Epigenetic regulation of chronic pain - Review - Megat Pharmaceutical

Page created by Brent Marquez
 
CONTINUE READING
Epigenetic regulation of chronic pain - Review - Megat Pharmaceutical
Review
For reprint orders, please contact: reprints@futuremedicine.com

Epigenetic regulation of chronic pain

Chronic pain arising from peripheral inflammation and tissue or nerve injury is a common              Lingli Liang1, Brianna Marie
clinical symptom. Although intensive research on the neurobiological mechanisms                       Lutz2, Alex Bekker1
of chronic pain has been carried out during previous decades, this disorder is still                  & Yuan-Xiang Tao*,1,3,4,5
                                                                                                      1
                                                                                                       Department of Anesthesiology, New
poorly managed by current drugs such as opioids and nonsteroidal anti-inflammatory
                                                                                                      Jersey Medical School, Rutgers, The State
drugs. Inflammation, tissue injury and/or nerve injury-induced changes in gene                        University of New Jersey, 185 S. Orange
expression in sensory neurons of the dorsal root ganglion, spinal cord dorsal horn                    Avenue, MSB F-548, Newark,
and pain-associated brain regions are thought to participate in chronic pain genesis;                 NJ 07103, USA
however, how these changes occur is still elusive. Epigenetic modifications including
                                                                                                      2
                                                                                                        Rutgers Graduate School of Biomedical
                                                                                                      Sciences, New Jersey Medical School,
DNA methylation and covalent histone modifications control gene expression. Recent
                                                                                                      Rutgers, The State University of New
studies have shown that peripheral noxious stimulation changes DNA methylation                        Jersey, Newark, NJ 07103, USA
and histone modifications and that these changes may be related to the induction                      3
                                                                                                        Department of Cell Biology & Molecular
of pain hypersensitivity under chronic pain conditions. This review summarizes the                    Medicine, New Jersey Medical School,
current knowledge and progress in epigenetic research in chronic pain and discusses                   Rutgers, The State University of New
                                                                                                      Jersey, Newark, NJ 07103, USA
the potential role of epigenetic modifications as therapeutic antinociceptive targets                 4
                                                                                                        Department of Neurology &
in this disorder.                                                                                     Neuroscience, New Jersey Medical
                                                                                                      School, Rutgers, The State University of
Keywords: chronic pain • DNA methylation • dorsal root ganglion • histone acetylation                 New Jersey, Newark, NJ 07103, USA
• histone methylation • inflammatory pain • neuropathic pain • spinal cord                            5
                                                                                                        Department of Physiology &
                                                                                                      Pharmacology, New Jersey Medical
                                                                                                      School, Rutgers, The State University of
Chronic pain is a major public health prob-         rotransmitters, neuromodulators and struc-        New Jersey, Newark, NJ 07103, USA
lem that affects approximately 30% of the           tural proteins in primary sensory neurons of      *Author for correspondence:
general population in the USA. It is a cause of     dorsal root ganglion (DRG), spinal cord and       Tel.: +1 973 972 9812
                                                                                                      Fax: +1 973 972 1644
grave physiological and psychological distress      other pain-related regions in the brain [2–4] .   yt211@ njms.rutgers.edu
in those affected, and it places significant        These changes contribute to the induction
pressures on the healthcare system. About           and maintenance of chronic pain; however,
100 billion US dollars are spent on chronic         how these changes are regulated by peripheral
pain-related healthcare expenses, and many          noxious stimuli is still not fully understood.
patients experience a loss of productivity [1] .       Recent studies have suggested that the
Chronic pain usually arises from inflam-            mechanism for gene regulation involves
mation or tissue and nerve injury. Although         epigenetic modifications. Environmental
intensive research on the neurobiological           toxins, medications, diet and psychological
mechanisms of chronic pain has been carried         stress alter epigenetic processes such as DNA
out during previous decades, this disorder is       methylation, covalent histone modifica-
still poorly managed by current drugs such as       tion (e.g., acetylation and methylation), and
opioids and nonsteroidal anti-inflammatory          noncoding RNA expression. Accumulating
drugs, which are ineffective and/or produce         evidence demonstrates that these processes
severe side effects [2] . Peripheral inflamma-      play an important role in synaptic plastic-
tion and nerve injury produce transcriptional       ity during memory formation as epigenetic
and translational changes in the expression         changes correlate with hippocampal activ-
                                                                                                                           part of
of receptors, enzymes, ion channels, neu-           ity [5–10] . Given that peripheral and central

10.2217/EPI.14.75 © 2015 Future Medicine Ltd       Epigenomics (2015) 7(2), 235–245                   ISSN 1750-1911                      235
Review   Liang, Lutz, Bekker & Tao

               sensitization under chronic pain conditions share           promotes gene transcription. In contrast, histone
               common mechanisms with the neuronal plasticity of           deacetylation tightly condenses chromatin resulting in
               memory formation, it is very likely that similar epigen-    gene silencing (Figure 1) [16] .
               etic mechanisms occur under both conditions. Indeed,
               peripheral inflammation and nerve injury drive              Effect of HDAC inhibitors on inflammatory pain
               changes in DNA methylation, histone modifications           Evidence from a pharmacological study has shown that
               and noncoding RNAs in pain-related regions [8,9,11–14] .    HDAC inhibitors can relieve inflammatory pain [20–22] .
               These changes might be responsible for inflammation/        A 5-day subcutaneous treatment with either of the two
               nerve injury-induced alterations of some pain-associ-       HDAC inhibitors, MS-275 and SAHA, substantially
               ated genes in central neurons. The evidence suggests        reduced nociceptive behaviors in the second phase of
               that modification of epigenetic processes participates      the formalin test and led to an increase in mGluR2
               in the mechanisms that underlie the induction and           (but not mGluR1a, mGluR4 or mGluR5) in the DRG
               maintenance of chronic pain.                                (Table 1) [21] . This antinociception could be abrogated
                  The role of noncoding RNAs including microR-             by an mGlu2/3 receptor antagonist. The induction
               NAs and long noncoding RNAs in chronic pain has             of DRG mGlu2 receptors in response to suberoyl-
               recently been discussed [8] . This article focuses on the   anilide hydroxamic acid (SAHA) was associated with
               evidence for the changes in DNA methylation and             increased acetylation of p65/RelA on lysine 310, a
               histone modification, mostly in DRG and spinal cord,        process that enhances the transcriptional activity of
               under chronic pain conditions. We explore how these         p65/RelA at NF-κB-regulated genes [21] . Given that
               changes are induced by peripheral noxious stimuli and       transcription of the mGlu2 receptor gene is activated
               how these epigenetic processes regulate pain-related        by p65/RelA in DRG neurons, HDAC inhibition
               genes. We finally deduce potential mechanisms of how        may produce antinociception by upregulating mGlu2
               the changes in DNA methylation and histone modifi-          receptor expression in DRG. However, it is not clear
               cation contribute to the development and maintenance        whether formalin injection changes the histone acety-
               of chronic pain.                                            lation conditions and HDAC expression and activity
                                                                           in the DRG.
               Histone modification in chronic pain                           It was reported that complete Freund’s adjuvant
               The process of histone modification                         (CFA)-induced peripheral inflammation increased
               The nucleosome is the basic unit of chromatin,              the levels of class IIa HDAC members (HDAC4, 5,
               composed of about 140 base pairs of DNA wrapped             7, 9), but not class I HDAC members (HDAC1, 2, 3),
               around a histone octamer. Histones are small, alka-         in the spinal dorsal horn [20] . Intrathecal administra-
               line proteins categorized into five major families: H1/     tion of HDAC inhibitors targeting class II (SAHA,
               H5, H2A, H2B, H3 and H4. Histones H2A, H2B,                 TSA, LAQ824) or IIa (VPA, 4-PB) significantly
               H3 and H4 are known as the core histones, while             delayed the development of thermal hyperalgesia and
               histones H1 and H5 are known as linker histones.            attenuated existing thermal hyperalgesia in a CFA-
               The N-terminal histone tail protrudes from the              induced inflammatory pain model (Table 1) [20] . It
               nucleosome and can be post-translationally modi-            appears that class I and II or IIa HDAC members
               fied, including acetylation, methylation, phosphory-        function differentially in inflammatory pain models.
               lation, citrullination, SUMOylation, ubiquitination         A recent investigation from Zhang and colleagues
               and ADP-ribosylation [15] . These modifications result      revealed the epigenetic mechanism of inflamma-
               in changes in the 3D chromatin structure and gene           tory pain in central pain-modulating neurons. CFA
               expression [15] .                                           induced the increase of global histone H3 and H4
                                                                           acetylation in brainstem nucleus raphe magnus
               Histone acetylation & deacetylation in                      (NRM), a crucial supraspinal site for maintenance
               chronic pain                                                of pain hypersensitivity [22] . However, acetylated H3
               Histone acetylation and deacetylation are the processes     was reduced in the Gad2 gene promoter region which
               by which the histones on lysine residues within the         epigenetically suppresses the transcription of Gad2
               N-terminal tail and on the surface of the nucleosome        (encoding glutamic acid decarboxylase 65) and con-
               core are acetylated by histone acetyltransferase (HAT)      sequently causes impaired inhibitory function. Local
               or deacetylated by histone deacetylases (HDACs) [16] .      injection of HDAC inhibitors Trichostatin A (TSA)
               Acetyl-Coenzyme A is the major source of the acetyl         and SAHA into nucleus raphe magnus reversed
               group in histone acetylation [16–19] . Conventionally,      this effect and produced a similar analgesic effect
               histone acetylation makes the condensed chroma-             on CFA-induced inflammatory pain using systemic
               tin into a more relaxed structure, and consequently         administration (Table 1) [22] .

236            Epigenomics (2015) 7(2)                                                                         future science group
Epigenetic regulation of chronic pain   Review

     A
                                           Condensed chromatin: transcription repression

                                 Histone
                                                                                    Histone tail

                             Me                     Me                 Me                     HDAC

                                                                                                             DNA
                      Me
                                                 Me                                           Me

                                                               Promoter regions

     B
                                              Loose chromatin: transcription activation

                           HAT
                                                                 HAT                                 HAT
                           Ac
                                                                  Ac                                 Ac
                                             Promoter region

                                               TF
                                                                                                                         DNA

Figure 1. Histone modification regulates gene expression. (A) Methylation of histones and deacetylation of histones
with HDAC results in a condensed chromatin. Under this condition, TFs cannot bind to the promoter region of the
gene, thereby gene transcription is repressed. (B) Histone acetylation with HAT results in loose chromatin that allows
the TF to bind to the promoter region of the gene. Consequently, gene transcription is activated.
Ac: Acetylation; HAT: Histone acetyltransferase; HDAC: Histone deacetylase; Me: Methylation; TF: Transcription factor.

Effect of HDAC inhibitors on visceral pain                             sciatic nerve (Table 1) [24] . However, how HDAC inhi-
One study on visceral pain also provided evidence to                   bition affects TNF-α expression under CCI-induced
support the involvement of central epigenetic mecha-                   neuropathic pain conditions is unknown. Intrathecal
nisms in pain [23] . Intracerebroventricular adminis-                  pretreatment with class I HDAC inhibitors (MS-275
tration of TSA significantly attenuated water avoid-                   or MGDC0103) attenuated mechanical and thermal
ance stress-induced visceral hypersensitivity in rats                  hypersensitivity in models of traumatic nerve injury
(Table 1) [23] . However, the targets of histone acetylation           and antiretroviral drug (stavudine)-induced periph-
were not known in this study.                                          eral neuropathy (Table 1) [25] . This analgesic effect
                                                                       may be related to the increase in global H3K9ac in
Effects of HAT inhibitors & HDAC inhibitors on                         the spinal cord but not in DRG, suggesting that any
neuropathic pain                                                       potential mechanism could be found in the CNS [25] .
Several groups reported that the HDAC inhibitors                       Interestingly, the acetylation changes at the promot-
had an antinociceptive effect in neuropathic pain.                     ers of some pain-related genes, like μ opioid receptor,
Oral administration of sodium butyrate, an HDAC                        Kv4.3, Nav1.8, and brain-derived neurotrophic factor
inhibitor, not only attenuated chronic constriction                    (BDNF), in DRG neurons have been reported in a
injury (CCI)-induced pain hypersensitivity but also                    neuropathic pain model (Table 1) [26 -29] . Nerve injury-
reduced the CCI-induced increase in TNF-α in the                       induced reductions of histone H3 and H4 acetylation

    future science group                                                                                   www.futuremedicine.com             237
Review        Liang, Lutz, Bekker & Tao

                      at the promoter regions of μ receptor, Nav1.8, and                    symptoms associated with neuropathic pain [27,28] ,
                      Kv4.3 silence their expression in DRG and may rep-                    whereas nerve injury-promoted increases in histone H3
                      resent an underlying cause of common negative                         and H4 acetylation at the promoter regions of BDNF

 Table 1. Summary of studies on histone acetylation and deacetylation.
Pain model                     Changes of                 Tissue           Inhibitors                 Nociceptive behavior            Target              Ref.
                               acetylation or                                                         response to inhibitors          genes
                               enzyme expression
Formalin                       NA                         DRG              MS-275, SAHA (sc.)         Second phase (↓)                mGluR2/3                 [21]

CFA                            HDAC4, 5, 7, 9 (↑),        Spinal      SAHA, TSA, LAQ824, Thermal (↓)                                  NA                       [20]
                               HDAC1, 2, 3 (-)            Dorsal horn VPA, 4-PB (it.)
                               Global histone H3          NRM              TSA and SAHA               Thermal (↓)                     GAD65                    [22]
                               and H4 acetylation                          (NRM)
                               (↑)
Water avoidance stress NA                                 NA               TSA (icv.)                 Visceral                        NA                       [23]
                                                                                                      hypersensitivity(↓)
CCI                            NA                         Sciatic          Sodium                     Thermal (↓),                    TNF-α                    [24]
                                                          nerve            butyrate(oral)             mechanical (↓), cold(↓)
Traumatic nerve injury Global H3K9ac(↑)                   Spinal cord MS-275, MS-275 or               Thermal (↓),                    NA                       [25]
and stavudine-induced                                                 MGDC0103 (it.)                  mechanical (↓)
peripheral neuropathy
SNL                            HDAC1(↑) Acetyl            Spinal      Baicalin                        Thermal (↓),                                             [41]
                               H3 (↓)                     dorsal horn                                 mechanical (↓)
                               Acetylation changes DRG                     NA                         NA                              μ receptor, [27–29]
                               on promoter of                                                                                         Nav1.8,
                               genes                                                                                                  Kv4.3, BDNF
                               H3K9 ac(↑) on              Injured          Anacardic acid (ip.)       Thermal (↓),                    MIP-2 and          [31,32]
                               promoter of MIP-2          sciatic                                     mechanical (↓)                  its receptor
                               and CXCR2                  nerve                                                                       CXCR2
CCI                            p300 (↑)                   Spinal cord p300 shRNA or                   Thermal (↓),                    COX-2              [33,34]
                                                                      C646 (it.)                      mechanical (↓)
                               H4 acetylation at          Spinal cord                                                                 Cdk5                     [30]
                               Cdk5 promoter (↑)
                               Sirt (↓), Acetyl H3(↑) Spinal cord Resveratrol                         Thermal (↓),                    NA                       [35]
                                                                                                      mechanical (↓)
Incision                       NA                         NA               Anacardic acid (ip.)       Mechanical (↓), thermal NA                         [36,37]
                                                                                                      (-)
                                                                           SAHA                       Mechanical (↓), thermal NA
                                                                                                      (-)
Morphine                       Sirt (↓), Acetyl H3(↑) Spinal cord Resveratrol                         ↓Tolerance                      NA                       [40]

                               NA                         NA               Curcumin                   ↓mechanical allodynia,          NA                       [39]
                                                                                                      thermal hyperalgesia,
                                                                                                      tolerance, and physical
                                                                                                      dependence
                                                                           SAHA                       ↑mechanical allodynia,          NA
                                                                                                      thermal hyperalgesia,
                                                                                                      tolerance, and physical
                                                                                                      dependence
BDNF: Brain-derived neurotrophic factor; CCI: Chronic constriction injury; CFA: Complete Freund’s adjuvant; COX-2: Cyclooxygenase-2; CXCR2: Chemokine CC motif
receptor 2; DRG: Dorsal root ganglion; HDAC: Histone deacetylase; icv.: Intracerebroventricular injection; ip.: Intraperitoneal injection; it.: Intrathecally;
MIP-2: Macrophage inflammatory protein 2; NA: Not applicable; NRM: Nucleus raphe magnus; SAHA: Suberoylanilide hydroxamic acid; sc.: Subcutaneous injection;
SNL: Spinal nerve ligation, TSA: Trichostatin A; VPA: Valproic acid.

238                   Epigenomics (2015) 7(2)                                                                                           future science group
Epigenetic regulation of chronic pain   Review

in DRG and in histone H4 acetylation at the promoter            Given that the degree of histone acetylation is con-
regions of Cdk5 in spinal cord upregulates the expres-       trolled by the enzymes HATs and HDACs, conven-
sion of BDNF and CdK5, respectively, and may con-            tionally, HAT inhibitors or HDAC activators should
tribute to the induction or maintenance of neuropathic       have opposite effects compared with HDAC inhibi-
pain (Table 1) [29,30] .                                     tors. Based on previous observations described above,
   Interestingly, data from other groups showed that         how HATs and HDACs are involved in neuropathic
HAT inhibitors also had an antinociceptive effect in         pain is still elusive. The role of histone acetylation
neuropathic pain. The HAT inhibitor anacardic acid           and deacetylation in neuropathic pain remains to be
relieved spinal nerve ligation (SNL)-induced neuro-          verified.
pathic pain by suppressing the hyperacetylation of
histone H3 in the promoter region of macrophage              Histone methylation & demethylation in
inflammatory protein 2 (MIP-2) and its receptor che-         chronic pain
mokine CC motif receptor 2, resulting in the blockade        Histone methylation is another process of histone
of SNL-induced upregulation of MIP-2 and chemo-              modification by which methyl groups are transferred
kine CC motif receptor 2 in the injured sciatic nerve        to amino acids of histone proteins in chromosomes
(Table 1) [31,32] . In another report, CCI increased the     (Figure 1) . Histone methylation is catalyzed by S-ade-
expression of p300, a HAT E1A binding protein, in the        nosylmethionine-dependent histone lysine methyl-
lumbar spinal cord (Table 1) [33,34] . Intrathecal admin-    transferases and protein arginine methyltransferases,
istration of p300 shRNA or an inhibitor of p300 HAT          whereas histone demethylation is catalyzed by histone
reversed CCI-induced mechanical allodynia and thermal        N-methylated lysine residue demethylases and the pep-
hyperalgesia and suppressed the expression of cyclooxy-      tidyl arginine deiminases [15,42] . Histone methylation
genase-2 (COX-2) in spinal cord (Table 1) [33,34] . Con-     could repress or activate gene transcription depending
sistently, the intrathecal administration of resveratrol,    on the sites and content being methylated. In general,
an activator of Sirt1 (a classic III HDAC), attenuated       methylation of histone H3 at Lys9 or Lys27 (H3K9 or
CCI-induced mechanical allodynia and thermal hyper-          H3K27) or histone H4 at Lys20 (H4K20) correlates
algesia, reversed the CCI-induced decrease in spinal         with transcriptional repression, whereas methylation of
Sirt1, and blocked the CCI-induced increase in spinal        H3K4, H3K36 and H3K79 correlates with enhanced
histone H3 acetylation [35] . The analgesic effect of HAT    transcription [15] .
inhibition was also reported in other persistent pain           Although histone methylation has been reported
conditions. In an incision model, injection of the HAT       to participate in the mechanism of formation of long-
inhibitor anacardic acid intraperitoneally reduced inci-     term memories and learning [7,43–45] , the role of histone
sion-induced pain hypersensitivity [36,37] . As expected,    methylation in chronic pain is still unclear. Evidence
the HDAC inhibitor SAHA exacerbated mechanical               indicates that histone methylation may be related to the
hypersensitivity after incision (Table 1) [36,37] . Given    expression of chemokine (CC motif) ligands (CCLs),
that neuropathic pain and opioid tolerance/opioid-           a class of small cytokines, in neuropathic pain [32,46] .
induced hyperalgesia share some common intracellular         The peripheral nerve injury-induced reduction in
pathways in their mechanisms [38] , the evidence sug-        H3K27me3 in the promoter region of monocyte che-
gests that histone modification is also involved in the      motactic protein 3 (MCP-3, known as CCL7) might
development and maintenance of opioid tolerance and          be responsible for the nerve injury-induced increase in
opioid-induced hyperalgesia. Indeed, daily administra-       the expression of MCP-3 in spinal cord [46] . IL-6 may
tion of the HAT inhibitor curcumin with morphine for         be involved in this response as the increased MCP-3
4 days reduced the development of morphine-induced           expression was almost abolished in IL-6 knockout mice
mechanical allodynia, thermal hyperalgesia, tolerance        with partial sciatic nerve ligation [46] . Peripheral nerve
and physical dependence [39] . Conversely, the HDAC          injury also increased the mRNA levels of CCL2, CCL3
inhibitor SAHA enhanced these responses (Table 1) [39] .     and their receptors (CCR2 and CCR1/CCR5, respec-
The intrathecal injection of resveratrol suppressed the      tively) in the injured sciatic nerve. These increases
established morphine analgesic tolerance, reversed the       could be related to the increased H3K4me3 in the pro-
morphine-induced decrease in spinal Sirt1, and attenu-       moter regions of these cytokine genes [32] . An increase
ated the morphine-induced increase in spinal histone         in global histone methylation was also observed in
H3 acetylation [40] . Interestingly, baicalin, a flavonoid   spinal cord after intrathecal injection of pertussis
compound isolated from Huang Qin, ameliorated                toxin, which induced significant thermal hyperalge-
SNL-induced neuropathic pain by suppressing HDAC1            sia [47] . However, whether these methylation sites are
expression and preventing histone-H3 acetylation in the      really required for these changes in gene expression is
spinal cord dorsal horn (Table 1) [41] .                     unknown. Furthermore, whether nerve injury-induced

    future science group                                                                        www.futuremedicine.com             239
Review   Liang, Lutz, Bekker & Tao

               changes in histone methylation contribute to neuro-         DNA methytransferases & DNA methylation in
               pathic pain remains to be investigated. A recent study      chronic pain
               showed that an increase in the expression of methyl-        Effect of the DNMT inhibitors on
               CpG-binding protein 2 (MeCP2) in mouse central              inflammatory pain
               nucleus of the amygdala (CeA) was caused by both            Although the function of DNA methylation has been
               CFA-induced chronic inflammatory pain and repeated          reported in other pathological states, so far, only a few
               morphine exposure [48] . The increased MeCP2 bound          studies have demonstrated the potential role of DNA
               to and repressed the transcriptional repressor histone      methylation and the activity and expressional levels of
               dimethyltransferase G9a, resulting in a reduction in        DNMTs in pain. Cystathionine-β-synthase (Cbs) syn-
               G9a-catalyzed repressive marker H3K9me2 and an              thesizes hydrogen sulfide, an endogenous gas molecule,
               increase in the expression of BDNF in CeA [48] . Over-      which is necessary and sufficient to elicit mechanical
               expression of CeA MeCP2 or knockdown of CeA                 pain hypersensitivity and increased excitability of DRG
               G9a facilitated behavior of morphine reward, whereas        neurons. Peripheral inflammation induced by CFA
               knockdown of CeA MeCP2 inhibited behavior of mor-           leads to demethylation of the cystathionine-β-synthase
               phine reward [48] . Whether such epigenetic cascade         (cbs) gene in DRG [58] . This demethylation may be
               occurs in neuropathic pain needs to be confirmed.           associated with the CFA-induced increase in expression
                                                                           of Cbs mRNA and protein in DRG and could influ-
               DNA methylation & chronic pain                              ence the induction of inflammation-induced mechani-
               Process of DNA methylation                                  cal hypersensitivity [58] . Given the fact that peripheral
               In mammalian cells, DNA methylation is a biochemi-          inflammation did not decrease DNMT expression and
               cal process, in which a methyl group is added to the        activity in DRG [12] , it is unclear how demethylation
               fifth carbon of cytosine residues situated adjacent to      occurs in DRG under chronic inflammatory pain con-
               a guanine residue (CpG site). DNA sequences with            ditions. The level of DNA methylation is controlled by
               a high concentration of CpG residues are referred to        both DNMTs and demethylation enzymes (e.g., ten-
               as CpG islands and are generally located at the start       eleven translocation dioxygenases). Whether periph-
               of the gene sequence within the promoter region             eral inflammation changes the expression and activity
               (Figure 2) . The process of DNA methylation is medi-        of DNA demethylation enzymes in DRG remains to
               ated by a group of DNA methyltransferases (DNMTs)           be determined. Interestingly, a recent study reported
               that includes DNMT1, DNMT3a and DNMT3b;                     the CFA-induced hypermethylation of CpG islands in
               this protein family also includes DNMT2, an inacti-         the miR-129 promoter in spinal cord neurons [59] . This
               vated isoform, and DNMT3L, which lacks the con-             methylation may regulate chronic inflammatory pain
               served catalytic domain [49–51] . DNMT1 maintains the       by targeting CaMKIIγ [59] .
               methylation of DNA that is already established at the
               genome and is considered to be the primary mainte-          Effect of the DNMT inhibitors on
               nance DNMT [50,51] . Both DNMT3a and DNMT3b                 neuropathic pain
               act as de novo methyltransferases and methylate             In addition to peripheral inflammation, peripheral
               unmethylated DNA [50,51] . Evidence has now expanded        nerve injury caused by sciatic nerve CCI increased the
               the role of DNMT1 to also include facilitation of de        level of global DNA methylation in the spinal cord [60] .
               novo DNA methylation by DNMT3a and DNMT3b                   Blocking spinal cord DNA methylation with intrathe-
               at gene promoters [6,50,52–54] .                            cal 5-azacytidine attenuated CCI-induced thermal
                  DNA methylation interferes with gene transcrip-          and mechanical pain hypersensitivities [60] . CCI also
               tion by physically interfering with the binding of          increased the level of DNA methylation in the proxi-
               transcription factors and serving as docking sites          mal promoter region of the μ opioid receptor gene in
               for methyl-CpG-binding domain proteins (MBDs)               DRG [61] . This increase may be related to the CCI-
               (Figure 2) [55,56] . MBDs contain a specific domain of      induced decrease in the analgesic effect of opioids [61] .
               approximately 70 residues, the methyl-CpG-binding           In the spared nerve injury-induced neuropathic pain
               domain, which directly binds to one or more meth-           model, DNMT1 and DNMT3a (but not DNMT3b)
               ylated CpGs of a gene promoter. MBDs function               transcripts were upregulated in the injured DRGs [62] .
               as docking sites, in which they recruit other tran-         Interestingly, the level of global DNA methylation was
               scriptional corepressors, such as HDACs, to the tar-        reduced in the prefrontal cortex and amygdala (but
               geted gene for gene silencing, or coactivators, such as     not the visual cortex and thalamus) following spared
               CREB1, for transcription activation [55–57] . The ability   nerve injury [63] . This reduction strongly correlated
               to recruit these proteins may be why DNA methylation        with the severity of pain behaviors [63] . It appears that
               has such a profound effect on gene expression.              nerve injury-induced changes in DNA methylation are

240            Epigenomics (2015) 7(2)                                                                          future science group
Epigenetic regulation of chronic pain   Review

   A                                                                    TSS    Transcriptional
                                                                               activation
                                     TF     RNAPII

                            CpG           CpG         CpG                         Gene

   B                                       DNMT

                                          MBD                           TSS    Transcriptional
                                                                               repression
                           Me        Me      Me

                           CpG            CpG        CpG                          Gene
                                Me         Me

Figure 2. DNA methylation represses gene transcriptional processes. (A) Without methylation at the CpG sites of
the gene promoter, the transcription factor and RNAPII bind to the promoter region of the gene, thereby gene
transcription is activated. (B) When methyl groups are added at the CpG islands by methyl-CpG-binding domain
protein-mediated DNA methyltransferases, the TF and RNAPII cannot bind the promoter region of the gene,
resulting in the repression of gene transcription.
CpG: -C-phosphate-G-; Me: Methylation; TF: Transcription factor; TSS: Transcription start site.

spatially different in the CNS and are implicated in dis-   gene promoter was reported in patients experiencing
tinct functions in spinal and supraspinal levels under      chronic low back pain associated with disc degenera-
neuropathic pain conditions. However, which type            tion [65] . The endothelin B receptor gene promoter was
of cells in the nervous system expresses these changes      heavily methylated in human oral squamous cell car-
is elusive. A recent study reported that DNMT1 was          cinoma lesions, which are highly painful, whereas this
found in both neurons and satellite glial cells of DRG,     promoter was not methylated in human oral dysplasia
DNMT3a in DRG satellite glial cells and DNMT3b in           lesions, which are typically not painful [66] . A regula-
DRG neurons [62] , but the conclusion remains uncer-        tory DNA methylation region in the CpG-island shore
tain because no specific neuronal and glial markers         of the TRPA1 promoter was reported to have a pos-
were used [62] . Additionally, the specificity and selec-   sible impact on TRPA1 gene expression and thermal
tivity of the antibodies used were not addressed [62] .     sensitivity [67] . Joint resident synovial fibroblasts from
These earlier studies raise several unanswered ques-        patients with rheumatoid arthritis exhibited a global
tions. For example, is the expression and/or activity       hypomethylation or both hypomethylation and hyper-
of DNMTs and demethylation enzymes spatially and            methylation patterns compared with patients with
temporally changed following peripheral inflammation        osteoarthritis or healthy controls [68–70] . This hypo-
or nerve injury? If so, which type of DNMT? What            methylation was identified in key genes relevant for
are the downstream targeted genes of DNMTs under            rheumatoid arthritis, related to multiple pathways,
chronic pain conditions? Do DNMT inhibitors lead            and associated with increased gene expression [70] . A
to side effects in addition to antinociception given that   reduction of DNMT1 and an increase in the expres-
they are pharmacologically nonselective for specific        sion of S-adenosyl methionine decarboxylase, spermi-
DNMTs? Broader future investigations are required.          dine/spermine N1-acetyltransferase and polya-mine-
                                                            modulated factor1-binding protein1 may be associated
DNA methylation in patients with chronic pain               with hypomethylation in rheumatoid arthritis [68,71] .
Alterations of DNA methylation have also been observed      Additionally, promoter methylation states of the death
in patients with painful diseases. Women with fibro-        receptor 3, IL-6, IL-10, IL-R2 and chemokine ligand
myalgia showed significant differences in DNA meth-         12 genes were altered in blood mononuclear cells and
ylation patterns compared with age-matched healthy          synovial fibroblasts in rheumatoid arthritis [42,72] . It
controls, when genomic DNA isolated from whole              appears that DNA methylation has the potential to
blood was examined [64] . Fibromyalgia-associated           serve as a biomarker for some types of painful disorders
genes with differential methylation include BDNF,           (e.g., autoimmune disorders or inflammation).
histone deacetylase 4, N-Acetyltransferase 15, protein
kinase C alpha and protein kinase G1 [64] . Increased       MBDs in chronic pain
methylation at the extracellular matrix protein             As discussed above, DNA methylation-triggered gene
SPARC (Secreted Protein, Acidic, Rich in Cysteine)          transcriptional changes require a family of MBDs. The

    future science group                                                                         www.futuremedicine.com             241
Review   Liang, Lutz, Bekker & Tao

               MBD family is composed of MeCP2 and MBD1–4.                        depends on the behavioral observations following phar-
               Each of these proteins, with the exception of MBD3, is             macological inhibitor administration. These inhibitors
               capable of binding specifically to methylated DNA [73] .           have varying degrees of specificity and selectivity for
               Accumulating evidence indicates that MeCp2 may be                  the corresponding epigenetic enzymes. They may also
               related to chronic pain. MeCP2 is associated closely               exert their effects through nonepigenetic mechanisms,
               with Rett syndrome, a neurodevelopmental disorder,                 resulting in potential side effects. For example, HATs
               which is primarily caused by mutations in the MeCP2                and HDACs are not histone specific and can also
               locus and patients display decreased pain sensitiv-                acetylate and deacetylate, respectively, other targets in
               ity [74,75] . Preclinical studies showed that the expres-          the cytoplasm. These factors call for careful interpreta-
               sion of MeCP2 and the level of its phosphorylation                 tion of current findings claiming the role of a particu-
               were increased in the superficial dorsal horn under                lar epigenetic enzyme in chronic pain. Therefore, the
               CFA-induced inflammatory pain conditions [76,77] .                 development of pharmacologic inhibitors for isoform
               This phosphorylation is controlled by a descending                 or subtype-specific epigenetic enzymes and/or the use
               serotonergic pathway as serotonergic depletion pre-                of targeted genetic inhibition of isoform or subtype-
               vented CFA-induced MeCP2 phosphorylation [76] . It                 specific epigenetic enzymes will be required. More-
               has been demonstrated that once MeCP2 is phosphor-                 over, whether these epigenetic enzymes are activated
               ylated, it can be dissociated from the promoter regions            by peripheral noxious insults and how their activation
               of genes that have been repressed [78] . Therefore, the            contributes to chronic pain remain to be investigated.
               CFA-induced increase in MeCP2 phosphorylation                      Given that chronic pain remains a challenging condi-
               may facilitate gene expression in the superficial dorsal           tion to manage and that the contribution of epigen-
               horn under inflammatory pain conditions. However,                  etic mechanisms underlying this disorder is becoming
               the significance of how this increased phosphorylation             increasingly recognized, it is conceivable that the sig-
               is related to descending inhibitory serotonergic func-             nificance of histone modification and DNA methyla-
               tion in this model is unclear. The changes in MeCP2                tion in chronic pain will become even more apparent
               expression following peripheral nerve injury are incon-            in the coming years.
               sistent. An increase in MeCP2 expression was observed
               in the spinal cord of CCI rats [60] , whereas a decrease           Financial & competing interests disclosure
               was detected in the superficial dorsal horn of rats                This work was supported by grants from the NIH (NS072206,
               after spared nerve injury [77] . The role of MeCP2 in              HL117684 and DA033390) and the Rita Allen Foundation. The
               neuropathic pain remains to be further clarified.                  authors have no other relevant affiliations or financial involve-
                                                                                  ment with any organization or entity with a financial inter-
               Conclusion & future perspective                                    est in or financial conflict with the subject matter or materials
               The evidence described above suggests that histone                 discussed in the manuscript apart from those disclosed.
               modifications and DNA methylation in DRG and spi-                     No writing assistance was utilized in the production of this
               nal cord are involved in chronic pain. The conclusion              manuscript.

                Exclusive summary
                Histone modification in chronic pain
                • Histone deacetylase inhibitors reduce inflammatory pain and visceral pain.
                • Both histone acetyltransferase inhibitors and histone deacetylase inhibitors show antinociceptive effects in
                  neuropathic pain.
                • Peripheral nerve injury or inflammation alters histone methylation in the promoter regions of some pain-
                  related genes.
                DNA methylation & chronic pain
                • DNA methyltransferase inhibitors blocks inflammatory pain.
                • DNA methyltransferase inhibitors attenuates neuropathic pain.
                • Alterations of DNA methylation have been found in patients with some painful diseases.

               References                                                         2    Latremoliere A, Woolf CJ. Central sensitization: a generator
               Papers of special note have been highlighted as:                        of pain hypersensitivity by central neural plasticity. J. Pain
               • of interest; •• of considerable interest                              10(9), 895–926 (2009).
               1    Morley S. Psychology of pain. Br. J. Anaesth. 101(1), 25–31   3    Campbell JN, Meyer RA. Mechanisms of neuropathic pain.
                    (2008).                                                            Neuron 52(1), 77–92 (2006).

242            Epigenomics (2015) 7(2)                                                                                        future science group
Epigenetic regulation of chronic pain   Review

4    Wang W, Gu J, Li YQ, Tao YX. Are voltage-gated sodium           22   Zhang Z, Cai YQ, Zou F, Bie B, Pan ZZ. Epigenetic
     channels on the dorsal root ganglion involved in the                 suppression of GAD65 expression mediates persistent pain.
     development of neuropathic pain? Mol. Pain 7, 16 (2011).             Nat. Med. 17(11), 1448–1455 (2011).
5    Bali P, Im HI, Kenny PJ. Methylation, memory and                •    Recognized the significant role of histone modefication in
     addiction. Epigenetics 6(6), 671–674 (2011).                         inflammatory pain
6    Feng J, Zhou Y, Campbell SL et al. Dnmt1 and Dnmt3a             23   Tran L, Chaloner A, Sawalha AH, Greenwood Van-
     maintain DNA methylation and regulate synaptic function              Meerveld B. Importance of epigenetic mechanisms in
     in adult forebrain neurons. Nat. Neurosci. 13(4), 423–430            visceral pain induced by chronic water avoidance stress.
     (2010).                                                              Psychoneuroendocrinology 38(6), 898–906 (2013).
7    Kramer JM, Kochinke K, Oortveld MA et al. Epigenetic            24   Kukkar A, Singh N, Jaggi AS. Attenuation of neuropathic
     regulation of learning and memory by Drosophila EHMT/                pain by sodium butyrate in an experimental model of
     G9a. PLoS Biol. 9(1), e1000569 (2011).                               chronic constriction injury in rats. J. Formos. Med. Assoc.
8    Lutz BM, Bekker A, Tao YX. Noncoding RNAs: new players               S0929–S6646(13), 00180 (2013).
     in chronic pain. Anesthesiology 121(2), 409–417 (2014).         25   Denk F, Huang W, Sidders B et al. HDAC inhibitors
••   More updated review regarding the role of noncoding                  attenuate the development of hypersensitivity in models of
     RNAs in chronic pain.                                                neuropathic pain. Pain 154(9), 1668–1679 (2013).

9    Wang F, Stefano GB, Kream RM. Epigenetic modification           26   Matsushita Y, Araki K, Omotuyi O, Mukae T, Ueda H.
     of DRG neuronal gene expression subsequent to nerve injury:          HDAC inhibitors restore C-fibre sensitivity in experimental
     etiological contribution to complex regional pain syndromes          neuropathic pain model. Br. J. Pharmacol. 170(5), 991–998
     (Part II). Med. Sci. Monit. 20, 1188–1200 (2014).                    (2013).

10   Zovkic IB, Guzman-Karlsson MC, Sweatt JD. Epigenetic            27   Uchida H, Ma L, Ueda H. Epigenetic gene silencing
     regulation of memory formation and maintenance. Learn.               underlies C-fiber dysfunctions in neuropathic pain.
     Mem. 20(2), 61–74 (2013).                                            J. Neurosci. 30(13), 4806–4814 (2010).

11   Mauck M, Van de Ven T, Shaw AD. Epigenetics of chronic          ••   The first study to identify the role of histone modefication
     pain after thoracic surgery. Curr. Opin. Anaesthesiol. 27(1),        in neuropathic pain.
     1–5 (2014).                                                     28   Uchida H, Sasaki K, Ma L, Ueda H. Neuron-restrictive
12   Rahn EJ, Guzman-Karlsson MC, David SJ. Cellular,                     silencer factor causes epigenetic silencing of Kv4.3 gene after
     molecular, and epigenetic mechanisms in non-associative              peripheral nerve injury. Neuroscience 166(1), 1–4 (2010).
     conditioning: implications for pain and memory. Neurobiol.      29   Uchida H, Matsushita Y, Ueda H. Epigenetic regulation
     Learn. Mem. 105, 133–150 (2013).                                     of BDNF expression in the primary sensory neurons after
13   Seo S, Grzenda A, Lomberk G, Ou XM, Cruciani RA,                     peripheral nerve injury: implications in the development of
     Urrutia R. Epigenetics: a promising paradigm for better              neuropathic pain. Neuroscience 240, 147–154 (2013).
     understanding and managing pain. J. Pain 14(6), 549–557         30   Li K, Zhao GQ, Li LY, Wu GZ, Cui SS. Epigenetic
     (2013).                                                              upregulation of Cdk5 in the dorsal horn contributes to
14   Stone LS, Szyf M. The emerging field of pain epigenetics.            neuropathic pain in rats. Neuroreport 25(14), 1116–1121
     Pain 154(1), 1–2 (2013).                                             (2014).
15   Kouzarides T. Chromatin modifications and their function.       31   Kiguchi N, Kobayashi Y, Maeda T et al. Epigenetic
     Cell 128(4), 693–705 (2007).                                         augmentation of the macrophage inflammatory protein
                                                                          2/C-X-C chemokine receptor type 2 axis through histone H3
16   Kuo MH, Allis CD. Roles of histone acetyltransferases and
                                                                          acetylation in injured peripheral nerves elicits neuropathic
     deacetylases in gene regulation. Bioessays 20(8), 615–626
                                                                          pain. J. Pharmacol. Exp. Ther. 340(3), 577–587 (2012).
     (1998).
                                                                     32   Kiguchi N, Kobayashi Y, Saika F, Kishioka S. Epigenetic
17   Bannister AJ, Kouzarides T. Regulation of chromatin by
                                                                          upregulation of CCL2 and CCL3 via histone modifications
     histone modifications. Cell Res. 21(3), 381–395 (2011).
                                                                          in infiltrating macrophages after peripheral nerve injury.
18   Contestabile A, Sintoni S. Histone acetylation in                    Cytokine 64(3), 666–672 (2013).
     neurodevelopment. Curr. Pharm. Des. 19(28), 5043–5050
                                                                     33   Zhu XY, Huang CS, Li Q et al. p300 exerts an epigenetic
     (2013).
                                                                          role in chronic neuropathic pain through its acetyltransferase
19   Gong F, Miller KM. Mammalian DNA repair: HATs and                    activity in rats following chronic constriction injury (CCI).
     HDACs make their mark through histone acetylation.                   Mol. Pain 8, 84 (2012).
     Mutat. Res. 750(1–2), 23–30 (2013).
                                                                     34   Zhu XY, Huang CS, Li Q et al. Temporal distribution of
20   Bai G, Wei D, Zou S, Ren K, Dubner R. Inhibition of                  p300/CBP immunoreactivity in the adult rat spinal dorsal
     class II histone deacetylases in the spinal cord attenuates          horn following chronic constriction injury (CCI). Cell Mol.
     inflammatory hyperalgesia. Mol. Pain 6, 51 (2010).                   Neurobiol. 33(2), 197–204 (2013).
21   Chiechio S, Zammataro M, Morales ME et al. Epigenetic           35   Yin Q, Lu FF, Zhao Y et al. Resveratrol facilitates pain
     modulation of mGlu2 receptors by histone deacetylase                 attenuation in a rat model of neuropathic pain through the
     inhibitors in the treatment of inflammatory pain. Mol.               activation of spinal Sirt1. Reg. Anesth. Pain Med. 38(2),
     Pharmacol. 75(5), 1014–1020 (2009).                                  93–99 (2013).

     future science group                                                                                     www.futuremedicine.com            243
Review   Liang, Lutz, Bekker & Tao

               36   Sun Y, Liang D, Sahbaie P, Clark JD. Effects of methyl donor        54   Liang G, Chan MF, Tomigahara Y et al. Cooperativity
                    diets on incisional pain in mice. PLoS One 8(10), e77881                 between DNA methyltransferases in the maintenance
                    (2013).                                                                  methylation of repetitive elements. Mol. Cell Biol. 22(2),
               37   Sun Y, Sahbaie P, Liang DY et al. Epigenetic regulation of               480–491 (2002).
                    spinal CXCR2 signaling in incisional hypersensitivity in mice.      55   Lubin FD, Gupta S, Parrish RR, Grissom NM, Davis RL.
                    Anesthesiology 119(5), 1198–1208 (2013).                                 Epigenetic mechanisms: critical contributors to long-term
               38   Mayer DJ, Mao J, Holt J, Price DD. Cellular mechanisms of                memory formation. Neuroscientist17(6), 616–632 (2011).
                    neuropathic pain, morphine tolerance, and their interactions.       56   Mifsud KR, Gutierrez-Mecinas M, Trollope AF, Collins A,
                    Proc. Natl Acad. Sci. USA 96(14), 7731–7736 (1999).                      Saunderson EA, Reul JM. Epigenetic mechanisms in stress
               39   Liang DY, Li X, Clark JD. Epigenetic regulation of opioid-               and adaptation. Brain Behav. Immun. 25(7), 1305–1315
                    induced hyperalgesia, dependence, and tolerance in mice.                 (2011).
                    J. Pain 14(1), 36–47 (2013).                                        57   Turek-Plewa J, Jagodzinski PP. The role of mammalian DNA
               40   He X, Ou P, Wu K et al. Resveratrol attenuates morphine                  methyltransferases in the regulation of gene expression. Cell
                    antinociceptive tolerance via SIRT1 regulation in the rat                Mol. Biol. Lett. 10(4), 631–647 (2005).
                    spinal cord. Neurosci. Lett. 566, 55–60 (2014).                     58   Qi F, Zhou Y, Xiao Y et al. Promoter demethylation
               41   Cherng CH, Lee KC, Chien CC et al. Baicalin ameliorates                  of cystathionine-beta-synthetase gene contributes to
                    neuropathic pain by suppressing HDAC1 expression in the                  inflammatory pain in rats. Pain 154(1), 34–45 (2013).
                    spinal cord of spinal nerve ligation rats. J. Formos. Med. Assoc.   59   Pan Z, Zhu LJ, Li YQ et al. Epigenetic modification of spinal
                    113(8), 513–520 (2014).                                                  miR-219 expression regulates chronic inflammation pain by
               42   Klein K, Ospelt C, Gay S. Epigenetic contributions in the                targeting CaMKIIgamma. J. Neurosci 34(29), 9476–9483
                    development of rheumatoid arthritis. Arthritis Res. Ther.                (2014).
                    14(6), 227 (2012).                                                  60   Wang Y, Liu C, Guo QL et al. Intrathecal 5-azacytidine
               43   Covington HE III, Maze I, Sun H et al. A role for repressive             inhibits global DNA methylation and methyl-CpG-binding
                    histone methylation in cocaine-induced vulnerability to stress.          protein 2 expression and alleviates neuropathic pain in
                    Neuron 71(4), 656–670 (2011).                                            rats following chronic constriction injury. Brain Res. 1418,
                                                                                             64–69 (2011).
               44   Gupta S, Kim SY, Artis S et al. Histone methylation regulates
                    memory formation. J. Neurosci. 30(10), 3589–3599 (2010).            •    The first study to demonstrate the role of DNA
                                                                                             methylation in neuropathic pain.
               45   Maze I, Covington HE III, Dietz DM et al. Essential role
                    of the histone methyltransferase G9a in cocaine-induced             61   Zhou XL, Yu LN, Wang Y et al. Increased methylation
                    plasticity. Science 327(5962), 213–216 (2010).                           of the MOR gene proximal promoter in primary sensory
                                                                                             neurons plays a crucial role in the decreased analgesic effect
               46   Imai S, Ikegami D, Yamashita A et al. Epigenetic
                                                                                             of opioids in neuropathic pain. Mol. Pain 10, 51 (2014).
                    transcriptional activation of monocyte chemotactic protein 3
                    contributes to long-lasting neuropathic pain. Brain 136(Pt 3),      62   Pollema-Mays SL, Centeno MV, Apkarian AV, Martina M.
                    828–843 (2013).                                                          Expression of DNA methyltransferases in adult dorsal root
                                                                                             ganglia is cell-type specific and up regulated in a rodent
               47   Tsai RY, Shen CH, Feng YP et al. Ultra-low-dose naloxone
                                                                                             model of neuropathic pain. Front Cell Neurosci. 8, 217
                    enhances the antinociceptive effect of morphine in PTX-
                                                                                             (2014).
                    treated rats: regulation on global histone methylation. Acta
                    Anaesthesiol. Taiwan. 50(3), 106–111 (2012).                        63   Tajerian M, Alvarado S, Millecamps M et al. Peripheral
                                                                                             nerve injury is associated with chronic, reversible changes
               48   Zhang Z, Tao W, Hou YY, Wang W, Kenny PJ, Pan ZZ.
                                                                                             in global DNA methylation in the mouse prefrontal cortex.
                    MeCP2 repression of G9a in regulation of pain and morphine
                                                                                             PLoS One 8(1), e55259 (2013).
                    reward. J. Neurosci 34(27), 9076–9087 (2014).
                                                                                        64   Menzies V, Lyon DE, Archer KJ et al. Epigenetic alterations
               49   Chedin F, Lieber MR, Hsieh CL. The DNA
                                                                                             and an increased frequency of micronuclei in women with
                    methyltransferase-like protein DNMT3L stimulates de novo
                                                                                             fibromyalgia. Nurs. Res. Pract. 2013, 795784 (2013).
                    methylation by Dnmt3a. Proc. Natl Acad. Sci. USA 99(26),
                    16916–16921 (2002).                                                 65   Tajerian M, Alvarado S, Millecamps M et al. DNA
                                                                                             methylation of SPARC and chronic low back pain. Mol. Pain
               50   Jeltsch A. Molecular enzymology of mammalian DNA
                                                                                             7, 65 (2011).
                    methyltransferases. Curr. Top. Microbiol. Immunol. 301,
                    203–225 (2006).                                                     66   Viet CT, Ye Y, Dang D et al. Re-expression of the
                                                                                             methylated EDNRB gene in oral squamous cell carcinoma
               51   Siedlecki P, Zielenkiewicz P. Mammalian DNA
                                                                                             attenuates cancer-induced pain. Pain 152(10), 2323–2332
                    methyltransferases. Acta Biochim. Pol. 53(2), 245–256 (2006).
                                                                                             (2011).
               52   Fatemi M, Hermann A, Gowher H, Jeltsch A. Dnmt3a and
                                                                                        67   Bell JT, Loomis AK, Butcher LM et al. Differential
                    Dnmt1 functionally cooperate during de novo methylation of
                                                                                             methylation of the TRPA1 promoter in pain sensitivity. Nat.
                    DNA. Eur. J. Biochem. 269(20), 4981–4984 (2002).
                                                                                             Commun. 5, 2978 (2014).
               53   Gowher H, Stockdale CJ, Goyal R, Ferreira H, Owen-Hughes
                                                                                        68   Karouzakis E, Gay RE, Michel BA, Gay S, Neidhart M.
                    T, Jeltsch A. De novo methylation of nucleosomal DNA by the
                                                                                             DNA hypomethylation in rheumatoid arthritis synovial
                    mammalian Dnmt1 and Dnmt3A DNA methyltransferases.
                                                                                             fibroblasts. Arthritis Rheum. 60(12), 3613–3622 (2009).
                    Biochemistry 44(29), 9899–9904 (2005).

244            Epigenomics (2015) 7(2)                                                                                              future science group
Epigenetic regulation of chronic pain   Review

69   Liu CC, Fang TJ, Ou TT et al. Global DNA methylation,           75   Downs J, Geranton SM, Bebbington A et al. Linking
     DNMT1, and MBD2 in patients with rheumatoid arthritis.               MECP2 and pain sensitivity: the example of Rett syndrome.
     Immunol. Lett. 135(1–2), 96–99 (2011).                               Am. J. Med. Genet. A 152A(5), 1197–1205 (2010).
70   Nakano K, Whitaker JW, Boyle DL, Wang W, Firestein GS.          76   Geranton SM, Fratto V, Tochiki KK, Hunt SP. Descending
     DNA methylome signature in rheumatoid arthritis. Ann.                serotonergic controls regulate inflammation-induced
     Rheum. Dis. 72(1), 110–117 (2013).                                   mechanical sensitivity and methyl-CpG-binding protein 2
71   Karouzakis E, Gay RE, Gay S, Neidhart M. Increased                   phosphorylation in the rat superficial dorsal horn. Mol. Pain
     recycling of polyamines is associated with global DNA                4, 350 (2008).
     hypomethylation in rheumatoid arthritis synovial fibroblasts.   •    First study that demonstrated the involvement of MeCP2
     Arthritis Rheum. 64(6), 1809–1817 (2012).                            in inflammatory pain.
72   Klein K, Gay S. Epigenetic modifications in rheumatoid          77   Tochiki KK, Cunningham J, Hunt SP, Geranton SM. The
     arthritis, a review. Curr. Opin. Pharmacol. 13(3), 420–425           expression of spinal methyl-CpG-binding protein 2, DNA
     (2013).                                                              methyltransferases and histone deacetylases is modulated in
73   Sansom OJ, Maddison K, Clarke AR. Mechanisms of                      persistent pain states. Mol. Pain 8, 14 (2012).
     disease: methyl-binding domain proteins as potential            78   Chen WG, Chang Q, Lin Y et al. Derepression of BDNF
     therapeutic targets in cancer. Nat. Clin Pract. Oncol. 4(5),         transcription involves calcium-dependent phosphorylation of
     305–315 (2007).                                                      MeCP2. Science 302(5646), 885–889 (2003).
74   Cuddapah VA, Pillai RB, Shekar KV et al. Methyl-CpG-
     binding protein 2 (MECP2) mutation type is associated
     with disease severity in Rett syndrome. J. Med. Genet. 51(3),
     152–158 (2014).

     future science group                                                                                    www.futuremedicine.com            245
You can also read