Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

PD-L1 inhibits acute and chronic pain by suppressing nociceptive neuron activity via PD-1

An Author Correction to this article was published on 14 January 2019

Abstract

Programmed cell death ligand-1 (PD-L1) is typically produced by cancer cells and suppresses immunity through the receptor PD-1 expressed on T cells. However, the role of PD-L1 and PD-1 in regulating pain and neuronal function is unclear. Here we report that both melanoma and normal neural tissues including dorsal root ganglion (DRG) produce PD-L1 that can potently inhibit acute and chronic pain. Intraplantar injection of PD-L1 evoked analgesia in naive mice via PD-1, whereas PD-L1 neutralization or PD-1 blockade induced mechanical allodynia. Mice lacking Pd1 (Pdcd1) exhibited thermal and mechanical hypersensitivity. PD-1 activation in DRG nociceptive neurons by PD-L1 induced phosphorylation of the tyrosine phosphatase SHP-1, inhibited sodium channels and caused hyperpolarization through activation of TREK2 K+ channels. PD-L1 also potently suppressed nociceptive neuron excitability in human DRGs. Notably, blocking PD-L1 or PD-1 elicited spontaneous pain and allodynia in melanoma-bearing mice. Our findings identify a previously unrecognized role of PD-L1 as an endogenous pain inhibitor and a neuromodulator.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Exogenous PD-L1 inhibits formalin-induced inflammatory pain and increases pain threshold in naive mice.
Figure 2: Endogenous PD-L1 regulates pain sensitivity in naive mice via PD-1.
Figure 3: PD-1 is expressed by mouse DRG neurons and nerve axons.
Figure 4: PD-L1 suppresses neuronal excitability in mouse DRG neurons via PD-1.
Figure 5: PD-L1 inhibits neuronal hyperexcitability and neuropathic pain after nerve injury.
Figure 6: PD-L1 modulates neuronal excitability and pain via SHP-1.
Figure 7: PD-L1 suppresses action potential firing and sodium currents and regulates RMP in human DRG neurons.
Figure 8: Blocking of PD-L1 or PD-1 signaling induces spontaneous pain and allodynia in a mouse melanoma model.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Mantyh, P.W. Cancer pain and its impact on diagnosis, survival and quality of life. Nat. Rev. Neurosci. 7, 797–809 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Mantyh, P. Bone cancer pain: causes, consequences, and therapeutic opportunities. Pain 154 (Suppl. 1), S54–S62 (2013).

    Article  PubMed  Google Scholar 

  3. Selvaraj, D. et al. A functional role for VEGFR1 expressed in peripheral sensory neurons in cancer pain. Cancer Cell 27, 780–796 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jimenez-Andrade, J.M., Ghilardi, J.R., Castañeda-Corral, G., Kuskowski, M.A. & Mantyh, P.W. Preventive or late administration of anti-NGF therapy attenuates tumor-induced nerve sprouting, neuroma formation, and cancer pain. Pain 152, 2564–2574 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cain, D.M. et al. Functional interactions between tumor and peripheral nerve: changes in excitability and morphology of primary afferent fibers in a murine model of cancer pain. J. Neurosci. 21, 9367–9376 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Schweizerhof, M. et al. Hematopoietic colony-stimulating factors mediate tumor-nerve interactions and bone cancer pain. Nat. Med. 15, 802–807 (2009).

    Article  CAS  PubMed  Google Scholar 

  7. Schmidt, B.L. The neurobiology of cancer pain. Neuroscientist 20, 546–562 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Brahmer, J.R. et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 366, 2455–2465 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Negin, B.P. et al. Symptoms and signs of primary melanoma: important indicators of Breslow depth. Cancer 98, 344–348 (2003).

    Article  PubMed  Google Scholar 

  10. Ji, R.R., Chamessian, A. & Zhang, Y.Q. Pain regulation by non-neuronal cells and inflammation. Science 354, 572–577 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sharma, P. & Allison, J.P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Butte, M.J., Keir, M.E., Phamduy, T.B., Sharpe, A.H. & Freeman, G.J. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 27, 111–122 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Keir, M.E., Butte, M.J., Freeman, G.J. & Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Day, C.L. et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443, 350–354 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Herbst, R.S. et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515, 563–567 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Topalian, S.L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ansell, S.M. et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N. Engl. J. Med. 372, 311–319 (2015).

    Article  PubMed  CAS  Google Scholar 

  18. Hamanishi, J. et al. Safety and antitumor activity of anti-PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer. J. Clin. Oncol. 33, 4015–4022 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Postow, M.A. et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N. Engl. J. Med. 372, 2006–2017 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Talbot, S., Foster, S.L. & Woolf, C.J. Neuroimmunity: physiology and pathology. Annu. Rev. Immunol. 34, 421–447 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. McMahon, S.B., La Russa, F. & Bennett, D.L. Crosstalk between the nociceptive and immune systems in host defence and disease. Nat. Rev. Neurosci. 16, 389–402 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Chiu, I.M. et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature 501, 52–57 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ji, R.R., Xu, Z.Z. & Gao, Y.J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 13, 533–548 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Li, Y. et al. Toll-like receptor 4 signaling contributes to paclitaxel-induced peripheral neuropathy. J. Pain 15, 712–725 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xu, Z.Z. et al. Inhibition of mechanical allodynia in neuropathic pain by TLR5-mediated A-fiber blockade. Nat. Med. 21, 1326–1331 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Park, C.K. et al. Extracellular microRNAs activate nociceptor neurons to elicit pain via TLR7 and TRPA1. Neuron 82, 47–54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Berta, T. et al. Extracellular caspase-6 drives murine inflammatory pain via microglial TNF-α secretion. J. Clin. Invest. 124, 1173–1186 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Patel, S.P. & Kurzrock, R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol. Cancer Ther. 14, 847–856 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Weber, J.S. et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 16, 375–384 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Brahmer, J.R., Hammers, H. & Lipson, E.J. Nivolumab: targeting PD-1 to bolster antitumor immunity. Future Oncol. 11, 1307–1326 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Hucho, T. & Levine, J.D. Signaling pathways in sensitization: toward a nociceptor cell biology. Neuron 55, 365–376 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Reichling, D.B. & Levine, J.D. Critical role of nociceptor plasticity in chronic pain. Trends Neurosci. 32, 611–618 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Basbaum, A.I., Bautista, D.M., Scherrer, G. & Julius, D. Cellular and molecular mechanisms of pain. Cell 139, 267–284 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Devor, M., Wall, P.D. & Catalan, N. Systemic lidocaine silences ectopic neuroma and DRG discharge without blocking nerve conduction. Pain 48, 261–268 (1992).

    Article  CAS  PubMed  Google Scholar 

  35. Chen, G., Park, C.K., Xie, R.G. & Ji, R.R. Intrathecal bone marrow stromal cells inhibit neuropathic pain via TGF-β secretion. J. Clin. Invest. 125, 3226–3240 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Todd, A.J. Neuronal circuitry for pain processing in the dorsal horn. Nat. Rev. Neurosci. 11, 823–836 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Braz, J., Solorzano, C., Wang, X. & Basbaum, A.I. Transmitting pain and itch messages: a contemporary view of the spinal cord circuits that generate gate control. Neuron 82, 522–536 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yang, Y. et al. Delayed activation of spinal microglia contributes to the maintenance of bone cancer pain in female Wistar rats via P2X7 receptor and IL-18. J. Neurosci. 35, 7950–7963 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Hebeisen, M. et al. SHP-1 phosphatase activity counteracts increased T cell receptor affinity. J. Clin. Invest. 123, 1044–1056 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bennett, D.L. & Woods, C.G. Painful and painless channelopathies. Lancet Neurol. 13, 587–599 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Acosta, C. et al. TREK2 expressed selectively in IB4-binding C-fiber nociceptors hyperpolarizes their membrane potentials and limits spontaneous pain. J. Neurosci. 34, 1494–1509 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Woolf, C.J. Overcoming obstacles to developing new analgesics. Nat. Med. 16, 1241–1247 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Mogil, J.S. Animal models of pain: progress and challenges. Nat. Rev. Neurosci. 10, 283–294 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Han, Q. et al. SHANK3 deficiency impairs heat hyperalgesia and TRPV1 signaling in primary sensory neurons. Neuron 92, 1279–1293 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kleffel, S. et al. Melanoma cell-intrinsic PD-1 receptor functions promote tumor growth. Cell 162, 1242–1256 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Scholz, J. & Woolf, C.J. The neuropathic pain triad: neurons, immune cells and glia. Nat. Neurosci. 10, 1361–1368 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Uçeyler, N. et al. Deficiency of the negative immune regulator B7-H1 enhances inflammation and neuropathic pain after chronic constriction injury of mouse sciatic nerve. Exp. Neurol. 222, 153–160 (2010).

    Article  PubMed  CAS  Google Scholar 

  48. Guan, Z. et al. Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain. Nat. Neurosci. 19, 94–101 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Grace, P.M., Hutchinson, M.R., Maier, S.F. & Watkins, L.R. Pathological pain and the neuroimmune interface. Nat. Rev. Immunol. 14, 217–231 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Sorge, R.E. et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat. Neurosci. 18, 1081–1083 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study is supported by NIH RO1 grants NS87988, DE17794 and DE22743 and National Science Fund of China (NSFC) 31420103903. Y.H.K. was supported by the National Research Foundation of Korea (NRF) 2013R1A6A3A04065858.

Author information

Authors and Affiliations

Authors

Contributions

G.C. developed the project, performed behavioral and histochemical experiments and prepared the final figures. Y.H.K. conducted electrophysiology in mouse and human DRG neurons. H. Li and H. Luo performed spinal cord recordings and behavioral test in bonce caner model under the guidance of Y.-Q.Z. D.-L.L. performed recordings in whole-mount mouse DRGs. Z.-J.Z. contributed to histochemistry in Pd1 knockout mice. M.L. did some in situ hybridization experiment. W.C. conducted some electrophysiology in mouse DRG neurons. R.-R.J. and Y.-Q.Z. supervised the project. R.-R.J., G.C. and Y.-Q.Z. wrote the paper.

Corresponding authors

Correspondence to Yu-Qiu Zhang or Ru-Rong Ji.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 PD-L1 secretion in melanoma cells and Pdl1 miRNA expression in mouse DRG neurons

(a) ELISA analysis showing PD-L1 secretion in culture medium collected from B16F10 mouse melanoma cell line or control medium (without cells). 1~1.5 × 106 cells were included per well. *P<0.05, two-tailed student t-test, n=3 cultures. Data are mean ± s.e.m. (b,c) In situ hybridization (ISH) image showing Pdl1 mRNA expression in mouse DRG neurons. (b) Left and middle panels, low and high magnification images of ISH with anti-sense probe. Scales, 50 and 20 μm. Right, high magnification image of double ISH and Nissl staining in mouse DRG neurons. Scale, 20 μm. (c) ISH image of sense probe showing the absence of Pdl1 mRNA expression in mouse DRG neurons. Scale, 50 μm.

Supplementary Figure 2 Spontaneous pain and mechanical sensitivity in naive mice and nivolumab binding in mouse DRG neurons and sciatic nerve

(a) Soluble PD-1 (sPD-1, 5 μg, i.pl.) does not induce spontaneous pain. n.s., not significant; n=5 mice/group. (b) Prevention of PD-L1 (5 μg, i.pl.) induced analgesia (increase in paw withdrawal threshold) by pretreatment of RMP1-14 (mouse anti-PD-1 antibody, 5 μg, i.pl.) or Nivolumab (human anti-PD-1 antibody, 10 μg, i.pl.). Human IgG was included as a control. *P<0.05, vs. human IgG/PD-L1, repeated measures Two-Way ANOVA, n = 4 mice/group. Arrows indicate drug injections. Human IgG or monoclonal antibody was injected 30 min prior to the injection of PD-L1. BL, baseline. (c) Nivolumab (10 mg/ml) binds DRG neurons and sciatic nerve axons in WT but not Pd1 KO mice. Nivolumab is detected by the 2nd antibody (mouse monoclonal HP6025 Anti-Human IgG4, FITC; 1.25 mg/ml). Arrows indicate nerve fibers. Data are mean ± s.e.m.

Supplementary Figure 3 PD-L1 or anti-PD-1 treatment fails to change mechanical sensitivity in Pd1−/− mice

(a,b) von Frey test showing the effects of PD-L1 (5 μg, i.pl.) and RMP1-14 (mouse anti-PD-1 antibody, 5 μg, i.pl.) on paw withdrawal threshold in WT and KO mice. (a) PD-L1 increases withdrawal threshold in WT but not KO mice. (b) RMP1-14 decreases withdrawal threshold in WT but not KO mice. *P<0.05, #P<0.05, vs. baseline (BL), two-way repeated ANOVA, n = 6 mice/group. Data are mean ± s.e.m.

Supplementary Figure 4 Pd1−/− mice display normal central innervations in the spinal cord dorsal horn

(a) Immunostaining of IB4, CGRP and NF200 on L4-spinal cord sections from WT or Pd1−/− mice. Scale bar, 100 μm. Note the central innervations of primary afferents in the spinal cord are normal in KO mice. (b) Nissl staining on L4-spinal cord sections showing normal morphology of spinal cord dorsal horn of KO mice. Scale bar, 100 μm. (c) Quantification of immunofluorescence of IB4, CGRP, and NF200 staining in the dorsal horn of WT and KO mice. n.s., not significant., Two-tailed Student’s t-test, n = 4 mice/group. Three to five sections from each animal were included for quantification. Data are mean ± s.e.m.

Supplementary Figure 5 Pd1−/− mice display normal distribution patterns of C-fiber and A-fiber neurons and have no neuronal loss in DRGs

(a) Immunostaining of IB4, CGRP and NF200 and Nissl staining on L4-DRG sections from WT or Pd1−/− mice. Scale bar, 100 μm. Note that KO mice have normal distribution patterns of different sensory populations of DRG neurons. (b, c) Quantification of the percentages of IB4-binding, CGRP-IR, and NF200-IR neurons (b) and the total numbers of neurons with Nissl staining (c) in DRG sections from WT and Pd1−/− mice. All the DRG sections (14 μm) were collected and every 5th section was used for respective immunostaining or Nissl staining. n = 4 mice/group. n.s., not significant; two-tailed Student’s t-test. Data are mean ± s.e.m.

Supplementary Figure 6 Spinal application of PD-L1 suppresses excitatory synaptic transmission in lamina IIo neurons in spinal cord slices and inhibits neuropathic pain and baseline pain in mice

(a-d) Patch clamp recordings of excitatory synaptic transmission and quantification of frequency and amplitude of spontaneous excitatory postsynaptic synaptic currents (sEPSCs) in lamina IIo neurons of spinal cord slices of naïve mice. (a) Perfusion with PD-L1 (30 ng/ml) reduces the frequency and amplitude of sEPSCs. Left, traces of sEPSCs before (1) and after (2) PD-L1 perfusion. Right, frequency (upper) and amplitude (bottom) of sEPSCs. *P < 0.05, before vs. after the treatment, paired two-tailed Student’s t-test, n =14 neurons/3-4 mice. (b) Perfusion with sPD-1 (30 ng/ml) increases the frequency (upper) not the amplitude (bottom) of sEPSCs. *P < 0.05, compared with control, n.s., no significance, paired two-tailed Student’s t-test, n = 9 neurons/3 mice. (c) Incubation of spinal cord slices with Nivolumab (300 ng/ml, 3 h) increases the frequency of sEPSCs. *P < 0.05, one-way ANOVA, followed by Bonferroni’s post-hoc test, n = 21 neurons/3-4 mice. (d) Incubation with Nivolumab (300 ng/ml, 3 h) blocks the effects of PD-L1. n.s., no significance, n = 6 neurons/3 mice. The data are mean ± s.e.m.

Supplementary Figure 7 Spinal application of PD-L1 inhibits mechanical hypersensitivity and firing of spinal WDR neurons in a model of bone cancer in rats.

(a) Inhibition of bone cancer-induced mechanical allodynia by i.t. PD-L1 in rats. *P < 0.05, vs. baseline (BL), §P < 0.05, vs. pre-injection baseline on post-tumor implantation day 14 (PTD 14), #P < 0.05, vs. vehicle, repeated measures two-Way ANOVA, n = 5 rats/group. Arrow indicates drug injection. (b,c) Suppression of brush, von Frey filaments, and pinch evoked spikes of spinal WDR neurons by PD-L1 (20 μg, i.t., 3 h) on post-tumor implantation day 14. (b) Histograms of evoked spikes of WDR neuron firing by brush, von Frey filaments, and pinch stimulation. (c) Mean spikes of WDR neurons following low and high intensity mechanical stimuli. *P < 0.05, vs. vehicle control, paired student’s t-test, n = 5 rats/group. (d,e) Enhancement of brush, von Frey filaments, and pinch evoked spikes of WDR neurons by intrathecal Nivolumab (10 μg, 1 h) on post-tumor implantation day 8. (d) Histograms of evoked spikes of WDR neurons firing by brush, von Frey filaments, and pinch stimulation. (e) Mean spikes of WDR neurons following low and high intensity mechanical stimuli. *P < 0.05, vs. vehicle; #P < 0.05 vs. human IgG, one-Way ANOVA, n = 4-5 rats/group. Data are mean ± s.e.m.

Supplementary Figure 8 PD-L1 induces phosphorylation of SHP1 in mouse DRG neurons

(a) Double IHC and ISH staining shows co-localization of pSHP-1 and Pd1 mRNA in DRG neurons 30 min after intrathecal PD-L1 injection (1 μg). Scale, 50 μm. (b) Enlarged images of the boxes in a. Arrows indicate the double-labeled neurons. Scale, 20 μm. (c) PD-L1 treatment (10 ng/ml, 30 min) increases phosphorylation of SHP-1 (pSHP-1) in dissociated mouse DRG neurons. Left, pSHP-1 immunostaining and the effects of PD-L1 and the SHP-1 inhibitor SSG (11 μM). Scales, 50 μm. Right, intensity of immunofluorescence of pSHP-1-positive neurons. *P < 0.05, n = 98-104 neurons from 3 separate dishes, one-Way ANOVA, n.s., no significance. Data are mean ± s.e.m.

Supplementary Figure 9 TREK2 activation by PD-L1 in CHO cells and schematic illustration of PD-L1-induced silencing of nociceptive neurons

(a) TREK2 immunostaining in mouse DRG. Scale, 50 μm. (b) PD-L1 increases TREK2-mediated outward currents (up-left and up-right), causes a negative shift in the reversal potential (Erev) of the outward currents (low-left), and changes RMP (low-right) via PD-1. CHO cells were co-transfected with KCNK10 cDNA (encoding TREK2) and PDCD1 cDNA (encoding PD-1) or transfected with KCNK10 or PDCD1 cDNA alone. Note that PD-L1 fails to cause these changes when CHO cells only express TREK2 or PD-1. *P < 0.05, One-Way ANOVA, n = 6-8 cells/2 cultures. Also see Figure 6d,e. Data are mean ± s.e.m. (c) Schematic illustration of the mechanisms by which PD-L1 silences nociceptive neurons, via suppression of action potentials through voltage-gated sodium channels (VGSCs) or/and via hyperpolarization through TREK2 potassium channels.

Supplementary Figure 10 PD-1 immunofluorescence in DRG neurons and nerve axons of human tissue sections

(a,b) PD-1 immunofluorescence in human DRG neurons and dorsal root axons. Right panels in (a) and (b) showing the absence of PD-1 immunostaining by the blocking peptide. Blue DAPI staining shows all cell nuclei in DRG and nerve sections. Scales, 50 μm. (c) Double immunostaining of PD-1 and NF200 in human spinal nerve axons. The Box in the left panel is enlarged in three panels. Scales, 50 μm. Arrows indicate the double-labeled axon.

Supplementary Figure 11 Intraplantar (i.pl.) injection of soluble PD-1 (sPD-1) evokes spontaneous pain and mechanical allodynia in melanoma mice

(a,b) Induction of spontaneous pain (a, flinching/licking behavior) and evoked pain (b, mechanical allodynia) by soluble PD-1 (sPD-1) following i.pl. injection at MCI-4w. Note a rapid onset of spontaneous pain by sPD-1 within 30 min. Arrow indicates drug injection. *P<0.05, compared with vehicle, repeated measures two-way ANOVA (b). n = 6 and 7 mice per group.

Supplementary Figure 12 Intraplantar (i.pl.) injection of soluble PD-1 (sPD1) does not change immune responses in melanoma-bearing hindpaw skin in the acute phase

(a) sPD1 (5 μg, i.pl.), given to melanoma mice at 4w does not change immune responses in hindpaw skins at 3 h after the injection. Note that relative mRNA levels (shown by quantitative RT-PCR and normalized with GAPDH) of TNF, IL-1B, Il-6, IFNG, CCL2, CD2, CD8, and CD68 all increase in tumor bearing skins. *P<0.05, vs. contralateral control (for all 8 markers), n.s., no significance (for all 8 markers), one-Way ANOVA, n = 5 mice/group. All the data are expressed as mean ± s.e.m. (b) Primer sequences used for qPCR in the experiment.

Supplementary Figure 13 Pd1-targeting siRNA decreases PD-1 expression in DRG and sciatic nerve but not spinal cord dorsal horn tissues

(a) Western blot analysis showing the effects of Pd1-targeting siRNA and non-targeting (NT) control siRNA on PD-1 expression in DRG, sciatic nerve, and spinal cord tissues. Low panels, quantification of PD-1 expression in different mouse tissues. *P < 0.05, PD-1 vs. NT, n.s., no significance, two-tailed student’s t-test, n = 5 mice/group. siRNA was applied via peri sciatic injection (2 μg) given at MCI-4w. (b) Uncut gels for PD-1 and GAPDH western blots of DRG, sciatic nerve, and dorsal horn tissues. The represented blots are highlighted in the red boxes, respectively. The images (flipped) show non-targeting (NT) siRNA treatment on the left and PD-1 siRNA treatment on the right.

Supplementary Figure 14 Induction of mechanical allodynia and spontaneous pain by systemic or local injection of anti-PD-1 antibodies and SHP-1 inhibitor in melanoma mice

(a,b) Intravenous RMP1-14 (mouse anti-PD-1 antibody, 10 mg/kg) induces mechanical allodynia (a) and spontaneous pain (b) in melanoma-bearing mice at MCI-4w. *P < 0.05, compared to control rat IgG2A, two-way ANOVA, repeated measures (a), or student’s t-test (b), n = 6 mice/group. Drug injection is indicated by arrow. (c) Intrathecal injection of Nivolumab (1 and 10 μg, n = 6 and 7 mice/group), given at MCI-4w (shown with the arrow), induces mechanical allodynia. *P < 0.05, compared with control human IgG4, repeated measures two-way ANOVA. (d) Intraplantar injection of Nivolumab (10 μg, n = 4 and 5 mice/group), given at MCI-4w (shown with the arrow), induces mechanical allodynia. *P < 0.05, compared with control human IgG4, repeated measures two-way ANOVA. (e) Induction of spontaneous pain (flinching/licking behavior) by SHP-1 inhibitor SSG (5 μg, i.pl.) given at MCI-4w. *P < 0.05, two-tailed student’s t-test. n = 5 mice/group. (f) Schematic illustration of PD-L1 evoked pain masking in melanoma. The pharmacological agents used in this study for targeting the PD-L1/PD-1 pathway are highlighted in purple. Data are mean ± s.e.m.

Supplementary Figure 15 Numbers of animals (mice and rats) used in this study

A total of 528 mice and 51 rats were used in this study. We also collected human DRGs from 9 donors.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 (PDF 2961 kb)

Supplementary Methods Checklist (PDF 373 kb)

Spontaneous pain in top left mouse

Video 1 (40 s) shows spontaneous pain in top left mouse. The bottom two control mice received vehicle injection and did not show spontaneous pain, despite massive growth of melanoma in a hindpaw. (AVI 19097 kb)

Spontaneous pain in top right mouse

Video 2 (40 s) shows spontaneous pain in top right mouse. The bottom two control mice received vehicle injection and did not show spontaneous pain, despite massive growth of melanoma in a hindpaw. (AVI 18443 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, G., Kim, Y., Li, H. et al. PD-L1 inhibits acute and chronic pain by suppressing nociceptive neuron activity via PD-1. Nat Neurosci 20, 917–926 (2017). https://doi.org/10.1038/nn.4571

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4571

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer