1
|
Fang Q, Xu Y, Tan X, Wu X, Li S, Yuan J, Chen X, Huang Q, Fu K, Xiao S. The Role and Therapeutic Potential of Pyroptosis in Colorectal Cancer: A Review. Biomolecules 2024; 14:874. [PMID: 39062587 PMCID: PMC11274949 DOI: 10.3390/biom14070874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related mortality worldwide. The unlimited proliferation of tumor cells is one of the key features resulting in the malignant development and progression of CRC. Consequently, understanding the potential proliferation and growth molecular mechanisms and developing effective therapeutic strategies have become key in CRC treatment. Pyroptosis is an emerging type of regulated cell death (RCD) that has a significant role in cells proliferation and growth. For the last few years, numerous studies have indicated a close correlation between pyroptosis and the occurrence, progression, and treatment of many malignancies, including CRC. The development of effective therapeutic strategies to inhibit tumor growth and proliferation has become a key area in CRC treatment. Thus, this review mainly summarized the different pyroptosis pathways and mechanisms, the anti-tumor (tumor suppressor) and protective roles of pyroptosis in CRC, and the clinical and prognostic value of pyroptosis in CRC, which may contribute to exploring new therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Qing Fang
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yunhua Xu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xiangwen Tan
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xiaofeng Wu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Shuxiang Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Jinyi Yuan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Xiguang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Qiulin Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuai Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.F.); (Y.X.); (X.T.); (X.W.)
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.L.); (J.Y.); (X.C.); (Q.H.)
| |
Collapse
|
2
|
Liu Z, Li Y, Zhu Y, Li N, Li W, Shang C, Song G, Li S, Cong J, Li T, Xiu Z, Lu J, Ge C, Yang X, Li Y, Sun L, Li X, Jin N. Apoptin induces pyroptosis of colorectal cancer cells via the GSDME-dependent pathway. Int J Biol Sci 2022; 18:717-730. [PMID: 35002520 PMCID: PMC8741846 DOI: 10.7150/ijbs.64350] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Apoptin is a small molecular weight protein encoded by the VP3 gene of chicken anemia virus (CAV). It can induce apoptosis of tumor cells and play anti-tumorigenic functions. In this study, we identified a time-dependent inhibitory role of apoptin on the viability of HCT116 cells. We also demonstrated that apoptin induces pyroptosis through cleaved caspase 3, and with a concomitant cleavage of gasdermin E (GSDME) rather than GSDMD. GSDME knockdown switched the apoptin-induced cell death from pyroptosis to apoptosis in vitro. Furthermore, we demonstrated that the effect of apoptin on GSDME-dependent pyroptosis could be mitigated by caspase-3 and caspase-9 siRNA knockdown. Additionally, apoptin enhanced the intracellular reactive oxygen species (ROS), causing aggregation of the mitochondrial membrane protein Tom20. Moreover, bax and cytochrome c were released to the activating caspase-9, eventually triggering pyroptosis. Therefore, GSDME mediates the apoptin-induced pyroptosis through the mitochondrial apoptotic pathway. Finally, using nude mice xenografted with HCT116 cells, we found that apoptin induces pyroptosis and significantly inhibits tumor growth. Based on this mechanism, apoptin may provide a new strategy for colorectal cancer therapy.
Collapse
Affiliation(s)
- Zirui Liu
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yilong Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Nan Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Wenjie Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Chao Shang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Gaojie Song
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Shanzhi Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Jianan Cong
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Tingyu Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Zhiru Xiu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Jing Lu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Chenchen Ge
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Xia Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Yaru Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China
| | - Lili Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China.,Department of Head and Neck Surgery, Tumor Hospital of Jilin Province, Changchun, 130012, China
| | - Xiao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Ningyi Jin
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 130122, China.,Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, 130021, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| |
Collapse
|
3
|
Ali T, Lei X, Barbour SE, Koizumi A, Chalfant CE, Ramanadham S. Alterations in β-Cell Sphingolipid Profile Associated with ER Stress and iPLA 2β: Another Contributor to β-Cell Apoptosis in Type 1 Diabetes. Molecules 2021; 26:molecules26216361. [PMID: 34770770 PMCID: PMC8587436 DOI: 10.3390/molecules26216361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) development, in part, is due to ER stress-induced β-cell apoptosis. Activation of the Ca2+-independent phospholipase A2 beta (iPLA2β) leads to the generation of pro-inflammatory eicosanoids, which contribute to β-cell death and T1D. ER stress induces iPLA2β-mediated generation of pro-apoptotic ceramides via neutral sphingomyelinase (NSMase). To gain a better understanding of the impact of iPLA2β on sphingolipids (SLs), we characterized their profile in β-cells undergoing ER stress. ESI/MS/MS analyses followed by ANOVA/Student’s t-test were used to assess differences in sphingolipids molecular species in Vector (V) control and iPLA2β-overexpressing (OE) INS-1 and Akita (AK, spontaneous model of ER stress) and WT-littermate (AK-WT) β-cells. As expected, iPLA2β induction was greater in the OE and AK cells in comparison with V and WT cells. We report here that ER stress led to elevations in pro-apoptotic and decreases in pro-survival sphingolipids and that the inactivation of iPLA2β restores the sphingolipid species toward those that promote cell survival. In view of our recent finding that the SL profile in macrophages—the initiators of autoimmune responses leading to T1D—is not significantly altered during T1D development, we posit that the iPLA2β-mediated shift in the β-cell sphingolipid profile is an important contributor to β-cell death associated with T1D.
Collapse
Affiliation(s)
- Tomader Ali
- Research Department, Imperial College London Diabetes Center, Abu Dhabi 51133, United Arab Emirates;
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Suzanne E. Barbour
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Akio Koizumi
- Department of Health and Environmental Sciences, Kyoto Graduate School of Medicine, Kyoto 606-8501, Japan;
| | - Charles E. Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA;
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: ; Tel.: +1-205-996-5973; Fax: +1-205-996-5220
| |
Collapse
|
4
|
Wu JMF, Schulze PC. Cardiovascular prognosis: a new role for ceramides and other cardiometabolites. ESC Heart Fail 2020; 7:3285-3287. [PMID: 33040495 PMCID: PMC7754998 DOI: 10.1002/ehf2.13025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Jasmine M F Wu
- Division of Cardiology, Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | - P Christian Schulze
- Division of Cardiology, Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| |
Collapse
|
5
|
Malla WA, Arora R, Khan RIN, Mahajan S, Tiwari AK. Apoptin as a Tumor-Specific Therapeutic Agent: Current Perspective on Mechanism of Action and Delivery Systems. Front Cell Dev Biol 2020; 8:524. [PMID: 32671070 PMCID: PMC7330108 DOI: 10.3389/fcell.2020.00524] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer remains one of the leading causes of death worldwide in humans and animals. Conventional treatment regimens often fail to produce the desired outcome due to disturbances in cell physiology that arise during the process of transformation. Additionally, development of treatment regimens with no or minimum side-effects is one of the thrust areas of modern cancer research. Oncolytic viral gene therapy employs certain viral genes which on ectopic expression find and selectively destroy malignant cells, thereby achieving tumor cell death without harming the normal cells in the neighborhood. Apoptin, encoded by Chicken Infectious Anemia Virus' VP3 gene, is a proline-rich protein capable of inducing apoptosis in cancer cells in a selective manner. In normal cells, the filamentous Apoptin becomes aggregated toward the cell margins, but is eventually degraded by proteasomes without harming the cells. In malignant cells, after activation by phosphorylation by a cancer cell-specific kinase whose identity is disputed, Apoptin accumulates in the nucleus, undergoes aggregation to form multimers, and prevents the dividing cancer cells from repairing their DNA lesions, thereby forcing them to undergo apoptosis. In this review, we discuss the present knowledge about the structure of Apoptin protein, elaborate on its mechanism of action, and summarize various strategies that have been used to deliver it as an anticancer drug in various cancer models.
Collapse
Affiliation(s)
- Waseem Akram Malla
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Richa Arora
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Sonalika Mahajan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Ashok Kumar Tiwari
- Division of Biological Standardisation, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| |
Collapse
|
6
|
Gilbert S, Loranger A, Omary MB, Marceau N. Keratin impact on PKCδ- and ASMase-mediated regulation of hepatocyte lipid raft size - implication for FasR-associated apoptosis. J Cell Sci 2016; 129:3262-73. [PMID: 27422101 DOI: 10.1242/jcs.171124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/13/2016] [Indexed: 11/20/2022] Open
Abstract
Keratins are epithelial cell intermediate filament (IF) proteins that are expressed as pairs in a cell-differentiation-regulated manner. Hepatocytes express the keratin 8 and 18 pair (denoted K8/K18) of IFs, and a loss of K8 or K18, as in K8-null mice, leads to degradation of the keratin partner. We have previously reported that a K8/K18 loss in hepatocytes leads to altered cell surface lipid raft distribution and more efficient Fas receptor (FasR, also known as TNFRSF6)-mediated apoptosis. We demonstrate here that the absence of K8 or transgenic expression of the K8 G62C mutant in mouse hepatocytes reduces lipid raft size. Mechanistically, we find that the lipid raft size is dependent on acid sphingomyelinase (ASMase, also known as SMPD1) enzyme activity, which is reduced in absence of K8/K18. Notably, the reduction of ASMase activity appears to be caused by a less efficient redistribution of surface membrane PKCδ toward lysosomes. Moreover, we delineate the lipid raft volume range that is required for an optimal FasR-mediated apoptosis. Hence, K8/K18-dependent PKCδ- and ASMase-mediated modulation of lipid raft size can explain the more prominent FasR-mediated signaling resulting from K8/K18 loss. The fine-tuning of ASMase-mediated regulation of lipid rafts might provide a therapeutic target for death-receptor-related liver diseases.
Collapse
Affiliation(s)
- Stéphane Gilbert
- Centre de recherche sur le cancer de l'Université Laval and Centre de recherche du CHU de Québec (HDQ), Québec, Canada G1R 2J6
| | - Anne Loranger
- Centre de recherche sur le cancer de l'Université Laval and Centre de recherche du CHU de Québec (HDQ), Québec, Canada G1R 2J6
| | - M Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Normand Marceau
- Centre de recherche sur le cancer de l'Université Laval and Centre de recherche du CHU de Québec (HDQ), Québec, Canada G1R 2J6
| |
Collapse
|
7
|
Lezhnin YN, Kravchenko YE, Frolova EI, Chumakov PM, Chumakov SP. Oncotoxic proteins in cancer therapy: Mechanisms of action. Mol Biol 2015. [DOI: 10.1134/s0026893315020077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Abstract
The virus-derived protein Apoptin has the ability to induce p53-independent apoptosis in a variety of human cancer cells while leaving normal cells unharmed. It thus represents a potential anti-cancer therapeutic agent of the future but a proper understanding of Apoptin-induced signalling events is necessary prior to clinical application. The tumor-specific nuclear translocation and phosphorylation of Apoptin by a cellular kinase such as protein kinase C seem to be required for its function but otherwise the mode of tumor selectivity remains unknown. Apoptin has been shown to interact with several cellular proteins including Akt and the anaphase-promoting complex that regulate its activity and promote caspase-dependent apoptosis. This chapter summarizes the available data on tumor-specific pathways sensed by Apoptin and the mechanism of Apoptin-induced cell death.
Collapse
Affiliation(s)
- Jessica Bullenkamp
- Kings College London, Guy's Hospital, Floor 2 Room 2.66S, Hodgkin Building, London, UK
| | | |
Collapse
|
9
|
Yun SH, Park ES, Shin SW, Na YW, Han JY, Jeong JS, Shastina VV, Stonik VA, Park JI, Kwak JY. Stichoposide C induces apoptosis through the generation of ceramide in leukemia and colorectal cancer cells and shows in vivo antitumor activity. Clin Cancer Res 2013; 18:5934-48. [PMID: 23132899 DOI: 10.1158/1078-0432.ccr-12-0655] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Marine triterpene glycosides that are physiologically active natural compounds isolated from sea cucumbers (holothurians) and sponges have antifungal, cytotoxic, and antitumor activities, whose specific molecular mechanisms remain to be elucidated. In this study, we examined if and through which mechanisms stichoposide C (STC) from Thelenota anax (family Stichopodidae) induces apoptosis in leukemia and colorectal cancer cells. EXPERIMENTAL DESIGN We examined STC-induced apoptosis in human leukemia and colorectal cancer cells in the context of mitochondrial injury and signaling pathway disturbances, and investigated the antitumor effect of STC in mouse CT-26 subcutaneous tumor and HL-60 leukemia xenograft models. RESULTS We found that STC induces apoptosis in these cells in a dose-dependent manner and leads to the activation of Fas and caspase-8, cleavage of Bid, mitochondrial damage, and activation of caspase-3. STC activates acid sphingomyelinase (SMase) and neutral SMase, which resulted in the generation of ceramide. Specific inhibition of acid SMase or neutral SMase and siRNA knockdown experiments partially blocked STC-induced apoptosis. Moreover, STC markedly reduced tumor growth of HL-60 xenograft and CT-26 subcutaneous tumors and increased ceramide generation in vivo. CONCLUSIONS Ceramide generation by STC, through activation of acid and neutral SMase, may in part contribute to STC-induced apoptosis and antitumor activity. Thus, STC may have therapeutic relevance for human leukemia and colorectal cancer.
Collapse
Affiliation(s)
- Seong-Hoon Yun
- Department of Biochemistry, Dong-A University College of Medicine, Busan, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Taebunpakul P, Sayan BS, Flinterman M, Klanrit P, Gäken J, Odell EW, Melino G, Tavassoli M. Apoptin induces apoptosis by changing the equilibrium between the stability of TAp73 and ΔNp73 isoforms through ubiquitin ligase PIR2. Apoptosis 2012; 17:762-76. [PMID: 22484480 DOI: 10.1007/s10495-012-0720-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Apoptin, a protein derived from the chicken anaemia virus, induces cell death in various cancer cells but shows little or no cytotoxicity in normal cells. The mechanism of apoptin-induced cell death is currently unknown but it appears to induce apoptosis independent of p53 status. Here we show that p73, a p53 family member, is important in apoptin-induced apoptosis. In p53 deficient and/or mutated cells, apoptin induced the expression of TAp73 leading to the induction of apoptosis. Knockdown of p73 using siRNA resulted in a significant reduction in apoptin-induced cytotoxicity. The p53 and p73 pro-apoptotic target PUMA plays an important role in apoptin-induced cell death as knockdown of PUMA significantly reduced cell sensitivity to apoptin. Importantly, apoptin expression resulted in a marked increase in TAp73 protein stability. Investigation into the mechanisms of TAp73 stability showed that apoptin induced the expression of the ring finger domain ubiquitin ligase PIR2 which is involved in the degradation of the anti-apoptotic ∆Np73 isoform. Collectively, our results suggest a novel mechanism of apoptin-induced apoptosis through increased TAp73 stability and induction of PIR2 resulting in the degradation of ∆Np73 and activation of pro-apoptotic targets such as PUMA causing cancer cell death.
Collapse
Affiliation(s)
- P Taebunpakul
- Head and Neck Oncology Group, King's College London Dental Institute, Floor 28 Tower Wing, Guy's Hospital Campus, London, SE1 9RT, UK
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Human Gyrovirus Apoptin shows a similar subcellular distribution pattern and apoptosis induction as the chicken anaemia virus derived VP3/Apoptin. Cell Death Dis 2012; 3:e296. [PMID: 22495351 PMCID: PMC3358012 DOI: 10.1038/cddis.2012.34] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The chicken anaemia virus-derived protein Apoptin/VP3 (CAV-Apoptin) has the important ability to induce tumour-selective apoptosis in a variety of human cancer cells. Recently the first human Gyrovirus (HGyV) was isolated from a human skin swab. It shows significant structural and organisational resemblance to CAV and encodes a homologue of CAV-Apoptin/VP3. Using overlapping primers we constructed a synthetic human Gyrovirus Apoptin (HGyV-Apoptin) fused to green fluorescent protein in order to compare its apoptotic function in various human cancer cell lines to CAV-Apoptin. HGyV-Apoptin displayed a similar subcellular expression pattern as observed for CAV-Apoptin, marked by translocation to the nucleus of cancer cells, although it is predominantly located in the cytosol of normal human cells. Furthermore, expression of either HGyV-Apoptin or CAV-Apoptin in several cancer cell lines triggered apoptosis at comparable levels. These findings indicate a potential anti-cancer role for HGyV-Apoptin.
Collapse
|
12
|
Mechanisms of Apoptin-induced cell death. Med Oncol 2011; 29:2985-91. [DOI: 10.1007/s12032-011-0119-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 11/09/2011] [Indexed: 12/22/2022]
|
13
|
Potent inhibition of Acid ceramidase by novel B-13 analogues. J Lipids 2010; 2011:971618. [PMID: 21490813 PMCID: PMC3066644 DOI: 10.1155/2011/971618] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 10/01/2010] [Accepted: 10/26/2010] [Indexed: 01/08/2023] Open
Abstract
The lipid-signalling molecule ceramide is known to induce apoptosis in a variety of cell types. Inhibition of the lysosomal acid ceramidase can increase cellular ceramide levels and thus induce apoptosis. Indeed, inhibitors of acid ceramidase have been reported to induce cell death and to display potentiating effects to classical radio- or chemo therapy in a number of in vitro and in vivo cancer models. The most potent in vitro inhibitor of acid ceramidase, B-13, recently revealed to be virtually inactive towards lysosomal acid ceramidase in living cells. In contrast, a number of weakly basic B-13 analogues have been shown to accumulate in the acidic compartments of living cells and to efficiently inhibit lysosomal acid ceramidase. However, introduction of weakly basic groups at the ω-position of the fatty acid moiety of B-13 led to a significant reduction of potency towards acid ceramidase from cellular extracts. Herein, we report a novel B-13-derived scaffold for more effective inhibitors of acid ceramidase. Furthermore, we provide hints for an introduction of basic functional groups at an alternative site of the B-13 scaffold that do not interfere with acid ceramidase inhibition in vitro.
Collapse
|
14
|
Noteborn MHM. Proteins selectively killing tumor cells. Eur J Pharmacol 2009; 625:165-73. [PMID: 19836376 DOI: 10.1016/j.ejphar.2009.06.068] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/24/2009] [Accepted: 06/25/2009] [Indexed: 01/04/2023]
Abstract
All human cells have a genetic program that upon activation will cause cell death, named apoptosis. Cancer cells can grow due to unbalances in proliferation, cell cycle regulation and their apoptosis machinery: genomic mutations resulting in non-functional pro-apoptosis proteins or over-expression of anti-apoptosis proteins form the basis of tumor formation. Surprisingly, lessons learned from viruses show that cancer cannot be regarded simply as the opposite of apoptosis. For instance, adenovirus can only transform cells when both its anti- and pro-apoptotic proteins are produced. Oncolytic viruses are known to replicate selectively in tumor cells resulting in cell death. Proteins derived from viruses, i.e. chicken anemia virus (CAV)-derived apoptosis-inducing protein (apoptin), adenovirus early region 4 open reading frame (E4orf4) and parvovirus-H1 derived non-structural protein 1 (NS1), the human alpha-lactalbumin made lethal to tumor cells (HAMLET), which is present in human milk or the human cytokines melanoma differentiation-associated gene-7 (mda-7) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) have all the ability to induce tumor-selective apoptosis. The tumor-selective apoptosis-inducing proteins seem to interact with transforming survival processes, which can become redirected by these proteins into cell death. Transformation-related processes have been identified, which seem to be crucial for the tumor-selectively killing activity of these proteins. For instance, the transformation-related protein phosphatase 2A (PP2A) plays a role in the induction of tumor-selective apoptosis. The proteins mda-7, TRAIL and HAMLET are already successfully tested in first clinical trials. Proteins harboring tumor-selective apoptosis characteristics represent, therefore, a therapeutic potential and a tool for unraveling tumor-related processes. Fundamental molecular and (pre)clinical therapeutic studies of the various tumor-selective apoptosis-inducing proteins apoptin, E4orf4, HAMLET, mda-7, NS1, TRAIL and related proteins will be discussed.
Collapse
Affiliation(s)
- Mathieu H M Noteborn
- Molecular Genetics, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
15
|
Sun J, Yan Y, Wang XT, Liu XW, Peng DJ, Wang M, Tian J, Zong YQ, Zhang YH, Noteborn MHM, Qu S. PTD4-apoptin protein therapy inhibits tumor growth in vivo. Int J Cancer 2009; 124:2973-81. [PMID: 19326452 DOI: 10.1002/ijc.24279] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Apoptin protein harbors tumor-selective cell death activity, which makes it a potential anticancer therapy candidate. This study reports an apoptin therapy approach based on protein transduction domain 4 (PTD4)-mediated transduction of recombinant apoptin protein. In vitro, the PTD4-apoptin fusion protein is located in the nucleus and induces cell death in, e.g., human hepatocarcinoma HepG2 cells. In normal human L-02 hepatocytes, PTD4-apoptin protein retained mainly cytoplasmic and did not induce detectable levels of cell death, illustrating that the PTD4 domain does not affect apoptin's tumor-selective characteristics. In vivo, liver, cervix and gastric carcinoma xenografts treated with PTD4-apoptin protein for 6 days via the tumor epidermis exhibited a significant tumor growth inhibition because of apoptin-mediated cell death. In addition, treatment of human hepatocarcinoma xenografts during 3 weeks showed that PTD4-apoptin protein has significant anticancer activity, whereas control treatment with PTD4-enhanced green fluorescence protein or saline did not. Cell death and disruption of the tumor integrity were apparent in the PTD4-apoptin transduced xenografted tumors. As important, although PTD4-apoptin protein could be detected in the epidermal tissue covering the subcutaneous tumor tissue and in several organs, such as liver and brain, of the treated mice, no tissue disruption or signs of cell death could be detected. Our in vivo data reveal that apoptin protein delivery constitutes a novel powerful and safe anticancer therapy.
Collapse
Affiliation(s)
- Jun Sun
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mahdy AEM, Cheng JC, Li J, Elojeimy S, Meacham WD, Turner LS, Bai A, Gault CR, McPherson AS, Garcia N, Beckham TH, Saad A, Bielawska A, Bielawski J, Hannun YA, Keane TE, Taha MI, Hammouda HM, Norris JS, Liu X. Acid ceramidase upregulation in prostate cancer cells confers resistance to radiation: AC inhibition, a potential radiosensitizer. Mol Ther 2008; 17:430-8. [PMID: 19107118 DOI: 10.1038/mt.2008.281] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Radiation resistance in a subset of prostate tumors remains a challenge to prostate cancer radiotherapy. The current study on the effects of radiation on prostate cancer cells reveals that radiation programs an unpredicted resistance mechanism by upregulating acid ceramidase (AC). Irradiated cells demonstrated limited changes of ceramide levels while elevating levels of sphingosine and sphingosine-1-phosphate. By genetically downregulating AC with small interfering RNA (siRNA), we observed radiosensitization of cells using clonogenic and cytotoxicity assays. Conversely, AC overexpression further decreased sensitivity to radiation. We also observed that radiation-induced AC upregulation was sufficient to create cross-resistance to chemotherapy as demonstrated by decreased sensitivity to Taxol and C(6) ceramide compared to controls. Lower levels of caspase 3/7 activity were detected in cells pretreated with radiation, also indicating increased resistance. Finally, utilization of the small molecule AC inhibitor, LCL385, sensitized PPC-1 cells to radiation and significantly decreased tumor xenograft growth. These data suggest a new mechanism of cancer cell resistance to radiation, through upregulation of AC that is, in part, mediated by application of the therapy itself. An improved understanding of radiotherapy and the application of combination therapy achieved in this study offer new opportunities for the modulation of radiation effects in the treatment of cancer.
Collapse
Affiliation(s)
- Ayman E M Mahdy
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Bedia C, Canals D, Matabosch X, Harrak Y, Casas J, Llebaria A, Delgado A, Fabriás G. Cytotoxicity and acid ceramidase inhibitory activity of 2-substituted aminoethanol amides. Chem Phys Lipids 2008; 156:33-40. [DOI: 10.1016/j.chemphyslip.2008.07.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 07/09/2008] [Accepted: 07/23/2008] [Indexed: 12/14/2022]
|
18
|
Backendorf C, Visser AE, de Boer AG, Zimmerman R, Visser M, Voskamp P, Zhang YH, Noteborn M. Apoptin: therapeutic potential of an early sensor of carcinogenic transformation. Annu Rev Pharmacol Toxicol 2008; 48:143-69. [PMID: 17848136 DOI: 10.1146/annurev.pharmtox.48.121806.154910] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The avian virus-derived protein apoptin induces p53-independent apoptosis in a tumor-specific way. Apoptin acts as a multimeric complex and forms superstructures upon binding to DNA. In tumor cells, apoptin is phosphorylated and mainly nuclear, whereas in normal cells it is unphosphorylated, cytoplasmic, and becomes readily neutralized. Interestingly, apoptin phosphorylation, nuclear translocation, and apoptosis can transiently be induced in normal cells by cotransfecting SV40 large T oncogene, indicating that apoptin recognizes early stages of oncogenic transformation. In cancer cells, apoptin appears to recognize survival signals, which it is able to redirect into cell death impulses. Apoptin targets include DEDAF, Nur77, Nmi, Hippi, and the potential drug target APC1. Apoptin-transgenic mice and animal tumor models have revealed apoptin as a safe and efficient antitumor agent, resulting in significant tumor regression. Future antitumor therapies could use apoptin either as a therapeutic bullet or as an early sensor of druggable tumor-specific processes.
Collapse
Affiliation(s)
- Claude Backendorf
- Molecular Genetics, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Visser AE, Backendorf C, Noteborn M. Viral protein apoptin as a molecular tool and therapeutic bullet: implications for cancer control. Future Virol 2007. [DOI: 10.2217/17460794.2.5.519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The chicken anemia virus-derived protein apoptin induces apoptosis in human tumor cells via a p53-independent pathway, while leaving normal cells intact. Moreover, apoptin treatment in preclinical animal studies leads to reduced tumor growth or remission without a detectable effect on healthy tissues. Apoptin is activated by a still unknown tumor-specific kinase activity. The mode of action of apoptin is under intense investigation, as certain features make it a promising tool for discovering early events in tumorigenesis, identifying druggable targets for antitumor treatment and possibly serving as an antitumor therapy in itself.
Collapse
Affiliation(s)
- Astrid E Visser
- Leiden University, Molecular Genetics, Leiden Institute of Chemistry, 2300 RA Leiden, The Netherlands
| | - Claude Backendorf
- Leiden University, Molecular Genetics, Leiden Institute of Chemistry, 2300 RA Leiden, The Netherlands
| | - Mathieu Noteborn
- Leiden University, Biological Chemistry, Leiden Institute of Chemistry, 2300 RA Leiden, The Netherlands
| |
Collapse
|
20
|
Zeidan YH, Hannun YA. Activation of Acid Sphingomyelinase by Protein Kinase Cδ-mediated Phosphorylation. J Biol Chem 2007; 282:11549-61. [PMID: 17303575 DOI: 10.1074/jbc.m609424200] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although important for cellular stress signaling pathways, the molecular mechanisms of acid sphingomyelinase (ASMase) activation remain poorly understood. Previous studies showed that treatment of MCF-7 mammary carcinoma cells with the potent protein kinase C (PKC) agonist, phorbol 12-myristate 13-acetate (PMA), induces a transient drop in sphingomyelin concomitant with an increase in cellular ceramide levels (Becker, K. P., Kitatani, K., Idkowiak-Baldys, J., Bielawski, J., and Hannun, Y. A. (2005) J. Biol. Chem. 280, 2606-2612). Here we show that PMA selectively activates ASMase and that ASMase accounts for the majority of PMA-induced ceramide. Pharmacologic inhibition and RNA interference experiments indicated that the novel PKC, PKCdelta, is required for ASMase activation. Immunoprecipitation experiments revealed the formation of a novel PKCdelta-ASMase complex after PMA stimulation, and PKCdelta was able to phosphorylate ASMase in vitro and in cells. Using site-directed mutagenesis, we identify serine 508 as the key residue phosphorylated in response to PMA. Phosphorylation of Ser(508) proved to be an indispensable step for ASMase activation and membrane translocation in response to PMA. The relevance of the proposed mechanism of ASMase regulation is further validated in a model of UV radiation. UV radiation also induced phosphorylation of ASMase at serine 508. Moreover, when transiently overexpressed, ASMase(S508A) blocked the ceramide formation after PMA treatment, suggesting a dominant negative function for this mutant. Taken together, these results establish a novel direct biochemical mechanism for ASMase activation in which PKCdelta serves as a key upstream kinase.
Collapse
Affiliation(s)
- Youssef H Zeidan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | |
Collapse
|
21
|
Chada S, Ramesh R. Apoptin studies illuminate intersection between lipidomics and tumor suppressors. Mol Ther 2007. [DOI: 10.1038/sj.mt.6300055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
22
|
Liu X, Elojeimy S, El-Zawahry AM, Holman DH, Bielawska A, Bielawski J, Rubinchik S, Guo GW, Dong JY, Keane T, Hannun YA, Tavassoli M, Norris JS. Modulation of ceramide metabolism enhances viral protein apoptin's cytotoxicity in prostate cancer. Mol Ther 2006; 14:637-46. [PMID: 16887394 DOI: 10.1016/j.ymthe.2006.06.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Revised: 06/06/2006] [Accepted: 06/06/2006] [Indexed: 12/19/2022] Open
Abstract
Despite local and systemic therapies, the National Cancer Institute estimates that prostate cancer will cause over 30,000 deaths in 2006. This suggests that additional therapeutic approaches are needed. The chicken anemia viral protein Apoptin causes tumor-selective apoptosis in human tumor lines independent of p53 and Bcl-2 status. Tet-regulated expression of Apoptin from an adenoviral vector showed cytotoxicity in DU145, PC-3, and LNCaP tumor cells regardless of expression of p53, Bcl-2, Bcl-xL, Bax, survivin, FLIP(S), XIAP, or CIAP. Apoptin expression caused an increase in the tumor suppressor lipid ceramide, which regulates the cellular stress response. Interestingly, 10 of 15 primary prostate cancers examined by Western blotting overexpressed acid ceramidase (AC), suggesting that ceramide deacylation might serve to negate elevated levels of ceramide, creating a more antiapoptotic phenotype. This was confirmed in AC-overexpressing cells in which we observed decreased sensitivity to apoptosis following treatment with Apoptin. Addition of the AC inhibitor LCL204, in combination with Apoptin, augmented cell killing. This effect was also demonstrated in vivo in that Apoptin and LCL204 cotreatment significantly reduced tumor growth in DU145 xenografts (P<0.05). Taken together, our data demonstrated that Apoptin is a promising therapeutic agent for prostate cancer and that its function is improved when combined with acid ceramidase inhibitors.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|