Macrophage- and RIP3-dependent inflammasome activation exacerbates retinal detachment-induced photoreceptor cell death

K Kataoka, H Matsumoto, H Kaneko, S Notomi, K Takeuchi, J H Sweigard, A Atik, Y Murakami, K M Connor, H Terasaki, J W Miller, D G Vavvas, K Kataoka, H Matsumoto, H Kaneko, S Notomi, K Takeuchi, J H Sweigard, A Atik, Y Murakami, K M Connor, H Terasaki, J W Miller, D G Vavvas

Abstract

Detachment of photoreceptors from the retinal pigment epithelium is seen in various retinal disorders, resulting in photoreceptor death and subsequent vision loss. Cell death results in the release of endogenous molecules that activate molecular platforms containing caspase-1, termed inflammasomes. Inflammasome activation in retinal diseases has been reported in some cases to be protective and in others to be detrimental, causing neuronal cell death. Moreover, the cellular source of inflammasomes in retinal disorders is not clear. Here, we demonstrate that patients with photoreceptor injury by retinal detachment (RD) have increased levels of cleaved IL-1β, an end product of inflammasome activation. In an animal model of RD, photoreceptor cell death led to activation of endogenous inflammasomes, and this activation was diminished by Rip3 deletion. The major source of Il1b expression was found to be infiltrating macrophages in the subretinal space, rather than dying photoreceptors. Inflammasome inhibition attenuated photoreceptor death after RD. Our data implicate the infiltrating macrophages as a source of damaging inflammasomes after photoreceptor detachment in a RIP3-dependent manner and suggest a novel therapeutic target for treatment of retinal diseases.

Figures

Figure 1
Figure 1
Concentration of IL-1β in human eyes with rhegmatogenous retinal detachment (RRD). IL-1β levels in vitreous fluid of control eyes (n=7) versus subretinal fluid (SRF) of eyes with RRD (n=8) and vitreous fluid of eyes with RRD (n=8). Horizontal line indicates median in each group. ***P<0.001
Figure 2
Figure 2
Retinal detachment (RD) induces activation of IL-1β, which is diminished in Rip3-deficient mice. (a) The data show IL-1β levels detected by ELISA at 0 h (non-treated eyes, n=6) and from 6 to 120 h after induction of RD. n=6–9. (b) Western blotting image shows representative data of cleaved IL-1β and β-actin in wild-type (WT) and Rip3−/− mouse eyes after RD. (-): non-treated eyes. (c) Densitometry analyses of the western blotting data of (b) normalized to the intensity of β-actin (n=4). Data are presented as mean±s.d. *P<0.05, **P<0.01, ***P<0.001
Figure 3
Figure 3
IL-1β is produced by the cells migrating into subretinal space. (a) ONL, subretinal space, and RPE including choroid were circled as indicated in the left column (red circles), followed by cutting with LCM. The right column shows the images after cutting out the samples from the sections. Nuclei were stained with Toluidine blue. INL, inner nuclear layer; ONL, outer nuclear layer; RPE, retinal pigment epithelium. Scale bars: 100 μm. (b) The graph shows the Il1b mRNA expression in each layers with (+) RD (n=12) or without (−) RD (n=6). ND, not detectable. Data are presented as mean±s.d. *P<0.05
Figure 4
Figure 4
Macrophage recruitment is not altered in Rip3−/− mice. (a) The images show macrophages stained with CD11b (green) that have migrated into the subretinal space after 12 and 24 h in WT mice and Rip3−/− mice. Nuclei staining: TO-PRO-3 (blue). Scale bar: 50 μm. (b) The number of macrophages in the subretinal space at 12 and 24 h after RD (n=6). (-): non-treated eyes. Data are presented as mean±s.d. *P<0.05
Figure 5
Figure 5
Inhibition of caspase-1 reduces photoreceptor cell death after RD. (a) Inhibitor of caspase-1 (YVAD) was administered into the subretinal space, and cleaved IL-1β was measured with western blotting to evaluate the efficiency of caspase-1 inhibition. (b) Densitometry analyses of the western blotting data of (a) normalized to the intensity of β-actin (n=4). (c) TUNEL staining (green) in WT and Rip3−/− mouse retinal sections. The eyes were treated with vehicle and YVAD and analyzed at 24 h after RD. Nuclei staining: TO-PRO-3 (blue), scale bar: 50 μm. (d) Quantification of TUNEL-positive cells in the ONL after RD. n=4–10 at each groups. (a and b) Data are presented as mean±s.d. *P<0.05, **P<0.01
Figure 6
Figure 6
Inhibition of IL-1β through Nlrp3 depletion or a neutralizing antibody diminishes photoreceptor cell death after RD. (a) Cleaved IL-1β was assessed at 24 h after RD in WT and Nlrp3−/− mice eyes with western blotting. (b) Densitometry analyses of the western blotting data of (b) normalized to the intensity of β-actin (n=4). (c and d) TUNEL staining (green) in WT and Nlrp3−/− mouse retinal sections at 24 h after RD (c) and quantification of TUNEL-positive cells in the ONL (d). WT: n=8, Nlrp3−/− mice: n=7. (e and f) Neutralizing anti-IL-1β antibody (10 and 100 μg/ml) and control IgG were administered into subretina and analyzed at 24 h after RD. TUNEL staining (green) in mouse retinal sections treated with antibody (e) and quantification of TUNEL-positive cells in the ONL (f). n=8. Nuclei staining: TO-PRO-3 (blue), scale bar: 50 μm. Data are presented as mean±s.d. *P<0.05, **P<0.01
Figure 7
Figure 7
Model of mechanism in RD-induced photoreceptor cell death. After photoreceptor separation from RPE, caspase and RIPK mediated cell death ensues as well as release of inflammatory mediators and DAMPs. Ensuing macrophage recruitment results in inflammasome activation and release of IL-1b. Rip3 deletion attenuates both directly and indirectly inflammasome activation. Neutralization of IL-1b or inhibition of inflammasome activation partially diminishes cell death

References

    1. 1Bird A, Bressler N, Bressler S, Chisholm IH, Coscas G, Davis M et al. An international classification and grading system for age-related maculopathy and age-related macular degeneration. Surv Ophthalmol 1995; 39: 367–374.
    1. 2Aiello L. Perspectives on diabetic retinopathy. Am J Ophthalmol 2003; 136: 122–135.
    1. 3Arroyo J, Yang L, Bula D, Chen D. Photoreceptor apoptosis in human retinal detachment. Am J Ophthalmol 2005; 139: 605–610.
    1. 4Cook B, Lewis G, Fisher S, Adler R. Apoptotic photoreceptor degeneration in experimental retinal detachment. Invest Ophthalmol Vis Sci 1995; 36: 990–996.
    1. 5Trichonas G, Murakami Y, Thanos A, Morizane Y, Kayama M, Debouck C et al. Receptor interacting protein kinases mediate retinal detachment-induced photoreceptor necrosis and compensate for inhibition of apoptosis. Proc Natl Acad Sci USA 2010; 107: 21695–21700.
    1. 6Mervin K, Valter K, Maslim J, Lewis G, Fisher S, Stone J. Limiting photoreceptor death and deconstruction during experimental retinal detachment: the value of oxygen supplementation. Am J Ophthalmol 1999; 128: 155–164.
    1. 7Sakai T, Lewis G, Linberg K, Fisher S. The ability of hyperoxia to limit the effects of experimental detachment in cone-dominated retina. Invest Ophthalmol Vis Sci 2001; 42: 3264–3273.
    1. 8Cho Y, Challa S, Moquin D, Genga R, Ray T, Guildford M et al. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 2009; 137: 1112–1123.
    1. 9He S, Wang L, Miao L, Wang T, Du F, Zhao L et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 2009; 137: 1100–1111.
    1. 10Murakami Y, Matsumoto H, Roh M, Suzuki J, Hisatomi T, Ikeda Y et al. Receptor interacting protein kinase mediates necrotic cone but not rod cell death in a mouse model of inherited degeneration. Proc Natl Acad Sci USA 2012; 109: 14598–14603.
    1. 11Murakami Y, Matsumoto H, Roh M, Giani A, Kataoka K, Morizane Y et al. Programmed necrosis, not apoptosis, is a key mediator of cell loss and DAMP-mediated inflammation in dsRNA-induced retinal degeneration. Cell Death Differ 2013; 21: 270–277.
    1. 12Upton J, Kaiser W, Mocarski E. Virus inhibition of RIP3-dependent necrosis. Cell Host Microbe 2010; 7: 302–313.
    1. 13Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S et al. Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol 2000; 1: 489–495.
    1. 14He S, Liang Y, Shao F, Wang X. Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3-mediated pathway. Proc Natl Acad Sci USA 2011; 108: 20054–20059.
    1. 15Sun L, Wang H, Wang Z, He S, Chen S, Liao D et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 2012; 148: 213–227.
    1. 16Chen X, Li W, Ren J, Huang D, He W, Song Y et al. Translocation of mixed lineage kinase domain-like protein to plasma membrane leads to necrotic cell death. Cell Res 2013; 24: 105–121.
    1. 17Cai Z, Jitkaew S, Zhao J, Chiang H, Choksi S, Liu J et al. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol 2014; 16: 55–65.
    1. 18Wang H, Sun L, Su L, Rizo J, Liu L, Wang LF et al. Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol Cell 2014; 54: 133–146.
    1. 19Scaffidi P, Misteli T, Bianchi M. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002; 418: 191–195.
    1. 20Di Virgilio F. Dr. Jekyll/Mr. Hyde: the dual role of extracellular ATP. J Auton Nerv Syst 2000; 81: 59–63.
    1. 21Bianchi M. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol 2007; 81: 1–5.
    1. 22Shi Y, Evans J, Rock K. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature 2003; 425: 516–521.
    1. 23Srinivasula S, Poyet J-L, Razmara M, Datta P, Zhang Z, Alnemri E. The PYRIN-CARD protein ASC is an activating adaptor for caspase-1. J Biol Chem 2002; 277: 21119–21122.
    1. 24Thornberry N, Bull H, Calaycay J, Chapman K, Howard A, Kostura M et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature 1992; 356: 768–774.
    1. 25Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 2002; 10: 417–426.
    1. 26Mariathasan S, Weiss D, Newton K, McBride J, O'Rourke K, Roose-Girma M et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 2006; 440: 228–232.
    1. 27Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006; 440: 237–241.
    1. 28Iyer S, Pulskens W, Sadler J, Butter L, Teske G, Ulland T et al. Necrotic cells trigger a sterile inflammatory response through the Nlrp3 inflammasome. Proc Natl Acad Sci USA 2009; 106: 20388–20393.
    1. 29Arimura N, Ki-i Y, Hashiguchi T, Kawahara K, Biswas K, Nakamura M et al. Intraocular expression and release of high-mobility group box 1 protein in retinal detachment. Lab Invest 2009; 89: 278–289.
    1. 30Kawano H, Ito T, Yamada S, Hashiguchi T, Maruyama I, Hisatomi T et al. Toxic effects of extracellular histones and their neutralization by vitreous in retinal detachment. Lab Invest 2014; 94: 569–585.
    1. 31Tarallo V, Hirano Y, Gelfand B, Dridi S, Kerur N, Kim Y et al. DICER1 loss and Alu RNA induce age-related macular degeneration via the NLRP3 inflammasome and MyD88. Cell 2012; 149: 847–859.
    1. 32Yoneda S, Tanihara H, Kido N, Honda Y, Goto W, Hara H et al. Interleukin-1beta mediates ischemic injury in the rat retina. Exp Eye Res 2001; 73: 661–667.
    1. 33Doyle SL, Campbell M, Ozaki E, Salomon RG, Mori A, Kenna PF et al. NLRP3 has a protective role in age-related macular degeneration through the induction of IL-18 by drusen components. Nat Med 2012; 18: 791–798.
    1. 34Lavalette S, Raoul W, Houssier M, Camelo S, Levy O, Calippe B et al. Interleukin-1β inhibition prevents choroidal neovascularization and does not exacerbate photoreceptor degeneration. Am J Pathol 2011; 178: 2416–2423.
    1. 35Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 2010; 140: 805–820.
    1. 36Yoshimura T, Sonoda KH, Sugahara M, Mochizuki Y. Comprehensive analysis of inflammatory immune mediators in vitreoretinal diseases. PLoS One 2009; 4: e8158.
    1. 37Ricker L, Kijlstra A, Jager W, Liem A, Hendrikse F, Hiji E. Chemokine levels in subretinal fluid obtained during scleral buckling surgery after rhegmatogenous retinal detachment. Invest Ophthalmol Vis Sci 2010; 51: 4143–4150.
    1. 38Ricker L, Kijlstra A, Kessels A, Jager W, Liem A, Hendrikse F et al. Interleukin and growth factor levels in subretinal fluid in rhegmatogenous retinal detachment: a case-control study. PLoS One 2011.
    1. 39Rasier R, Gormus U, Artunay O, Yuzbasioglu E, Oncel M, Bahcecioglu H. Vitreous levels of VEGF, IL-8, and TNF-alpha in retinal detachment. Curr Eye Res 2010; 35: 505–509.
    1. 40Nakazawa T, Hisatomi T, Nakazawa C, Noda K, Maruyama K, She H et al. Monocyte chemoattractant protein 1 mediates retinal detachment-induced photoreceptor apoptosis. Proc Natl Acad Sci USA 2007; 104: 2425–2430.
    1. 41Nakazawa T, Matsubara A, Noda K, Hisatomi T, She H, Skondra D et al. Characterization of cytokine responses to retinal detachment in rats. Mol Vis 2006; 12: 867–878.
    1. 42Tseng W, Thein T, Kinnunen K, Lashkari K, Gregory M, D'Amore P et al. NLRP3 inflammasome activation in retinal pigment epithelial cells by lysosomal destabilization: implications for age-related macular degeneration. Invest Ophthalmol Vis Sci 2013; 54: 110–120.
    1. 43Ghazi N, Green W. Pathology and pathogenesis of retinal detachment. Eye (London, England) 2002; 16: 411–421.
    1. 44Minami M, Kuraishi Y, Yabuuchi K, Yamazaki A, Satoh M. Induction of interleukin-1 beta mRNA in rat brain after transient forebrain ischemia. J Neurochem 1992; 58: 390–392.
    1. 45Fan L, Young P, Barone F, Feuerstein G, Smith D, McIntosh T. Experimental brain injury induces expression of interleukin-1 beta mRNA in the rat brain. Brain Res Mol Brain Res 1995; 30: 125–130.
    1. 46Hisatomi T, Sakamoto T, Murata T, Yamanaka I, Oshima Y, Hata Y et al. Relocalization of apoptosis-inducing factor in photoreceptor apoptosis induced by retinal detachment in vivo. Am J Pathol 2001; 158: 1271–1278.
    1. 47Zacks DN, Hänninen V, Pantcheva M, Ezra E, Grosskreutz C, Miller JW. Caspase activation in an experimental model of retinal detachment. Invest Ophthalmol Vis Sci 2003; 44: 1262–1267.
    1. 48Yang L, Bula D, Arroyo J, Chen D. Preventing retinal detachment–associated photoreceptor cell loss in bax-deficient mice. Invest Ophthalmol Vis Sci 2004; 45: 648–654.
    1. 49Matsumoto H, Miller J, Vavvas D. Retinal detachment model in rodents by subretinal injection of sodium hyaluronate. J Vis Exp 2013; 79: e50660.
    1. 50Matsumoto H, Murakami Y, Kataoka K, Lin H, Connor K, Miller J et al. Mammalian STE20-like kinase 2, not kinase 1, mediates photoreceptor cell death during retinal detachment. Cell Death Dis 2014; 29: e1269.
    1. 51Matsumoto H, Kataoka K, Tsoka PA, Connor KM, Miller JW, Vavvas DG. Strain difference in photoreceptor cell death after retinal detachment in mice. Invest Ophthalmol Vis Sci 2014; 55: 4165–4174.
    1. 52Vince J, Wong W, Gentle I, Lawlor K, Allam R, O'Reilly L et al. Inhibitor of apoptosis proteins limit RIP3 kinase-dependent interleukin-1 activation. Immunity 2012; 36: 215–227.
    1. 53Kang T-B, Yang S-H, Toth B, Kovalenko A, Wallach D. Caspase-8 blocks kinase RIPK3-mediated activation of the NLRP3 inflammasome. Immunity 2013; 38: 27–40.
    1. 54Watanabe H, Gaide O, Pétrilli V, Martinon F, Contassot E, Roques S et al. Activation of the IL-1beta-processing inflammasome is involved in contact hypersensitivity. J Invest Dermatol 2007; 127: 1956–1963.
    1. 55Hisatomi T, Sakamoto T, Sonoda K-H, Tsutsumi C, Qiao H, Enaida H et al. Clearance of apoptotic photoreceptors: elimination of apoptotic debris into the subretinal space and macrophage-mediated phagocytosis via phosphatidylserine receptor and integrin alphavbeta3. Am J Pathol 2003; 162: 1869–1879.
    1. 56Johnson N, Foulds W. Observations on the retinal pigment epithelium and retinal macrophages in experimental retinal detachment. Br J Ophthalmol 1977; 61: 564–572.
    1. 57Thornton P, Pinteaux E, Gibson R, Allan S, Rothwell N. Interleukin-1-induced neurotoxicity is mediated by glia and requires caspase activation and free radical release. J Neurochem 2006; 98: 258–266.
    1. 58Rothwell N, Strijbos P. Cytokines in neurodegeneration and repair. Int J Dev Neurosci 1995; 13: 179–185.
    1. 59Pinteaux E, Trotter P, Simi A. Cell-specific and concentration-dependent actions of interleukin-1 in acute brain inflammation. Cytokine 2009; 45: 1–7.
    1. 60LaVail M, Unoki K, Yasumura D, Matthes M, Yancopoulos G, Steinberg R. Multiple growth factors, cytokines, and neurotrophins rescue photoreceptors from the damaging effects of constant light. Proc Natl Acad Sci USA 1992; 89: 11249–11253.
    1. 61Fogal B, Hewett J, Hewett S. Interleukin-1beta potentiates neuronal injury in a variety of injury models involving energy deprivation. J Neuroimmunol 2005; 161: 93–100.
    1. 62Vandenabeele P, Berghe V, Festjens N. Caspase inhibitors promote alternative cell death pathways. Sci Signal 2006; 2006: pe44.
    1. 63Smith J, Douty E. Electrophoresis of subretinal fluid. Arch Ophthalmol 1960; 64: 114–119.
    1. 64Heath H, Beck T, Foulds W. Chemical composition of subretinal fluid. Br J Ophthalmol 1962; 46: 385–396.
    1. 65Bishop P. The biochemical structure of mammalian vitreous. Eye 1996; 10: 664–670.
    1. 66Lam K, Constable I, Schepens C. Subretinal fluid: Isoenzymes and cytologic studies. Invest Ophthalmol Vis Sci 1972; 11: 1037–1043.
    1. 67Feeney L, Burns R, Mixon R. Human subretinal fluid: its cellular and subcellular components. Arch Ophthalmol 1975; 93: 62–69.

Source: PubMed

3
Prenumerera