Elsevier

Cellular Signalling

Volume 44, April 2018, Pages 51-61
Cellular Signalling

Wnt5a mediates chronic post-thoracotomy pain by regulating non-canonical pathways, nerve regeneration, and inflammation in rats

https://doi.org/10.1016/j.cellsig.2018.01.017Get rights and content

Highlights

  • The non-canonical Wnt5a pathways but not the canonical one are activated in CPTP.

  • The non-canonical Wnt5a pathways and nerve regeneration are activated in the chronic phase of CPTP.

  • Wnt5a, Ror2 receptor and inflammation are activated through the development of CPTP.

  • Wnt5a could regulate nerve regeneration and inflammation in CPTP.

Abstract

As well-characterized ligands involved in neurogenesis, Wnts are emerging as promising targets in pain pathogenesis. Our previous pilot study showed that intrathecal inhibition of Wnt5a, but not Wnts, relieves chronic post-thoracotomy pain (CPTP) in rats. In the present study, we aimed to further explore the regulatory mechanism of Wnt5a in CPTP development. Increased protein levels of Wnt5a, transmembrane receptor Ror2, and activated non-canonical Wnt pathway members were found in the thoracic dorsal root ganglions from postoperative day (POD) 7 to POD 21. However, the levels of canonical Wnt pathway members showed no change by reverse transcriptase-PCR. In addition, elevated nerve regeneration, activated pro-inflammatory factors, and glial cells were detected from POD 7 to POD 21. Furthermore, intrathecal Wnt5a blockade during the early phase (POD 0 to POD 9) significantly increased the pain threshold, and intervention in the late phase (POD 14 to POD 16) alleviated pain; however, the analgesic response was not as effective as that in the early phase. Additionally, early but not late Wnt5a blockade significantly reversed CPTP-induced activation of the non-canonical Wnt pathways, nerve regeneration, and inflammation. In contrast, a Wnt5a agonist decreased the pain threshold in both naïve and painless rats. These results suggest that Wnt5a promotes the development of CPTP by activating non-canonical Wnt pathways, nerve regeneration, and inflammation. Therapeutic intervention by targeting Wnt5a may represent an effective strategy for preventing and treating CPTP.

Introduction

Management strategies for chronic pain after surgery, particularly amputation, breast surgery, and thoracotomy, are inadequate considering the high incidence of such chronic pain and insensitivity to typical therapies [1, 2]. Chronic post-thoracotomy pain (CPTP), which recurs or persists along a thoracotomy incision at least 2 months following the surgical procedure [3], with a high incidence of CPTP (50%) for 3–6 postoperative months [4], has become a major research focus in the past two decades. Surgical-associated intercostal nerve injury is believed to be a dominant factor in the pathogenesis of CPTP, as demonstrated by intraoperative electrophysiology examinations [5] and the high proportion of neuropathic pain in CPTP [6, 7]. However, strategies aimed at protecting the intercostal nerve do not always prevent CPTP, indicating that complex additional factors are involved. Thus, additional research into the effective management CPTP pathogenesis are needed.

In a pilot study [8], we showed that Wnt5a inhibition alleviates CPTP. This finding was consistent data showing that levels of circulating microRNAs targeting Wnt pathway members in rodent models of both neuropathic pain and inflammatory pain were changed, suggesting Wnt as a possible biomarker for chronic pain [9]. Moreover, Wnts are involved in both the induction and persistence of neuropathic pain [10, 11], accompanied with a rapid-onset and long-lasting upregulation of pro-inflammatory factors and calcium signals in the spinal cord [10]. Wnts also play a role in cancer pain by activating non-canonical Wnt pathways [12]. As a secreted glycoprotein, Wnt5a acts mainly via the non-canonical planar cell polarity, or Ca2+ pathway, rather than via the canonical β-catenin pathway [13], whose effects are often opposite [14]. The downstream cellular signals of Wnt5a in CPTP remain to be elucidated.

Quite recently, it was reported that the inability of regenerating nerves to re-innervate target tissues results in the persistence of neuropathic pain [15], suggesting a crucial role of nerve regeneration in the condition. As critical ligands in maintaining neuronal functions and tissue morphogenesis, dysregulated Wnts have been identified in various degenerative and inflammatory central nervous system disorders [13, 16, 17]. Thus, we further tested the involvement of nerve regeneration in the maintenance of CPTP and its regulation by Wnt5a. Additionally, Wnt expression in the immune system and in immune-like cells of the central nervous system suggested its involvement in inflammation-related damage or repair [13]. Similarly, we found that the activated pro-inflammatory response in CPTP could be regulated by Wnt5a [8]. Interestingly, patients with more serious acute post-thoracotomy pain were more likely to develop chronic pain [18], indicating that alterations in the acute phase may contribute to the development of CPTP. Moreover, in our retrospective study of CPTP, we found that the difference between pre- and post-operative day 2 white blood cell counts could predict the occurrence of CPTP [19]. Thus, we further examined the dynamic changes of systemic inflammation and neuroinflammation to determine these changes are correlated with CPTP development.

In this study, we focused on the elusive mechanism of Wnt5a in the induction and persistence of CPTP, and its regulation of Wnt pathways, nerve regeneration, and inflammation.

Section snippets

Animals

Adult male Sprague-Dawley rats (250–350 g, Experimental Animal Center of the Chinese Academy of Medical Sciences, Beijing, China) were used in this study. Rats were housed at a temperature of 22–24 °C under a 12 h light-dark cycle. All animal protocols were approved by the Institutional Animal Care and Use Committee in the Chinese Academy of Medical Sciences. The procedures were performed in accordance to the National Institutes of Health Guide for Care and Use of Laboratory Animals. Rats were

CPTP induced time-course activation of Wnt5a and Ror2 in the DRG

Rats were divided into the naïve group (n = 10), sham group (n = 10), and model group (n = 20), which was further divided into the CPTP group and non-CPTP group according to the pain state observed on POD 21. Rats with an MWT ≦4 g on POD 21 were assigned to the CPTP group (n = 11), and the remaining rats were assigned to the non-CPTP group (n = 9). The pain incidence in all our model group was consistent with that reported in previous studies [20, 21]. The MWT in the CPTP group showed a sharp

Discussion

Emerging evidence has shown that Wnts play a critical role in the pathogenesis of neuropathic pain [10], the persistence of inflammatory pain [10], and the pathogenesis of human immunodeficiency virus-induced pain [28]. In this study, we found that Wnt5a was activated on POD 7 and upregulated over time in CPTP. Inhibiting Wnt5a by antagonizing its binding site significantly reduced pain-related behavior. Moreover, early intervention during the preliminary and acute phases resulted in enhanced

Conclusions

Collectively, we found that the activated Wnt5a/Ca2+ and Wnt5a/planar cell polarity pathways, along with increased axonal regeneration and sympathetic sprouting (which could be regulated by Wnt5a), are involved in CPTP persistence. The accompanying activation of inflammation modulated by Wnt5a may play a role in CPTP induction and persistence. Further, this study suggests that early (but not late) therapy targeting Wnt5a may represent an effective treatment for CPTP.

Funding

This study was supported by grants from National Natural Science Foundation of China (NO: 81671098).

Competing interests

The authors declare no competing interests.

Author contributions

YH designed the experiments and reviewed the manuscript. AF carried out the thoracotomy, intrathecal catheterization and injection, western blot, RT-PCR, immunohistochemistry, ELISA and prepared the manuscript. LS, LX and WC conducted the pain behavior measurements, analyzed the data, and reviewed the manuscript. All authors have read and approved the final manuscript.

Acknowledgements

We thank Dr. Xin Zhao for supplying the animal ventilator. We also thank Professor Chao Ma and post-doctoral fellow Fan Liu for helpful comments on the experiments.

References (40)

  • M.S. Ramer et al.

    Rapid sprouting of sympathetic axons in dorsal root ganglia of rats with a chronic constriction injury

    Pain

    (1997)
  • K. Schaale et al.

    Wnt signaling in macrophages: augmenting and inhibiting mycobacteria-induced inflammatory responses

    Eur. J. Cell Biol.

    (2011)
  • D. Reddi

    Preventing chronic postoperative pain

    Anaesthesia

    (2016)
  • H. Merskey et al.

    Classification of chronic pain

  • M.L. Rogers et al.

    Preliminary findings in the neurophysiological assessment of intercostal nerve injury during thoracotomy

    Eur. J. Cardiol.

    (2002)
  • M.F. Maguire et al.

    A questionnaire study investigating the prevalence of the neuropathic component of chronic pain after thoracic surgery

    Eur. J. Cardiol.

    (2006)
  • R.A. Qureshi et al.

    Circulating microRNAs signatures in rodent models of pain

    Mol. Neurobiol.

    (2016)
  • Y.K. Zhang et al.

    WNT signaling underlies the pathogenesis of neuropathic pain in rodents

    J. Clin. Invest.

    (2013)
  • S. Liu et al.

    Wnt/Ryk signaling contributes to neuropathic pain by regulating sensory neuron excitability and spinal synaptic plasticity in rats

    Pain

    (2015)
  • R. van Amerongen et al.

    Alternative wnt signaling is initiated by distinct receptors

    Sci. Signal.

    (2008)
  • Cited by (16)

    • C3aR in astrocytes mediates post-thoracotomy pain by inducing A1 astrocytes in male rats

      2023, Biochimica et Biophysica Acta - Molecular Basis of Disease
    • Wnt signaling: A prospective therapeutic target for chronic pain

      2022, Pharmacology and Therapeutics
      Citation Excerpt :

      In another study, Zhong et al. revealed that dexmedetomidine remarkably ameliorated mechanical allodynia and thermal hyperalgesia in streptozotocin-induced diabetic neuropathic pain rats via inhibiting Wnt10a/β-catenin signaling pathway and astrocytic activation in the spinal cord (Zhong et al., 2018). In a rat model of chronic post-thoracotomy pain, Zhu et al. showed that Box5, a Wnt5a antagonist, considerably inhibited the activation of astrocyte in the spinal cord (Zhu et al., 2017, 2018). Moreover, Box5 also repressed the spinal activation of astrocyte in nucleoside reverse transcriptase inhibitors-induce chronic pain mice (S. Yuan, Shi, Guo, & Tang, 2018).

    • Ror2 mediates chronic post-thoracotomy pain by inducing the transformation of A1/A2 reactive astrocytes in rats

      2022, Cellular Signalling
      Citation Excerpt :

      Rats with an MWT of ≤4 g on POD 21 were assigned to the CPTP group (n = 17), and the remaining rats were assigned to the non-CPTP group (n = 13). The pain incidence in all model groups was consistent with that reported in previous studies [21,8]. However, not all thoracotomized rats developed CPTP, which was consistent with clinical practice.

    • 5.34 - Pathophysiology of Postoperative Pain

      2020, The Senses: A Comprehensive Reference: Volume 1-7, Second Edition
    View all citing articles on Scopus
    View full text