1
|
Ding Q, Rha H, Yoon C, Kim Y, Hong SJ, Kim HJ, Li Y, Lee MH, Kim JS. Regulated cell death mechanisms in mitochondria-targeted phototherapy. J Control Release 2025; 382:113720. [PMID: 40228665 DOI: 10.1016/j.jconrel.2025.113720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Phototherapy, comprising photodynamic therapy (PDT) and photothermal therapy (PTT), was first introduced over a century ago and has since evolved into a versatile cancer treatment modality. While numerous studies have explored regulated cell death (RCD) mechanisms induced by phototherapy, a comprehensive synthesis centered on mitochondria-targeted phototherapeutic strategies and agents as mediators of RCD is still lacking. This review provides a systematic and in-depth analysis of recent advances in mitochondria-centered mechanisms driving phototherapy-induced death pathways, including apoptosis, autophagy, pyroptosis, immunogenic cell death, ferroptosis, and cuproptosis. We highlight the critical role of mitochondria as central regulators of these death pathways in response to phototherapeutic interventions. Moreover, we discuss fundamental design strategies for developing precision-targeted phototherapeutic materials to enhance efficacy and minimize off-target effects. Finally, we identify prevailing challenges and propose future research directions to address these hurdles, paving the way for next-generation mitochondria-targeted phototherapy as a highly effective strategy for cancer management.
Collapse
Affiliation(s)
- Qihang Ding
- School of Chemical Engineering & Pharmacy, Pharmaceutical Research Institute, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Wuhan Institute of Technology, Wuhan 430205, China; Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Hyeonji Rha
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Changyu Yoon
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Yujin Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - So Jin Hong
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hui Ju Kim
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yang Li
- School of Chemical Engineering & Pharmacy, Pharmaceutical Research Institute, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Min Hee Lee
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
2
|
Murtazina A, Jimenez-Martinez Y, Ruiz Alcala G, Marchal JA, Tarabayeva A, Bitanova E, Rakhimbayev I, McDougall GJ, Bishimbayeva N, Boulaiz H. In Vitro Inhibition of Colon Cancer Stem Cells by Natural Polysaccharides Obtained from Wheat Cell Culture. Polymers (Basel) 2025; 17:1048. [PMID: 40284312 PMCID: PMC12030112 DOI: 10.3390/polym17081048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Natural polysaccharides (PSs) have shown inhibitory effects on differentiated cancer cells (DCCs), but their activity against cancer stem cells (CSCs) remains poorly understood. Here, we report that PSs from wheat cell cultures (WCCPSs) inhibit the proliferation of both DCCs and CSCs derived from HCT-116 colorectal cancer cells. Among them, NA and DC fractions showed the strongest anti-CSC activity. NA, rich in xylose, was effective at lower concentrations, while DC, enriched in xylose and galacturonic acid (GalUA), exhibited higher potency, with a lower IC50 and preferential activity against CSCs at higher doses. WCCPSs reduced β-catenin levels, and some fractions also downregulated Ep-CAM, CD44, and c-Myc. Notably, DC increased caspase-3 without inducing cytochrome C and caspase-8 overexpression, suggesting a mechanism promoting CSC differentiation rather than apoptosis. Correlation analysis linked xylose content to reduced c-Myc expression, and GalUA levels to increased caspase-3. These results suggest that WCCPS bioactivity may be related to their monosaccharide composition. Overall, our findings support the potential of wheat-derived PSs as CSC-targeting agents that suppress self-renewal and promote differentiation, offering a promising approach to reduce tumor aggressiveness and recurrence.
Collapse
Affiliation(s)
- Alima Murtazina
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
| | - Yaiza Jimenez-Martinez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
| | - Gloria Ruiz Alcala
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
| | - Anel Tarabayeva
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
| | - Elmira Bitanova
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
| | | | - Gordon J. McDougall
- Plant Biochemistry and Food Quality Group, Environmental and Biochemical Sciences Department, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, UK;
| | - Nazira Bishimbayeva
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
- Research Institute for Problems of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Houria Boulaiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| |
Collapse
|
3
|
Tetramethylpyrazine Protects against Early Brain Injury after Experimental Subarachnoid Hemorrhage by Affecting Mitochondrial-Dependent Caspase-3 Apoptotic Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:3514914. [PMID: 28337226 PMCID: PMC5350396 DOI: 10.1155/2017/3514914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/27/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022]
Abstract
This study was to test the hypothesis that tetramethylpyrazine (TMP) protected against early brain injury after subarachnoid hemorrhage (SAH) by affecting the mitochondrial-dependent caspase-3 apoptotic pathway. TMP was administrated after the rats' prechiasmatic SAH mode. Animal neurobehavioral functions were assessed and the mitochondrial morphology, mitochondrial and cytoplasmic calcium, and mitochondrial membrane potential changes (Δψm) of the brain tissues were measured. The expressions of cytoplasmic cytochrome c (cyt c), second mitochondria-derived activator of caspases (Smac), and cleaved caspase-3 B-cell lymphoma 2 (bcl-2) in cells were determined and cellular apoptosis was detected. The treatment of TMP resulted in less apoptotic cells and milder mitochondrial injury and potentially performed better in the neurobehavioral outcome compared to those with saline. Also, TMP ameliorated calcium overload in mitochondria and cytoplasm and alleviated the decrease of Δψm. In addition, TMP inhibited the expression of cytoplasmic cyt c, Smac, and cleaved caspase-3, yet it upregulated the expression of bcl-2. These findings suggest that TMP exerts an antiapoptosis property in the SAH rat model and this is probably mediated by the caspase-3 apoptotic pathway triggered by mitochondrial calcium overload. The finding offers a new therapeutic candidate for early brain injury after SAH.
Collapse
|
4
|
Cui L, Song J, Wu L, Huang L, Wang Y, Huang Y, Yu H, Huang Y, You C, Ye J. Smac is another pathway in the anti-tumour activity of Trichosanthin and reverses Trichosanthin resistance in CaSki cervical cancer cells. Biomed Pharmacother 2015; 69:119-24. [DOI: 10.1016/j.biopha.2014.10.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/27/2014] [Indexed: 10/24/2022] Open
|
5
|
Ramachandiran S, Cain J, Liao A, He Y, Guo X, Boise LH, Fu H, Ratner L, Khoury HJ, Bernal-Mizrachi L. The Smac mimetic RMT5265.2HCL induces apoptosis in EBV and HTLV-I associated lymphoma cells by inhibiting XIAP and promoting the mitochondrial release of cytochrome C and Smac. Leuk Res 2012; 36:784-90. [PMID: 22325366 PMCID: PMC3331941 DOI: 10.1016/j.leukres.2011.12.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/29/2011] [Accepted: 12/30/2011] [Indexed: 12/14/2022]
Abstract
The inhibitors of apoptosis (IAP) are important regulators of apoptosis. However, little is known about the capacity of Smac mimetics (IAP inhibitor) to overcome virally associated-lymphoma's (VAL) resistance to apoptosis. Here, we explored the pro-apoptotic effect of a novel Smac mimetic, RMT5265.2HCL (RMT) in VAL cells. RMT improved the sensitivity to apoptosis in EBV- and to some extend in HTLV-1- but not in HHV-8-VAL. Furthermore, we identified that RMT promotes caspase 3 and 9 cleavage by inhibiting XIAP and inducing the mitochondrial efflux of Smac and cytochrome C. This investigation further support exploring the use of Smac inhibitors in VAL.
Collapse
Affiliation(s)
- Sampath Ramachandiran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Joan Cain
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Albert Liao
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Yanjuan He
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Xiangxue Guo
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Haian Fu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Pharmacology, Emory University, Atlanta, GA, USA
| | - Lee Ratner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hanna Jean Khoury
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Leon Bernal-Mizrachi
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
6
|
Le Quement ST, Ishoey M, Petersen MT, Thastrup J, Hagel G, Nielsen TE. Solid-phase synthesis of smac peptidomimetics incorporating triazoloprolines and biarylalanines. ACS COMBINATORIAL SCIENCE 2011; 13:667-75. [PMID: 21905744 DOI: 10.1021/co200078u] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Apoptotic induction mechanisms are of crucial importance for the general homeostasis of multicellular organisms. In cancer the apoptotic pathways are downregulated, which, at least partly, is due to an abundance of inhibitors of apoptosis proteins (IAPs) that block the apoptotic cascade by deactivating proteolytic caspases. The Smac protein has an antagonistic effect on IAPs, thus providing structural clues for the synthesis of new pro-apoptotic compounds. Herein, we report a solid-phase approach for the synthesis of Smac-derived tetrapeptide libraries. On the basis of a common (N-Me)AVPF sequence, peptides incorporating triazoloprolines and biarylalanines were synthesized by means of Cu(I)-catalyzed azide-alkyne cycloaddition and Pd-catalyzed Suzuki cross-coupling reactions. Solid-phase procedures were optimized to high efficiency, thus accessing all products in excellent crude purities and yields (both typically above 90%). The peptides were subjected to biological evaluation in a live/dead cellular assay which revealed that structural decorations on the AVPF sequence indeed are highly important for cytotoxicity toward HeLa cells.
Collapse
Affiliation(s)
| | - Mette Ishoey
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Mette T. Petersen
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Jacob Thastrup
- 2CureX, Department of Surgery, K, Bispebjerg Hospital, Bispebjerg Bakke 23, DK-2400 Copenhagen NV, Denmark
| | - Grith Hagel
- 2CureX, Department of Surgery, K, Bispebjerg Hospital, Bispebjerg Bakke 23, DK-2400 Copenhagen NV, Denmark
| | - Thomas E. Nielsen
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
7
|
Hu W, Wang F, Tang J, Liu X, Yuan Z, Nie C, Wei Y. Proapoptotic protein Smac mediates apoptosis in cisplatin-resistant ovarian cancer cells when treated with the anti-tumor agent AT101. J Biol Chem 2011; 287:68-80. [PMID: 22052903 DOI: 10.1074/jbc.m111.271205] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Chemoresistance of ovarian cancer has been previously attributed to the expression and activation of Bcl-2 family proteins. BH3-mimetic molecules possessing potential anticancer activity are able to inhibit antiapoptotic Bcl-2 family proteins. AT101 (R-(-)-gossypol), a natural BH3-mimetic molecule, has shown anti-tumor activity as a single agent and in combination with standard anticancer therapies in a variety of tumor models. Here, we report the effect of AT101 on apoptosis in cisplatin-resistant ovarian cancer cells and identify the major molecular events that determine sensitivity. AT101 induced cell apoptosis by activating Bax through a conformational change, translocation, and oligomerization. The inhibition of Bax expression only partially prevented caspase-3 cleavage. However, the gene silencing of Bax had no effect on mitochondrial Smac release. Further experiments demonstrated that Smac reduction inhibited caspase-3 activation and attenuated cell apoptosis. More importantly, the inhibition of Smac or overexpression of XIAP attenuated Bax activation in ovarian cells. Furthermore, our data indicate that the Akt-p53 pathway is involved in the regulation of Smac release. Taken together, our data demonstrate the role of Smac and the molecular mechanisms of AT101-induced apoptosis of chemoresistant ovarian cancer cells. Our findings suggest that AT101 not only triggers Bax activation but also induces mitochondrial Smac release. Activated Smac can enhance Bax-mediated cellular apoptosis. Therefore, Smac mediates Bax activation to determine the threshold for overcoming cisplatin resistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Wenbin Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 4 Keyuan Road, Hitech District, Chengdu 610041, China
| | - Fang Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 4 Keyuan Road, Hitech District, Chengdu 610041, China
| | - Jingsheng Tang
- ChongQing NewFine Biology Technology Co., Ltd., 8 Lingfang Road, Banqiao District, Chongqing 402460, China
| | - Xinyu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 4 Keyuan Road, Hitech District, Chengdu 610041, China
| | - Zhu Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 4 Keyuan Road, Hitech District, Chengdu 610041, China
| | - Chunlai Nie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 4 Keyuan Road, Hitech District, Chengdu 610041, China.
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, 4 Keyuan Road, Hitech District, Chengdu 610041, China
| |
Collapse
|
8
|
Abstract
Apoptosis is a critical process for the maintenance of tissue homeostasis and prevention of tumorigenesis. The members of the Bcl-2 family of proteins are the central regulators of the intrinsic apoptotic pathway. Within the Bcl-2 family, the BH3-only subfamily of proteins is tasked with sensing a broad range of apoptotic stimuli and transmitting this signal to other Bcl-2 proteins to initiate programmed cell death. This family of proteins is highly regulated at both transcriptional and post-translational levels, as well as by prominent protein-protein interactions among the family members. Bcl-2 family proteins are often deregulated in cancer, with overexpression of antiapoptotic members, as well as mutations or defects in proapoptotic members. These proteins have been the subject of intensive study for many years and the complex relationships between their regulation and tumorigenesis have spawned a new thinking about cancer treatment. New generations of small molecule Bcl-2 family inhibitors and BH3 and SMAC mimetics have brought new optimism to the pursuit of more individualized and effective cancer therapies.
Collapse
Affiliation(s)
- Brian Leibowitz
- University of Pittsburgh Cancer Institute; University of Pittsburgh; and Department of Pathology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA
| | - Jian Yu
- University of Pittsburgh Cancer Institute; University of Pittsburgh; and Department of Pathology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA
| |
Collapse
|
9
|
Bank A, Wang P, Du C, Yu J, Zhang L. SMAC mimetics sensitize nonsteroidal anti-inflammatory drug-induced apoptosis by promoting caspase-3-mediated cytochrome c release. Cancer Res 2008; 68:276-84. [PMID: 18172320 DOI: 10.1158/0008-5472.can-07-5242] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAID) are effective in suppressing the formation of colorectal tumors. However, the mechanisms underlying the antineoplastic effects of NSAIDs remain unclear. The effects of NSAIDs are incomplete, and resistance to NSAIDs is often developed. Growing evidence has indicated that the chemopreventive activity of NSAIDs is mediated by induction of apoptosis. Our previous studies showed that second mitochondria-derived activator of caspase (SMAC)/Diablo, a mitochondrial apoptogenic protein, plays an essential role in NSAID-induced apoptosis in colon cancer cells. In this study, we found that SMAC mediates NSAID-induced apoptosis through a feedback amplification mechanism involving interactions with inhibitor of apoptosis proteins, activation of caspase-3, and induction of cytosolic release of cytochrome c. Small-molecule SMAC mimetics at nanomolar concentrations significantly sensitize colon cancer cells to NSAID-induced apoptosis by promoting caspase-3 activation and cytochrome c release. Furthermore, SMAC mimetics overcome NSAID resistance in Bax-deficient or SMAC-deficient colon cancer cells by restoring caspase-3 activation and cytochrome c release. Together, these results suggest that SMAC is useful as a target for the development of more effective chemopreventive strategies and agents.
Collapse
Affiliation(s)
- Alexander Bank
- Department of Pharmacology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | |
Collapse
|
10
|
Meng XW, Lee SH, Dai H, Loegering D, Yu C, Flatten K, Schneider P, Dai NT, Kumar SK, Smith BD, Karp JE, Adjei AA, Kaufmann SH. MCL-1 as a Buffer for Proapoptotic BCL-2 Family Members during TRAIL-induced Apoptosis. J Biol Chem 2007; 282:29831-46. [PMID: 17698840 DOI: 10.1074/jbc.m706110200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies have suggested that Mcl-1, an antiapoptotic Bcl-2 homolog that does not exhibit appreciable affinity for the caspase 8-generated C-terminal Bid fragment (tBid), diminishes sensitivity to tumor necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL). This study was performed to determine the mechanism by which Mcl-1 confers TRAIL resistance and to evaluate methods for overcoming this resistance. Affinity purification/immunoblotting assays using K562 human leukemia cells, which contain Mcl-1 and Bcl-x(L) as the predominant antiapoptotic Bcl-2 homologs, demonstrated that TRAIL treatment resulted in binding of tBid to Bcl-x(L) but not Mcl-1. In contrast, TRAIL caused increased binding between Mcl-1 and Bak that was diminished by treatment with the caspase 8 inhibitor N-(N(alpha)-acetylisoleucylglutamylthreonyl) aspartic acid (O-methyl ester)-fluoromethyl ketone (IETD(OMe)-fmk) or the c-Jun N-terminal kinase inhibitor SP600125. In addition, TRAIL caused increased binding of Bim and Puma to Mcl-1 that was inhibited by IETD(OMe)-fmk but not SP600125. Further experiments demonstrated that down-regulation of Mcl-1 by short hairpin RNA or the kinase inhibitor sorafenib increased TRAIL-induced Bak activation and death ligand-induced apoptosis in a wide variety of neoplastic cell lines as well as clinical acute myelogenous leukemia specimens. Collectively, these observations not only suggest a model in which Mcl-1 confers TRAIL resistance by serving as a buffer for Bak, Bim, and Puma, but also identify sorafenib as a potential modulator of TRAIL sensitivity.
Collapse
Affiliation(s)
- Xue Wei Meng
- Divisions of Oncology Research, Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Yu J, Wang P, Ming L, Wood MA, Zhang L. SMAC/Diablo mediates the proapoptotic function of PUMA by regulating PUMA-induced mitochondrial events. Oncogene 2007; 26:4189-98. [PMID: 17237824 DOI: 10.1038/sj.onc.1210196] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
p53-upregulated modulator of apoptosis (PUMA) is a BH3-only Bcl-2 family protein and an essential mediator of DNA damage-induced apoptosis. PUMA is localized in the mitochondria and induces apoptosis through the mitochondrial pathway. However, the mechanisms of PUMA-induced apoptosis remain unclear. In this study, we found that second mitochondria-derived activator of caspase (SMAC)/Diablo, a mitochondrial apoptogenic protein, mediates the proapoptotic function of PUMA by regulating PUMA-induced mitochondrial events. SMAC is consistently released into the cytosol in colon cancer cells undergoing PUMA-induced apoptosis. In SMAC-deficient cells, execution of PUMA-induced apoptosis is abrogated, in company with decreases in caspase activation, cytosolic release of cytochrome c and collapse of mitochondrial membrane potential. Reconstituting SMAC expression restored these events in the SMAC-deficient cells. Furthermore, SMAC and agents that mimic the inhibitor of apoptosis proteins (IAPs) inhibition function of SMAC significantly sensitize cells to PUMA-induced apoptosis. These results demonstrate an important role of SMAC in executing DNA damage-induced and PUMA-mediated apoptosis and suggest that SMAC participates in a feedback amplification loop to promote cytochrome c release and other mitochondrial events in apoptosis.
Collapse
Affiliation(s)
- J Yu
- Department of Pharmacology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
12
|
Meng XW, Lee SH, Kaufmann SH. Apoptosis in the treatment of cancer: a promise kept? Curr Opin Cell Biol 2006; 18:668-76. [PMID: 17049222 DOI: 10.1016/j.ceb.2006.10.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Accepted: 10/03/2006] [Indexed: 10/24/2022]
Abstract
A common feature of cancer cells is their ability to evade apoptosis as a result of alterations that block cell death signaling pathways. The extensive research efforts that elucidated these signaling pathways over the past decade have set the stage for the development of therapeutic agents that either kill cancer cells selectively or reset their apoptotic threshold. Over the past two years a number of these agents have been evaluated in preclinical and clinical trials. The results of these studies suggest that it might soon be possible to modulate apoptosis in cancer cells for therapeutic benefit.
Collapse
Affiliation(s)
- Xue Wei Meng
- Division of Oncology Research, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | |
Collapse
|
13
|
Le SB, Holmuhamedov EL, Narayanan VL, Sausville EA, Kaufmann SH. Adaphostin and other anticancer drugs quench the fluorescence of mitochondrial potential probes. Cell Death Differ 2006; 13:151-9. [PMID: 16052237 DOI: 10.1038/sj.cdd.4401732] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Fluorescent dyes are widely used to monitor changes in mitochondrial transmembrane potential (DeltaPsim). When MitoTracker Red CMXRos, tetramethylrhodamine methyl ester (TMRM), and 3,3'dihexyloxacarbocyanine iodide (DiOC6(3)) were utilized to examine the effects of the experimental anticancer drug adaphostin on intact cells or isolated mitochondria, decreased fluorescence was observed. In contrast, measurement of tetraphenylphosphonium uptake by the mitochondria using an ion-selective microelectrode failed to show any effect of adaphostin on DeltaPsim. Instead, further experiments demonstrated that adaphostin quenches the fluorescence of the mitochondrial dyes. Structure-activity analysis revealed that the adamantyl and p-aminobenzoic acid moieties of adaphostin are critical for this quenching. Anticancer drugs containing comparable structural motifs, including mitoxantrone, aminoflavone, and amsacrine, also quenched the mitochondrial probes. These results indicate the need for caution when mitochondrial dyes are utilized to examine the effects of xenobiotics on DeltaPsim and suggest that some previously reported direct effects of anticancer drugs on mitochondria might need re-evaluation.
Collapse
Affiliation(s)
- S B Le
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
14
|
Mendoza FJ, Henson ES, Gibson SB. MEKK1-induced apoptosis is mediated by Smac/Diablo release from the mitochondria. Biochem Biophys Res Commun 2005; 331:1089-98. [PMID: 15882989 DOI: 10.1016/j.bbrc.2005.03.234] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2005] [Indexed: 11/22/2022]
Abstract
During apoptotic stimulation, the serine threonine kinase, MEKK1, is cleaved into an activated 91 kDa kinase fragment. This cleavage is mediated by caspase 3 and leads to further caspase 3 activation and apoptosis. Forced expression of the 91 kDa kinase fragment induces apoptosis through changes in membrane potential of the mitochondria mediated by permeability transition pore opening. MEKK1 activation, however, fails to release cytochrome c from the mitochondria. Herein, we determined that overexpression of MEKK1 causes mitochondrial Smac/Diablo release correlating with MEKK1-induced apoptosis. Furthermore, using siRNA that lowers Smac/Diablo expression, MEKK1-induced apoptosis was significantly reduced. Mouse embryonic fibroblast cells lacking MEKK1 expression are also resistant to etoposide-induced mitochondrial Smac/Diablo release. In contrast, etoposide-induced mitochondrial cytochrome c release was not inhibited. MEKK1 also activates the MAP kinase JNK, but MEKK1-induced mitochondrial Smac/Diablo release and apoptosis are independent of MEKK1 mediated JNK activation. Taken together, release of Smac/Diablo from the mitochondria plays a role in MEKK1-induced apoptosis.
Collapse
Affiliation(s)
- Francisco J Mendoza
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, Man., Canada R3N 0V9
| | | | | |
Collapse
|
15
|
Kohli M, Yu J, Seaman C, Bardelli A, Kinzler KW, Vogelstein B, Lengauer C, Zhang L. SMAC/Diablo-dependent apoptosis induced by nonsteroidal antiinflammatory drugs (NSAIDs) in colon cancer cells. Proc Natl Acad Sci U S A 2004; 101:16897-902. [PMID: 15557007 PMCID: PMC534714 DOI: 10.1073/pnas.0403405101] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Accepted: 10/22/2004] [Indexed: 11/18/2022] Open
Abstract
Nonsteroidal antiinflammatory drugs (NSAIDs) form a paradigm for the chemoprevention of cancer, preventing colonic tumor progression in both experimental animals and humans. However, the mechanisms underlying the antineoplastic effects of NSAIDs are currently unclear. We found that the mitochondrial second mitochondrial-derived activator of caspase (SMAC)/direct inhibitor of apoptosis protein-binding protein with low pI (Diablo) protein translocates into the cytosol during NSAID-induced apoptosis in colon cancer cells. When SMAC/Diablo is disrupted by homologous recombination and RNA interference in these cells, the NSAID-induced apoptosis is abrogated. Biochemical markers of apoptosis, such as caspase activation, cytosolic release of cytochrome c and apoptosis-inducing factor, and mitochondrial membrane potential change, are accordingly decreased. These results establish that SMAC/Diablo is essential for the apoptosis induced by NSAIDs in colon cancer cells.
Collapse
Affiliation(s)
- Manu Kohli
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Cummins JM, Kohli M, Rago C, Kinzler KW, Vogelstein B, Bunz F. X-Linked Inhibitor of Apoptosis Protein (XIAP) Is a Nonredundant Modulator of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL)- Mediated Apoptosis in Human Cancer Cells. Cancer Res 2004; 64:3006-8. [PMID: 15126334 DOI: 10.1158/0008-5472.can-04-0046] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to play an important role in the immunosurveillance of neoplasia, apoptotic factors that modulate the sensitivity of cancer cells to TRAIL are poorly understood. The inhibitor of apoptosis proteins (IAPs) have generated considerable interest as potential targets for cancer therapy, but the lack of a phenotype in X-linked IAP (XIAP) knockout mice has generated speculation that IAP function may be redundant. Using gene targeting technology, we show that disruption of the gene encoding XIAP in human cancer cells did not interfere with basal proliferation, but caused a remarkable sensitivity to TRAIL. These results demonstrate that XIAP is a nonredundant modulator of TRAIL-mediated apoptosis and provide a rationale for XIAP as a therapeutic target.
Collapse
Affiliation(s)
- Jordan M Cummins
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|