Naltrexone Inhibits IL-6 and TNFα Production in Human Immune Cell Subsets following Stimulation with Ligands for Intracellular Toll-Like Receptors

Rachel Cant, Angus G Dalgleish, Rachel L Allen, Rachel Cant, Angus G Dalgleish, Rachel L Allen

Abstract

The opioid antagonist naltrexone hydrochloride has been suggested to be a potential therapy at low dosage for multiple inflammatory conditions and cancers. Little is known about the immune-modulating effects of naltrexone, but an effect on the activity of toll-like receptor 4 (TLR4) has been reported. We analyzed the effects of naltrexone hydrochloride on IL-6 secretion by peripheral blood mononuclear cells (PBMC) in vitro following stimulation with ligands for TLR4 and for the intracellular receptors TLR7, TLR8, and TLR9. Naltrexone did not affect cell viability or induce apoptosis of PBMC. Intracellular staining demonstrated that naltrexone inhibited production of IL-6 and TNFα by monocyte and plasmacytoid dendritic cell subsets within the PBMC population following treatment with ligands for TLR7/8 and TLR9, respectively. No effect of cytokine production by PBMC following stimulation of TLR4 was observed. Additionally, naltrexone inhibited IL-6 production in isolated monocytes and B cells after TLR7/8 and TLR9 stimulation, respectively, but no effect on IL-6 production in isolated monocytes after TLR4 stimulation was observed. These findings indicate that naltrexone has the potential to modulate the secretion of inflammatory cytokines in response to intracellular TLR activity, supporting the hypothesis that it may have potential for use as an immunomodulator.

Keywords: B cells; interleukin-6; monocytes; naltrexone; plasmacytoid dendritic cells; toll-like receptor; tumor necrosis factor alpha.

Figures

Figure 1
Figure 1
Naltrexone inhibits IL-6 production after toll-like receptor (TLR)7/8 and TLR9 stimulation but not after TLR4 or IL-1 stimulation. 1 × 106 peripheral blood mononuclear cells were incubated with (A) 1 ng/ml LPS (TLR4-L), (B) 1µM R848 (TLR7/8-L), (C) 1µM CpG (TLR9-L), (D) 100 ng/ml IL-1 (IL-1R) in the presence or absence of 1–200µM naltrexone for 24 h. Cell-free supernatants were collected and analyzed for IL-6 by ELISA. Data show the mean; SD values are shown and were analyzed using an one-way ANOVA and Tukey’s multiple comparison test (n = 5 TLR ligand experiments and n = 3 IL-1). *p < 0.05.
Figure 2
Figure 2
Intracellular cytokine staining for TNFα and IL-6 in monocytes and plasmacytoid dendritic cells. 1 × 106 peripheral blood mononuclear cells (PBMC) were incubated with either LPS 1 ng/ml (A,C), R848 1µM (B), or CpG 1µM (D) and 200 µM naltrexone for 6 h in the presence of brefeldin A for 4 of those hours. After 6 h, PBMC were stained using antibody panel shown in Figure S4 in Supplementary Material and stained for either intracellular IL-6 or TNF-α. Results show the mean fluorescence intensity (MFI) of IL-6 or TNF-α within that subset from 5 donors. Histograms are representative of 5 independent experiments.
Figure 3
Figure 3
Naltrexone inhibits IL-6 production in isolated monocytes and B cells after toll-like receptor (TLR)7/8 and TLR9 stimulation, respectively, but has no effect on IL-6 production in isolated monocytes after TLR4 stimulation. (A) CD14+ monocytes were isolated from peripheral blood mononuclear cells (PBMC) using magnetic bead isolation. 1 × 105 CD14+ cells were incubated with 1 ng/ml LPS (TLR4-L) or 1µM R848 (TLR7/8-L), in the presence or absence of 200µM naltrexone for 24 h. Cell-free supernatants were collected and analyzed for IL-6 by ELISA. (B) CD19+ B cells were isolated from PBMC using magnetic bead isolation. 105 B cells were incubated with 1µM CpG or 3 µg/ml CD40-L and 20 ng/ml IL-4, with or without 200µM naltrexone for 24 h. IL-6 production was measured in cell-free supernatants by ELISA. Data show the mean and SD values (n = 4).
Figure 4
Figure 4
Toll-like receptor ligand (TLR-L) and naltrexone does not affect the viability of peripheral blood mononuclear cells (PBMC). (A) 1 × 106 PBMC were incubated with 1–200µM naltrexone for 24 h before percentage viability was assessed using trypan blue exclusion. (B,C) 1 × 106 PBMC were incubated with 1 ng/ml LPS (TLR4-L), 1µM R848 (TLR7/8-L), 1µM CpG (TLR9-L), and 200µM naltrexone for 24 h. PBMC were incubated with annexin V and 7AAD before being analyzed by flow cytometry. Figure 4B shows the gating strategy, and Figure 4C shows results from 4 donors. AV−7AAD− are viable cells, AV+7AAD− are in early apoptosis, and AV+7AAD+ are in late apoptosis.

References

    1. Krystal JH, Cramer JA, Krol WF, Kirk GF, Rosenheck RA. Naltrexone in the treatment of alcohol dependence. N Engl J Med (2001) 345:1734–9.10.1056/NEJMoa011127
    1. Lee JD, Friedmann PD, Kinlock TW, Nunes EV, Boney TY, Hoskinson RA, Jr, et al. Extended-release naltrexone to prevent opioid relapse in criminal justice offenders. N Engl J Med (2016) 374:1232–42.10.1056/NEJMoa1505409
    1. Weerts EM, Kim YK, Wand GS, Dannals RF, Lee JS, Frost JJ, et al. Differences in δ- and µ-opioid receptor blockade measured by positron emission tomography in naltrexone-treated recently abstinent alcohol-dependent subjects. Neuropsychopharmacology (2008) 33:653–65.10.1038/sj.npp.1301440
    1. Smith JP, Bingaman SI, Ruggiero F, Mauger DT, Mukherjee A, McGovern CO, et al. Therapy with the opioid antagonist naltrexone promotes mucosal healing in active Crohn’s disease: a randomized placebo-controlled trial. Dig Dis Sci (2011) 56:2088–97.10.1007/s10620-011-1653-7
    1. Cree BAC, Kornyeyeva E, Goodin DS. Pilot trial of low-dose naltrexone and quality of life in multiple sclerosis. Ann Neurol (2010) 68:145–50.10.1002/ana.22006
    1. Younger JW, Zautra AJ, Cummins ET. Effects of naltrexone on pain sensitivity and mood in fibromyalgia: no evidence for endogenous opioid pathophysiology. PLoS One (2009) 4:e5180.10.1371/journal.pone.0005180
    1. Younger J, Mackey S. Fibromyalgia symptoms are reduced by low-dose naltrexone: a pilot study. Pain Med (2009) 10:663–72.10.1111/j.1526-4637.2009.00613.x
    1. Younger J, Noor N, McCue R, Mackey S. Low-dose naltrexone for the treatment of fibromyalgia: findings of a small, randomized, double-blind, placebo-controlled, counterbalanced, crossover trial assessing daily pain levels. Arthritis Rheum (2013) 65:529–38.10.1002/art.37734
    1. Berkson BM, Rubin DM, Berkson AJ. Reversal of signs and symptoms of a B-cell lymphoma in a patient using only low-dose naltrexone. Integr Cancer Ther (2007) 6:293–6.10.1177/1534735407306358
    1. Berkson BM, Rubin DM, Berkson AJ. Revisiting the ALA/N (alpha-lipoic acid/low-dose naltrexone) protocol for people with metastatic and nonmetastatic pancreatic cancer: a report of 3 new cases. Integr Cancer Ther (2009) 8:416–22.10.1177/1534735409352082
    1. Berkson BM, Rubin DM, Berkson AJ. The long-term survival of a patient with pancreatic cancer with metastases to the liver after treatment with the intravenous α-lipoic acid/low-dose naltrexone protocol. Integr Cancer Ther (2006) 5:83–9.10.1177/1534735405285901
    1. Donahue RN, McLaughlin PJ, Zagon IS. Cell proliferation of human ovarian cancer is regulated by the opioid growth factor-opioid growth factor receptor axis. Am J Physiol Regul Integr Comp Physiol (2009) 296:R1716–25.10.1152/ajpregu.00075.2009
    1. Donahue RN, McLaughlin PJ, Zagon IS. Low-dose naltrexone suppresses ovarian cancer and exhibits enhanced inhibition in combination with cisplatin. Exp Biol Med (Maywood) (2011) 236:883–95.10.1258/ebm.2011.011096
    1. Cheng F, McLaughlin PJ, Verderame MF, Zagon IS. The OGF-OGFr axis utilizes the p16INK4a and p21WAF1/CIP1 pathways to restrict normal cell proliferation. Mol Biol Cell (2009) 20:319–27.10.1091/mbc.E08-07-0681
    1. Grace PM, Shimizu K, Strand KA, Rice KC, Deng G, Watkins LR, et al. (+)-Naltrexone is neuroprotective and promotes alternative activation in the mouse hippocampus after cardiac arrest/cardiopulmonary resuscitation. Brain Behav Immun (2015) 48:115–22.10.1016/j.bbi.2015.03.005
    1. Hutchinson MR, Zhang Y, Brown K, Coats BD, Shridhar M, Sholar PW, et al. Non-stereoselective reversal of neuropathic pain by naloxone and naltrexone: involvement of toll-like receptor 4 (TLR4). Eur J Neurosci (2008) 28:20–9.10.1111/j.1460-9568.2008.06321.x
    1. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on toll-like receptors. Nat Immunol (2010) 11:373–84.10.1038/ni.1863
    1. Gay NJ, Symmons MF, Gangloff M, Bryant CE. Assembly and localization of toll-like receptor signalling complexes. Nat Rev Immunol (2014) 14:546–58.10.1038/nri3713
    1. Hornung V, Rothenfusser S, Britsch S, Krug A, Jahrsdörfer B, Giese T, et al. Quantitative expression of toll-like receptor 1–10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. J Immunol (2002) 168:4531–7.10.4049/jimmunol.168.9.4531
    1. Visintin A, Mazzoni A, Spitzer JH, Wyllie DH, Dower SK, Segal DM. Regulation of toll-like receptors in human monocytes and dendritic cells. J Immunol (2001) 166:249–55.10.4049/jimmunol.166.1.249
    1. Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol (2011) 29:163–83.10.1146/annurev-immunol-031210-101345
    1. Fischer M, Ehlers M. Toll-like receptors in autoimmunity. Ann N Y Acad Sci (2008) 1143:21–34.10.1196/annals.1443.012
    1. Berkowitz D, Peri R, Lavy A, Kessel A. Increased toll-like receptor 9 expression by B cells from inflammatory bowel disease patients. Hum Immunol (2013) 74(12):1519–23.10.1016/j.humimm.2013.08.285
    1. Lande R, Gregorio J, Facchinetti V, Chatterjee B, Wang YH, Homey B, et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature (2007) 449:564–9.10.1038/nature06116
    1. Sato Y, Goto Y, Narita N, Hoon DSB. Cancer cells expressing toll-like receptors and the tumor microenvironment. Cancer Microenviron (2009) 2(Suppl 1):205–14.10.1007/s12307-009-0022-y
    1. Tolentino YFM, Elia PP, Fogaça HS, Carneiro AJV, Zaltman C, Moura-Neto R, et al. Common NOD2/CARD15 and TLR4 polymorphisms are associated with Crohn’s disease phenotypes in southeastern Brazilians. Dig Dis Sci (2016) 61:2636–47.10.1007/s10620-016-4172-8
    1. Bank S, Andersen PS, Burisch J, Pedersen N, Roug S, Galsgaard J, et al. Polymorphisms in the toll-like receptor and the IL-23/IL-17 pathways were associated with susceptibility to inflammatory bowel disease in a Danish cohort. PLoS One (2015) 10:e0145302.10.1371/journal.pone.0145302
    1. Kanzler H, Barrat FJ, Hessel EM, Coffman RL. Therapeutic targeting of innate immunity with toll-like receptor agonists and antagonists. Nat Med (2007) 13:552–9.10.1038/nm1589
    1. Rahmani F, Rezaei N. Therapeutic targeting of toll-like receptors: a review of toll-like receptors and their signaling pathways in psoriasis. Expert Rev Clin Immunol (2016) 12(12):1289–98.10.1080/1744666X.2016.1204232
    1. O’Neill LAJ. When signaling pathways collide: positive and negative regulation of toll-like receptor signal transduction. Immunity (2008) 29:12–20.10.1016/j.immuni.2008.06.004
    1. Jiang W, Zhu FG, Bhagat L, Yu D, Tang JX, Kandimalla ER, et al. A toll-like receptor 7, 8, and 9 antagonist inhibits Th1 and Th17 responses and inflammasome activation in a model of IL-23-induced psoriasis. J Invest Dermatol (2013) 133:1777–84.10.1038/jid.2013.57
    1. Mohamed FE, Al-Jehani RM, Minogue SS, Andreola F, Winstanley A, Olde Damink SW, et al. Effect of toll-like receptor 7 and 9 targeted therapy to prevent the development of hepatocellular carcinoma. Liver Int (2015) 35:1063–76.10.1111/liv.12626
    1. Junt T, Barchet W. Translating nucleic acid-sensing pathways into therapies. Nat Rev Immunol (2015) 15:529–44.10.1038/nri3875
    1. Wang X, Zhang Y, Peng Y, Hutchinson MR, Rice KC, Yin H, et al. Pharmacological characterization of the opioid inactive isomers (+)-naltrexone and (+)-naloxone as antagonists of toll-like receptor 4. Br J Pharmacol (2016) 173:856–69.10.1111/bph.13394
    1. Northcutt AL, Hutchinson MR, Wang X, Baratta MV, Hiranita T, Cochran TA, et al. DAT isn’t all that: cocaine reward and reinforcement require toll-like receptor 4 signaling. Mol Psychiatry (2015) 20:1525–37.10.1038/mp.2014.177
    1. Hutchinson MR, Northcutt AL, Hiranita T, Wang X, Lewis SS, Thomas J, et al. Opioid activation of toll-like receptor 4 contributes to drug reinforcement. J Neurosci (2012) 32:11187–200.10.1523/JNEUROSCI.0684-12.2012
    1. McLaughlin PJ, McHugh DP, Magister MJ, Zagon IS. Endogenous opioid inhibition of proliferation of T and B cell subpopulations in response to immunization for experimental autoimmune encephalomyelitis. BMC Immunol (2015) 16:24.10.1186/s12865-015-0093-0
    1. Zagon IS, Donahue RN, Bonneau RH, McLaughlin PJ. T lymphocyte proliferation is suppressed by the opioid growth factor ([Met5]-enkephalin)-opioid growth factor receptor axis: implication for the treatment of autoimmune diseases. Immunobiology (2011) 216:579–90.10.1016/j.imbio.2010.06.001
    1. Pinto A, Morello S, Sorrentino R. Lung cancer and toll-like receptors. Cancer Immunol Immunother (2011) 60:1211–20.10.1007/s00262-011-1057-8
    1. Sandholm J, Kauppila JH, Pressey C, Tuomela J, Jukkola-Vuorinen A, Vaarala M, et al. Estrogen receptor-α and sex steroid hormones regulate toll-like receptor-9 expression and invasive function in human breast cancer cells. Breast Cancer Res Treat (2012) 132:411–9.10.1007/s10549-011-1590-3
    1. Qiu J, Shao S, Yang G, Shen Z, Zhang Y. Association of toll like receptor 9 expression with lymph node metastasis in human breast cancer. Neoplasma (2011) 58:251–5.10.4149/neo_2011_03_251
    1. Wu HQ, Wang B, Zhu SK, Tian Y, Zhang JH, Wu HS. Effects of CPG ODN on biological behavior of PANC-1 and expression of TLR9 in pancreatic cancer. World J Gastroenterol (2011) 17:996–1003.10.3748/wjg.v17.i8.996
    1. Gribar SC, Anand RJ, Sodhi CP, Hackam DJ. The role of epithelial toll-like receptor signaling in the pathogenesis of intestinal inflammation. J Leukoc Biol (2008) 83:493–8.10.1189/jlb.0607358
    1. McLaughlin PJ, Stucki JK, Zagon IS. Modulation of the opioid growth factor ([Met(5)]-enkephalin)-opioid growth factor receptor axis: novel therapies for squamous cell carcinoma of the head and neck. Head Neck (2012) 34:513–9.10.1002/hed.21759
    1. Bigliardi PL, Stammer H, Jost G, Rufli T, Büchner S, Bigliardi-Qi M. Treatment of pruritus with topically applied opiate receptor antagonist. J Am Acad Dermatol (2007) 56:979–88.10.1016/j.jaad.2007.01.007
    1. Liu WM, Scott KA, Dennis JL, Kaminska E, Levett AJ, Dalgleish AG. Naltrexone at low doses upregulates a unique gene expression not seen with normal doses: implications for its use in cancer therapy. Int J Oncol (2016) 49:793–802.10.3892/ijo.2016.3567

Source: PubMed

3
Subskrybuj