HMGB1 release and redox regulates autophagy and apoptosis in cancer cells

D Tang, R Kang, C-W Cheh, K M Livesey, X Liang, N E Schapiro, R Benschop, L J Sparvero, A A Amoscato, K J Tracey, H J Zeh, M T Lotze, D Tang, R Kang, C-W Cheh, K M Livesey, X Liang, N E Schapiro, R Benschop, L J Sparvero, A A Amoscato, K J Tracey, H J Zeh, M T Lotze

Abstract

The functional relationship and cross-regulation between autophagy and apoptosis is complex. In this study we show that the high-mobility group box 1 protein (HMGB1) is a redox-sensitive regulator of the balance between autophagy and apoptosis. In cancer cells, anticancer agents enhanced autophagy and apoptosis, as well as HMGB1 release. HMGB1 release may be a prosurvival signal for residual cells after various cytotoxic cancer treatments. Diminished HMGB1 by short hairpin RNA transfection or inhibition of HMGB1 release by ethyl pyruvate or other small molecules led predominantly to apoptosis and decreased autophagy in stressed cancer cells. In this setting, reducible HMGB1 binds to the receptor for advanced glycation end products (RAGEs), but not to Toll-like receptor 4, induces Beclin1-dependent autophagy and promotes tumor resistance to alkylators (melphalan), tubulin disrupting agents (paclitaxel), DNA crosslinkers (ultraviolet light) and DNA intercalators (oxaliplatin or adriamycin). On the contrary, oxidized HMGB1 increases the cytotoxicity of these agents and induces apoptosis mediated by the caspase-9/-3 intrinsic pathway. HMGB1 release, as well as its redox state, thus links autophagy and apoptosis, representing a suitable target when coupled with conventional tumor treatments.

Conflict of interest statement

CONFLICT OF INTEREST

The authors declare no conflict of interest.

Figures

Figure 1. Cell injury/stress promotes HMGB1 release…
Figure 1. Cell injury/stress promotes HMGB1 release from cancer cell lines
(A) Small molecule anticancer agents decreased cell viability and induced both apoptotic and autophagic pathways. Panc02 and HCT116 cancer cells were treated with either a DNA alkylating agent or a tubulin depolymerization inhibitor (melphalan, “ME”, 160 μg/ml; paclitaxel, “PA”, 10 μg/ml respectively) for 0–24 h, and then assayed for cell viability using measures of NADH dehydrogenases [CCK8], apoptosis by flow cytometric analaysis (right panel) using Annexin V/PI stain and autophagy by quantification of the percentage of cells with GFP-LC3 punctae as described in methods (N=3, * p

Figure 2. Inhibition of autophagy diminishes HMGB1…

Figure 2. Inhibition of autophagy diminishes HMGB1 release and enhances selective apoptosis

(A) Immunoblots are…

Figure 2. Inhibition of autophagy diminishes HMGB1 release and enhances selective apoptosis
(A) Immunoblots are shown for Beclin1 and ATG5 knockdown performed in Panc02 cells. (B–D) Panc02 cells as indicated were treated with the anticancer agents (melphalan, “ME”, 160 μg/ml; paclitaxel, “PA”, 10 μg/ml) for 6 h. and then assayed for early apoptosis (annexin V+/PI−) by flow cytometry (B), autophagy by quantification of the percentage of cells with GFP-LC3 punctae (C) and HMGB1 release, by western blotting analysis (LDH and H3 were both used as controls for protein leakage from damaged cells) (D). PI3-kinase inhibitor 3-methyladenine (3MA, 5mm) was used as a nominal autophagy inhibitor. Representative western blots of the indicated proteins are presented.

Figure 3. HMGB1 release and autophagy is…

Figure 3. HMGB1 release and autophagy is detected in the absence of measurable apoptosis

(A)…

Figure 3. HMGB1 release and autophagy is detected in the absence of measurable apoptosis
(A) Immunoblots are shown for Bax and p53 knockout in HCT116 cells. (B-D) WT, Bax knock out, p53 knockout or pan-caspase inhibitor treated (ZVAD-FMK, 20 μm) HCT116 cells were treated with melphalan, “ME”, 160 μg/ml or paclitaxel, “PA”, 10 μg/ml for 6 h. and then assayed for measures of early apoptosis (annexin V+/PI−) by flow cytometry (B), autophagy by quantification of the percentage of cells with GFP-LC3 punctae (C) and HMGB1 release by western blot analysis (LDH and H3 were both used as controls for protein leakage from damaged cells) (D). Representative western blots of the indicated proteins are presented.

Figure 4. Inhibition of HMGB1 release increases…

Figure 4. Inhibition of HMGB1 release increases tumor cell sensitivity to anticancer agents

(A) Inhibition…

Figure 4. Inhibition of HMGB1 release increases tumor cell sensitivity to anticancer agents
(A) Inhibition of HMGB1 release with small molecule drugs increases tumor cell sensitivity to anticancer agents. Panc2.03 and HCT116 cells were pretreated with the HMGB1-release inhibitors ethyl pyruvate (EP, 10 mm) or glycyrrhizin (Gly, 500 μm) for 2 h and then cultured in the presence of melphalan for an additional 24 h. Representative western blotting analysis of protein levels are presented. In parallel, measures of apoptosis (annexin V+/PI−) were assayed by flow cytometry and autophagy by quantifying the percentage of cells with GFP-LC3 punctae. (B) Panc2.03 and HCT116 cells were knocked down for HMGB1 using shRNA for 48 h, and then stimulated with melphalan for 24 h. Representative western blotting analysis of protein levels is presented. In parallel, apoptosis (annexin V+/PI−) was assayed by flow cytometry (right panel) and autophagy by quantifying the percentage of cells with GFP-LC3 punctae (N=3, p

Figure 5. Provision of exogenous reduced HMGB1…

Figure 5. Provision of exogenous reduced HMGB1 increases autophagy in cancer cells

(A) Relative amounts…

Figure 5. Provision of exogenous reduced HMGB1 increases autophagy in cancer cells
(A) Relative amounts of oxidized Cys106 (as Cys sulfonic acid) in Lilly Pool #2 and #3. MALDI-TOF Mass Spectrum of tryptic fragments of Lilly Pool #2 (top) and Pool #3 (bottom). The Cys106 containing fragment is amino acids 97–112. The free sulfhydryl (-SH) of total reducible cysteine is at a mass of 1944.9 Da. The monoxide is faintly seen at a mass of 1960.9 Da. The di- and tri- oxides are at masses of 1976.9 Da and 1992.9 Da, respectively. The peak at 1962.9 Da is the free sulfhydryl of the 13–28 fragments, used as an internal standard to verify the DTT reduction went to completion. (B) Reduced HMGB1 protein induces autophagy and oxidized HMGB1 mildly induces apoptosis. Panc2.03 and HCT116 cancer cells were treated with oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml) for 24 h, and then assayed for apoptosis by FACS using Annexin V/PI stain and autophagy by quantification of the percentage of cells with GFP-LC3 dots as described in methods. (C) Western analysis of LC3 processing in the presence or absence of lysosomal protease inhibitors pepstatin A (10 μg/ml) and E64D (10 μg/ml) and degradation of p62 by autophagy after HMGB1 or HMGB1 C106S mutant treatment. (D) Reduced HMGB1 protein regulates Beclin1/Bcl-2 complex formation in autophagy. Panc2.03 cells were treated with oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml), for 6 h, then cell lysates were prepared for IP with anti-Beclin1/-Bcl-2 or IgG. The resulting immune complexes and inputs were analyzed by western blotting as indicated. Representative western blotting analysis of protein levels is presented. (E) RAGE/Beclin1 but not TLR4 is required for HMGB1 mediated autophagy. Cells were transfected with the indicated shRNA for 48 h and then were treated with reduced HMGB1 (“R”, 10 μg/ml) for 24 h. Representative western blotting analysis of protein levels is presented. In parallel, autophagy was assayed by the percentage of cells with GFP-LC3 dots (N=3, * p

Figure 6. Redox of HMGB1 regulates chemotherapy…

Figure 6. Redox of HMGB1 regulates chemotherapy effectiveness

(A) Cell viability and apoptosis assay. Panc2.03…

Figure 6. Redox of HMGB1 regulates chemotherapy effectiveness
(A) Cell viability and apoptosis assay. Panc2.03 and HCT116 cells were treated with oxaliplatin (160 μg/ml), melphalan (320 μg/ml), adriamycin (1.6 μg/ml), paclitaxel (20 μg/ml) with or without oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml). Cell death was analysis at indicated time by CCK-8 cell viability assay (n=3, * and #, p
Similar articles
Cited by
References
    1. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 2007;13:7271–9. - PubMed
    1. Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68:1485–94. - PubMed
    1. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. - PubMed
    1. Azad MB, Chen Y, Gibson SB. Regulation of autophagy by reactive oxygen species (ROS): implications for cancer progression and treatment. Antioxid Redox Signal. 2009;11:777–90. - PubMed
    1. Bell CW, Jiang W, Reich CF, 3rd, Pisetsky DS. The extracellular release of HMGB1 during apoptotic cell death. Am J Physiol Cell Physiol. 2006;291:C1318–25. - PubMed
Show all 49 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2. Inhibition of autophagy diminishes HMGB1…
Figure 2. Inhibition of autophagy diminishes HMGB1 release and enhances selective apoptosis
(A) Immunoblots are shown for Beclin1 and ATG5 knockdown performed in Panc02 cells. (B–D) Panc02 cells as indicated were treated with the anticancer agents (melphalan, “ME”, 160 μg/ml; paclitaxel, “PA”, 10 μg/ml) for 6 h. and then assayed for early apoptosis (annexin V+/PI−) by flow cytometry (B), autophagy by quantification of the percentage of cells with GFP-LC3 punctae (C) and HMGB1 release, by western blotting analysis (LDH and H3 were both used as controls for protein leakage from damaged cells) (D). PI3-kinase inhibitor 3-methyladenine (3MA, 5mm) was used as a nominal autophagy inhibitor. Representative western blots of the indicated proteins are presented.
Figure 3. HMGB1 release and autophagy is…
Figure 3. HMGB1 release and autophagy is detected in the absence of measurable apoptosis
(A) Immunoblots are shown for Bax and p53 knockout in HCT116 cells. (B-D) WT, Bax knock out, p53 knockout or pan-caspase inhibitor treated (ZVAD-FMK, 20 μm) HCT116 cells were treated with melphalan, “ME”, 160 μg/ml or paclitaxel, “PA”, 10 μg/ml for 6 h. and then assayed for measures of early apoptosis (annexin V+/PI−) by flow cytometry (B), autophagy by quantification of the percentage of cells with GFP-LC3 punctae (C) and HMGB1 release by western blot analysis (LDH and H3 were both used as controls for protein leakage from damaged cells) (D). Representative western blots of the indicated proteins are presented.
Figure 4. Inhibition of HMGB1 release increases…
Figure 4. Inhibition of HMGB1 release increases tumor cell sensitivity to anticancer agents
(A) Inhibition of HMGB1 release with small molecule drugs increases tumor cell sensitivity to anticancer agents. Panc2.03 and HCT116 cells were pretreated with the HMGB1-release inhibitors ethyl pyruvate (EP, 10 mm) or glycyrrhizin (Gly, 500 μm) for 2 h and then cultured in the presence of melphalan for an additional 24 h. Representative western blotting analysis of protein levels are presented. In parallel, measures of apoptosis (annexin V+/PI−) were assayed by flow cytometry and autophagy by quantifying the percentage of cells with GFP-LC3 punctae. (B) Panc2.03 and HCT116 cells were knocked down for HMGB1 using shRNA for 48 h, and then stimulated with melphalan for 24 h. Representative western blotting analysis of protein levels is presented. In parallel, apoptosis (annexin V+/PI−) was assayed by flow cytometry (right panel) and autophagy by quantifying the percentage of cells with GFP-LC3 punctae (N=3, p

Figure 5. Provision of exogenous reduced HMGB1…

Figure 5. Provision of exogenous reduced HMGB1 increases autophagy in cancer cells

(A) Relative amounts…

Figure 5. Provision of exogenous reduced HMGB1 increases autophagy in cancer cells
(A) Relative amounts of oxidized Cys106 (as Cys sulfonic acid) in Lilly Pool #2 and #3. MALDI-TOF Mass Spectrum of tryptic fragments of Lilly Pool #2 (top) and Pool #3 (bottom). The Cys106 containing fragment is amino acids 97–112. The free sulfhydryl (-SH) of total reducible cysteine is at a mass of 1944.9 Da. The monoxide is faintly seen at a mass of 1960.9 Da. The di- and tri- oxides are at masses of 1976.9 Da and 1992.9 Da, respectively. The peak at 1962.9 Da is the free sulfhydryl of the 13–28 fragments, used as an internal standard to verify the DTT reduction went to completion. (B) Reduced HMGB1 protein induces autophagy and oxidized HMGB1 mildly induces apoptosis. Panc2.03 and HCT116 cancer cells were treated with oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml) for 24 h, and then assayed for apoptosis by FACS using Annexin V/PI stain and autophagy by quantification of the percentage of cells with GFP-LC3 dots as described in methods. (C) Western analysis of LC3 processing in the presence or absence of lysosomal protease inhibitors pepstatin A (10 μg/ml) and E64D (10 μg/ml) and degradation of p62 by autophagy after HMGB1 or HMGB1 C106S mutant treatment. (D) Reduced HMGB1 protein regulates Beclin1/Bcl-2 complex formation in autophagy. Panc2.03 cells were treated with oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml), for 6 h, then cell lysates were prepared for IP with anti-Beclin1/-Bcl-2 or IgG. The resulting immune complexes and inputs were analyzed by western blotting as indicated. Representative western blotting analysis of protein levels is presented. (E) RAGE/Beclin1 but not TLR4 is required for HMGB1 mediated autophagy. Cells were transfected with the indicated shRNA for 48 h and then were treated with reduced HMGB1 (“R”, 10 μg/ml) for 24 h. Representative western blotting analysis of protein levels is presented. In parallel, autophagy was assayed by the percentage of cells with GFP-LC3 dots (N=3, * p

Figure 6. Redox of HMGB1 regulates chemotherapy…

Figure 6. Redox of HMGB1 regulates chemotherapy effectiveness

(A) Cell viability and apoptosis assay. Panc2.03…

Figure 6. Redox of HMGB1 regulates chemotherapy effectiveness
(A) Cell viability and apoptosis assay. Panc2.03 and HCT116 cells were treated with oxaliplatin (160 μg/ml), melphalan (320 μg/ml), adriamycin (1.6 μg/ml), paclitaxel (20 μg/ml) with or without oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml). Cell death was analysis at indicated time by CCK-8 cell viability assay (n=3, * and #, p
Similar articles
Cited by
References
    1. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 2007;13:7271–9. - PubMed
    1. Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68:1485–94. - PubMed
    1. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. - PubMed
    1. Azad MB, Chen Y, Gibson SB. Regulation of autophagy by reactive oxygen species (ROS): implications for cancer progression and treatment. Antioxid Redox Signal. 2009;11:777–90. - PubMed
    1. Bell CW, Jiang W, Reich CF, 3rd, Pisetsky DS. The extracellular release of HMGB1 during apoptotic cell death. Am J Physiol Cell Physiol. 2006;291:C1318–25. - PubMed
Show all 49 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 5. Provision of exogenous reduced HMGB1…
Figure 5. Provision of exogenous reduced HMGB1 increases autophagy in cancer cells
(A) Relative amounts of oxidized Cys106 (as Cys sulfonic acid) in Lilly Pool #2 and #3. MALDI-TOF Mass Spectrum of tryptic fragments of Lilly Pool #2 (top) and Pool #3 (bottom). The Cys106 containing fragment is amino acids 97–112. The free sulfhydryl (-SH) of total reducible cysteine is at a mass of 1944.9 Da. The monoxide is faintly seen at a mass of 1960.9 Da. The di- and tri- oxides are at masses of 1976.9 Da and 1992.9 Da, respectively. The peak at 1962.9 Da is the free sulfhydryl of the 13–28 fragments, used as an internal standard to verify the DTT reduction went to completion. (B) Reduced HMGB1 protein induces autophagy and oxidized HMGB1 mildly induces apoptosis. Panc2.03 and HCT116 cancer cells were treated with oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml) for 24 h, and then assayed for apoptosis by FACS using Annexin V/PI stain and autophagy by quantification of the percentage of cells with GFP-LC3 dots as described in methods. (C) Western analysis of LC3 processing in the presence or absence of lysosomal protease inhibitors pepstatin A (10 μg/ml) and E64D (10 μg/ml) and degradation of p62 by autophagy after HMGB1 or HMGB1 C106S mutant treatment. (D) Reduced HMGB1 protein regulates Beclin1/Bcl-2 complex formation in autophagy. Panc2.03 cells were treated with oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml), for 6 h, then cell lysates were prepared for IP with anti-Beclin1/-Bcl-2 or IgG. The resulting immune complexes and inputs were analyzed by western blotting as indicated. Representative western blotting analysis of protein levels is presented. (E) RAGE/Beclin1 but not TLR4 is required for HMGB1 mediated autophagy. Cells were transfected with the indicated shRNA for 48 h and then were treated with reduced HMGB1 (“R”, 10 μg/ml) for 24 h. Representative western blotting analysis of protein levels is presented. In parallel, autophagy was assayed by the percentage of cells with GFP-LC3 dots (N=3, * p

Figure 6. Redox of HMGB1 regulates chemotherapy…

Figure 6. Redox of HMGB1 regulates chemotherapy effectiveness

(A) Cell viability and apoptosis assay. Panc2.03…

Figure 6. Redox of HMGB1 regulates chemotherapy effectiveness
(A) Cell viability and apoptosis assay. Panc2.03 and HCT116 cells were treated with oxaliplatin (160 μg/ml), melphalan (320 μg/ml), adriamycin (1.6 μg/ml), paclitaxel (20 μg/ml) with or without oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml). Cell death was analysis at indicated time by CCK-8 cell viability assay (n=3, * and #, p
Similar articles
Cited by
References
    1. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 2007;13:7271–9. - PubMed
    1. Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68:1485–94. - PubMed
    1. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. - PubMed
    1. Azad MB, Chen Y, Gibson SB. Regulation of autophagy by reactive oxygen species (ROS): implications for cancer progression and treatment. Antioxid Redox Signal. 2009;11:777–90. - PubMed
    1. Bell CW, Jiang W, Reich CF, 3rd, Pisetsky DS. The extracellular release of HMGB1 during apoptotic cell death. Am J Physiol Cell Physiol. 2006;291:C1318–25. - PubMed
Show all 49 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6. Redox of HMGB1 regulates chemotherapy…
Figure 6. Redox of HMGB1 regulates chemotherapy effectiveness
(A) Cell viability and apoptosis assay. Panc2.03 and HCT116 cells were treated with oxaliplatin (160 μg/ml), melphalan (320 μg/ml), adriamycin (1.6 μg/ml), paclitaxel (20 μg/ml) with or without oxidized HMGB1 (“O”, 10 μg/ml) or reduced HMGB1 (“R”, 10 μg/ml). Cell death was analysis at indicated time by CCK-8 cell viability assay (n=3, * and #, p

References

    1. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 2007;13:7271–9.
    1. Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68:1485–94.
    1. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9.
    1. Azad MB, Chen Y, Gibson SB. Regulation of autophagy by reactive oxygen species (ROS): implications for cancer progression and treatment. Antioxid Redox Signal. 2009;11:777–90.
    1. Bell CW, Jiang W, Reich CF, 3rd, Pisetsky DS. The extracellular release of HMGB1 during apoptotic cell death. Am J Physiol Cell Physiol. 2006;291:C1318–25.
    1. Bierhaus A, Schiekofer S, Schwaninger M, Andrassy M, Humpert PM, Chen J, et al. Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB. Diabetes. 2001;50:2792–808.
    1. Choi JJ, Reich CF, 3rd, Pisetsky DS. Release of DNA from dead and dying lymphocyte and monocyte cell lines in vitro. Scand J Immunol. 2004;60:159–66.
    1. Criollo A, Senovilla L, Authier H, Maiuri MC, Morselli E, Vitale I, et al. The IKK complex contributes to the induction of autophagy. Embo J. 29:619–31.
    1. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 2006;10:51–64.
    1. Geft D, Schwartzenberg S, Rogowsky O, Finkelstein A, Ablin J, Maysel-Auslender S, et al. Circulating apoptotic progenitor cells in patients with congestive heart failure. PLoS ONE. 2008;3:e3238.
    1. Hamamoto R, Furukawa Y, Morita M, Iimura Y, Silva FP, Li M, et al. SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells. Nat Cell Biol. 2004;6:731–40.
    1. Han J, Hou W, Goldstein LA, Lu C, Stolz DB, Yin XM, et al. Involvement of protective autophagy in TRAIL resistance of apoptosis-defective tumor cells. J Biol Chem. 2008;283:19665–77.
    1. Hoppe G, Talcott KE, Bhattacharya SK, Crabb JW, Sears JE. Molecular basis for the redox control of nuclear transport of the structural chromatin protein Hmgb1. Exp Cell Res. 2006;312:3526–38.
    1. Ito N, Demarco RA, Mailliard RB, Han J, Rabinowich H, Kalinski P, et al. Cytolytic cells induce HMGB1 release from melanoma cell lines. J Leukoc Biol. 2007;81:75–83.
    1. Kang R, Tang D, Schapiro NE, Livesey KM, Farkas A, Loughran P, et al. The receptor for advanced glycation end products (RAGE) sustains autophagy and limits apoptosis, promoting pancreatic tumor cell survival. Cell Death Differ. 2010a;17:666–76.
    1. Kang R, Tang D, Yu Y, Wang Z, Hu T, Wang H, et al. WAVE1 regulates Bcl-2 localization and phosphorylation in leukemia cells. Leukemia. 2010b;24:177–86.
    1. Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR, Ferguson TA. Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity. 2008;29:21–32.
    1. Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell Biol. 2008;9:1004–10.
    1. Levine B. Cell biology: autophagy and cancer. Nature. 2007;446:745–7.
    1. Liliensiek B, Weigand MA, Bierhaus A, Nicklas W, Kasper M, Hofer S, et al. Receptor for advanced glycation end products (RAGE) regulates sepsis but not the adaptive immune response. J Clin Invest. 2004;113:1641–50.
    1. Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol. 2005;5:331–42.
    1. Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, et al. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120:237–48.
    1. Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 2007;8:741–52.
    1. Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, et al. Autophagy suppresses tumorigenesis through elimination of p62. Cell. 2009;137:1062–75.
    1. Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting. Autophagy. 2007;3:542–5.
    1. Mollica L, De Marchis F, Spitaleri A, Dallacosta C, Pennacchini D, Zamai M, et al. Glycyrrhizin binds to high-mobility group box 1 protein and inhibits its cytokine activities. Chem Biol. 2007;14:431–41.
    1. Muller S, Scaffidi P, Degryse B, Bonaldi T, Ronfani L, Agresti A, et al. New EMBO members’ review: the double life of HMGB1 chromatin protein: architectural factor and extracellular signal. Embo J. 2001;20:4337–40.
    1. Ohndorf UM, Rould MA, He Q, Pabo CO, Lippard SJ. Basis for recognition of cisplatin-modified DNA by high-mobility-group proteins. Nature. 1999;399:708–12.
    1. Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 2007;282:24131–45.
    1. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, et al. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122:927–39.
    1. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–5.
    1. Sparvero LJ, Asafu-Adjei D, Kang R, Tang D, Amin N, Im J, et al. RAGE (Receptor for Advanced Glycation Endproducts), RAGE ligands, and their role in cancer and inflammation. J Transl Med. 2009;7:17.
    1. Sy LK, Yan SC, Lok CN, Man RY, Che CM. Timosaponin A-III induces autophagy preceding mitochondria-mediated apoptosis in HeLa cancer cells. Cancer Res. 2008;68:10229–37.
    1. Tang D, Kang R, Cao L, Zhang G, Yu Y, Xiao W, et al. A pilot study to detect high mobility group box 1 and heat shock protein 72 in cerebrospinal fluid of pediatric patients with meningitis. Crit Care Med. 2008;36:291–295.
    1. Tang D, Kang R, Xiao W, Jiang L, Liu M, Shi Y, et al. Nuclear Heat Shock Protein 72 as a Negative Regulator of Oxidative Stress (Hydrogen Peroxide)-Induced HMGB1 Cytoplasmic Translocation and Release. J Immunol. 2007a;178:7376–7384.
    1. Tang D, Kang R, Xiao W, Wang H, Calderwood SK, Xiao X. 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. 2007b;179:1236–1244.
    1. Tang D, Kang R, Xiao W, Zhang H, Lotze MT, Wang H, et al. Quercetin prevents LPS-induced high-mobility group box 1 release and proinflammatory function. Am J Respir Cell Mol Biol. 2009;41:651–60.
    1. Tang D, Kang R, Zeh HJ, 3rd, Lotze MT. High-mobility group box 1 and cancer. Biochim Biophys Acta. 2010;1799:131–140.
    1. Tang D, Shi Y, Jang L, Wang K, Xiao W, Xiao X. Heat shock response inhibits release of high mobility group box 1 protein induced by endotoxin in murine macrophages. Shock. 2005;23:434–40.
    1. Tang D, Shi Y, Kang R, Li T, Xiao W, Wang H, et al. Hydrogen peroxide stimulates macrophages and monocytes to actively release HMGB1. J Leukoc Biol. 2007c;81:741–7.
    1. Tesniere A, Panaretakis T, Kepp O, Apetoh L, Ghiringhelli F, Zitvogel L, et al. Molecular characteristics of immunogenic cancer cell death. Cell Death Differ. 2008;15:3–12.
    1. Thorburn J, Horita H, Redzic J, Hansen K, Frankel AE, Thorburn A. Autophagy regulates selective HMGB1 release in tumor cells that are destined to die. Cell Death Differ. 2009;16:175–83.
    1. Tian J, Avalos AM, Mao SY, Chen B, Senthil K, Wu H, et al. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat Immunol. 2007;8:487–96.
    1. Ulloa L, Ochani M, Yang H, Tanovic M, Halperin D, Yang R, et al. Ethyl pyruvate prevents lethality in mice with established lethal sepsis and systemic inflammation. Proc Natl Acad Sci U S A. 2002;99:12351–6.
    1. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science. 1999;285:248–51.
    1. Wang P, Yu J, Zhang L. The nuclear function of p53 is required for PUMA-mediated apoptosis induced by DNA damage. Proc Natl Acad Sci U S A. 2007;104:4054–9.
    1. White E, DiPaola RS. The double-edged sword of autophagy modulation in cancer. Clin Cancer Res. 2009;15:5308–16.
    1. Yousefi S, Perozzo R, Schmid I, Ziemiecki A, Schaffner T, Scapozza L, et al. Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol. 2006;8:1124–32.
    1. Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B. Role of BAX in the apoptotic response to anticancer agents. Science. 2000;290:989–92.

Source: PubMed

Подписаться