1
|
Iba T, Helms J, Maier CL, Ferrer R, Levy JH. Mitochondrial dysfunction is a major cause of thromboinflammation and inflammatory cell death in critical illnesses. Inflamm Res 2025; 74:17. [PMID: 39806233 DOI: 10.1007/s00011-025-01994-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/02/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Mitochondria generate the adenosine triphosphate (ATP) necessary for eukaryotic cells, serving as their primary energy suppliers, and contribute to host defense by producing reactive oxygen species. In many critical illnesses, including sepsis, major trauma, and heatstroke, the vicious cycle between activated coagulation and inflammation results in tissue hypoxia-induced mitochondrial dysfunction, and impaired mitochondrial function contributes to thromboinflammation and cell death. METHODS A computer-based online search was performed using the PubMed and Web of Science databases for published articles concerning sepsis, trauma, critical illnesses, cell death, mitochondria, inflammation, coagulopathy, and organ dysfunction. RESULTS Mitochondrial outer membrane permeabilization triggers apoptosis by releasing cytochrome c and activating caspases. Apoptosis is a non-inflammatory programmed cell death but requires sufficient ATP supply. Therefore, conversion to inflammatory necrosis may occur due to a lack of ATP in critical illness. Severely damaged mitochondria release excess reactive oxygen species and injurious mitochondrial DNA, inducing cell death. Besides non-programmed necrosis, mitochondrial damage can trigger programmed inflammatory cell death, including necroptosis, pyroptosis, and ferroptosis. Additionally, a unique form of DNA-ejecting cell death, known as etosis, occurs in monocytes and granulocytes following external stimuli and mitochondrial damage. The type of cell death chosen remains uncertain but is known to depend on the cell type, the nature of the injury, and the degree of damage. CONCLUSIONS Mitochondria damage is the major contributor to the cell death mechanism that leads to organ damage in critical illnesses. Regulating and restoring mitochondrial function holds promise for developing new therapeutic approaches for mitigating critical diseases.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Julie Helms
- Medical Intensive Care Unit - NHC, Strasbourg University (UNISTRA), Strasbourg University Hospital, INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ricard Ferrer
- Intensive Care Department, Hospital Universitari Vall d'Hebron Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Ward GA, Zhang Z, Jueliger S, Potapov IS, Davis MP, Boxall AR, Taylor J, Keer H, Biondo A, Lyons JF, Sims M, Smyth T. Epigenetic Priming by Hypomethylation Enhances the Immunogenic Potential of Tolinapant in T-cell Lymphoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1441-1453. [PMID: 38727208 PMCID: PMC11155518 DOI: 10.1158/2767-9764.crc-23-0415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/02/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
Programmed cell death mechanisms are important for the regulation of tumor development and progression. Evasion of and resistance to apoptosis are significant factors in tumorigenesis and drug resistance. Bypassing apoptotic pathways and eliciting another form of regulated cell death, namely necroptosis, an immunogenic cell death (ICD), may override apoptotic resistance. Here, we present the mechanistic rationale for combining tolinapant, an antagonist of the inhibitor of apoptosis proteins (IAP), with decitabine, a hypomethylating agent (HMA), in T-cell lymphoma (TCL). Tolinapant treatment alone of TCL cells in vitro and in syngeneic in vivo models demonstrated that ICD markers can be upregulated, and we have shown that epigenetic priming with decitabine further enhances this effect. The clinical relevance of ICD markers was confirmed by the direct measurement of plasma proteins from patients with peripheral TCL treated with tolinapant. We showed increased levels of necroptosis in TCL lines, along with the expression of cancer-specific antigens (such as cancer testis antigens) and increases in genes involved in IFN signaling induced by HMA treatment, together deliver a strong adaptive immune response to the tumor. These results highlight the potential of a decitabine and tolinapant combination for TCL and could lead to clinical evaluation. SIGNIFICANCE The IAP antagonist tolinapant can induce necroptosis, a key immune-activating event, in TCL. Combination with DNA hypomethylation enhances tolinapant sensitivity and primes resistant cells by re-expressing necrosome proteins. In addition, this combination leads to increases in genes involved in IFN signaling and neoantigen expression, providing further molecular rationale for this novel therapeutic option.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jason Taylor
- Astex Pharmaceuticals, Inc., Pleasanton, California
| | - Harold Keer
- Astex Pharmaceuticals, Inc., Pleasanton, California
| | | | | | - Martin Sims
- Astex Pharmaceuticals, Cambridge, United Kingdom
| | - Tomoko Smyth
- Astex Pharmaceuticals, Cambridge, United Kingdom
| |
Collapse
|
3
|
Tang Y, Zhuang C. Design, synthesis and anti-necroptosis activity of fused heterocyclic MLKL inhibitors. Bioorg Med Chem 2024; 102:117659. [PMID: 38442525 DOI: 10.1016/j.bmc.2024.117659] [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: 02/03/2024] [Revised: 02/18/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024]
Abstract
Necroptosis is an important form of programmed cell death (PCD), which is mediated by a death receptor and independent of the caspase proteolytic enzyme. Mixed lineage kinase domain-like (MLKL) is the final effector of necroptosis, playing an irreplaceable role in the execution of necroptosis. However, the studies on MLKL inhibitors are in their infancy. Necrosulfonamide (NSA) is an early-discovered covalent MLKL inhibitor, possessing medium anti-necroptosis activity and a structure-activity relationship (SAR) not widely disclosed. In this study, with the covalent motif maintained, we aim to improve the activity by introducing the terminal fused heterocycles and meanwhile revealing the SAR on the part. As a result, compounds 9 and 14 showed the best activity (EC50 = 148.4 and 595.9 nM) against necroptosis among the analogues by covalently binding to MLKL. The SAR was also concluded to guide further structural optimization in this field.
Collapse
Affiliation(s)
- Yining Tang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
4
|
Ghanavatian P, Salehi-Sedeh H, Ataei F, Hosseinkhani S. Bioluminescent RIPoptosome Assay for FADD/RIPK1 Interaction Based on Split Luciferase Assay in a Human Neuroblastoma Cell Line SH-SY5Y. BIOSENSORS 2023; 13:297. [PMID: 36832063 PMCID: PMC9954477 DOI: 10.3390/bios13020297] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Different programed cell death (PCD) modalities involve protein-protein interactions in large complexes. Tumor necrosis factor α (TNFα) stimulated assembly of receptor-interacting protein kinase 1 (RIPK1)/Fas-associated death domain (FADD) interaction forms Ripoptosome complex that may cause either apoptosis or necroptosis. The present study addresses the interaction of RIPK1 and FADD in TNFα signaling by fusion of C-terminal (CLuc) and N-terminal (NLuc) luciferase fragments to RIPK1-CLuc (R1C) or FADD-NLuc (FN) in a caspase 8 negative neuroblastic SH-SY5Y cell line, respectively. In addition, based on our findings, an RIPK1 mutant (R1C K612R) had less interaction with FN, resulting in increasing cell viability. Moreover, presence of a caspase inhibitor (zVAD.fmk) increases luciferase activity compared to Smac mimetic BV6 (B), TNFα -induced (T) and non-induced cell. Furthermore, etoposide decreased luciferase activity, but dexamethasone was not effective in SH-SY5Y. This reporter assay might be used to evaluate basic aspects of this interaction as well as for screening of necroptosis and apoptosis targeting drugs with potential therapeutic application.
Collapse
|
5
|
Chen F, Zhong Z, Tan HY, Guo W, Zhang C, Cheng C, Wang N, Ren J, Feng Y. Suppression of lncRNA MALAT1 by betulinic acid inhibits hepatocellular carcinoma progression by targeting IAPs via miR-22-3p. Clin Transl Med 2020; 10:e190. [PMID: 33135336 PMCID: PMC7586994 DOI: 10.1002/ctm2.190] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Betulinic acid (BA) is a natural product extracted from a broad range of medicinal and edible herbal plants. Previous studies showed that BA induces cell death in tumors derived from multiple tissues; however, the underlying mechanism remains obscure. The present study aimed to study the effects of BA on autophagy and apoptosis of hepatocellular carcinoma (HCC). Human HCC cell lines and orthotopic HCC implanted mice were employed to examine the BA-induced tumor suppression; RT2 long noncoding RNA (lncRNA) PCR array and database analysis were used to explore the possible mechanisms; validation of pathways was performed using siRNA and miRNA inhibitors. The results indicated that BA regulated autophagy and induced apoptosis in HCC. The degradation of inhibitor of apoptosis proteins (IAPs), the conversion of LC3-I to LC3-II, and p62 accumulation were enhanced by BA, thereby suggesting that the downregulation of IAPs and autophagic cell death are induced by BA. The addition of autophagy and lysosomal inhibitors indicated that BA induced autophagy-independent apoptosis via degradation of IAPs. Moreover, RT2 lncRNA PCR array and database analysis suggested that BA downregulated the levels of lncRNA MALAT1, which is considered to be an oncogene. Further investigations demonstrated that lncRNA MALAT1 functioned as a ceRNA (competing endogenous RNA) to contribute to BA-mediated degradation of IAPs by sponging miR-22-3p. Therefore, BA could be developed as a potential anticancer agent for HCC.
Collapse
Affiliation(s)
- Feiyu Chen
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Zhangfeng Zhong
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Hor Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Wei Guo
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Chien‐Shan Cheng
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| | - Junguo Ren
- Institute of Basic Medical Sciences, Xiyuan HospitalChina Academy of Chinese Medical SciencesBeijingP. R. China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.P. R. China
| |
Collapse
|
6
|
Kumar S, Fairmichael C, Longley DB, Turkington RC. The Multiple Roles of the IAP Super-family in cancer. Pharmacol Ther 2020; 214:107610. [PMID: 32585232 DOI: 10.1016/j.pharmthera.2020.107610] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/16/2020] [Accepted: 06/08/2020] [Indexed: 12/22/2022]
Abstract
The Inhibitor of Apoptosis proteins (IAPs) are a family of proteins that are mainly known for their anti-apoptotic activity and ability to directly bind and inhibit caspases. Recent research has however revealed that they have extensive roles in governing numerous other cellular processes. IAPs are known to modulate ubiquitin (Ub)-dependent signaling pathways through their E3 ligase activity and influence activation of nuclear factor κB (NF-κB). In this review, we discuss the involvement of IAPs in individual hallmarks of cancer and the current status of therapies targeting these critical proteins.
Collapse
Affiliation(s)
- Swati Kumar
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, United Kingdom
| | - Ciaran Fairmichael
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, United Kingdom
| | - Daniel B Longley
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, United Kingdom
| | - Richard C Turkington
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, United Kingdom.
| |
Collapse
|
7
|
Abbas R, Larisch S. Targeting XIAP for Promoting Cancer Cell Death-The Story of ARTS and SMAC. Cells 2020; 9:E663. [PMID: 32182843 PMCID: PMC7140716 DOI: 10.3390/cells9030663] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/18/2022] Open
Abstract
Inhibitors of apoptosis (IAPs) are a family of proteins that regulate cell death and inflammation. XIAP (X-linked IAP) is the only family member that suppresses apoptosis by directly binding to and inhibiting caspases. On the other hand, cIAPs suppress the activation of the extrinsic apoptotic pathway by preventing the formation of pro-apoptotic signaling complexes. IAPs are negatively regulated by IAP-antagonist proteins such as Smac/Diablo and ARTS. ARTS can promote apoptosis by binding and degrading XIAP via the ubiquitin proteasome-system (UPS). Smac can induce the degradation of cIAPs but not XIAP. Many types of cancer overexpress IAPs, thus enabling tumor cells to evade apoptosis. Therefore, IAPs, and in particular XIAP, have become attractive targets for cancer therapy. In this review, we describe the differences in the mechanisms of action between Smac and ARTS, and we summarize efforts to develop cancer therapies based on mimicking Smac and ARTS. Several Smac-mimetic small molecules are currently under evaluation in clinical trials. Initial efforts to develop ARTS-mimetics resulted in a novel class of compounds, which bind and degrade XIAP but not cIAPs. Smac-mimetics can target tumors with high levels of cIAPs, whereas ARTS-mimetics are expected to be effective for cancers with high levels of XIAP.
Collapse
Affiliation(s)
| | - Sarit Larisch
- Laboratory of Cell Death and Cancer Research, Biology& Human Biology Departments, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel;
| |
Collapse
|
8
|
Epigenetic Regulation of RIP3 Suppresses Necroptosis and Increases Resistance to Chemotherapy in NonSmall Cell Lung Cancer. Transl Oncol 2019; 13:372-382. [PMID: 31887632 PMCID: PMC6938879 DOI: 10.1016/j.tranon.2019.11.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION The efficacy of chemotherapeutic agents in killing cancer cells is mainly attributed to the induction of apoptosis. However, the tremendous efforts on enhancing apoptosis-related mechanisms have only moderately improved lung cancer chemotherapy, suggesting that other cell death mechanisms such as necroptosis could be involved. In this study, we investigated the role of the necroptosis pathway in the responsiveness of nonsmall cell lung cancer (NSCLC) to chemotherapy. METHODS In vitro cell culture and in vivo xenograft tumor therapy models and clinical sample studies are combined in studying the role of necroptosis in chemotherapy and mechanism of necroptosis suppression involving RIP3 expression regulation. RESULTS While chemotherapeutic drugs were able to induce necroptotic cell death, this pathway was suppressed in lung cancer cells at least partly through downregulation of RIP3 expression. Ectopic RIP3 expression significantly sensitized lung cancer cells to the cytotoxicity of anticancer drugs such as cisplatin, etoposide, vincristine, and adriamycin. In addition, RIP3 suppression was associated with RIP3 promoter methylation, and demethylation partly restored RIP3 expression and increased chemotherapeutic-induced necroptotic cell death. In a xenograft tumor therapy model, ectopic RIP3 expression significantly sensitized anticancer activity of cisplatin in vivo. Furthermore, lower RIP3 expression was associated with worse chemotherapy response in NSCLC patients. CONCLUSION Our results indicate that the necroptosis pathway is suppressed in lung cancer through RIP3 promoter methylation, and reactivating this pathway should be exploited for improving lung cancer chemotherapy.
Collapse
|
9
|
The Molecular Links between Cell Death and Inflammasome. Cells 2019; 8:cells8091057. [PMID: 31509938 PMCID: PMC6769855 DOI: 10.3390/cells8091057] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death pathways and inflammasome activation pathways can be genetically and functionally separated. Inflammasomes are specialized protein complexes that process pro-inflammatory cytokines, interleukin-1β (IL-1β), and IL-18 to bioactive forms for protection from a wide range of pathogens, as well as environmental and host-derived danger molecules. Programmed cell death has been extensively studied, and its role in the development, homeostasis, and control of infection and danger is widely appreciated. Apoptosis and the recently recognized necroptosis are the best-characterized forms of programmed death, and the interplay between them through death receptor signaling is also being studied. Moreover, growing evidence suggests that many of the signaling molecules known to regulate programmed cell death can also modulate inflammasome activation in a cell-intrinsic manner. Therefore, in this review, we will discuss the current knowledge concerning the role of the signaling molecules originally associated with programmed cell death in the activation of inflammasome and IL-1β processing.
Collapse
|
10
|
Boddu P, Carter BZ, Verstovsek S, Pemmaraju N. SMACmimetics as potential cancer therapeutics in myeloid malignancies. Br J Haematol 2019; 185:219-231. [DOI: 10.1111/bjh.15829] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Prajwal Boddu
- Department of Hematology and Oncology Yale University School of Medicine New Haven CTUSA
| | - Bing Z. Carter
- Department of Leukemia University of Texas MD Anderson Cancer Center Houston TX USA
| | - Srdan Verstovsek
- Department of Leukemia University of Texas MD Anderson Cancer Center Houston TX USA
| | - Naveen Pemmaraju
- Department of Leukemia University of Texas MD Anderson Cancer Center Houston TX USA
| |
Collapse
|
11
|
Grant S. Rational combination strategies to enhance venetoclax activity and overcome resistance in hematologic malignancies. Leuk Lymphoma 2018; 59:1292-1299. [PMID: 28838268 PMCID: PMC5826810 DOI: 10.1080/10428194.2017.1366999] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Venetoclax (ABT-199) is a Bcl-2-specific BH3-mimetic that has shown significant promise in certain subtypes of CLL as well as in several other hematologic malignancies. As in the case of essentially all targeted agents, intrinsic or acquired resistance to this agent generally occurs, prompting the search for new strategies capable of circumventing this problem. A logical approach to this challenge involves rational combination strategies designed to disable preexisting or induced compensatory survival pathways. Many of these strategies involve downregulation of Mcl-1, a pro-survival Bcl-2 family member that is not targeted by venetoclax, and which often confers resistance to this agent. Given encouraging clinical results involving venetoclax in both lymphoid and myeloid malignancies, it is likely that such combination approaches will be incorporated into the therapeutic armamentarium for multiple hematologic malignancies in the near future.
Collapse
Affiliation(s)
- Steven Grant
- Department of Medicine, Biochemistry, Pharmacology, and Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Translational Research, Developmental Therapeutics Program, Massey Cancer Center, Richmond, VA, USA
- Shirley Carter and Sture Gordon Olsson Professor of Oncology, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| |
Collapse
|
12
|
Otkur W, Liu W, Wang J, Jia X, Huang D, Wang F, Hayashi T, Tashiro SI, Onodera S, Ikejima T. Sub-lethal ultraviolet B irradiation and Poly I:C treatment synergistically induced apoptosis of HaCaT cells through NF-κB pathway. Mol Immunol 2018; 99:19-29. [PMID: 29674236 DOI: 10.1016/j.molimm.2018.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/24/2018] [Accepted: 04/02/2018] [Indexed: 12/23/2022]
Abstract
Ultraviolet B (UVB) irradiation exerts multiple effects on skin cells, inducing apoptosis, senescence and carcinogenesis. Toll-like receptor 3, a member of pattern recognition receptors, is reported to initiate inflammation by recognizing double-strand RNA (dsRNA) released from UVB-irradiated cells. It has not been studied, however, whether apoptosis induction in UVB irradiation is attributed to TLR3 activation. Here, we report on the pro-apoptotic role of TLR3 in UVB-irradiated epidermal cells. Poly I:C, an analogue of dsRNA that activates TLR3, was used in combination with sub-lethal UVB (4.8 mJ/cm2) irradiation for investigating the effects of TLR3 activation on human immortalized keratinocyte HaCaT cells. Although sub-lethal dose of either Poly I:C or UVB alone did not induce cell death, UVB-Poly I:C co-treatment synergistically induced cell death by activation of caspase-3 and cleavages of ICAD and PARP, with apoptotic features when stained with Annexin V/PI or Hoechst 33342. Treatment with pan-caspase inhibitor, Z-VAD, attenuated UVB-Poly I:C-induced cell death. Silencing TLR3 by siRNA rescued HaCaT cells from UVB-Poly I:C-induced apoptosis. NF-κB, a major downstream component of TLR3 pathway, that usually negatively regulates the classical TLR3 apoptotic pathway, was analyzed by western blotting and immunofluorescence confocal microscopy. The results indicate to our surprise that NF-κB is translocated to nucleus in the cells co-treated with UVB-Poly I:C. The nuclear translocation of NF-κB is attenuated by TLR3 silencing. Treatment with BAY, an inhibitor of NF-κB pathway, blocked UVB-Poly I:C-induced apoptosis. Therefore, we conclude that NF-κB pathway plays a cytotoxic role in UVB-Poly I:C-treated HaCaT cells, mediating TLR3-related apoptosis.
Collapse
Affiliation(s)
- Wuxiyar Otkur
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Weiwei Liu
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinda Wang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xingfan Jia
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dianchao Huang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fang Wang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Toshihiko Hayashi
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shin-Ichi Tashiro
- Department of Medical Education & Primary Care, Kyoto Prefectural University of Medicine, Kajiicho 465, Kamikyo-ku, Kyoto City, Kyoto 602-8566, Japan
| | - Satoshi Onodera
- Department of Clinical and Pharmaceutical Sciences, Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Takashi Ikejima
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
13
|
Ballweg R, Paek AL, Zhang T. A dynamical framework for complex fractional killing. Sci Rep 2017; 7:8002. [PMID: 28808338 PMCID: PMC5556027 DOI: 10.1038/s41598-017-07422-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/26/2017] [Indexed: 11/09/2022] Open
Abstract
When chemotherapy drugs are applied to tumor cells with the same or similar genotypes, some cells are killed, while others survive. This fractional killing contributes to drug resistance in cancer. Through an incoherent feedforward loop, chemotherapy drugs not only activate p53 to induce cell death, but also promote the expression of apoptosis inhibitors which inhibit cell death. Consequently, cells in which p53 is activated early undergo apoptosis while cells in which p53 is activated late survive. The incoherent feedforward loop and the essential role of p53 activation timing makes fractional killing a complex dynamical challenge, which is hard to understand with intuition alone. To better understand this process, we have constructed a representative model by integrating the control of apoptosis with the relevant signaling pathways. After the model was trained to recapture the observed properties of fractional killing, it was analyzed with nonlinear dynamical tools. The analysis suggested a simple dynamical framework for fractional killing, which predicts that cell fate can be altered in three possible ways: alteration of bifurcation geometry, alteration of cell trajectories, or both. These predicted categories can explain existing strategies known to combat fractional killing and facilitate the design of novel strategies.
Collapse
Affiliation(s)
- Richard Ballweg
- Department of Molecular and Cellular Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Andrew L Paek
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| | - Tongli Zhang
- Department of Molecular and Cellular Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
14
|
Wassmer SJ, Leonard BC, Coupland SG, Baker AN, Hamilton J, Hauswirth WW, Tsilfidis C. Overexpression of the X-Linked Inhibitor of Apoptosis Protects Against Retinal Degeneration in a Feline Model of Retinal Detachment. Hum Gene Ther 2017; 28:482-492. [PMID: 28335619 PMCID: PMC5488383 DOI: 10.1089/hum.2016.161] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/16/2017] [Indexed: 01/13/2023] Open
Abstract
Retinal detachment is an acute disorder in humans that is caused by trauma or disease, and it can often lead to permanent visual deficits that result from the death of photoreceptors in the retina. The final pathway for photoreceptor cell death is apoptosis and necroptosis. The X-linked inhibitor of apoptosis (XIAP) has been shown to block both of these cell death pathways. This study tested the effects of XIAP on photoreceptor survival in a feline model of retinal detachment. The study was performed in 12 cats, divided into two experimental groups. Six animals received a subretinal injection of adeno-associated virus (AAV) carrying XIAP, and six animals received AAV carrying green fluorescent protein (GFP) as a control. Three weeks after viral delivery, retinas were detached by injecting C3F8 gas into the subretinal space. Optical coherence tomography revealed that the retinal detachments resolved within 3-6 weeks as the gas was slowly resorbed. Analysis of histological sections through the plane of the detachment showed significant preservation of the photoreceptor layer in AAV-XIAP-treated animals compared to AAV-GFP-treated animals at 9 weeks after the detachment. XIAP-treated detached retinas were similar to intact controls. These studies support the potential for XIAP therapy in the treatment of human retinal detachment.
Collapse
Affiliation(s)
- Sarah J. Wassmer
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Brian C. Leonard
- Department of Ophthalmology, University of Ottawa, Ottawa, Canada
- Ottawa Hospital Research Institute, Regenerative Medicine, Ottawa, Canada
- Ottawa Hospital, Eye Institute, Ottawa, Canada
| | - Stuart G. Coupland
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Department of Ophthalmology, University of Ottawa, Ottawa, Canada
- Ottawa Hospital Research Institute, Regenerative Medicine, Ottawa, Canada
- Ottawa Hospital, Eye Institute, Ottawa, Canada
| | - Adam N. Baker
- Ottawa Hospital Research Institute, Regenerative Medicine, Ottawa, Canada
| | | | - William W. Hauswirth
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida
| | - Catherine Tsilfidis
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Department of Ophthalmology, University of Ottawa, Ottawa, Canada
- Ottawa Hospital Research Institute, Regenerative Medicine, Ottawa, Canada
- Ottawa Hospital, Eye Institute, Ottawa, Canada
| |
Collapse
|
15
|
Ma J, Guo W, Li C. Ubiquitination in melanoma pathogenesis and treatment. Cancer Med 2017; 6:1362-1377. [PMID: 28544818 PMCID: PMC5463089 DOI: 10.1002/cam4.1069] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022] Open
Abstract
Melanoma is one of the most aggressive skin cancers with fiercely increasing incidence and mortality. Since the progressive understanding of the mutational landscape and immunologic pathogenic factors in melanoma, the targeted therapy and immunotherapy have been recently established and gained unprecedented improvements for melanoma treatment. However, the prognosis of melanoma patients remains unoptimistic mainly due to the resistance and nonresponse to current available drugs. Ubiquitination is a posttranslational modification which plays crucial roles in diverse cellular biological activities and participates in the pathogenesis of various cancers, including melanoma. Through the regulation of multiple tumor promoters and suppressors, ubiquitination is emerging as the key contributor and therefore a potential therapeutic target for melanoma. Herein, we summarize the current understanding of ubiquitination in melanoma, from mechanistic insights to clinical progress, and discuss the prospect of ubiquitination modification in melanoma treatment.
Collapse
Affiliation(s)
- Jinyuan Ma
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
16
|
Finlay D, Teriete P, Vamos M, Cosford NDP, Vuori K. Inducing death in tumor cells: roles of the inhibitor of apoptosis proteins. F1000Res 2017; 6:587. [PMID: 28529715 PMCID: PMC5414821 DOI: 10.12688/f1000research.10625.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2017] [Indexed: 12/17/2022] Open
Abstract
The heterogeneous group of diseases collectively termed cancer results not just from aberrant cellular proliferation but also from a lack of accompanying homeostatic cell death. Indeed, cancer cells regularly acquire resistance to programmed cell death, or apoptosis, which not only supports cancer progression but also leads to resistance to therapeutic agents. Thus, various approaches have been undertaken in order to induce apoptosis in tumor cells for therapeutic purposes. Here, we will focus our discussion on agents that directly affect the apoptotic machinery itself rather than on drugs that induce apoptosis in tumor cells indirectly, such as by DNA damage or kinase dependency inhibition. As the roles of the Bcl-2 family have been extensively studied and reviewed recently, we will focus in this review specifically on the inhibitor of apoptosis protein (IAP) family. IAPs are a disparate group of proteins that all contain a baculovirus IAP repeat domain, which is important for the inhibition of apoptosis in some, but not all, family members. We describe each of the family members with respect to their structural and functional similarities and differences and their respective roles in cancer. Finally, we also review the current state of IAPs as targets for anti-cancer therapeutics and discuss the current clinical state of IAP antagonists.
Collapse
Affiliation(s)
- Darren Finlay
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Peter Teriete
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Mitchell Vamos
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Nicholas D P Cosford
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Kristiina Vuori
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
17
|
Lin CY, Chang TW, Hsieh WH, Hung MC, Lin IH, Lai SC, Tzeng YJ. Simultaneous induction of apoptosis and necroptosis by Tanshinone IIA in human hepatocellular carcinoma HepG2 cells. Cell Death Discov 2016; 2:16065. [PMID: 27752362 PMCID: PMC5045965 DOI: 10.1038/cddiscovery.2016.65] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 06/29/2016] [Accepted: 07/14/2016] [Indexed: 01/13/2023] Open
Abstract
Tanshinone IIA (Tan IIA), a constituent of the traditional medicinal plant Salvia miltiorrhiza BUNGE, has been reported to possess anticancer activity through induction of apoptosis in many cancer cells. Surprisingly, the present study finds that Tan IIA simultaneously causes apoptosis and necroptosis in human hepatocellular carcinoma HepG2 cells. We further find that apoptosis can be converted to necroptosis by pan-caspase inhibitor Z-VAD-fmk, and the two death modes can be blocked by necroptotic inhibitor necrostatin-1. The underlying mechanisms are revealed by analysis of the signaling molecules using western blotting. In control cells, FLICE inhibitory protein in short form (FLIPS) is expressed in relatively high levels and binds to caspase 8 in ripoptosome, which supposedly sustains cell survival. However, in Tan IIA-treated cells, FLIPS is down-regulated and may thus cause homodimer formation of cleaved caspase 8, cleavage of receptor-interacting serine/threonine-protein kinases 1, 3 (RIP1, RIP3), and mixed-lineage kinase domain-like (MLKL), in turn leads to cell apoptosis. In parallel, Tan IIA causes necroptosis by forming a suggested necrosomal complex composed of RIP1/RIP3. Regarding the inhibitors, z-VAD-fmk diminishes the cleaved caspase 8, RIP1, RIP3, and MLKL induced by Tan IIA, and reconstructs the ripoptosome complex, which marks cells moving from apoptosis to necroptosis. Nec-1 recovers the Tan IIA down-regulated FLIPS, consequently causes FLIPS to form heterodimer with caspase 8 and thus block apoptosis. Meanwhile, cleaved forms of RIP1 and RIP3 were observed preventing necroptosis. Intriguingly, the cytotoxicity of tumor necrosis factor-related apoptosis-inducing ligand to HepG2 cells is enhanced by Tan IIA in a pilot study, which may be attributed to low FLIPS levels induced by Tan IIA. In short, Tan IIA simultaneously induces both Nec-1 inhibition and FLIPS regulation-mediated apoptosis/necroptosis, which has not been previously documented. Moreover, the involvement of the cleavage type of MLKL in executing necroptosis warrants further investigation.
Collapse
Affiliation(s)
- C-Y Lin
- Institute of Medical Sciences, Tzu Chi University , Hualien, Taiwan
| | - T-W Chang
- Division of Crop Improvement, Hualien District Agricultural Research and Extension Station, Council of Agriculture , Hualien, Taiwan
| | - W-H Hsieh
- Department of Public Health, Tzu Chi University , Hualien, Taiwan
| | - M-C Hung
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University of Science and Technology , Hualien, Taiwan
| | - I-H Lin
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan; Department of Chinese Medicine, Buddhist Hualien Tzu Chi General Hospital, Hualien, Taiwan
| | - S-C Lai
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan; Department of Pharmacy, Buddhist Hualien Tzu Chi General Hospital, Hualien, Taiwan
| | - Y-J Tzeng
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan; Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan; Department of Life Science, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
18
|
ZHU GUANGWEI, YE JIANXIN, HUANG YONGJIAN, ZHENG WEI, HUA JIN, YANG SHUGANG, ZHUANG JINFU, WANG JINZHOU. Receptor-interacting protein-1 promotes the growth and invasion in gastric cancer. Int J Oncol 2016; 48:2387-98. [DOI: 10.3892/ijo.2016.3455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/23/2016] [Indexed: 11/06/2022] Open
|
19
|
Ratovitski EA. Phospho-ΔNp63α-responsive microRNAs contribute to the regulation of necroptosis in squamous cell carcinoma upon cisplatin exposure. FEBS Lett 2015; 589:1352-8. [PMID: 25910754 DOI: 10.1016/j.febslet.2015.04.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 12/13/2022]
Abstract
This study shows that specific microRNAs differentially regulated by ΔNp63α in cisplatin-sensitive and resistant squamous cell carcinoma (SSC) cells of larynx and tongue affect the expression of members of the necroptotic pathway CYLD, RIPK1, and MLKL. Different degrees of protein interaction between necroptotic signaling intermediates were also observed in SCC cells sensitive or resistant to cisplatin. Modulation of RIPK1 with miR-101-3p mimic or inhibitor, as well as with siRNA, or chemical inhibitors was shown to affect sensitivity of SCC cells to cisplatin. This is the first report showing the modulatory effect of ΔNp63α-responsive microRNAs on the specific members of necroptotic pathway in SCC tumor cells variably responding to platinum chemotherapy.
Collapse
Affiliation(s)
- Edward A Ratovitski
- Head and Neck Cancer Research Division, Department of Otolaryngology/Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
20
|
Liu XY, Lai F, Yan XG, Jiang CC, Guo ST, Wang CY, Croft A, Tseng HY, Wilmott JS, Scolyer RA, Jin L, Zhang XD. RIP1 Kinase Is an Oncogenic Driver in Melanoma. Cancer Res 2015; 75:1736-48. [PMID: 25724678 DOI: 10.1158/0008-5472.can-14-2199] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/01/2015] [Indexed: 11/16/2022]
Abstract
Although many studies have uncovered an important role for the receptor-binding protein kinase RIP1 in controlling cell death signaling, its possible contributions to cancer pathogenesis have been little explored. Here, we report that RIP1 functions as an oncogenic driver in human melanoma. Although RIP1 was commonly upregulated in melanoma, RIP1 silencing inhibited melanoma cell proliferation in vitro and retarded the growth of melanoma xenografts in vivo. Conversely, while inducing apoptosis in a small proportion of melanoma cells, RIP1 overexpression enhanced proliferation in the remaining cells. Mechanistic investigations revealed that the proliferative effects of RIP1 overexpression were mediated by NF-κB activation. Strikingly, ectopic expression of RIP1 enhanced the proliferation of primary melanocytes, triggering their anchorage-independent cell growth in an NF-κB-dependent manner. We identified DNA copy-number gain and constitutive ubiquitination by a TNFα autocrine loop mechanism as two mechanisms of RIP1 upregulation in human melanomas. Collectively, our findings define RIP1 as an oncogenic driver in melanoma, with potential implications for targeting its NF-κB-dependent activation mechanism as a novel approach to treat this disease.
Collapse
Affiliation(s)
- Xiao Ying Liu
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia. School of Life Science, Anhui Medical University, Anhui, China
| | - Fritz Lai
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia
| | - Xu Guang Yan
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia
| | - Chen Chen Jiang
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia
| | - Su Tang Guo
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia. Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Shanxi, China
| | - Chun Yan Wang
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia. Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Shanxi, China
| | - Amanda Croft
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia
| | - Hsin-Yi Tseng
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia
| | - James S Wilmott
- Discipline of Pathology, The University of Sydney, and Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Discipline of Pathology, The University of Sydney, and Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Lei Jin
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia.
| | - Xu Dong Zhang
- School of Medicine and Public Health, The University of Newcastle, New South Wales, Australia.
| |
Collapse
|
21
|
Synergistic anticancer effect of cisplatin and Chal-24 combination through IAP and c-FLIPL degradation, Ripoptosome formation and autophagy-mediated apoptosis. Oncotarget 2015; 6:1640-51. [PMID: 25682199 PMCID: PMC4359321 DOI: 10.18632/oncotarget.2746] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/08/2014] [Indexed: 01/26/2023] Open
Abstract
Drug resistance is a major hurdle in anticancer chemotherapy. Combined therapy using drugs with distinct mechanisms of function may increase anticancer efficacy. We have recently identified the novel chalcone derivative, chalcone-24 (Chal-24), as a potential therapeutic that kills cancer cells through activation of an autophagy-mediated necroptosis pathway. In this report, we investigated if Chal-24 can be combined with the frontline genotoxic anticancer drug, cisplatin for cancer therapy. The combination of Chal-24 and cisplatin synergistically induced apoptotic cytotoxicity in lung cancer cell lines, which was dependent on Chal-24-induced autophagy. While cisplatin slightly potentiated the JNK/Bcl2/Beclin1 pathway for autophagy activation, its combination with Chal-24 strongly triggered proteasomal degradation of the cellular inhibitor of apoptosis proteins (c-IAPs) and formation of the Ripoptosome complex that contains RIP1, FADD and caspase 8. Furthermore, the cisplatin and Chal-24 combination induced dramatic degradation of cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein large (cFLIPL) which suppresses Ripoptosome-mediated apoptosis activation. These results establish a novel mechanism for potentiation of anticancer activity with the combination of Chal-24 and cisplatin: to enhance apoptosis signaling through Ripoptosome formation and to release the apoptosis brake through c-FLIPL degradation. Altogether, our work suggests that the combination of Chal-24 and cisplatin could be employed to improve chemotherapy efficacy.
Collapse
|
22
|
Cho YS, Park SY. Harnessing of Programmed Necrosis for Fighting against Cancers. Biomol Ther (Seoul) 2014; 22:167-75. [PMID: 25009696 PMCID: PMC4060077 DOI: 10.4062/biomolther.2014.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/11/2014] [Accepted: 05/12/2014] [Indexed: 12/17/2022] Open
Abstract
Chemotherapy has long been considered as one of useful strategies for cancer treatment. It is primarily based on the apoptosis that can selectively kill cancer cells. However, cancer cells can progressively develop an acquired resistance to apoptotic cell death, rendering refractory to chemo- and radiotherapies. Although the mechanism by which cells attained resistance to drug remains to be clarified, it might be caused by either pumping out of them or interfering with apoptotic signal cascades in response to cancer drugs. In case that cancer cells are defective in some part of apoptotic machinery by repeated exposure to anticancer drugs, alternative cell death mechanistically distinct from apoptosis could be adopted to remove cancer cells refractory to apoptosis-inducing agents. This review will mainly deal with harnessing of necrotic cell death, specifically, programmed necrosis and practical uses. Here, we begin with various defects of apoptotic death machinery in cancer cells, and then provide new perspective on programmed necrosis as an alternative anticancer approach.
Collapse
Affiliation(s)
- Young Sik Cho
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Seung Yeon Park
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| |
Collapse
|
23
|
He W, Wang Q, Srinivasan B, Xu J, Padilla MT, Li Z, Wang X, Liu Y, Gou X, Shen HM, Xing C, Lin Y. A JNK-mediated autophagy pathway that triggers c-IAP degradation and necroptosis for anticancer chemotherapy. Oncogene 2014; 33:3004-13. [PMID: 23831571 PMCID: PMC3912228 DOI: 10.1038/onc.2013.256] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 04/16/2013] [Accepted: 04/21/2013] [Indexed: 02/07/2023]
Abstract
Killing cancer cells through the induction of apoptosis is one of the main mechanisms of chemotherapy. However, numerous cancer cells have primary or acquired apoptosis resistance, resulting in chemoresistance. In this study, using a novel chalcone derivative chalcone-24 (Chal-24), we identified a novel anticancer mechanism through autophagy-mediated necroptosis (RIP1- and RIP3-dependent necrosis). Chal-24 potently killed different cancer cells with induction of necrotic cellular morphology while causing no detectable caspase activation. Blocking the necroptosis pathway with either necrostatin-1 or by knockdown of RIP1 and RIP3 effectively blocked the cytotoxicity of Chal-24, suggesting that Chal-24-induced cell death is associated with necroptosis. Chal-24 robustly activated JNK and ERK and blockage of which effectively suppressed Chal-24-induced cytotoxicity. In addition, Chal-24 strongly induced autophagy that is dependent on JNK-mediated phosphorylation of Bcl-2 and Bcl-xL and dissociation of Bcl-2 or Bcl-xL from Beclin-1. Importantly, suppression of autophagy, with either pharmacological inhibitors or small interfering RNAs targeting the essential autophagy components ATG7 and Beclin-1, effectively attenuated Chal-24-induced cell death. Furthermore, we found that autophagy activation resulted in c-IAP1 and c-IAP2 degradation and formation of the Ripoptosome that contributes to necroptosis. These results thus establish a novel mechanism for killing cancer cells that involves autophagy-mediated necroptosis, which may be employed for overcoming chemoresistance.
Collapse
Affiliation(s)
- Weiyang He
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM87108, USA
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Qiong Wang
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM87108, USA
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | | | - Jennings Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM87108, USA
| | - Mabel T. Padilla
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM87108, USA
| | - Zi Li
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM87108, USA
| | - Xia Wang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yushi Liu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM87108, USA
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN55455, USA
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM87108, USA
| |
Collapse
|
24
|
Wang D, Zhao M, Chen G, Cheng X, Han X, Lin S, Zhang X, Yu X. The histone deacetylase inhibitor vorinostat prevents TNFα-induced necroptosis by regulating multiple signaling pathways. Apoptosis 2014; 18:1348-1362. [PMID: 23708756 DOI: 10.1007/s10495-013-0866-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are novel anticancer reagents that have recently been reported to have anti-inflammatory and neuroprotective effects; however, the mechanisms underlying their activities are largely undefined. The data from this study show that the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) can protect L929 cells from TNFα-induced necroptosis. This effect involves multiple mechanisms, including the upregulation of cFLIPL expression, the enhanced activation of NFκB and p38 MAPK, and the inactivation of JNK. In addition, SAHA could initiate cell autophagy by inhibiting Akt and mTOR, which also play important roles in protecting cells from necroptosis. Because cell necroptosis is important for inflammation-related deterioration and neurodegenerative disease, our results indicate that preventing cell necrosis may be an important mechanism through which HDAC inhibitor compounds exert their anti-inflammatory or neuroprotective effects.
Collapse
Affiliation(s)
- Di Wang
- Department of Stress Medicine, Beijing Institute of Basic Medical Sciences, Cognitive and Mental Health Research Center, #27 Taiping Road, Beijing, 100850, China
| | - Ming Zhao
- Department of Stress Medicine, Beijing Institute of Basic Medical Sciences, Cognitive and Mental Health Research Center, #27 Taiping Road, Beijing, 100850, China
| | - Guozhu Chen
- Department of Stress Medicine, Beijing Institute of Basic Medical Sciences, Cognitive and Mental Health Research Center, #27 Taiping Road, Beijing, 100850, China
| | - Xiang Cheng
- Department of Stress Medicine, Beijing Institute of Basic Medical Sciences, Cognitive and Mental Health Research Center, #27 Taiping Road, Beijing, 100850, China
| | - Xiaoxi Han
- Department of Biochemistry, China Medical University, Shenyang, China
| | - Song Lin
- Department of Stress Medicine, Beijing Institute of Basic Medical Sciences, Cognitive and Mental Health Research Center, #27 Taiping Road, Beijing, 100850, China
| | - Xuhui Zhang
- Department of Stress Medicine, Beijing Institute of Basic Medical Sciences, Cognitive and Mental Health Research Center, #27 Taiping Road, Beijing, 100850, China
| | - Xiaodan Yu
- Department of Stress Medicine, Beijing Institute of Basic Medical Sciences, Cognitive and Mental Health Research Center, #27 Taiping Road, Beijing, 100850, China.
| |
Collapse
|
25
|
MacKay C, Carroll E, Ibrahim AFM, Garg A, Inman GJ, Hay RT, Alpi AF. E3 ubiquitin ligase HOIP attenuates apoptotic cell death induced by cisplatin. Cancer Res 2014; 74:2246-2257. [PMID: 24686174 DOI: 10.1158/0008-5472.can-13-2131] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The genotoxin cisplatin is commonly used in chemotherapy to treat solid tumors, yet our understanding of the mechanism underlying the drug response is limited. In a focused siRNA screen, using an siRNA library targeting genes involved in ubiquitin and ubiquitin-like signaling, we identified the E3 ubiquitin ligase HOIP as a key regulator of cisplatin-induced genotoxicity. HOIP forms, with SHARPIN and HOIL-1L, the linear ubiquitin assembly complex (LUBAC). We show that cells deficient in the HOIP ligase complex exhibit hypersensitivity to cisplatin. This is due to a dramatic increase in caspase-8/caspase-3-mediated apoptosis that is strictly dependent on ATM-, but not ATR-mediated DNA damage checkpoint activation. Moreover, basal and cisplatin-induced activity of the stress response kinase JNK is enhanced in HOIP-depleted cells and, conversely, JNK inhibition can increase cellular resistance to cisplatin and reverse the apoptotic hyperactivation in HOIP-depleted cells. Furthermore, we show that HOIP depletion sensitizes cancer cells, derived from carcinomas of various origins, through an enhanced apoptotic cell death response. We also provide evidence that ovarian cancer cells classified as cisplatin-resistant can regain sensitivity following HOIP downregulation. Cumulatively, our study identifies a HOIP-regulated antiapoptotic signaling pathway, and we envisage HOIP as a potential target for the development of combinatorial chemotherapies to potentiate the efficacy of platinum-based anticancer drugs.
Collapse
Affiliation(s)
- Craig MacKay
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Science, University of Dundee, UK
| | - Eilís Carroll
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Science, University of Dundee, UK
| | - Adel F M Ibrahim
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Science, University of Dundee, UK
| | - Amit Garg
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Science, University of Dundee, UK
| | - Gareth J Inman
- Division of Cancer Research, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Ronald T Hay
- Wellcome Trust Centre for Gene Regulation and Expression, University of Dundee, UK
| | - Arno F Alpi
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Science, University of Dundee, UK
| |
Collapse
|
26
|
Inhibitor of apoptosis proteins as therapeutic targets in multiple myeloma. Leukemia 2014; 28:1519-28. [PMID: 24402161 PMCID: PMC4090267 DOI: 10.1038/leu.2014.2] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 12/20/2013] [Indexed: 12/26/2022]
Abstract
The inhibitor of apoptosis (IAP) proteins plays a critical role in the control of apoptotic machinery, and has been explored as a therapeutic target. Here, we have examined the functional importance of IAPs in multiple myeloma (MM) by using a Smac-mimetic LCL161. We observed that LCL161 was able to potently induce apoptosis in some MM cell lines but not in others. Examining the levels of XIAP, cIAP1 and cIAP2 post LCL161 treatment indicated clear down regulation of both XIAP activity and cIAP1 levels in both the sensitive and less sensitive (resistant) cell lines. cIAP2, however, was not down regulated in the cell line resistant to the drug. siRNA mediated silencing of cIAP2 significantly enhanced the effect of LCL161 indicating the importance of down regulating all IAPs simultaneously for induction of apopotsis in MM cells. LCL161 induced marked up regulation of the Jak2/Stat3 pathway in the resistant MM cell lines. Combining LCL161 with a Jak2 specific inhibitor resulted in synergistic cell death in MM cell lines and patient cells. In addition, combining LCL161 with death inducing ligands clearly showed that LCL161 sensitized MM cells to both FAS-L and TRAIL.
Collapse
|
27
|
Carter SR, Zahs A, Palmer JL, Wang L, Ramirez L, Gamelli RL, Kovacs EJ. Intestinal barrier disruption as a cause of mortality in combined radiation and burn injury. Shock 2013; 40:281-9. [PMID: 24045418 PMCID: PMC3780610 DOI: 10.1097/shk.0b013e3182a2c5b5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nuclear disaster associated with combined radiation injury (CRI) and trauma or burns results in higher mortality than component injuries. Early death is caused by sequelae of gastrointestinal (GI) leakiness such as bacterial translocation and shock. We developed a murine model to characterize GI injury after CRI and determine the extent of barrier disruption. Animals received radiation (5.5 Gy) alone or with 15% total body surface area (TBSA) scald burn and were euthanized at 24, 48, and 72 h. Mesenteric lymph node homogenate was plated on tryptic soy agar to assess for bacterial translocation. Tight junction protein, occludin, was characterized by Western blot and immunofluorescence. Intestinal histology was evaluated, and apoptosis was quantified using histone-associated DNA fragmentation enzyme-linked immunosorbent assay and Western blot for caspase-3 and caspase-8. At 72 h, a 100-fold increase in bacterial growth after CRI was observed. Occludin colocalization was reduced by radiation exposure, with largest differences in CRI at 24 and 48 h. Histopathology exhibited increased apoptosis in radiation alone and CRI animals at 24 and 48 h (P < 0.05). Further evidence of apoptotic activity in CRI was seen at 48 h, with 3-fold increases in enzyme-linked immunosorbent assay detection relative to all groups and caspase-8 activity relative to radiation alone and sham (P < 0.05). Prolonged epithelial apoptosis and disruption of tight junctions likely contribute to gut leakiness after CRI. Subsequent bacterial translocation to mesenteric lymph node potentially leads to sepsis and death and could serve as a target for mitigating agents to improve survival from CRI.
Collapse
Affiliation(s)
- Stewart R Carter
- *Burn and Shock Trauma Research Institute, and †Departments of Surgery, ‡Pathology, and §Microbiology and Immunology, Loyola University Chicago Health Sciences Division, Maywood, Illinois
| | | | | | | | | | | | | |
Collapse
|
28
|
Zhou Y, Dai W, Lin C, Wang F, He L, Shen M, Chen P, Wang C, Lu J, Xu L, Xu X, Guo C. Protective effects of necrostatin-1 against concanavalin A-induced acute hepatic injury in mice. Mediators Inflamm 2013; 2013:706156. [PMID: 24198446 PMCID: PMC3806455 DOI: 10.1155/2013/706156] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/15/2013] [Accepted: 08/14/2013] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Necrostatin-1 (Nec-1) inhibits receptor-interacting protein 1 (RIP1) kinase and programmed necrosis. This study was designed to examine the protective effects and mechanisms of Nec-1 in concanavalin A- (ConA-) induced hepatitis in mice. METHODS C57BL/6 mice were exposed to ConA via tail vein injection and injected intraperitoneally with Nec-1 or vehicle. Levels of serum liver enzymes and histopathology were determined. Levels of inflammatory cytokines with ConA-induced hepatitis were determined with real-time polymerase chain reaction (real-time PCR). The expression of TNF- α , RIP1, and LC3 was detected with immunohistochemical staining. The expression of TNF- α , IFN- γ , IL2, IL6, caspase 3, RIP1, beclin-1, and LC3 protein was assessed by immunofluorescence and western blotting. Autophagosomes were observed with transmission electron microscopy (TEM). RESULTS Amelioration in liver functions and histopathological changes and the suppression of inflammatory cytokine production were observed in Nec-1-injected mice. Western blotting analysis showed that the expression of TNF- α , IFN- γ , IL2, IL6, and RIP1 was significantly reduced in the Nec-1-injected mice, which was confirmed by immunofluorescence and immunohistochemistry. Autophagosome formation was significantly reduced by Nec-1 treatment, as the expression of beclin-1 and LC3, determined with immunofluorescence and western blotting. CONCLUSION These results demonstrate that Nec-1 prevents ConA-induced liver injury via RIP1-related and autophagy-related pathways.
Collapse
Affiliation(s)
- Yingqun Zhou
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chunlei Lin
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Fan Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Lei He
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Miao Shen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ping Chen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chenfen Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ling Xu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Xuanfu Xu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| |
Collapse
|
29
|
Roscioli E, Hamon R, Lester S, Murgia C, Grant J, Zalewski P. Zinc-rich inhibitor of apoptosis proteins (IAPs) as regulatory factors in the epithelium of normal and inflamed airways. Biometals 2013; 26:205-27. [PMID: 23460081 DOI: 10.1007/s10534-013-9618-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 02/25/2013] [Indexed: 11/25/2022]
Abstract
Integrity of the airway epithelium (AE) is important in the context of inhaled allergens and noxious substances, particularly during asthma-related airway inflammation where there is increased vulnerability of the AE to cell death. Apoptosis involves a number of signaling pathways which activate procaspases leading to cleavage of critical substrates. Understanding the factors which regulate AE caspases is important for development of strategies to minimize AE damage and airway inflammation, and therefore to better control asthma. One such factor is the essential dietary metal zinc. Zinc deficiency results in enhanced AE apoptosis, and worsened airway inflammation. This has implications for asthma, where abnormalities in zinc homeostasis have been observed. Zinc is thought to suppress the steps involved in caspase-3 activation. One target of zinc is the family of inhibitor of apoptosis proteins (IAPs) which are endogenous regulators of caspases. More studies are needed to identify the roles of IAPs in regulating apoptosis in normal and inflamed airways and to study their interaction with labile zinc ions. This new information will provide a framework for future clinical studies aimed at monitoring and management of airway zinc levels as well as minimising airway damage and inflammation in asthma.
Collapse
Affiliation(s)
- Eugene Roscioli
- Discipline of Medicine, The Basil Hetzel Institute for Translational Research, The Queen Elizabeth Hospital, University of Adelaide, Woodville, SA, 5011, Australia.
| | | | | | | | | | | |
Collapse
|
30
|
Vérillaud B, Gressette M, Morel Y, Paturel C, Herman P, Lo KW, Tsao SW, Wassef M, Jimenez-Pailhes AS, Busson P. Toll-like receptor 3 in Epstein-Barr virus-associated nasopharyngeal carcinomas: consistent expression and cytotoxic effects of its synthetic ligand poly(A:U) combined to a Smac-mimetic. Infect Agent Cancer 2012. [PMID: 23198710 PMCID: PMC3599303 DOI: 10.1186/1750-9378-7-36] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Nasopharyngeal carcinomas (NPC) are consistently associated with the Epstein-Barr virus (EBV). Though NPCs are more radiosensitive and chemosensitive than other tumors of the upper aero-digestive tract, many therapeutic challenges remain. In a previous report, we have presented data supporting a possible therapeutic strategy based on artificial TLR3 stimulation combined to the inhibition of the IAP protein family (Inhibitor of Apoptosis Proteins). The present study was designed to progress towards practical applications of this strategy pursuing 2 main objectives: 1) to formally demonstrate expression of the TLR3 protein by malignant NPC cells; 2) to investigate the effect of poly(A:U) as a novel TLR3-agonist more specific than poly(I:C) which was used in our previous study. Methods TLR3 expression was investigated in a series of NPC cell lines and clinical specimens by Western blot analysis and immunohistochemistry, respectively. The effects on NPC cells growth of the TLR3 ligand poly(A:U) used either alone or in combination with RMT5265, an IAP inhibitor based on Smac-mimicry, were assessed using MTT assays and clonogenic assays. Results TLR3 was detected at a high level in all NPC cell lines and clinical specimens. Low concentrations of poly(A:U) were applied to several types of NPC cells including cells from the C17 xenograft which for the first time have been adapted to permanent propagation in vitro. As a single agent, poly(A:U) had no significant effects on cell growth and cell survival. In contrast, dramatic effects were obtained when it was combined with the IAP inhibitor RMT5265. These effects were obtained using concentrations as low as 0.5 μg/ml (poly(A:U)) and 50 nM (RMT5265). Conclusion These data confirm that TLR3 expression is a factor of vulnerability for NPC cells. They suggest that in some specific pathological and pharmacological contexts, it might be worth to use Smac-mimetics at very low doses, allowing a better management of secondary effects. In light of our observations, combined use of both types of compounds should be considered for treatment of nasopharyngeal carcinomas.
Collapse
Affiliation(s)
- Benjamin Vérillaud
- CNRS-UMR 8126, Institut de Cancérologie Gustave Roussy, University Paris-Sud 11, 39 rue Camille Desmoulins, 94805, Villejuif cedex, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kenneth NS, Duckett CS. IAP proteins: regulators of cell migration and development. Curr Opin Cell Biol 2012; 24:871-5. [DOI: 10.1016/j.ceb.2012.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 11/16/2012] [Accepted: 11/19/2012] [Indexed: 10/27/2022]
|
32
|
Dunai ZA, Imre G, Barna G, Korcsmaros T, Petak I, Bauer PI, Mihalik R. Staurosporine induces necroptotic cell death under caspase-compromised conditions in U937 cells. PLoS One 2012; 7:e41945. [PMID: 22860037 PMCID: PMC3409216 DOI: 10.1371/journal.pone.0041945] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 06/27/2012] [Indexed: 11/25/2022] Open
Abstract
For a long time necrosis was thought to be an uncontrolled process but evidences recently have revealed that necrosis can also occur in a regulated manner. Necroptosis, a type of programmed necrosis is defined as a death receptor-initiated process under caspase-compromised conditions. The process requires the kinase activity of receptor-interacting protein kinase 1 and 3 (RIPK1 and RIPK3) and mixed lineage kinase domain-like protein (MLKL), as a substrate of RIPK3. The further downstream events remain elusive. We applied known inhibitors to characterize the contributing enzymes in necroptosis and their effect on cell viability and different cellular functions were detected mainly by flow cytometry. Here we report that staurosporine, the classical inducer of intrinsic apoptotic pathway can induce necroptosis under caspase-compromised conditions in U937 cell line. This process could be hampered at least partially by the RIPK1 inhibitor necrotstin-1 and by the heat shock protein 90 kDa inhibitor geldanamycin. Moreover both the staurosporine-triggered and the classical death ligand-induced necroptotic pathway can be effectively arrested by a lysosomal enzyme inhibitor CA-074-OMe and the recently discovered MLKL inhibitor necrosulfonamide. We also confirmed that the enzymatic role of poly(ADP-ribose)polymerase (PARP) is dispensable in necroptosis but it contributes to membrane disruption in secondary necrosis. In conclusion, we identified a novel way of necroptosis induction that can facilitate our understanding of the molecular mechanisms of necroptosis. Our results shed light on alternative application of staurosporine, as a possible anticancer therapeutic agent. Furthermore, we showed that the CA-074-OMe has a target in the signaling pathway leading to necroptosis. Finally, we could differentiate necroptotic and secondary necrotic processes based on participation of PARP enzyme.
Collapse
Affiliation(s)
- Zsuzsanna A Dunai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
| | | | | | | | | | | | | |
Collapse
|
33
|
Bacterium-generated nitric oxide hijacks host tumor necrosis factor alpha signaling and modulates the host cell cycle in vitro. J Bacteriol 2012; 194:4059-68. [PMID: 22636782 DOI: 10.1128/jb.00476-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In mammalian cells, nitric oxide (NO·) is an important signal molecule with concentration-dependent and often controversial functions of promoting cell survival and inducing cell death. An inducible nitric oxide synthase (iNOS) in various mammalian cells produces higher levels of NO· from l-arginine upon infections to eliminate pathogens. In this study, we reveal novel pathogenic roles of NO· generated by bacteria in bacterium-host cell cocultures using Moraxella catarrhalis, a respiratory tract disease-causing bacterium, as a biological producer of NO·. We recently demonstrated that M. catarrhalis cells that express the nitrite reductase (AniA protein) can produce NO· by reducing nitrite. Our study suggests that, in the presence of pathophysiological levels of nitrite, this opportunistic pathogen hijacks host cell signaling and modulates host gene expression through its ability to produce NO· from nitrite. Bacterium-generated NO· significantly increases the secretion of tumor necrosis factor alpha (TNF-α) and modulates the expression of apoptotic proteins, therefore triggering host cell programmed death partially through TNF-α signaling. Furthermore, our study reveals that bacterium-generated NO· stalls host cell division and directly results in the death of dividing cells by reducing the levels of an essential regulator of cell division. This study provides unique insight into why NO· may exert more severe cytotoxic effects on fast growing cells, providing an important molecular basis for NO·-mediated pathogenesis in infections and possible therapeutic applications of NO·-releasing molecules in tumorigenesis. This study strongly suggests that bacterium-generated NO· can play important pathogenic roles during infections.
Collapse
|