Jump to content

Epigenetics in psychology: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
ref reformatting
Owleye769 (talk | contribs)
Reverting a second hand information citation to re-insert Globe and Mail newspaper as sole reference as the content for the reference was extracted from this newspaper article, not a journal article.
Line 1: Line 1:
'''Epigenetics in psychology''' helps to explain how nurture shapes nature,<ref name="Powledge_2011">{{cite journal | author = Powledge T | title = Behavioral epigenetics: How nurture shapes nature | journal = BioScience | year = 2011 | volume = 61 | issue = 8 | pages = 588–592 | doi = 10.1525/bio.2011.61.8.4 }}</ref> where nature refers to biological [[heredity]]<ref name="isbn0-13-255770-3">{{cite book | author = Kail RV, Barnfield A| title = Children and Their Development, Second Canadian Edition with MyDevelopmentLab | edition = | language = | publisher = Pearson Education Canada | location = Toronto | year = 2011 | isbn = 0-13-255770-3 }}</ref> and nurture refers to virtually everything that occurs during the life-span (e.g., social-experience, diet and nutrition, and exposure to toxins).<ref name="Powledge_2011"/> '''Epigenetics in psychology''' provides a framework for understanding how the expression of [[gene]]s is influenced by experiences and the environment<ref name="Champagne">{{cite journal | author = Champagne FA, Mashoodh R | title = Genes in context: Gene-environment interplay and the origins of individual differences in behaviour | journal = Current Directions in Psychological Science | year = 2012 | volume = 18 | issue = 3 | pages = 127–131 | doi = 10.1111/j.1467-8721.2009.01622.x | url = http://champagnelab.psych.columbia.edu/docs/champ17.pdf }}</ref> to produce individual differences in [[personality]],<ref name="Bagot_2010"/> [[behaviour]],<ref name="Zhang_2010">{{cite journal | author = Zhang TY, Meaney MJ | title = Epigenetics and the environmental regulation of the genome and its function | journal = Annu Rev Psychol | volume = 61 | issue = | pages = 439–66, C1–3 | year = 2010 | pmid = 19958180 | doi = 10.1146/annurev.psych.60.110707.163625 }}</ref> [[cognition]]<ref name="Powledge_2011"/> and [[mental health]].<ref name="pmid18197504">{{cite journal | author = Stuffrein-Roberts S, Joyce PR, Kennedy MA | title = Role of epigenetics in mental disorders | journal = Aust N Z J Psychiatry | volume = 42 | issue = 2 | pages = 97–107 | year = 2008 | month = February | pmid = 18197504 | doi = 10.1080/00048670701787495 }}</ref><ref name="pmid18319075">{{cite journal | author = Mill J, Tang T, Kaminsky Z, Khare T, Yazdanpanah S, Bouchard L, Jia P, Assadzadeh A, Flanagan J, Schumacher A, Wang SC, Petronis A | title = Epigenomic profiling reveals DNA-methylation changes associated with major psychosis | journal = Am. J. Hum. Genet. | volume = 82 | issue = 3 | pages = 696–711 | year = 2008 | month = March | pmid = 18319075 | pmc = 2427301 | doi = 10.1016/j.ajhg.2008.01.008 | laysummary = http://www.theglobeandmail.com/news/technology/science/article26436.ece | laysource = The Globe and Mail }}</ref>
'''Epigenetics in psychology''' helps to explain how nurture shapes nature,<ref name="Powledge_2011">{{cite journal | author = Powledge T | title = Behavioral epigenetics: How nurture shapes nature | journal = BioScience | year = 2011 | volume = 61 | issue = 8 | pages = 588–592 | doi = 10.1525/bio.2011.61.8.4 }}</ref> where nature refers to biological [[heredity]]<ref name="isbn0-13-255770-3">{{cite book | author = Kail RV, Barnfield A| title = Children and Their Development, Second Canadian Edition with MyDevelopmentLab | edition = | language = | publisher = Pearson Education Canada | location = Toronto | year = 2011 | isbn = 0-13-255770-3 }}</ref> and nurture refers to virtually everything that occurs during the life-span (e.g., social-experience, diet and nutrition, and exposure to toxins).<ref name="Powledge_2011"/> '''Epigenetics in psychology''' provides a framework for understanding how the expression of [[gene]]s is influenced by experiences and the environment<ref name="Champagne">{{cite journal | author = Champagne FA, Mashoodh R | title = Genes in context: Gene-environment interplay and the origins of individual differences in behaviour | journal = Current Directions in Psychological Science | year = 2012 | volume = 18 | issue = 3 | pages = 127–131 | doi = 10.1111/j.1467-8721.2009.01622.x | url = http://champagnelab.psych.columbia.edu/docs/champ17.pdf }}</ref> to produce individual differences in [[personality]],<ref name="Bagot_2010"/> [[behaviour]],<ref name="Zhang_2010">{{cite journal | author = Zhang TY, Meaney MJ | title = Epigenetics and the environmental regulation of the genome and its function | journal = Annu Rev Psychol | volume = 61 | issue = | pages = 439–66, C1–3 | year = 2010 | pmid = 19958180 | doi = 10.1146/annurev.psych.60.110707.163625 }}</ref> [[cognition]]<ref name="Powledge_2011"/> and [[mental health]].<ref name="pmid18197504">{{cite journal | author = Stuffrein-Roberts S, Joyce PR, Kennedy MA | title = Role of epigenetics in mental disorders | journal = Aust N Z J Psychiatry | volume = 42 | issue = 2 | pages = 97–107 | year = 2008 | month = February | pmid = 18197504 | doi = 10.1080/00048670701787495 }}</ref><ref name=McIlroy>{{cite news|last=McIlroy|first=Anne|title=Scientists solve a psychiatric mystery|url=http://www.theglobeandmail.com/news/technology/science/article26436.ece|newspaper=The Globe and Mail|date=March 12, 2008}}</ref>


The subdiscipline of '''behavioral epigenetics''' is the field of study examining the role of [[epigenetic]]s in shaping [[animal]] (including [[human]]) [[behaviour]].<ref name="Miller_2010">{{cite journal | author = Miller G | title = Epigenetics. The seductive allure of behavioral epigenetics | journal = Science | volume = 329 | issue = 5987 | pages = 24–7 | year = 2010 | month = July | pmid = 20595592 | doi = 10.1126/science.329.5987.24 }}</ref> Epigenetic [[regulation of gene expression|gene regulation]] involves changes other than to the [[Nucleic acid sequence |sequence]] of [[DNA]] and includes changes to [[histone|histones]] (proteins around which DNA is wrapped) and [[DNA methylation]].<ref name="pmid11498570">{{cite journal | author = Pennisi E | title = Behind the scenes of gene expression | journal = Science | volume = 293 | issue = 5532 | pages = 1064–7 | year = 2001 | month = August | pmid = 11498570 | doi = 10.1126/science.293.5532.1064 }}</ref> These epigenetic changes can influence the growth of [[neuron|neurons]] in the developing brain<ref name="pmid20608952">{{cite journal | author = Juliandi B, Abematsu M, Nakashima K | title = Epigenetic regulation in neural stem cell differentiation | journal = Dev. Growth Differ. | volume = 52 | issue = 6 | pages = 493–504 | year = 2010 | month = August | pmid = 20608952 | doi = 10.1111/j.1440-169X.2010.01175.x }}</ref> as well as modify activity of the neurons in the adult brain.<ref name="pmid20975758">{{cite journal | author = Ma DK, Marchetto MC, Guo JU, Ming GL, Gage FH, Song H | title = Epigenetic choreographers of neurogenesis in the adult mammalian brain | journal = Nat. Neurosci. | volume = 13 | issue = 11 | pages = 1338–44 | year = 2010 | month = November | pmid = 20975758 | doi = 10.1038/nn.2672 }}</ref><ref name="pmid21395852">{{cite journal | author = Sun J, Sun J, Ming GL, Song H | title = Epigenetic regulation of neurogenesis in the adult mammalian brain | journal = Eur. J. Neurosci. | volume = 33 | issue = 6 | pages = 1087–93 | year = 2011 | month = March | pmid = 21395852 | pmc = 3076719 | doi = 10.1111/j.1460-9568.2011.07607.x }}</ref> Together, these epigenetic changes on neuron structure and function can have a marked influence on an organism's behavior.<ref name="Miller_2010"/>
The subdiscipline of '''behavioral epigenetics''' is the field of study examining the role of [[epigenetic]]s in shaping [[animal]] (including [[human]]) [[behaviour]].<ref name="Miller_2010">{{cite journal | author = Miller G | title = Epigenetics. The seductive allure of behavioral epigenetics | journal = Science | volume = 329 | issue = 5987 | pages = 24–7 | year = 2010 | month = July | pmid = 20595592 | doi = 10.1126/science.329.5987.24 }}</ref> Epigenetic [[regulation of gene expression|gene regulation]] involves changes other than to the [[Nucleic acid sequence |sequence]] of [[DNA]] and includes changes to [[histone|histones]] (proteins around which DNA is wrapped) and [[DNA methylation]].<ref name="pmid11498570">{{cite journal | author = Pennisi E | title = Behind the scenes of gene expression | journal = Science | volume = 293 | issue = 5532 | pages = 1064–7 | year = 2001 | month = August | pmid = 11498570 | doi = 10.1126/science.293.5532.1064 }}</ref> These epigenetic changes can influence the growth of [[neuron|neurons]] in the developing brain<ref name="pmid20608952">{{cite journal | author = Juliandi B, Abematsu M, Nakashima K | title = Epigenetic regulation in neural stem cell differentiation | journal = Dev. Growth Differ. | volume = 52 | issue = 6 | pages = 493–504 | year = 2010 | month = August | pmid = 20608952 | doi = 10.1111/j.1440-169X.2010.01175.x }}</ref> as well as modify activity of the neurons in the adult brain.<ref name="pmid20975758">{{cite journal | author = Ma DK, Marchetto MC, Guo JU, Ming GL, Gage FH, Song H | title = Epigenetic choreographers of neurogenesis in the adult mammalian brain | journal = Nat. Neurosci. | volume = 13 | issue = 11 | pages = 1338–44 | year = 2010 | month = November | pmid = 20975758 | doi = 10.1038/nn.2672 }}</ref><ref name="pmid21395852">{{cite journal | author = Sun J, Sun J, Ming GL, Song H | title = Epigenetic regulation of neurogenesis in the adult mammalian brain | journal = Eur. J. Neurosci. | volume = 33 | issue = 6 | pages = 1087–93 | year = 2011 | month = March | pmid = 21395852 | pmc = 3076719 | doi = 10.1111/j.1460-9568.2011.07607.x }}</ref> Together, these epigenetic changes on neuron structure and function can have a marked influence on an organism's behavior.<ref name="Miller_2010"/>
Line 32: Line 32:


=== Comparison with evolution ===
=== Comparison with evolution ===
According to [[On the Origin of Species|Darwin’s classic theory of evolution]], several generations are required for [[Adaptation|adaptation]] to occur. Also known as [[natural selection]], this process contributes to humans' (and other species') ability to survive and reproduce in their particular environments. Adaptation may occur through physiological, structural (anatomical) and/or behavioural changes. In the context of natural selection and psychology, more successful (i.e., better adapted) traits and behaviours are more likely to be passed down from an individual to subsequent generations.<ref name="isbn0-205-83256-3">{{cite book | author =Pinel J | title = Biopsychology (8th Edition) | edition = | language = | publisher = Prentice Hall | location = Englewood Cliffs, N.J | year = 2010 | isbn = 0-205-83256-3 }}</ref> Thus, most [[Evolutionary psychology|evolutionary psychologists]] agree that modern human traits and behaviours can be characterized as beneficial adaptations to the [[Evolutionary psychology#Environment of evolutionary adaptedness|environment]].<ref name="isbn0-13-255770-3">{{cite book | author = Kail RV, Barnfield A| title = Children and Their Development, Second Canadian Edition with MyDevelopmentLab | edition = | language = | publisher = Pearson Education Canada | location = Toronto | year = 2011 | isbn = 0-13-255770-3 }}</ref> However, other theorists argue that some traits are maladaptive.<ref name="Keller and Miller, 2006">{{cite journal | author = Keller MC, Miller G | title = Resolving the paradox of common, harmful, heritable mental disorders: which evolutionary genetic models work best? | journal = Behav Brain Sci | volume = 29 | issue = 4 | pages = 385–404; discussion 405–52 | year = 2006 | month = August | pmid = 17094843 | doi = 10.1017/S0140525X06009095 | url = }}</ref> Not to be confused with evolution, epigenetics provides a mechanism for immediate adaptations to ever-changing environments<ref name="pmid20535202">{{cite journal | author = Dulac C | title = Brain function and chromatin plasticity | journal = Nature | volume = 465 | issue = 7299 | pages = 728–35 | year = 2010 | month = June | pmid = 20535202 | pmc = 3075582 | doi = 10.1038/nature09231 }}</ref><ref name="Campbell_2011"/> throughout the human life-span.<ref name="Campbell_2011"/>
According to [[On the Origin of Species|Darwin’s classic theory of evolution]], several generations are required for [[Adaptation|adaptation]] to occur. Also known as [[natural selection]], this process contributes to humans' (and other species') ability to survive and reproduce in their particular environments. Adaptation may occur through physiological, structural (anatomical) and/or behavioural changes. In the context of natural selection and psychology, more successful (i.e., better adapted) traits and behaviours are more likely to be passed down from an individual to subsequent generations.<ref name="isbn0-205-83256-3">{{cite book | author =Pinel J | title = Biopsychology (8th Edition) | edition = | language = | publisher = Prentice Hall | location = Englewood Cliffs, N.J | year = 2010 | isbn = 0-205-83256-3 }}</ref> Thus, most [[Evolutionary psychology|evolutionary psychologists]] agree that modern human traits and behaviours can be characterized as beneficial adaptations to the [[Evolutionary psychology#Environment of evolutionary adaptedness|environment]].<ref name="isbn0-13-255770-3">{{cite book | author = Kail RV, Barnfield A| title = Children and Their Development, Second Canadian Edition with MyDevelopmentLab | edition = | language = | publisher = Pearson Education Canada | location = Toronto | year = 2011 | isbn = 0-13-255770-3 }}</ref> However, other theorists argue that some traits are maladaptive.<ref name="Keller and Miller, 2006">{{cite journal|last=Keller|first=M.C.|coauthors=Miller, G.|title=Resolving the paradox of common, harmful, heritable mental disorders: which evolutionary genetic models work best?|journal=The behavioral and brain science|year=2006|month=Aug|volume=29|issue=4|pages=405-452|pmid=17094843|url=http://www.ncbi.nlm.nih.gov/pubmed/17094843}}</ref> Not to be confused with evolution, epigenetics provides a mechanism for immediate adaptations to ever-changing environments<ref name="pmid20535202">{{cite journal | author = Dulac C | title = Brain function and chromatin plasticity | journal = Nature | volume = 465 | issue = 7299 | pages = 728–35 | year = 2010 | month = June | pmid = 20535202 | pmc = 3075582 | doi = 10.1038/nature09231 }}</ref><ref name="Campbell_2011"/> throughout the human life-span.<ref name="Campbell_2011"/>


Individual diversity, stemming from epigenetic differentiation, cannot be attributed to random mutations. Therefore, one view suggests that epigenetics is inconsistent with Darwinian theory of evolution’s concept of random mutations resulting in diversity.<ref name="Mostafavi-Abdolmaleky" /> Although [[Lamarckism]] is not appropriate to explain evolution, it might offer insight into epigenetic-related individuation. [[Evolutionary developmental biology]], for example, accounts for epigenetic and pseudo-Lamarckian mechanisms whereby environmentally induced variation is inherited.<ref name="kirschner and gerhart">{{cite book | title=The Plausibility of Life: Resolving Darwin's Dilemma | publisher=Yale University Press | author=Kirschner, M. W., & Gerhart, J. C. | year=2005 | location=New Haven, CT | isbn=978-0300108651}}</ref><ref name="Mostafavi-Abdolmaleky">{{cite book | editor = Bronner F, Helmtrud I | title = Epigenetic Aspects of Chronic Diseases | publisher = Springer | location = Berlin | year = 2011 | pages = | isbn = 1-84882-643-5 | author = Mostafavi-Abdolmaleky H, Glatt SJ, Tsuang MT | chapter = Epigenetics in Psychiatry | pages = 163–174 }}</ref> Another view maintains that epigenetics can be accomodated within a neo-Darwinian framework. In this context, inheritable epigenetic differences are viewed as part of long-term development, whereby the number of generations through which epigenetic modifications traverse is dependent on the duration of the influence on the genes.<ref name="pmid12547675">{{cite journal | author = Jablonka E, Lamb MJ | title = The changing concept of epigenetics | journal = Ann. N. Y. Acad. Sci. | volume = 981 | issue = | pages = 82–96 | year = 2002 | month = December | pmid = 12547675 | doi = 10.1111/j.1749-6632.2002.tb04913.x }}</ref>
Individual diversity, stemming from epigenetic differentiation, cannot be attributed to random mutations. Therefore, one view suggests that epigenetics is inconsistent with Darwinian theory of evolution’s concept of random mutations resulting in diversity.<ref name="Mostafavi-Abdolmaleky" /> Although [[Lamarckism]] is not appropriate to explain evolution, it might offer insight into epigenetic-related individuation. [[Evolutionary developmental biology]], for example, accounts for epigenetic and pseudo-Lamarckian mechanisms whereby environmentally induced variation is inherited.<ref name="kirschner and gerhart">{{cite book | title=The Plausibility of Life: Resolving Darwin's Dilemma | publisher=Yale University Press | author=Kirschner, M. W., & Gerhart, J. C. | year=2005 | location=New Haven, CT | isbn=978-0300108651}}</ref><ref name="Mostafavi-Abdolmaleky">{{cite book | editor = Bronner F, Helmtrud I | title = Epigenetic Aspects of Chronic Diseases | publisher = Springer | location = Berlin | year = 2011 | pages = | isbn = 1-84882-643-5 | author = Mostafavi-Abdolmaleky H, Glatt SJ, Tsuang MT | chapter = Epigenetics in Psychiatry | pages = 163–174 }}</ref> Another view maintains that epigenetics can be accomodated within a neo-Darwinian framework. In this context, inheritable epigenetic differences are viewed as part of long-term development, whereby the number of generations through which epigenetic modifications traverse is dependent on the duration of the influence on the genes.<ref name="pmid12547675">{{cite journal | author = Jablonka E, Lamb MJ | title = The changing concept of epigenetics | journal = Ann. N. Y. Acad. Sci. | volume = 981 | issue = | pages = 82–96 | year = 2002 | month = December | pmid = 12547675 | doi = 10.1111/j.1749-6632.2002.tb04913.x }}</ref>
Line 42: Line 42:
Epigenetic differences have been linked to differences in risk-taking and reactions to stress. Specifically, epigenetic differentiation in [[Twin#Monozygotic ("identical") twins| monozygotic twins]] has been shown to play a role in individual differences in:
Epigenetic differences have been linked to differences in risk-taking and reactions to stress. Specifically, epigenetic differentiation in [[Twin#Monozygotic ("identical") twins| monozygotic twins]] has been shown to play a role in individual differences in:
* risk-taking that is characterized by impulsive or disinhibited decision making.<ref name="Kaminsky_2008">{{cite journal | author = Kaminsky Z, Petronis A, Wang SC, Levine B, Ghaffar O, Floden D, Feinstein A | title = Epigenetics of personality traits: an illustrative study of identical twins discordant for risk-taking behavior | journal = Twin Res Hum Genet | volume = 11 | issue = 1 | pages = 1–11 | year = 2008 | month = February | pmid = 18251670 | doi = 10.1375/twin.11.1.1 | url = }}</ref>
* risk-taking that is characterized by impulsive or disinhibited decision making.<ref name="Kaminsky_2008">{{cite journal | author = Kaminsky Z, Petronis A, Wang SC, Levine B, Ghaffar O, Floden D, Feinstein A | title = Epigenetics of personality traits: an illustrative study of identical twins discordant for risk-taking behavior | journal = Twin Res Hum Genet | volume = 11 | issue = 1 | pages = 1–11 | year = 2008 | month = February | pmid = 18251670 | doi = 10.1375/twin.11.1.1 | url = }}</ref>
* the levels of [[anxiety]] (a state of apprehension or fear<ref name="pmid10235121">{{cite journal | author = Bakish D | title = The patient with comorbid depression and anxiety: the unmet need | journal = J Clin Psychiatry | volume = 60 Suppl 6 | issue = | pages = 20–4 | year = 1999 | pmid = 10235121 | doi = }}</ref> that arises during stress) and somatic complaints<ref name="Kaminsky_2008"/> (physical ailments, symptoms and/or pain in the absence of underlying medical conditions – usually associated with anxiety).<ref name="isbn0-553-59226-2"/> Anxiety and somatic complaints have been associated with the broad personality trait of neuroticism.<ref name="Costa and McRae, 1987">{{cite journal | author = Costa PT, McCrae RR | title = Neuroticism, somatic complaints, and disease: is the bark worse than the bite? | journal = J Pers | volume = 55 | issue = 2 | pages = 299–316 | year = 1987 | month = June | pmid = 3612472 | doi = }}</ref>
* the levels of [[anxiety]] (a state of apprehension or fear<ref name="pmid10235121">{{cite journal | author = Bakish D | title = The patient with comorbid depression and anxiety: the unmet need | journal = J Clin Psychiatry | volume = 60 Suppl 6 | issue = | pages = 20–4 | year = 1999 | pmid = 10235121 | doi = }}</ref> that arises during stress) and somatic complaints<ref name="Kaminsky_2008"/> (physical ailments, symptoms and/or pain in the absence of underlying medical conditions – usually associated with anxiety).<ref name="isbn0-553-59226-2"/> Anxiety and somatic complaints have been associated with the broad personality trait of neuroticism.<ref name="Costa and McRae, 1987">{{cite journal|last=Costa|first=P.T.|coauthors=McCrae, R.R.|title=Neuroticism, somatic complaints, and disease: is the bark worse than the bite?|journal=Journal of Personality|year=1987|month=Jun|volume=55|issue=2|pages=299-316|pmid=3612472|url=http://www.ncbi.nlm.nih.gov/pubmed/3612472}}</ref>


Epigenetic variations, associated with risk-taking behaviours and anxiety, have been linked to individual differences in lifestyle and career choices. In one study, each member of a pair of twins had chosen two very different life paths. One twin, who displayed high-risk behaviours (like gambling), chose a dangerous and high-sensation career as a war journalist. Despite the dangerous profession, this twin showed no elevated anxiety levels when confronted with stress. This twin married late in life (to an individual who also chose a dangerous profession), had no children, and consumed more alcohol than is considered medically healthy. In contrast, the other twin was considered to be a polar opposite. The second twin was prone to anxiety and developing somatic complaints when faced with stress.<ref name="Kaminsky_2008"/> Instead of displaying risk-taking behaviours, the second twin displayed risk-averse behaviours. The second twin chose a safe and predictable career as a manager in a law firm, married a lawyer, had children at a young age and had only traveled out of country once (to another English-speaking country).<ref name="Kaminsky_2008"/>
Epigenetic variations, associated with risk-taking behaviours and anxiety, have been linked to individual differences in lifestyle and career choices. In one study, each member of a pair of twins had chosen two very different life paths. One twin, who displayed high-risk behaviours (like gambling), chose a dangerous and high-sensation career as a war journalist. Despite the dangerous profession, this twin showed no elevated anxiety levels when confronted with stress. This twin married late in life (to an individual who also chose a dangerous profession), had no children, and consumed more alcohol than is considered medically healthy. In contrast, the other twin was considered to be a polar opposite. The second twin was prone to anxiety and developing somatic complaints when faced with stress.<ref name="Kaminsky_2008"/> Instead of displaying risk-taking behaviours, the second twin displayed risk-averse behaviours. The second twin chose a safe and predictable career as a manager in a law firm, married a lawyer, had children at a young age and had only traveled out of country once (to another English-speaking country).<ref name="Kaminsky_2008"/>
Line 66: Line 66:
Evidence shows that the environment exerts an influence on epigenetic changes related to [[cognition]], in terms of learning and memory.<ref name="Fischer and Tsai">{{cite journal | author = Fischer A, Sananbenesi F, Wang X, Dobbin M, Tsai LH | title = Recovery of learning and memory is associated with chromatin remodelling | journal = Nature | volume = 447 | issue = 7141 | pages = 178–82 | year = 2007 | month = May | pmid = 17468743 | doi = 10.1038/nature05772 }}</ref><ref name="Miller_2010" /> Animal studies using mice with extensive [[neurodegeneration]] and [[Synapse|synaptic]] loss in the [[forebrain]] indicated that environmental enrichment helped to restore [[spatial learning]] and [[Long-term memory|long-term memories]]. Specifically, mice with brain damage were subjected to a fear paradigm that consisted of a single exposure to a conditioning context followed by an electric foot shock that elicited a freezing (i.e., staying still) response. After training, the mice were allowed to rest for four weeks in order to develop long-term memories. Mice were then housed in regular animal cages or in an enriched environment for ten weeks. The enriched environment consisted of cages with running wheels, play tunnels, and climbing apparatuses. When re-exposed to the conditioned context, mice housed in regular cages displayed impaired freezing responses. Conversely, the environmentally-enriched mice displayed the freezing behaviour, indicating a recovery of long-term memories.<ref name="Fischer and Tsai" />
Evidence shows that the environment exerts an influence on epigenetic changes related to [[cognition]], in terms of learning and memory.<ref name="Fischer and Tsai">{{cite journal | author = Fischer A, Sananbenesi F, Wang X, Dobbin M, Tsai LH | title = Recovery of learning and memory is associated with chromatin remodelling | journal = Nature | volume = 447 | issue = 7141 | pages = 178–82 | year = 2007 | month = May | pmid = 17468743 | doi = 10.1038/nature05772 }}</ref><ref name="Miller_2010" /> Animal studies using mice with extensive [[neurodegeneration]] and [[Synapse|synaptic]] loss in the [[forebrain]] indicated that environmental enrichment helped to restore [[spatial learning]] and [[Long-term memory|long-term memories]]. Specifically, mice with brain damage were subjected to a fear paradigm that consisted of a single exposure to a conditioning context followed by an electric foot shock that elicited a freezing (i.e., staying still) response. After training, the mice were allowed to rest for four weeks in order to develop long-term memories. Mice were then housed in regular animal cages or in an enriched environment for ten weeks. The enriched environment consisted of cages with running wheels, play tunnels, and climbing apparatuses. When re-exposed to the conditioned context, mice housed in regular cages displayed impaired freezing responses. Conversely, the environmentally-enriched mice displayed the freezing behaviour, indicating a recovery of long-term memories.<ref name="Fischer and Tsai" />


Other memory-related research has yielded similar results. For example, when placed in a tub of water, brain-damaged mice learned to swim to a platform to escape from the water in the [[Morris water navigation task]]. After training, the brain-damaged mice that had resided in regular cages could not swim directly to the platform. However, brain-damaged mice living in an enriched environment were still able to remember where the platform was located by swimming to it – indicating a recovery of long-term memories. The environmental enrichment induced epigenetic changes in the hippocampus and cortex regions (via [[Histone acetylation and deacetylation|histone acetylation]]) of the brain. Injection of [[Histone deacetylase inhibitor|histone deacetylase inhibitor]] was shown to increase hippocampal acetylation and mimic the effects of environmental enrichment in brain-damaged rats. This was apparent through reinstating learning and a reduction in long-term memory loss related to the freezing-fear response and performance on the [[Morris water navigation task|Morris water maze]]. These findings are consistent with human studies that have noted fluctuations in cognition, including temporary periods of clear memories, in individuals with [[dementia]].<ref name="Fischer and Tsai" /> Research has also linked learning and long-term memory formation to reversible epigenetic changes in the hippocampus and cortex in animals with normal-functioning, non-damaged brains.<ref name="Miller_2010" /><ref name=Gupat>{{cite journal | author = Gupta S, Kim SY, Artis S, Molfese DL, Schumacher A, Sweatt JD, Paylor RE, Lubin FD | title = Histone methylation regulates memory formation | journal = J. Neurosci. | volume = 30 | issue = 10 | pages = 3589–99 | year = 2010 | month = March | pmid = 20219993 | pmc = 2859898 | doi = 10.1523/JNEUROSCI.3732-09.2010 }}</ref> In human studies, post-mortem brains from Alzheimer patients show increased histone de-acetylase levels.<ref name="Graff_2012">{{cite journal | author = Gräff J, Rei D, Guan JS, Wang WY, Seo J, Hennig KM, Nieland TJ, Fass DM, Kao PF, Kahn M, Su SC, Samiei A, Joseph N, Haggarty SJ, Delalle I, Tsai LH | title = An epigenetic blockade of cognitive functions in the neurodegenerating brain | journal = Nature | volume = 483 | issue = 7388 | pages = 222–6 | year = 2012 | month = March | pmid = 22388814 | doi = 10.1038/nature10849 }}</ref>
Other memory-related research has yielded similar results. For example, when placed in a tub of water, brain-damaged mice learned to swim to a platform to escape from the water in the [[Morris water navigation task]]. After training, the brain-damaged mice that had resided in regular cages could not swim directly to the platform. However, brain-damaged mice living in an enriched environment were still able to remember where the platform was located by swimming to it – indicating a recovery of long-term memories. The environmental enrichment induced epigenetic changes in the hippocampus and cortex regions (via [[Histone acetylation and deacetylation|histone acetylation]]) of the brain. Injection of [[Histone deacetylase inhibitor|histone deacetylase inhibitor]] was shown to increase hippocampal acetylation and mimic the effects of environmental enrichment in brain-damaged rats. This was apparent through reinstating learning and a reduction in long-term memory loss related to the freezing-fear response and performance on the [[Morris water navigation task|Morris water maze]]. These findings are consistent with human studies that have noted fluctuations in cognition, including temporary periods of clear memories, in individuals with [[dementia]].<ref name="Fischer and Tsai" /> Research has also linked learning and long-term memory formation to reversible epigenetic changes in the hippocampus and cortex in animals with normal-functioning, non-damaged brains.<ref name="Miller_2010" /><ref name=Gupat>{{cite journal | author = Gupta S, Kim SY, Artis S, Molfese DL, Schumacher A, Sweatt JD, Paylor RE, Lubin FD | title = Histone methylation regulates memory formation | journal = J. Neurosci. | volume = 30 | issue = 10 | pages = 3589–99 | year = 2010 | month = March | pmid = 20219993 | pmc = 2859898 | doi = 10.1523/JNEUROSCI.3732-09.2010 }}</ref> In human studies, post-mortem brains from Alzheimer patients show increased histone de-acetylase levels.<ref name="Graff et al, 2012">{{cite journal|last=Graff|first=J.|coauthors=Rei, D., Guan, H., Wang, W., Seo, J.,Hennig, K., Nieland, T., Fass, D., Kao, P., Kahn, M., Su, S., Samiei, A., Joseph, N., Haggarty, S., Delalle, I., Tsai, L.|title=An epigenetic blockade of cognitive functions in the neurodegenerating brain|journal=Nature|year=2012|volume=483|issue=7388|doi=10.1038/nature10849|url=http://www.nature.com/nature/journal/v483/n7388/abs/nature10849.html}}</ref>


Animal studies have also provided evidence for epigenetic, age-associated memory decline.<ref name="Miller_2010" /> Older mice show increased epigenetic changes (i.e., decreased histone acetylation and reduced gene expression) in the hippocampus that is related to poorer learning and memory in the Morris water maze. Specifically, older mice spend more time to find the platform and escape from the water. Agents used to increase histone acetylation (i.e., reverse the age-related epigenetic effects) lead to improved performance in the older rats.<ref name=Peleg>{{cite journal | author = Peleg S, Sananbenesi F, Zovoilis A, Burkhardt S, Bahari-Javan S, Agis-Balboa RC, Cota P, Wittnam JL, Gogol-Doering A, Opitz L, Salinas-Riester G, Dettenhofer M, Kang H, Farinelli L, Chen W, Fischer A | title = Altered histone acetylation is associated with age-dependent memory impairment in mice | journal = Science | volume = 328 | issue = 5979 | pages = 753–6 | year = 2010 | month = May | pmid = 20448184 | doi = 10.1126/science.1186088 }}</ref>
Animal studies have also provided evidence for epigenetic, age-associated memory decline.<ref name="Miller_2010" /> Older mice show increased epigenetic changes (i.e., decreased histone acetylation and reduced gene expression) in the hippocampus that is related to poorer learning and memory in the Morris water maze. Specifically, older mice spend more time to find the platform and escape from the water. Agents used to increase histone acetylation (i.e., reverse the age-related epigenetic effects) lead to improved performance in the older rats.<ref name=Peleg>{{cite journal | author = Peleg S, Sananbenesi F, Zovoilis A, Burkhardt S, Bahari-Javan S, Agis-Balboa RC, Cota P, Wittnam JL, Gogol-Doering A, Opitz L, Salinas-Riester G, Dettenhofer M, Kang H, Farinelli L, Chen W, Fischer A | title = Altered histone acetylation is associated with age-dependent memory impairment in mice | journal = Science | volume = 328 | issue = 5979 | pages = 753–6 | year = 2010 | month = May | pmid = 20448184 | doi = 10.1126/science.1186088 }}</ref>
Line 136: Line 136:
* {{cite journal | author = McGowan PO, Meaney MJ, Szyf M | title = Diet and the epigenetic (re)programming of phenotypic differences in behavior | journal = Brain Res. | volume = 1237 | issue = | pages = 12–24 | year = 2008 | month = October | pmid = 18694740 | pmc = 2951010 | doi = 10.1016/j.brainres.2008.07.074 }}
* {{cite journal | author = McGowan PO, Meaney MJ, Szyf M | title = Diet and the epigenetic (re)programming of phenotypic differences in behavior | journal = Brain Res. | volume = 1237 | issue = | pages = 12–24 | year = 2008 | month = October | pmid = 18694740 | pmc = 2951010 | doi = 10.1016/j.brainres.2008.07.074 }}
* {{cite journal | author = Szyf M, Weaver I, Meaney M | title = Maternal care, the epigenome and phenotypic differences in behavior | journal = Reprod. Toxicol. | volume = 24 | issue = 1 | pages = 9–19 | year = 2007 | month = July | pmid = 17561370 | doi = 10.1016/j.reprotox.2007.05.001 }}
* {{cite journal | author = Szyf M, Weaver I, Meaney M | title = Maternal care, the epigenome and phenotypic differences in behavior | journal = Reprod. Toxicol. | volume = 24 | issue = 1 | pages = 9–19 | year = 2007 | month = July | pmid = 17561370 | doi = 10.1016/j.reprotox.2007.05.001 }}
* Harmon K (2010-08-26). [http://www.scientificamerican.com/article.cfm?id=first-ant-genomes-epigenetics First Ant Genomes Promise Insight into Epigenetics and Longevity]. ''Scientific American Magazine.''
* {{cite journal | author = Bonasio R, Zhang G, Ye C, Mutti NS, Fang X, Qin N, Donahue G, Yang P, Li Q, Li C, Zhang P, Huang Z, Berger SL, Reinberg D, Wang J, Liebig J | title = Genomic comparison of the ants Camponotus floridanus and Harpegnathos saltator | journal = Science | volume = 329 | issue = 5995 | pages = 1068–71 | year = 2010 | month = August | pmid = 20798317 | doi = 10.1126/science.1192428 | laysource = Scientific American Magazine | laysummary = http://www.scientificamerican.com/article.cfm?id=first-ant-genomes-epigenetics }}
{{refend}}
{{refend}}



Revision as of 02:25, 5 April 2012

Epigenetics in psychology helps to explain how nurture shapes nature,[1] where nature refers to biological heredity[2] and nurture refers to virtually everything that occurs during the life-span (e.g., social-experience, diet and nutrition, and exposure to toxins).[1] Epigenetics in psychology provides a framework for understanding how the expression of genes is influenced by experiences and the environment[3] to produce individual differences in personality,[4] behaviour,[5] cognition[1] and mental health.[6][7]

The subdiscipline of behavioral epigenetics is the field of study examining the role of epigenetics in shaping animal (including human) behaviour.[8] Epigenetic gene regulation involves changes other than to the sequence of DNA and includes changes to histones (proteins around which DNA is wrapped) and DNA methylation.[9] These epigenetic changes can influence the growth of neurons in the developing brain[10] as well as modify activity of the neurons in the adult brain.[11][12] Together, these epigenetic changes on neuron structure and function can have a marked influence on an organism's behavior.[8]

Discovery

The first documented example of epigenetics affecting behavior was provided by Meaney and Szyf. While working at McGill University in Montréal in 2004, they discovered that the type of mothering a rat receives in infancy determines how that rat responds to stress later in life. Rat pups that receive a less nurturing upbringing are more sensitive to stress throughout their life span. This stress sensitivity is linked to a down-regulation in the expression of the glucocorticoid receptor in the brain. In turn, this down-regulation was found to be a consequence of the extent of methylation in the promoter region of the glucocorticoid receptor gene.[13][8] This pioneering work in rodents has been hard to replicate in humans because of a general lack of human brain tissue for measurement of epigenetic changes.[8] The first study that has directly linked epigenetic changes in human brain tissue to behavior involved post-mortem brains of people who committed suicide, half of whom had been abused as children. Those who had been abused had a lower expression of glucocorticoid receptor due to increased methylation in the promoter region of the glucocorticoid receptor gene. These observations in humans closely parallel the earlier rat studies.[14][8]

Background

History

For more details on this topic, see Epigenetic etymology and definitions.

British biologist Conrad Waddington is credited with developing the term epigenetics in 1947.[15] Epi is a Greek term meaning upon or above. Hence, epigenetics reflects the effects that take place upon, above[4] or in addition to genetics.[3][16] This original definition implied that something aside from genes was involved in defining the phenotype. When Waddington created the term epigenetics, little was known about the expression of genes. Waddington and others eventually realized that development involved networks of interactions between genes. By the 1990s, Hall wrote that epigenetics involves genetic and non-genetic factors that affect gene expression. Hall further noted that the code of heredity includes an extra layer in addition to DNA.[16] By the 2000s, epigenetics was used to describe how experiences shape heritable genetic expression.[17] Psychosocial and environmental factors have been shown to alter these epigenetic mechanisms which, in turn, influence normal and abnormal psychology.[18] The current definition still maintains its continuity to Waddington’s, as epigenetics is used to explain how genes and the environment work in tandem.[16] However, epigenetic changes do not alter or mutate the DNA. Instead, epigenetic mechanisms affect DNA by regulating the expression of genes.[19] Therefore, due to their regulatory function, epigenetic mechanisms are still considered to occur upon or above the DNA[4] without actually altering it.[15]

Epigenetic mechanisms

Modifications of the epigenome do not alter DNA.

Two of the best understood mechanisms of epigenetic modification include DNA methylation[20] and histone modification.[21] DNA methylation, the best understood epigenetic modification,[17] results in the inability of genetic information to be read from DNA.[22] In other words, epigenetic modification can "turn off" a gene.[23] This mechanism of turning off genes is reversible.[17][23]

Genes can be likened to books, categorized and arranged, in a library. The books contain enormous amounts of accessible information that can be utilized in an infinite number of ways. Therefore, books are similar to DNA in that both are “waiting to be read.” [3]: 128  In a library, many factors may contribute to whether a book will or will not be read. If a book is difficult to reach or being blocked by a piece of furniture, the book may not be read. Like an unread gene, an obstructed book is not altered or discarded. Both are still present, but if they are not accessible, neither the gene nor the book can be read.[3] In this manner, epigenetics can silence a gene.[24]

Genes contained within DNA constitute a complete instruction set. Epigenetics, in part, controls if and when these individual genes are expressed.[9] The epigenome, a set of chemical markers attached to the DNA, is a secondary code that acts like a referee in activating and de-activating genes.[25] Hence, epigenetics has a strong influence on the development of an organism and can alter the expression of individual traits.[9]

Epigenetics across the life-span

Epigenetic changes occur not only in the developing fetus, but also in individuals throughout the human life-span.[17] Increased levels of epigenetic changes have been found in older monozygotic ("identical") twins than in younger twins. Since identical twins become more epigenetically dissimilar as they age, epigenetic changes occur cumulatively over the life-span.[26] Epigenetic changes, affecting the regulation of gene expression, are also reversible.[15]

Inheritance

Epigenetic modifications can also be inherited from one generation to the next.[27] Specifically, the effects of epigenetics may be transmitted from parents to children through meiosis[28][17] and mitosis[29][17] - processes that pass genetic information from the father into the sperm and from the mother into the egg.[30] Hence, subsequent generations may be affected by the epigenetic changes that took place in the parents. [27] Epigenetic modifications in offspring, resulting from parental environments, may also be passed down from generation to generation.[31] Epigenetic research counters the long-standing notion that children are born with an uncontaminated genetic slate.[32]

Comparison with evolution

According to Darwin’s classic theory of evolution, several generations are required for adaptation to occur. Also known as natural selection, this process contributes to humans' (and other species') ability to survive and reproduce in their particular environments. Adaptation may occur through physiological, structural (anatomical) and/or behavioural changes. In the context of natural selection and psychology, more successful (i.e., better adapted) traits and behaviours are more likely to be passed down from an individual to subsequent generations.[33] Thus, most evolutionary psychologists agree that modern human traits and behaviours can be characterized as beneficial adaptations to the environment.[2] However, other theorists argue that some traits are maladaptive.[34] Not to be confused with evolution, epigenetics provides a mechanism for immediate adaptations to ever-changing environments[35][17] throughout the human life-span.[17]

Individual diversity, stemming from epigenetic differentiation, cannot be attributed to random mutations. Therefore, one view suggests that epigenetics is inconsistent with Darwinian theory of evolution’s concept of random mutations resulting in diversity.[36] Although Lamarckism is not appropriate to explain evolution, it might offer insight into epigenetic-related individuation. Evolutionary developmental biology, for example, accounts for epigenetic and pseudo-Lamarckian mechanisms whereby environmentally induced variation is inherited.[37][36] Another view maintains that epigenetics can be accomodated within a neo-Darwinian framework. In this context, inheritable epigenetic differences are viewed as part of long-term development, whereby the number of generations through which epigenetic modifications traverse is dependent on the duration of the influence on the genes.[38]

Personality

Anxiety and risk taking

Monozygotic Twins are identical twins. Twin studies help to reveal epigenetic differences related to various aspects of psychology.

Epigenetic differences have been linked to differences in risk-taking and reactions to stress. Specifically, epigenetic differentiation in monozygotic twins has been shown to play a role in individual differences in:

  • risk-taking that is characterized by impulsive or disinhibited decision making.[39]
  • the levels of anxiety (a state of apprehension or fear[40] that arises during stress) and somatic complaints[39] (physical ailments, symptoms and/or pain in the absence of underlying medical conditions – usually associated with anxiety).[19] Anxiety and somatic complaints have been associated with the broad personality trait of neuroticism.[41]

Epigenetic variations, associated with risk-taking behaviours and anxiety, have been linked to individual differences in lifestyle and career choices. In one study, each member of a pair of twins had chosen two very different life paths. One twin, who displayed high-risk behaviours (like gambling), chose a dangerous and high-sensation career as a war journalist. Despite the dangerous profession, this twin showed no elevated anxiety levels when confronted with stress. This twin married late in life (to an individual who also chose a dangerous profession), had no children, and consumed more alcohol than is considered medically healthy. In contrast, the other twin was considered to be a polar opposite. The second twin was prone to anxiety and developing somatic complaints when faced with stress.[39] Instead of displaying risk-taking behaviours, the second twin displayed risk-averse behaviours. The second twin chose a safe and predictable career as a manager in a law firm, married a lawyer, had children at a young age and had only traveled out of country once (to another English-speaking country).[39]

In this twin study, epigenetic differences in DNA methylation of the CpG islands proximal to the DLX1 gene have been implicated as affecting risk-taking behaviours and coping with stress in terms of anxiety.[39] DLX1 exerts its effects by helping to regulate the production of the neurotransmitter GABA (gamma-aminobutyric acid), and the hormone neuropeptide Y. GABA decreases stress[42] by acting on what is often referred to as the stress centre of the brain - the hypothalamic-pituitary-adrenal axis, or HPA axis.[43] Conversely, neuropeptide Y negates the effect of GABA on the HPA axis and results in an increased stress response.[44] In general, activation of the HPA axis, as part of the stress response, has been well-established in resulting in increased anxiety[45] and depression.[46] The epigenetic differences pertaining to GABA and neuropeptide Y, and their associated physiological HPA processes, have been posited to help explain individual differences in risk-taking behaviour and anxiety. In turn, this helps to explain why one twin functioned without anxiety under extremely dangerous situations.[39]

Despite the associations between epigenetic markers for individual differences in personality traits and behaviours that might influence occupational choices, epigenetics cannot, on their own, predict complex decision-making processes like career choices. Such complex decisions transcend genetic predication.[39]

Stress

The hypothalamic pituitary adrenal axis is involved in the human stress response.

The early parental-caregiving environment has long been a central focus of human psychological development. Early abuse and neglect can lead to a wide range of cognitive and emotional impairment. Animal studies reveal that the influence of early maternal care also affects genetic expression.[18] In their review of a series of studies, Szyf, McGowan and Meaney[47] indicated that early maternal care affected offspring's reactivity to stress. Specifically, the effects of maternal care, in terms of the parental licking-grooming of offspring, were examined. High licking-grooming resulted in a positive, long-term effect on the reactivity of the HPA axis when compared to pups who received low levels of licking-grooming. This was evidenced by decreased adrenocorticotropic hormone (ACTH) and corticosterone in response to stress in offspring. The effects of parental care on the HPA and ACTH resulted from epigenetic changes, whereby high licking-grooming led to decreased DNA methylation. This allowed for increased access to the hippocampal glucocorticoid receptor genes in the offspring. Increased expression of hippocampal glucocorticoid receptors causes the hippocampus to release less ACTH-releasing hormone, which acts on the pituitary gland to release less ACTH, which in turn acts on the adrenal glands to release less cortisol. In contrast, less parental care (low licking-grooming) ultimately results in increased cortisol release.[18] The release of increased cortisol, via the HPA axis, as a reaction to psychological stress is well-established.[30][48] Therefore, pups that received less licking-grooming were more prone to react to stress.[18] Since humans also show the same cortisol response to stress and heritable cortisol levels,[49] the animal models have implications for humans.[47]

Research has also demonstrated that the maternal genotype did not determine stress reactions in offspring. Instead, the early parental care environment affected genetic expression to influence how offspring react to stress. When rat offspring were removed from their low licking-grooming mothers and placed with high licking-grooming mothers, the epigenetic changes related to an increased stress response were reversed. The reverse was observed when pups were removed from their high licking-grooming parents and placed with low licking-grooming mothers. This research provides evidence for an underlying epigenetic mechanism, as the stress response in offspring was determined by the foster mother, not the biological, genetic mother.[18]

Research has also demonstrated the reversibility of epigenetic changes related to maternal care in mice. When adult offspring of low licking-grooming mothers were injected with an agent that decreased DNA methylation to match levels of adult pups of high licking-grooming mothers, their responsivity to stress decreased to match the levels seen in the offspring of high licking-grooming mothers.[13] Conversely, when adult offspring of high licking-grooming mothers were injected with an agent to increase DNA methylation, their response to stress increased to match adult offspring of low licking-grooming parents.[50]

Stable epigenetic variations in parental care can be passed down from one generation to the next, from mother to female offspring. Female offspring who received increased parental care (i.e., high licking-grooming) became mothers who engaged in high licking-grooming. Conversely, offspring who received less licking-grooming became mothers who engaged in less licking-grooming. Research has also provided the transgenerational epigenetic link related to maternal care. To this end, offspring of mothers with high licking-grooming behaviour evidenced DNA methylation-related increased expression of an estrogen receptor gene in the medial preoptic region of the hypothalamus – an area of the brain known to be linked to maternal nurturing and care.[51]

In humans, the relationship between prenatal exposure to maternal mood and genetic expression resulting in increased reactivity to stress in offspring has been confirmed. Three groups of infants were examined: those born to mothers medicated for depression with serotonin reuptake inhibitors; those born to mothers not being treated for depression; and those born to non-depressed mothers. Prenatal exposure to depressed/anxious mood was associated with increased epigenetic changes in terms of increased DNA methylation at the glucorticoid receptor gene. In turn, this was related to increased HPA axis stress reactivity - as measured by increased salivary cortisol - in three month old infants.[52] Research has well-established that increased cortisol levels are associated with increased stress.[48] The findings were independent of whether the mothers were being pharmaceutically treated for depression.[52]

Cognition

Learning and memory

Evidence shows that the environment exerts an influence on epigenetic changes related to cognition, in terms of learning and memory.[53][8] Animal studies using mice with extensive neurodegeneration and synaptic loss in the forebrain indicated that environmental enrichment helped to restore spatial learning and long-term memories. Specifically, mice with brain damage were subjected to a fear paradigm that consisted of a single exposure to a conditioning context followed by an electric foot shock that elicited a freezing (i.e., staying still) response. After training, the mice were allowed to rest for four weeks in order to develop long-term memories. Mice were then housed in regular animal cages or in an enriched environment for ten weeks. The enriched environment consisted of cages with running wheels, play tunnels, and climbing apparatuses. When re-exposed to the conditioned context, mice housed in regular cages displayed impaired freezing responses. Conversely, the environmentally-enriched mice displayed the freezing behaviour, indicating a recovery of long-term memories.[53]

Other memory-related research has yielded similar results. For example, when placed in a tub of water, brain-damaged mice learned to swim to a platform to escape from the water in the Morris water navigation task. After training, the brain-damaged mice that had resided in regular cages could not swim directly to the platform. However, brain-damaged mice living in an enriched environment were still able to remember where the platform was located by swimming to it – indicating a recovery of long-term memories. The environmental enrichment induced epigenetic changes in the hippocampus and cortex regions (via histone acetylation) of the brain. Injection of histone deacetylase inhibitor was shown to increase hippocampal acetylation and mimic the effects of environmental enrichment in brain-damaged rats. This was apparent through reinstating learning and a reduction in long-term memory loss related to the freezing-fear response and performance on the Morris water maze. These findings are consistent with human studies that have noted fluctuations in cognition, including temporary periods of clear memories, in individuals with dementia.[53] Research has also linked learning and long-term memory formation to reversible epigenetic changes in the hippocampus and cortex in animals with normal-functioning, non-damaged brains.[8][54] In human studies, post-mortem brains from Alzheimer patients show increased histone de-acetylase levels.[55]

Animal studies have also provided evidence for epigenetic, age-associated memory decline.[8] Older mice show increased epigenetic changes (i.e., decreased histone acetylation and reduced gene expression) in the hippocampus that is related to poorer learning and memory in the Morris water maze. Specifically, older mice spend more time to find the platform and escape from the water. Agents used to increase histone acetylation (i.e., reverse the age-related epigenetic effects) lead to improved performance in the older rats.[56]

One review also noted the role of DNA methylation in memory formation and storage, but the precise mechanisms involving neuronal function, memory, and methylation reversal remain unclear.[57]

Psychopathology and mental health

Substance use and addiction

Environmental and epigenetic influences seem to work together to increase the risk of addiction.[15] For example, environmental stress has been shown to increase the risk of substance use.[58] Specifically, in an attempt to cope with stress, alcohol and drugs can be used as an escape.[59] Once substance use commences, however, epigenetic alterations may further exacerbate the biological and behavioural changes associated with addiction.[15]

Even short-term substance use can produce long-lasting epigenetic changes in the brain,[15] via DNA methylation and histone modification.[21] Epigenetic modifications have been observed in studies using alcohol, nicotene, cocaine, amphetamines, and opiates. Specifically, these epigenetic changes modify gene expression. In turn, this increases the vulnerability of an individual to engage in future, repeated substance use and even greater epigenetic changes in the brain's pleasure-reward areas[15] (e.g., in the nucleus accumbens[60]). Hence, a cycle emerges whereby changes in the pleasure-reward areas contribute to the long-lasting neural and behavioural changes associated with the increased likelihood of addiction, the maintenance of addiction and relapse.[15] For example, in humans, alcohol consumption has been shown to produce epigenetic changes that contribute to the increased craving of alcohol. As such, epigenetic modifications may play a part in the progression from the controlled intake to the loss of control of alcohol consumption.[61] These alterations may be long-term, as is evidenced in smokers who still possess nicotine-related epigenetic changes ten years after cessation.[62] Therefore, epigenetic modifactions[15] may account for some of the behavioural changes generally associated with addiction. These include: repetitive habits that increase the risk of disease and personal and social problems; need for immediate gratification; high rates of relapse following treatment; and the feeling of loss of control.[63]

Evidence for related epigenetic changes have come from human studies on alcohol,[64] nicotine and opiate use. Evidence for epigenetic changes stemming from amphetamine and cocaine use derives from animal studies. In animals, drug-related epigenetic changes in fathers have also been shown to negatively affect offspring in terms of poorer spatial working memory, decreased attention and decreased cerebral volume.[65]

Eating disorders and obesity

Epigenetics help to facilitate the development and maintenance of eating disorders via influences in the early environment and throughout the lifespan.[17] Pre-natal epigenetic changes due to maternal stress, behaviour and diet may later predispose offspring to persistent, increased anxiety and anxiety disorders. These anxiety issues can precipitate the onset of eating disorders and obesity and persist even after recovery from the eating disorders.[66]

In addition, epigenetic differences may accumulate over the life-span, thus accounting for the often incongruent differences in eating disorders observed in monozygotic twins. At puberty, sex hormones may exert epigenetic changes (via DNA methylation) on gene expression, thus accounting for higher rates of eating disorders in men as compared to women. Overall, epigenetics contribute to persistent, unregulated self-control behaviours related to the urge to binge.[17]

Schizophrenia

Epigenetic changes, including hypomethylation of glutamatergic genes (i.e., NMDA-receptor-subunit gene NR3B and the promoter of the AMPA-receptor-subunit gene GRIA2) have been observed in the post-mortem human brains of schizophrenics. These are associated with increased levels of the neurotoxin glutamate.[36] Since glutamate is the most prevalent fast excitatory neurotoxin, increased levels have been noted to result in the psychotic episodes related to schizophrenia. Interestingly, epigenetic changes affecting a greater number of genes have been detected in men with schizophrenia as compared to women with the illness.[67]

Population studies have also established a strong association linking schizophrenia in children to older fathers.[68][69][67] Specifically, children born to fathers over the age of 35 years are up to three times more likely to develop schizophrenia.[69] Epigenetic dysfunction in human male sperm cells, affecting numerous genes, have been shown to increase with age. This provides an explanation for increased rates of the disease in men.[69][67] To this end, toxins[69][67] (e.g., air pollutants) have been shown to increase epigenetic differentiation. Animals exposed to ambient air from steel mills and highways show drastic epigenetic changes that persist after removal from the exposure.[70] Therefore, similar epigenetic changes in older human fathers is likely.[69] Further, schizophrenia studies provide evidence that the nature versus nurture debate in the field of psychopathology should be re-evaluated to accommodate the concept that genes and the environment work in tandem. As such, many other environmental factors (e.g., nutritional deficiencies and cannabis use) have been proposed to increase the susceptability of psychotic disorders like schizophrenia via epigenetics.[69]

Bipolar disorder

Evidence for epigenetic modifications for bipolar disorder is unclear.[71] One study found hypomethylation of a gene promoter of a prefrontal lobe enzyme (i.e., membrane-bound catechol-O-methyl transferase, or COMT) in post-mortem brain samples from individuals with bipolar disorder. COMT is an enzyme that metabolizes dopamine in the synapse. These findings suggest that the hypomethylation of the promoter results in over-expression of the enzyme. In turn, this results in increased degradation of dopamine levels in the brain. Further, these findings provide evidence that epigenetic modification in the prefrontal lobe is a risk factor for bipolar disorder.[72] However, a second study found no epigenetic differences in post-mortem brains from bipolar individuals.[73]

Limitations and future direction

Many researchers are contributing information to the Human Epigenome Consortium.[74] The aim of future research is to reprogram epigenetic changes that will help with addiction-related issues, mental illnesses, age related changes[1] and memory decline.[8] However, the sheer volume of consortium-based data makes analysis difficult.[1] Most studies also focus on one gene.[49] In actuality, many genes and interactions between them likely contribute to individual differences in personality, behaviour and health.[75] As social scientists often work with more than ten variables, determining the number of affected genes also poses methodological challenges. More collaboration between medical researchers, geneticists and social scientists has been advocated to increase knowledge in this field of study.[76]

Some researchers note that pharmacological therapies may eventually be used to reverse epigenetic changes.[6] Others caution that more research is necessary as drugs are known to modify the activity of multiple genes and may, therefore, cause serious side effects.[8] However, the ultimate goal is to find patterns of epigenetic changes that can be targeted to treat mental illness, and reverse the effects of childhood stressors, for example. If such treatable patterns eventually become well-established, the inability to access brains in living humans to identify them poses an obstacle to pharmacological treatment.[74]

Although some research has translated findings from animals to humans,[14] others caution about the extrapolation of animal epigenetic studies to humans.[8] Limited access to human brain tissue has posed the greatest challenge to conducting human research.[1] Not yet knowing if epigenetic changes in the blood and (non-brain) tissues parallel modifications in the brain, places even greater reliance on brain research.[74]

Much research is cross-sectional in that it establishes associations. More longitudinal research is necessary to help establish causation.[77] Lack of resources has also limited the number of intergenerational studies.[1] Therefore, advancing longitudinal[76] and multigenerational, experience-dependent studies will be critical to further understanding the role of epigenetics in psychology.[3]

See also

References

  1. ^ a b c d e f g Powledge T (2011). "Behavioral epigenetics: How nurture shapes nature". BioScience. 61 (8): 588–592. doi:10.1525/bio.2011.61.8.4.
  2. ^ a b Kail RV, Barnfield A (2011). Children and Their Development, Second Canadian Edition with MyDevelopmentLab. Toronto: Pearson Education Canada. ISBN 0-13-255770-3.
  3. ^ a b c d e Champagne FA, Mashoodh R (2012). "Genes in context: Gene-environment interplay and the origins of individual differences in behaviour" (PDF). Current Directions in Psychological Science. 18 (3): 127–131. doi:10.1111/j.1467-8721.2009.01622.x.
  4. ^ a b c Bagot RC, Meaney MJ (2010). "Epigenetics and the biological basis of gene x environment interactions". J Am Acad Child Adolesc Psychiatry. 49 (8): 752–71. doi:10.1016/j.jaac.2010.06.001. PMID 20643310. {{cite journal}}: Unknown parameter |month= ignored (help)
  5. ^ Zhang TY, Meaney MJ (2010). "Epigenetics and the environmental regulation of the genome and its function". Annu Rev Psychol. 61: 439–66, C1–3. doi:10.1146/annurev.psych.60.110707.163625. PMID 19958180.
  6. ^ a b Stuffrein-Roberts S, Joyce PR, Kennedy MA (2008). "Role of epigenetics in mental disorders". Aust N Z J Psychiatry. 42 (2): 97–107. doi:10.1080/00048670701787495. PMID 18197504. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  7. ^ McIlroy, Anne (March 12, 2008). "Scientists solve a psychiatric mystery". The Globe and Mail.
  8. ^ a b c d e f g h i j k Miller G (2010). "Epigenetics. The seductive allure of behavioral epigenetics". Science. 329 (5987): 24–7. doi:10.1126/science.329.5987.24. PMID 20595592. {{cite journal}}: Unknown parameter |month= ignored (help)
  9. ^ a b c Pennisi E (2001). "Behind the scenes of gene expression". Science. 293 (5532): 1064–7. doi:10.1126/science.293.5532.1064. PMID 11498570. {{cite journal}}: Unknown parameter |month= ignored (help)
  10. ^ Juliandi B, Abematsu M, Nakashima K (2010). "Epigenetic regulation in neural stem cell differentiation". Dev. Growth Differ. 52 (6): 493–504. doi:10.1111/j.1440-169X.2010.01175.x. PMID 20608952. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  11. ^ Ma DK, Marchetto MC, Guo JU, Ming GL, Gage FH, Song H (2010). "Epigenetic choreographers of neurogenesis in the adult mammalian brain". Nat. Neurosci. 13 (11): 1338–44. doi:10.1038/nn.2672. PMID 20975758. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  12. ^ Sun J, Sun J, Ming GL, Song H (2011). "Epigenetic regulation of neurogenesis in the adult mammalian brain". Eur. J. Neurosci. 33 (6): 1087–93. doi:10.1111/j.1460-9568.2011.07607.x. PMC 3076719. PMID 21395852. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  13. ^ a b Weaver IC, Cervoni N, Champagne FA, D'Alessio AC, Sharma S, Seckl JR, Dymov S, Szyf M, Meaney MJ (2004). "Epigenetic programming by maternal behavior". Nat. Neurosci. 7 (8): 847–54. doi:10.1038/nn1276. PMID 15220929. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  14. ^ a b McGowan PO, Sasaki A, D'Alessio AC, Dymov S, Labonté B, Szyf M, Turecki G, Meaney MJ (2009). "Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse". Nat. Neurosci. 12 (3): 342–8. doi:10.1038/nn.2270. PMC 2944040. PMID 19234457. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  15. ^ a b c d e f g h i Wong CC, Mill J, Fernandes C (2011). "Drugs and addiction: an introduction to epigenetics". Addiction. 106 (3): 480–9. doi:10.1111/j.1360-0443.2010.03321.x. PMID 21205049. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  16. ^ a b c Jablonka E, Lamb MJ (2002). "The changing concept of epigenetics". Ann. N. Y. Acad. Sci. 981: 82–96. PMID 12547675. {{cite journal}}: Unknown parameter |month= ignored (help)
  17. ^ a b c d e f g h i j Campbell IC, Mill J, Uher R, Schmidt U (2011). "Eating disorders, gene-environment interactions and epigenetics". Neurosci Biobehav Rev. 35 (3): 784–93. doi:10.1016/j.neubiorev.2010.09.012. PMID 20888360. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  18. ^ a b c d e Masterpasqua F (2009). "Psychology and epigenetics". Reveiw of General Psychology. 13 (3): 194–201. doi:10.1037/a0016301.
  19. ^ a b Urdang, Laurence (2009). The Bantam Medical Dictionary, Sixth Edition. London: Bantam. ISBN 0-553-59226-2.
  20. ^ Mehler MF (2008). "Epigenetic principles and mechanisms underlying nervous system functions in health and disease". Prog. Neurobiol. 86 (4): 305–41. doi:10.1016/j.pneurobio.2008.10.00. PMC 2636693. PMID 18940229. {{cite journal}}: Unknown parameter |month= ignored (help)
  21. ^ a b Maze I, Nestler EJ (2011). "The epigenetic landscape of addiction". Ann. N. Y. Acad. Sci. 1216: 99–113. doi:10.1111/j.1749-6632.2010.05893.x. PMC 3071632. PMID 21272014. {{cite journal}}: Unknown parameter |month= ignored (help)
  22. ^ Reik W (2007). "Stability and flexibility of epigenetic gene regulation in mammalian development". Nature. 447 (7143): 425–32. doi:10.1038/nature05918. PMID 17522676. {{cite journal}}: Unknown parameter |month= ignored (help)
  23. ^ a b Gottesman II, Hanson DR (2005). "Human development: biological and genetic processes". Annu Rev Psychol. 56: 263–86. doi:10.1146/annurev.psych.56.091103.070208. PMID 15709936.
  24. ^ Bell JT, Spector TD (2011). "A twin approach to unraveling epigenetics". Trends Genet. 27 (3): 116–25. doi:10.1016/j.tig.2010.12.005. PMC 3063335. PMID 21257220. {{cite journal}}: Unknown parameter |month= ignored (help)
  25. ^ Dance, Amber (May 3, 2010). "DNA Referees". Los Angeles Times.
  26. ^ Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M (2005). "Epigenetic differences arise during the lifetime of monozygotic twins". Proc. Natl. Acad. Sci. U.S.A. 102 (30): 10604–9. doi:10.1073/pnas.0500398102. PMC 1174919. PMID 16009939. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  27. ^ a b Goto T, Monk M (1998). "Regulation of X-chromosome inactivation in development in mice and humans". Microbiol. Mol. Biol. Rev. 62 (2): 362–78. PMC 98919. PMID 9618446. {{cite journal}}: Unknown parameter |month= ignored (help)
  28. ^ Rakyan VK, Blewitt ME, Druker R, Preis JI, Whitelaw E (2002). "Metastable epialleles in mammals". Trends Genet. 18 (7): 348–51. doi:10.1016/S0168-9525(02)02709-9. PMID 12127774. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  29. ^ Lim AL, Ferguson-Smith AC (2010). "Genomic imprinting effects in a compromised in utero environment: implications for a healthy pregnancy". Semin. Cell Dev. Biol. 21 (2): 201–8. doi:10.1016/j.semcdb.2009.10.008. PMID 19879952. {{cite journal}}: Unknown parameter |month= ignored (help)
  30. ^ a b Guyton AC (2000). Textbook of Medical Physiology - 10th Edition. Philadelphia: W.B. Saunders. pp. 14, 33–36, 914, 916. ISBN 0-7216-8677-X.
  31. ^ Oz M, Roizen MF (2009). You: Having a baby: The owner's manual to a happy and healthy pregnancy. New York: Free Press. pp. 27–43. ISBN 1-4165-7236-8.
  32. ^ Cloud J (2010-01-06). "Why your DNA isn't your destiny". Time Magazine.
  33. ^ Pinel J (2010). Biopsychology (8th Edition). Englewood Cliffs, N.J: Prentice Hall. ISBN 0-205-83256-3.
  34. ^ Keller, M.C. (2006). "Resolving the paradox of common, harmful, heritable mental disorders: which evolutionary genetic models work best?". The behavioral and brain science. 29 (4): 405–452. PMID 17094843. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help); Unknown parameter |month= ignored (help)
  35. ^ Dulac C (2010). "Brain function and chromatin plasticity". Nature. 465 (7299): 728–35. doi:10.1038/nature09231. PMC 3075582. PMID 20535202. {{cite journal}}: Unknown parameter |month= ignored (help)
  36. ^ a b c Mostafavi-Abdolmaleky H, Glatt SJ, Tsuang MT (2011). "Epigenetics in Psychiatry". In Bronner F, Helmtrud I (ed.). Epigenetic Aspects of Chronic Diseases. Berlin: Springer. pp. 163–174. ISBN 1-84882-643-5.{{cite book}}: CS1 maint: multiple names: authors list (link)
  37. ^ Kirschner, M. W., & Gerhart, J. C. (2005). The Plausibility of Life: Resolving Darwin's Dilemma. New Haven, CT: Yale University Press. ISBN 978-0300108651.{{cite book}}: CS1 maint: multiple names: authors list (link)
  38. ^ Jablonka E, Lamb MJ (2002). "The changing concept of epigenetics". Ann. N. Y. Acad. Sci. 981: 82–96. doi:10.1111/j.1749-6632.2002.tb04913.x. PMID 12547675. {{cite journal}}: Unknown parameter |month= ignored (help)
  39. ^ a b c d e f g Kaminsky Z, Petronis A, Wang SC, Levine B, Ghaffar O, Floden D, Feinstein A (2008). "Epigenetics of personality traits: an illustrative study of identical twins discordant for risk-taking behavior". Twin Res Hum Genet. 11 (1): 1–11. doi:10.1375/twin.11.1.1. PMID 18251670. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  40. ^ Bakish D (1999). "The patient with comorbid depression and anxiety: the unmet need". J Clin Psychiatry. 60 Suppl 6: 20–4. PMID 10235121.
  41. ^ Costa, P.T. (1987). "Neuroticism, somatic complaints, and disease: is the bark worse than the bite?". Journal of Personality. 55 (2): 299–316. PMID 3612472. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help); Unknown parameter |month= ignored (help)
  42. ^ Kovács KJ, Miklós IH, Bali B (2004). "GABAergic mechanisms constraining the activity of the hypothalamo-pituitary-adrenocortical axis". Ann. N. Y. Acad. Sci. 1018: 466–76. doi:10.1196/annals.1296.057. PMID 15240403. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  43. ^ Cobos I, Broccoli V, Rubenstein JL (2005). "The vertebrate ortholog of Aristaless is regulated by Dlx genes in the developing forebrain". J. Comp. Neurol. 483 (3): 292–303. doi:10.1002/cne.20405. PMID 15682394. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  44. ^ Kash TL, Winder DG (2006). "Neuropeptide Y and corticotropin-releasing factor bi-directionally modulate inhibitory synaptic transmission in the bed nucleus of the stria terminalis". Neuropharmacology. 51 (5): 1013–22. doi:10.1016/j.neuropharm.2006.06.011. PMID 16904135. {{cite journal}}: Unknown parameter |month= ignored (help)
  45. ^ Gutman DA, Nemeroff DB (2010). "Stress and depression". In Baum A, Contrada RJ (ed.). The Handbook of Stress Science: Biology, Psychology, and Health. New York: Springer Publishing Company. p. 347. ISBN 0-8261-1471-7.
  46. ^ Amsterdam JD, Marinelli DL, Arger P, Winokur A (1987). "Assessment of adrenal gland volume by computed tomography in depressed patients and healthy volunteers: a pilot study". Psychiatry Res. 21 (3): 189–97. doi:10.1016/0165-1781(87)90022-9. PMID 3628606. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  47. ^ a b Szyf M, McGowan P, Meaney MJ (2008). "The social environment and the epigenome" (PDF). Environ. Mol. Mutagen. 49 (1): 46–60. doi:10.1002/em.20357. PMID 18095330. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  48. ^ a b Parkinson CF, McCance KL, Huether SE (1997). Pathophysiology : The Biologic Basis for Disease in Adults and Children (Study Guide & Workbook). St. Louis, USA: Mosby Elsevier Health Science. ISBN 0-8151-3766-4.{{cite book}}: CS1 maint: multiple names: authors list (link)
  49. ^ a b Baum A, Contrada RJ (2010). The Handbook of Stress Science: Biology, Psychology, and Health. New York: Springer Publishing Company. pp. 77–100. ISBN 0-8261-1471-7. Cite error: The named reference "McCaffery" was defined multiple times with different content (see the help page).
  50. ^ Weaver IC, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ, Szyf M (2005). "Reversal of maternal programming of stress responses in adult offspring through methyl supplementation: altering epigenetic marking later in life". J. Neurosci. 25 (47): 11045–54. doi:10.1523/JNEUROSCI.3652-05.2005. PMID 16306417. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  51. ^ Champagne FA, Weaver IC, Diorio J, Dymov S, Szyf M, Meaney MJ (2006). "Maternal care associated with methylation of the estrogen receptor-alpha1b promoter and estrogen receptor-alpha expression in the medial preoptic area of female offspring". Endocrinology. 147 (6): 2909–15. doi:10.1210/en.2005-1119. PMID 16513834. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  52. ^ a b Oberlander TF, Weinberg J, Papsdorf M, Grunau R, Misri S, Devlin AM (2008). "Prenatal exposure to maternal depression, neonatal methylation of human glucocorticoid receptor gene (NR3C1) and infant cortisol stress responses". Epigenetics. 3 (2): 97–106. PMID 18536531.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  53. ^ a b c Fischer A, Sananbenesi F, Wang X, Dobbin M, Tsai LH (2007). "Recovery of learning and memory is associated with chromatin remodelling". Nature. 447 (7141): 178–82. doi:10.1038/nature05772. PMID 17468743. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  54. ^ Gupta S, Kim SY, Artis S, Molfese DL, Schumacher A, Sweatt JD, Paylor RE, Lubin FD (2010). "Histone methylation regulates memory formation". J. Neurosci. 30 (10): 3589–99. doi:10.1523/JNEUROSCI.3732-09.2010. PMC 2859898. PMID 20219993. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  55. ^ Graff, J. (2012). "An epigenetic blockade of cognitive functions in the neurodegenerating brain". Nature. 483 (7388). doi:10.1038/nature10849. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  56. ^ Peleg S, Sananbenesi F, Zovoilis A, Burkhardt S, Bahari-Javan S, Agis-Balboa RC, Cota P, Wittnam JL, Gogol-Doering A, Opitz L, Salinas-Riester G, Dettenhofer M, Kang H, Farinelli L, Chen W, Fischer A (2010). "Altered histone acetylation is associated with age-dependent memory impairment in mice". Science. 328 (5979): 753–6. doi:10.1126/science.1186088. PMID 20448184. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  57. ^ Day JJ, Sweatt JD (2010). "DNA methylation and memory formation". Nat. Neurosci. 13 (11): 1319–23. doi:10.1038/nn.2666. PMC 3130618. PMID 20975755. {{cite journal}}: Unknown parameter |month= ignored (help)
  58. ^ Andersen SL, Teicher MH (2009). "Desperately driven and no brakes: developmental stress exposure and subsequent risk for substance abuse". Neurosci Biobehav Rev. 33 (4): 516–24. doi:10.1016/j.neubiorev.2008.09.009. PMC 2688959. PMID 18938197. {{cite journal}}: Unknown parameter |month= ignored (help)
  59. ^ Carver C (2010). "Coping". In Baum A, Contrada RJ (ed.). The Handbook of Stress Science: Biology, Psychology, and Health. New York: Springer Publishing Company. p. 223. ISBN 0-8261-1471-7.
  60. ^ Renthal W, Nestler EJ (2008). "Epigenetic mechanisms in drug addiction". Trends Mol Med. 14 (8): 341–50. doi:10.1016/j.molmed.2008.06.004. PMC 2753378. PMID 18635399. {{cite journal}}: Unknown parameter |month= ignored (help)
  61. ^ Naassila M (2011). "Epigenetics in alcohol addiction: New mechanisms for new treatments?". Alcohol and Alcoholism. 46 (Suppl 1): i1–63. doi:10.1093/alcalc/agr085. PMID 21863600.
  62. ^ Launay JM, Del Pino M, Chironi G, Callebert J, Peoc'h K, Mégnien JL, Mallet J, Simon A, Rendu F (2009). "Smoking induces long-lasting effects through a monoamine-oxidase epigenetic regulation". PLoS ONE. 4 (11): e7959. doi:10.1371/journal.pone.0007959. PMC 2775922. PMID 19956754.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  63. ^ Marlatt GA, Baer JS, Donovan DM, Kivlahan DR (1988). "Addictive behaviors: etiology and treatment". Annu Rev Psychol. 39: 223–52. doi:10.1146/annurev.ps.39.020188.001255. PMID 3278676.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  64. ^ Bönsch D, Lenz B, Reulbach U, Kornhuber J, Bleich S (2004). "Homocysteine associated genomic DNA hypermethylation in patients with chronic alcoholism". J Neural Transm. 111 (12): 1611–6. doi:10.1007/s00702-004-0232-x. PMID 15565495. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  65. ^ He F, Lidow IA, Lidow MS (2006). "Consequences of paternal cocaine exposure in mice". Neurotoxicol Teratol. 28 (2): 198–209. doi:10.1016/j.ntt.2005.12.003. PMID 16458479.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  66. ^ Raney TJ, Thornton LM, Berrettini W, Brandt H, Crawford S, Fichter MM, Halmi KA, Johnson C, Kaplan AS, LaVia M, Mitchell J, Rotondo A, Strober M, Woodside DB, Kaye WH, Bulik CM (2008). "Influence of overanxious disorder of childhood on the expression of anorexia nervosa". Int J Eat Disord. 41 (4): 326–32. doi:10.1002/eat.20508. PMID 18213688. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  67. ^ a b c d Abdolmaleky HM, Thiagalingam S, Wilcox M (2005). "Genetics and epigenetics in major psychiatric disorders: dilemmas, achievements, applications, and future scope". Am J Pharmacogenomics. 5 (3): 149–60. PMID 15952869.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  68. ^ Malaspina D, Harlap S, Fennig S, Heiman D, Nahon D, Feldman D, Susser ES (2001). "Advancing paternal age and the risk of schizophrenia". Arch. Gen. Psychiatry. 58 (4): 361–7. PMID 11296097. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  69. ^ a b c d e f Rutten BP, Mill J (2009). "Epigenetic mediation of environmental influences in major psychotic disorders". Schizophr Bull. 35 (6): 1045–56. doi:10.1093/schbul/sbp104. PMC 2762629. PMID 19783603. {{cite journal}}: Unknown parameter |month= ignored (help)
  70. ^ Yauk C, Polyzos A, Rowan-Carroll A, Somers CM, Godschalk RW, Van Schooten FJ, Berndt ML, Pogribny IP, Koturbash I, Williams A, Douglas GR, Kovalchuk O (2008). "Germ-line mutations, DNA damage, and global hypermethylation in mice exposed to particulate air pollution in an urban/industrial location". Proc. Natl. Acad. Sci. U.S.A. 105 (2): 605–10. doi:10.1073/pnas.0705896105. PMC 2206583. PMID 18195365. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  71. ^ McGowan PO, Kato T (2008). "Epigenetics in mood disorders". Environ Health Prev Med. 13 (1): 16–24. doi:10.1007/s12199-007-0002-0. PMC 2698240. PMID 19568875. {{cite journal}}: Unknown parameter |month= ignored (help)
  72. ^ Abdolmaleky HM, Cheng KH, Faraone SV, Wilcox M, Glatt SJ, Gao F, Smith CL, Shafa R, Aeali B, Carnevale J, Pan H, Papageorgis P, Ponte JF, Sivaraman V, Tsuang MT, Thiagalingam S (2006). "Hypomethylation of MB-COMT promoter is a major risk factor for schizophrenia and bipolar disorder". Hum. Mol. Genet. 15 (21): 3132–45. doi:10.1093/hmg/ddl253. PMC 2799943. PMID 16984965. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  73. ^ Dempster EL, Mill J, Craig IW, Collier DA (2006). "The quantification of COMT mRNA in post mortem cerebellum tissue: diagnosis, genotype, methylation and expression". BMC Med. Genet. 7: 10. doi:10.1186/1471-2350-7-10. PMC 1456954. PMID 16483362.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  74. ^ a b c Albert PR (2010). "Epigenetics in mental illness: hope or hype?". J Psychiatry Neurosci. 35 (6): 366–8. doi:10.1503/jpn.100148. PMC 2964366. PMID 20964959. {{cite journal}}: Unknown parameter |month= ignored (help)
  75. ^ Kalant H (2010). "What neurobiology cannot tell us about addiction". Addiction. 105 (5): 780–9. doi:10.1111/j.1360-0443.2009.02739.x. PMID 19919596. {{cite journal}}: Unknown parameter |month= ignored (help)
  76. ^ a b Guo G (2010). "Family influences on children's well being: potential roles of molecular genetics and epigenetics". In Landale N, Booth A, McHale S (ed.). Biosocial Foundations of Family Processes (National Symposium on Family Issues). Berlin: Springer. pp. 181–204. ISBN 1-4419-7360-5.{{cite book}}: CS1 maint: multiple names: editors list (link)
  77. ^ Dempster EL, Pidsley R, Schalkwyk LC, Owens S, Georgiades A, Kane F, Kalidindi S, Picchioni M, Kravariti E, Toulopoulou T, Murray RM, Mill J (2011). "Disease-associated epigenetic changes in monozygotic twins discordant for schizophrenia and bipolar disorder". Hum. Mol. Genet. 20 (24): 4786–96. doi:10.1093/hmg/ddr416. PMC 3221539. PMID 21908516. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)

Further reading

  • McDonald B (2011). "The Fingerprints of Poverty". Quirks & Quarks. CBC Radio. Audio interview with Dr. Moshe Szyf, a professor of Pharmacology and Therapeutics at McGill University, discusses how epigenetic changes are related to differences in socioeconomic status.
  • Paylor B (2010). "Epigenetic Landscapes". This video addresses how, in principle, accumulated epigenetic changes may result in personality differences in identical twins. This science film was made by a Ph.D candidate in experimental medicine and award winning filmmaker Ben Paylor.
  • Oz M (2011). "Control Your Pregnancy". The Dr. Oz Show. Video explaining how epigenetics can affect the unborn fetus.