Elsevier

Brain Research

Volume 1444, 20 March 2012, Pages 96-111
Brain Research

Review
Epigenetic programming of neurodegenerative diseases by an adverse environment

https://doi.org/10.1016/j.brainres.2012.01.038Get rights and content

Abstract

Experience and environment can critically influence the risk and progression of neurodegenerative disorders. Epigenetic mechanisms, such as miRNA expression, DNA methylation, and histone modifications, readily respond to experience and environmental factors. Here we propose that epigenetic regulation of gene expression and environmental modulation thereof may play a key role in the onset and course of common neurological conditions, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. For example, epigenetic mechanisms may mediate long-term responses to adverse experience, such as stress, to affect disease susceptibility and the course of neurodegenerative events. This review introduces the epigenetic components and their possible role in mediating neuropathological processes in response to stress. We argue that epigenetic modifications will affect neurodegenerative events through altered gene function. The study of epigenetic states in neurodegenerative diseases presents an opportunity to gain new insights into risk factors and pathogenic mechanisms. Moreover, research into epigenetic regulation of disease may revolutionize health care by opening new avenues of personalized, preventive and curative medicine.

Introduction

The notion that the environment influences the phenotype of a living organism goes back to Jean–Baptiste Lamarck's concept of the driving forces of evolution (Lamarck, 1914). Rapid progress in genetic and epigenetic analysis technologies over the recent years has revolutionized the depth of understanding of the environmental impact on brain function and adaptation far beyond the early notions of Lamarck. In addition, the completion of the Human Genome Project and the commencement of the Human Epigenome Project opened new avenues for life sciences and biomedical research (http://www.epigenome.org).

The nature of genetic and epigenetic changes in response to environmental stimuli has received particular attention in the quest to understand the origins of disease. Whereas genetics involves permanent and mostly irreversible changes to DNA sequence, epigenetics is thought to concern mainly transient and reversible changes to chromatin and gene expression (Wu and Morris, 2001). Although tremendous breakthroughs have been made in the search for genetic and epigenetic patterns linked to disease, a comprehensive explanation of the mechanisms that underlie phenotypic plasticity is still far from being realistic. However, epigenetic regulation of gene expression has been identified as a key player in producing rapid adaptation to changing environmental conditions both within a single lifespan as well as across multiple generations. These mechanisms particularly apply to the brain, which is capable of changing readily in response to experience throughout a lifetime.

The discovery that the brain changes in response to experience began with early studies by Ramon y Cajal (Cajal, 1928). It is assumed that experience and environment are major factors to determine neuronal plasticity and susceptibility to neurological diseases (Crews, 2008). Although the concept of brain plasticity has now become central to neuroscientific studies, the enormous power of epigenetic regulation to determine brain plasticity and its output behavior has become recognized mainly in the past ten years. Ultimately, the epigenome may represent a critical link to understanding not only adaptive responses of the brain to beneficial experience, but also to elucidate mechanisms of pathological processes induced by adverse experience, such as stress.

Stress likely represents a critical influence on neuronal function and disease. The term stress refers to perturbation of homeostasis by a stressful event causing the onset of complex neural and endocrine adaptations characterized by activation of the hypothalamic–pituitary–adrenal (HPA) axis, which prompts the release of glucocorticoid hormones from the adrenal gland. Glucocorticoids act throughout the body, but very prominently their actions are focused on the brain. Excessive or chronically elevated glucocorticoid levels in particular can compromise neuronal survival and diminish the plastic capacity of the brain (Sapolsky, 2000a, Sapolsky, 2000b, Sapolsky, 2000c). The effects of physiological stress on neuronal integrity and survival involve cellular and biochemical processes including compromised chemoresistance to oxidative stress (Landriscina et al., 2009), exaggerated activation of inflammatory cascades (Matsunaga et al., 2011, Sorrells et al., 2009), altered neurotrophic factor expression (Smith et al., 1995) or facilitated cytoplasmic protein accumulation (Sotiropoulos et al., 2011). Epigenetic programming through previous experience may determine the ability of the brain to adapt to a stressful environment in the long term. Maladaptive epigenetic programming by chronic stress, however, may enhance the vulnerability to stress and place the brain at a higher risk of neurodegenerative events.

The aim of this review is to provide an overview of possible epigenetic mechanisms involved in the pathology of neurodegenerative disease. A specific focus will be on the role of stressful experiences in altering epigenetic regulation of gene expression. We propose that chronic stress may affect the course of neurodegenerative events through DNA methylation, chromatin remodeling and histone modification, and microRNA (miRNA) expression. We hypothesize that epigenetic regulation in response to chronic stress may contribute to the pathogenesis of common neurodegenerative diseases.

Section snippets

The genome processes and integrates environmental signals via epigenetic regulation

The new discipline of epigenetics presents a critical level in molecular mechanisms that allow the brain to integrate environmental signals and process stressful experiences. Epigenetics is the study of heritable changes in gene expression that are not caused by changes in the nucleotide sequence of the DNA. In this review we will discuss the main mechanisms of epigenetic regulation of neuronal function and survival, such as DNA methylation, histone modification and chromatin remodeling, as

Epigenetic mechanisms underlying neurodegenerative diseases

Neurodegenerative diseases are associated with progressive and irreversible loss of neuronal structure and function in the central nervous system. The progressive damage and loss of neurons ultimately results in gradual onset of functional disability. This review will discuss the potential relevance of epigenetic mechanisms in disease etiology and progression for the three most common neurodegenerative conditions, Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease

Physiological, molecular, behavioral, and pathological correlates of stress

Stress may be the single most important influence on behavior, performance and disease (Selye, 1975a, Selye, 1975b). The stress response serves as the physiological and behavioral adaptation to an emotional or physical threat (de Kloet et al., 2009) through activation of components of the central nervous system and peripheral nervous system (Weinstock, 2010). In response to stress, the paraventricular nucleus of the hypothalamus releases corticotropin-releasing hormone that activates the

Conclusions

A growing body of evidence suggests that epigenetic mechanisms play a central role in the regulation of gene expression in the nervous system during development and in adulthood. Hence, the epigenetic makeup of an organism significantly determines ageing process of the central nervous system, thus being able to influence the onset and progression of neurodegenerative diseases. Therefore it is not surprising that many neurological disorders are accompanied by changes in epigenetic patterns in

Conflict of interest

The authors declare that there is no conflict of interest.

Acknowledgments

The authors acknowledge support by the University of Lethbridge Faculty of Arts & Science, the Canadian Institutes of Health Research and the AI-HS Interdisciplinary Team Grant #200700595 “Preterm Birth and Healthy Outcomes”. GM is a Senior Scholar of Alberta Innovates-Health Solutions.

References (233)

  • H. Coirini et al.

    Further studies of brain aldosterone binding sites employing new mineralocorticoid and glucocorticoid receptor markers in vitro

    Brain Res.

    (1985)
  • A. Compston et al.

    Multiple sclerosis

    Lancet

    (2002)
  • A. Compston et al.

    Multiple sclerosis

    Lancet

    (2008)
  • L.A. Craig et al.

    Revisiting the cholinergic hypothesis in the development of Alzheimer's disease

    Neurosci. Biobehav. Rev.

    (2011)
  • D. Crews

    Epigenetics and its implications for behavioral neuroendocrinology

    Front. Neuroendocrinol.

    (2008)
  • B. Czeh et al.

    Chronic psychosocial stress and concomitant repetitive transcranial magnetic stimulation: effects on stress hormone levels and adult hippocampal neurogenesis

    Biol. Psychiatry

    (2002)
  • A. Dagnino-Subiabre et al.

    Chronic stress induces upregulation of brain-derived neurotrophic factor (BDNF) mRNA and integrin alpha5 expression in the rat pineal gland

    Brain Res.

    (2006)
  • E.R. de Kloet et al.

    Glucocorticoid signaling and stress-related limbic susceptibility pathway: about receptors, transcription machinery and microRNA

    Brain Res.

    (2009)
  • J. Faraji et al.

    Stress and corticosterone enhance cognitive recovery from hippocampal stroke in rats

    Neurosci. Lett.

    (2009)
  • J. Faraji et al.

    Chronic stress prior to hippocampal stroke enhances post-stroke spatial deficits in the ziggurat task

    Neurobiol. Learn. Mem.

    (2011)
  • J. Faraji et al.

    Stress precipitates functional deficits following striatal silent stroke: a synergistic effect

    Exp. Neurol.

    (2011)
  • E.L. Gershey et al.

    Chemical studies of histone acetylation. The occurrence of epsilon-N-acetyllysine in the f2a1 histone

    J. Biol. Chem.

    (1968)
  • I.L. Goldknopf et al.

    Remarkable similarities of peptide fingerprints of histone 2A and nonhistone chromosomal protein A24

    Biochem. Biophys. Res. Commun.

    (1975)
  • A.E. Handel et al.

    No evidence for an effect of DNA methylation on multiple sclerosis severity at HLA-DRB1*15 or HLA-DRB5

    J. Neuroimmunol.

    (2010)
  • M.M. Harraz et al.

    MicroRNAs in Parkinson's disease

    J. Chem. Neuroanat.

    (2011)
  • R.B. Harris et al.

    Chronic disruption of body weight but not of stress peptides or receptors in rats exposed to repeated restraint stress

    Horm. Behav.

    (2006)
  • J.P. Herman et al.

    Central mechanisms of stress integration: hierarchical circuitry controlling hypothalamo–pituitary–adrenocortical responsiveness

    Front. Neuroendocrinol.

    (2003)
  • R.E. Hughes

    Polyglutamine disease: acetyltransferases awry

    Curr. Biol.

    (2002)
  • N.M. Jadavji et al.

    Sex differences in skilled movement in response to restraint stress and recovery from stress

    Behav. Brain Res.

    (2008)
  • M. Joels et al.

    Chronic stress: implications for neuronal morphology, function and neurogenesis

    Front. Neuroendocrinol.

    (2007)
  • R. Johnson et al.

    A microRNA-based gene dysregulation pathway in Huntington's disease

    Neurobiol. Dis.

    (2008)
  • H. Kimura et al.

    Methyl-CpG-binding protein, MeCP2, is a target molecule for maintenance DNA methyltransferase, Dnmt1

    J. Biol. Chem.

    (2003)
  • S.W. Kirkland et al.

    Delayed recovery and exaggerated infarct size by post-lesion stress in a rat model of focal cerebral stroke

    Brain Res.

    (2008)
  • R.J. Klose et al.

    Genomic DNA methylation: the mark and its mediators

    Trends Biochem. Sci.

    (2006)
  • D. Aarsland et al.

    Prevalence and clinical correlates of psychotic symptoms in Parkinson disease: a community-based study

    Arch. Neurol.

    (1999)
  • V. Ambros

    The functions of animal microRNAs

    Nature

    (2004)
  • S.E. Baranzini et al.

    Genome, epigenome and RNA sequences of monozygotic twins discordant for multiple sclerosis

    Nature

    (2010)
  • M. Barrachina et al.

    DNA methylation of Alzheimer disease and tauopathy-related genes in postmortem brain

    J. Neuropathol. Exp. Neurol.

    (2009)
  • E.A. Bates et al.

    Differential contributions of Caenorhabditis elegans histone deacetylases to huntingtin polyglutamine toxicity

    J. Neurosci.

    (2006)
  • S.K. Bhattacharya et al.

    A mammalian protein with specific demethylase activity for mCpG DNA

    Nature

    (1999)
  • M.B. Bracken et al.

    A randomized, controlled trial of methylprednisolone or naloxone in the treatment of acute spinal-cord injury. Results of the Second National Acute Spinal Cord Injury Study

    N. Engl. J. Med.

    (1990)
  • J. Brohede et al.

    A DNA methylation study of the amyloid precursor protein gene in several brain regions from patients with familial Alzheimer disease

    J. Neurogenet.

    (2010)
  • A.M. Burrell et al.

    Epigenetic mechanisms in multiple sclerosis and the major histocompatibility complex (MHC)

    Discov. Med.

    (2011)
  • R. Cacabelos

    Pharmacogenomics and therapeutic prospects in Alzheimer's disease

    Expert Opin. Pharmacother.

    (2005)
  • R. Cacabelos

    Pharmacogenetic basis for therapeutic optimization in Alzheimer's disease

    Mol. Diagn. Ther.

    (2007)
  • R. Cajal

    Degeneration and Regeneration of the Nervous System

    (1928)
  • C. Caldji et al.

    Variations in maternal care alter GABA(A) receptor subunit expression in brain regions associated with fear

    Neuropsychopharmacology

    (2003)
  • E. Castilla-Ortega et al.

    Aggravation of chronic stress effects on hippocampal neurogenesis and spatial memory in LPA(1) receptor knockout mice

    PLoS One

    (2011)
  • J.J. Cerqueira et al.

    The prefrontal cortex as a key target of the maladaptive response to stress

    J. Neurosci.

    (2007)
  • M. Chahrour et al.

    MeCP2, a key contributor to neurological disease, activates and represses transcription

    Science

    (2008)
  • Cited by (71)

    • Epigenetic regulons in Alzheimer's disease

      2023, Progress in Molecular Biology and Translational Science
    • Stress system and related biomarkers in Parkinson's disease

      2022, Advances in Clinical Chemistry
      Citation Excerpt :

      Such factors are: first, DNA methylation, which may regulates α-synuclein expression [61], as well as, significant shortening of leucocyte telomeres in PD affecting health and lifespan [61]. Aberrant DNA methylation of key clock genes in the PD brain may also promote widespread circadian deregulation and neuronal dysfunction [61,62]. Second, increased baseline circulating GCs due to prolonged exposure to stress early in life, has also been associated with compromised neuronal survival.

    • Stress and cortisol in Parkinson's disease

      2020, International Review of Neurobiology
    View all citing articles on Scopus
    1

    Authors contributed equally.

    View full text