Cannabinoid receptor 2‑selective agonist JWH015 attenuates bone cancer pain through the amelioration of impaired autophagy flux induced by inflammatory mediators in the spinal cord

Yanting Mao, Yulin Huang, Ying Zhang, Chenchen Wang, Hao Wu, Xinyu Tian, Yue Liu, Bailing Hou, Ying Liang, Hui Rong, Xiaoping Gu, Zhengliang Ma, Yanting Mao, Yulin Huang, Ying Zhang, Chenchen Wang, Hao Wu, Xinyu Tian, Yue Liu, Bailing Hou, Ying Liang, Hui Rong, Xiaoping Gu, Zhengliang Ma

Abstract

Bone cancer pain (BCP) is a severe complication of advanced bone cancer. Although cannabinoid receptor 2 (CB2) agonists may have an analgesic effect, the underlying mechanism remains unclear. CB2 serves a protective role in various pathological states through the activation of autophagy. Therefore, the present study aimed to determine whether the analgesic effects of the selective CB2 agonist JWH015 was mediated by the activation of autophagy in BCP. BCP was induced by the intra‑femur implantation of NCTC2472 fibrosarcoma cells in C3H/HeN mice. The pain behaviors were assessed on the following postoperative days. The selective CB2 agonist JWH015 (1 and 2 µg) was intrathecally administered on day 14 following implantation. AM630 (1 µg), a CB2 antagonist, was injected 30 min before JWH015 administration. Lipopolysaccharide (LPS; 100 nM)‑stimulated primary neurons were treated with JWH015 (1 µM) and AM630 (1 µM) to further verify the mechanism by which CB2 affects autophagy. The results demonstrated that autophagy flux was impaired in spinal neurons during BCP, as indicated by the increased ratio of microtubule‑associated protein 1 light chain 3β (LC3B)‑II/LC3B‑I and increased expression of p62. Intrathecal administration of JWH015 attenuated BCP, which was accompanied by the amelioration of impaired autophagy flux (decreased LC3B‑II/LC3B‑I ratio and decreased p62expression). In addition, the activation of glia cells and upregulation of the glia‑derived inflammatory mediators, interleukin (IL)‑1β and IL‑6 were suppressed by JWH015. In LPS‑stimulated primary neurons, IL‑1β and IL‑6 were increased, and autophagy flux was impaired; whereas treatment with JWH015 decreased the expression of IL‑1β and IL‑6, LC3B‑II/LC3B‑I ratio and expression of p62. These effects were by pretreatment with the CB2‑selective antagonist AM630. The results of the present study suggested that the impairment of autophagy flux was induced by glia‑derived inflammatory mediators in spinal neurons. Intrathecal administration of the selective CB2 agonist JWH015 ameliorated autophagy flux through the downregulation of IL‑1β and IL‑6 and attenuated BCP.

Figures

Figure 1.
Figure 1.
Intrathecal administration of JWH015 attenuates bone cancer pain. Intra-femur implantation of NCTC 2476 cells induced mechanical hypersensitivity in the ipsilateral hind paw. The (A) PWMT and (B) NSF were measured on days 0, 4, 7, 10, 14, 21 and 28 after operation in Control, Sham and Tumor group mice. JWH015 and AM630 were intrathecally injected on day 14 after the operation. (C) PWMT and (D) NSF were tested before administration (baseline, 0 h) and at 4, 8, 12, 24, 48 and 72 h after administration of JWH015. Data are expressed as the mean ± SD; n=8; *P#P<0.05 vs. Sham at each time point. PWMT, paw withdrawal mechanical threshold; NSF, Number of spontaneous flinches.
Figure 2.
Figure 2.
Autophagy flux is impaired in bone cancer pain mice. (A) Representative western blotting images of LC3BI/II and p62 protein expression in the spinal cord of mice in the Sham and Tumor groups on day 0, 4, 7, 10, 14, 21 and 28 post-operation. (B) Ratio of LC3B-II/LC3B-I and (C) quantification of p62 in the spinal cord in BCP. *P

Figure 3.

Autophagy flux is impaired in…

Figure 3.

Autophagy flux is impaired in neurons in the spinal cord. (A-C) LC3B-immunoreactive cells…

Figure 3.
Autophagy flux is impaired in neurons in the spinal cord. (A-C) LC3B-immunoreactive cells (green) in the spinal cord were (A) NeuN-positive (red), (B) Iba1-negative (red), and (C) GFAP-negative (red). Scale bar, 50 µm; magnification, ×400. GFAP, glial fibrillary acidic protein; Iba1, ionized calcium-binding adaptor molecule 1; LC3B, microtubule-associated protein 1 light chain 3β; NeuN, neuronal nuclei antigen.

Figure 4.

Intrathecal administration of JWH015 ameliorates…

Figure 4.

Intrathecal administration of JWH015 ameliorates impaired autophagy flux in bone cancer pain. (A)…

Figure 4.
Intrathecal administration of JWH015 ameliorates impaired autophagy flux in bone cancer pain. (A) Representative western blotting images of LC3B and p62 in the spinal cord 12 h after treatment with JWH015. (B) Ratio of LC3B-II/LC3B-I and (C) quantification of p62 protein expression levels. #P<0.05 vs. Sham + vehicle group; *P<0.05 vs. Tumor + vehicle group. (D) Representative western blots of LC3B and p62 protein expression in the spinal cord at 0, 4, 8, 12, 24, 48 and 72 h after JWH015 (2 µg) injection. (E) Ratio of LC3B-II/LC3B-I and (F) quantification of p62. *P<0.05 vs. 0 h. (G) Representative blots of LC3B and p62 in the spinal cord with treatment of vehicle, Baf-A1 (10 nM), JWH015 (2 µg), and Baf-A1 (10 nM) + JWH015 (2 µg) in BCP mice on day 14. (H) Ratio of LC3B-II/LC3B-I. (I) Quantification of p62. *P<0.05 vs. tumor + vehicle group. For all western blots, β-actin was used as a loading control; data are expressed as the mean ± SD n=3 per group. LC3B, microtubule-associated protein 1 light chain 3β.

Figure 5.

Intrathecal administration of JWH015 inhibits…

Figure 5.

Intrathecal administration of JWH015 inhibits activation of astrocytes and microglia induced by bone…

Figure 5.
Intrathecal administration of JWH015 inhibits activation of astrocytes and microglia induced by bone cancer pain. (A) Images and quantification of immunostaining for GFAP (red) and Iba1 (green) in lumbar spinal dorsal horn were captured on day 14 after operation in mice in the Sham and Tumor groups. n=3; *P#P<0.05 vs. Sham + vehicle group; *P<0.05 vs. Tumor + vehicle group; n=3 per group. Scale bar, 50 µm; magnification, × 200. GFAP, glial fibrillary acidic protein; Iba1, ionized calcium binding adaptor molecule 1.

Figure 6.

Intrathecal administration of JWH015 downregulated…

Figure 6.

Intrathecal administration of JWH015 downregulated the expression of pro-inflammatory mediators IL-1β and IL-6.…

Figure 6.
Intrathecal administration of JWH015 downregulated the expression of pro-inflammatory mediators IL-1β and IL-6. (A) Representative western blotting images and (B) quantification of IL-1β and IL-6 protein expression in the spinal cord in Sham and Tumor mice on day 0, 4, 7, 10, 14, 21 and 28 after the operation. *P#P<0.05 vs. Sham + vehicle group; *P<0.05 vs. Tumor + vehicle. (E) Representative blots and (F) quantification of IL-1β and IL-6 in the spinal cord at 0, 4, 8, 12, 24, 48 and 72 h after JWH015 (2 µg) injection. *P<0.05 vs. 0 h. For all blots, β-actin was used as a loading control; data are expressed as the mean ± SD; n=3. IL, interleukin.

Figure 7.

Effects of JWH015 on inflammatory…

Figure 7.

Effects of JWH015 on inflammatory factors and autophagy in primary neurons. Primary neurons…

Figure 7.
Effects of JWH015 on inflammatory factors and autophagy in primary neurons. Primary neurons were stimulated with LPS (100 nM) for 0, 3, 6, 9, 12 and 24 h. (A) Western blotting demonstrated the upregulation of IL-1β and IL-6 after LPS-stimulation. (B) Quantification of IL-1β and IL-6 in the primary neurons after LPS-stimulation. *P#P<0.05 vs. LPS group; $P<0.05 vs. LPS + JWH015 group. (G) Decreased ratio of LC3B-II/LC3B-I and decreased expression of p62 after treatment with JWH015 in LPS-stimulated primary neurons. (H) Quantification of LC3B-II/LC3B-I and p62 in different groups. *P<0.05 vs. control group; #P<0.05 vs. LPS group; $P<0.05 vs. LPS + JWH015 group. In all western blots, β-actin was used as a loading control; data are expressed as the mean ± SD; n=3. IL, interleukin; LC3B, microtubule-associated protein 1 light chain 3β; LPS, lipopolysaccharide.
All figures (7)
Figure 3.
Figure 3.
Autophagy flux is impaired in neurons in the spinal cord. (A-C) LC3B-immunoreactive cells (green) in the spinal cord were (A) NeuN-positive (red), (B) Iba1-negative (red), and (C) GFAP-negative (red). Scale bar, 50 µm; magnification, ×400. GFAP, glial fibrillary acidic protein; Iba1, ionized calcium-binding adaptor molecule 1; LC3B, microtubule-associated protein 1 light chain 3β; NeuN, neuronal nuclei antigen.
Figure 4.
Figure 4.
Intrathecal administration of JWH015 ameliorates impaired autophagy flux in bone cancer pain. (A) Representative western blotting images of LC3B and p62 in the spinal cord 12 h after treatment with JWH015. (B) Ratio of LC3B-II/LC3B-I and (C) quantification of p62 protein expression levels. #P<0.05 vs. Sham + vehicle group; *P<0.05 vs. Tumor + vehicle group. (D) Representative western blots of LC3B and p62 protein expression in the spinal cord at 0, 4, 8, 12, 24, 48 and 72 h after JWH015 (2 µg) injection. (E) Ratio of LC3B-II/LC3B-I and (F) quantification of p62. *P<0.05 vs. 0 h. (G) Representative blots of LC3B and p62 in the spinal cord with treatment of vehicle, Baf-A1 (10 nM), JWH015 (2 µg), and Baf-A1 (10 nM) + JWH015 (2 µg) in BCP mice on day 14. (H) Ratio of LC3B-II/LC3B-I. (I) Quantification of p62. *P<0.05 vs. tumor + vehicle group. For all western blots, β-actin was used as a loading control; data are expressed as the mean ± SD n=3 per group. LC3B, microtubule-associated protein 1 light chain 3β.
Figure 5.
Figure 5.
Intrathecal administration of JWH015 inhibits activation of astrocytes and microglia induced by bone cancer pain. (A) Images and quantification of immunostaining for GFAP (red) and Iba1 (green) in lumbar spinal dorsal horn were captured on day 14 after operation in mice in the Sham and Tumor groups. n=3; *P#P<0.05 vs. Sham + vehicle group; *P<0.05 vs. Tumor + vehicle group; n=3 per group. Scale bar, 50 µm; magnification, × 200. GFAP, glial fibrillary acidic protein; Iba1, ionized calcium binding adaptor molecule 1.
Figure 6.
Figure 6.
Intrathecal administration of JWH015 downregulated the expression of pro-inflammatory mediators IL-1β and IL-6. (A) Representative western blotting images and (B) quantification of IL-1β and IL-6 protein expression in the spinal cord in Sham and Tumor mice on day 0, 4, 7, 10, 14, 21 and 28 after the operation. *P#P<0.05 vs. Sham + vehicle group; *P<0.05 vs. Tumor + vehicle. (E) Representative blots and (F) quantification of IL-1β and IL-6 in the spinal cord at 0, 4, 8, 12, 24, 48 and 72 h after JWH015 (2 µg) injection. *P<0.05 vs. 0 h. For all blots, β-actin was used as a loading control; data are expressed as the mean ± SD; n=3. IL, interleukin.
Figure 7.
Figure 7.
Effects of JWH015 on inflammatory factors and autophagy in primary neurons. Primary neurons were stimulated with LPS (100 nM) for 0, 3, 6, 9, 12 and 24 h. (A) Western blotting demonstrated the upregulation of IL-1β and IL-6 after LPS-stimulation. (B) Quantification of IL-1β and IL-6 in the primary neurons after LPS-stimulation. *P#P<0.05 vs. LPS group; $P<0.05 vs. LPS + JWH015 group. (G) Decreased ratio of LC3B-II/LC3B-I and decreased expression of p62 after treatment with JWH015 in LPS-stimulated primary neurons. (H) Quantification of LC3B-II/LC3B-I and p62 in different groups. *P<0.05 vs. control group; #P<0.05 vs. LPS group; $P<0.05 vs. LPS + JWH015 group. In all western blots, β-actin was used as a loading control; data are expressed as the mean ± SD; n=3. IL, interleukin; LC3B, microtubule-associated protein 1 light chain 3β; LPS, lipopolysaccharide.

References

    1. Mantyh P. Bone cancer pain: Causes, consequences, and therapeutic opportunities. Pain. 2013;154(Suppl 1):S54–S62. doi: 10.1016/j.pain.2013.07.044.
    1. Mantyh PW. Cancer pain and its impact on diagnosis, survival and quality of life. Nat Rev Neurosci. 2006;7:797–809. doi: 10.1038/nrn1914.
    1. Honore P, Luger NM, Sabino MA, Schwei MJ, Rogers SD, Mach DB, O'keefe PF, Ramnaraine ML, Clohisy DR, Mantyh PW. Osteoprotegerin blocks bone cancer-induced skeletal destruction, skeletal pain and pain-related neurochemical reorganization of the spinal cord. Nat Med. 2000;6:521–528. doi: 10.1038/74999.
    1. Miller K, Steger GG, Niepel D, Luftner D. Harnessing the potential of therapeutic agents to safeguard bone health in prostate cancer. Prostate Cancer Prostatic Dis. 2018;21:461–472. doi: 10.1038/s41391-018-0060-y.
    1. Ji RR, Xu ZZ, Gao YJ. Emerging targets in neuroinflammation-driven chronic pain. Nat Rev Drug Discov. 2014;13:533–548. doi: 10.1038/nrd4334.
    1. Ligresti A, De Petrocellis L, Di Marzo V. From phytocannabinoids to cannabinoid receptors and endocannabinoids: Pleiotropic physiological and pathological roles through complex pharmacology. Physiol Rev. 2016;96:1593–1659. doi: 10.1152/physrev.00002.2016.
    1. Kendall DA, Yudowski GA. Cannabinoid receptors in the central nervous system: Their signaling and roles in disease. Front Cell Neurosci. 2016;10:294.
    1. Devane WA, Dysarz FA, III, Johnson MR, Melvin LS, Howlett AC. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol. 1988;34:605–613.
    1. Pernia-Andrade AJ, Kato A, Witschi R, Nyilas R, Katona I, Freund TF, Watanabe M, Filitz J, Koppert W, Schüttler J, et al. Spinal endocannabinoids and CB1 receptors mediate C-fiber-induced heterosynaptic pain sensitization. Science. 2009;325:760–764. doi: 10.1126/science.1171870.
    1. Racz I, Nadal X, Alferink J, Baños JE, Rehnelt J, Martín M, Pintado B, Gutierrez-Adan A, Sanguino E, Manzanares J, et al. Crucial role of CB(2) cannabinoid receptor in the regulation of central immune responses during neuropathic pain. J Neurosci. 2008;28:12125–12135. doi: 10.1523/JNEUROSCI.3400-08.2008.
    1. Pertwee RG. Targeting the endocannabinoid system with cannabinoid receptor agonists: Pharmacological strategies and therapeutic possibilities. Philos Trans R Soc Lond B Biol Sci. 2012;367:3353–3363. doi: 10.1098/rstb.2011.0381.
    1. Fernandez-Ruiz J, Romero J, Velasco G, Tolon RM, Ramos JA, Guzman M. Cannabinoid CB2 receptor: A new target for controlling neural cell survival? Trends Pharmacol Sci. 2007;28:39–45. doi: 10.1016/j.tips.2006.11.001.
    1. Kalant H. Adverse effects of cannabis on health: An update of the literature since 1996. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:849–863. doi: 10.1016/j.pnpbp.2004.05.027.
    1. Gao F, Xiang HC, Li HP, Jia M, Pan XL, Pan HL, Li M. Electroacupuncture inhibits NLRP3 inflammasome activation through CB2 receptors in inflammatory pain. Brain Behav Immun. 2018;67:91–100. doi: 10.1016/j.bbi.2017.08.004.
    1. Niu J, Huang D, Zhou R, Yue M, Xu T, Yang J, He L, Tian H, Liu X, Zeng J. Activation of dorsal horn cannabinoid CB2 receptor suppresses the expression of P2Y12 and P2Y13 receptors in neuropathic pain rats. J Neuroinflammation. 2017;14:185. doi: 10.1186/s12974-017-0960-0.
    1. La Porta C, Bura SA, Aracil-Fernandez A, Manzanares J, Maldonado R. Role of CB1 and CB2 cannabinoid receptors in the development of joint pain induced by monosodium iodoacetate. Pain. 2013;154:160–174. doi: 10.1016/j.pain.2012.10.009.
    1. Li AL, Lin X, Dhopeshwarkar AS, Thomaz AC, Carey LM, Liu Y, Nikas SP, Makriyannis A, Mackie K, Hohmann AG. Cannabinoid CB2 Agonist AM1710 differentially suppresses distinct pathological pain states and attenuates morphine tolerance and withdrawal. Mol Pharmacol. 2019;95:155–168. doi: 10.1124/mol.118.113233.
    1. Wu J, Hocevar M, Bie B, Foss JF, Naguib M. Cannabinoid Type 2 receptor system modulates Paclitaxel-induced microglial dysregulation and central sensitization in rats. J Pain. 2018;20:501–514. doi: 10.1016/j.jpain.2018.10.007.
    1. Feng XL, Deng HB, Wang ZG, Wu Y, Ke JJ, Feng XB. Suberoylanilide hydroxamic acid triggers autophagy by influencing the mTOR pathway in the spinal dorsal horn in a rat neuropathic pain model. Neurochem Res. 2018;44:450–464. doi: 10.1007/s11064-018-2698-1.
    1. Coccurello R, Nazio F, Rossi C, De Angelis F, Vacca V, Giacovazzo G, Procacci P, Magnaghi V, Ciavardelli D, Marinelli S. Effects of caloric restriction on neuropathic pain, peripheral nerve degeneration and inflammation in normometabolic and autophagy defective prediabetic Ambra1 mice. PLoS One. 2018;13:e0208596. doi: 10.1371/journal.pone.0208596.
    1. Shao BZ, Wei W, Ke P, Xu ZQ, Zhou JX, Liu C. Activating cannabinoid receptor 2 alleviates pathogenesis of experimental autoimmune encephalomyelitis via activation of autophagy and inhibiting NLRP3 inflammasome. CNS Neurosci Ther. 2014;20:1021–1028. doi: 10.1111/cns.12349.
    1. Denaes T, Lodder J, Chobert MN, Ruiz I, Pawlotsky JM, Lotersztajn S, Teixeira-Clerc F. The Cannabinoid Receptor 2 protects against alcoholic liver disease via a macrophage autophagy-dependent pathway. Sci Rep. 2016;6:28806. doi: 10.1038/srep28806.
    1. Vara D, Salazar M, Olea-Herrero N, Guzman M, Velasco G, Diaz-Laviada I. Anti-tumoral action of cannabinoids on hepatocellular carcinoma: Role of AMPK-dependent activation of autophagy. Cell Death Differ. 2011;18:1099–1111. doi: 10.1038/cdd.2011.67.
    1. Salazar M, Carracedo A, Salanueva IJ, Hernández-Tiedra S, Lorente M, Egia A, Vázquez P, Blázquez C, Torres S, García S, et al. Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest. 2009;119:1359–1372. doi: 10.1172/JCI37948.
    1. Netea-Maier RT, Plantinga TS, van de Veerdonk FL, Smit JW, Netea MG. Modulation of inflammation by autophagy: Consequences for human disease. Autophagy. 2016;12:245–260. doi: 10.1080/15548627.2015.1071759.
    1. Tian R, Li Y, Yao X. PGRN suppresses inflammation and promotes autophagy in keratinocytes through the Wnt/β-Catenin signaling pathway. Inflammation. 2016;39:1387–1394. doi: 10.1007/s10753-016-0370-y.
    1. Sil S, Niu F, Tom E, Liao K, Periyasamy P, Buch S. Cocaine mediated neuroinflammation: Role of dysregulated autophagy in pericytes. Mol Neurobiol. 2019;56:3576–3590. doi: 10.1007/s12035-018-1325-0.
    1. Piippo N, Korhonen E, Hytti M, Kinnunen K, Kaarniranta K, Kauppinen A. Oxidative stress is the principal contributor to inflammasome activation in retinal pigment epithelium cells with defunct proteasomes and autophagy. Cell Physiol Biochem. 2018;49:359–367. doi: 10.1159/000492886.
    1. Lu C, Liu Y, Sun B, Sun Y, Hou B, Zhang Y, Ma Z, Gu X. Intrathecal injection of JWH-015 attenuates bone cancer pain via time-dependent modification of pro-inflammatory cytokines expression and astrocytes activity in spinal cord. Inflammation. 2015;38:1880–1890. doi: 10.1007/s10753-015-0168-3.
    1. Demers G, Griffin G, De Vroey G, Haywood JR, Zurlo J, Bédard M. Animal research. Harmonization of animal care and use guidance. Science. 2006;312:700–701. doi: 10.1126/science.1124036.
    1. Schwei MJ, Honore P, Rogers SD, Salak-Johnson JL, Finke MP, Ramnaraine ML, Clohisy DR, Mantyh PW. Neurochemical and cellular reorganization of the spinal cord in a murine model of bone cancer pain. J Neurosci. 1999;19:10886–10897. doi: 10.1523/JNEUROSCI.19-24-10886.1999.
    1. Lu C, Shi L, Sun B, Zhang Y, Hou B, Sun Y, Ma Z, Gu X. A single intrathecal or intraperitoneal injection of CB2 receptor agonist attenuates bone cancer pain and induces a time-dependent modification of GRK2. Cell Mol Neurobiol. 2017;37:101–109. doi: 10.1007/s10571-016-0349-0.
    1. Gu X, Mei F, Liu Y, Zhang R, Zhang J, Ma Z. Intrathecal administration of the cannabinoid 2 receptor agonist JWH015 can attenuate cancer pain and decrease mRNA expression of the 2B subunit of N-methyl-D-aspartic acid. Anesth Analg. 2011;113:405–411. doi: 10.1213/ANE.0b013e31821d1062.
    1. Hylden JL, Wilcox GL. Intrathecal morphine in mice: A new technique. Eur J Pharmacol. 1980;67:313–316. doi: 10.1016/0014-2999(80)90515-4.
    1. Mizushima N, Klionsky DJ. Protein turnover via autophagy: Implications for metabolism. Annu Rev Nutr. 2007;27:19–40. doi: 10.1146/annurev.nutr.27.061406.093749.
    1. Bjorkoy G, Lamark T, Brech A, Outzen H, Perander M, Overvatn A, Stenmark H, Johansen T. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol. 2005;171:603–614. doi: 10.1083/jcb.200507002.
    1. Hu Y, Li G, Zhang Y, Liu N, Zhang P, Pan C, Nie H, Li Q, Tang Z. Upregulated TSG-6 expression in ADSCs inhibits the BV2 Microglia-mediated inflammatory response. Biomed Res Int. 2018;2018:7239181. doi: 10.1155/2018/7239181.
    1. Ye J, Guan M, Lu Y, Zhang D, Li C, Zhou C. Arbutin attenuates LPS-induced lung injury via Sirt1/Nrf2/NF-kappaBp65 pathway. Pulm Pharmacol Ther. 2019;54:53–59. doi: 10.1016/j.pupt.2018.12.001.
    1. Rubens RD. Bone metastases-the clinical problem. Eur J Cancer. 1998;34:210–213. doi: 10.1016/S0959-8049(97)10128-9.
    1. Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: A fatal attraction. Nat Rev Cancer. 2011;11:411–425. doi: 10.1038/nrc3055.
    1. Sturge J, Caley MP, Waxman J. Bone metastasis in prostate cancer: Emerging therapeutic strategies. Nat Rev Clin Oncol. 2011;8:357–368. doi: 10.1038/nrclinonc.2011.127.
    1. Li MH, Suchland KL, Ingram SL. Compensatory Activation of cannabinoid CB2 receptor inhibition of GABA release in the rostral ventromedial medulla in inflammatory pain. J Neurosci. 2017;37:626–636. doi: 10.1523/JNEUROSCI.1310-16.2016.
    1. Luongo L, Palazzo E, Tambaro S, Giordano C, Gatta L, Scafuro MA, Rossi FS, Lazzari P, Pani L, de Novellis V, et al. 1-(2′,4′-dichlorophenyl)-6-methyl-N-cyclohexylamine-1,4-dihydroindeno[1,2-c]pyrazole-3-carboxamide, a novel CB2 agonist, alleviates neuropathic pain through functional microglial changes in mice. Neurobiol Dis. 2010;37:177–185. doi: 10.1016/j.nbd.2009.09.021.
    1. Glick D, Barth S, Macleod KF. Autophagy: Cellular and molecular mechanisms. J Pathol. 2010;221:3–12. doi: 10.1002/path.2697.
    1. Kumar D, Shankar S, Srivastava RK. Rottlerin-induced autophagy leads to the apoptosis in breast cancer stem cells: Molecular mechanisms. Mol Cancer. 2013;12:171. doi: 10.1186/1476-4598-12-171.
    1. Deretic V, Saitoh T, Akira S. Autophagy in infection, inflammation and immunity. Nat Rev Immunol. 2013;13:722–737. doi: 10.1038/nri3532.
    1. Piao Y, Gwon DH, Kang DW, Hwang TW, Shin N, Kwon HH, Shin HJ, Yin Y, Kim JJ, Hong J, et al. TLR4-mediated autophagic impairment contributes to neuropathic pain in chronic constriction injury mice. Mol Brain. 2018;11:11. doi: 10.1186/s13041-018-0354-y.
    1. Weng W, Yao C, Poonit K, Zhou X, Sun C, Zhang F, Yan H. Metformin relieves neuropathic pain after spinal nerve ligation via autophagy flux stimulation. J Cell Mol Med. 2019;23:1313–1324. doi: 10.1111/jcmm.14033.
    1. Yin Y, Yi MH, Kim DW. Impaired autophagy of GABAergic interneurons in neuropathic pain. Pain Res Manag. 2018;2018:9185368. doi: 10.1155/2018/9185368.
    1. Dando I, Donadelli M, Costanzo C, Dalla Pozza E, D'Alessandro A, Zolla L, Palmieri M. Cannabinoids inhibit energetic metabolism and induce AMPK-dependent autophagy in pancreatic cancer cells. Cell Death Dis. 2013;4:e664. doi: 10.1038/cddis.2013.151.
    1. Kimura T, Isaka Y, Yoshimori T. Autophagy and kidney inflammation. Autophagy. 2017;13:997–1003. doi: 10.1080/15548627.2017.1309485.
    1. Chen ZH, Wu YF, Wang PL, Wu YP, Li ZY, Zhao Y, Zhou JS, Zhu C, Cao C, Mao YY, et al. Autophagy is essential for ultrafine particle-induced inflammation and mucus hyperproduction in airway epithelium. Autophagy. 2016;12:297–311. doi: 10.1080/15548627.2015.1124224.

Source: PubMed

3
Subscribe