Skip to main content

Advertisement

Log in

Neuronal allodynic mechanisms of Slc7a5 (LAT1) in the spared nerve injury rodent model of neuropathic pain

  • Original Article
  • Published:
Pflügers Archiv - European Journal of Physiology Aims and scope Submit manuscript

Abstract

High-impact chronic pain is suffered by 1 in 5 patients in the USA and globally. Effective, non-addictive, non-opioid therapeutics are urgently needed for the treatment of chronic pain. Slc7a5 (Lat1), also known as system L-neutral amino acid transporter, is involved in a number of physiological processes related to inflammation. Transcriptomics studies have shown that Slc7a5 and its binding partner Slc3a2 are expressed in neurons of the dorsal root ganglia (DRG) and spinal dorsal horn, which are critical to the initiation and maintenance of nociception and pathophysiology of chronic pain. In addition, Slc7a5 is a transporter for the first-line anti-allodynic gabapentinoid drugs and binds to ion channels implicated in nociception and chronic pain including the voltage-gated sodium channel Nav1.7 and the voltage-gated potassium channels Kv1.1 and Kv1.2. We found that blocking Slc7a5 with intrathecal administration of the drug JPH203 alleviated allodynia in the spared nerve injury (SNI) rodent model of neuropathic pain. Western blot and immunohistochemistry studies revealed an increase in Slc7a5 protein levels in the spinal cord and DRGs of SNI mice compared to control mice. Using whole-cell current-clamp electrophysiology, we observed that JPH203 treatment reduced excitability of small-diameter (< 30 µm) DRG neurons from SNI mice, in agreement with its behavioral effects. Voltage-clamp recordings from JPH203-treated naïve rat DRGs identified an effect on tetrodotoxin-resistant (TTX-R) sodium currents. Altogether, these results demonstrate that Slc7a5 is dysregulated in chronic neuropathic pain and can be targeted to provide relief of hypersensitivity.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Alles SRA, Gomez K, Moutal A, Khanna R (2020) Putative roles of SLC7A5 (LAT1) transporter in pain. Neurobiol Pain 8:100050. https://doi.org/10.1016/j.ynpai.2020.100050

    Article  PubMed  PubMed Central  Google Scholar 

  2. Baronas VA, Yang RY, Morales LC, Sipione S, Kurata HT (2018) Slc7a5 regulates Kv1.2 channels and modifies functional outcomes of epilepsy-linked channel mutations. Nat Commun 9:4417. https://doi.org/10.1038/s41467-018-06859-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD (2019) The role of voltage-gated sodium channels in pain signaling. Physiol Rev 99:1079–1151. https://doi.org/10.1152/physrev.00052.2017

    Article  CAS  PubMed  Google Scholar 

  4. Biggs JE, Boakye PA, Ganesan N, Stemkowski PL, Garcia AL, Ballanyi K, Smith PA (2014) Analysis of the long-term actions of gabapentin and pregabalin in dorsal root ganglia and substantia gelatinosa. J Neurophysiol. https://doi.org/10.1152/jn.00168.2014

    Article  PubMed  Google Scholar 

  5. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL (1994) Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 53:55–63. https://doi.org/10.1016/0165-0270(94)90144-9

    Article  CAS  PubMed  Google Scholar 

  6. Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts E, Springell K, Karbani G, Jafri H, Mannan J, Raashid Y, Al-Gazali L, Hamamy H, Valente EM, Gorman S, Williams R, McHale DP, Wood JN, Gribble FM, Woods CG (2006) An SCN9A channelopathy causes congenital inability to experience pain. Nature 444:894–898. https://doi.org/10.1038/nature05413

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Decosterd I, Woolf CJ (2000) Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 87:149–158

    Article  Google Scholar 

  8. Dickens D, Webb SD, Antonyuk S, Giannoudis A, Owen A, Rädisch S, Hasnain SS, Pirmohamed M (2013) Transport of gabapentin by LAT1 (SLC7A5). Biochem Pharmacol 85:1672–1683. https://doi.org/10.1016/j.bcp.2013.03.022

    Article  CAS  Google Scholar 

  9. François-Moutal L, Wang Y, Moutal A, Cottier KE, Melemedjian OK, Yang X, Wang Y, Ju W, Largent-Milnes TM, Khanna M, Vanderah TW, Khanna R (2015) A membrane-delimited N-myristoylated CRMP2 peptide aptamer inhibits CaV2.2 trafficking and reverses inflammatory and postoperative pain behaviors. Pain 156:1247–1264. https://doi.org/10.1097/j.pain.0000000000000147

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Häring M, Zeisel A, Hochgerner H, Rinwa P, Jakobsson JET, Lönnerberg P, Manno G, Sharma N, Borgius L, Kiehn O, Lagerström MC, Linnarsson S, Ernfors P (2018) Neuronal atlas of the dorsal horn defines its architecture and links sensory input to transcriptional cell types. Nat Neurosci 21(6):869–880. https://doi.org/10.1038/s41593-018-0141-1

  11. Kanellopoulos AH, Koenig J, Huang H, Pyrski M, Millet Q, Lolignier S, Morohashi T, Gossage SJ, Jay M, Linley JE, Baskozos G, Kessler BM, Cox JJ, Dolphin AC, Zufall F, Wood JN, Zhao J (2018) Mapping protein interactions of sodium channel NaV1.7 using epitope‐tagged gene‐targeted mice. EMBO J 37(3):427–445. https://doi.org/10.15252/embj.201796692

  12. Lamothe SM, Kurata HT (2020) Slc7a5 alters Kvβ-mediated regulation of Kv1.2. J Gen Physiol 152(7):e201912524. https://doi.org/10.1085/jgp.201912524

  13. Lamothe SM, Sharmin N, Silver G, Satou M, Hao Y, Tateno T, Baronas VA, Kurata HT (2020) Control of Slc7a5 sensitivity by the voltage-sensing domain of Kv1 channels. eLife 9:e54916. https://doi.org/10.7554/eLife.54916

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Luo ZD, Calcutt NA, Higuera ES, Valder CR, Song YH, Svensson CI, Myers RR (2002) Injury type-specific calcium channel alpha 2 delta-1 subunit up-regulation in rat neuropathic pain models correlates with antiallodynic effects of gabapentin. J Pharmacol Exp Ther 303:1199–1205. https://doi.org/10.1124/jpet.102.041574

    Article  CAS  PubMed  Google Scholar 

  15. Luo ZD, Chaplan SR, Higuera ES, Sorkin LS, Stauderman KA, Williams ME, Yaksh TL (2001) Upregulation of dorsal root ganglion (alpha)2(delta) calcium channel subunit and its correlation with allodynia in spinal nerve-injured rats. J Neurosci 21:1868–1875

    Article  CAS  Google Scholar 

  16. Moutal A, Chew LA, Yang X, Wang Y, Yeon SK, Telemi E, Meroueh S, Park KD, Shrinivasan R, Gilbraith KB, Qu C, Xie JY, Patwardhan A, Vanderah TW, Khanna M, Porreca F, Khanna R (2016) (S)-lacosamide inhibition of CRMP2 phosphorylation reduces postoperative and neuropathic pain behaviors through distinct classes of sensory neurons identified by constellation pharmacology. Pain 157:1448–1463. https://doi.org/10.1097/j.pain.0000000000000555

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Powell JD (2013) Slc7a5 helps T cells get with the program. Nat Immunol 14:422–424. https://doi.org/10.1038/ni.2594

    Article  CAS  PubMed  Google Scholar 

  18. Sinclair LV, Rolf J, Emslie E, Shi Y-B, Taylor PM, Cantrell DA (2013) Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat Immunol 14:500–508. https://doi.org/10.1038/ni.2556

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Torigoe M, Maeshima K, Ozaki T, Omura Y, Gotoh K, Tanaka Y, Ishii K, Shibata H (2019) l-Leucine influx through Slc7a5 regulates inflammatory responses of human B cells via mammalian target of rapamycin complex 1 signaling. Mod Rheumatol 29:885–891. https://doi.org/10.1080/14397595.2018.1510822

    Article  CAS  PubMed  Google Scholar 

  20. Toyooka T, Nawashiro H, Shinomiya N, Yano A, Ooigawa H, Ohsumi A, Uozumi Y, Yanagawa Y, Matsuo H, Shima K (2008) Up-regulation of L type amino acid transporter 1 after spinal cord injury in rats. Acta Neurochir Suppl 102:385–388. https://doi.org/10.1007/978-3-211-85578-2_74

    Article  PubMed  Google Scholar 

  21. Usoskin D, Furlan A, Islam S, Abdo H, Lönnerberg P, Lou D, Hjerling-Leffler J, Haeggström J, Kharchenko O, Kharchenko PV, Linnarsson S, Ernfors P (2014) Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat Neurosci 18:145–153. https://doi.org/10.1038/nn.3881

    Article  CAS  PubMed  Google Scholar 

  22. Wempe MF, Rice PJ, Lightner JW, Jutabha P, Hayashi M, Anzai N, Wakui S, Kusuhara H, Sugiyama Y, Endou H (2012) Metabolism and pharmacokinetic studies of JPH203, an L-amino acid transporter 1 (LAT1) selective compound. Drug Metab Pharmacokinet 27:155–161

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Prof. Karin N. Westlund and Dr. Sabrina McIlwrath for assistance with immunohistochemistry experiments. We thank Marena Montera for assistance with western blot experiments and lab management.

Funding

S.R.A.A received funding from the Research Endowment Fund of the Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine. Supported by National Institutes of Health awards (NINDS (NS098772 and NS120663 to RK).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Rajesh Khanna or Sascha R. A. Alles.

Ethics declarations

Conflict of interest

R. Khanna is the co-founder of Regulonix LLC, a company developing non-opioids drugs for chronic pain. In addition, R. Khanna 481 has patents US10287334 (Non-narcotic CRMP2 peptides targeting sodium channels for chronic 482 pain) and US10441586 (SUMOylation inhibitors and uses thereof) issued to Regulonix LLC. R. Khanna is also a co-founder of ElutheriaTx Inc., a company developing gene therapy approaches for chronic pain.

Additional information

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the special issue on Nociception in Pflügers Archiv—European Journal of Physiology

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Goins, A.E., Gomez, K., Ran, D. et al. Neuronal allodynic mechanisms of Slc7a5 (LAT1) in the spared nerve injury rodent model of neuropathic pain. Pflugers Arch - Eur J Physiol 474, 397–403 (2022). https://doi.org/10.1007/s00424-021-02653-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00424-021-02653-9

Keywords

Navigation