Elsevier

Experimental Neurology

Volume 283, Part A, September 2016, Pages 276-286
Experimental Neurology

Research Paper
Increased miR-132-3p expression is associated with chronic neuropathic pain

https://doi.org/10.1016/j.expneurol.2016.06.025Get rights and content

Highlight

  • Pain and inflammation up-regulates miR-132-3p in humans

  • SNI causes spinal and DRG miR-132-3p up-regulation in rats

  • Spinal miR-132-3p antagonism induces long-lasting anti-hyperalgesia

  • Spinal miR-132-3p mimicry induces thermal and mechanical hyperalgesia

Abstract

Alterations in the neuro-immune balance play a major role in the pathophysiology of chronic neuropathic pain. MicroRNAs (miRNA) can regulate both immune and neuronal processes and may function as master switches in chronic pain development and maintenance. We set out to analyze the role of miR-132-3p, first in patients with peripheral neuropathies and second in an animal model of neuropathic pain. We initially determined miR-132-3p expression by measuring its levels in white blood cells (WBC) of 30 patients and 30 healthy controls and next in sural nerve biopsies of 81 patients with painful or painless inflammatory or non-inflammatory neuropathies based on clinical diagnosis. We found a 2.6 fold increase in miR-132-3p expression in WBC of neuropathy patients compared to healthy controls (p < 0.001). MiR-132-3p expression was also slightly up-regulated in sural nerve biopsies from neuropathy patients suffering from neuropathic pain compared to those without pain (1.2 fold; p < 0.001).

These promising findings were investigated further in an animal model of neuropathic pain, the spared nerve injury model (SNI). For this purpose miR-132-3p expression levels were measured in dorsal root ganglia and spinal cord of rats. Subsequently, miR-132-3p expression was pharmacologically modulated with miRNA antagonists or mimetics, and evoked pain and pain aversion were assessed.

Spinal miR-132-3p levels were highest 10 days after SNI, a time when persistent allodynia was established (p < 0.05). Spinal administration of miR-132-3p antagonists via intrathecal (i.t.) catheters dose dependently reversed mechanical allodyina (p < 0.001) and eliminated pain behavior in the place escape avoidance paradigm (p < 0.001). Intrathecal administration of miR-132-3p mimetic dose-dependently induced pain behavior in naïve rats (p < 0.001). Taken together these results indicate a pro-nociceptive effect of miR-132-3p in chronic neuropathic pain.

Introduction

Neuropathic pain is characteristically severe and persistent and may greatly impair health related quality of life by additionally inducing anxiety, depression, and cognitive impairment (Breivik et al., 2006). There is ample evidence for a potential role of the immune system and particularly of pro- and anti-inflammatory mediators in the pathophysiology of neuropathic pain (Kuner, 2010, McMahon and Malcangio, 2009). Peripheral neuropathies of the same etiology can either be painful or painless (Üçeyler et al., 2007). The mechanism for this discrepancy is unknown.

In recent years, non-coding RNAs have been studied in normal cellular functioning as well as in pathological processes (Huttenhofer and Schattner, 2006, Mattick, 2004). Micro-RNAs (miRNAs) are a family of non-coding RNAs that post-transcriptionally regulate gene expression by inhibiting mRNA translation or inhibiting mRNA and protein degradation (Mattick, 2004). Various diseases, including neuropathic pain disorders, appear to possess unique miRNA expression signatures. Recent reports on modulation of miRNA function in both neuronal and immune processes predict the therapeutic potential of manipulating miRNAs in diseases affecting the immune system and the brain (O'Connor et al., 2012, Soreq and Wolf, 2011). miRNAs that communicate between the nervous and immune system have been termed “neurimmiRs” and primarily target transcription factors or other regulatory genes, which enable simultaneous cross-communication between neural and immune compartments (Soreq and Wolf, 2011). Thus, miRNAs possibly control cellular pathways in multiple systems and act as “master-switches” (Soreq and Wolf, 2011).

Aberrant expression of several miRNAs has been reported throughout many peripheral and central nervous system loci associated with pain perception (Aldrich et al., 2009, Bai et al., 2007, Imai et al., 2011, Kusuda et al., 2011, von Schack et al., 2011). First reports describing characteristic miRNA expression profiles in blood or cerebrospinal fluid of patients with distinct pain conditions are starting to emerge (Andersen et al., 2016, Beyer et al., 2015, Bjersing et al., 2013, Orlova et al., 2011), however evidence linking specific miRNA expression profiles to specific pain disorders is still insufficient.

miR-132 is abundantly expressed in the brain and is emerging as a regulator of cognition, neuronal plasticity, and memory. It can regulate synapse structure and function (Bredy et al., 2011, Miller et al., 2012, Schratt, 2009, Soreq and Wolf, 2011). Hippocampal miR-132 mediates stress-induced cognitive deficits through suppression of acetylcholinesterase (Haramati et al., 2011) and miR-132 has recently been implicated in neuropathic pain after chronic constriction injury (CCI) (Arai et al., 2013). Similarly, spinal cord miR-132 is now proposed as a mediator of neuropathic pain following spared nerve injury (SNI) (Zhang et al., 2015). However, direct links between pain and miR-132 expression levels in human and/or animal models of neuropathic pain still remain elusive.

The current studies evaluated blood and sural nerve miR-132-3p, a splice variant of miR-132, expression in patients suffering from chronic neuropathic pain accompanying peripheral neuropathy and analyzed the role of miR-132-3p in pain behavior in an animal model of neuropathic pain.

Section snippets

Patient assessment and diagnostic classification

Patients with neuropathies of different etiologies were recruited at the Department of Neurology, University of Würzburg between 2014 and 2015, where they underwent diagnostic work-up, including sural nerve biopsy. The study was approved by the Würzburg Medical Faculty Ethics Committee and written informed consent was obtained from every study participant before recruitment. The diagnosis of neuropathy was based on characteristic symptoms and signs in the neurological examination and typical

Basic description of the patient cohort

We included 81 patients with neuropathies of different etiology. The study cohort had a median age of 66 years (range 33–84 years) and consisted of 55 men (median age: 66 years, range 33–84 years) and 26 women (median age: 67 years, range 47–84 years). Clinical characteristics of the cohort and diagnostic subgroups are summarized in Table 1. In 23/81 (28%) patients an inflammatory neuropathy was diagnosed (CIDP, vasculitic neuropathy, PIAN, paraproteinemic neuropathy), while in 24/81 patients (30%)

Discussion

miRNAs have a vital role in post-transcriptional regulation, are widely expressed throughout the brain, are regulated by neuronal activity and some, including miR-132 are thought to be necessary for neuronal plasticity required for memory consolidation and pathological pain (Elramah et al., 2014, Soreq and Wolf, 2011). For example, overexpression of hippocampal miR-132 increased local excitatory postsynaptic currents and impaired learning and memory processes (Edbauer et al., 2010), and

Conclusion

Taken together these findings imply that aberrant neural miR-132-3p expression is associated with human neuropathic pain, and that in animals targeted antagonism of miR-132-3p results in dose-dependent reversal of pain behavior while miR-132-3p mimicry results in a dose-dependent induction of pain behavior allowing us to designate miR-132-3p as a pro-nociceptive miRNA in our study. Importantly, alterations in miRNA levels may be indicative of their functional involvement in pain

Conflicts of interest

The authors have no conflicts of interest.

Acknowledgement

The authors would like to thank Joanne Steinauer for assistance with the confocal microscope, Dr. Gary Firestein for the use of his RT-PCR and Tony Yaksh for helpful discussions. This work was supported by NIH NINDS 067459 (LSS) and BaCaTeC® (No. 20; 2013-2) (ML, CS, LSS). NÜ and CS received funding by the European Union's Seventh Framework Programme (“ncRNAPain”, grant agreement number 602133). This work is part of the doctoral thesis of ML.

References (65)

  • D.M. Dirig et al.

    Characterization of variables defining hindpaw withdrawal latency evoked by radiant thermal stimuli

    J. Neurosci. Methods

    (1997)
  • D. Edbauer et al.

    Regulation of synaptic structure and function by FMRP-associated microRNAs miR-125b and miR-132

    Neuron

    (2010)
  • B. Hartmann et al.

    The AMPA receptor subunits GluR-A and GluR-B reciprocally modulate spinal synaptic plasticity and inflammatory pain

    Neuron

    (2004)
  • R.R. Ji et al.

    Glia and pain: is chronic pain a gliopathy?

    Pain

    (2013)
  • S.T. Lee et al.

    Altered microRNA regulation in Huntington's disease models

    Exp. Neurol.

    (2011)
  • S.B. McMahon et al.

    Current challenges in glia-pain biology

    Neuron

    (2009)
  • H. Soreq et al.

    NeurimmiRs: microRNAs in the neuroimmune interface

    Trends Mol. Med.

    (2011)
  • C.A. Stein

    Exploiting the potential of antisense: beyond phosphorothioate oligodeoxynucleotides

    Chem. Biol.

    (1996)
  • J. Tsang et al.

    MicroRNA-mediated feedback and feedforward loops are recurrent network motifs in mammals

    Mol. Cell

    (2007)
  • M. Von Korff et al.

    Grading the severity of chronic pain

    Pain

    (1992)
  • Z. Zhang et al.

    MicroRNAs: potential regulators involved in human anencephaly

    Int. J. Biochem. Cell Biol.

    (2010)
  • H.H. Andersen et al.

    Serum MicroRNA Signatures in Migraineurs During Attacks and in Pain-Free Periods

    Mol. Neurobiol.

    (2016)
  • M. Arai et al.

    The miRNA and mRNA changes in rat hippocampi after chronic constriction injury

    Pain Med.

    (2013)
  • G. Bai et al.

    Downregulation of selective microRNAs in trigeminal ganglion neurons following inflammatory muscle pain

    Mol. Pain

    (2007)
  • D. Betel et al.

    The microRNA.org resource: targets and expression

    Nucleic Acids Res.

    (2008)
  • C. Beyer et al.

    Signature of circulating microRNAs in osteoarthritis

    Ann. Rheum. Dis.

    (2015)
  • J.L. Bjersing et al.

    Profile of cerebrospinal microRNAs in fibromyalgia

    PLoS One

    (2013)
  • G.A. Calin et al.

    MicroRNA signatures in human cancers

    Nat. Rev. Cancer

    (2006)
  • J.P. Cogswell et al.

    Identification of miRNA changes in Alzheimer's disease brain and CSF yields putative biomarkers and insights into disease pathways

    J. Alzheimers Dis.

    (2008)
  • P. Dyck et al.

    Peripheral Neuropathy

    (2005)
  • S. Elramah et al.

    MicroRNAs regulate neuronal plasticity and are involved in pain mechanisms

    Front. Cell. Neurosci.

    (2014)
  • R.C. Friedman et al.

    Most mammalian mRNAs are conserved targets of microRNAs

    Genome Res.

    (2009)
  • Cited by (81)

    • miRNA contributes to neuropathic pains

      2023, International Journal of Biological Macromolecules
    • The contribution of neuro-immune crosstalk to pain in the peripheral nervous system and the spinal cord

      2022, International Immunopharmacology
      Citation Excerpt :

      In a rat model of spared nerve injury (SNI), the miR-132-3p expression is upregulated in the spinal cord and DRGs. Application of miR-132-3p could induce mechanical hypersensitivity, whereas the intrathecal injection of miR-132-3p antagonist relieves SNI-induced neuropathic pain [27]. In addition, a clinical study has also highlighted that miR-132-3p, miR-98-5p, miR-let-7d-5p are upregulated in patients with neuropathic pain [28].

    View all citing articles on Scopus
    View full text