1
|
Zhu W, Pan S, Zhang J, Xu J, Zhang R, Zhang Y, Fu Z, Wang Y, Hu C, Xu Z. The role of hyperthermia in the treatment of tumor. Crit Rev Oncol Hematol 2024; 204:104541. [PMID: 39461607 DOI: 10.1016/j.critrevonc.2024.104541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
Despite recent advancements in the diagnosis and treatment options for cancer, it remains one of the most serious threats to health. Hyperthermia (HT) has emerged as a highly promising area of research due to its safety and cost-effectiveness. Currently, based on temperature, HT can be categorized into thermal ablation and mild hyperthermia. Thermal ablation involves raising the temperature within the tumor to over 60°C, resulting in direct necrosis in the central region of the tumor. In contrast, mild hyperthermia operates at relatively lower temperatures, typically in the range of 41-45°C, to induce damage to tumor cells. Furthermore, HT also serves as an immune adjuvant strategy in radiotherapy, chemotherapy, and immunotherapy, enhancing the effectiveness of radiotherapy, increasing the uptake of chemotherapy drugs, and reprogramming the tumor microenvironment through the induction of immunogenic cell death, thereby promoting the recruitment of endogenous immune cells. This article reviews the current status and development of hyperthermia, outlines potential mechanisms by which hyperthermia inhibits tumors, describes clinical trial attempts combining hyperthermia with radiotherapy, chemotherapy, and immunotherapy, and discusses the relationship between nanoparticles and hyperthermia.
Collapse
Affiliation(s)
- Weiwei Zhu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Siwei Pan
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jiaqing Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jingli Xu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ruolan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yanqiang Zhang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhenjie Fu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yuqi Wang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Can Hu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| | - Zhiyuan Xu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| |
Collapse
|
2
|
Shah RB, Li Y, Yu H, Kini E, Sidi S. Stepwise phosphorylation and SUMOylation of PIDD1 drive PIDDosome assembly in response to DNA repair failure. Nat Commun 2024; 15:9195. [PMID: 39448602 PMCID: PMC11502896 DOI: 10.1038/s41467-024-53412-0] [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: 06/05/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
SUMOylation regulates numerous cellular stress responses, yet targets in the apoptotic machinery remain elusive. We show that a single, DNA damage-induced monoSUMOylation event controls PIDDosome (PIDD1/RAIDD/caspase-2) formation and apoptotic death in response to unresolved DNA interstrand crosslinks (ICLs). SUMO-1 conjugation occurs on conserved K879 in the PIDD1 death domain (DD); is catalyzed by PIAS1 and countered by SENP3; and is triggered by ATR phosphorylation of neighboring T788 in the PIDD1 DD, which enables PIAS1 docking. Phospho/SUMO-PIDD1 proteins are captured by nucleolar RAIDD monomers via a SUMO-interacting motif (SIM) in the RAIDD DD, thus compartmentalizing nascent PIDDosomes for caspase-2 recruitment. Denying SUMOylation or the SUMO-SIM interaction spares the onset of PIDDosome assembly but blocks its completion, thus eliminating the apoptotic response to ICL repair failure. Conversely, removal of SENP3 forces apoptosis, even in cells with tolerable ICL levels. SUMO-mediated PIDDosome control is also seen in response to DNA breaks but not supernumerary centrosomes. These results illuminate PIDDosome formation in space and time and identify a direct role for SUMOylation in the assembly of a major pro-apoptotic device.
Collapse
Affiliation(s)
- Richa B Shah
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuanyuan Li
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Honglin Yu
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ela Kini
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samuel Sidi
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
3
|
Chang C, Tang X, Woodley DT, Chen M, Li W. Previously unrecognized and potentially consequential challenges facing Hsp90 inhibitors in cancer clinical trials. Cell Stress Chaperones 2024; 29:642-653. [PMID: 39181529 PMCID: PMC11402043 DOI: 10.1016/j.cstres.2024.08.002] [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: 05/27/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
Targeting the heat shock protein-90 (Hsp90) chaperone machinery in various cancers with 200 monotherapy or combined-therapy clinical trials since 1999 has not yielded any success of food and drug administration approval. Blames for the failures were unanimously directed at the Hsp90 inhibitors or tumors or both. However, analyses of recent cellular and genetic studies together with the Hsp90 data from the Human Protein Atlas database suggest that the vast variations in Hsp90 expression among different organs in patients might have been the actual cause. It is evident now that Hsp90β is the root of dose-limiting toxicity (DLT), whereas Hsp90α is a buffer of penetrated Hsp90 inhibitors. The more Hsp90α, the safer Hsp90β, and the lower DLT are for the host. Unfortunately, the dramatic variations of Hsp90, from total absence in the eye, muscle, pancreas, and heart to abundance in reproduction organs, lung, liver, and gastrointestinal track, would cause the selection of any fair toxicity biomarker and an effective maximum tolerable dose (MTD) of Hsp90 inhibitor extremely challenging. In theory, a safe MTD for the organs with high Hsp90 could harm the organs with low Hsp90. In reverse, a safe MTD for organs with low or undetectable Hsp90 would have little impact on the tumors, whose cells exhibit average 3-7% Hsp90 over the average 2-3% Hsp90 in normal cells. Moreover, not all tumor cell lines tested follow the "inhibitor binding-client protein degradation" paradigm. It is likely why the oral Hsp90 inhibitor TAS-116 (Pimitespib), which bypasses blood circulation and other organs, showed some beneficiary efficacy by conveniently hitting tumors along the gastrointestinal track. The critical question is what the next step will be for the Hsp90 chaperone as a cancer therapeutic target.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - Xin Tang
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - David T Woodley
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - Mei Chen
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - Wei Li
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA.
| |
Collapse
|
4
|
Sakthivel D, Brown-Suedel AN, Lopez KE, Salgar S, Coutinho LE, Keane F, Huang S, Sherry KM, Charendoff CI, Dunne KP, Robichaux DJ, Vargas-Hernández A, Le B, Shin CS, Carisey AF, Poreba M, Flanagan JM, Bouchier-Hayes L. Caspase-2 is essential for proliferation and self-renewal of nucleophosmin-mutated acute myeloid leukemia. SCIENCE ADVANCES 2024; 10:eadj3145. [PMID: 39093977 PMCID: PMC11296348 DOI: 10.1126/sciadv.adj3145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
Mutation in nucleophosmin (NPM1) causes relocalization of this normally nucleolar protein to the cytoplasm (NPM1c+). Despite NPM1 mutation being the most common driver mutation in cytogenetically normal adult acute myeloid leukemia (AML), the mechanisms of NPM1c+-induced leukemogenesis remain unclear. Caspase-2 is a proapoptotic protein activated by NPM1 in the nucleolus. Here, we show that caspase-2 is also activated by NPM1c+ in the cytoplasm and DNA damage-induced apoptosis is caspase-2 dependent in NPM1c+ but not in NPM1wt AML cells. Strikingly, in NPM1c+ cells, caspase-2 loss results in profound cell cycle arrest, differentiation, and down-regulation of stem cell pathways that regulate pluripotency including impairment of the AKT/mTORC1 pathways, and inhibition of Rictor cleavage. In contrast, there were minimal differences in proliferation, differentiation, or the transcriptional profile of NPM1wt cells lacking caspase-2. Our results show that caspase-2 is essential for proliferation and self-renewal of AML cells expressing mutated NPM1. This study demonstrates that caspase-2 is a major effector of NPM1c+ function.
Collapse
Affiliation(s)
- Dharaniya Sakthivel
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandra N. Brown-Suedel
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Karla E. Lopez
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Suruchi Salgar
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Luiza E. Coutinho
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Francesca Keane
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shixia Huang
- Advanced Technology Cores, Department of Molecular and Cellular Biology, Huffington Department of Education, Innovation & Technology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kenneth Mc Sherry
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chloé I. Charendoff
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin P. Dunne
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dexter J. Robichaux
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander Vargas-Hernández
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - BaoChau Le
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Crystal S. Shin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre F. Carisey
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Marcin Poreba
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw 50370, Poland
| | - Jonathan M. Flanagan
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Lisa Bouchier-Hayes
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Shkarina K, Broz P. Selective induction of programmed cell death using synthetic biology tools. Semin Cell Dev Biol 2024; 156:74-92. [PMID: 37598045 DOI: 10.1016/j.semcdb.2023.07.012] [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: 05/05/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/21/2023]
Abstract
Regulated cell death (RCD) controls the removal of dispensable, infected or malignant cells, and is thus essential for development, homeostasis and immunity of multicellular organisms. Over the last years different forms of RCD have been described (among them apoptosis, necroptosis, pyroptosis and ferroptosis), and the cellular signaling pathways that control their induction and execution have been characterized at the molecular level. It has also become apparent that different forms of RCD differ in their capacity to elicit inflammation or an immune response, and that RCD pathways show a remarkable plasticity. Biochemical and genetic studies revealed that inhibition of a given pathway often results in the activation of back-up cell death mechanisms, highlighting close interconnectivity based on shared signaling components and the assembly of multivalent signaling platforms that can initiate different forms of RCD. Due to this interconnectivity and the pleiotropic effects of 'classical' cell death inducers, it is challenging to study RCD pathways in isolation. This has led to the development of tools based on synthetic biology that allow the targeted induction of RCD using chemogenetic or optogenetic methods. Here we discuss recent advances in the development of such toolset, highlighting their advantages and limitations, and their application for the study of RCD in cells and animals.
Collapse
Affiliation(s)
- Kateryna Shkarina
- Institute of Innate Immunity, University Hospital Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Switzerland.
| |
Collapse
|
6
|
Dawar S, Benitez MC, Lim Y, Dite TA, Yousef JM, Thio N, Garciaz S, Jackson TD, Milne JV, Dagley LF, Phillips WA, Kumar S, Clemons NJ. Caspase-2 protects against ferroptotic cell death. Cell Death Dis 2024; 15:182. [PMID: 38429264 PMCID: PMC10907636 DOI: 10.1038/s41419-024-06560-6] [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: 01/05/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 03/03/2024]
Abstract
Caspase-2, one of the most evolutionarily conserved members of the caspase family, is an important regulator of the cellular response to oxidative stress. Given that ferroptosis is suppressed by antioxidant defense pathways, such as that involving selenoenzyme glutathione peroxidase 4 (GPX4), we hypothesized that caspase-2 may play a role in regulating ferroptosis. This study provides the first demonstration of an important and unprecedented function of caspase-2 in protecting cancer cells from undergoing ferroptotic cell death. Specifically, we show that depletion of caspase-2 leads to the downregulation of stress response genes including SESN2, HMOX1, SLC7A11, and sensitizes mutant-p53 cancer cells to cell death induced by various ferroptosis-inducing compounds. Importantly, the canonical catalytic activity of caspase-2 is not required for its role and suggests that caspase-2 regulates ferroptosis via non-proteolytic interaction with other proteins. Using an unbiased BioID proteomics screen, we identified novel caspase-2 interacting proteins (including heat shock proteins and co-chaperones) that regulate cellular responses to stress. Finally, we demonstrate that caspase-2 limits chaperone-mediated autophagic degradation of GPX4 to promote the survival of mutant-p53 cancer cells. In conclusion, we document a novel role for caspase-2 as a negative regulator of ferroptosis in cells with mutant p53. Our results provide evidence for a novel function of caspase-2 in cell death regulation and open potential new avenues to exploit ferroptosis in cancer therapy.
Collapse
Affiliation(s)
- Swati Dawar
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Mariana C Benitez
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Yoon Lim
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
| | - Toby A Dite
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jumana M Yousef
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Niko Thio
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sylvain Garciaz
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Thomas D Jackson
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Julia V Milne
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Laura F Dagley
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Wayne A Phillips
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Surgery (St Vincent's Hospital), The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5000, Australia
| | - Nicholas J Clemons
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
7
|
Amason ME, Li L, Harvest CK, Lacey CA, Miao EA. Validation of the Intermolecular Disulfide Bond in Caspase-2. BIOLOGY 2024; 13:49. [PMID: 38248479 PMCID: PMC10813798 DOI: 10.3390/biology13010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Caspases are a family of proteins involved in cell death. Although several caspase members have been well characterized, caspase-2 remains enigmatic. Caspase-2 has been implicated in several phenotypes, but there has been no consensus in the field about its upstream activating signals or its downstream protein targets. In addition, the unique ability of caspase-2 to form a disulfide-bonded dimer has not been studied in depth. Herein, we investigate the disulfide bond in the context of inducible dimerization, showing that disulfide bond formation is dimerization dependent. We also explore and review several stimuli published in the caspase-2 field, test ferroptosis-inducing stimuli, and study in vivo infection models. We hypothesize that the disulfide bond will ultimately prove to be essential for the evolved function of caspase-2. Proving this will require the discovery of cell death phenotypes where caspase-2 is definitively essential.
Collapse
Affiliation(s)
- Megan E. Amason
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lupeng Li
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Carissa K. Harvest
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Carolyn A. Lacey
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Edward A. Miao
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
8
|
Li Y, Shah RB, Sarti S, Belcher AL, Lee BJ, Gorbatenko A, Nemati F, Yu H, Stanley Z, Rahman M, Shao Z, Silva JM, Zha S, Sidi S. A noncanonical IRAK4-IRAK1 pathway counters DNA damage-induced apoptosis independently of TLR/IL-1R signaling. Sci Signal 2023; 16:eadh3449. [PMID: 38113335 PMCID: PMC11111193 DOI: 10.1126/scisignal.adh3449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
Interleukin-1 receptor (IL-1R)-associated kinases (IRAKs) are core effectors of Toll-like receptors (TLRs) and IL-1R in innate immunity. Here, we found that IRAK4 and IRAK1 together inhibited DNA damage-induced cell death independently of TLR or IL-1R signaling. In human cancer cells, IRAK4 was activated downstream of ATR kinase in response to double-strand breaks (DSBs) induced by ionizing radiation (IR). Activated IRAK4 then formed a complex with and activated IRAK1. The formation of this complex required the E3 ubiquitin ligase Pellino1, acting structurally but not catalytically, and the activation of IRAK1 occurred independently of extracellular signaling, intracellular TLRs, and the TLR/IL-1R signaling adaptor MyD88. Activated IRAK1 translocated to the nucleus in a Pellino2-dependent manner. In the nucleus, IRAK1 bound to the PIDD1 subunit of the proapoptotic PIDDosome and interfered with platform assembly, thus supporting cell survival. This noncanonical IRAK signaling pathway was also activated in response to other DSB-inducing agents. The loss of IRAK4, of IRAK4 kinase activity, of either Pellino protein, or of the nuclear localization sequence in IRAK1 sensitized p53-mutant zebrafish to radiation. Thus, the findings may lead to strategies for overcoming tumor resistance to conventional cancer treatments.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Richa B. Shah
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Samanta Sarti
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alicia L. Belcher
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian J. Lee
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Andrej Gorbatenko
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Current address: Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Francesca Nemati
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Honglin Yu
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zoe Stanley
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mahbuba Rahman
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhengping Shao
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Jose M. Silva
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shan Zha
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Division of Pediatric Oncology, Hematology and Stem Cell Transplantation, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Samuel Sidi
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
9
|
Volik PI, Kopeina GS, Zhivotovsky B, Zamaraev AV. Total recall: the role of PIDDosome components in neurodegeneration. Trends Mol Med 2023; 29:996-1013. [PMID: 37716905 DOI: 10.1016/j.molmed.2023.08.008] [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: 07/12/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/18/2023]
Abstract
The PIDDosome is a multiprotein complex that includes p53-induced protein with a death domain 1 (PIDD1), receptor-interacting protein-associated ICH-1/CED-3 homologous protein with a death domain (RAIDD), and caspase-2, the activation of which is driven by PIDDosome assembly. In addition to the key role of the PIDDosome in the regulation of cell differentiation, tissue homeostasis, and organogenesis and regeneration, caspase-2, RAIDD and PIDD1 engagement in neuronal development was shown. Here, we focus on the involvement of PIDDosome components in neurodegenerative disorders, including retinal neuropathies, different types of brain damage, and Alzheimer's disease (AD), Huntington's disease (HD), and Lewy body disease. We also discuss pathogenic variants of PIDD1, RAIDD, and caspase-2 that are associated with intellectual, behavioral, and psychological abnormalities, together with prospective PIDDosome inhibition strategies and their potential clinical application.
Collapse
Affiliation(s)
- Pavel I Volik
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia
| | - Gelina S Kopeina
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia
| | - Boris Zhivotovsky
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia; Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden.
| | - Alexey V Zamaraev
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia.
| |
Collapse
|
10
|
Sahoo G, Samal D, Khandayataray P, Murthy MK. A Review on Caspases: Key Regulators of Biological Activities and Apoptosis. Mol Neurobiol 2023; 60:5805-5837. [PMID: 37349620 DOI: 10.1007/s12035-023-03433-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 06/06/2023] [Indexed: 06/24/2023]
Abstract
Caspases are proteolytic enzymes that belong to the cysteine protease family and play a crucial role in homeostasis and programmed cell death. Caspases have been broadly classified by their known roles in apoptosis (caspase-3, caspase-6, caspase-7, caspase-8, and caspase-9 in mammals) and in inflammation (caspase-1, caspase-4, caspase-5, and caspase-12 in humans, and caspase-1, caspase-11, and caspase-12 in mice). Caspases involved in apoptosis have been subclassified by their mechanism of action as either initiator caspases (caspase-8 and caspase-9) or executioner caspases (caspase-3, caspase-6, and caspase-7). Caspases that participate in apoptosis are inhibited by proteins known as inhibitors of apoptosis (IAPs). In addition to apoptosis, caspases play a role in necroptosis, pyroptosis, and autophagy, which are non-apoptotic cell death processes. Dysregulation of caspases features prominently in many human diseases, including cancer, autoimmunity, and neurodegenerative disorders, and increasing evidence shows that altering caspase activity can confer therapeutic benefits. This review covers the different types of caspases, their functions, and their physiological and biological activities and roles in different organisms.
Collapse
Affiliation(s)
- Gayatri Sahoo
- Department of Zoology, PSSJ College, Banarpal, 759128, Odisha, India
| | - Dibyaranjan Samal
- Department of Biotechnology, Academy of Management and Information Technology (AMIT, affiliated to Utkal University), Khurda, 752057, Odisha, India
| | | | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
11
|
Sakthivel D, Brown-Suedel AN, Keane F, Huang S, Sherry KM, Charendoff CI, Dunne KP, Robichaux DJ, Le B, Shin CS, Carisey AF, Flanagan JM, Bouchier-Hayes L. Caspase-2 is essential for proliferation and self-renewal of nucleophosmin-mutated acute myeloid leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542723. [PMID: 37398413 PMCID: PMC10312440 DOI: 10.1101/2023.05.29.542723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Mutation in nucleophosmin (NPM1) causes relocalization of this normally nucleolar protein to the cytoplasm ( NPM1c+ ). Despite NPM1 mutation being the most common driver mutation in cytogenetically normal adult acute myeloid leukemia (AML), the mechanisms of NPM1c+-induced leukemogenesis remain unclear. Caspase-2 is a pro-apoptotic protein activated by NPM1 in the nucleolus. Here, we show that caspase-2 is also activated by NPM1c+ in the cytoplasm, and DNA damage-induced apoptosis is caspase-2-dependent in NPM1c+ AML but not in NPM1wt cells. Strikingly, in NPM1c+ cells, loss of caspase-2 results in profound cell cycle arrest, differentiation, and down-regulation of stem cell pathways that regulate pluripotency including impairment in the AKT/mTORC1 and Wnt signaling pathways. In contrast, there were minimal differences in proliferation, differentiation, or the transcriptional profile of NPM1wt cells with and without caspase-2. Together, these results show that caspase-2 is essential for proliferation and self-renewal of AML cells that have mutated NPM1. This study demonstrates that caspase-2 is a major effector of NPM1c+ function and may even be a druggable target to treat NPM1c+ AML and prevent relapse.
Collapse
|
12
|
Li Y, Shah RB, Sarti S, Belcher AL, Lee BJ, Gorbatenko A, Nemati F, Yu I, Stanley Z, Shao Z, Silva JM, Zha S, Sidi S. A Non-Canonical IRAK Signaling Pathway Triggered by DNA Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527716. [PMID: 36798275 PMCID: PMC9934671 DOI: 10.1101/2023.02.08.527716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Interleukin-1 receptor (IL-1R)-associated kinases (IRAKs) are core effectors of Toll-like receptor (TLR) and IL-1R signaling, with no reported roles outside of innate immunity. We find that vertebrate cells exposed to ionizing radiation (IR) sequentially activate IRAK4 and IRAK1 through a phosphorylation cascade mirroring that induced by TLR/IL-1R, resulting in a potent anti-apoptotic response. However, IR-induced IRAK1 activation does not require the receptors or the IRAK4/1 adaptor protein MyD88, and instead of remaining in the cytoplasm, the activated kinase is immediately transported to the nucleus via a conserved nuclear localization signal. We identify: double-strand DNA breaks (DSBs) as the biologic trigger for this pathway; the E3 ubiquitin ligase Pellino1 as the scaffold enabling IRAK4/1 activation in place of TLR/IL-1R-MyD88; and the pro-apoptotic PIDDosome (PIDD1-RAIDD-caspase-2) as a critical downstream target in the nucleus. The data delineate a non-canonical IRAK signaling pathway derived from, or ancestral to, TLR signaling. This DSB detection pathway, which is also activated by genotoxic chemotherapies, provides multiple actionable targets for overcoming tumor resistance to mainstay cancer treatments.
Collapse
|
13
|
Chang C, Tang X, Woodley DT, Chen M, Li W. The Distinct Assignments for Hsp90α and Hsp90β: More Than Skin Deep. Cells 2023; 12:277. [PMID: 36672211 PMCID: PMC9857327 DOI: 10.3390/cells12020277] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
For decades, the undisputable definition of the cytosolic Hsp90α and hsp90β proteins being evolutionarily conserved, ATP-driven chaperones has ruled basic research and clinical trials. The results of recent studies, however, have fundamentally challenged this paradigm, not to mention the spectacular failures of the paradigm-based clinical trials in cancer and beyond. We now know that Hsp90α and Hsp90β are both ubiquitously expressed in all cell types but assigned for distinct and irreplaceable functions. Hsp90β is essential during mouse development and Hsp90α only maintains male reproductivity in adult mice. Neither Hsp90β nor Hsp90α could substitute each other under these biological processes. Hsp90β alone maintains cell survival in culture and Hsp90α cannot substitute it. Hsp90α also has extracellular functions under stress and Hsp90β does not. The dramatic difference in the steady-state expression of Hsp90 in different mouse organs is due to the variable expressions of Hsp90α. The lowest expression of Hsp90 is less than 2% and the highest expression of Hsp90 is 9% among non-transformed cell lines. The two linker regions only take up less than 5% of the Hsp90 proteins, but harbor 21% of the total amino acid substitutions, i.e., 40% in comparison to the 86% overall amino acid homology. A full understanding of the distinctions between Hsp90α and Hsp90β could lead to new, safe and effective therapeutics targeting Hsp90 in human disorders such as cancer. This is the first comprehensive review of a comparison between the two cytosolic Hsp90 isoforms.
Collapse
Affiliation(s)
| | | | | | | | - Wei Li
- Department of Dermatology and the Norris Comprehensive Cancer Centre, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| |
Collapse
|
14
|
Tie H, Lu X, Yu D, Yang F, Jiang Q, Xu Y, Xia W. Apoptosis Inducing Factors Involved in the Changes of Flesh Quality in Postmortem Grass Carp ( Ctenopharyngodon idella) Muscle. Foods 2022; 12:foods12010140. [PMID: 36613356 PMCID: PMC9818144 DOI: 10.3390/foods12010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 12/29/2022] Open
Abstract
Alterations of apoptosis have notable influences on flesh quality, but the mechanism is still unclear. Thus, apoptotic behaviors and related triggering mechanisms need to be explored. Fish muscle was prepared and stored at 4 °C for 0, 24, 48, 72, 96, and 120 h for apoptosis analysis. Results showed that positive apoptotic nuclei were positively correlated with drop loss and negatively correlated with shear force and water holding capacity (p < 0.05). Results showed that the triggering apoptotic mechanisms were involved with enhanced transcriptional levels of caspase-2, 3, 7, 8, and 9 through mitochondria and death receptor pathways in the muscle of grass carp. The decreased ATP content, changed cytochrome c redox state, increased protein levels of HSP27 and HSP 90, and enhanced activity of cathepsin (B, L, and D), calpain, and serine proteinase were involved in apoptosis activations. Results indicated that caspases, energy metabolism, cytochrome c redox state, heat shock protein expressions, and protease activities played critical roles in apoptosis alterations in carp muscle during refrigerated storage.
Collapse
Affiliation(s)
- Huaimao Tie
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Xuan Lu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Dawei Yu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- Correspondence:
| | - Fang Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Qixing Jiang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Yanshun Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Wenshui Xia
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
15
|
Bosc E, Anastasie J, Soualmia F, Coric P, Kim JY, Wang LQ, Lacin G, Zhao K, Patel R, Duplus E, Tixador P, Sproul AA, Brugg B, Reboud-Ravaux M, Troy CM, Shelanski ML, Bouaziz S, Karin M, El Amri C, Jacotot ED. Genuine selective caspase-2 inhibition with new irreversible small peptidomimetics. Cell Death Dis 2022; 13:959. [PMID: 36379916 PMCID: PMC9666555 DOI: 10.1038/s41419-022-05396-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Caspase-2 (Casp2) is a promising therapeutic target in several human diseases, including nonalcoholic steatohepatitis (NASH) and Alzheimer's disease (AD). However, the design of an active-site-directed inhibitor selective to individual caspase family members is challenging because caspases have extremely similar active sites. Here we present new peptidomimetics derived from the VDVAD pentapeptide structure, harboring non-natural modifications at the P2 position and an irreversible warhead. Enzyme kinetics show that these new compounds, such as LJ2 or its specific isomers LJ2a, and LJ3a, strongly and irreversibly inhibit Casp2 with genuine selectivity. In agreement with the established role of Casp2 in cellular stress responses, LJ2 inhibits cell death induced by microtubule destabilization or hydroxamic acid-based deacetylase inhibition. The most potent peptidomimetic, LJ2a, inhibits human Casp2 with a remarkably high inactivation rate (k3/Ki ~5,500,000 M-1 s-1), and the most selective inhibitor, LJ3a, has close to a 1000 times higher inactivation rate on Casp2 as compared to Casp3. Structural analysis of LJ3a shows that the spatial configuration of Cα at the P2 position determines inhibitor efficacy. In transfected human cell lines overexpressing site-1 protease (S1P), sterol regulatory element-binding protein 2 (SREBP2) and Casp2, LJ2a and LJ3a fully inhibit Casp2-mediated S1P cleavage and thus SREBP2 activation, suggesting a potential to prevent NASH development. Furthermore, in primary hippocampal neurons treated with β-amyloid oligomers, submicromolar concentrations of LJ2a and of LJ3a prevent synapse loss, indicating a potential for further investigations in AD treatment.
Collapse
Affiliation(s)
- Elodie Bosc
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Julie Anastasie
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Feryel Soualmia
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Pascale Coric
- Université de Paris, CNRS, CiTCoM, F-75006, Paris, France
| | - Ju Youn Kim
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Lily Q Wang
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Gullen Lacin
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
- MicroBrain Biotech S.A.S. 52 Avenue de l'Europe, Marly-Le-Roi, F-78160, France
| | - Kaitao Zhao
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Ronak Patel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Eric Duplus
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Philippe Tixador
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Andrew A Sproul
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Bernard Brugg
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Michelle Reboud-Ravaux
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Carol M Troy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Michael L Shelanski
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Serge Bouaziz
- Université de Paris, CNRS, CiTCoM, F-75006, Paris, France
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Chahrazade El Amri
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France
| | - Etienne D Jacotot
- INSERM U1164, CNRS UMR 8256, Sorbonne Université, Campus Pierre et Marie Curie, Paris, F-75005, France.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
16
|
Hååg P, Olsson M, Forsberg J, Lindberg ML, Stenerlöw B, Zong D, Kanter L, Lewensohn R, Viktorsson K, Zhivotovsky B, Stenke L. Caspase-2 is a mediator of apoptotic signaling in response to gemtuzumab ozogamicin in acute myeloid leukemia. Cell Death Discov 2022; 8:284. [PMID: 35690610 PMCID: PMC9188552 DOI: 10.1038/s41420-022-01071-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/02/2022] [Accepted: 05/26/2022] [Indexed: 11/27/2022] Open
Abstract
The antibody conjugate gemtuzumab ozogamicin (GO; Mylotarg®) provides targeted therapy of acute myeloid leukemia (AML), with recent approvals for patients with CD33-positive disease at diagnosis or relapse, as monotherapy or combined with chemotherapeutics. While its clinical efficacy is well documented, the molecular routes by which GO induces AML cell death warrant further analyses. We have earlier reported that this process is initiated via mitochondria-mediated caspase activation. Here we provide additional data, focusing on the involvement of caspase-2 in this mechanism. We show that this enzyme plays an important role in triggering apoptotic death of human AML cells after exposure to GO or its active moiety calicheamicin. Accordingly, the caspase-2 inhibitor z-VDVAD-fmk reduced GO-induced caspase-3 activation. This finding was validated with shRNA and siRNA targeting caspase-2, resulting in reduced caspase-3 activation and cleavage of poly [ADP-ribose] polymerase 1 (PARP-1). We previously demonstrated that GO-induced apoptosis included a conformational change of Bax into a pro-apoptotic state. Present data reveal that GO-treatment also induced Bid cleavage, which was partially reduced by caspase-2 specific inhibition while the effect on GO-induced Bax conformational change remained unaltered. In mononuclear cells isolated from AML patients that responded to GO treatment in vitro, processing of caspase-2 was evident, whereas in cells from an AML patient refractory to treatment no such processing was seen. When assessing diagnostic samples from 22 AML patients, who all entered complete remission (CR) following anthracycline-based induction therapy, and comparing patients with long versus those with short CR duration no significant differences in baseline caspase-2 or caspase-3 full-length protein expression levels were found. In summary, we demonstrate that GO triggers caspase-2 cleavage in human AML cells and that the subsequent apoptosis of these cells in part relies on caspase-2. These findings may have future clinical implications.
Collapse
Affiliation(s)
- Petra Hååg
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64, Solna, Sweden.
| | - Magnus Olsson
- Institute of Environmental Medicine, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Jeremy Forsberg
- Institute of Environmental Medicine, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | | | - Bo Stenerlöw
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-75185, Uppsala, Sweden
| | - Dali Zong
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64, Solna, Sweden
- Laboratory of Genome Integrity, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Lena Kanter
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64, Solna, Sweden
| | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64, Solna, Sweden
- Theme Cancer, Medical Unit head and neck, lung and skin tumors, Thoracic Oncoflogy Center, Karolinska University Hospital, SE-171 64, Solna, Sweden
| | - Kristina Viktorsson
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64, Solna, Sweden
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, SE-171 77, Stockholm, Sweden
- Faculty of Medicine, Lomonosov Moscow State University, 191992, Moscow, Russia
| | - Leif Stenke
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64, Solna, Sweden
- Theme Cancer, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, SE-171 64, Solna, Sweden
| |
Collapse
|
17
|
Kong X, Gao M, Liu Y, Zhang P, Li M, Ma P, Shang P, Wang W, Liu H, Zhang Q, Feng F. GSDMD-miR-223-NLRP3 axis involved in B(a)P-induced inflammatory injury of alveolar epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 232:113286. [PMID: 35144130 DOI: 10.1016/j.ecoenv.2022.113286] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 06/14/2023]
Abstract
Benzo(a)pyrene [B(a)P], a ubiquitous environmental pollutant, causes lung inflammatory damage. Pyroptosis,a new inflammation-dependent programmed cell death, happened when pyroptosis-related GSDMD is activated mediated by NLRP3 inflammasome. microRNA-223 (miRNA-223) is involved in inflammatory diseases by regulating NLRP3. However, whether GSDMD regulate NLRP3 inflammasome through miR-223 in B(a)P induced lung inflammatory injury remain unknown. In this study, alveolar epithelial cells (A549) were stimulated with 0, 2, 4, 8 μM B(a)P for 12 h or 24 h. The inflammatory injury and pyroptosis were determined. And the activation of NLRP3 inflammasomes and the level of miRNA-223 were detected. Then, the change of inflammatory injury and activation of NLRP3 inflammasomes in B(a)P-induced A549 cells were detected after inhibiting of GSDMD or miR-223 using siRNA-GSDMD (siGSDMD) or miR-223 inhibitor, respectively. Our results indicated that after B(a)P exposure, TNF-α and IL-6 in the supernatant were increased. Transmission electron microscope (TEM) results showed that A549 cells were obviously swollen and the cell membrane ruptured. Hoechest33342/PI staining showed that pyroptosis occurred. NLRP3, IL-1β, IL-18, GSDMD, GSDMD-N, pro caspase-1 and cleaved caspase-1 were significantly increased. Additionally, after transfecting with siGSDMD in B(a)P-induced A549 cells, the expression level of miR-223 was significantly increased. But IL-6 and TNF-α in the supernatant, the expression of NLRP3, IL-1β, IL-18 and cleaved caspase-1 protein were also decreased. And after inhibiting miR-223 in B(a)P-induced A549 cells, the expression of TNF-α and IL-6 in the supernatant, the protein expression of NLRP3, IL-1β, IL-18 and cleaved caspase-1 were increased. In conclusion, GSDMD may regulate NLRP3 inflammasome through miR-223, which is involved in B(a)P induced inflammatory damage in A549 cells.
Collapse
Affiliation(s)
- Xiangbing Kong
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province, China
| | - Min Gao
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province, China
| | - Yitong Liu
- College of Public Health, University of Southern California, Los Angeles, USA
| | - Peng Zhang
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, Henan Province, China
| | - Mengyuan Li
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province, China
| | - Pengwei Ma
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province, China
| | - Pingping Shang
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute, CNC, Zhengzhou, Henan Province, China
| | - Wei Wang
- Department of Occupational Medicine, Zhengzhou University School of Public Health, Zhengzhou, Henan Province, China
| | - Hong Liu
- Department of Pulmonary Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiao Zhang
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province, China
| | - Feifei Feng
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province, China.
| |
Collapse
|
18
|
Boice AG, Lopez KE, Pandita RK, Parsons MJ, Charendoff CI, Charaka V, Carisey AF, Pandita TK, Bouchier-Hayes L. Caspase-2 regulates S-phase cell cycle events to protect from DNA damage accumulation independent of apoptosis. Oncogene 2022; 41:204-219. [PMID: 34718349 PMCID: PMC8738157 DOI: 10.1038/s41388-021-02085-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 11/09/2022]
Abstract
In addition to its classical role in apoptosis, accumulating evidence suggests that caspase-2 has non-apoptotic functions, including regulation of cell division. Loss of caspase-2 is known to increase proliferation rates but how caspase-2 is regulating this process is currently unclear. We show that caspase-2 is activated in dividing cells in G1-phase of the cell cycle. In the absence of caspase-2, cells exhibit numerous S-phase defects including delayed exit from S-phase, defects in repair of chromosomal aberrations during S-phase, and increased DNA damage following S-phase arrest. In addition, caspase-2-deficient cells have a higher frequency of stalled replication forks, decreased DNA fiber length, and impeded progression of DNA replication tracts. This indicates that caspase-2 protects from replication stress and promotes replication fork protection to maintain genomic stability. These functions are independent of the pro-apoptotic function of caspase-2 because blocking caspase-2-induced cell death had no effect on cell division, DNA damage-induced cell cycle arrest, or DNA damage. Thus, our data supports a model where caspase-2 regulates cell cycle and DNA repair events to protect from the accumulation of DNA damage independently of its pro-apoptotic function.
Collapse
Affiliation(s)
- Ashley G Boice
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Karla E Lopez
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Raj K Pandita
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas A&M Institute of Biosciences and Technology, Houston, TX, 77030, USA
| | - Melissa J Parsons
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chloe I Charendoff
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
| | - Vijay Charaka
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Alexandre F Carisey
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
- Department of Pediatrics, Section of Allergy and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tej K Pandita
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas A&M Institute of Biosciences and Technology, Houston, TX, 77030, USA
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Shah RB, Kernan JL, van Hoogstraten A, Ando K, Li Y, Belcher AL, Mininger I, Bussenault AM, Raman R, Ramanagoudr-Bhojappa R, Huang TT, D'Andrea AD, Chandrasekharappa SC, Aggarwal AK, Thompson R, Sidi S. FANCI functions as a repair/apoptosis switch in response to DNA crosslinks. Dev Cell 2021; 56:2207-2222.e7. [PMID: 34256011 DOI: 10.1016/j.devcel.2021.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 05/12/2021] [Accepted: 06/10/2021] [Indexed: 12/16/2022]
Abstract
Cells counter DNA damage through repair or apoptosis, yet a direct mechanism for this choice has remained elusive. When facing interstrand crosslinks (ICLs), the ICL-repair protein FANCI heterodimerizes with FANCD2 to initiate ICL excision. We found that FANCI alternatively interacts with a pro-apoptotic factor, PIDD1, to enable PIDDosome (PIDD1-RAIDD-caspase-2) formation and apoptotic death. FANCI switches from FANCD2/repair to PIDD1/apoptosis signaling in the event of ICL-repair failure. Specifically, removing key endonucleases downstream of FANCI/FANCD2, increasing ICL levels, or allowing damaged cells into mitosis (when repair is suppressed) all suffice for switching. Reciprocally, apoptosis-committed FANCI reverts from PIDD1 to FANCD2 after a failed attempt to assemble the PIDDosome. Monoubiquitination and deubiquitination at FANCI K523 impact interactor selection. These data unveil a repair-or-apoptosis switch in eukaryotes. Beyond ensuring the removal of unrepaired genomes, the switch's bidirectionality reveals that damaged cells can offset apoptotic defects via de novo attempts at lesion repair.
Collapse
Affiliation(s)
- Richa B Shah
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer L Kernan
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anya van Hoogstraten
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiyohiro Ando
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuanyuan Li
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alicia L Belcher
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ivy Mininger
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrei M Bussenault
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Renuka Raman
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ramanagouda Ramanagoudr-Bhojappa
- Cancer Genomics Unit, Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tony T Huang
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Settara C Chandrasekharappa
- Cancer Genomics Unit, Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aneel K Aggarwal
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruth Thompson
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology & Metabolism, University of Sheffield Medical School, Sheffield, UK
| | - Samuel Sidi
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
20
|
Bolívar BE, Brown-Suedel AN, Rohrman BA, Charendoff CI, Yazdani V, Belcher JD, Vercellotti GM, Flanagan JM, Bouchier-Hayes L. Noncanonical Roles of Caspase-4 and Caspase-5 in Heme-Driven IL-1β Release and Cell Death. THE JOURNAL OF IMMUNOLOGY 2021; 206:1878-1889. [PMID: 33741688 DOI: 10.4049/jimmunol.2000226] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 01/28/2021] [Indexed: 12/21/2022]
Abstract
Excessive release of heme from RBCs is a key pathophysiological feature of several disease states, including bacterial sepsis, malaria, and sickle cell disease. This hemolysis results in an increased level of free heme that has been implicated in the inflammatory activation of monocytes, macrophages, and the endothelium. In this study, we show that extracellular heme engages the human inflammatory caspases, caspase-1, caspase-4, and caspase-5, resulting in the release of IL-1β. Heme-induced IL-1β release was further increased in macrophages from patients with sickle cell disease. In human primary macrophages, heme activated caspase-1 in an inflammasome-dependent manner, but heme-induced activation of caspase-4 and caspase-5 was independent of canonical inflammasomes. Furthermore, we show that both caspase-4 and caspase-5 are essential for heme-induced IL-1β release, whereas caspase-4 is the primary contributor to heme-induced cell death. Together, we have identified that extracellular heme is a damage-associated molecular pattern that can engage canonical and noncanonical inflammasome activation as a key mediator of inflammation in macrophages.
Collapse
Affiliation(s)
- Beatriz E Bolívar
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
| | - Alexandra N Brown-Suedel
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
| | - Brittany A Rohrman
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Chloé I Charendoff
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Vanda Yazdani
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
| | - John D Belcher
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55455; and
| | - Gregory M Vercellotti
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55455; and
| | - Jonathan M Flanagan
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
| | - Lisa Bouchier-Hayes
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030; .,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
21
|
Brown-Suedel AN, Bouchier-Hayes L. Caspase-2 Substrates: To Apoptosis, Cell Cycle Control, and Beyond. Front Cell Dev Biol 2020; 8:610022. [PMID: 33425918 PMCID: PMC7785872 DOI: 10.3389/fcell.2020.610022] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/03/2020] [Indexed: 01/12/2023] Open
Abstract
Caspase-2 belongs to the caspase family of proteins responsible for essential cellular functions including apoptosis and inflammation. Uniquely, caspase-2 has been identified as a tumor suppressor, but how it regulates this function is still unknown. For many years, caspase-2 has been considered an “orphan” caspase because, although it is able to induce apoptosis, there is an abundance of conflicting evidence that questions its necessity for apoptosis. Recent evidence supports that caspase-2 has non-apoptotic functions in the cell cycle and protection from genomic instability. It is unclear how caspase-2 regulates these opposing functions, which has made the mechanism of tumor suppression by caspase-2 difficult to determine. As a protease, caspase-2 likely exerts its functions by proteolytic cleavage of cellular substrates. This review highlights the known substrates of caspase-2 with a special focus on their functional relevance to caspase-2’s role as a tumor suppressor.
Collapse
Affiliation(s)
- Alexandra N Brown-Suedel
- Hematology-Oncology Section, Department of Pediatrics, Department of Molecular Cell Biology, Baylor College of Medicine, Houston, TX, United States.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX, United States
| | - Lisa Bouchier-Hayes
- Hematology-Oncology Section, Department of Pediatrics, Department of Molecular Cell Biology, Baylor College of Medicine, Houston, TX, United States.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
22
|
Phosphorylation by Aurora B kinase regulates caspase-2 activity and function. Cell Death Differ 2020; 28:349-366. [PMID: 32811973 PMCID: PMC7852673 DOI: 10.1038/s41418-020-00604-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Mitotic catastrophe (MC) is an important oncosuppressive mechanism that serves to eliminate cells that become polyploid or aneuploid due to aberrant mitosis. Previous studies have demonstrated that the activation and catalytic function of caspase-2 are key steps in MC to trigger apoptosis and/or cell cycle arrest of mitotically defective cells. However, the molecular mechanisms that regulate caspase-2 activation and its function are unclear. Here, we identify six new phosphorylation sites in caspase-2 and show that a key mitotic kinase, Aurora B kinase (AURKB), phosphorylates caspase-2 at the highly conserved residue S384. We demonstrate that phosphorylation at S384 blocks caspase-2 catalytic activity and apoptosis function in response to mitotic insults, without affecting caspase-2 dimerisation. Moreover, molecular modelling suggests that phosphorylation at S384 may affect substrate binding by caspase-2. We propose that caspase-2 S384 phosphorylation by AURKB is a key mechanism that controls caspase-2 activation during mitosis.
Collapse
|
23
|
Hiregange D, Naick H, Rao BJ. ATR signalling mediates the prosurvival function of phospho-NPM against PIDDosome mediated cell death. Cell Signal 2020; 71:109602. [DOI: 10.1016/j.cellsig.2020.109602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 03/14/2020] [Indexed: 12/14/2022]
|
24
|
Vigneswara V, Ahmed Z. The Role of Caspase-2 in Regulating Cell Fate. Cells 2020; 9:cells9051259. [PMID: 32438737 PMCID: PMC7290664 DOI: 10.3390/cells9051259] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Caspase-2 is the most evolutionarily conserved member of the mammalian caspase family and has been implicated in both apoptotic and non-apoptotic signaling pathways, including tumor suppression, cell cycle regulation, and DNA repair. A myriad of signaling molecules is associated with the tight regulation of caspase-2 to mediate multiple cellular processes far beyond apoptotic cell death. This review provides a comprehensive overview of the literature pertaining to possible sophisticated molecular mechanisms underlying the multifaceted process of caspase-2 activation and to highlight its interplay between factors that promote or suppress apoptosis in a complicated regulatory network that determines the fate of a cell from its birth and throughout its life.
Collapse
|
25
|
Roque W, Cuevas-Mora K, Romero F. Mitochondrial Quality Control in Age-Related Pulmonary Fibrosis. Int J Mol Sci 2020; 21:ijms21020643. [PMID: 31963720 PMCID: PMC7013724 DOI: 10.3390/ijms21020643] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/11/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is age-related interstitial lung disease of unknown etiology. About 100,000 people in the U.S have IPF, with a 3-year median life expectancy post-diagnosis. The development of an effective treatment for pulmonary fibrosis will require an improved understanding of its molecular pathogenesis and the “normal” and “pathological’ hallmarks of the aging lung. An important characteristic of the aging organism is its lowered capacity to adapt quickly to, and counteract, disturbances. While it is likely that DNA damage, chronic endoplasmic reticulum (ER) stress, and accumulation of heat shock proteins are capable of initiating tissue repair, recent studies point to a pathogenic role for mitochondrial dysfunction in the development of pulmonary fibrosis. These studies suggest that damage to the mitochondria induces fibrotic remodeling through a variety of mechanisms including the activation of apoptotic and inflammatory pathways. Mitochondrial quality control (MQC) has been demonstrated to play an important role in the maintenance of mitochondrial homeostasis. Different factors can induce MQC, including mitochondrial DNA damage, proteostasis dysfunction, and mitochondrial protein translational inhibition. MQC constitutes a complex signaling response that affects mitochondrial biogenesis, mitophagy, fusion/fission and the mitochondrial unfolded protein response (UPRmt) that, together, can produce new mitochondria, degrade the components of the oxidative complex or clearance the entire organelle. In pulmonary fibrosis, defects in mitophagy and mitochondrial biogenesis have been implicated in both cellular apoptosis and senescence during tissue repair. MQC has also been found to have a role in the regulation of other protein activity, inflammatory mediators, latent growth factors, and anti-fibrotic growth factors. In this review, we delineated the role of MQC in the pathogenesis of age-related pulmonary fibrosis.
Collapse
Affiliation(s)
- Willy Roque
- Department of Medicine, Rutgers New Jersey Medical School, 185 S Orange Ave, Newark, NJ 07103, USA;
| | - Karina Cuevas-Mora
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Freddy Romero
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
- Correspondence:
| |
Collapse
|
26
|
Xu ZX, Tan JW, Xu H, Hill CJ, Ostrovskaya O, Martemyanov KA, Xu B. Caspase-2 promotes AMPA receptor internalization and cognitive flexibility via mTORC2-AKT-GSK3β signaling. Nat Commun 2019; 10:3622. [PMID: 31399584 PMCID: PMC6689033 DOI: 10.1038/s41467-019-11575-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 07/23/2019] [Indexed: 01/22/2023] Open
Abstract
Caspase-2 is the most evolutionarily conserved member in the caspase family of proteases and is constitutively expressed in most cell types including neurons; however, its physiological function remains largely unknown. Here we report that caspase-2 plays a critical role in synaptic plasticity and cognitive flexibility. We found that caspase-2 deficiency led to deficits in dendritic spine pruning, internalization of AMPA receptors and long-term depression. Our results indicate that caspase-2 degrades Rictor, a key mTOR complex 2 (mTORC2) component, to inhibit Akt activation, which leads to enhancement of the GSK3β activity and thereby long-term depression. Furthermore, we found that mice lacking caspase-2 displayed elevated levels of anxiety, impairment in reversal water maze learning, and little memory loss over time. These results not only uncover a caspase-2-mTORC2-Akt-GSK3β signaling pathway, but also suggest that caspase-2 is important for memory erasing and normal behaviors by regulating synaptic number and transmission.
Collapse
Affiliation(s)
- Zhi-Xiang Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Ji-Wei Tan
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Haifei Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Cassandra J Hill
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA.
| |
Collapse
|
27
|
Radke JR, Routes JM, Cook JL. E1A oncogene induced sensitization to NK cell induced apoptosis requires PIDD and Caspase-2. Cell Death Discov 2019; 5:110. [PMID: 31285853 PMCID: PMC6602934 DOI: 10.1038/s41420-019-0189-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 11/09/2022] Open
Abstract
Expression of the adenovirus E1A oncogene sensitizes tumor cells to innate immune rejection by NK cells. This increased NK sensitivity is only partly explained by an E1A-induced increase in target cell surface expression of NKG2D ligands. The post-recognition mechanisms by which E1A sensitizes cells to the apoptotic cell death response to NK injury remains to be defined. E1A sensitizes cells to apoptotic stimuli through two distinct mechanisms-repression of NF-κB-dependent antiapoptotic responses and enhancement of caspase-2 activation and related mitochondrial injury. The current studies examined the roles of each of these post-NKG2D-recognition pathways in the increased sensitivity of E1A-positive target cells to NK killing. Sensitization to NK-induced apoptosis was independent of E1A-mediated repression of cellular NF-κB responses but was dependent on the expression of both caspase-2 and the upstream, caspase-2 activating molecule, PIDD. Target cells lacking caspase-2 or PIDD expression retained E1A-induced increased expression of the NKG2D ligand, RAE-1. NK cell-induced mitochondrial injury of E1A-expressing cells did not require expression of the mitochondrial molecules, Bak or Bax. These results define a PIDD/caspase-2-dependent pathway, through which E1A sensitizes cells to NK-mediated cytolysis independently of and complementarily to E1A-enhanced NKG2D/RAE-1 ligand expression.
Collapse
Affiliation(s)
- Jay R Radke
- 1Research Section, Boise VA Hospital and Idaho Veterans Research and Education Foundation, Boise, ID 83702 USA
| | - John M Routes
- 2Section of Allergy and Clinical Immunology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - James L Cook
- 3Research Section, Edward Hines, Jr. VA Hospital, Hines, Maywood, IL 60141 USA.,4Division of Infectious Diseases, Department of Microbiology and Immunology, and the Infectious Diseases and Immunology Research Institute, Loyola University Chicago-Stritch School of Medicine, Maywood, IL 60153 USA
| |
Collapse
|
28
|
Potent and selective caspase-2 inhibitor prevents MDM-2 cleavage in reversine-treated colon cancer cells. Cell Death Differ 2019; 26:2695-2709. [PMID: 30976094 DOI: 10.1038/s41418-019-0329-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/17/2019] [Accepted: 03/25/2019] [Indexed: 01/02/2023] Open
Abstract
Most caspases can be positioned unambiguously within the regulated cell death networks of apoptosis and pyroptosis, but the role of caspase-2, a highly conserved protease within the family, remains enigmatic. This is mainly due to lack of selective chemical and biochemical tools for the investigation of this protease. In this study, we used our hybrid combinatorial substrate library (HyCoSuL) approach to broadly profile caspase-2 substrate specificity using peptide scanning libraries. This screen uncovered previously unknown caspase-2 peptidyl substrate preferences, which were further used to develop caspase-2 selective fluorogenic substrates and covalent, irreversible AOMK inhibitors. Finally, we used the champion inhibitor (NH-23-C2) in reversine-treated HCT-116 colon cancer cells to selectively block caspase-2 activity and caspase-2-mediated MDM-2 cleavage. In addition, we showed that NH-23-C2 does not block caspase-3 or caspase-8, which makes it a powerful chemical tool to dissect the true role of caspase-2 in various biological setups.
Collapse
|
29
|
Song N, Li T. Regulation of NLRP3 Inflammasome by Phosphorylation. Front Immunol 2018; 9:2305. [PMID: 30349539 PMCID: PMC6186804 DOI: 10.3389/fimmu.2018.02305] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/17/2018] [Indexed: 01/01/2023] Open
Abstract
The cytosolic pattern recognition receptor (PRR) NOD-like receptor family, pyrin domain containing 3 (NLRP3) senses a wide range of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Upon activation, NLRP3 triggers the assembly of inflammasome via the self-oligomerization and the recruitment of apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) and pro-caspase-1, facilitating the robust immune responses including the secretion of proinflammatory cytokines and pyroptosis. The NLRP3 inflammasome must be well orchestrated to prevent the aberrant activations under physiological and pathological conditions, because uncontrolled activation of NLRP3 inflammasome is one of the major causes of a variety of autoimmune diseases and metabolic disorders. Therefore, understanding the molecular mechanisms for controlling NLRP3 inflammasome activation may provide novel strategies for the treatment of NLRP3-related diseases. Although NLRP3 inflammasome can be regulated at the transcriptional level, the post-translational modification (PTM) of NLRP3 as well as other inflammasome components has also been showed to be critical for the regulation of its activation. Several kinases and phosphatases have been shown to control NLRP3 inflammasome activation in response to either exogenous pathogen infections or endogenous molecules, such as bile acids. In this review, we summarize our current knowledge of phosphorylation patterns and their functional role in the regulation of NLRP3 inflammasome, and suggest interesting areas for future research.
Collapse
Affiliation(s)
- Nan Song
- Beijing Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,State Key Laboratory of Proteomics, Beijing Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Tao Li
- State Key Laboratory of Proteomics, Beijing Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
30
|
Ha HJ, Park HH. RAIDD mutations underlie the pathogenesis of thin lissencephaly (TLIS). PLoS One 2018; 13:e0205042. [PMID: 30281648 PMCID: PMC6169973 DOI: 10.1371/journal.pone.0205042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/18/2018] [Indexed: 11/18/2022] Open
Abstract
Abnormal regulation of caspase-2-mediated neuronal cell death causes neurodegenerative diseases and defective brain development. PIDDosome is caspase-2 activating complex composed of PIDD, RAIDD, and caspase-2. Recent whole-exome sequencing study showed that the RAIDD mutations in the death domain (DD), including G128R, F164C, R170C, and R170H mutations, cause thin lissencephaly (TLIS) by reducing caspase-2-mediated neuronal apoptosis. Given that the molecular structure of the RAIDD DD:PIDD DD complex is available, in this study, we analyzed the molecular mechanisms underlying TLIS caused by the RAIDD TLIS variants by performing mutagenesis and biochemical assays.
Collapse
Affiliation(s)
- Hyun Ji Ha
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
- * E-mail:
| |
Collapse
|
31
|
PIDD-dependent activation of caspase-2-mediated mitochondrial injury in E1A-induced cellular sensitivity to macrophage nitric oxide-induced apoptosis. Cell Death Discov 2018; 4:35. [PMID: 30245858 PMCID: PMC6135794 DOI: 10.1038/s41420-018-0100-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023] Open
Abstract
Expression of the adenovirus E1A oncogene sensitizes tumor cells to innate immune rejection by apoptosis induced by macrophage-produced tumor necrosis factor (TNF)-α and nitric oxide (NO). E1A sensitizes cells to TNF-α and NO through two distinct mechanisms, by repressing NF-κB-dependent antiapoptotic responses and enhancing caspase-2 activation and mitochondrial injury, respectively. The mechanisms through which E1A enhances caspase-2 activation in response to NO were unknown. Here, we report that E1A-induced sensitization to NO-induced apoptosis is dependent on expression of PIDD (p53-inducible protein with a death domain) and enhancement of primary immunodeficiency diseases (PIDD) processing for formation of the PIDDosome, the core component of the caspase-2 activation complex. NO-induced apoptosis in E1A-expressing cells did not require expression Bak or Bax, indicating that NO-induced caspase-2-mediated mitochondrial injury does not proceed through the activities of typical, proapoptotic Bcl-2 family members that induce mitochondrial cytochrome C release. These results define a PIDD-dependent pathway, through which E1A enhances casapse-2-mediated mitochondrial injury, resulting in increased sensitivity of mammalian cells to macrophage-induced, NO-mediated apoptosis.
Collapse
|
32
|
Robeson AC, Lindblom KR, Wojton J, Kornbluth S, Matsuura K. Dimer-specific immunoprecipitation of active caspase-2 identifies TRAF proteins as novel activators. EMBO J 2018; 37:e97072. [PMID: 29875129 PMCID: PMC6043850 DOI: 10.15252/embj.201797072] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 12/13/2022] Open
Abstract
Caspase-2 has been shown to initiate apoptotic cell death in response to specific intracellular stressors such as DNA damage. However, the molecular mechanisms immediately upstream of its activation are still poorly understood. We combined a caspase-2 bimolecular fluorescence complementation (BiFC) system with fluorophore-specific immunoprecipitation to isolate and study the active caspase-2 dimer and its interactome. Using this technique, we found that tumor necrosis factor receptor-associated factor 2 (TRAF2), as well as TRAF1 and 3, directly binds to the active caspase-2 dimer. TRAF2 in particular is necessary for caspase-2 activation in response to apoptotic cell death stimuli. Furthermore, we found that dimerized caspase-2 is ubiquitylated in a TRAF2-dependent manner at K15, K152, and K153, which in turn stabilizes the active caspase-2 dimer complex, promotes its association with an insoluble cellular fraction, and enhances its activity to fully commit the cell to apoptosis. Together, these data indicate that TRAF2 positively regulates caspase-2 activation and consequent cell death by driving its activation through dimer-stabilizing ubiquitylation.
Collapse
Affiliation(s)
- Alexander C Robeson
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Kelly R Lindblom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey Wojton
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Sally Kornbluth
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Kenkyo Matsuura
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
33
|
Kopeina GS, Prokhorova EA, Lavrik IN, Zhivotovsky B. Alterations in the nucleocytoplasmic transport in apoptosis: Caspases lead the way. Cell Prolif 2018; 51:e12467. [PMID: 29947118 DOI: 10.1111/cpr.12467] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Apoptosis is a mode of regulated cell death that is indispensable for the morphogenesis, development and homeostasis of multicellular organisms. Caspases are cysteine-dependent aspartate-specific proteases, which function as initiators and executors of apoptosis. Caspases are cytosolic proteins that can cleave substrates located in different intracellular compartments during apoptosis. Many years ago, the involvement of caspases in the regulation of nuclear changes, a hallmark of apoptosis, was documented. Accumulated data suggest that apoptosis-associated alterations in nucleocytoplasmic transport are also linked to caspase activity. Here, we aim to discuss the current state of knowledge regarding this process. Particular attention will be focused on caspase nuclear entry and their functions in the demolition of the nucleus upon apoptotic stimuli.
Collapse
Affiliation(s)
- Gelina S Kopeina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | | | - Inna N Lavrik
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Translational Inflammation Research, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Boris Zhivotovsky
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Hashimoto R, Gupte S. Pentose Shunt, Glucose-6-Phosphate Dehydrogenase, NADPH Redox, and Stem Cells in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:47-55. [PMID: 29047080 DOI: 10.1007/978-3-319-63245-2_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Redox signaling plays a critical role in the pathophysiology of cardiovascular diseases. The pentose phosphate pathway is a major source of NADPH redox in the cell. The activities of glucose-6-phosphate dehydrogenase (the rate-limiting enzyme in the pentose shunt) and glucose flux through the shunt pathway is increased in various lung cells including, the stem cells, in pulmonary hypertension. This chapter discusses the importance of the shunt pathway and glucose-6-phosphate dehydrogenase in the pathogenesis of pulmonary artery remodeling and occlusive lesion formation within the hypertensive lungs.
Collapse
Affiliation(s)
- Ryota Hashimoto
- Department of Pharmacology, New York Medical College, School of Medicine, Basic Science Building, Rm. 546, 15 Dana Road, Valhalla, NY, 10595, USA
| | - Sachin Gupte
- Department of Pharmacology, New York Medical College, School of Medicine, Basic Science Building, Rm. 546, 15 Dana Road, Valhalla, NY, 10595, USA.
| |
Collapse
|
35
|
Badawi A, Biyanee A, Nasrullah U, Winslow S, Schmid T, Pfeilschifter J, Eberhardt W. Inhibition of IRES-dependent translation of caspase-2 by HuR confers chemotherapeutic drug resistance in colon carcinoma cells. Oncotarget 2018; 9:18367-18385. [PMID: 29719611 PMCID: PMC5915078 DOI: 10.18632/oncotarget.24840] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 02/23/2018] [Indexed: 12/31/2022] Open
Abstract
HuR plays an important role in tumor cell survival mainly through posttranscriptional upregulation of prominent anti-apoptotic genes. In addition, HuR can inhibit the translation of pro-apoptotic factors as we could previously report for caspase-2. Here, we investigated the mechanisms of caspase-2 suppression by HuR and its contribution to chemotherapeutic drug resistance of colon carcinoma cells. In accordance with the significant drug-induced increase in cytoplasmic HuR abundance, doxorubicin and paclitaxel increased the interaction of cytoplasmic HuR with the 5ʹuntranslated region (5ʹUTR) of caspase-2 as shown by RNA pull down assay. Experiments with bicistronic reporter genes furthermore indicate the presence of an internal ribosome entry site (IRES) within the caspase-2-5ʹUTR. Luciferase activity was suppressed either by chemotherapeutic drugs or ectopic expression of HuR. IRES-driven luciferase activity was significantly increased upon siRNA-mediated knockdown of HuR implicating an inhibitory effect of HuR on caspase-2 translation which is further reinforced by chemotherapeutic drugs. Comparison of RNA-binding affinities of recombinant HuR to two fragments of the caspase-2-5ʹUTR by EMSA revealed a critical HuR-binding site residing between nucleotides 111 and 241 of caspase-2-5ʹUTR. Mapping of critical RNA binding domains within HuR revealed that a fusion of RNA recognition motif 2 (RRM2) plus the hinge region confers a full caspase-2-5ʹUTR-binding. Functionally, knockdown of HuR significantly increased the sensitivity of colon cancer cells to drug-induced apoptosis. Importantly, the apoptosis sensitizing effects by HuR knockdown were rescued after silencing of caspase-2. The negative caspase-2 regulation by HuR offers a novel therapeutic target for sensitizing colon carcinoma cells to drug-induced apoptosis.
Collapse
Affiliation(s)
- Amel Badawi
- Pharmazentrum Frankfurt/ZAFES, Medical School, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Abhiruchi Biyanee
- Pharmazentrum Frankfurt/ZAFES, Medical School, Goethe-University Frankfurt, Frankfurt/Main, Germany.,Present address: Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Usman Nasrullah
- Pharmazentrum Frankfurt/ZAFES, Medical School, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Sofia Winslow
- Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Medical School, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Wolfgang Eberhardt
- Pharmazentrum Frankfurt/ZAFES, Medical School, Goethe-University Frankfurt, Frankfurt/Main, Germany
| |
Collapse
|
36
|
Charendoff CI, Bouchier-Hayes L. Lighting Up the Pathways to Caspase Activation Using Bimolecular Fluorescence Complementation. J Vis Exp 2018. [PMID: 29553529 DOI: 10.3791/57316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The caspase family of proteases play essential roles in apoptosis and innate immunity. Among these, a subgroup known as initiator caspases are the first to be activated in these pathways. This group includes caspase-2, -8, and -9, as well as the inflammatory caspases, caspase-1, -4, and -5. The initiator caspases are all activated by dimerization following recruitment to specific multiprotein complexes called activation platforms. Caspase Bimolecular Fluorescence Complementation (BiFC) is an imaging-based approach where split fluorescent proteins fused to initiator caspases are used to visualize the recruitment of initiator caspases to their activation platforms and the resulting induced proximity. This fluorescence provides a readout of one of the earliest steps required for initiator caspase activation. Using a number of different microscopy-based approaches, this technique can provide quantitative data on the efficiency of caspase activation on a population level as well as the kinetics of caspase activation and the size and number of caspase activating complexes on a per cell basis.
Collapse
Affiliation(s)
- Chloé I Charendoff
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology and Department of Molecular and Cellular Biology, Baylor College of Medicine;
| |
Collapse
|
37
|
Carroll BL, Bonica J, Shamseddine AA, Hannun YA, Obeid LM. A role for caspase-2 in sphingosine kinase 1 proteolysis in response to doxorubicin in breast cancer cells - implications for the CHK1-suppressed pathway. FEBS Open Bio 2017; 8:27-40. [PMID: 29321954 PMCID: PMC5757171 DOI: 10.1002/2211-5463.12344] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/29/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Sphingosine kinase 1 (SK1) is a lipid kinase whose activity produces sphingosine 1‐phosphate, a prosurvival lipid associated with proliferation, angiogenesis, and invasion. SK1 overexpression has been observed in numerous cancers. Recent studies have demonstrated that SK1 proteolysis occurs downstream of the tumor suppressor p53 in response to several DNA‐damaging agents. Moreover, loss of SK1 in p53‐knockout mice resulted in complete protection from thymic lymphoma, providing evidence that regulation of SK1 constitutes a major tumor suppressor function of p53. Given this profound phenotype, this study aimed to investigate the mechanism by which wild‐type p53 regulates proteolysis of SK1 in response to the DNA‐damaging agent doxorubicin in breast cancer cells. We find that p53‐mediated activation of caspase‐2 was required for SK1 proteolysis and that caspase‐2 activity significantly alters the levels of endogenous sphingolipids. As p53 is mutated in 50% of all cancers, we extended our studies to investigate whether SK1 is deregulated in the context of triple‐negative breast cancer cells (TNBC) harboring a mutation in p53. Indeed, caspase‐2 was not activated in these cells and SK1 was not degraded. Moreover, caspase‐2 activation was recently shown to be downstream of the CHK1‐suppressed pathway in p53‐mutant cells, whereby inhibition of the cell cycle kinase CHK1 leads to caspase‐2 activation and apoptosis. Indeed, knockdown and inhibition of CHK1 led to the loss of SK1 in p53‐mutant TNBC cells, providing evidence that SK1 may be the first identified effector of the CHK1‐suppressed pathway.
Collapse
Affiliation(s)
- Brittany L Carroll
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Joseph Bonica
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Achraf A Shamseddine
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Yusuf A Hannun
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Lina M Obeid
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA.,Northport Veterans Affairs Medical Center NY USA
| |
Collapse
|
38
|
Sidi S, Bouchier-Hayes L. Direct pro-apoptotic role for NPM1 as a regulator of PIDDosome formation. Mol Cell Oncol 2017; 4:e1348325. [PMID: 29057309 DOI: 10.1080/23723556.2017.1348325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 06/23/2017] [Accepted: 06/24/2017] [Indexed: 10/19/2022]
Abstract
Despite being frequently mutated or deregulated in acute myeloid leukemia (AML) and many other cancers, the mechanisms by which nucleophosmin (NPM1) regulates oncogenesis remain elusive. We found that NPM1 plays a direct and conserved role in DNA damage-induced assembly of the PIDDosome complex, the activating platform for caspase-2. This function is carried in the nucleolus and is essential for caspase-2-mediated apoptosis in response to a variety of DNA injuries.
Collapse
Affiliation(s)
- Samuel Sidi
- Department of Medicine, Division of Hematology/Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Developmental and Regenerative Biology Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
39
|
Abstract
O’Byrne and Richard preview results from the Bouchier-Hayes group delineating two distinct pathways for caspase-2 activation in response to different stimuli Caspase-2 triggers apoptosis, but how it is activated by different stimuli is unclear. In this issue, Ando et al. (2017. J. Cell Biol.https://doi.org/10.1083/jcb.201608095) delineate two pathways of caspase-2 activation and show that, in response to DNA damage, caspase-2 forms a complex with the PIDDosome and NPM1 within the nucleolus.
Collapse
Affiliation(s)
- Kenneth J O'Byrne
- School of Biomedical Research, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Woolloongabba QLD 4102, Australia
| | - Derek J Richard
- School of Biomedical Research, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Woolloongabba QLD 4102, Australia
| |
Collapse
|
40
|
Ando K, Parsons MJ, Shah RB, Charendoff CI, Paris SL, Liu PH, Fassio SR, Rohrman BA, Thompson R, Oberst A, Sidi S, Bouchier-Hayes L. NPM1 directs PIDDosome-dependent caspase-2 activation in the nucleolus. J Cell Biol 2017; 216:1795-1810. [PMID: 28432080 PMCID: PMC5461015 DOI: 10.1083/jcb.201608095] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/19/2017] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
The PIDDosome (PIDD-RAIDD-caspase-2 complex) is considered to be the primary signaling platform for caspase-2 activation in response to genotoxic stress. Yet studies of PIDD-deficient mice show that caspase-2 activation can proceed in the absence of PIDD. Here we show that DNA damage induces the assembly of at least two distinct activation platforms for caspase-2: a cytoplasmic platform that is RAIDD dependent but PIDD independent, and a nucleolar platform that requires both PIDD and RAIDD. Furthermore, the nucleolar phosphoprotein nucleophosmin (NPM1) acts as a scaffold for PIDD and is essential for PIDDosome assembly in the nucleolus after DNA damage. Inhibition of NPM1 impairs caspase-2 processing, apoptosis, and caspase-2-dependent inhibition of cell growth, demonstrating that the NPM1-dependent nucleolar PIDDosome is a key initiator of the caspase-2 activation cascade. Thus we have identified the nucleolus as a novel site for caspase-2 activation and function.
Collapse
Affiliation(s)
- Kiyohiro Ando
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute at Mount Sinai, New York, NY 10029.,Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Melissa J Parsons
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030
| | - Richa B Shah
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute at Mount Sinai, New York, NY 10029.,Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Chloé I Charendoff
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030
| | - Sheré L Paris
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030
| | - Peter H Liu
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute at Mount Sinai, New York, NY 10029.,Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sara R Fassio
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030
| | - Brittany A Rohrman
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030
| | - Ruth Thompson
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute at Mount Sinai, New York, NY 10029.,Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Samuel Sidi
- Department of Medicine, Division of Hematology/Oncology, Tisch Cancer Institute at Mount Sinai, New York, NY 10029 .,Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030 .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
41
|
Imre G, Berthelet J, Heering J, Kehrloesser S, Melzer IM, Lee BI, Thiede B, Dötsch V, Rajalingam K. Apoptosis inhibitor 5 is an endogenous inhibitor of caspase-2. EMBO Rep 2017; 18:733-744. [PMID: 28336776 DOI: 10.15252/embr.201643744] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/09/2017] [Accepted: 02/14/2017] [Indexed: 11/09/2022] Open
Abstract
Caspases are key enzymes responsible for mediating apoptotic cell death. Across species, caspase-2 is the most conserved caspase and stands out due to unique features. Apart from cell death, caspase-2 also regulates autophagy, genomic stability and ageing. Caspase-2 requires dimerization for its activation which is primarily accomplished by recruitment to high molecular weight protein complexes in cells. Here, we demonstrate that apoptosis inhibitor 5 (API5/AAC11) is an endogenous and direct inhibitor of caspase-2. API5 protein directly binds to the caspase recruitment domain (CARD) of caspase-2 and impedes dimerization and activation of caspase-2. Interestingly, recombinant API5 directly inhibits full length but not processed caspase-2. Depletion of endogenous API5 leads to an increase in caspase-2 dimerization and activation. Consistently, loss of API5 sensitizes cells to caspase-2-dependent apoptotic cell death. These results establish API5/AAC-11 as a direct inhibitor of caspase-2 and shed further light onto mechanisms driving the activation of this poorly understood caspase.
Collapse
Affiliation(s)
- Gergely Imre
- MSU-FZI, Institute of Immunology, University Medical Center Mainz, JGU, Mainz, Germany
| | - Jean Berthelet
- MSU-FZI, Institute of Immunology, University Medical Center Mainz, JGU, Mainz, Germany
| | - Jan Heering
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Sebastian Kehrloesser
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Inga Maria Melzer
- MSU-FZI, Institute of Immunology, University Medical Center Mainz, JGU, Mainz, Germany
| | - Byung Il Lee
- Division of Convergence Technology, Biomolecular Function Research Branch, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Krishnaraj Rajalingam
- MSU-FZI, Institute of Immunology, University Medical Center Mainz, JGU, Mainz, Germany .,UCT, Mainz, German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
42
|
Nguyen MT, Knieß RA, Daturpalli S, Le Breton L, Ke X, Chen X, Mayer MP. Isoform-Specific Phosphorylation in Human Hsp90β Affects Interaction with Clients and the Cochaperone Cdc37. J Mol Biol 2017; 429:732-752. [DOI: 10.1016/j.jmb.2017.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/15/2017] [Accepted: 01/16/2017] [Indexed: 11/28/2022]
|
43
|
McConkey DJ. The integrated stress response and proteotoxicity in cancer therapy. Biochem Biophys Res Commun 2017; 482:450-453. [PMID: 28212730 DOI: 10.1016/j.bbrc.2016.11.047] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022]
Abstract
A variety of different forms of cellular stress can cause protein misfolding and aggregation and proteotoxicity. The cytoprotective response to proteotoxicity is termed the integrated stress response and involves 4 distinct serine/threonine protein kinases that converge on the translation initiation factor eIF2α, resulting in phosphorylation at S51, cell cycle arrest, and a general inhibition of global protein synthesis. Phosphorylation of eIF2α also promotes translation of ATF4 and the expression of ATF4 target genes that ameliorate proteotoxic stress but can also promote apoptosis. This mini review provides a general overview of these mechanisms and discusses how the inter-tumor heterogeneity that involves them affects sensitivity and resistance to proteasome inhibitors, a new class of cancer therapeutics that promotes tumor cell killing via proteotoxic stress.
Collapse
Affiliation(s)
- David J McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Brady Urological Institute, 600 North Wolfe Street, Baltimore, MD 21287, United States.
| |
Collapse
|
44
|
Carroll BL, Pulkoski-Gross MJ, Hannun YA, Obeid LM. CHK1 regulates NF-κB signaling upon DNA damage in p53- deficient cells and associated tumor-derived microvesicles. Oncotarget 2017; 7:18159-70. [PMID: 26921248 PMCID: PMC4951279 DOI: 10.18632/oncotarget.7566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/23/2016] [Indexed: 12/15/2022] Open
Abstract
The recently discovered CHK1-Suppressed (CS) pathway is activated by inhibition or loss of the checkpoint kinase CHK1, promoting an apoptotic response to DNA damage mediated by caspase-2 in p53-deficient cells. Although functions of the CS-pathway have been investigated biochemically, it remains unclear whether and how CHK1 inhibition can be regulated endogenously and whether this constitutes a key component of the DNA damage response (DDR). Here, we present data that define the first endogenous activation of the CS-pathway whereby, upon DNA damage, wild type p53 acts as an endogenous regulator of CHK1 levels that modulates caspase-2 activation. Moreover, we demonstrate that persistence of CHK1 levels in response to DNA damage in p53-deficient cancer cells, leads to CHK1-mediated activation of NF-κB and induction of NF-κB-regulated genes in cells and in associated tumor-derived microvesicles (TMVs), both of which are abrogated by loss or inhibition of CHK1. These data define a novel role for CHK1 in the DDR pathway as a regulator NF-κB activity. Our data provide evidence that targeting CHK1 in p53-deficient cancers may abrogate NF-κB signaling that is associated with increased cellular survival and chemoresistance.
Collapse
Affiliation(s)
- Brittany L Carroll
- Stony Brook Cancer Center and The Department of Medicine, Stony Brook, New York, USA
| | - Michael J Pulkoski-Gross
- Stony Brook Cancer Center and The Department of Medicine, Stony Brook, New York, USA.,Pharmacological Sciences, Stony Brook University, Health Sciences Center, Stony Brook, New York, USA
| | - Yusuf A Hannun
- Stony Brook Cancer Center and The Department of Medicine, Stony Brook, New York, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and The Department of Medicine, Stony Brook, New York, USA.,Northport Veterans Affairs Medical Center, Northport, New York, USA
| |
Collapse
|
45
|
Miles M, Kitevska-Ilioski T, Hawkins C. Old and Novel Functions of Caspase-2. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 332:155-212. [DOI: 10.1016/bs.ircmb.2016.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Dawar S, Lim Y, Puccini J, White M, Thomas P, Bouchier-Hayes L, Green DR, Dorstyn L, Kumar S. Caspase-2-mediated cell death is required for deleting aneuploid cells. Oncogene 2016; 36:2704-2714. [PMID: 27991927 PMCID: PMC5442422 DOI: 10.1038/onc.2016.423] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/06/2016] [Accepted: 10/03/2016] [Indexed: 12/22/2022]
Abstract
Caspase-2, one of the most evolutionarily conserved of the caspase family, has been implicated in maintenance of chromosomal stability and tumour suppression. Caspase-2 deficient (Casp2−/−) mice develop normally but show premature ageing-related traits and when challenged by certain stressors, succumb to enhanced tumour development and aneuploidy. To test how caspase-2 protects against chromosomal instability, we utilized an ex vivo system for aneuploidy where primary splenocytes from Casp2−/− mice were exposed to anti-mitotic drugs and followed up by live cell imaging. Our data show that caspase-2 is required for deleting mitotically aberrant cells. Acute silencing of caspase-2 in cultured human cells recapitulated these results. We further generated Casp2C320S mutant mice to demonstrate that caspase-2 catalytic activity is essential for its function in limiting aneuploidy. Our results provide direct evidence that the apoptotic activity of caspase-2 is necessary for deleting cells with mitotic aberrations to limit aneuploidy.
Collapse
Affiliation(s)
- S Dawar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Y Lim
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - J Puccini
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia.,Departments of Biochemistry and Molecular Pharmacology and Medicine, New York University, New York City, NY, USA
| | - M White
- SA Genome Editing Facility, School of Biological Sciences and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - P Thomas
- SA Genome Editing Facility, School of Biological Sciences and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - L Bouchier-Hayes
- Department of Pediatrics-Hematology, Baylor College of Medicine, Houston, TX, USA
| | - D R Green
- Immunology Department, St Jude Children's Research Hospital, Memphis, TN, USA
| | - L Dorstyn
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - S Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
47
|
Radke JR, Siddiqui ZK, Figueroa I, Cook JL. E1A enhances cellular sensitivity to DNA-damage-induced apoptosis through PIDD-dependent caspase-2 activation. Cell Death Discov 2016; 2:16076. [PMID: 27833761 PMCID: PMC5086486 DOI: 10.1038/cddiscovery.2016.76] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/19/2016] [Indexed: 11/22/2022] Open
Abstract
Expression of the adenoviral protein, E1A, sensitizes mammalian cells to a wide variety of apoptosis-inducing agents through multiple cellular pathways. For example, E1A sensitizes cells to apoptosis induced by TNF-superfamily members by inhibiting NF-kappa B (NF-κB)-dependent gene expression. In contrast, E1A sensitization to nitric oxide, an inducer of the intrinsic apoptotic pathway, is not dependent upon repression of NF-κB-dependent transcription but rather is dependent upon caspase-2 activation. The latter observation suggested that E1A-induced enhancement of caspase-2 activation might be a critical factor in cellular sensitization to other intrinsic apoptosis pathway-inducing agents. Etoposide and gemcitabine are two DNA damaging agents that induce intrinsic apoptosis. Here we report that E1A-induced sensitization to both of these agents, like NO, is independent of NF-κB activation but dependent on caspase-2 activation. The results show that caspase-2 is a key mitochondrial-injuring caspase during etoposide and gemcitabine-induced apoptosis of E1A-positive cells, and that caspase-2 is required for induction of caspase-3 activity by both chemotherapeutic agents. Expression of PIDD was required for caspase-2 activation, mitochondrial injury and enhanced apoptotic cell death. Furthermore, E1A-enhanced sensitivity to injury-induced apoptosis required PIDD cleavage to PIDD-CC. These results define the PIDD/caspase-2 pathway as a key apical, mitochondrial-injuring mechanism in E1A-induced sensitivity of mammalian cells to chemotherapeutic agents.
Collapse
Affiliation(s)
- Jay R Radke
- Research Section, Edward Hines, Jr. VA Hospital, 5000 S 5th Ave., Hines, IL 60141, USA; Division of Infectious Diseases, Loyola University Medical Center; Infectious Diseases and Immunology Research Institute, Loyola University Chicago-Stritch School of Medicine, Maywood, IL, USA; Department of Microbiology and Immunology, Loyola University Chicago-Stritch School of Medicine, Maywood, IL, USA
| | - Zeba K Siddiqui
- Department of Medicine, Section of Infectious Diseases, University of Illinois at Chicago , Chicago, IL, USA
| | - Iris Figueroa
- Department of Microbiology and Immunology, Loyola University Chicago-Stritch School of Medicine , Maywood, IL, USA
| | - James L Cook
- Research Section, Edward Hines, Jr. VA Hospital, 5000 S 5th Ave., Hines, IL 60141, USA; Division of Infectious Diseases, Loyola University Medical Center; Infectious Diseases and Immunology Research Institute, Loyola University Chicago-Stritch School of Medicine, Maywood, IL, USA; Department of Microbiology and Immunology, Loyola University Chicago-Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
48
|
Jang TH, Lim IH, Kim CM, Choi JY, Kim EA, Lee TJ, Park HH. Rescuing neuronal cell death by RAIDD- and PIDD- derived peptides and its implications for therapeutic intervention in neurodegenerative diseases. Sci Rep 2016; 6:31198. [PMID: 27502430 PMCID: PMC4977500 DOI: 10.1038/srep31198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/14/2016] [Indexed: 11/28/2022] Open
Abstract
Caspase-2 is known to be involved in oxidative-stress mediated neuronal cell death. In this study, we demonstrated that rotenone-induced neuronal cell death is mediated by caspase-2 activation via PIDDosome formation. Our newly designed TAT-fused peptides, which contains wild-type helix number3 (H3) from RAIDD and PIDD, blocked the PIDDosome formation in vitro. Furthermore, peptides inhibited rotenone-induced caspase-2-dependent apoptosis in neuronal cells. These results suggest that PIDD- or RAIDD-targeted peptides might be effective at protecting against rotenone-induced neurotoxicity. Our peptides are novel neuronal cell apoptosis inhibitors that might serve as a prototype for development of drugs for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Tae-Ho Jang
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan 38541, South Korea
| | - In-Hye Lim
- Department of Anatomy, College of Medicine, Yeungnam University, 317-1 Daemyung-Dong Nam-Gu, Daegu 42415, South Korea
| | - Chang Min Kim
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan 38541, South Korea
| | - Jae Young Choi
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan 38541, South Korea
| | - Eun-Ae Kim
- Department of Anatomy, College of Medicine, Yeungnam University, 317-1 Daemyung-Dong Nam-Gu, Daegu 42415, South Korea
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, 317-1 Daemyung-Dong Nam-Gu, Daegu 42415, South Korea
| | - Hyun Ho Park
- School of Biotechnology and Graduate School of Biochemistry at Yeungnam University, Gyeongsan 38541, South Korea
| |
Collapse
|
49
|
Ahmed K, Tabuchi Y, Kondo T. Hyperthermia: an effective strategy to induce apoptosis in cancer cells. Apoptosis 2016; 20:1411-9. [PMID: 26354715 DOI: 10.1007/s10495-015-1168-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Heat has been used as a medicinal and healing modality throughout human history. The combination of hyperthermia (HT) with radiation and anticancer agents has been used clinically and has shown positive results to a certain extent. However, the clinical results of HT treatment alone have been only partially satisfactory. Cell death following HT treatment is a function of both temperature and treatment duration. HT induces cancer cell death through apoptosis; the degree of apoptosis and the apoptotic pathway vary in different cancer cell types. HT-induced reactive oxygen species production are responsible for apoptosis in various cell types. However, the underlying mechanism of signal transduction and the genes related to this process still need to be elucidated. In this review, we summarize the molecular mechanism of apoptosis induced by HT, enhancement of heat-induced apoptosis, and the genetic network involved in HT-induced apoptosis.
Collapse
Affiliation(s)
- Kanwal Ahmed
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetic Research, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
50
|
Hirasaka K, Mills EM, Haruna M, Bando A, Ikeda C, Abe T, Kohno S, Nowinski SM, Lago CU, Akagi KI, Tochio H, Ohno A, Teshima-Kondo S, Okumura Y, Nikawa T. UCP3 is associated with Hax-1 in mitochondria in the presence of calcium ion. Biochem Biophys Res Commun 2016; 472:108-13. [PMID: 26915802 DOI: 10.1016/j.bbrc.2016.02.075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 10/22/2022]
Abstract
Uncoupling protein 3 (UCP3) is known to regulate energy dissipation, proton leakage, fatty acid oxidation, and oxidative stress. To identify the putative protein regulators of UCP3, we performed yeast two-hybrid screens. Here we report that UCP3 interacted with HS-1 associated protein X-1 (Hax-1), an anti-apoptotic protein that was localized in the mitochondria, and is involved in cellular responses to Ca(2+). The hydrophilic sequences within loop 2, and the matrix-localized hydrophilic domain of mouse UCP3, were necessary for binding to Hax-1 at the C-terminal domain, adjacent to the mitochondrial inner membrane. Interestingly, interaction of these proteins occurred in a calcium-dependent manner. Moreover, the NMR spectrum of the C-terminal domain of Hax-1 was dramatically changed by removal of Ca(2+), suggesting that the C-terminal domain of Hax-1 underwent a Ca(2+)-induced conformational change. In the Ca(2+)-free state, the C-terminal Hax-1 tended to unfold, suggesting that Ca(2+) binding may induce protein folding of the Hax-1 C-terminus. These results suggested that the UCP3-Hax-1 complex may regulate mitochondrial functional changes caused by mitochondrial Ca(2+).
Collapse
Affiliation(s)
- Katsuya Hirasaka
- Graduate School of Fisheries and Environmental Sciences, Nagasaki University, Nagasaki, Japan; Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan.
| | - Edward M Mills
- Division of Pharmacology/Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Marie Haruna
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Aki Bando
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Chika Ikeda
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Tomoki Abe
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Shohei Kohno
- Division of Pharmacology/Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Sara M Nowinski
- Division of Pharmacology/Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Cory U Lago
- Translational Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ken-Ichi Akagi
- Section of Laboratory Equipment, National Institute of Biomedical Innovation, Osaka, Japan
| | - Hidehito Tochio
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Ayako Ohno
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Shigetada Teshima-Kondo
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | - Yuushi Okumura
- Department of Nutrition and Health, Sagami Woman's University, Kanagawa, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| |
Collapse
|