Endogenous HMGB1 regulates autophagy

Daolin Tang, Rui Kang, Kristen M Livesey, Chun-Wei Cheh, Adam Farkas, Patricia Loughran, George Hoppe, Marco E Bianchi, Kevin J Tracey, Herbert J Zeh 3rd, Michael T Lotze, Daolin Tang, Rui Kang, Kristen M Livesey, Chun-Wei Cheh, Adam Farkas, Patricia Loughran, George Hoppe, Marco E Bianchi, Kevin J Tracey, Herbert J Zeh 3rd, Michael T Lotze

Abstract

Autophagy clears long-lived proteins and dysfunctional organelles and generates substrates for adenosine triphosphate production during periods of starvation and other types of cellular stress. Here we show that high mobility group box 1 (HMGB1), a chromatin-associated nuclear protein and extracellular damage-associated molecular pattern molecule, is a critical regulator of autophagy. Stimuli that enhance reactive oxygen species promote cytosolic translocation of HMGB1 and thereby enhance autophagic flux. HMGB1 directly interacts with the autophagy protein Beclin1 displacing Bcl-2. Mutation of cysteine 106 (C106), but not the vicinal C23 and C45, of HMGB1 promotes cytosolic localization and sustained autophagy. Pharmacological inhibition of HMGB1 cytoplasmic translocation by agents such as ethyl pyruvate limits starvation-induced autophagy. Moreover, the intramolecular disulfide bridge (C23/45) of HMGB1 is required for binding to Beclin1 and sustaining autophagy. Thus, endogenous HMGB1 is a critical pro-autophagic protein that enhances cell survival and limits programmed apoptotic cell death.

Figures

Figure 1.
Figure 1.
Autophagy promotes extranuclear HMGB1 translocation and is dependent on ROS generation. (A) HMGB1 translocates from the nucleus to the cytosol during autophagy, but not apoptosis. Mouse embryonic fibroblasts (MEFs) and the human Panc2.03 tumor cell line were treated with 1 µM rapamycin (Rap) for 12 h, starvation (HBSS) for 3 h, or UV irradiation at 50 mJ/cm2 for 5 min before a 12-h recovery and then were immunostained with HMGB1-specific antibody (green) and Hoechst 33342 (blue). The mean nuclear (Nuc) and cytosolic (Cyt) HMGB1 intensity per cell was determined by imaging cytometric analysis as described in Materials and methods. *, P < 0.05 and **, P < 0.005 versus untreated (UT) group; n = 3. Representative images are depicted (right). (B) Inhibition of autophagy blocks HMGB1 translocation. Cells were pretreated as indicated with 100 nM wortmannin or 10 µM Ly294002 for 1 h or ATG5-specific shRNA for 48 h and were stimulated with starvation (HBSS) for 3 h and immunostained with HMGB1- or LC3-specific antibody and Hoechst 33342. The mean nuclear/cytosolic HMGB1 intensity and LC3 punctae per cell were determined by imaging cytometric analysis. A representative Western blot of ATG5 level after shRNA and HMGB1 staining is depicted (right). In parallel, the indicated cells were transfected with GFP-LC3 plasmid and assayed for autophagy by quantifying the percentage of cells with GFP-LC3 punctae. *, P < 0.05, **, P < 0.005, and ***, P < 0.0005 versus HBSS group; n = 3. Ctrl, control. (C) Knockdown of ATG5 inhibits LC3-II expression. Western blot analysis of LC3-I/II expression in Panc02 cells under the conditions described in B. Actin was used as a loading control. (D) Hmgb1−/− does not influence ATG5 staining. Hmgb1−/− and Hmgb1+/+ MEFs were immunostained with HMGB1-specific antibody (green), ATG-5–specific antibody (red), and Hoechst 33342 (dark blue). Representative images are depicted in the right panels. (E) mETC inhibitors promote ROS production, autophagy, and HMGB1 translocation. Panc2.03 cells were stimulated with rotenone (Rot), thenoyltrifluoroacetone (TTFA), and antimycin A (AA) at indicated doses for 12 h, and ROS production was assessed by measuring the fluorescent intensity of CM-H2DCFDA in a fluorescent plate reader. In parallel experiments, cells were then immunostained with HMGB1- or LC3-specific antibody and Hoechst 33342. The mean nuclear/cytosolic HMGB1 intensity and LC3 punctae per cell were determined by imaging cytometric analysis. *, P < 0.05, **, P < 0.005, and ***, P < 0.0005 versus untreated group; n = 3. (F) mETC inhibitors increase LC3-II expression and promote HMGB1 translocation. Western blot analysis of LC3-I/II and nuclear/cytosolic HMGB1 expression as indicated in E. Fibrillarin is a nuclear fraction control, and tubulin is a cytoplasmic fraction control. (G) Antioxidant and SOD RNAi limit starvation-induced autophagy and HMGB1 translocation. Panc2.03 cells were pretreated with the antioxidant (NAC) at the indicated concentrations for 1 h or with SOD1 or SOD2 siRNA for 48 h. Then cells were starved (HBSS) for 3 h and analyzed by imaging cytometry to determine the mean nuclear/cytosolic HMGB1 intensity and LC3 punctae per cell. *, P < 0.05; **, P < 0.005; and ***, P < 0.0005 versus HBSS group; n = 3. A representative Western blot for SOD1 and SOD2 level after siRNA is depicted here. (H) Antioxidant and SOD RNAi limit starvation-induced autophagy as measured by LC3-II expression. Western blot analysis of LC3-I/II expression under the conditions indicated in G. Actin was used as a loading control. Data are means ± SEM.
Figure 2.
Figure 2.
Depletion of HMGB1 inhibits autophagy. (A) HMGB1 knockout inhibits LC3 punctae formation. HMGB1−/− and HMGB1+/+ MEFs were treated with autophagic stimuli as indicated, and LC3 punctae formation was detected by LC3 antibody or GFP-LC3 as described in Materials and methods. *, P < 0.05; **, P < 0.005; and ***, P < 0.0005 versus Hmgb1+/+ group; n = 3. UT, untreated. (B) Representative images of LC3 punctae in Hmgb1−/− and HMGB1+/+ MEFs with the indicated treatments are depicted. The percentage of cells showing accumulation of LC3 punctae was reported in A. (C) Analysis of LC3 processing by autophagy in the presence or absence of lysosomal protease inhibitors pepstatin A (pepA) at 10 µg/ml and E64D at 10 µg/ml after starvation treatment for 3 h. **, P < 0.05 versus Hmgb1+/+ group; n = 3. Actin was used as a loading control. AU, arbitrary units. (D) Up-regulation of HMGB1 protein expression restores starvation-induced autophagy. Hmgb1−/− MEFs were transfected with HMGB1 plasmid or empty vector and then were treated with starvation for 3 h. LC3 punctae formation was assayed by imaging cytometric analysis. **, P < 0.05 versus vector group; n = 3. Non, nontransfected. (E) Analysis of p62 processing by autophagy in the presence or absence of lysosomal protease inhibitors pepstatin A at 10 µg/ml and E64D at 10 µg/ml after starvation treatment for 3 h. **, P < 0.005 and ***, P < 0.0005 versus Hmgb1+/+ group; n = 3. Representative images are depicted (left). (F) Ultrastructural features in Hmgb1−/− and Hmgb1+/+ MEFs with or without starvation (HBSS for 3 h) treatment (a–e point to autophagosomes and autolysosomes). Data are means ± SEM.
Figure 3.
Figure 3.
Inhibition of autophagy by cytoplasmic HMGB1. Hmgb1−/− and Hmgb1+/+ MEFs were enucleated by centrifugation after cytochalasin B treatment as described in Materials and methods and then were treated with HBSS for 3 h, and LC3 punctae formation was detected by a confocal microscope. (A) Representative images of LC3 punctae (white arrows) and HMGB1 (red arrows) in cytoplasts of Hmgb1−/− and Hmgb1+/+ MEFs are depicted. (B) The percentage of cells showing accumulation of LC3 punctae was reported (*, P < 0.05; n = 3). Data are means ± SEM.
Figure 4.
Figure 4.
Absence of HMGB1 sustains Beclin1–Bcl-2 interactions. (A) MEK inhibitors block starvation-induced p-ERK and Bcl-2. HMGB1−/− and HMGB1+/+ MEFs were starved in the presence or absence of 10 µM U0126 and 20 µM PD98059 for 6 h. Protein expression levels were assessed as indicated by co-IP or Western blotting. (B) Knockout of HMGB1 limits the disassociation of the Bcl-2–Beclin1 complex during treatment with autophagic stimuli. HMGB1−/− and HMGB1+/+ MEFs were starved in the presence or absence of 10 µM U0126 for 3 h. Protein expression levels were then assayed as indicated by co-IP or Western blotting. (C) HMGB1 interacts directly with Beclin1 during autophagy. HCT116 and Panc02 cells were treated with HBSS for 3 h and then assayed for protein expression levels as indicated by co-IP or Western blotting. (D) HMGB1 direct interactions with Bcl-2 are dependent on Beclin1. Knockdown of Beclin1 and Bcl-2 by siRNA in HMGB1 wild-type MEFs was performed, and protein expression levels were then assayed as indicated by co-IP or Western blotting. Ctrl, control. (E and F) Quantitative data demonstrating the interaction between HMGB1–Beclin1 and Beclin1–HMGB1 using densitometry software to assay the relative protein band intensity in co-IP experiments as shown in A–D. **, P < 0.005 and ***, P < 0.0005; n = 3. UT, untreated. Data are means ± SEM.
Figure 5.
Figure 5.
Oxidation of HMGB1 regulates HMGB1 subcellular localization and autophagy. (A) The C106 mutation (C106S) of HMGB1 impairs its nuclear localization. Hmgb1−/− MEFs were transfected with wild-type and cysteine mutant HMGB1-GFP plasmids as indicated and then were treated with 1 µM rapamycin for 12 h or starved (HBSS) for 3 h. The mean nuclear (Nuc) and cytosolic (Cyt) HMGB1 intensity per cell was analyzed by imaging cytometric analysis. *, P < 0.05 versus HMGB1+/+ group; n = 3. Representative images of HMGB1 location are shown on the left (green, HMGB1; blue, nucleus). The right panel is a schematic diagram of HMGB1 structure illustrating the basic A box and B box as well as the acidic C-terminal domain, with the cysteine mutation locations identified. (B and C) Cytoplasmic HMGB1 enhances autophagy and limits apoptosis. HMGB1−/− MEFs were transfected with wild-type or cysteine mutant HMGB1-GFP plasmids as indicated and then were starved (HBSS) for the indicated time. In a parallel experiment, Hmgb1+/+ MEFs were pretreated with 5 mM ethyl pyruvate (EP) for 2 h and then starved as indicated. LC3 punctae formation was assayed by imaging cytometric analysis (B), and apoptosis was assayed by FACS (C) as described in Materials and methods. *, P < 0.05 and **, P < 0.005; n = 3. UT, untreated. Non, nontransfection. (D) C23/C45 is required for the binding of HMGB1 to Beclin1. Hmgb1−/− MEFs were transfected with wild-type or cysteine mutant HMGB1-GFP plasmids as indicated and stimulated with starvation (HBSS) for 3 h. These cells were then assayed for protein expression levels as indicated by IP or Western blotting. Blots are representative of three independent experiments with similar results. (E) Reducing reagents disrupt the interaction between wild-type/C106 HMGB1 and Beclin1. As a control, before IP, samples were incubated with 50 mM DTT (+DTT) and assayed for protein expression levels as indicated by IP or Western blotting. Blots are representative of two independent experiments with similar results. (F) Knockdown of Beclin1 by siRNA inhibits autophagy under conditions of HMGB1 translocation from the nucleus to the cytosol. Cells were stimulated with HBSS, rapamycin (Rap), rotenone (Rot), or thenoyltrifluoroacetone (TTFA) for 3 h or 12 h, and LC3 punctae formation was assayed as indicated. **, P < 0.005 versus Beclin1 shRNA group; n = 3. Ctrl, control. Data are means ± SEM.
Figure 6.
Figure 6.
Beclin1 is required for HMGB1-mediated autophagy. (A and B) Knockdown of Beclin1 in MEFs by shRNA inhibits exogenous HMGB1-induced autophagy. Cells as indicated were stimulated with 1 µg/ml HMGB1 protein (rHMGB1) for 24 h, and LC3 expression was detected by Western blotting (A). LC3 punctae formation (arrows) was assayed by immunofluorescence (B; n = 3; *, P < 0.05). (C and D) Knockdown of Beclin1 in MEFs by shRNA inhibits HBSS-induced autophagy with and without pUNO1-HMGB1 transfection. Cells as indicated were stimulated with Earle’s balanced salt solution for 3 h, and LC3 expression was detected by Western blotting (C). LC3 punctae formation (arrows) was assayed by immunofluorescence (D; n = 3; *, P < 0.05). Data are means ± SEM.
Figure 7.
Figure 7.
Expression of HMGB1 or the C106S mutant, but not C23S and C45S mutants, restore autophagic flux in Hmgb1−/− MEFs. (A) HMGB1−/− MEFs were transfected with wild-type (WT) or the cysteine mutant HMGB1-GFP plasmids as indicated, starved (HBSS) for 3 h in the presence or absence of 100 nM bafilomycin A1, and then immunostained with LAMP2-specific antibody/Alexa Fluor 594 secondary antibody (shown in red), LC3-specific antibody/Alexa Fluor 647 secondary antibody (shown in green), and Hoechst 33342 (shown in blue). Images were acquired digitally from a randomly selected pool of 10–15 fields under each condition. (B) Quantitative analysis of the percentage of LAMP2/LC3 colocalization was detected by Image-Pro Plus 5.1 software. Data are means ± SEM.
Figure 8.
Figure 8.
Conceptual relationships between endogenous HMGB1 and autophagy. ROS trigger HMGB1 translocation to the cytosol in the setting of starvation-mediated autophagy. Cytosolic HMGB1 then binds Beclin1, which requires C23/45. This results in dissociation of Beclin1–Bcl-2 and subsequent induction of autophagy. C106 mutation (C106S) in HMGB1 impairs its nuclear localization and promotes autophagy. Inhibition of HMGB1 translocation by ethyl pyruvate (EP) blocks autophagy. Additionally, HMGB1 promotes phosphorylation and activation of the ERK1/2 (p-ERK1/2) pathway, which is an important autophagy-dependent signal pathway.

References

    1. Anathy V., Aesif S.W., Guala A.S., Havermans M., Reynaert N.L., Ho Y.S., Budd R.C., Janssen-Heininger Y.M. 2009. Redox amplification of apoptosis by caspase-dependent cleavage of glutaredoxin 1 and S-glutathionylation of Fas. J. Cell Biol. 184:241–252 10.1083/jcb.200807019
    1. Bjørkøy G., Lamark T., Brech A., Outzen H., Perander M., Overvatn A., Stenmark H., Johansen T. 2005. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J. Cell Biol. 171:603–614 10.1083/jcb.200507002
    1. Goldman R.D., Pollack R., Hopkins N.H. 1973. Preservation of normal behavior by enucleated cells in culture. Proc. Natl. Acad. Sci. USA. 70:750–754 10.1073/pnas.70.3.750
    1. Hoppe G., Talcott K.E., Bhattacharya S.K., Crabb J.W., Sears J.E. 2006. Molecular basis for the redox control of nuclear transport of the structural chromatin protein Hmgb1. Exp. Cell Res. 312:3526–3538 10.1016/j.yexcr.2006.07.020
    1. Ishihara K., Tsutsumi K., Kawane S., Nakajima M., Kasaoka T. 2003. The receptor for advanced glycation end-products (RAGE) directly binds to ERK by a D-domain-like docking site. FEBS Lett. 550:107–113 10.1016/S0014-5793(03)00846-9
    1. Kang R., Tang D., Schapiro N.E., Livesey K.M., Farkas A., Loughran P., Bierhaus A., Lotze M.T., Zeh H.J. 2010a. The receptor for advanced glycation end products (RAGE) sustains autophagy and limits apoptosis, promoting pancreatic tumor cell survival. Cell Death Differ. 17:666–676 10.1038/cdd.2009.149
    1. Kang R., Tang D., Yu Y., Wang Z., Hu T., Wang H., Cao L. 2010b. WAVE1 regulates Bcl-2 localization and phosphorylation in leukemia cells. Leukemia. 24:177–186 10.1038/leu.2009.224
    1. Kazama H., Ricci J.E., Herndon J.M., Hoppe G., Green D.R., Ferguson T.A. 2008. Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity. 29:21–32 10.1016/j.immuni.2008.05.013
    1. Levine B., Kroemer G. 2008. Autophagy in the pathogenesis of disease. Cell. 132:27–42 10.1016/j.cell.2007.12.018
    1. Livesey K.M., Tang D., Zeh H.J., Lotze M.T. 2009. Autophagy inhibition in combination cancer treatment. Curr. Opin. Investig. Drugs. 10:1269–1279
    1. Lotze M.T., Tracey K.J. 2005. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat. Rev. Immunol. 5:331–342 10.1038/nri1594
    1. Maiuri M.C., Zalckvar E., Kimchi A., Kroemer G. 2007. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 8:741–752 10.1038/nrm2239
    1. Miller R.A., Ruddle F.H. 1974. Enucleated neuroblastoma cells form neurites when treated with dibutyryl cyclic AMP. J. Cell Biol. 63:295–299 10.1083/jcb.63.1.295
    1. Mizushima N., Yoshimori T. 2007. How to interpret LC3 immunoblotting. Autophagy. 3:542–545
    1. Pattingre S., Tassa A., Qu X., Garuti R., Liang X.H., Mizushima N., Packer M., Schneider M.D., Levine B. 2005. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 122:927–939 10.1016/j.cell.2005.07.002
    1. Scaffidi P., Misteli T., Bianchi M.E. 2002. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 418:191–195 10.1038/nature00858
    1. Scherz-Shouval R., Elazar Z. 2007. ROS, mitochondria and the regulation of autophagy. Trends Cell Biol. 17:422–427 10.1016/j.tcb.2007.07.009
    1. Scherz-Shouval R., Shvets E., Fass E., Shorer H., Gil L., Elazar Z. 2007. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J. 26:1749–1760 10.1038/sj.emboj.7601623
    1. Shao Y., Gao Z., Marks P.A., Jiang X. 2004. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc. Natl. Acad. Sci. USA. 101:18030–18035 10.1073/pnas.0408345102
    1. Sims G.P., Rowe D.C., Rietdijk S.T., Herbst R., Coyle A.J. 2010. HMGB1 and RAGE in inflammation and cancer. Annu. Rev. Immunol. 28:367–388 10.1146/annurev.immunol.021908.132603
    1. Subramanian M., Shaha C. 2007. Up-regulation of Bcl-2 through ERK phosphorylation is associated with human macrophage survival in an estrogen microenvironment. J. Immunol. 179:2330–2338
    1. Tang D., Kang R., Xiao W., Jiang L., Liu M., Shi Y., Wang K., Wang H., Xiao X. 2007a. Nuclear heat shock protein 72 as a negative regulator of oxidative stress (hydrogen peroxide)-induced HMGB1 cytoplasmic translocation and release. J. Immunol. 178:7376–7384
    1. Tang D., Kang R., Xiao W., Wang H., Calderwood S.K., Xiao X. 2007b. The anti-inflammatory effects of heat shock protein 72 involve inhibition of high-mobility-group box 1 release and proinflammatory function in macrophages. J. Immunol. 179:1236–1244
    1. Tang D., Shi Y., Kang R., Li T., Xiao W., Wang H., Xiao X. 2007c. Hydrogen peroxide stimulates macrophages and monocytes to actively release HMGB1. J. Leukoc. Biol. 81:741–747 10.1189/jlb.0806540
    1. Tang D., Kang R., Xiao W., Zhang H., Lotze M.T., Wang H., Xiao X. 2009. Quercetin prevents LPS-induced high-mobility group box 1 release and proinflammatory function. Am. J. Respir. Cell Mol. Biol. 41:651–660 10.1165/rcmb.2008-0119OC
    1. Tang D., Kang R., Cheh C.W., Livesey K.M., Liang X., Schapiro N.E., Benschop R., Sparvero L.J., Amoscato A.A., Tracey K.J., et al. 2010a. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene. 10.1038/onc.2010.261
    1. Tang D., Kang R., Zeh H.J., III, Lotze M.T. 2010b. High-mobility group box 1 and cancer. Biochim. Biophys. Acta. 1799:131–140
    1. Tasdemir E., Maiuri M.C., Galluzzi L., Vitale I., Djavaheri-Mergny M., D’Amelio M., Criollo A., Morselli E., Zhu C., Harper F., et al. 2008. Regulation of autophagy by cytoplasmic p53. Nat. Cell Biol. 10:676–687 10.1038/ncb1730
    1. Tsung A., Klune J.R., Zhang X., Jeyabalan G., Cao Z., Peng X., Stolz D.B., Geller D.A., Rosengart M.R., Billiar T.R. 2007. HMGB1 release induced by liver ischemia involves Toll-like receptor 4 dependent reactive oxygen species production and calcium-mediated signaling. J. Exp. Med. 204:2913–2923 10.1084/jem.20070247
    1. Ulloa L., Ochani M., Yang H., Tanovic M., Halperin D., Yang R., Czura C.J., Fink M.P., Tracey K.J. 2002. Ethyl pyruvate prevents lethality in mice with established lethal sepsis and systemic inflammation. Proc. Natl. Acad. Sci. USA. 99:12351–12356 10.1073/pnas.192222999
    1. Wang H., Bloom O., Zhang M., Vishnubhakat J.M., Ombrellino M., Che J., Frazier A., Yang H., Ivanova S., Borovikova L., et al. 1999. HMG-1 as a late mediator of endotoxin lethality in mice. Science. 285:248–251 10.1126/science.285.5425.248
    1. Wei Y., Pattingre S., Sinha S., Bassik M., Levine B. 2008. JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol. Cell. 30:678–688 10.1016/j.molcel.2008.06.001
    1. Yanai H., Ban T., Wang Z., Choi M.K., Kawamura T., Negishi H., Nakasato M., Lu Y., Hangai S., Koshiba R., et al. 2009. HMGB proteins function as universal sentinels for nucleic-acid-mediated innate immune responses. Nature. 462:99–103 10.1038/nature08512
    1. Zhang Q., Raoof M., Chen Y., Sumi Y., Sursal T., Junger W., Brohi K., Itagaki K., Hauser C.J. 2010. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 464:104–107 10.1038/nature08780

Source: PubMed

Подписаться