Melatonin receptor agonist protects against acute lung injury induced by ventilator through up-regulation of IL-10 production

Geng-Chin Wu, Chung-Kan Peng, Wen-I Liao, Hsin-Ping Pao, Kun-Lun Huang, Shi-Jye Chu, Geng-Chin Wu, Chung-Kan Peng, Wen-I Liao, Hsin-Ping Pao, Kun-Lun Huang, Shi-Jye Chu

Abstract

Background: It is well known that ventilation with high volume or pressure may damage healthy lungs or worsen injured lungs. Melatonin has been reported to be effective in animal models of acute lung injury. Melatonin exerts its beneficial effects by acting as a direct antioxidant and via melatonin receptor activation. However, it is not clear whether melatonin receptor agonist has a protective effect in ventilator-induced lung injury (VILI). Therefore, in this study, we determined whether ramelteon (a melatonin receptor agonist) can attenuate VILI and explore the possible mechanism for protection.

Methods: VILI was induced by high tidal volume ventilation in a rat model. The rats were randomly allotted into the following groups: control, control+melatonin, control+ramelteon, control+luzindole, VILI, VILI+luzindole, VILI + melatonin, VILI + melatonin + luzindole (melatonin receptor antagonist), VILI + ramelteon, and VILI + ramelteon + luzindole (n = 6 per group). The role of interleukin-10 (IL-10) in the melatonin- or ramelteon-mediated protection against VILI was also investigated.

Results: Ramelteon treatment markedly reduced lung edema, serum malondialdehyde levels, the concentration of inflammatory cytokines in bronchoalveolar lavage fluid (BALF), NF-κB activation, iNOS levels, and apoptosis in the lung tissue. Additionally, ramelteon treatment significantly increased heat shock protein 70 expression in the lung tissue and IL-10 levels in BALF. The protective effect of ramelteon was mitigated by the administration of luzindole or an anti-IL-10 antibody.

Conclusions: Our results suggest that a melatonin receptor agonist has a protective effect against VILI, and its protective mechanism is based on the upregulation of IL-10 production.

Keywords: Interleukin-10; Melatonin receptor agonist; Ramelteon; Ventilator-induced lung injury.

Conflict of interest statement

All authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Effect of melatonin and ramelteon on lung edema. VILI significantly increased the lung weight/body weight ratio (a), W/D weight ratio (b), and protein levels in BALF (c). Melatonin or ramelteon treatment significantly reduced these increases. The protective effect of melatonin and ramelteon was abrogated by luzindole treatment. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p-values < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 2
Fig. 2
Effect of melatonin and ramelteon on serum malondialdehyde (MDA) levels, carbonyl content and myeloperoxidase (MPO)-positive cell numbers in lung tissue. Serum MDA levels (a), carbonyl content (b) and MPO-positive cell numbers in lung tissue (c) were significantly increased in the VILI group. Melatonin or ramelteon treatment significantly attenuated these increases. The protective effect of melatonin and ramelteon was abrogated by luzindole treatment. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p-values < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 3
Fig. 3
Effect of melatonin and ramelteon on TNF-α, IL-1β, IL-6, and CXCL-1 concentrations in BALF. TNF-α (a), IL-1β (b), IL-6 (C), and CXCL-1 (d) levels in BALF increased significantly in the VILI group. The increases in the BALF protein concentrations were significantly attenuated by treatment with melatonin or ramelteon. The protective effect of melatonin and ramelteon was abrogated by luzindole treatment. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 4
Fig. 4
Effect of melatonin and ramelteon on IL-10 levels in BALF and STAT3 expression in the lung tissue. The p-STAT 3 level in lung tissue was determined by western blotting. β-Actin served as a loading control for cytoplasmic proteins. VILI significantly decreased IL-10 levels in BALF (a) and increased p-STAT3 protein expression in the lung tissue (b). Melatonin or ramelteon treatment significantly reversed this phenomenon. When luzindole was added, the protective effect of melatonin and ramelteon was blocked. Data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 5
Fig. 5
Effect of melatonin and ramelteon on iNOS expression. iNOS levels in the lung tissue were determined by western blotting. β-Actin served as a loading control for cytoplasmic proteins. VILI significantly increased iNOS protein expression. Melatonin or ramelteon treatment significantly attenuated the increase. When luzindole was added, the protective effect of melatonin and ramelteon was blocked. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p-values < 0.05); # significantly different from the VILI group (p < 0.05)
Fig. 6
Fig. 6
Effect of melatonin and ramelteon on lung pathology. a Hematoxylin and eosin staining analysis of lung pathological injury. Representative photomicrographs were taken at a magnification of × 400. b The numbers of neutrophils per high-power field (× 400 magnification), and c lung injury score. Histological evaluation of lung tissues showed that neutrophil infiltration and the lung injury score were increased in the VILI group. Melatonin or ramelteon treatment significantly attenuated these histopathological changes, but the protective effect of melatonin and ramelteon was abrogated by luzindole treatment. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 7
Fig. 7
Effect of melatonin and ramelteon on the NF-κB signaling pathway. a The NF-κB p65 level and b IκBα level in the lung tissue were determined by western blotting. PCNA and β-actin served as loading controls for nuclear and cytoplasmic proteins, respectively. Representative blots are shown. Melatonin or ramelteon treatment reduced NF-κB p65 levels and increased IκB-α levels in VILI. When luzindole was added, the protective effect was blocked. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 8
Fig. 8
Effect of melatonin and ramelteon on apoptosis. Hsp70 (a), Bcl-2 (b), caspase-3 (c), and cleaved PARP (d) levels in the lung tissue were determined by western blotting. Melatonin or ramelteon treatment increased Hsp70 and Bcl-2 levels and reduced caspase-3 and cleaved PARP levels in VILI. When luzindole was added, the protective effect was blocked. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 9
Fig. 9
Effect of an anti-IL-10 antibody on melatonin- and ramelteon-mediated BALF IL-10 levels. VILI significantly decreased IL-10 levels in BALF. Melatonin or ramelteon treatment significantly reversed this phenomenon. When an anti-IL-10 antibody was administered, the protective effect of melatonin and ramelteon was blocked. Data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p #significantly different from the VILI group (p < 0.05)
Fig. 10
Fig. 10
Effect of anti-IL-10 antibody on melatonin- and ramelteon-mediated VILI protection against lung edema. a Lung weight/body weight ratio. b W/D weight ratio. c Protein levels in BALF. Melatonin or ramelteon treatment significantly attenuated the increase in LW/BW and W/D weight ratios and protein levels in VILI. When an anti-IL-10 antibody was administered, the protective effect was blocked. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 11
Fig. 11
Effect of an anti-IL-10 antibody on the melatonin- and ramelteon-mediated cytokines concentrations in BALF. TNF-α (a), IL-1β (b), and IL-6 (c) levels in BALF increased significantly in the VILI group. The increases in the BALF protein concentrations were significantly attenuated by treatment with melatonin or ramelteon. The protective effect of melatonin and ramelteon was abrogated by anti-IL-10 antibody treatment. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 12
Fig. 12
Effect of an anti-IL-10 antibody on the melatonin- and ramelteon-mediated lung pathology. a Hematoxylin and eosin staining analysis of lung pathological injury. b The numbers of neutrophils per high-power field, and c lung injury score. Representative photomicrographs were taken at a magnification of × 400. Histological evaluation of lung tissues showed that neutrophil infiltration and the lung injury score were increased in the VILI group. Melatonin or ramelteon treatment significantly attenuated these histopathological changes, but the protective effect of melatonin and ramelteon was abrogated by anti-IL-10 antibody treatment. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)
Fig. 13
Fig. 13
Effect of an anti-IL-10 antibody on the melatonin- and ramelteon-mediated apoptosis and NF-κB signaling pathway. a Bcl2, b cleaved PARP,and c NF-κB p65 levels in the lung tissue were determined by western blotting. Lamin B and β-actin served as loading controls for nuclear and cytoplasmic proteins, respectively. Melatonin or ramelteon treatment increased Bcl2, and reduced cleaved PARP and NF-κB p65 levels in VILI. When anti-IL-10 antibody was added, the protective effect was blocked. The data are expressed as the mean ± SD (n = 6 per group). *Significantly different from the control (p < 0.05); #significantly different from the VILI group (p < 0.05)

References

    1. Matthay MA, McAuley DF, Ware LB. Clinical trials in acute respiratory distress syndrome: challenges and opportunities. Lancet Respir Med. 2017;5:524–534. doi: 10.1016/S2213-2600(17)30188-1.
    1. Hoegl S, Boost KA, Czerwonka H, Dolfen A, Scheiermann P, Muhl H, Zwissler B, Hofstetter C. Inhaled IL-10 reduces biotrauma and mortality in a model of ventilator-induced lung injury. Respir Med. 2009;103:463–470. doi: 10.1016/j.rmed.2008.09.020.
    1. Jockers R, Delagrange P, Dubocovich ML, Markus RP, Renault N, Tosini G, Cecon E, Zlotos DP. Update on melatonin receptors: IUPHAR review 20. Br J Pharmacol. 2016;173:2702–2725. doi: 10.1111/bph.13536.
    1. Radogna F, Paternoster L, Albertini MC, Cerella C, Accorsi A, Bucchini A, Spadoni G, Diamantini G, Tarzia G, De Nicola M, et al. Melatonin antagonizes apoptosis via receptor interaction in U937 monocytic cells. J Pineal Res. 2007;43:154–162. doi: 10.1111/j.1600-079X.2007.00455.x.
    1. Lochner A, Genade S, Davids A, Ytrehus K, Moolman JA. Short- and long-term effects of melatonin on myocardial post-ischemic recovery. J Pineal Res. 2006;40:56–63. doi: 10.1111/j.1600-079X.2005.00280.x.
    1. Rezzani R, Rodella LF, Bonomini F, Tengattini S, Bianchi R, Reiter RJ. Beneficial effects of melatonin in protecting against cyclosporine A-induced cardiotoxicity are receptor mediated. J Pineal Res. 2006;41:288–295. doi: 10.1111/j.1600-079X.2006.00368.x.
    1. Fink T, Glas M, Wolf A, Kleber A, Reus E, Wolff M, Kiefer D, Wolf B, Rensing H, Volk T, Mathes AM. Melatonin receptors mediate improvements of survival in a model of polymicrobial sepsis. Crit Care Med. 2014;42:e22–e31. doi: 10.1097/CCM.0b013e3182a63e2b.
    1. Hokenson MA, Wang Y, Hawwa RL, Huang Z, Sharma S, Sanchez-Esteban J. Reduced IL-10 production in fetal type II epithelial cells exposed to mechanical stretch is mediated via activation of IL-6-SOCS3 signaling pathway. PLoS One. 2013;8:e59598. doi: 10.1371/journal.pone.0059598.
    1. Ren Wenkai, Liu Gang, Chen Shuai, Yin Jie, Wang Jing, Tan Bie, Wu Guoyao, Bazer Fuller W., Peng Yuanyi, Li Tiejun, Reiter Russel J., Yin Yulong. Melatonin signaling in T cells: Functions and applications. Journal of Pineal Research. 2017;62(3):e12394. doi: 10.1111/jpi.12394.
    1. Mathes AM, Kubulus D, Waibel L, Weiler J, Heymann P, Wolf B, Rensing H. Selective activation of melatonin receptors with ramelteon improves liver function and hepatic perfusion after hemorrhagic shock in rat. Crit Care Med. 2008;36:2863–2870. doi: 10.1097/CCM.0b013e318187b863.
    1. Shimizu N, Nozawa M, Sugimoto K, Yamamoto Y, Minami T, Hayashi T, Yoshimura K, Ishii T, Uemura H. Therapeutic efficacy and anti-inflammatory effect of ramelteon in patients with insomnia associated with lower urinary tract symptoms. Res Rep Urol. 2013;5:113–119.
    1. Zhou W, Zhang X, Zhu CL, He ZY, Liang JP, Song ZC. Melatonin receptor agonists as the “Perioceutics” agents for periodontal disease through modulation of Porphyromonas gingivalis virulence and inflammatory response. PLoS One. 2016;11:e0166442. doi: 10.1371/journal.pone.0166442.
    1. Syrkina O, Jafari B, Hales CA, Quinn DA. Oxidant stress mediates inflammation and apoptosis in ventilator-induced lung injury. Respirology. 2008;13:333–340. doi: 10.1111/j.1440-1843.2008.01279.x.
    1. Wang ML, Wei CH, Wang WD, Wang JS, Zhang J, Wang JJ. Melatonin attenuates lung ischaemia-reperfusion injury via inhibition of oxidative stress and inflammation. Interact Cardiovasc Thorac Surg. 2018;26:761–767. doi: 10.1093/icvts/ivx440.
    1. Cevik-Aras H, Godoy T, Ekstrom J. Melatonin-induced protein synthesis in the rat parotid gland. J Physiol Pharmacol. 2011;62:95–99.
    1. Jaworek J, Konturek SJ, Leja-Szpak A, Nawrot K, Bonior J, Tomaszewska R, Stachura J, Pawlik WW. Role of endogenous melatonin and its MT2 receptor in the modulation of caerulein-induced pancreatitis in the rat. J Physiol Pharmacol. 2002;53:791–804.
    1. Lee BS, Jun IG, Kim SH, Park JY. Intrathecal gabapentin increases interleukin-10 expression and inhibits pro-inflammatory cytokine in a rat model of neuropathic pain. J Korean Med Sci. 2013;28:308–314. doi: 10.3346/jkms.2013.28.2.308.
    1. Wu SY, Tang SE, Ko FC, Wu GC, Huang KL, Chu SJ. Valproic acid attenuates acute lung injury induced by ischemia-reperfusion in rats. Anesthesiology. 2015;122:1327–1337. doi: 10.1097/ALN.0000000000000618.
    1. Wu GC, Liao WI, Wu SY, Pao HP, Tang SE, Li MH, Huang KL, Chu SJ. Targeting of nicotinamide phosphoribosyltransferase enzymatic activity ameliorates lung damage induced by ischemia/reperfusion in rats. Respir Res. 2017;18:71. doi: 10.1186/s12931-017-0557-2.
    1. Pan X, Zhu L, Lu H, Wang D, Lu Q, Yan H. Melatonin attenuates oxidative damage induced by acrylamide in vitro and in vivo. Oxidative Med Cell Longev. 2015;2015:703709. doi: 10.1155/2015/703709.
    1. Rosen RB, Hu DN, Chen M, McCormick SA, Walsh J, Roberts JE. Effects of melatonin and its receptor antagonist on retinal pigment epithelial cells against hydrogen peroxide damage. Mol Vis. 2012;18:1640–1648.
    1. Zhang HM, Zhang Y. Melatonin: a well-documented antioxidant with conditional pro-oxidant actions. J Pineal Res. 2014;57:131–146. doi: 10.1111/jpi.12162.
    1. Pedreira PR, Garcia-Prieto E, Parra D, Astudillo A, Diaz E, Taboada F, Albaiceta GM. Effects of melatonin in an experimental model of ventilator-induced lung injury. Am J Physiol Lung Cell Mol Physiol. 2008;295:L820–L827. doi: 10.1152/ajplung.90211.2008.
    1. Genovese T, Di Paola R, Mazzon E, Muia C, Caputi AP, Cuzzocrea S. Melatonin limits lung injury in bleomycin treated mice. J Pineal Res. 2005;39:105–112. doi: 10.1111/j.1600-079X.2005.00229.x.
    1. Wu X, Ji H, Wang Y, Gu C, Gu W, Hu L, Zhu L. Melatonin alleviates radiation-induced lung injury via regulation of miR-30e/NLRP3 Axis. Oxidative Med Cell Longev. 2019;2019:4087298.
    1. Stroethoff M, Christoph I, Behmenburg F, Raupach A, Bunte S, Senpolat S, Heinen A, Hollmann MW, Mathes A, Huhn R. Melatonin receptor agonist Ramelteon reduces ischemia-reperfusion injury through activation of mitochondrial potassium channels. J Cardiovasc Pharmacol. 2018;72:106–111. doi: 10.1097/FJC.0000000000000600.
    1. Yanling Qiu, Xiaoning Cheng, Fei Bai, Liyun Fang, Huizhong Hu, Daqing Sun. Inhibition of NLRP9b attenuates acute lung injury through suppressing inflammation, apoptosis and oxidative stress in murine and cell models. Biochemical and Biophysical Research Communications. 2018;503(2):436–443. doi: 10.1016/j.bbrc.2018.04.079.
    1. Kim YH, Park JW, Lee JY, Surh YJ, Kwon TK. Bcl-2 overexpression prevents daunorubicin-induced apoptosis through inhibition of XIAP and Akt degradation. Biochem Pharmacol. 2003;66:1779–1786. doi: 10.1016/S0006-2952(03)00545-8.
    1. Prado Natalia Jorgelina, Casarotto Mariana, Calvo Juan Pablo, Mazzei Luciana, Ponce Zumino Amira Zulma, García Isabel Mercedes, Cuello-Carrión Fernando Darío, Fornés Miguel Walter, Ferder León, Diez Emiliano Raúl, Manucha Walter. Antiarrhythmic effect linked to melatonin cardiorenal protection involves AT1 reduction and Hsp70-VDR increase. Journal of Pineal Research. 2018;65(4):e12513. doi: 10.1111/jpi.12513.
    1. Aschkenasy G, Bromberg Z, Raj N, Deutschman CS, Weiss YG. Enhanced Hsp70 expression protects against acute lung injury by modulating apoptotic pathways. PLoS One. 2011;6:e26956. doi: 10.1371/journal.pone.0026956.
    1. Kondrikov D, Fulton D, Dong Z, Su Y. Heat shock protein 70 prevents Hyperoxia-induced disruption of lung endothelial barrier via Caspase-dependent and AIF-dependent pathways. PLoS One. 2015;10:e0129343. doi: 10.1371/journal.pone.0129343.
    1. Sabirzhanov B, Stoica BA, Hanscom M, Piao CS, Faden AI. Over-expression of HSP70 attenuates caspase-dependent and caspase-independent pathways and inhibits neuronal apoptosis. J Neurochem. 2012;123:542–554. doi: 10.1111/j.1471-4159.2012.07927.x.
    1. Sinha Bharati, Wu Qiaofeng, Li Wei, Tu Yanyang, Sirianni Ana C., Chen Yanchun, Jiang Jiying, Zhang Xinmu, Chen Wu, Zhou Shuanhu, Reiter Russel J., Manning Simon M., Patel Nirav J., Aziz-Sultan Ali M., Inder Terrie E., Friedlander Robert M., Fu Jianfang, Wang Xin. Protection of melatonin in experimental models of newborn hypoxic-ischemic brain injury through MT1 receptor. Journal of Pineal Research. 2017;64(1):e12443. doi: 10.1111/jpi.12443.
    1. Espino J, Ortiz A, Bejarano I, Lozano GM, Monllor F, Garcia JF, Rodriguez AB, Pariente JA. Melatonin protects human spermatozoa from apoptosis via melatonin receptor- and extracellular signal-regulated kinase-mediated pathways. Fertil Steril. 2011;95:2290–2296. doi: 10.1016/j.fertnstert.2011.03.063.
    1. Radogna F, Cristofanon S, Paternoster L, D'Alessio M, De Nicola M, Cerella C, Dicato M, Diederich M, Ghibelli L. Melatonin antagonizes the intrinsic pathway of apoptosis via mitochondrial targeting of Bcl-2. J Pineal Res. 2008;44:316–325. doi: 10.1111/j.1600-079X.2007.00532.x.
    1. Leja-Szpak A, Pierzchalski P, Goralska M, Nawrot-Porabka K, Bonior J, Link-Lenczowski P, Jastrzebska M, Jaworek J. Kynuramines induce overexpression of heat shock proteins in pancreatic cancer cells via 5-hydroxytryptamine and MT1/MT2 receptors. J Physiol Pharmacol. 2015;66:711–718.
    1. Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011;21:103–115. doi: 10.1038/cr.2010.178.
    1. Moniruzzaman M, Ghosal I, Das D, Chakraborty SB. Melatonin ameliorates H2O2-induced oxidative stress through modulation of Erk/Akt/NFkB pathway. Biol Res. 2018;51:17. doi: 10.1186/s40659-018-0168-5.
    1. Peng X, Abdulnour RE, Sammani S, Ma SF, Han EJ, Hasan EJ, Tuder R, Garcia JG, Hassoun PM. Inducible nitric oxide synthase contributes to ventilator-induced lung injury. Am J Respir Crit Care Med. 2005;172:470–479. doi: 10.1164/rccm.200411-1547OC.
    1. Jung KH, Hong SW, Zheng HM, Lee DH, Hong SS. Melatonin downregulates nuclear erythroid 2-related factor 2 and nuclear factor-kappaB during prevention of oxidative liver injury in a dimethylnitrosamine model. J Pineal Res. 2009;47:173–183. doi: 10.1111/j.1600-079X.2009.00698.x.
    1. Hsu JT, Le PH, Lin CJ, Chen TH, Kuo CJ, Chiang KC, Yeh TS. Mechanism of salutary effects of melatonin-mediated liver protection after trauma-hemorrhage: p38 MAPK-dependent iNOS/HIF-1alpha pathway. Am J Physiol Gastrointest Liver Physiol. 2017;312:G427–g433. doi: 10.1152/ajpgi.00440.2016.
    1. Radogna F, Diederich M, Ghibelli L. Melatonin: a pleiotropic molecule regulating inflammation. Biochem Pharmacol. 2010;80:1844–1852. doi: 10.1016/j.bcp.2010.07.041.
    1. Carrillo-Vico A, Lardone PJ, Alvarez-Sanchez N, Rodriguez-Rodriguez A, Guerrero JM. Melatonin: buffering the immune system. Int J Mol Sci. 2013;14:8638–8683. doi: 10.3390/ijms14048638.
    1. Zhao J, Yu H, Liu Y, Gibson SA, Yan Z, Xu X, Gaggar A, Li PK, Li C, Wei S, et al. Protective effect of suppressing STAT3 activity in LPS-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2016;311:L868–l880. doi: 10.1152/ajplung.00281.2016.
    1. Severgnini M, Takahashi S, Rozo LM, Homer RJ, Kuhn C, Jhung JW, Perides G, Steer M, Hassoun PM, Fanburg BL, et al. Activation of the STAT pathway in acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2004;286:L1282–L1292. doi: 10.1152/ajplung.00349.2003.
    1. Braun DA, Fribourg M, Sealfon SC. Cytokine response is determined by duration of receptor and signal transducers and activators of transcription 3 (STAT3) activation. J Biol Chem. 2013;288:2986–2993. doi: 10.1074/jbc.M112.386573.
    1. Hofstetter C, Flondor M, Hoegl S, Muhl H, Zwissler B. Interleukin-10 aerosol reduces proinflammatory mediators in bronchoalveolar fluid of endotoxemic rat. Crit Care Med. 2005;33:2317–2322. doi: 10.1097/01.CCM.0000182815.78568.B2.
    1. Li HD, Zhang QX, Mao Z, Xu XJ, Li NY, Zhang H. Exogenous interleukin-10 attenuates hyperoxia-induced acute lung injury in mice. Exp Physiol. 2015;100:331–340. doi: 10.1113/expphysiol.2014.083337.
    1. Dokka S, Shi X, Leonard S, Wang L, Castranova V, Rojanasakul Y. Interleukin-10-mediated inhibition of free radical generation in macrophages. Am J Physiol Lung Cell Mol Physiol. 2001;280:L1196–L1202. doi: 10.1152/ajplung.2001.280.6.L1196.
    1. Huang CJ, Stevens BR, Nielsen RB, Slovin PN, Fang X, Nelson DR, Skimming JW. Interleukin-10 inhibition of nitric oxide biosynthesis involves suppression of CAT-2 transcription. Nitric Oxide. 2002;6:79–84. doi: 10.1006/niox.2001.0402.
    1. Lee HS, Wang Y, Maciejewski BS, Esho K, Fulton C, Sharma S, Sanchez-Esteban J. Interleukin-10 protects cultured fetal rat type II epithelial cells from injury induced by mechanical stretch. Am J Physiol Lung Cell Mol Physiol. 2008;294:L225–L232. doi: 10.1152/ajplung.00370.2007.
    1. John T, Muller RD, Oberholzer A, Zreiqat H, Kohl B, Ertel W, Hostmann A, Tschoeke SK, Schulze-Tanzil G. Interleukin-10 modulates pro-apoptotic effects of TNF-alpha in human articular chondrocytes in vitro. Cytokine. 2007;40:226–234. doi: 10.1016/j.cyto.2007.10.002.
    1. Borges TJ, Lopes RL, Pinho NG, Machado FD, Souza AP, Bonorino C. Extracellular Hsp70 inhibits pro-inflammatory cytokine production by IL-10 driven down-regulation of C/EBPbeta and C/EBPdelta. Int J Hyperth. 2013;29:455–463. doi: 10.3109/02656736.2013.798037.
    1. Yang Y, Takeda A, Yoshimura T, Oshima Y, Sonoda KH, Ishibashi T. IL-10 is significantly involved in HSP70-regulation of experimental subretinal fibrosis. PLoS One. 2013;8:e80288. doi: 10.1371/journal.pone.0080288.
    1. Wu WS, Chou MT, Chao CM, Chang CK, Lin MT, Chang CP. Melatonin reduces acute lung inflammation, edema, and hemorrhage in heatstroke rats. Acta Pharmacol Sin. 2012;33:775–782. doi: 10.1038/aps.2012.29.
    1. Pandi-Perumal SR, Trakht I, Srinivasan V, Spence DW, Maestroni GJ, Zisapel N, Cardinali DP. Physiological effects of melatonin: role of melatonin receptors and signal transduction pathways. Prog Neurobiol. 2008;85:335–353. doi: 10.1016/j.pneurobio.2008.04.001.
    1. Liu J, Clough SJ, Hutchinson AJ, Adamah-Biassi EB, Popovska-Gorevski M, Dubocovich ML. MT1 and MT2 melatonin receptors: a therapeutic perspective. Annu Rev Pharmacol Toxicol. 2016;56:361–383. doi: 10.1146/annurev-pharmtox-010814-124742.

Source: PubMed

3
Suscribir