Adoptive transfer of M2 macrophages reduces neuropathic pain via opioid peptides

Maria Pannell, Dominika Labuz, Melih Ö Celik, Jacqueline Keye, Arvind Batra, Britta Siegmund, Halina Machelska, Maria Pannell, Dominika Labuz, Melih Ö Celik, Jacqueline Keye, Arvind Batra, Britta Siegmund, Halina Machelska

Abstract

Background: During the inflammation which occurs following nerve damage, macrophages are recruited to the site of injury. Phenotypic diversity is a hallmark of the macrophage lineage and includes pro-inflammatory M1 and anti-inflammatory M2 populations. Our aim in this study was to investigate the ability of polarized M0, M1, and M2 macrophages to secrete opioid peptides and to examine their relative contribution to the modulation of neuropathic pain.

Methods: Mouse bone marrow-derived cells were cultured as unstimulated M0 macrophages or were stimulated into an M1 phenotype using lipopolysaccharide and interferon-γ or into an M2 phenotype using interleukin-4. The macrophage phenotypes were verified using flow cytometry for surface marker analysis and cytokine bead array for cytokine profile assessment. Opioid peptide levels were measured by radioimmunoassay and enzyme immunoassay. As a model of neuropathic pain, a chronic constriction injury (CCI) of the sciatic nerve was employed. Polarized M0, M1, and M2 macrophages (5 × 105 cells) were injected perineurally twice, on days 14 and 15 following CCI or sham surgery. Mechanical and heat sensitivity were measured using the von Frey and Hargreaves tests, respectively. To track the injected macrophages, we also transferred fluorescently stained polarized cells and analyzed the surface marker profile of endogenous and injected cells in the nerves ex vivo.

Results: Compared to M0 and M1 cells, M2 macrophages contained and released higher amounts of opioid peptides, including Met-enkephalin, dynorphin A (1-17), and β-endorphin. M2 cells transferred perineurally at the nerve injury site reduced mechanical, but not heat hypersensitivity following the second injection. The analgesic effect was reversed by the perineurally applied opioid receptor antagonist naloxone methiodide. M2 cells did not affect sensitivity following sham surgery. Neither M0 nor M1 cells altered mechanical and heat sensitivity in CCI or sham-operated animals. Tracing the fluorescently labeled M0, M1, and M2 cells ex vivo showed that they remained in the nerve and preserved their phenotype.

Conclusions: Perineural transplantation of M2 macrophages resulted in opioid-mediated amelioration of neuropathy-induced mechanical hypersensitivity, while M1 macrophages did not exacerbate pain. Therefore, rather than focusing on macrophage-induced pain generation, promoting opioid-mediated M2 actions may be more relevant for pain control.

Keywords: Analgesia; Dynorphin; Endorphin; Enkephalin; Macrophage; Neuropathic pain.

Figures

Fig. 1
Fig. 1
Analysis of surface markers on cultured macrophages following polarization. a Representative dot blots showing cell populations expressing F4/80 and CD163. The blots represent the CD163 FMO-negative control (far left), M0-polarized cells (second left), M1 cells (third left), and M2 cells (far right). The percentage of macrophages negative for CD163 (F4/80+CD163−) are shown in top left quadrants, while macrophages expressing CD163 (F4/80+CD163+) are shown in top right quadrants. b Representative dot blots showing cell populations expressing F4/80 and MHC II. The blots show the MHC II FMO-negative control (far left), the M0 cells (second left), the M1 cells (third left), and the M2 cells (far right). The percentage of macrophages negative for MHC II (F4/80+MHC II−) are shown in top left quadrants, while macrophages expressing MHC II (F4/80+MHC II+) are shown in top right quadrants. c Percentage of polarized M0, M1, and M2 macrophages expressing CD163. d Percentage of polarized M0, M1, and M2 macrophages expressing MHC II. The data were analyzed using flow cytometry and FlowJo software. In c and d, **p < 0.01, ***p < 0.001 (one-way RM ANOVA, Bonferroni’s test). Data show the mean ± SEM (n = 8 samples per group)
Fig. 2
Fig. 2
Pro-inflammatory cytokine release profiles of cultured macrophages following polarization. Released levels of MCP-1 (a), TNF-α (b), IL-6 (c), and IFN-γ (d). The insert in d shows the graph with smaller Y-axis scale since IFN-γ levels were very low; only levels for M1 were above detection limit. The cytokine concentrations were measured in media using CBA, ~18 h after polarization was completed. *p < 0.05, **p < 0.01, ***p < 0.001 (one-way RM ANOVA, Bonferroni’s test). Data show the mean ± SEM (n = 8 samples per group)
Fig. 3
Fig. 3
Opioid peptide content and release from cultured macrophages following polarization. Intracellular content (a) and extracellular, released levels (b) of Met-enkephalin, dynorphin, and β-endorphin. Opioid peptide levels were measured by RIA (Met-enkephalin and dynorphin) or EIA (β-endorphin), ~18 h after polarization was completed. *p < 0.05, **p < 0.01, ***p < 0.001 (one-way RM ANOVA, Bonferroni’s test). Data show the mean ± SEM (n = 8 samples per group)
Fig. 4
Fig. 4
Effects of polarized macrophages transferred at the nerves on sensitivity following CCI and sham surgery. a, b Effects of M0, M1, and M2 macrophages on mechanical (a) and heat hypersensitivity (b) following CCI. c, d Effects of M0, M1, and M2 macrophages on mechanical (c) and heat sensitivity (d) following sham surgery. Macrophages or control DMEM were injected at the site of CCI or sham surgery on days 14 and 15 following surgeries (indicated by arrows). Mechanical and heat sensitivity were measured using von Frey filaments and Hargreaves test, respectively. ***p < 0.001 vs. control (DMEM); two-way RM ANOVA, Bonferroni’s test. Data show the mean ± SEM (n = 9 animals per group)
Fig. 5
Fig. 5
Contribution of peripheral opioid receptors to M2 macrophage-mediated analgesia in neuropathy-induced mechanical hypersensitivity. M2 macrophages were injected on days 14 and 15 following CCI, and opioid receptor antagonist NLXM was applied on day 16 after CCI (i.e., 24 h after the second injection of macrophages). All injections (indicated by arrows) were performed at the CCI site, and the effects were assessed using von Frey filaments. ***p < 0.001 vs. vehicle; ###p < 0.001 vs. baseline measured 14 days after CCI (two-way RM ANOVA, Bonferroni’s test). Data show the mean ± SEM (n = 6 animals per group)
Fig. 6
Fig. 6
CFSE labeling of cultured macrophages following polarization. CFSE (0.5 μM) was added to cells immediately after polarization and CFSE staining was measured ~24 h later using flow cytometry. p > 0.05 (one-way RM ANOVA). Data show the mean ± SEM (n = 8 samples per group)
Fig. 7
Fig. 7
Quantification of CD45+ cells in injured nerves following perineural transfer of CFSE-labeled polarized macrophages. a Representative dot blots of CD45 staining: (left panel) CD45 FMO-negative control; (middle panel) CD45+ cells after one injection of DMEM only; (right panel) CD45+ cells following one injection of M2 macrophages. b Total number of CD45+ cells per nerve after one injection of DMEM only or CFSE-labeled M0, M1, and M2 cells. c Total number of CD45+ cells per nerve after two injections of DMEM only or CFSE-labeled M0, M1, and M2 cells. In all experiments, DMEM- or CFSE-labeled macrophages were injected at the injury site on day 14 (one injection) or on days 14 and 15 (two injections) after CCI. Animals were killed 24 h after the first injection and 24 h after the second injection, and cells from the nerves were analyzed using flow cytometry and FlowJo software. *p < 0.05, **p < 0.01, ***p < 0.001 (one-way ANOVA, Bonferroni’s test); ##p < 0.01; ###p < 0.001 compared to same treatment 24 h after the first injection (unpaired t test). Data show the mean ± SEM (n = 6 animals per group)
Fig. 8
Fig. 8
Quantification of endogenous and injected macrophages in injured nerves following perineural transfer of CFSE-labeled polarized macrophages. a Representative dot blot of F4/80 and CFSE staining after one injection of DMEM only, showing endogenous macrophages (F4/80+CFSE−) in the top left quadrant, and the lack of injected macrophages (F4/80+CFSE+) in the top right quadrant. b Representative dot blot of F4/80 and CFSE staining after one injection of CFSE-labeled M2 cells, showing endogenous macrophages (F4/80+CFSE−) in the top left quadrant and injected macrophages (F4/80+CFSE+) in the top right quadrant. c Total number of endogenous macrophages (F4/80+CFSE−) after one injection of DMEM alone or CFSE-labeled M0, M1, and M2 cells. d Total number of endogenous macrophages (F4/80+CFSE−) after two injections of DMEM alone or CFSE-labeled M0, M1, and M2 cells. e Total number of injected macrophages (F4/80+CFSE+) after one injection of DMEM alone or CFSE-labeled M0, M1, and M2 cells. f Total number of injected macrophages (F4/80+CFSE+) after two injections of DMEM alone or CFSE-labeled M0, M1, and M2 cells. In all experiments, DMEM- or CFSE-labeled macrophages were injected at the injury site on day 14 (one injection) or on days 14 and 15 (two injections) after CCI. Animals were killed 24 h after the first injection and 24 h after the second injection, and cells from the nerves were analyzed using flow cytometry and FlowJo software. *p < 0.05, **p < 0.01, ***p < 0.001 (one-way ANOVA, Bonferroni’s test); ##p < 0.01, ###p < 0.001 compared to the same treatment 24 h after the first injection (unpaired t test). Data show the mean ± SEM (n = 6 animals per group)
Fig. 9
Fig. 9
Analysis of CD163 and MHC II expression in endogenous macrophages in injured nerves following perineural transfer of CFSE-labeled polarized macrophages. Endogenous macrophages were identified as F4/80+CFSE− cells. a Percentage of endogenous macrophages expressing CD163 following one injection of DMEM only or CFSE-labeled M0, M1, and M2 cells. b Percentage of endogenous macrophages expressing CD163 following two injections of DMEM only or CFSE-labeled M0, M1, and M2 cells. c Percentage of endogenous macrophages expressing MHC II following one injection of DMEM only or CFSE-labeled M0, M1, and M2 cells. d Percentage of endogenous macrophages expressing MHC II following two injections of DMEM only or CFSE-labeled M0, M1, and M2 cells. In all experiments, DMEM- or CFSE-labeled macrophages were injected at the injury site on day 14 (one injection) or on days 14 and 15 (two injections) after CCI. Animals were killed 24 h after the first injection and 24 h after the second injection, and cells from the nerves were analyzed using flow cytometry and FlowJo software. *p < 0.05, **p < 0.01, ***p < 0.001 (one-way ANOVA, Bonferroni’s test); #p < 0.05, ##p < 0.01 compared to the same treatment 24 h after the first injection (unpaired t test). Data show the mean ± SEM (n = 6 animals per group)
Fig. 10
Fig. 10
Analysis of CD163 and MHC II expression in perineurally injected CFSE-labeled polarized macrophages in injured nerves. Injected macrophages were identified as F4/80+CFSE+ cells. a Percentage of injected macrophages expressing CD163 following one injection of DMEM only or CFSE-labeled M0, M1, and M2 cells. b Percentage of injected macrophages expressing CD163 following one injections of DMEM only or CFSE-labeled M0, M1, and M2 cells. c Percentage of injected macrophages expressing MHC II following one injection of DMEM only or CFSE-labeled M0, M1, and M2 cells. d Percentage of injected macrophages expressing MHC II following two injections of DMEM only or CFSE-labeled M0, M1, and M2 cells. In all experiments, DMEM medium or CFSE-labeled macrophages were injected at the injury site on day 14 (one injection) or on days 14 and 15 (two injections) after CCI. Animals were killed 24 h after the first injection and 24 h after the second injection, and cells from the nerves were analyzed using flow cytometry and FlowJo software. **p < 0.01, ***p < 0.001 (one-way ANOVA, Bonferroni’s test); #p < 0.05 compared to the same treatment 24 h after the first injection (unpaired t test). Data show the mean ± SEM (n = 6 animals per group)

References

    1. Torrance N, Smith BH, Bennett MI, Lee AJ. The epidemiology of chronic pain of predominantly neuropathic origin. Results from a general population survey. J Pain. 2006;7:281–289. doi: 10.1016/j.jpain.2005.11.008.
    1. Jensen TS, Finnerup NB. Allodynia and hyperalgesia in neuropathic pain: clinical manifestations and mechanisms. Lancet Neurol. 2014;13:924–935. doi: 10.1016/S1474-4422(14)70102-4.
    1. Woolf CJ, Mannion RJ. Neuropathic pain: aetiology, symptoms, mechanisms, and management. Lancet. 1999;353:1959–1964. doi: 10.1016/S0140-6736(99)01307-0.
    1. Dworkin RH, O’Connor AB, Backonja M, Farrar JT, Finnerup NB, Jensen TS, et al. Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain. 2007;132:237–251. doi: 10.1016/j.pain.2007.08.033.
    1. Eisenberg E, McNicol E, Carr DB. Opioids for neuropathic pain. Cochrane Database Syst Rev. 2006;3:CD006146.
    1. Benyamin R, Trescot AM, Datta S, Buenaventura R, Adlaka R, Sehgal N, et al. Opioid complications and side effects. Pain Physician. 2008;11:S105–S120.
    1. Volkow ND, Frieden TR, Hyde PS, Cha SS. Medication-assisted therapies—tackling the opioid-overdose epidemic. N Engl J Med. 2014;370:2063–2066. doi: 10.1056/NEJMp1402780.
    1. Basso L, Bourreille A, Dietrich G. Intestinal inflammation and pain management. Curr Opin Pharmacol. 2015;25:50–55. doi: 10.1016/j.coph.2015.11.004.
    1. Gavériaux-Ruff C. Opiate-induced analgesia: contributions from mu, delta and kappa opioid receptors mouse mutants. Curr Pharm Des. 2013;19:7373–7381. doi: 10.2174/138161281942140105163727.
    1. Stein C, Schäfer M, Machelska H. Attacking pain at its source: new perspectives on opioids. Nat Med. 2003;9:1003–1008. doi: 10.1038/nm908.
    1. Stein C, Machelska H. Modulation of peripheral sensory neurons by the immune system: implications for pain therapy. Pharmacol Rev. 2011;63:860–881. doi: 10.1124/pr.110.003145.
    1. Austin PJ, Moalem-Taylor G. The neuro-immune balance in neuropathic pain: involvement of inflammatory immune cells, immune-like glial cells and cytokines. J Neuroimmunol. 2010;229:26–50. doi: 10.1016/j.jneuroim.2010.08.013.
    1. Calvo M, Dawes JM, Bennett DLH. The role of the immune system in the generation of neuropathic pain. Lancet Neurol. 2012;11:629–642. doi: 10.1016/S1474-4422(12)70134-5.
    1. Machelska H. Dual peripheral actions of immune cells in neuropathic pain. Arch Immunol Ther Exp (Warsz) 2011;59:11–24. doi: 10.1007/s00005-010-0106-x.
    1. Barclay J, Clark AK, Ganju P, Gentry C, Patel S, Wotherspoon G, et al. Role of the cysteine protease cathepsin S in neuropathic hyperalgesia. Pain. 2007;130:225–234. doi: 10.1016/j.pain.2006.11.017.
    1. Liu T, van Rooijen N, Tracey DJ. Depletion of macrophages reduces axonal degeneration and hyperalgesia following nerve injury. Pain. 2000;86:25–32. doi: 10.1016/S0304-3959(99)00306-1.
    1. Rutkowski MD, Pahl JL, Sweitzer S, van Rooijen N, DeLeo JA. Limited role of macrophages in generation of nerve injury-induced mechanical allodynia. Physiol Behav. 2000;71:225–235. doi: 10.1016/S0031-9384(00)00333-4.
    1. Trevisan G, Benemei S, Materazzi S, Logu FD, Siena GD, Fusi C, et al. TRPA1 mediates trigeminal neuropathic pain in mice downstream of monocytes/macrophages and oxidative stress. Brain. 2016;139:1361–1377. doi: 10.1093/brain/aww038.
    1. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448.
    1. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122:787–795. doi: 10.1172/JCI59643.
    1. Bouhlel MA, Derudas B, Rigamonti E, Dièvart R, Brozek J, Haulon S, et al. PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007;6:137–143. doi: 10.1016/j.cmet.2007.06.010.
    1. Hasegawa-Moriyama M, Ohnou T, Godai K, Kurimoto T, Nakama M, Kanmura Y. Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuates postincisional pain by regulating macrophage polarization. Biochem Biophys Res Commun. 2012;426:76–82. doi: 10.1016/j.bbrc.2012.08.039.
    1. Hasegawa-Moriyama M, Kurimoto T, Nakama M, Godai K, Kojima M, Kuwaki T, et al. Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuates inflammatory pain through the induction of heme oxygenase-1 in macrophages. Pain. 2013;154:1402–1412. doi: 10.1016/j.pain.2013.04.039.
    1. Napimoga MH, Souza GR, Cunha TM, Ferrari LF, Clemente-Napimoga JT, Parada CA, et al. 15d-prostaglandin J2 inhibits inflammatory hypernociception: involvement of peripheral opioid receptor. J Pharmacol Exp Ther. 2008;324:313–321. doi: 10.1124/jpet.107.126045.
    1. Sauer R-S, Hackel D, Morschel L, Sahlbach H, Wang Y, Mousa SA, et al. Toll like receptor (TLR)-4 as a regulator of peripheral endogenous opioid-mediated analgesia in inflammation. Mol Pain. 2014;10:10. doi: 10.1186/1744-8069-10-10.
    1. Komori T, Morikawa Y, Inada T, Hisaoka T, Senba E. Site-specific subtypes of macrophages recruited after peripheral nerve injury. Neuroreport. 2011;22:911–917. doi: 10.1097/WNR.0b013e32834cd76a.
    1. Takahashi Y, Hasegawa-Moriyama M, Sakurai T, Inada E. The macrophage-mediated effects of the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuate tactile allodynia in the early phase of neuropathic pain development. Anesth Analg. 2011;113:398–404. doi: 10.1213/ANE.0b013e31821b220c.
    1. Kiguchi N, Kobayashi Y, Saika F, Sakaguchi H, Maeda T, Kishioka S. Peripheral interleukin-4 ameliorates inflammatory macrophage-dependent neuropathic pain. Pain. 2015;156:684–693. doi: 10.1097/j.pain.0000000000000097.
    1. Celik MÖ, Labuz D, Henning K, Busch-Dienstfertig M, Gaveriaux-Ruff C, Kieffer BL, et al. Leukocyte opioid receptors mediate analgesia via Ca(2+)-regulated release of opioid peptides. Brain Behav Immun. 2016;57:227–242. doi: 10.1016/j.bbi.2016.04.018.
    1. Labuz D, Schmidt Y, Schreiter A, Rittner HL, Mousa SA, Machelska H. Immune cell-derived opioids protect against neuropathic pain in mice. J Clin Invest. 2009;119:278–286. doi: 10.1172/JCI36246C1.
    1. Liou J-T, Liu F-C, Mao C-C, Lai Y-S, Day Y-J. Inflammation confers dual effects on nociceptive processing in chronic neuropathic pain model. Anesthesiology. 2011;114:660–672. doi: 10.1097/ALN.0b013e31820b8b1e.
    1. Schmidt Y, Gavériaux-Ruff C, Machelska H. μ-Opioid receptor antibody reveals tissue-dependent specific staining and increased neuronal μ-receptor immunoreactivity at the injured nerve trunk in mice. PloS One. 2013;8:e79099. doi: 10.1371/journal.pone.0079099.
    1. Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8:e1000412. doi: 10.1371/journal.pbio.1000412.
    1. Zhang X, Goncalves R, Mosser DM. The isolation and characterization of murine macrophages. Curr Protoc Immunol. 2008;Chapter 14:Unit 14.1.
    1. Gracey E, Lin A, Akram A, Chiu B, Inman RD. Intracellular survival and persistence of Chlamydia muridarum is determined by macrophage polarization. PLoS One. 2013;8:e69421. doi: 10.1371/journal.pone.0069421.
    1. Jablonski KA, Amici SA, Webb LM, Ruiz-Rosado JD, Popovich PG, Partida-Sanchez S, et al. Novel markers to delineate murine M1 and M2 macrophages. PloS One. 2015;10:e0145342. doi: 10.1371/journal.pone.0145342.
    1. Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009;29:13435–13444. doi: 10.1523/JNEUROSCI.3257-09.2009.
    1. Roederer M. Compensation in flow cytometry. Curr Protoc Cytom. 2002;Chapter 1:Unit 1.14.
    1. Pannell M, Meier MA, Szulzewsky F, Matyash V, Endres M, Kronenberg G, et al. The subpopulation of microglia expressing functional muscarinic acetylcholine receptors expands in stroke and Alzheimer’s disease. Brain Struct Funct. 2016;221:1157–1172. doi: 10.1007/s00429-014-0962-y.
    1. Rustenhoven J, Aalderink M, Scotter EL, Oldfield RL, Bergin PS, Mee EW, et al. TGF-beta1 regulates human brain pericyte inflammatory processes involved in neurovasculature function. J Neuroinflammation. 2016;13:37. doi: 10.1186/s12974-016-0503-0.
    1. Schreiter A, Gore C, Labuz D, Fournie-Zaluski M-C, Roques BP, Stein C, et al. Pain inhibition by blocking leukocytic and neuronal opioid peptidases in peripheral inflamed tissue. FASEB J. 2012;26:5161–5171. doi: 10.1096/fj.12-208678.
    1. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994;53:55–63. doi: 10.1016/0165-0270(94)90144-9.
    1. Labuz D, Schreiter A, Schmidt Y, Brack A, Machelska H. T lymphocytes containing β-endorphin ameliorate mechanical hypersensitivity following nerve injury. Brain Behav Immun. 2010;24:1045–1053. doi: 10.1016/j.bbi.2010.04.001.
    1. Labuz D, Machelska H. Stronger antinociceptive efficacy of opioids at the injured nerve trunk than at its peripheral terminals in neuropathic pain. J Pharmacol Exp Ther. 2013;346:535–544. doi: 10.1124/jpet.113.205344.
    1. Labuz D, Spahn V, Celik MÖ, Machelska H. Opioids and TRPV1 in the peripheral control of neuropathic pain—defining a target site in the injured nerve. Neuropharmacology. 2016;101:330–340. doi: 10.1016/j.neuropharm.2015.10.003.
    1. Shi H, Yang W, Cui Z-H, Lu C-W, Li X-H, Liang L-L, et al. Tracking of CFSE-labeled endothelial progenitor cells in laser-injured mouse retina. Chin Med J (Engl) 2011;124:751–757.
    1. Xaus J, Comalada M, Barrachina M, Herrero C, Goñalons E, Soler C, et al. The expression of MHC class II genes in macrophages is cell cycle dependent. J Immunol. 2000;165:6364–6371. doi: 10.4049/jimmunol.165.11.6364.
    1. Yang H, Qiu Q, Gao B, Kong S, Lin Z, Fang D. Hrd1-mediated BLIMP-1 ubiquitination promotes dendritic cell MHCII expression for CD4 T cell priming during inflammation. J Exp Med. 2014;211:2467–2479. doi: 10.1084/jem.20140283.
    1. Davis MJ, Tsang TM, Qiu Y, Dayrit JK, Freij JB, Huffnagle GB, et al. Macrophage M1/M2 polarization dynamically adapts to changes in cytokine microenvironments in Cryptococcus neoformans infection. mBio. 2013;4:e00264–13. doi: 10.1128/mBio.00264-13.
    1. Schaer DJ, Schaer CA, Buehler PW, Boykins RA, Schoedon G, Alayash AI, et al. CD163 is the macrophage scavenger receptor for native and chemically modified hemoglobins in the absence of haptoglobin. Blood. 2006;107:373–380. doi: 10.1182/blood-2005-03-1014.
    1. Lau SK, Chu PG, Weiss LM. CD163: a specific marker of macrophages in paraffin-embedded tissue samples. Am J Clin Pathol. 2004;122:794–801. doi: 10.1309/QHD6YFN81KQXUUH6.
    1. Edin S, Wikberg ML, Dahlin AM, Rutegård J, Öberg Å, Oldenborg P-A, et al. The distribution of macrophages with a M1 or M2 phenotype in relation to prognosis and the molecular characteristics of colorectal cancer. PLoS One. 2012;7:e47045. doi: 10.1371/journal.pone.0047045.
    1. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000prime Rep. 2014;6:13. doi: 10.12703/P6-13.
    1. Chen W, Liu J, Meng J, Lu C, Li X, Wang E, et al. Macrophage polarization induced by neuropeptide methionine enkephalin (MENK) promotes tumoricidal responses. Cancer Immunol Immunother. 2012;61:1755–1768. doi: 10.1007/s00262-012-1240-6.
    1. Takeuchi H, Tanaka M, Tanaka A, Tsunemi A, Yamamoto H. Predominance of M2-polarized macrophages in bladder cancer affects angiogenesis, tumor grade and invasiveness. Oncol Lett. 2016;11:3403–3408.
    1. Rőszer T. Understanding the mysterious M2 macrophage through activation markers and effector mechanisms. Mediators Inflamm. 2015;2015:816460.
    1. Corna G, Campana L, Pignatti E, Castiglioni A, Tagliafico E, Bosurgi L, et al. Polarization dictates iron handling by inflammatory and alternatively activated macrophages. Haematologica. 2010;95:1814–1822. doi: 10.3324/haematol.2010.023879.
    1. Bacci M, Capobianco A, Monno A, Cottone L, Di Puppo F, Camisa B, et al. Macrophages are alternatively activated in patients with endometriosis and required for growth and vascularization of lesions in a mouse model of disease. Am J Pathol. 2009;175:547–556. doi: 10.2353/ajpath.2009.081011.
    1. Garg K, Pullen NA, Oskeritzian CA, Ryan JJ, Bowlin GL. Macrophage functional polarization (M1/M2) in response to varying fiber and pore dimensions of electrospun scaffolds. Biomaterials. 2013;34:4439–4451. doi: 10.1016/j.biomaterials.2013.02.065.
    1. Mia S, Warnecke A, Zhang X-M, Malmström V, Harris RA. An optimized protocol for human M2 macrophages using M-CSF and IL-4/IL-10/TGF-β yields a dominant immunosuppressive phenotype. Scand J Immunol. 2014;79:305–314. doi: 10.1111/sji.12162.
    1. Kuroda E, Ho V, Ruschmann J, Antignano F, Hamilton M, Rauh MJ, et al. SHIP represses the generation of IL-3-induced M2 macrophages by inhibiting IL-4 production from basophils. J Immunol. 2009;183:3652–3660. doi: 10.4049/jimmunol.0900864.
    1. Murray PJ, Wynn TA. Obstacles and opportunities for understanding macrophage polarization. J Leukoc Biol. 2011;89:557–563. doi: 10.1189/jlb.0710409.
    1. Shechter R, London A, Varol C, Raposo C, Cusimano M, Yovel G, et al. Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice. PLoS Med. 2009;6:e1000113. doi: 10.1371/journal.pmed.1000113.
    1. Brack A, Labuz D, Schiltz A, Rittner HL, Machelska H, Schäfer M, et al. Tissue monocytes/macrophages in inflammation: hyperalgesia versus opioid-mediated peripheral antinociception. Anesthesiology. 2004;101:204–211. doi: 10.1097/00000542-200407000-00031.
    1. Godai K, Hasegawa-Moriyama M, Kurimoto T, Saito T, Yamada T, Sato T, et al. Peripheral administration of morphine attenuates postincisional pain by regulating macrophage polarization through COX-2-dependent pathway. Mol Pain. 2014;10:36. doi: 10.1186/1744-8069-10-36.
    1. Abbadie C, Lindia JA, Cumiskey AM, Peterson LB, Mudgett JS, Bayne EK, et al. Impaired neuropathic pain responses in mice lacking the chemokine receptor CCR2. Proc Natl Acad Sci U S A. 2003;100:7947–7952. doi: 10.1073/pnas.1331358100.
    1. Ghasemlou N, Chiu IM, Julien J-P, Woolf CJ. CD11b + Ly6G- myeloid cells mediate mechanical inflammatory pain hypersensitivity. Proc Natl Acad Sci U S A. 2015;112:E6808–E6817. doi: 10.1073/pnas.1501372112.
    1. Brack A, Rittner HL, Machelska H, Leder K, Mousa SA, Schäfer M, et al. Control of inflammatory pain by chemokine-mediated recruitment of opioid-containing polymorphonuclear cells. Pain. 2004;112:229–238. doi: 10.1016/j.pain.2004.08.029.
    1. Rittner HL, Hackel D, Yamdeu R-S, Mousa SA, Stein C, Schäfer M, et al. Antinociception by neutrophil-derived opioid peptides in noninflamed tissue—role of hypertonicity and the perineurium. Brain Behav Immun. 2009;23:548–557. doi: 10.1016/j.bbi.2009.02.007.
    1. Dubový P. Wallerian degeneration and peripheral nerve conditions for both axonal regeneration and neuropathic pain induction. Ann Anat. 2011;193:267–275. doi: 10.1016/j.aanat.2011.02.011.
    1. Willemen HLDM, Eijkelkamp N, Garza Carbajal A, Wang H, Mack M, Zijlstra J, et al. Monocytes/macrophages control resolution of transient inflammatory pain. J Pain. 2014;15:496–506. doi: 10.1016/j.jpain.2014.01.491.
    1. Ma S-F, Chen Y-J, Zhang J-X, Shen L, Wang R, Zhou J-S, et al. Adoptive transfer of M2 macrophages promotes locomotor recovery in adult rats after spinal cord injury. Brain Behav Immun. 2015;45:157–170. doi: 10.1016/j.bbi.2014.11.007.
    1. Rapalino O, Lazarov-Spiegler O, Agranov E, Velan GJ, Yoles E, Fraidakis M, et al. Implantation of stimulated homologous macrophages results in partial recovery of paraplegic rats. Nat Med. 1998;4:814–821. doi: 10.1038/nm0798-814.

Source: PubMed

3
Abonnere