You are on page 1of 8

NEUROLOGICAL REVIEW

Understanding Neurological Disease Mechanisms


in the Era of Epigenetics
Irfan A. Qureshi, MD; Mark F. Mehler, MD

T
he burgeoning field of epigenetics is making a significant impact on our understand-
ing of brain evolution, development, and function. In fact, it is now clear that epigen-
etic mechanisms promote seminal neurobiological processes, ranging from neural stem
cell maintenance and differentiation to learning and memory. At the molecular level,
epigenetic mechanisms regulate the structure and activity of the genome in response to intracel-
lular and environmental cues, including the deployment of cell type–specific gene networks and
those underlying synaptic plasticity. Pharmacological and genetic manipulation of epigenetic fac-
tors can, in turn, induce remarkable changes in neural cell identity and cognitive and behavioral
phenotypes. Not surprisingly, it is also becoming apparent that epigenetics is intimately involved
in neurological disease pathogenesis. Herein, we highlight emerging paradigms for linking epi-
genetic machinery and processes with neurological disease states, including how (1) mutations in
genes encoding epigenetic factors cause disease, (2) genetic variation in genes encoding epigen-
etic factors modify disease risk, (3) abnormalities in epigenetic factor expression, localization, or
function are involved in disease pathophysiology, (4) epigenetic mechanisms regulate disease-
associated genomic loci, gene products, and cellular pathways, and (5) differential epigenetic pro-
files are present in patient-derived central and peripheral tissues.
JAMA Neurol. 2013;70(6):703-710.
Published online April 9, 2013. doi:10.1001/jamaneurol.2013.1443

The hallmarks of the human brain are its ers, and alternative exons) represent prime
extraordinary degree of cellular diversity, advances toward this goal.1,2 This postge-
capacity for synaptic and neural network nomic era has been defined by the rise of
connectivity and plasticity, and intellec- epigenetics—the scientific discipline fo-
tual abilities. Ongoing efforts have sought cused on interrogating how genomic pro-
to better understand this hierarchical or- cesses, such as gene transcription and DNA
ganization and the molecular, cellular, and replication and repair, are mediated in dif-
environmental mechanisms responsible for ferent cellular contexts.
generating it. The completion of the Hu- Epigenetics promises to provide insights
man Genome Project and the continuing thatwillhelpanswerseminalquestionsabout
characterization of functional genomic ele- the human brain. How did it evolve? How
ments (tissue-specific promoters, enhanc- does the human genome encode neural cel-
Author Affiliations: Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology lular diversity? How do genetic factors and
and Regenerative Medicine (Drs Qureshi and Mehler), Institute for Brain Disorders environmental stimuli interact to promote
and Neural Regeneration (Drs Qureshi and Mehler), Departments of Neurology synaptic and neural connectivity and plas-
(Drs Qureshi and Mehler), Neuroscience (Dr Mehler), and Psychiatry and ticity?Howdocognitiveandbehavioraltraits
Behavioral Sciences (Dr Mehler), Rose F. Kennedy Center for Research on
emerge? Most importantly, what mecha-
Intellectual and Developmental Disabilities (Drs Qureshi and Mehler), Einstein
Cancer Center (Dr Mehler), Ruth L. and David S. Gottesman Institute for Stem nisms are responsible for the pathogenesis
Cell Biology and Regenerative Medicine Research (Dr Mehler), Center for of complex neurological diseases?
Epigenomics (Dr Mehler), and Institute for Aging Research (Dr Mehler), Albert Further, the rapidly emerging era of
Einstein College of Medicine, Bronx, New York, New York. highly personalized epigenetic and epi-

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


703

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021
Environmental cues/neuromodulators/synaptic activity/
metabolic signals/stress responses

ADAR 1/2 miRNAs ESC

A RNA degradation
APOBECs Translational repression NSC
I

C
mRNA/ncRNA
U

Nucleosomes
Histones

DNA strand

N1 N2 N3 OL AS
me

P/Me/Ac/Ub
5′ ...NCGN...
...NGCN... 5′ H2A
me

H3 H4 SWI/SNF
H2B Adult neurogenesis

MBDs DG
DNMTs
HMTs/HDMs
HATs/HADCs

LTP
PcG/TrxG
ΔLTD/ΔLTP

ANRIL/HOTAIR
LTD

Figure 1. Environmental cues, neuromodulators, synaptic activity, metabolic signals, and stress responses lead to the activation of diverse epigenetic
mechanisms, including DNA methylation, histone modifications and chromatin remodeling, noncoding RNA (ncRNA) expression, and RNA editing. In turn, these
processes mediate embryonic stem cell (ESC) and neural stem cell (NSC) maintenance and maturation, adult neurogenesis, neural network formation, and
synaptic plasticity. Ac indicates acetylation; ADAR, adenosine deaminases that act on RNA enzymes; APOBECs, apolipoprotein B editing catalytic subunit enzymes;
AS, astrocyte; C, cytosine; DG, dentate gyrus; DNMTs, DNA methyltransferases; G, guanine; HATs, histone acetylases; HDACs, histone deacetylases; HDMs,
histone demethylases; HMTs, histone methyltransferases; LTD, long-term depression; LTP, long-term potentiation; MBDs, methyl-CpG-binding domain proteins;
me, methylation; miRNA, microRNA; mRNA, messenger RNA; N, any nucleotide; N1-3, neuronal subtypes 1-3; OL, oligodendrocyte; P, phosphorylation;
PcG, Polycomb group proteins; TrxG, Trithorax group proteins; and Ub, ubiquitination.

genomic medicine is poised to radically transform diag- ases (DNase I).3 Higher-order chromatin exists as rela-
nostic and therapeutic strategies for neurological dis- tively open (euchromatic) or highly condensed (hetero-
eases and to deliver innovative treatments to promote chromatic) structures. Euchromatin is generally associated
neural protection and repair. with active transcription, whereas heterochromatin is typi-
cally found in inactive regions, such as repressed genes
THE ERA OF EPIGENETICS and structural components of chromosomes (centro-
meres and telomeres). Chromatin structure is dynamic
Revolutionary Insights and subject to local modification at the level of indi-
vidual nucleotides, histone proteins, and nucleosomes
Among the most important insights to have emerged is and genome-wide by higher-order chromatin remodel-
an appreciation for chromatin organization in regulat- ing. Protein complexes mediate these processes by the
ing genomic function and establishing cellular memory capacity to “read,” “erase,” and “write” specific chroma-
states. Chromatin refers to the packaging of the genome tin states (“marks”). Inhibiting specific chromatin-
within the cell nucleus. DNA is wrapped around a his- modifying enzymes is a powerful tool for modulating gene
tone protein octamer, forming a fundamental chroma- expression programs and a strategy approved by the Food
tin structure, the nucleosome. Chromatin states play cen- and Drug Administration for select disease indications
tral roles in coordinating the accessibility of DNA and now in preclinical and clinical trials for cancer and
sequences to factors in the nucleus mediating critical cel- neurodegenerative diseases.
lular processes, including gene transcription. Nucleosome- Chromatin states are intimately linked to the estab-
free regions represent DNA actively engaged in regula- lishment and maintenance of cell identity (Figure 1).
tory and other functions. These regions can be identified, Chromatin exists in a relatively open conformation in em-
experimentally, by their relative hypersensitivity to nucle- bryonic stem cells, reflecting their pluripotent state. As

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


704

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021
cellular differentiation proceeds, chromatin becomes more
closed, with marks indicative of gene repression, par- Table 1. Principal Epigenetic Mechanisms and Factors
ticularly in chromatin associated with pluripotency genes.
Conversely, chromatin associated with lineage-specific Epigenetic Mechanism Epigenetic Factors
genes selectively opens and becomes decorated with marks DNA (de)methylation and DNA methyltransferase enzymes
of active gene transcription. Differences in chromatin hydroxymethylation Methyl-CpG-binding domain proteins
states explain how the genome gives rise to the plethora DNA excision repair enzymes
Cytidine deaminase enzymes
of organismal cell types, including the extraordinary di- Gadd45 proteins
versity of neurons and glia of the human brain. Agents Ten-Eleven Translocation enzymes
targeting epigenetic enzymes have, therefore, been used Histone and chromatin Histone-modifying (histone [de]acetylase
to reprogram cell identity and function and may also pro- modifications and [de]methylase) enzymes
mote neural regeneration and repair. SWI/SNF nucleosome remodeling
complexes
Another key insight is that the entire human genome
Polycomb group proteins
is actively transcribed, including regions that code for pro- Trithorax group proteins
teins (⬍2%) as well as those that do not. Such noncoding RE1-silencing transcription factor
regions give rise to numerous classes of noncoding RNAs CoREST
(ncRNAs)—a previously unexplored layer within the ge- Noncoding RNAs MicroRNAs
nome—that are interlaced with each other and with protein- Small nucleolar RNAs
coding messenger RNAs (mRNAs). The identification of Endogenous short-interfering RNAs
PIWI-interacting RNAs
large numbers of these ncRNAs embedded within the ge- Long noncoding RNAs
nome and recognition of their important biological roles RNA editing Adenosine deaminases that act on RNA
directly challenges the central dogma of molecular biol- enzymes
ogy (DNA makes RNA and RNA makes protein). Apolipoprotein B editing catalytic subunit
Noncoding RNAs are far more abundant than protein- enzymes
coding mRNAs in human cells, particularly in neural cells.
Abbreviations: CoREST, corepressor for element-1–silencing transcription
They engage in complex interactions with DNA, RNA, factor; RE1, repressor for element-1.
and proteins and exhibit a spectrum of sophisticated func-
tions. Certain classes of ncRNAs are implicated in gene
silencing whereas others are involved in regulating the ies, and repetitive elements) and DNA methylation seems
activity of retrotransposons. Notably, the expression of to be context specific. The effects of DNA methylation
ncRNAs is most prominent in the brain and subject to are mediated by methyl-CpG-binding domain proteins,
intricate temporal and spatial regulation during neural including methyl-CpG-binding protein 2 (MECP2), which
development, homeostasis, and plasticity, highlighting bind to 5mC and recruit additional regulatory factors to
their significance.4 In fact, it is becoming clear that non- methylated loci. 5-Hydroxymethylcytosine, another modi-
coding DNA sequences and associated ncRNAs serve as fied cytosine residue generated by the Ten-Eleven Trans-
substrates for evolutionary innovations in the human location family of enzymes that oxidizes 5mC, appears
brain. Human accelerated regions represent elements in to counteract 5mC by inhibiting the function of methyl-
the human genome that have evolved rapidly since di- CpG-binding domain proteins.
vergence from our common ancestor with chimpan- Each nucleosome contains 2 of each core histone pro-
zees. One of hundreds of such regions, HAR1, gives rise tein (H2A, H2B, H3, and H4). Specific classes of histone
to an ncRNA selectively expressed in Cajal-Retzius cells deacetylases (HDACs) as well as histone acetyltransfer-
in the developing fetus and implicated in neuronal sub- ases, histone demethylases, histone methyltransferases, and
type specification and cell migration in the neocortex. others catalyze histone posttranslational modifications.
These enzymes serve as “writers” of posttranslational modi-
Epigenetic Mechanisms fications, thereby altering nucleosome structure and func-
tion by modifying interactions with other proteins that
The major epigenetic mechanisms include DNA meth- “read,” “erase,” and “write” epigenetic marks. Macromo-
ylation, histone modifications and nucleosome and higher- lecular complexes containing various proteins with the abil-
order chromatin remodeling, ncRNAs, and RNA editing ity to simultaneously read, erase, and write epigenetic marks
(Table 1). DNA methylation refers to covalent modifi- promote nucleosome (SWI/SNF) and higher-order chro-
cation of cytosine residues to form 5-methylcytosine matin (Polycomb group and Trithorax group proteins) re-
(5mC). 5-Methylcytosine levels are dynamically regu- modeling. Epigenetic regulatory complexes (repressor for
lated during development and adulthood. Enzymes that element-1 [RE1]–silencing transcription factor [REST] and
catalyze DNA methylation are DNA methyltransferases; co-RE1–silencing transcription factor [CoREST] com-
those that catalyze DNA demethylation include DNA ex- plexes) can also be highly modular and have functions that
cision repair, cytidine deaminase, and Gadd45 proteins. range from recognizing specific nucleotide sequences and
The presence of 5mC at specific gene loci was previ- DNA methylation states to promoting local histone and
ously thought to promote transcriptional repression and higher-order chromatin modifications.
long-term gene silencing. However, high-resolution ge- Newly recognized classes of ncRNAs are classified as
nome-wide studies interrogating DNA methylation re- short or long. Salient classes of short ncRNAs include mi-
veal that 5mC is distributed throughout the genome (gene croRNAs (miRNAs), endogenous short-interfering RNAs,
regulatory regions, transcriptional start sites, gene bod- PIWI-interacting RNAs, and small nucleolar RNAs. Each

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


705

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021
class is associated with specific biogenesis pathways, repertoires of DNA methyltransferases, methyl-CpG-
mechanisms of action, and functions. MicroRNAs are the binding domain proteins, histone- and chromatin-
best-characterized class of short ncRNAs. They engage modifying enzymes, and ncRNA biogenesis factors are
in posttranscriptional gene regulation. Specifically, single- necessary for mediating neurogenesis and gliogenesis.
stranded, 20- to 23-nucleotide miRNAs bind to protein- Moreover, as embryonic stem cells differentiate into neu-
coding mRNAs in their 3' untranslated regions through ral lineage-committed progenitors, hundreds of gene pro-
complementary sequence-specific interactions and pre- moter regions are methylated, including those associ-
vent translation of these target mRNAs. Importantly, a ated with pluripotency and alternative lineage genes. These
single miRNA molecule can repress a network of mRNAs. DNA methylation events are responsible for silencing these
Long ncRNAs (lncRNAs) are the most abundant but least genes, thereby promoting neural lineage commitment.
well-characterized class of ncRNAs. The most common Histone modifications and chromatin remodeling are simi-
strategy for analyzing lncRNAs is to study their genomic larly involved in establishing neural cell identity. For ex-
context relative to protein-coding genes. Some lncRNAs ample, the developmental stage-specific deployment of
are derived from intergenic regions (long intergenic/ REST and CoREST complexes modulates neural lineage
intervening ncRNAs). Other lncRNAs are transcribed in elaboration, including neuronal and glial subtype speci-
antisense or overlapping orientations relative to protein- fication, and terminal differentiation. In terms of ncRNAs,
coding genes, and these transcripts are often function- both miRNAs and lncRNAs regulate neural stem cell self-
ally interrelated. For example, expression of a protein- renewal and differentiation programs. The balance be-
coding mRNA might be specifically regulated by an tween specific miRNAs is implicated in the execution of
antisense lncRNA from the same genomic locus. Never- these programs. For example, miR-184 promotes neural
theless, lncRNAs are highly versatile with a spectrum of stem cell proliferation and inhibits neuronal differentia-
novel activities. These include roles in recruiting non- tion, whereas miR-9 and miR-137 inhibit neural stem cell
selective transcriptional and epigenetic regulators to spe- proliferation and promote neural differentiation. Other
cific loci distributed throughout the genome, forming miRNAs, including miR-125b, miR-128, miR-132, miR-
nuclear bodies, modulating nuclear-cytoplasmic trans- 134, and miR-138, are associated with neuronal matu-
port, and controlling local protein synthesis at synapses. ration and neural network integration. Long ncRNAs also
RNA editing is a mechanism for modifying RNA mol-
influence the establishment of neural cell identity. For
ecules. Adenosine-to-inosine editing is catalyzed by aden-
example, mice lacking the Dlx6 antisense RNA 1 (Dlx6as1)
osine deaminases that act on RNA and cytidine-to-
lncRNA have reduced numbers of GABAergic interneu-
uridine editing is catalyzed by apolipoprotein B editing
rons selectively in the postnatal hippocampus and den-
catalytic subunit enzymes. Intriguingly, cytidine deami-
tate gyrus, because Dlx6as1 regulates the expression of
nases also act on DNA and are implicated in DNA demeth-
ylation pathways (see earlier). RNA editing modifies the DLX5 and DLX6, key transcription factors responsible
information content of RNA molecules and affects their for the development of GABAergic interneurons.5
intracellular dynamics. For example, RNA editing can Epigenetic mechanisms also underpin synaptic plas-
change codons in mRNAs, leading to expression of pro- ticity (long-term potentiation and depression) and learn-
tein molecules different from those encoded in the ge- ing and memory. Each epigenetic mechanism is respon-
nome. This process occurs primarily in proteins involved sible for regulating genes critical for learning and memory,
in neural excitability, such as ion channels and neurotrans- such as brain-derived neurotrophic factor and cAMP re-
mitter receptors, and is thought to fine-tune synaptic re- sponsive element-binding protein. In parallel, these
sponsiveness to environmental stimuli. RNA editing also mechanisms can themselves be deployed in response to
plays a role in controlling the subcellular localization of synaptic activity. Indeed, DNA methylation levels, his-
transcripts. RNAs subject to high levels of editing can be tone posttranslational modification profiles, ncRNA ex-
sequestered in nuclear bodies and released in response to pression patterns, and RNA editing in hippocampal neu-
specific signals including cellular differentiation, viral in- rons all vary with memory consolidation and storage.
fection, and stress. While RNA editing has been studied Animal models have also demonstrated the importance
primarily in protein-coding mRNAs, the majority of RNA of specific epigenetic factors in mediating cognitive and
editing occurs in noncoding regions. Lastly, RNA editing behavioral phenotypes. These exciting studies suggest that
is much more pronounced in the human brain than in other modulating the expression and function of epigenetic fac-
organ systems and other species, highlighting its respon- tors can modulate these processes. For example, Dnmt1
siveness to environmental stimuli. and Dnmt3a double knockout mice exhibit decreases in
DNA methylation selectively leading to abnormalities in
EVOLVING EPIGENETIC LANDSCAPE OF BRAIN plasticity in the hippocampal CA1 region and impair-
ments in learning and memory. Furthermore, the
A broad range of studies has focused on elucidating how DNMT3A2 isoform is involved in age-associated cogni-
epigenetic mechanisms together coordinate brain devel- tive decline, and upregulation of DNMT3A2 seems to re-
opment and functioning throughout the life span, includ- store cognitive function in mice.6 Similarly, abnormali-
ing neural stem cell maintenance and differentiation and ties in HDAC2-mediated acetylation of histones associated
synaptic plasticity underlying learning and memory. with genes involved in learning and memory contribute
Each epigenetic mechanism contributes to the estab- to cognitive symptoms in neurodegenerative diseases, such
lishment and maintenance of neural cell identity. In fact, as Alzheimer disease, and they can be rescued by inhib-
the appropriate expression and integrated functional iting HDAC2 activity.7

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


706

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021
PARADIGMS LINKING EPIGENETICS
WITH NEUROLOGICAL DISEASE PROCESSES Table 2. Emerging Paradigms Linking Epigenetic
Factors and Mechanisms With Diverse Neurological
Disease Processes
Here, we describe several distinct but nonmutually ex-
clusive paradigms linking epigenetic machinery and Paradigms
mechanisms with neurological disease states (Table 2
and Figure 2). Mutations in genes encoding epigenetic Mutations in genes encoding epigenetic factors cause disease
Genetic variations in genes encoding epigenetic factors modify
factors can cause neurological disease. Among the most disease risk
salient examples is the MECP2 gene, which encodes a mul- Abnormalities in epigenetic factor expression, localization, or function
tifunctional methyl-CpG-binding domain family pro- are involved in disease pathophysiology
tein. MECP2 mutations, including missense, nonsense, Epigenetic mechanisms regulate disease-associated genomic loci,
frameshift, and large deletion mutations, are the pri- gene products, and cellular pathways
Differential epigenetic profiles are present in patient-derived central
mary causes of Rett syndrome, an autism spectrum dis- and peripheral tissues
order. MECP2 mutations are also linked to a severe neo-
natal encephalopathy, syndromic forms of intellectual and
developmental disability, and an Angelman-like syn- bedded. Rather, the risk of large-vessel ischemic stroke
drome. Mutations in other epigenetic factors are simi- might be modified by the effect of this SNP on 1 or more
larly associated with neurodevelopmental disorders. For ncRNAs derived from the same locus. Indeed, it is well
example, mutations in an increasing array of genes en- documented that SNPs within disease-linked genomic loci
coding proteins that directly and indirectly regulate chro- harboring both protein-coding genes and ncRNA genes
matin structure cause syndromic and nonsyndromic forms can modify disease risk via effects on ncRNA genes. For
of intellectual and developmental disability.8 These emerg- example, CDKN2B-AS1, also known as ANRIL, is an an-
ing observations suggest that disruption of epigenetic net- tisense lncRNA transcribed from the CDKN2B-
works is one of the principal pathogenic mechanisms CDKN2A gene cluster on chromosome 9p21.3. A case-
underlying intellectual and developmental disability. Mu- control study demonstrated that SNPs at this locus,
tations in genes encoding epigenetic factors also pro- including those that preferentially influence expression
mote the onset and progression of various forms of can- of ANRIL transcripts, modify the risk of large-vessel is-
cer, including primary brain tumors. Recent studies have chemic and hemorrhagic stroke and, independently, the
detected a strong association between mutations in the risk of recurrent stroke and cardiovascular mortality.12
epigenetic machinery—particularly in factors mediat- Emerging data suggest that ANRIL interacts with other
ing histone lysine (de)methylation—and medulloblas- epigenetic regulatory factors, including PRC2, and can
toma.9 These findings are consistent with the observa- modulate gene expression programs underpinning di-
tion that the cellular functions of lysine methylation, verse cellular processes (proliferation, apoptosis, extra-
including roles in regulating cell identity, DNA repair, cellular matrix remodeling, and inflammation). Nota-
cell cycle, stress responses, and transcription, are all bly, other genome-wide association studies have found
known to be deregulated in cancer, in general, and me- that the ANRIL locus is a genomic hot spot influencing
dulloblastoma, in particular. These studies implicate many susceptibility to other disease states affecting the ner-
novel genes in medulloblastoma pathogenesis and also vous system and beyond.13 These observations highlight
correlate specific gene mutations with subgroups of tu- the interrelated nature of different epigenetic regulatory
mors with distinct histological and clinical features, sug- processes, their impact on critical cellular programs, and
gesting significant diagnostic, prognostic, and therapeu- their relevance to disease.
tic value in further characterizing these epigenetic targets. Abnormalities in epigenetic factor expression, local-
Genetic variation in genes encoding epigenetic fac- ization, or function can be involved in the molecular patho-
tors can modify the risk of neurological disease onset and physiology of neurological disease. Convergence be-
progression. For example, in patients with multiple scle- tween epigenetic factors and disease-associated pathways
rosis, single-nucleotide polymorphisms (SNPs) in 3 HDAC and proteins, including those that explicitly cause dis-
gene loci—rs2522129 (SIRT4), rs2675231 (HDAC11), and ease when mutated, results in this kind of deregulation.
rs2389963 (HDAC9)—are correlated with brain volume For example, mutations in the huntingtin protein (HTT),
measurements on neuroimaging, markers of disease se- which cause Huntington disease, lead to abnormal pro-
verity. 1 0 In another prominent example, a large files of REST expression, localization, and function. RE1-
genome-wide association study performed by the Inter- silencing transcription factor is upregulated in Hunting-
national Stroke Genetics Consortium demonstrated that ton disease and clearly contributes to the transcriptional
a SNP in an intron of the HDAC9 gene (rs11984041) on dysregulation that is a hallmark of the disease. Wild-
chromosome 7p21.1 is associated with risk of large- type HTT modulates the translocation of REST into the
vessel ischemic stroke (odds ratio, 1.38; 95% CI, neuronal cell nucleus by forming a complex with other
1.22-1.57).11 Although the precise pathogenic molecu- factors. Mutant HTT alters the conformation of this com-
lar mechanism is still unclear, HDAC9 is known to play plex, leading to aberrant nuclear accumulation of REST.14
numerous roles in developmental programs and adap- In turn, REST-regulated genes, including protein-
tive responses across many organ systems involved in the coding and ncRNA genes, are deregulated in Hunting-
pathophysiology of atherosclerosis and stroke. Given its ton disease.15 Other proteins that cause disease are simi-
intronic location, the effect of this SNP might not simply larly involved in modulating the nuclear-cytoplasmic
manifest via the protein-coding gene in which it is em- shuttling of epigenetic factors. The ataxia telangiectasia

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


707

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021
A Epigenetic gene mutations B Epigenetic gene variations

Missense Nonsense Frameshift Deletion SNP SNP

C Abnormal epigenetic factor expression, localization, and function D Epigenetic regulation of disease-associated genes and pathways

GAAn

me

me
CG CG

E Abnormal central and peripheral epigenetic profiles


CSF
AAA
Bloodstream

RISC
Lymphatics
Donor cell
Recipient cell

Figure 2. Emerging paradigms linking epigenetic factors and mechanisms with diverse neurological disease processes. A indicates adenosine; C, cytosine;
CSF, cerebrospinal fluid; G, guanine; me, methylation; RISC, RNA-induced silencing complex; and SNP, single-nucleotide polymorphism.

mutated protein indirectly regulates the subcellular lo- riety of functions including the ability to serve as a trans-
calization of HDAC4.16 Deficiency of ataxia telangiec- lational repressor for target neuronal mRNAs by inter-
tasia mutated leads to neurodegeneration as a conse- acting with select miRNAs and miRNA effector pathways
quence of HDAC4 accumulation in the neuronal cell (RNA-induced silencing complex). These pathways are
nucleus. Moreover, inhibiting HDAC4 activity or block- perturbed in fragile X syndrome and related disorders
ing its nuclear localization can rescue the pathological caused by mutations in the gene encoding FMRP. In ad-
phenotype. Intriguingly, perturbations of the subcellu- dition, disease-linked factors affect lncRNAs. TAR DNA-
lar localization of HDAC1 that occur in response to in- binding protein (TARDBP/TDP-43) is an RNA-binding
flammatory insults disrupt axonal integrity in animal mod- protein that forms inclusions in frontotemporal lobar de-
els of multiple sclerosis, and HDAC1 is mislocalized in generation and amyotrophic lateral sclerosis. Interest-
regions of demyelination in tissues derived from pa- ingly, interrogating the profiles of RNAs bound by TDP-43
tients with multiple sclerosis.17 These observations raise reveals that the most significant increases in binding in
the possibility that abnormalities in nuclear-cytoplas- frontotemporal lobar degeneration–TDP are to nuclear
mic shuttling of epigenetic factors represent a common paraspeckle assembly transcript 1 (NEAT1) and metastasis-
pathogenic mechanism for neuronal damage and loss. In associated lung adenocarcinoma transcript 1 (MALAT1;
addition to interactions with chromatin regulatory fac- also known as NEAT2).19 These lncRNAs localize to
tors, disease-associated pathways and proteins also in- nuclear bodies referred to as speckles (NEAT2) and para-
fluence ncRNAs. Mutations in leucine-rich repeat ki- speckles (NEAT1) that coordinate the execution of post-
nase 2 (LRRK2) that cause familial and sporadic forms transcriptional programs within the nuclear compart-
of Parkinson disease compromise the integrity of miRNA- ment. Speckles contain factors implicated in splicing. By
mediated translational repression, resulting in deregu- contrast, paraspeckles seem to serve as reservoirs for hy-
lation of pathways implicated in Parkinson disease patho- per adenosine-to-inosine edited RNAs that are rapidly ex-
genesis.18 Inhibiting the expression and function of let-7 ported into the cytoplasm in response to cellular stress.
and miR-184* reproduces these effects in wild-type LRRK2 The relationship between TDP-43 and these lncRNAs high-
cells and increasing let-7 and miR-184* expression lights the importance of TDP-43 for modulating post-
levels alleviates mutant LRRK2-induced pathology, sug- transcriptional RNA processing and transport in fronto-
gesting that these miRNAs are key players in LRRK2- temporal lobar degeneration and amyotrophic lateral
mediated Parkinson disease pathogenesis. Other disease- sclerosis.
associated factors, including fragile X mental retardation Epigenetic mechanisms can regulate disease-asso-
protein (FMRP), also act cooperatively with the miRNA ciated genomic loci, gene products, and cellular path-
machinery. FMRP is an RNA-binding protein with a va- ways. By definition, epigenetic mechanisms modulate the

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


708

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021
entire genome, which includes loci involved in the patho- tial levels of DNA methylation, histone and chromatin
genesis of every neurological disease with a genetic com- modifications, and ncRNA expression in a spectrum of
ponent. Imprinting is the classic example of this phenom- neurological disorders using patient-derived central and
enon. It refers to monoallelic gene expression and is peripheral tissues and animal models. Although the con-
mediated by the effects of DNA methylation, histone and tribution of these aberrant epigenetic profiles to disease
chromatin modifications, and ncRNAs that occur at the processes is sometimes unclear, deregulation of these mul-
imprinted locus in a parent-of-origin–dependent man- tilayered epigenetic processes is likely to be respon-
ner. Clinical phenotypes associated with perturbations in sible, either directly or indirectly, for disease pathophysi-
imprinting on chromosome 15q11-13 include Prader- ology by mediating cross talk between genetic
Willi and Angelman syndromes. Additionally, a rapidly in- susceptibilities and sex, environment, nutritional states,
creasing number of studies have focused on identifying and aging. These seemingly ubiquitously abnormal sig-
differential profiles of DNA methylation and histone modi- natures may, therefore, have clinical applications as in-
fications associated with disease-linked gene loci (in cen- dicators of disease risk, onset, progression, and respon-
tral and peripheral tissues) and correlating these patterns siveness to treatment. For example, the DNA methylation
with corresponding gene expression changes and clinical status of genomic repetitive elements in blood corre-
phenotypes. For example, an expansion repeat mutation lates with Alzheimer disease and Mini-Mental State Ex-
in the frataxin (FXN) gene causes Friedreich ataxia, and a amination scores, stroke and total mortality, and other
provocative study analyzed DNA methylation profiles as- clinical phenotypes. Moreover, in multiple sclerosis,
sociated with the FXN gene locus in peripheral blood mono- miRNA expression profiles have been measured in white
nuclear cells and buccal cells from patients with Fried- matter lesions, where they can discriminate between ac-
reich ataxia. It found that DNA methylation upstream of tive and inactive lesions, and blood, where they can dis-
the expansion repeat is correlated with decreased FXN criminate between relapsing-remitting and other forms
mRNA expression and increased disease severity and also of the disease and patients treated with different thera-
that DNA methylation downstream of the expansion re- pies (glatiramer acetate and natalizumab) and those who
peat is correlated with age at onset of disease.20 Comple- are untreated.23 The secretion of microvesicles, includ-
mentary studies have identified distinct profiles of his- ing exosomes, is one interesting mechanism that might
tone modifications, which likely regulate FXN expression, explain why peripheral levels of ncRNAs reflect central
flanking the mutant FXN expansion repeat in blood as well disease processes. Microvesicles are secreted by donor cells
as the brain and heart, the primary tissues affected in Fried- (neural, immune, and other cell types) into cerebrospi-
reich ataxia. Another recent study demonstrated that lev- nal fluid, lymphatics, and blood.24 Microvesicles trans-
els of an miRNA, miR-886-3p, are elevated in cell lines and fer DNA, RNA, and proteins and can actively “repro-
blood samples derived from patients with Friedreich ataxia gram” recipient cells. For example, transferred miRNAs
and that this miRNA plays a role in silencing FXN expres- can silence target mRNAs in recipient cells. The major-
sion.21 In addition, epigenetic mechanisms regulate genes ity of miRNAs found in the peripheral circulation are de-
and pathways involved in disease processes arising from rived from microvesicles, suggesting that these factors are
environmental exposures or genetic and environmental in- not simply bystanders but key effectors of dynamic central-
teractions. One study focusing on chronic pain syn- peripheral signaling. Long ncRNAs are similarly found
dromes caused by peripheral nervous system injuries dem- in microvesicles and their expression patterns are also
onstrated that C-fiber dysfunction is mediated by decreased deregulated in a range of neurological diseases,25 sug-
levels of sodium channel, voltage-gated, type X, alpha sub- gesting that interrogating central and peripheral lncRNA
unit and opioid receptor, mu 1 expression in the dorsal profiles may have diagnostic and prognostic value.
root ganglion and that REST is responsible for the down-
regulation of these genes in response to injury.22 Another PERSPECTIVES
study using animal models of chronic pain found that per-
sistent painful stimuli led to downregulation of the glu- Epigenetics is a swiftly advancing, but nascent, field. Much
tamate decarboxylase 2 gene (Gad2) in pain-modulating of the intricacy and nuance surrounding epigenetic mecha-
neurons of the brainstem nucleus raphe magnus. Hypo- nisms is yet to be defined fully. Nevertheless, epigenetics
acetylation of histones in the Gad2 gene promoter region has already revolutionized our understanding of how the
was responsible for the decreased expression of Gad2, and human brain evolved and how neural cellular diversity,
HDAC inhibition increased GAD2 protein expression and connectivity and plasticity, and advanced intellectual abili-
mitigated pain sensitization. In fact, a plethora of evi- ties emerge. There is also an evolving literature focused
dence suggests that pharmacological agents, such as HDAC on discovering whether epigenetics is involved in medi-
inhibitors, can target genes involved in disease-linked pro- ating the risk, onset, progression, and treatment respon-
cesses including apoptosis, blood-brain barrier function, siveness of neurological diseases. It is becoming clear from
cell-cycle control, DNA repair, excitotoxicity, heat shock, these studies that aberrations associated with epigenetic
inflammation, oxidative stress, and neurogenesis and glio- factors, pathways, and profiles can almost always be un-
genesis. These drugs have significant potential to modify covered, perhaps not surprisingly given the panoramic scope
disease pathogenesis and symptoms and are actively being of epigenetics. The contributions of epigenetic abnormali-
developed for these purposes. ties to neurological disease pathophysiology range from
Differential epigenetic profiles can be present in patient- being primary pathogenic mechanisms, protective re-
derived central and peripheral tissues. Indeed, a rapidly sponses, or simply passenger phenomena. Herein, we have
increasing number of studies have identified differen- presented one framework for understanding and inter-

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


709

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021
preting neurological disease mechanisms in the context 6. Oliveira AM, Hemstedt TJ, Bading H. Rescue of aging-associated decline in Dnmt3a2
expression restores cognitive abilities. Nat Neurosci. 2012;15(8):1111-1113.
of epigenetics and for promoting the development of para-
7. Gräff J, Rei D, Guan JS, et al. An epigenetic blockade of cognitive functions in
digm-shifting clinical applications. the neurodegenerating brain. Nature. 2012;483(7388):222-226.
8. van Bokhoven H. Genetic and epigenetic networks in intellectual disabilities. Annu
Accepted for Publication: October 17, 2012. Rev Genet. 2011;45:81-104.
Published Online: April 9, 2013. doi:10.1001/jamaneurol 9. Robinson G, Parker M, Kranenburg TA, et al. Novel mutations target distinct sub-
groups of medulloblastoma. Nature. 2012;488(7409):43-48.
.2013.1443 10. Inkster B, Strijbis EM, Vounou M, et al. Histone deacetylase gene variants pre-
Correspondence: Mark F. Mehler, MD, Albert Einstein dict brain volume changes in multiple sclerosis. Neurobiol Aging. 2013;34
College of Medicine, Rose F. Kennedy Center, 1410 Pel- (1):238-247.
ham Pkwy South, Room 401, Bronx, NY 10461 (mark 11. Bellenguez C, Bevan S, Gschwendtner A, et al; International Stroke Genetics Con-
.mehler@einstein.yu.edu). sortium (ISGC); Wellcome Trust Case Control Consortium 2 (WTCCC2).
Genome-wide association study identifies a variant in HDAC9 associated with
Author Contributions: Study concept and design: Qureshi large vessel ischemic stroke. Nat Genet. 2012;44(3):328-333.
and Mehler. Acquisition of data: Qureshi and Mehler. 12. Zhang W, Chen Y, Liu P, et al. Variants on chromosome 9p21.3 correlated with
Analysis and interpretation of data: Qureshi and Mehler. ANRIL expression contribute to stroke risk and recurrence in a large prospec-
Drafting of the manuscript: Qureshi and Mehler. Critical tive stroke population. Stroke. 2012;43(1):14-21.
revision of the manuscript for important intellectual con- 13. Pasmant E, Sabbagh A, Vidaud M, Bièche I. ANRIL, a long, noncoding RNA, is
an unexpected major hotspot in GWAS. FASEB J. 2011;25(2):444-448.
tent: Qureshi and Mehler. Obtained funding: Mehler. Ad- 14. Shimojo M. Huntingtin regulates RE1-silencing transcription factor/neuron-
ministrative, technical, and material support: Mehler. Study restrictive silencer factor (REST/NRSF) nuclear trafficking indirectly through a
supervision: Mehler. complex with REST/NRSF-interacting LIM domain protein (RILP) and dynactin
Conflict of Interest Disclosures: None reported. p150 Glued. J Biol Chem. 2008;283(50):34880-34886.
15. Zuccato C, Belyaev N, Conforti P, et al. Widespread disruption of repressor ele-
Funding/Support: Dr Mehler is supported by grants
ment-1 silencing transcription factor/neuron-restrictive silencer factor occu-
NS071571, HD071593, and MH66290 from the Na- pancy at its target genes in Huntington’s disease. J Neurosci. 2007;27(26):
tional Institutes of Health, as well as by the F. M. Kirby, 6972-6983.
Alpern Family, Mildred and Bernard H. Kayden, and Ro- 16. Li J, Chen J, Ricupero CL, et al. Nuclear accumulation of HDAC4 in ATM defi-
slyn and Leslie Goldstein foundations. ciency promotes neurodegeneration in ataxia telangiectasia. Nat Med. 2012;
18(5):783-790.
Additional Information: We regret that space con- 17. Kim JY, Shen S, Dietz K, et al. HDAC1 nuclear export induced by pathological
straints have prevented the citation of many relevant and conditions is essential for the onset of axonal damage. Nat Neurosci. 2010;
important references. 13(2):180-189.
18. Gehrke S, Imai Y, Sokol N, Lu B. Pathogenic LRRK2 negatively regulates microRNA-
mediated translational repression. Nature. 2010;466(7306):637-641.
REFERENCES 19. Tollervey JR, Curk T, Rogelj B, et al. Characterizing the RNA targets and position-
dependent splicing regulation by TDP-43. Nat Neurosci. 2011;14(4):452-458.
1. Lander ES, Linton LM, Birren B, et al; International Human Genome Sequencing 20. Evans-Galea MV, Carrodus N, Rowley SM, et al. FXN methylation predicts ex-
Consortium. Initial sequencing and analysis of the human genome [published pression and clinical outcome in Friedreich ataxia. Ann Neurol. 2012;71(4):
correction appears in Nature. 2001;411(6838):720]. Nature. 2001;409(6822): 487-497.
860-921. 21. Mahishi LH, Hart RP, Lynch DR, Ratan RR. miR-886-3p levels are elevated in
2. Dunham I, Kundaje A, Aldred SF, et al; ENCODE Project Consortium. An inte- Friedreich ataxia. J Neurosci. 2012;32(27):9369-9373.
grated encyclopedia of DNA elements in the human genome. Nature. 2012; 22. Uchida H, Ma L, Ueda H. Epigenetic gene silencing underlies C-fiber dysfunc-
489(7414):57-74. tions in neuropathic pain. J Neurosci. 2010;30(13):4806-4814.
3. Thurman RE, Rynes E, Humbert R, et al. The accessible chromatin landscape of 23. Thamilarasan M, Koczan D, Hecker M, Paap B, Zettl UK. MicroRNAs in multiple
the human genome. Nature. 2012;489(7414):75-82. sclerosis and experimental autoimmune encephalomyelitis. Autoimmun Rev. 2012;
4. Qureshi IA, Mehler MF. Emerging roles of non-coding RNAs in brain evolution, 11(3):174-179.
development, plasticity and disease. Nat Rev Neurosci. 2012;13(8):528-541. 24. Lai CP, Breakefield XO. Role of exosomes/microvesicles in the nervous system
5. Bond AM, Vangompel MJ, Sametsky EA, et al. Balanced gene regulation by an and use in emerging therapies. Front Physiol. 2012;3:228.
embryonic brain ncRNA is critical for adult hippocampal GABA circuitry. Nat 25. Qureshi IA, Mattick JS, Mehler MF. Long non-coding RNAs in nervous system
Neurosci. 2009;12(8):1020-1027. function and disease. Brain Res. 2010;1338:20-35.

JAMA NEUROL/ VOL 70 (NO. 6), JUNE 2013 WWW.JAMANEURO.COM


710

©2013 American Medical Association. All rights reserved.


Downloaded From: https://jamanetwork.com/ by a PUC/RS User on 12/26/2021

You might also like