social contact through a reduction of olfactory processing ability rather than change in the size of the mushroom bodies, which serve important functions in learning and memory, or the central complex, which controls movement.
Collapse
Affiliation(s)
- Billie C. Goolsby
- Department of Biology, Boston University, Boston, MA, 02215, USA
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - E. Jordan Smith
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | - Isabella B. Muratore
- Department of Biology, Boston University, Boston, MA, 02215, USA
- Department of Biological Sciences, New Jersey Institute of Technology, NJ, 07102, USA
| | - Zach N. Coto
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | | | | |
Collapse
|
8
|
Jang J, Kim HJ, Koh HY. Compensatory enhancement of paternal care in maternally neglected mice family. Anim Cells Syst (Seoul) 2023; 27:249-259. [PMID: 37818017 PMCID: PMC10561577 DOI: 10.1080/19768354.2023.2266006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023] Open
Abstract
Parental care strategies, ranging from biparental to uniparental, evolve based on factors affecting sexual conflict over care. Plasticity in how parents respond to reduction in each other's care effort is thus proposed to be important in the evolution of parental care behaviors. Models predict that 'obligate' biparental care is stable when a parent responds to reduced partner effort with 'partial' compensation, trading-off current and future reproduction. A meta-analysis of experimental studies on biparental birds also revealed partial compensation, supporting coevolution of parental care type and plasticity pattern. However, few studies have addressed this issue across different taxa and different parental care types. In laboratory mice, a female-biased 'facultative' biparental species, fathers paired with a competent mother rarely provide care. We show that, when mated with a pup-neglecting mutant mother, fathers increased care effort to 'fully' compensate for the lost maternal care in both pup survival rate and total care amount. Pup retrieval latency was significantly shorter, and neural activity in relevant brain regions twice as high, suggesting enhanced motivation. This study with mice not only opens a road to explore the neural correlates of paternal plasticity but will also help understand how behavioral plasticity contributes to adaptive evolution of parental care behaviors.
Collapse
Affiliation(s)
- Jaewon Jang
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Hea-jin Kim
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Hae-Young Koh
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Life Sciences, Korea University, Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| |
Collapse
|
9
|
Farmer AL, Lewis MH. Reduction of restricted repetitive behavior by environmental enrichment: Potential neurobiological mechanisms. Neurosci Biobehav Rev 2023; 152:105291. [PMID: 37353046 DOI: 10.1016/j.neubiorev.2023.105291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Restricted repetitive behaviors (RRB) are one of two diagnostic criteria for autism spectrum disorder and common in other neurodevelopmental and psychiatric disorders. The term restricted repetitive behavior refers to a wide variety of inflexible patterns of behavior including stereotypy, self-injury, restricted interests, insistence on sameness, and ritualistic and compulsive behavior. However, despite their prevalence in clinical populations, their underlying causes remain poorly understood hampering the development of effective treatments. Intriguingly, numerous animal studies have demonstrated that these behaviors are reduced by rearing in enriched environments (EE). Understanding the processes responsible for the attenuation of repetitive behaviors by EE should offer insights into potential therapeutic approaches, as well as shed light on the underlying neurobiology of repetitive behaviors. This review summarizes the current knowledge of the relationship between EE and RRB and discusses potential mechanisms for EE's attenuation of RRB based on the broader EE literature. Existing gaps in the literature and future directions are also discussed.
Collapse
Affiliation(s)
- Anna L Farmer
- Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Mark H Lewis
- Department of Psychology, University of Florida, Gainesville, FL, USA; Department of Psychiatry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Clark FE, Greggor AL, Montgomery SH, Plotnik JM. The endangered brain: actively preserving ex-situ animal behaviour and cognition will benefit in-situ conservation. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230707. [PMID: 37650055 PMCID: PMC10465207 DOI: 10.1098/rsos.230707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023]
Abstract
Endangered species have small, unsustainable population sizes that are geographically or genetically restricted. Ex-situ conservation programmes are therefore faced with the challenge of breeding sufficiently sized, genetically diverse populations earmarked for reintroduction that have the behavioural skills to survive and breed in the wild. Yet, maintaining historically beneficial behaviours may be insufficient, as research continues to suggest that certain cognitive-behavioural skills and flexibility are necessary to cope with human-induced rapid environmental change (HIREC). This paper begins by reviewing interdisciplinary studies on the 'captivity effect' in laboratory, farmed, domesticated and feral vertebrates and finds that captivity imposes rapid yet often reversible changes to the brain, cognition and behaviour. However, research on this effect in ex-situ conservation sites is lacking. This paper reveals an apparent mismatch between ex-situ enrichment aims and the cognitive-behavioural skills possessed by animals currently coping with HIREC. After synthesizing literature across neuroscience, behavioural biology, comparative cognition and field conservation, it seems that ex-situ endangered species deemed for reintroduction may have better chances of coping with HIREC if their natural cognition and behavioural repertoires are actively preserved. Evaluating the effects of environmental challenges rather than captivity per se is recommended, in addition to using targeted cognitive enrichment.
Collapse
Affiliation(s)
- Fay E. Clark
- School of Psychological Science, University of Bristol, Bristol, UK
| | | | | | - Joshua M. Plotnik
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA
- Department of Psychology, The Graduate Center, City University of New York, New York, NY, USA
| |
Collapse
|
11
|
Mashoodh R, Habrylo IB, Gudsnuk K, Champagne FA. Sex-specific effects of chronic paternal stress on offspring development are partially mediated via mothers. Horm Behav 2023; 152:105357. [PMID: 37062113 DOI: 10.1016/j.yhbeh.2023.105357] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/18/2023]
Abstract
Paternal stress exposure is known to impact the development of stress-related behaviors in offspring. Previous work has highlighted the importance of sperm mediated factors, such as RNAs, in transmitting the effects of parental stress. However, a key unanswered question is whether mothers behavior could drive or modulate the transmission of paternal stress effects on offspring development. Here we investigate how chronic variable stress in Balb/C mice influences the sex-specific development of anxiety- and depression-like neural and behavioral development in offspring. Moreover, we examined how stressed fathers influenced mate maternal investment towards their offspring and how this may modulate the transmission of paternal stress effects on offspring. We show that paternal stress leads to sex-specific effects on offspring behavior. Males that are chronically stressed sire female offspring that show increased anxiety and depression-like behaviors. However, male offspring of stressed fathers show reductions in anxiety- and depression-behaviors and are generally more exploratory. Moreover, we show that females mated with stressed males gain less weight during pregnancy and provide less care towards their offspring which additionally influenced offspring development. These data indicate that paternal stress can influence offspring development both directly and indirectly via changes in mothers, with implications for sex-specific offspring development.
Collapse
Affiliation(s)
- Rahia Mashoodh
- University of Cambridge, Department of Zoology, Downing Street, Cambridge CB2 3EJ, United Kingdom.
| | - Ireneusz B Habrylo
- Columbia University, Department of Psychology, 1190 Amsterdam Avenue, Schermerhorn Hall, New York, NY 10027, United States of America
| | - Kathryn Gudsnuk
- Columbia University, Department of Psychology, 1190 Amsterdam Avenue, Schermerhorn Hall, New York, NY 10027, United States of America
| | - Frances A Champagne
- Columbia University, Department of Psychology, 1190 Amsterdam Avenue, Schermerhorn Hall, New York, NY 10027, United States of America; University of Texas Austin, Department of Psychology, 108 Dean Keeton, Austin, TX 78712, United States of America
| |
Collapse
|
12
|
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: 2.0] [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
|
13
|
Patlar B. On the Role of Seminal Fluid Protein and Nucleic Acid Content in Paternal Epigenetic Inheritance. Int J Mol Sci 2022; 23:ijms232314533. [PMID: 36498858 PMCID: PMC9739459 DOI: 10.3390/ijms232314533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The evidence supports the occurrence of environmentally-induced paternal epigenetic inheritance that shapes the offspring phenotype in the absence of direct or indirect paternal care and clearly demonstrates that sperm epigenetics is one of the major actors mediating these paternal effects. However, in most animals, while sperm makes up only a small portion of the seminal fluid, males also have a complex mixture of proteins, peptides, different types of small noncoding RNAs, and cell-free DNA fragments in their ejaculate. These seminal fluid contents (Sfcs) are in close contact with the reproductive cells, tissues, organs, and other molecules of both males and females during reproduction. Moreover, their production and use are adjusted in response to environmental conditions, making them potential markers of environmentally- and developmentally-induced paternal effects on the next generation(s). Although there is some intriguing evidence for Sfc-mediated paternal effects, the underlying molecular mechanisms remain poorly defined. In this review, the current evidence regarding the links between seminal fluid and environmental paternal effects and the potential pathways and mechanisms that seminal fluid may follow in mediating paternal epigenetic inheritance are discussed.
Collapse
Affiliation(s)
- Bahar Patlar
- Animal Ecology, Department of Zoology, Martin-Luther University Halle-Wittenberg, 06099 Halle (Saale), Germany
| |
Collapse
|
14
|
Rearing in an Enriched Environment Ameliorates the ADHD-like Behaviors of Lister Hooded Rats While Suppressing Neuronal Activities in the Medial Prefrontal Cortex. Cells 2022; 11:cells11223649. [PMID: 36429076 PMCID: PMC9688563 DOI: 10.3390/cells11223649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
In addition to genetic factors, environmental factors play a role in the pathogenesis of attention deficit/hyperactivity disorder (ADHD). This study used Lister hooded rats (LHRs) as ADHD model animals to evaluate the effects of environmental factors. Male LHR pups were kept in four rearing conditions from postnatal day 23 (4 rats in a standard cage; 12 rats in a large flat cage; and 4 or 12 rats in an enriched environment [EE]) until 9 weeks of age. EE rearing but not rearing in a large flat cage decreased the activity of LHRs in the open field test that was conducted for 7 consecutive days. In the drop test, most rats reared in an EE remained on a disk at a height, whereas most rats reared in a standard cage fell off. RNA sequencing revealed that the immediate-early gene expression in the medial prefrontal cortex of LHRs reared in an EE was reduced. cFos-expressing neurons were reduced in number in LHRs reared in an EE. These results suggest that growing in an EE improves ADHD-like behaviors and that said improvement is due to the suppression of neuronal activity in the mPFC.
Collapse
|
15
|
Afseth C, Shim A, Anderson S, Bell AM, Hellmann JK. Vertical transmission of horizontally acquired social information in sticklebacks: implications for transgenerational plasticity. Proc Biol Sci 2022; 289:20220571. [PMID: 35855606 PMCID: PMC9297010 DOI: 10.1098/rspb.2022.0571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/27/2022] [Indexed: 12/25/2022] Open
Abstract
There is growing evidence that offspring receive information about their environment vertically, i.e. from their parents (environmental parental effects or transgenerational plasticity). For example, parents exposed to predation risk may produce offspring with heightened antipredator defences. At the same time, organisms can gain information about the environment horizontally, from conspecifics. In this study, we provide some of the first evidence that horizontally acquired social information can be transmitted vertically across generations. Three-spined stickleback (Gasterosteus aculeatus) fathers produced larval offspring with altered antipredator behaviour when fathers received visual and olfactory cues from predator-chased neighbours. Although fathers did not personally witness their neighbours being chased (i.e. they never saw the predator), changes in offspring traits were similar to those induced by direct paternal exposure to predation risk. These findings suggest that two different non-genetic pathways (horizontal transfer of social information, vertical transfer via sperm-mediated paternal effects) can combine to affect offspring phenotypes. The implications of simultaneous horizontal and vertical transmission are widely appreciated in the context of disease and culture; our results suggest that they could be equally important for the maintenance of phenotypic variation and could have profound consequences for the rate at which information flows within and across generations.
Collapse
Affiliation(s)
- Cassandra Afseth
- Department of Evolution, Ecology and Behavior, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| | - Andrew Shim
- Department of Evolution, Ecology and Behavior, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| | - Samantha Anderson
- Department of Evolution, Ecology and Behavior, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| | - Alison M. Bell
- Department of Evolution, Ecology and Behavior, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
- Program in Ecology, Evolution and Conservation, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| | - Jennifer K. Hellmann
- Department of Evolution, Ecology and Behavior, University of Illinois Urbana–Champaign, Urbana, IL 61801, USA
| |
Collapse
|
16
|
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: 3.0] [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
|
17
|
Manno FA, An Z, Kumar R, Su AJ, Liu J, Wu EX, He J, Feng Y, Lau C. Environmental enrichment leads to behavioral circadian shifts enhancing brain-wide functional connectivity between sensory cortices and eliciting increased hippocampal spiking. Neuroimage 2022; 252:119016. [DOI: 10.1016/j.neuroimage.2022.119016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/30/2021] [Accepted: 02/17/2022] [Indexed: 11/27/2022] Open
|
18
|
Li MF, Swaisgood RR, Owen MA, Zhang H, Zhang G, Qiu J, Martin MS. Consequences of nescient mating: artificial insemination increases cub rejection in the giant panda (Ailuropoda melanoleuca). Appl Anim Behav Sci 2022. [DOI: 10.1016/j.applanim.2022.105565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
19
|
Makowska IJ, Weary DM. A Good Life for Laboratory Rodents? ILAR J 2021; 60:373-388. [PMID: 32311030 DOI: 10.1093/ilar/ilaa001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/16/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
Most would agree that animals in research should be spared "unnecessary" harm, pain, or distress, and there is also growing interest in providing animals with some form of environmental enrichment. But is this the standard of care that we should aspire to? We argue that we need to work towards a higher standard-specifically, that providing research animals with a "good life" should be a prerequisite for their use. The aims of this paper are to illustrate our vision of a "good life" for laboratory rats and mice and to provide a roadmap for achieving this vision. We recognize that several research procedures are clearly incompatible with a good life but describe here what we consider to be the minimum day-to-day living conditions to be met when using rodents in research. A good life requires that animals can express a rich behavioral repertoire, use their abilities, and fulfill their potential through active engagement with their environment. In the first section, we describe how animals could be housed for these requirements to be fulfilled, from simple modifications to standard housing through to better cage designs and free-ranging options. In the second section, we review the types of interactions with laboratory rodents that are compatible with a good life. In the third section, we address the potential for the animals to have a life outside of research, including the use of pets in clinical trials (the animal-as-patient model) and the adoption of research animals to new homes when they are no longer needed in research. We conclude with a few suggestions for achieving our vision.
Collapse
Affiliation(s)
- I Joanna Makowska
- Animal Welfare Program, University of British Columbia, Vancouver, Canada.,Animal Welfare Institute, Washington, DC, USA
| | - Daniel M Weary
- Animal Welfare Program, University of British Columbia, Vancouver, Canada
| |
Collapse
|
20
|
Gapp K, Parada GE, Gross F, Corcoba A, Kaur J, Grau E, Hemberg M, Bohacek J, Miska EA. Single paternal dexamethasone challenge programs offspring metabolism and reveals multiple candidates in RNA-mediated inheritance. iScience 2021; 24:102870. [PMID: 34386731 PMCID: PMC8346661 DOI: 10.1016/j.isci.2021.102870] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/21/2021] [Accepted: 07/14/2021] [Indexed: 01/16/2023] Open
Abstract
Single traumatic events that elicit an exaggerated stress response can lead to the development of neuropsychiatric conditions. Rodent studies suggested germline RNA as a mediator of effects of chronic environmental exposures to the progeny. The effects of an acute paternal stress exposure on the germline and their potential consequences on offspring remain to be seen. We find that acute administration of an agonist for the stress-sensitive Glucocorticoid receptor, using the common corticosteroid dexamethasone, affects the RNA payload of mature sperm as soon as 3 hr after exposure. It further impacts early embryonic transcriptional trajectories, as determined by single-embryo sequencing, and metabolism in the offspring. We show persistent regulation of tRNA fragments in sperm and descendant 2-cell embryos, suggesting transmission from sperm to embryo. Lastly, we unravel environmentally induced alterations in sperm circRNAs and their targets in the early embryo, highlighting this class as an additional candidate in RNA-mediated inheritance of disease risk.
Collapse
Affiliation(s)
- Katharina Gapp
- Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, 8057, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, 8057, Switzerland
| | - Guillermo E. Parada
- Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Fridolin Gross
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, 8057, Switzerland
| | | | - Jasmine Kaur
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, 8057, Switzerland
| | - Evelyn Grau
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Department of Medicine, CITIID, University of Cambridge, Cambridge CB2 0AW, UK
| | - Martin Hemberg
- Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, 8057, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, 8057, Switzerland
| | - Eric A. Miska
- Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| |
Collapse
|
21
|
Chleilat F, Schick A, Deleemans JM, Ma K, Alukic E, Wong J, Noye Tuplin EW, Nettleton JE, Reimer RA. Paternal high protein diet modulates body composition, insulin sensitivity, epigenetics, and gut microbiota intergenerationally in rats. FASEB J 2021; 35:e21847. [PMID: 34405464 DOI: 10.1096/fj.202100198rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022]
Abstract
Mounting evidence demonstrates that paternal diet programs offspring metabolism. However, the contribution of a pre-conception paternal high protein (HP) diet to offspring metabolism, gut microbiota, and epigenetic changes remains unclear. Here we show that paternal HP intake in Sprague Dawley rats programs protective metabolic outcomes in offspring. Compared to paternal high fat/sucrose (HF/S), HP diet improved body composition and insulin sensitivity and improved circulating satiety hormones and cecal short-chain fatty acids compared to HF/S and control diet (P < .05). Further, using 16S rRNA gene sequencing to assess gut microbial composition, we observed increased alpha diversity, distinct bacterial clustering, and increased abundance of Bifidobacterium, Akkermansia, Bacteroides, and Marvinbryantia in HP fathers and/or male and female adult offspring. At the epigenetic level, DNMT1and 3b expression was altered intergenerationally. Our study identifies paternal HP diet as a modulator of gut microbial composition, epigenetic markers, and metabolic function intergenerationally.
Collapse
Affiliation(s)
- Faye Chleilat
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Alana Schick
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Julie M Deleemans
- Division of Medical Science and Psychosocial Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kyle Ma
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Erna Alukic
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Jolene Wong
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | | | - Jodi E Nettleton
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
22
|
Yang Y, Lagisz M, Foo YZ, Noble DWA, Anwer H, Nakagawa S. Beneficial intergenerational effects of exercise on brain and cognition: a multilevel meta-analysis of mean and variance. Biol Rev Camb Philos Soc 2021; 96:1504-1527. [PMID: 33783115 DOI: 10.1111/brv.12712] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
Physical exercise not only helps to improve physical health but can also enhance brain development and cognition. Recent reports on parental (both maternal and paternal) effects raise the possibility that parental exercise may provide benefits to offspring through intergenerational inheritance. However, the general magnitude and consistency of parental exercise effects on offspring is still controversial. Additionally, empirical research has long overlooked an important aspect of exercise: its effects on variability in neurodevelopmental and cognitive traits. Here, we compiled data from 52 studies involving 4786 rodents (412 effect sizes) to quantify the intergenerational transmission of exercise effects on brain and cognition. Using a multilevel meta-analytic approach, we found that, overall, parental exercise showed a tendency for increasing their offspring's brain structure by 12.7% (albeit statistically non-significant) probably via significantly facilitating neurogenesis (16.5%). Such changes in neural anatomy go in hand with a significant 20.8% improvement in neurobehaviour (improved learning and memory, and reduced anxiety). Moreover, we found parental exercise significantly reduces inter-individual differences (i.e. reduced variance in the treatment group) in progeny's neurobehaviour by 10.2% (coefficient of variation ratio, lnCVR), suggesting the existence of an individual by intervention interaction. The positive effects of exercise are modulated by several covariates (i.e. moderators), such as the exercised parent's sex, offspring's sex, and age, mode of exercise, and exercise timing. In particular, parental forced exercise is more efficient than voluntary exercise at significantly improving offspring neurobehaviour (26.0%) and reducing its variability (14.2%). We observed larger effects when parental exercise started before pregnancy. However, exercising only during pregnancy also had positive effects. Mechanistically, exercise significantly upregulated brain-derived neurotrophic factor (BDNF) by 28.9%, vascular endothelial growth factor (VEGF) by 35.8%, and significantly decreased hippocampal DNA methylation by 3.5%, suggesting that brain growth factor cascades and epigenetic modifications can moderate the transmission of parental exercise effects. Collectively, by coupling mean with variance effects, our analyses draw a more integrated picture of the benefits that parental exercise has on offspring: not only does it improve offspring brain development and cognitive performance, but it also reduces inter-individual differences in cognition-related traits. We advocate that meta-analysis of variation together with the mean of a trait provides novel insights for old controversies as well as emerging new questions, opening up a new era for generating variance-based hypotheses.
Collapse
Affiliation(s)
- Yefeng Yang
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.,Department of Biosystems Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Malgorzata Lagisz
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Yong Zhi Foo
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Daniel W A Noble
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.,Division of Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Hamza Anwer
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Shinichi Nakagawa
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
23
|
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: 11] [Impact Index Per Article: 2.8] [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
|
24
|
Salmanzadeh H, Ahmadi-Soleimani SM, Azadi M, Halliwell RF, Azizi H. Adolescent Substance Abuse, Transgenerational Consequences and Epigenetics. Curr Neuropharmacol 2021; 19:1560-1569. [PMID: 33655865 PMCID: PMC8762180 DOI: 10.2174/1570159x19666210303121519] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/13/2021] [Accepted: 02/12/2021] [Indexed: 11/22/2022] Open
Abstract
Adolescence is the transitional period between childhood and adulthood and a critical period in brain development. Adolescence in humans is also associated with increased expression of risk-taking behaviors. Epidemiological and clinical studies, for example, show a surge of drug abuse and raise the hypothesis that the adolescent brain undergoes critical changes resulting in diminished control. Determining how substance abuse during this critical period might cause longterm neurobiological changes in cognition and behavior is therefore critically important. The present work aims to provide an evaluation of the transgenerational and multi-generational phenotypes derived from parent animals exposed to drugs of abuse only during their adolescence. Specifically, we will consider changes found following the administration of cannabinoids, nicotine, alcohol and opiates. In addition, epigenetic modifications of the genome following drug exposure will be discussed as emerging evidence of the underlying adverse transgenerational effects. Notwithstanding, much of the new data discussed here is from animal models, indicating that future clinical studies are much needed to better understand the neurobiological consequences and mechanisms of drug actions on the human brains' development and maturation.
Collapse
Affiliation(s)
| | | | | | - Robert F. Halliwell
- Address correspondence to this author at the TJ Long School of Pharmacy, University of the Pacific, Stockton, California, USA; Tel: +1 (209) 946 2074; E-mail: and Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Tel: +98-21-82884587; Fax: +98-21-82884528; E-mail:
| | - Hossein Azizi
- Address correspondence to this author at the TJ Long School of Pharmacy, University of the Pacific, Stockton, California, USA; Tel: +1 (209) 946 2074; E-mail: and Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Tel: +98-21-82884587; Fax: +98-21-82884528; E-mail:
| |
Collapse
|
25
|
Development and Control of Behaviour. Anim Behav 2021. [DOI: 10.1007/978-3-030-82879-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Hellmann JK, Bukhari SA, Deno J, Bell AM. Sex-specific plasticity across generations I: Maternal and paternal effects on sons and daughters. J Anim Ecol 2020; 89:2788-2799. [PMID: 33191518 PMCID: PMC7902357 DOI: 10.1111/1365-2656.13364] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023]
Abstract
Intergenerational plasticity or parental effects-when parental environments alter the phenotype of future generations-can influence how organisms cope with environmental change. An intriguing, underexplored possibility is that sex-of both the parent and the offspring-plays an important role in driving the evolution of intergenerational plasticity in both adaptive and non-adaptive ways. Here, we evaluate the potential for sex-specific parental effects in a freshwater population of three-spined sticklebacks Gasterosteus aculeatus by independently and jointly manipulating maternal and paternal experiences and separately evaluating their phenotypic effects in sons versus daughters. We tested the adaptive hypothesis that daughters are more responsive to cues from their mother, whereas sons are more responsive to cues from their father. We exposed mothers, fathers or both parents to visual cues of predation risk and measured offspring antipredator traits and brain gene expression. Predator-exposed fathers produced sons that were more risk-prone, whereas predator-exposed mothers produced more anxious sons and daughters. Furthermore, maternal and paternal effects on offspring survival were non-additive: offspring with a predator-exposed father, but not two predator-exposed parents, had lower survival against live predators. There were also strong sex-specific effects on brain gene expression: exposing mothers versus fathers to predation risk activated different transcriptional profiles in their offspring, and sons and daughters strongly differed in the ways in which their brain gene expression profiles were influenced by parental experience. We found little evidence to support the hypothesis that offspring prioritize their same-sex parent's experience. Parental effects varied with both the sex of the parent and the offspring in complicated and non-additive ways. Failing to account for these sex-specific patterns (e.g. by pooling sons and daughters) would have underestimated the magnitude of parental effects. Altogether, these results draw attention to the potential for sex to influence patterns of intergenerational plasticity and raise new questions about the interface between intergenerational plasticity and sex-specific selective pressures, sexual conflict and sexual selection.
Collapse
Affiliation(s)
- Jennifer K Hellmann
- Department of Evolution, Ecology and Behavior, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
| | - Syed Abbas Bukhari
- Department of Evolution, Ecology and Behavior, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
| | - Jack Deno
- Department of Evolution, Ecology and Behavior, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
| | - Alison M Bell
- Department of Evolution, Ecology and Behavior, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
- Program in Ecology, Evolution and Conservation, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
| |
Collapse
|
27
|
Hellmann JK, Carlson ER, Bell AM. Sex-specific plasticity across generations II: Grandpaternal effects are lineage specific and sex specific. J Anim Ecol 2020; 89:2800-2812. [PMID: 33191513 PMCID: PMC7902365 DOI: 10.1111/1365-2656.13365] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 07/07/2020] [Indexed: 12/31/2022]
Abstract
Transgenerational plasticity (TGP) occurs when the environment encountered by one generation (F0) alters the phenotypes of one or more future generations (e.g. F1 and F2). Sex selective TGP, via specific lineages or to only male or female descendants, has been underexplored in natural systems, and may be adaptive if it allows past generations to fine-tune the phenotypes of future generations in response to sex-specific life-history strategies. We sought to understand if exposing males to predation risk can influence grandoffspring via sperm in three-spined stickleback Gasterosteus aculeatus. We specifically tested the hypothesis that grandparental effects are transmitted in a sex-specific way down the male lineage, from paternal grandfathers to F2 males. We reared F1 offspring of unexposed and predator-exposed F0 males under 'control' conditions and used them to generate F2s with control grandfathers, a predator-exposed maternal grandfather (i.e. predator-exposed F0 males to F1 daughters to F2s), a predator-exposed paternal grandfather (i.e. predator-exposed F0 males to F1 sons to F2s) or two predator-exposed grandfathers. We then assayed male and female F2s for a variety of traits related to antipredator defence. We found little evidence that transgenerational effects were mediated to only male descendants via the paternal lineage. Instead, grandpaternal effects depended on lineage and were mediated largely across sexes, from F1 males to F2 females and from F1 females to F2 males. When their paternal grandfather was exposed to predation risk, female F2s were heavier and showed a reduced change in behaviour in response to a simulated predator attack relative to grandoffspring of control, unexposed grandparents. In contrast, male F2s showed reduced antipredator behaviour when their maternal grandfather was exposed to predation risk. However, these patterns were only evident when one grandfather, but not both grandfathers, was exposed to predation risk, suggesting the potential for non-additive interactions across lineages. If sex-specific and lineage effects are common, then grandparental effects are likely underestimated in the literature. These results draw attention to the importance of sex-selective inheritance of environmental effects and raise new questions about the proximate and ultimate causes of selective transmission across generations.
Collapse
Affiliation(s)
- Jennifer K Hellmann
- Department of Evolution, Ecology and Behavior, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
| | - Erika R Carlson
- Department of Evolution, Ecology and Behavior, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
| | - Alison M Bell
- Department of Evolution, Ecology and Behavior, School of Integrative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
- Program in Ecology, Evolution and Conservation, University of Illinois Urbana-Champaign, Urbana, Illinois, USA, 61801
| |
Collapse
|
28
|
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: 3.2] [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
|
29
|
Sex-biased impact of endocrine disrupting chemicals on behavioral development and vulnerability to disease: Of mice and children. Neurosci Biobehav Rev 2020; 121:29-46. [PMID: 33248148 DOI: 10.1016/j.neubiorev.2020.11.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 10/16/2020] [Accepted: 11/14/2020] [Indexed: 12/14/2022]
Abstract
Sex is a fundamental biological characteristic that influences many aspects of an organism's phenotype, including neurobiological functions and behavior as a result of species-specific evolutionary pressures. Sex differences have strong implications for vulnerability to disease and susceptibility to environmental perturbations. Endocrine disrupting chemicals (EDCs) have the potential to interfere with sex hormones functioning and influence development in a sex specific manner. Here we present an updated descriptive review of findings from animal models and human studies regarding the current evidence for altered sex-differences in behavioral development in response to early exposure to EDCs, with a focus on bisphenol A and phthalates. Overall, we show that animal and human studies have a good degree of consistency and that there is strong evidence demonstrating that EDCs exposure during critical periods of development affect sex differences in emotional and cognitive behaviors. Results are more heterogeneous when social, sexual and parental behaviors are considered. In order to pinpoint sex differences in environmentally-driven disease vulnerabilities, researchers need to consider sex-biased developmental effects of EDCs.
Collapse
|
30
|
Azadi M, Gompf HS, Azizi H. Paternal exposure to morphine during adolescence potentiates morphine withdrawal in male offspring: Involvement of the lateral paragigantocellularis nucleus. J Psychopharmacol 2020; 34:1289-1299. [PMID: 33112218 DOI: 10.1177/0269881120953993] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Opiate exposure during adolescence perturbs the brain's maturation process and potentially confers long-term adverse consequences, not only in exposed individuals but also in their posterity. Here, we investigate the outcomes of adolescent paternal morphine exposure on morphine withdrawal profile in male offspring. METHODS Male Wistar rats were chronically subjected to 10 days of an escalating regimen of morphine during adolescence. After a 20-day washout period, adult males were allowed to copulate with naïve females. The adult male offspring were tested for somatic and affective components of naloxone-precipitated morphine withdrawal using conditioned place aversion. Moreover, electrical activity of the lateral paragigantocellularis (LPGi) nucleus, which is involved in development of opiate dependence, was recorded in response to a challenge dose of morphine via extracellular single-unit recordings. RESULTS Morphine-sired offspring exhibited augmented expression of naloxone-induced somatic and affective signs of opiate withdrawal compared to the control saline-sired counterparts. In vivo recording revealed that LPGi neurons displayed heterogeneous responses (inhibitory, excitatory, and no change) to acute morphine administration in both morphine- and saline-sired animals. The morphine-induced discharge inhibition was potentiated in morphine-sired offspring. However, the extent of discharge excitation in response to morphine did not reach significance in these subjects. Moreover, the lack of alteration in maternal behavior toward morphine-sired offspring indicates that this is due to germline-dependent transmission of epigenetic traits across generations. CONCLUSIONS Preconception paternal exposure to morphine during adolescence potentiates opiate withdrawal signs in male offspring which is mediated, at least in part, by epigenetic alteration of LPGi-related brain circuitry.
Collapse
Affiliation(s)
- Maryam Azadi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Heinrich S Gompf
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, USA
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
31
|
Krug A, Wöhr M, Seffer D, Rippberger H, Sungur AÖ, Dietsche B, Stein F, Sivalingam S, Forstner AJ, Witt SH, Dukal H, Streit F, Maaser A, Heilmann-Heimbach S, Andlauer TFM, Herms S, Hoffmann P, Rietschel M, Nöthen MM, Lackinger M, Schratt G, Koch M, Schwarting RKW, Kircher T. Advanced paternal age as a risk factor for neurodevelopmental disorders: a translational study. Mol Autism 2020; 11:54. [PMID: 32576230 PMCID: PMC7310295 DOI: 10.1186/s13229-020-00345-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 05/07/2020] [Indexed: 01/13/2023] Open
Abstract
Advanced paternal age (APA) is a risk factor for several neurodevelopmental disorders, including autism and schizophrenia. The potential mechanisms conferring this risk are poorly understood. Here, we show that the personality traits schizotypy and neuroticism correlated with paternal age in healthy subjects (N = 677). Paternal age was further positively associated with gray matter volume (VBM, N = 342) in the right prefrontal and the right medial temporal cortex. The integrity of fiber tracts (DTI, N = 222) connecting these two areas correlated positively with paternal age. Genome-wide methylation analysis in humans showed differential methylation in APA individuals, linking APA to epigenetic mechanisms. A corresponding phenotype was obtained in our rat model. APA rats displayed social-communication deficits and emitted fewer pro-social ultrasonic vocalizations compared to controls. They further showed repetitive and stereotyped patterns of behavior, together with higher anxiety during early development. At the neurobiological level, microRNAs miR-132 and miR-134 were both differentially regulated in rats and humans depending on APA. This study demonstrates associations between APA and social behaviors across species. They might be driven by changes in the expression of microRNAs and/or epigenetic changes regulating neuronal plasticity, leading to brain morphological changes and fronto-hippocampal connectivity, a network which has been implicated in social interaction.
Collapse
Affiliation(s)
- Axel Krug
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039, Marburg, Germany.
- Center for Mind, Brain and Behavior, Marburg, Germany.
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany.
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, Marburg, Germany
- Laboratory for Behavioral Neuroscience, Department of Biology, Faculty of Science, University of Southern Denmark, Odense, Denmark
| | - Dominik Seffer
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University Marburg, Marburg, Germany
| | - Henrike Rippberger
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University Marburg, Marburg, Germany
| | - A Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, Marburg, Germany
| | - Bruno Dietsche
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039, Marburg, Germany
| | - Sugirthan Sivalingam
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Mannheim, Germany
| | - Helene Dukal
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Mannheim, Germany
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Mannheim, Germany
| | - Anna Maaser
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Mannheim, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
| | - Martin Lackinger
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-University Marburg, 35043, Marburg, Germany
| | - Gerhard Schratt
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-University Marburg, 35043, Marburg, Germany
- Lab of Systems Neuroscience, Department of Health Science and Technology, Institute for Neuroscience, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Michael Koch
- Department of Neuropharmacology, Brain Research Institute, Centre for Cognitive Sciences, University of Bremen, 28334, Bremen, Germany
| | - Rainer K W Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, Marburg, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039, Marburg, Germany
- Center for Mind, Brain and Behavior, Marburg, Germany
| |
Collapse
|
32
|
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: 27] [Impact Index Per Article: 5.4] [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
|
33
|
Cauceglia JW, Nelson AC, Rubinstein ND, Kukreja S, Sasso LN, Beaufort JA, Rando OJ, Potts WK. Transitions in paternal social status predict patterns of offspring growth and metabolic transcription. Mol Ecol 2020; 29:624-638. [PMID: 31885115 DOI: 10.1111/mec.15346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 11/27/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
Abstract
One type of parental effect occurs when changes in parental phenotype or environment trigger changes to offspring phenotype. Such nongenetic parental effects can be precisely triggered in response to an environmental cue in time-locked fashion, or in other cases, persist for multiple generations after the cue has been removed, suggesting multiple timescales of action. For parental effects to serve as reliable signals of current environmental conditions, they should be reversible, such that when cues change, offspring phenotypes change in accordance. Social hierarchy is a prevalent feature of the environment, and current parental social status could signal the environment in which offspring will be born. Here, we sought to address parental effects of social status and their timescale of action in mice. We show that territorial competition in seminatural environments affects offspring growth. Although dominant males are not heavier than nondominant or control males, they produce faster growing offspring, particularly sons. The timing, effect-size, and sex-specificity of this association are modulated by maternal social experience. We show that a change in paternal social status is sufficient to modulate offspring weight: from one breeding cycle to the next, status-ascending males produce heavier sons than before, and status-descending males produce lighter sons than before. Current paternal status is also highly predictive of liver transcription in sons, including molecular pathways controlling oxidative phosphorylation and iron metabolism. These results are consistent with a parental effect of social experience, although alternative explanations are considered. In summary, changes in paternal social status are associated with changes in offspring growth and metabolism.
Collapse
Affiliation(s)
- Joseph W Cauceglia
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Adam C Nelson
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | | | - Shweta Kukreja
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lynsey N Sasso
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
| | - John A Beaufort
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Wayne K Potts
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
34
|
Abstract
In recent decades, human sociocultural changes have increased the numbers of fathers that are involved in direct caregiving in Western societies. This trend has led to a resurgence of interest in understanding the mechanisms and effects of paternal care. Across the animal kingdom, paternal caregiving has been found to be a highly malleable phenomenon, presenting with great variability among and within species. The emergence of paternal behaviour in a male animal has been shown to be accompanied by substantial neural plasticity and to be shaped by previous and current caregiving experiences, maternal and infant stimuli and ecological conditions. Recent research has allowed us to gain a better understanding of the neural basis of mammalian paternal care, the genomic and circuit-level mechanisms underlying paternal behaviour and the ways in which the subcortical structures that support maternal caregiving have evolved into a global network of parental care. In addition, the behavioural, neural and molecular consequences of paternal caregiving for offspring are becoming increasingly apparent. Future cross-species research on the effects of absence of the father and the transmission of paternal influences across generations may allow research on the neuroscience of fatherhood to impact society at large in a number of important ways.
Collapse
|
35
|
Fusco S, Spinelli M, Cocco S, Ripoli C, Mastrodonato A, Natale F, Rinaudo M, Livrizzi G, Grassi C. Maternal insulin resistance multigenerationally impairs synaptic plasticity and memory via gametic mechanisms. Nat Commun 2019; 10:4799. [PMID: 31641124 PMCID: PMC6805915 DOI: 10.1038/s41467-019-12793-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Metabolic diseases harm brain health and cognitive functions, but whether maternal metabolic unbalance may affect brain plasticity of next generations is still unclear. Here, we demonstrate that maternal high fat diet (HFD)-dependent insulin resistance multigenerationally impairs synaptic plasticity, learning and memory. HFD downregulates BDNF and insulin signaling in maternal tissues and epigenetically inhibits BDNF expression in both germline and hippocampus of progeny. Notably, exposure of the HFD offspring to novel enriched environment restores Bdnf epigenetic activation in the male germline and counteracts the transmission of cognitive impairment to the next generations. BDNF administration to HFD-fed mothers or preserved insulin sensitivity in HFD-fed p66Shc KO mice also prevents the intergenerational transmission of brain damage to the progeny. Collectively, our data suggest that maternal diet multigenerationally impacts on descendants' brain health via gametic mechanisms susceptible to lifestyle.
Collapse
Affiliation(s)
- Salvatore Fusco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy. .,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| | - Matteo Spinelli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Cristian Ripoli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy.,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Alessia Mastrodonato
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Francesca Natale
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Marco Rinaudo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Giulia Livrizzi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy. .,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| |
Collapse
|
36
|
Tuscher JJ, Day JJ. Multigenerational epigenetic inheritance: One step forward, two generations back. Neurobiol Dis 2019; 132:104591. [PMID: 31470104 DOI: 10.1016/j.nbd.2019.104591] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/22/2019] [Accepted: 08/26/2019] [Indexed: 02/08/2023] Open
Abstract
Modifications to DNA and histone proteins serve a critical regulatory role in the developing and adult brain, and over a decade of research has established the importance of these "epigenetic" modifications in a wide variety of brain functions across the lifespan. Epigenetic patterns orchestrate gene expression programs that establish the phenotypic diversity of various cellular classes in the central nervous system, play a key role in experience-dependent gene regulation in the adult brain, and are commonly implicated in neurodevelopmental, psychiatric, and neurodegenerative disease states. In addition to these established roles, emerging evidence indicates that epigenetic information can potentially be transmitted to offspring, giving rise to inter- and trans-generational epigenetic inheritance phenotypes. However, our understanding of the cellular events that participate in this information transfer is incomplete, and the ability of this transfer to overcome complete epigenetic reprogramming during embryonic development is highly controversial. This review explores the existing literature on multigenerational epigenetic mechanisms in the central nervous system. First, we focus on the cellular mechanisms that may perpetuate or counteract this type of information transfer, and consider how epigenetic modification in germline and somatic cells regulate important aspects of cellular and organismal development. Next, we review the potential phenotypes resulting from ancestral experiences that impact gene regulatory modifications, including how these changes may give rise to unique metabolic phenotypes. Finally, we discuss several caveats and technical limitations that influence multigenerational epigenetic effects. We argue that studies reporting multigenerational epigenetic changes impacting the central nervous system must be interpreted with caution, and provide suggestions for how epigenetic information transfer can be mechanistically disentangled from genetic and environmental influences on brain function.
Collapse
Affiliation(s)
- Jennifer J Tuscher
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Jeremy J Day
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
37
|
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: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
38
|
Nieto SJ, Kosten TA. Who's your daddy? Behavioral and epigenetic consequences of paternal drug exposure. Int J Dev Neurosci 2019; 78:109-121. [PMID: 31301337 DOI: 10.1016/j.ijdevneu.2019.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/11/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022] Open
Abstract
Substance use disorders (SUDs) reflect genetic and environmental factors. While identifying reliable genetic variants that predispose individuals to developing SUDs has been challenging, epigenetic factors may also contribute to the heritability of SUDs. Familial drug use associates with a wide range of problems in children, including an increased risk for developing a SUD. The implications of maternal drug use on offspring development are a well-studied area; however, paternal drug use prior to conception has received relatively little attention. Paternal exposure to several environmental stimuli (i.e. stress or diet manipulations) results in behavioral and epigenetic changes in offspring. The purpose of this review is to determine the state of the preclinical literature on the behavioral and epigenetic consequences of paternal drug exposure. Drug-sired offspring show several developmental and physiological abnormalities. These offspring also show deficits in cognitive and emotional domains. Examining sensitivity to drugs in offspring is a growing area of research. Drug-sired offspring are resistant to the rewarding and reinforcing properties of drugs. However, greater paternal motivation for the drug, combined with high drug intake, can result in addiction-like behaviors in offspring. Drug-sired offspring also show altered histone modifications and DNA methylation levels of imprinted genes and microRNAs; epigenetic-mediated changes were also noted in genes related to glutamatergic and neurotrophic factor signaling. In some instances, drug use resulted in aberrant epigenetic modifications in sire sperm, and these changes were maintained in the brains of offspring. Thus, paternal drug exposure has long-lasting consequences that include altered drug sensitivity in subsequent generations. We discuss factors (i.e. maternal behaviors) that may moderate these paternal drug-induced effects as well as ideas for future directions.
Collapse
Affiliation(s)
- Steven J Nieto
- University of Houston, Department of Psychology & Texas Institute for Measurement, Evaluation and Statistics (TIMES), Houston, TX, 77204-6022, United States
| | - Therese A Kosten
- University of Houston, Department of Psychology & Texas Institute for Measurement, Evaluation and Statistics (TIMES), Houston, TX, 77204-6022, United States
| |
Collapse
|
39
|
Rompala GR, Homanics GE. Intergenerational Effects of Alcohol: A Review of Paternal Preconception Ethanol Exposure Studies and Epigenetic Mechanisms in the Male Germline. Alcohol Clin Exp Res 2019; 43:1032-1045. [PMID: 30908630 PMCID: PMC6551262 DOI: 10.1111/acer.14029] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/13/2019] [Indexed: 12/11/2022]
Abstract
While alcohol use disorder (AUD) is a highly heritable psychiatric disease, efforts to elucidate that heritability by examining genetic variation (e.g., single nucleotide polymorphisms) have been insufficient to fully account for familial AUD risk. Perhaps not coincidently, there has been a burgeoning interest in novel nongenomic mechanisms of inheritance (i.e., epigenetics) that are shaped in the male or female germ cells by significant lifetime experiences such as exposure to chronic stress, malnutrition, or drugs of abuse. While many epidemiological and preclinical studies have long pointed to a role for the parental preconception environment in offspring behavior, over the last decade many studies have implicated a causal relationship between the environmentally sensitive sperm epigenome and intergenerational phenotypes. This critical review will detail the heritable effects of alcohol and the potential role for epigenetics.
Collapse
Affiliation(s)
- Gregory R Rompala
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gregg E Homanics
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School Medicine, Pittsburgh, Pennsylvania
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
40
|
Kentner AC, Cryan JF, Brummelte S. Resilience priming: Translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019; 61:350-375. [PMID: 30311210 PMCID: PMC6447439 DOI: 10.1002/dev.21775] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the increasing attention to early life adversity and its long-term consequences on health, behavior, and the etiology of neurodevelopmental disorders, our understanding of the adaptations and interventions that promote resiliency and rescue against such insults are underexplored. Specifically, investigations of the perinatal period often focus on negative events/outcomes. In contrast, positive experiences (i.e. enrichment/parental care//healthy nutrition) favorably influence development of the nervous and endocrine systems. Moreover, some stressors result in adaptations and demonstrations of later-life resiliency. This review explores the underlying mechanisms of neuroplasticity that follow some of these early life experiences and translates them into ideas for interventions in pediatric settings. The emerging role of the gut microbiome in mediating stress susceptibility is also discussed. Since many negative outcomes of early experiences are known, it is time to identify mechanisms and mediators that promote resiliency against them. These range from enrichment, quality parental care, dietary interventions and those that target the gut microbiota.
Collapse
Affiliation(s)
- Amanda C. Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115,
| | - John F. Cryan
- Dept. Anatomy & Neuroscience & APC Microbiome Institute, University College Cork, College Rd., Cork, Ireland,
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202,
| |
Collapse
|
41
|
Vågerö D, Pinger PR, Aronsson V, van den Berg GJ. Paternal grandfather's access to food predicts all-cause and cancer mortality in grandsons. Nat Commun 2018; 9:5124. [PMID: 30538239 PMCID: PMC6290014 DOI: 10.1038/s41467-018-07617-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
Studies of animals and plants suggest that nutritional conditions in one generation may affect phenotypic characteristics in subsequent generations. A small number of human studies claim to show that pre-pubertal nutritional experience trigger a sex-specific transgenerational response along the male line. A single historical dataset, the Överkalix cohorts in northern Sweden, is often quoted as evidence. To test this hypothesis on an almost 40 times larger dataset we collect harvest data during the pre-pubertal period of grandparents (G0, n = 9,039) to examine its potential association with mortality in children (G1, n = 7,280) and grandchildren (G2, n = 11,561) in the Uppsala Multigeneration Study. We find support for the main Överkalix finding: paternal grandfather's food access in pre-puberty predicts his male, but not female, grandchildren's all-cause mortality. In our study, cancer mortality contributes strongly to this pattern. We are unable to reproduce previous results for diabetes and cardiovascular mortality.
Collapse
Affiliation(s)
- Denny Vågerö
- CHESS, Centre for Health Equity Studies, Department of Public Health Sciences, SE-106 91 Stockholm University, Stockholm, Sweden.
| | - Pia R Pinger
- Department of Economics, University of Bonn, Adenauerallee 24-42, 53113, Bonn, Germany.,briq, Institute on Behavior & Inequality, Bonn, Germany
| | - Vanda Aronsson
- CHESS, Centre for Health Equity Studies, Department of Public Health Sciences, SE-106 91 Stockholm University, Stockholm, Sweden
| | - Gerard J van den Berg
- Department of Economics, Priory Rd Complex, University of Bristol, Bristol, BS8 ITU, United Kingdom.,IFAU, Institute for Evaluation of Labor Market and Education Policy, Uppsala, Sweden
| |
Collapse
|
42
|
Stenz L, Schechter DS, Serpa SR, Paoloni-Giacobino A. Intergenerational Transmission of DNA Methylation Signatures Associated with Early Life Stress. Curr Genomics 2018; 19:665-675. [PMID: 30532646 PMCID: PMC6225454 DOI: 10.2174/1389202919666171229145656] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/25/2017] [Accepted: 12/17/2017] [Indexed: 02/07/2023] Open
Abstract
Early life stress in humans (i.e. maltreatment, violence exposure, loss of a loved one) and in rodents (i.e. disrupted attachment or nesting, electric shock, restraint, predator odor) occurs during critical steps of neural circuit formation. ELS in humans is associated with increased risk for developmental psychopathology, including anxious and depressive phenotypes. The biological mechanisms underlying these potentially persistent maladaptive changes involve long-term epigenetic modifications, which have been suggested to be potentially transmissible to subsequent generations. DNA methylation is an epigenetic mechanism that modifies gene expression patterns in response to environmental challenges and influences mutation rates. It remains to be seen whether a functionally relevant fraction of DNA methylation marks can escape genome-wide erasures that occur in primordial germ cells and after fertilization within the zygote. Early life-stress-triggered changes in epigenetic mediated transmission of acquired behavioral traits among humans have been assessed mainly by targeting genes involved in the hypothalamic-pituitary-adrenal (HPA) axis, such as NR3C1 and FKBP5. Recently, researchers examining epigenetic transmission have begun to apply genome-wide approaches. In humans, reduced representation bisulfite sequencing (RRBS) was performed on peripheral samples that were obtained from individuals who were prenatally exposed to the "Dutch Hunger Winter", resulting in two Differentially Methylated Regions (DMRs) in INSR and CPTIA genes that were functionally, biologically and technically validated, and significantly associated with birth weights and LDL cholesterol levels in offspring. In rodents, non-genomic intergenerational transmission of anxiety which was associated with differentially methylated enhancers that were putatively involved in lipid signaling and synaptic/neurotransmission in hippocampal granule cells, was discovered also using RRBS. Finally, transgenerational transmission of altered behaviors was associated with sperm-derived microRNAs produced by ELS male mice. The field of epigenetic transmission is just beginning to enter the epigenomic era by using genome-wide analyses. Such approaches remain of strong interest to human studies, first in order to help to assess the relevance of the previous targeted studies, and second to discover new important epigenetic modifications of potential clinical importance. New discoveries may help to assess how transmittable the negative impact of stress may be to offspring. The latter may open doors for future treatments and resilience-promoting interventions, as well as new approaches to treat the effects of childhood trauma before the onset of psychiatric disorder.
Collapse
Affiliation(s)
| | - Daniel S. Schechter
- Address correspondence to this author at the Department of Child & Adolescent Psychiatry, University of Geneva Hospitals and Faculty of Medicine, Geneva, Switzerland; Tel: +41 (022) 372 5067; Fax: +91 (022) 372 5077; E-mail:
| | | | | |
Collapse
|
43
|
Baptissart M, Lamb HE, To K, Bradish C, Tehrani J, Reif D, Cowley M. Neonatal mice exposed to a high-fat diet in utero influence the behaviour of their nursing dam. Proc Biol Sci 2018; 285:rspb.2018.1237. [PMID: 30429298 DOI: 10.1098/rspb.2018.1237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/23/2018] [Indexed: 01/04/2023] Open
Abstract
The behaviour of a nursing dam influences the development, physiology, and behaviour of her offspring. Maternal behaviours can be modulated both by environmental factors, including diet, and by physical or behavioural characteristics of the offspring. In most studies of the effects of the environment on maternal behaviour, F0 dams nurse their own F1 offspring. Because the F1 are indirectly exposed to the environmental stressor in utero in these studies, it is not possible to differentiate between effects on maternal behaviour from direct exposure of the dam and those mediated by changes in the F1 as a consequence of in utero exposure. In this study, we used a mouse model of high-fat (HF) diet feeding, which has been shown to influence maternal behaviours, combined with cross-fostering to discriminate between these effects. We tested whether the diet of the F0 dam or the exposure experienced by the F1 pups in utero is the most significant predictor of maternal behaviour. Neither factor significantly influenced pup retrieval behaviours. However, strikingly, F1 in utero exposure was a significant predictor of maternal behaviour in the 15 min immediately following pup retrieval while F0 diet had no discernable effect. Our findings suggest that in utero exposure to HF diet programmes physiological changes in the offspring which influence the maternal behaviours of their dam after birth.
Collapse
Affiliation(s)
- Marine Baptissart
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Harold E Lamb
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Kimberly To
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | - Christine Bradish
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Jesse Tehrani
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - David Reif
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | - Michael Cowley
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA .,W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
44
|
Cutuli D, Berretta E, Laricchiuta D, Caporali P, Gelfo F, Petrosini L. Pre-reproductive Parental Enriching Experiences Influence Progeny's Developmental Trajectories. Front Behav Neurosci 2018; 12:254. [PMID: 30483072 PMCID: PMC6240645 DOI: 10.3389/fnbeh.2018.00254] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/10/2018] [Indexed: 01/09/2023] Open
Abstract
While the positive effects of environmental enrichment (EE) applied after weaning, in adulthood, during aging, or even in the presence of brain damage have been widely described, the transgenerational effects of pre-reproductive EE have been less examined. And yet, this issue is remarkable given that parental environmental experience may imprint offspring’s phenotype over generations through many epigenetic processes. Interactions between individual and environment take place lifelong even before conception. In fact, the environment pre-reproductively experienced by the mother and/or the father exerts a substantial impact on neural development and motor and cognitive performances of the offspring, even if not directly exposed to social, cognitive, physical and/or motor enrichment. Furthermore, pre-reproductive parental enrichment exerts a transgenerational impact on coping response to stress as well as on the social behavior of the offspring. Among the effects of pre-reproductive parental EE, a potentiation of the maternal care and a decrease in global methylation levels in the frontal cortex and hippocampus of the progeny have been described. Finally, pre-reproductive EE modifies different pathways of neuromodulation in the brain of the offspring (involving brain-derived neurotrophic factor, oxytocin and glucocorticoid receptors). The present review highlights the importance of pre-reproductive parental enrichment in altering the performances not only of animals directly experiencing it, but also of their progeny, thus opening the way to new hypotheses on the inheritance mechanisms of behavioral traits.
Collapse
Affiliation(s)
- Debora Cutuli
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Erica Berretta
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Daniela Laricchiuta
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Paola Caporali
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| | - Francesca Gelfo
- Fondazione Santa Lucia, Rome, Italy.,Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| | - Laura Petrosini
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
45
|
Kinnally EL, Gonzalez MN, Capitanio JP. Paternal line effects of early experiences persist across three generations in rhesus macaques. Dev Psychobiol 2018; 60:879-888. [DOI: 10.1002/dev.21771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/11/2018] [Accepted: 07/05/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Erin L. Kinnally
- California National Primate Research Center; University of California; Davis California
| | - Mireille N. Gonzalez
- California National Primate Research Center; University of California; Davis California
| | - John P. Capitanio
- California National Primate Research Center; University of California; Davis California
- Department of Psychology; University of California; Davis California
| |
Collapse
|
46
|
Gale T, Garratt M, Brooks RC. Perceived threats of infanticide reduce maternal allocation during lactation and lead to elevated oxidative damage in offspring. Funct Ecol 2018. [DOI: 10.1111/1365-2435.13146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Teagan Gale
- School of Biological, Earth and Environmental Sciences (BEES) Evolution and Ecology Research Centre University of New South Wales Kensington NSW Australia
| | - Michael Garratt
- Department of Pathology University of Michigan Medical School Ann Arbor Michigan
| | - Robert C. Brooks
- School of Biological, Earth and Environmental Sciences (BEES) Evolution and Ecology Research Centre University of New South Wales Kensington NSW Australia
| |
Collapse
|
47
|
Intergenerational Sex-Specific Transmission of Maternal Social Experience. Sci Rep 2018; 8:10529. [PMID: 30002484 PMCID: PMC6043535 DOI: 10.1038/s41598-018-28729-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/27/2018] [Indexed: 02/07/2023] Open
Abstract
The social environment is a major determinant of individual stress response and lifetime health. The present study shows that (1) social enrichment has a significant impact on neuroplasticity and behaviour particularly in females; and (2) social enrichment in females can be transmitted to their unexposed female descendants. Two generations (F0 and F1) of male and female rats raised in standard and social housing conditions were examined for neurohormonal and molecular alterations along with changes in four behavioural modalities. In addition to higher cortical neuronal density and cortical thickness, social experience in mothers reduced hypothalamic-pituitary-adrenal (HPA) axis activity in F0 rats and their F1 non-social housing offspring. Only F0 social mothers and their F1 non-social daughters displayed improved novelty-seeking exploratory behaviour and reduced anxiety-related behaviour whereas their motor and cognitive performance remained unchanged. Also, cortical and mRNA measurements in the F1 generation were affected by social experience intergenerationally via the female lineage (mother-to-daughter). These findings indicate that social experience promotes cortical neuroplasticity, neurohormonal and behavioural outcomes, and these changes can be transmitted to the F1 non-social offspring in a sexually dimorphic manner. Thus, a socially stimulating environment may form new biobehavioural phenotypes not only in exposed individuals, but also in their intergenerationally programmed descendants.
Collapse
|
48
|
Vega‐Trejo R, Kruuk LEB, Jennions MD, Head ML. What happens to offspring when parents are inbred, old or had a poor start in life? Evidence for sex‐specific parental effects. J Evol Biol 2018; 31:1138-1151. [DOI: 10.1111/jeb.13292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Regina Vega‐Trejo
- Division of Ecology & Evolution, Research School of Biology The Australian National University, Acton Canberra ACT Australia
| | - Loeske E. B. Kruuk
- Division of Ecology & Evolution, Research School of Biology The Australian National University, Acton Canberra ACT Australia
| | - Michael D. Jennions
- Division of Ecology & Evolution, Research School of Biology The Australian National University, Acton Canberra ACT Australia
| | - Megan L. Head
- Division of Ecology & Evolution, Research School of Biology The Australian National University, Acton Canberra ACT Australia
| |
Collapse
|
49
|
Korgan AC, O'Leary E, King JL, Weaver ICG, Perrot TS. Effects of paternal high-fat diet and rearing environment on maternal investment and development of defensive responses in the offspring. Psychoneuroendocrinology 2018. [PMID: 29518693 DOI: 10.1016/j.psyneuen.2018.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Paternal preconception risk factors (e.g. stress, diet, drug use) correlate with metabolic dysfunction in offspring, which is often comorbid with depressive and anxiety-like phenotypes. Detection of these risk factors or deleterious phenotypes informs a female about prevailing ecological demands, in addition to potential adverse environment-induced phenotypes that may be disseminated to her offspring. We examined whether a F0 male rat's prior exposure to an obesogenic high-fat diet (HFD) influences a female's attraction towards a male, subsequent mother-infant interactions and the development of defensive (emotional) responses in the F1 offspring. Females displayed less interest in the HFD exposed F0 males relative to control diet-exposed F0 males. Dams that reared F1 offspring in larger, semi-naturalistic housing provided more licking and grooming and active arched-back-nursing behavior. However, some of these effects interacted with paternal experience. F0 HFD and maternal rearing environment revealed sex-dependent, between group differences in F1 offspring wean weight, juvenile social interactions and anxiety-like behavior in adolescence. Our results show for the first time in mammals that male exposure to HFD may contribute to stable behavioral variation among females in courtship, maternal care, even when the females are not directly exposed to a HFD, and anxiety-like behavior in F1 offspring. Furthermore, when offspring were exposed to a predatory threat, hypothalamic Crf gene regulation was influenced by early housing. These results, together with our previous findings, suggest that paternal experience and maternal rearing conditions can influence maternal behavior and development of defensive responses of offspring.
Collapse
Affiliation(s)
- Austin C Korgan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Elizabeth O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Jillian L King
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Tara S Perrot
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada.
| |
Collapse
|
50
|
Sheriff MJ, Bell A, Boonstra R, Dantzer B, Lavergne SG, McGhee KE, MacLeod KJ, Winandy L, Zimmer C, Love OP. Integrating Ecological and Evolutionary Context in the Study of Maternal Stress. Integr Comp Biol 2018; 57:437-449. [PMID: 28957523 DOI: 10.1093/icb/icx105] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Maternal stress can prenatally influence offspring phenotypes and there are an increasing number of ecological studies that are bringing to bear biomedical findings to natural systems. This is resulting in a shift from the perspective that maternal stress is unanimously costly, to one in which maternal stress may be beneficial to offspring. However, this adaptive perspective is in its infancy with much progress to still be made in understanding the role of maternal stress in natural systems. Our aim is to emphasize the importance of the ecological and evolutionary context within which adaptive hypotheses of maternal stress can be evaluated. We present five primary research areas where we think future research can make substantial progress: (1) understanding maternal and offspring control mechanisms that modulate exposure between maternal stress and subsequent offspring phenotype response; (2) understanding the dynamic nature of the interaction between mothers and their environment; (3) integrating offspring phenotypic responses and measuring both maternal and offspring fitness outcomes under real-life (either free-living or semi-natural) conditions; (4) empirically testing these fitness outcomes across relevant spatial and temporal environmental contexts (both pre- and post-natal environments); (5) examining the role of maternal stress effects in human-altered environments-i.e., do they limit or enhance fitness. To make progress, it is critical to understand the role of maternal stress in an ecological context and to do that, we must integrate across physiology, behavior, genetics, and evolution.
Collapse
Affiliation(s)
- Michael J Sheriff
- Department of Ecosystem Science and Management, Huck Institute of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Alison Bell
- School of Integrative Biology, Program in Neuroscience, and Program in Ecology, Evolution and Conservation Biology, Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, IL 61821, USA
| | - Rudy Boonstra
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Ben Dantzer
- Department of Psychology, and Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sophia G Lavergne
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Katie E McGhee
- Department of Biology, the University of the South, Sewanee, TN 37383, USA
| | - Kirsty J MacLeod
- Department of Ecosystem Science and Management, Huck Institute of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.,Department of Biology, Mueller Laboratory, Pennsylvania State University, University Park, PA 16802, USA
| | - Laurane Winandy
- CNRS, Université Toulouse 3 Paul Sabatier, ENFA, UMR5174 (Laboratoire Évolution and Diversité Biologique), 31077 Toulouse, France.,CNRS, UMR5321, Station d'Ecologie Théorique et Expérimentale, 09200 Moulis, France
| | - Cedric Zimmer
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY 14853, USA
| | - Oliver P Love
- Department of Biological Sciences, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| |
Collapse
|