1
|
Marynowicz W, Borski N, Flis Z, Ptak A, Molik E. Orotic acid induces apoptotic death in ovarian adult granulosa tumour cells and increases mitochondrial activity in normal ovarian granulosa cells. Reprod Biol 2023; 23:100790. [PMID: 37478515 DOI: 10.1016/j.repbio.2023.100790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Orotic acid (OA) is a natural product that acts as a precursor in the pyrimidine nucleotide biosynthesis pathway. Most studies concerning administration of OA focus on its therapeutic effects; however, its effect on tumours is unclear. We aimed to determine whether treatment with OA influences the viability and apoptosis of normal (HGrC1) and tumour-derived (KGN) human ovarian granulosa cells. The effects of OA (10-250 μM) on viability and apoptosis of both cell lines were determined by using alamarBlue and assessing caspase-3/7 activity, respectively. Annexin V binding and loss of membrane integrity were evaluated in KGN cells. The cell cycle and proliferation of HGrC1 cells were assessed by performing flow cytometric and DNA content analyses, respectively. The influence of OA (10 and 100 μM) on cell cycle- and apoptosis-related gene expression was assessed by RT-qPCR in both cell lines. Mitochondrial activity was analysed by JC-1 staining in HGrC1 cells. In KGN cells, OA reduced viability and increased caspase-3/7 activity, but did not affect mRNA expression of Caspase 3, BAX, and BCL2. OA enhanced proliferation and mitochondrial activity in HGrC1 cells without activating apoptosis. This study demonstrates that the anti-cancer properties of OA in ovarian granulosa tumour cells are not related to changes in apoptosis-associated gene expression, but to increased caspase-3/7 activity. Thus, OA is a promising therapeutic agent for ovarian granulosa tumours. Further, our results suggest that differences in basal expression of cell cycle- and apoptosis-related genes between the two cell lines are responsible for their different responses to OA.
Collapse
Affiliation(s)
- Weronika Marynowicz
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - Norbert Borski
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - Zuzanna Flis
- Department of Animal Nutrition and Biotechnology, and Fisheries, Faculty of Animal Science, University of Agriculture in Krakow, al. Mickiewicza 21, 31-120 Krakow, Poland
| | - Anna Ptak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - Edyta Molik
- Department of Animal Nutrition and Biotechnology, and Fisheries, Faculty of Animal Science, University of Agriculture in Krakow, al. Mickiewicza 21, 31-120 Krakow, Poland.
| |
Collapse
|
2
|
Valdez L, Cheng B, Gonzalez D, Rodriguez R, Campano P, Tsin A, Fang X. Combined treatment with niclosamide and camptothecin enhances anticancer effect in U87 MG human glioblastoma cells. Oncotarget 2022; 13:642-658. [PMID: 35548329 PMCID: PMC9084225 DOI: 10.18632/oncotarget.28227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Laura Valdez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- These authors contributed equally to this work
| | - Benxu Cheng
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- These authors contributed equally to this work
| | - Daniela Gonzalez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Reanna Rodriguez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Paola Campano
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Andrew Tsin
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Xiaoqian Fang
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
3
|
The p53-caspase-2 axis in the cell cycle and DNA damage response. Exp Mol Med 2021; 53:517-527. [PMID: 33854186 PMCID: PMC8102494 DOI: 10.1038/s12276-021-00590-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/08/2023] Open
Abstract
Caspase-2 was discovered almost three decades ago. It was one of the first two mammalian homologs of CED-3, the other being interleukin 1β-converting enzyme (ICE/caspase-1). Despite high similarity with CED-3 and its fly and mammalian counterparts (DRONC and caspase-9, respectively), the function of caspase-2 in apoptosis has remained enigmatic. A number of recent studies suggest that caspase-2 plays an important role in the regulation of p53 in response to cellular stress and DNA damage to prevent the proliferation and accumulation of damaged or aberrant cells. Here, we review these recent observations and their implications in caspase-2-mediated cellular death, senescence, and tumor suppression.
Collapse
|
4
|
Machine learning and data mining frameworks for predicting drug response in cancer: An overview and a novel in silico screening process based on association rule mining. Pharmacol Ther 2019; 203:107395. [DOI: 10.1016/j.pharmthera.2019.107395] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
|
5
|
Egorshina AY, Zamaraev AV, Lavrik IN, Zhivotovsky BD, Kopeina GS. Caspase-2 as an Oncosupressor and Metabolism Regulator: What Life Will Bring over the Long Run? Mol Biol 2018. [DOI: 10.1134/s0026893318050060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Forsberg J, Li X, Zamaraev AV, Panaretakis T, Zhivotovsky B, Olsson M. Caspase-2 associates with FAN through direct interaction and overlapping functionality. Biochem Biophys Res Commun 2018; 499:822-828. [PMID: 29621545 DOI: 10.1016/j.bbrc.2018.03.230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 03/31/2018] [Indexed: 11/30/2022]
Abstract
Caspase-2 has been implicated in diverse cellular processes, and the identification of factors with which it interacts has steadily increased. In the present study, we report a direct interaction between caspase-2 and factor associated with neutral sphingomyelinase activation (FAN) using yeast two-hybrid screening and co-immunoprecipitation. Further, stable suppression of caspase-2 expression in HEK293T and HeLa cells enabled a systematic investigation of putative novel enzyme functionalities, especially with respect to ceramide production, cell migration, IL-6 production and vesicular homeostasis, all of which have been previously reported to be associated with FAN. Lipidomics excluded the involvement of caspase-2 in the generation of ceramide species, but caspase-2-dependent deregulation of IL-6 release, vesicular size and delayed cell relocation supported an association between caspase-2 and FAN. Collectively, these data identify a novel caspase-2-interacting factor, FAN, and expand the role for the enzyme in seemingly non-apoptotic cellular mechanisms.
Collapse
Affiliation(s)
- Jeremy Forsberg
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xinge Li
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Boris Zhivotovsky
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.
| | - Magnus Olsson
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Forsberg J, Li X, Akpinar B, Salvatori R, Ott M, Zhivotovsky B, Olsson M. A caspase-2-RFXANK interaction and its implication for MHC class II expression. Cell Death Dis 2018; 9:80. [PMID: 29362422 PMCID: PMC5833739 DOI: 10.1038/s41419-017-0144-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022]
Abstract
Despite recent achievements implicating caspase-2 in tumor suppression, the enzyme stands out from the apoptotic caspase family as a factor whose function requires further clarification. To specify enzyme characteristics through the definition of interacting proteins in apoptotic or non-apoptotic settings, a yeast 2-hybrid (Y2H) screen was performed using the full-length protein as bait. The current report describes the analysis of a captured prey and putative novel caspase-2 interacting factor, the regulatory factor X-associated ankyrin-containing protein (RFXANK), previously associated with CIITA, the transactivator regulating cell-type specificity and inducibility of MHC class II gene expression. The interaction between caspase-2 and RFXANK was verified by co-immunoprecipitations using both exogenous and endogenous proteins, where the latter approach suggested that binding of the components occurs in the cytoplasm. Cellular co-localization was confirmed by transfection of fluorescently conjugated proteins. Enhanced caspase-2 processing in RFXANK-overexpressing HEK293T cells treated with chemotherapeutic agents further supported Y2H data. Yet, no distinct differences with respect to MHC class II expression were observed in plasma membranes of antigen-presenting cells derived from wild type and caspase-2-/- mice. In contrast, increased levels of the total MHC class II protein was evident in protein lysates from caspase-2 RNAi-silenced leukemia cell lines and B-cells isolated from gene-targeted mice. Together, these data identify a novel caspase-2-interacting factor, RFXANK, and indicate a potential non-apoptotic role for the enzyme in the control of MHC class II gene regulation.
Collapse
Affiliation(s)
- Jeremy Forsberg
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xinge Li
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Birce Akpinar
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Roger Salvatori
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Martin Ott
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Boris Zhivotovsky
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden. .,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.
| | - Magnus Olsson
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Caspase-2 is required for skeletal muscle differentiation and myogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:95-104. [DOI: 10.1016/j.bbamcr.2017.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/20/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023]
|
9
|
Forsberg J, Zhivotovsky B, Olsson M. Caspase-2: an orphan enzyme out of the shadows. Oncogene 2017; 36:5441-5444. [PMID: 28581521 DOI: 10.1038/onc.2017.169] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/18/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
Abstract
Caspase-2 has been embodied as an initiator or executioner protease in diverse apoptotic scenarios. However, accumulating evidence is challenging this view, pertaining to its true role. The enzyme's catalytic activity is currently implicated in various functions required for correct cell proliferation, such as counteracting genomic instability, as well as suppressing tumorigenesis. Here, apart from summarizing the latest observations in caspase-2-related research, we make an attempt to reconcile these findings and discuss their implications for future directions.
Collapse
Affiliation(s)
- J Forsberg
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - B Zhivotovsky
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - M Olsson
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
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
|
11
|
Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat Rev Mol Cell Biol 2016; 17:280-92. [PMID: 27033256 DOI: 10.1038/nrm.2016.27] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The roles of cyclins and their catalytic partners, the cyclin-dependent kinases (CDKs), as core components of the machinery that drives cell cycle progression are well established. Increasing evidence indicates that mammalian cyclins and CDKs also carry out important functions in other cellular processes, such as transcription, DNA damage repair, control of cell death, differentiation, the immune response and metabolism. Some of these non-canonical functions are performed by cyclins or CDKs, independently of their respective cell cycle partners, suggesting that there was a substantial divergence in the functions of these proteins during evolution.
Collapse
|
12
|
Burnham ME, Esnault S, Roti Roti EC, Bates ME, Bertics PJ, Denlinger LC. Cholesterol selectively regulates IL-5 induced mitogen activated protein kinase signaling in human eosinophils. PLoS One 2014; 9:e103122. [PMID: 25121926 PMCID: PMC4133209 DOI: 10.1371/journal.pone.0103122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 06/26/2014] [Indexed: 01/21/2023] Open
Abstract
Eosinophils function contributes to human allergic and autoimmune diseases, many of which currently lack curative treatment. Development of more effective treatments for eosinophil-related diseases requires expanded understanding of eosinophil signaling and biology. Cell signaling requires integration of extracellular signals with intracellular responses, and is organized in part by cholesterol rich membrane microdomains (CRMMs), commonly referred to as lipid rafts. Formation of these organizational membrane domains is in turn dependent upon the amount of available cholesterol, which can fluctuate widely with a variety of disease states. We tested the hypothesis that manipulating membrane cholesterol content in primary human peripheral blood eosinophils (PBEos) would selectively alter signaling pathways that depend upon membrane-anchored signaling proteins localized within CRMMs (e.g., mitogen activated protein kinase [MAPK] pathway), while not affecting pathways that signal through soluble proteins, like the Janus Kinase/Signal Transducer and Activator of Transcription [JAK/STAT] pathway. Cholesterol levels were increased or decreased utilizing cholesterol-chelating methyl-β-cyclodextrin (MβCD), which can either extract membrane cholesterol or add exogenous membrane cholesterol depending on whether MβCD is preloaded with cholesterol. Human PBEos were pretreated with MβCD (cholesterol removal) or MβCD+Cholesterol (MβCD+Chol; cholesterol delivery); subsequent IL-5-stimulated signaling and physiological endpoints were assessed. MβCD reduced membrane cholesterol in PBEos, and attenuated an IL-5-stimulated p38 and extracellular-regulated kinase 1/2 phosphorylation (p-p38, p-ERK1/2), and an IL-5-dependent increase in interleukin-1β (IL-1β) mRNA levels. In contrast, MβCD+Chol treatment elevated PBEos membrane cholesterol levels and basal p-p38, but did not alter IL-5-stimulated phosphorylation of ERK1/2, STAT5, or STAT3. Furthermore, MβCD+Chol pretreatment attenuated an IL-5-induced increase in cell survival at 48 hours, measured as total cellular metabolism. The reduction in cell survival following cholesterol addition despite unaltered STAT phosphorylation contradicts the current dogma in which JAK/STAT activation is sufficient to promote eosinophil survival, and suggests an additional, unidentified mechanism critically regulates IL-5-mediated human PBEos survival.
Collapse
Affiliation(s)
- Mandy E. Burnham
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Stephane Esnault
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Elon C. Roti Roti
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Mary E. Bates
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Paul J. Bertics
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Loren C. Denlinger
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| |
Collapse
|
13
|
Cyclin D3 promotes pancreatic β-cell fitness and viability in a cell cycle-independent manner and is targeted in autoimmune diabetes. Proc Natl Acad Sci U S A 2014; 111:E3405-14. [PMID: 25092329 DOI: 10.1073/pnas.1323236111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is an autoimmune condition caused by the lymphocyte-mediated destruction of the insulin-producing β cells in pancreatic islets. We aimed to identify final molecular entities targeted by the autoimmune assault on pancreatic β cells that are causally related to β cell viability. Here, we show that cyclin D3 is targeted by the autoimmune attack on pancreatic β cells in vivo. Cyclin D3 is down-regulated in a dose-dependent manner in β cells by leukocyte infiltration into the islets of the nonobese diabetic (NOD) type 1 diabetes-prone mouse model. Furthermore, we established a direct in vivo causal link between cyclin D3 expression levels and β-cell fitness and viability in the NOD mice. We found that changes in cyclin D3 expression levels in vivo altered the β-cell apoptosis rates, β-cell area homeostasis, and β-cell sensitivity to glucose without affecting β-cell proliferation in the NOD mice. Cyclin D3-deficient NOD mice exhibited exacerbated diabetes and impaired glucose responsiveness; conversely, transgenic NOD mice overexpressing cyclin D3 in β cells exhibited mild diabetes and improved glucose responsiveness. Overexpression of cyclin D3 in β cells of cyclin D3-deficient mice rescued them from the exacerbated diabetes observed in transgene-negative littermates. Moreover, cyclin D3 overexpression protected the NOD-derived insulinoma NIT-1 cell line from cytokine-induced apoptosis. Here, for the first time to our knowledge, cyclin D3 is identified as a key molecule targeted by autoimmunity that plays a nonredundant, protective, and cell cycle-independent role in β cells against inflammation-induced apoptosis and confers metabolic fitness to these cells.
Collapse
|
14
|
Olsson M, Forsberg J, Zhivotovsky B. Caspase-2: the reinvented enzyme. Oncogene 2014; 34:1877-82. [DOI: 10.1038/onc.2014.139] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/16/2014] [Accepted: 04/16/2014] [Indexed: 12/11/2022]
|
15
|
Kim PY, Rahmanto AS, Tan O, Norris MD, Haber M, Marshall GM, Cheung BB. TRIM16 overexpression induces apoptosis through activation of caspase-2 in cancer cells. Apoptosis 2013; 18:639-51. [PMID: 23404198 PMCID: PMC3618413 DOI: 10.1007/s10495-013-0813-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
TRIM16 exhibits tumour suppressor functions by interacting with cytoplasmic vimentin and nuclear E2F1 proteins in neuroblastoma and squamous cell carcinoma cells, reducing cell migration and replication. Reduced TRIM16 expression in a range of human primary malignant tissues correlates with increased malignant potential. TRIM16 also induces apoptosis in breast and lung cancer cells, by unknown mechanisms. Here we show that overexpression of TRIM16 induces apoptosis in human breast cancer (MCF7) and neuroblastoma (BE(2)-C) cells, but not in non-malignant HEK293 cells. TRIM16 increased procaspase-2 protein levels in MCF7 and induced caspase-2 activity in both MCF7 and BE(2)-C cells. We show that TRIM16 and caspase-2 proteins directly interact in both MCF7 and BE(2)-C cells and co-localise in MCF7 cells. Most importantly, the induction of caspase-2 activity is required for TRIM16 to initiate apoptosis. Our data suggest a novel mechanism by which TRIM16 can promote apoptosis by directly modulating caspase-2 activity.
Collapse
Affiliation(s)
- Patrick Y Kim
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | | | | | | | | | | | | |
Collapse
|
16
|
Zhang F, Yin W, Chen J. Apoptosis in cerebral ischemia: executional and regulatory signaling mechanisms. Neurol Res 2013; 26:835-45. [PMID: 15727267 DOI: 10.1179/016164104x3824] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Programmed cell death, often in the form of apoptosis, is an important contributing mechanism in the pathogenesis of ischemic brain injury. Depending on the severity of the insult and the stage of the injury, the executional pathways that are directly responsible for cell death and the signaling mechanisms that participate in the regulation of these death pathways may vary. It is likely that molecular or pharmacological targeting of the upstream signaling mechanisms that control the death executional pathways may offer opportunities for more complete and long-term neuroprotection. This review summarizes the recent advancements in the understanding of the executional and regulatory signaling mechanisms in ischemic brain injury.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neurology and Institute of Neurodegenerative Disorders University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania 15213, USA
| | | | | |
Collapse
|
17
|
Poehlmann A, Reissig K, Just A, Walluscheck D, Hartig R, Schinlauer A, Lessel W, Guenther T, Silver A, Steinberg P, Roessner A. Non-apoptotic function of caspases in a cellular model of hydrogen peroxide-associated colitis. J Cell Mol Med 2013; 17:901-13. [PMID: 23742011 PMCID: PMC3822895 DOI: 10.1111/jcmm.12079] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 04/15/2013] [Indexed: 01/26/2023] Open
Abstract
Oxidative stress, caused by reactive oxygen species (ROS), is a major contributor to inflammatory bowel disease (IBD)-associated neoplasia. We mimicked ROS exposure of the epithelium in IBD using non-tumour human colonic epithelial cells (HCEC) and hydrogen peroxide (H2 O2 ). A population of HCEC survived H2 O2 -induced oxidative stress via JNK-dependent cell cycle arrests. Caspases, p21(WAF1) and γ-H2AX were identified as JNK-regulated proteins. Up-regulation of caspases was linked to cell survival and not, as expected, to apoptosis. Inhibition using the pan-caspase inhibitor Z-VAD-FMK caused up-regulation of γ-H2AX, a DNA-damage sensor, indicating its negative regulation via caspases. Cell cycle analysis revealed an accumulation of HCEC in the G1 -phase as first response to oxidative stress and increased S-phase population and then apoptosis as second response following caspase inhibition. Thus, caspases execute a non-apoptotic function by promoting cells through G1 - and S-phase by overriding the G1 /S- and intra-S checkpoints despite DNA-damage. This led to the accumulation of cells in the G2 /M-phase and decreased apoptosis. Caspases mediate survival of oxidatively damaged HCEC via γ-H2AX suppression, although its direct proteolytic inactivation was excluded. Conversely, we found that oxidative stress led to caspase-dependent proteolytic degradation of the DNA-damage checkpoint protein ATM that is upstream of γ-H2AX. As a consequence, undetected DNA-damage and increased proliferation were found in repeatedly H2 O2 -exposed HCEC. Such features have been associated with neoplastic transformation and appear here to be mediated by a non-apoptotic function of caspases. Overexpression of upstream p-JNK in active ulcerative colitis also suggests a potential importance of this pathway in vivo.
Collapse
Affiliation(s)
- Angela Poehlmann
- Department of Pathology, Otto-von-Guericke University, Magdeburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Taghiyev AF, Rokhlin OW, Glover RB. Caspase-2-Based Regulation of the Androgen Receptor and Cell Cycle in the Prostate Cancer Cell Line LNCaP. Genes Cancer 2012; 2:745-52. [PMID: 22207900 DOI: 10.1177/1947601911426007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 09/12/2011] [Indexed: 11/17/2022] Open
Abstract
Caspase-2 can induce apoptosis in response to extrinsic and intrinsic signals. Unlike other caspases, this protein is not expressed solely in nonnuclear compartments; a subpopulation is constitutively localized in the nucleus. As one of the most evolutionarily conserved caspases, caspase-2 may have roles in multiple cellular processes. However, its contribution to nonapoptotic processes remains a mystery. In this study, we show that caspase-2 activity is important for proliferation by cells of the androgen-dependent prostate cancer cell line LNCaP. LNCaP cells expressing either a dominant-negative (dn) form of caspase or an siRNA against caspase-2 had lower androgen receptor (AR)-dependent proliferative responses than control cells, and application of the siRNA resulted in downregulation of the expression of both AR-dependent prostate-specific antigen (PSA) and AR-dependent reporter luciferase. Also, caspase-2 formed complexes with the cell cycle regulatory proteins cyclin D3, CDK4, and p21/Cip1, and caspase-2 regulated AR transactivation by inhibiting the repressive function of cyclin D3. Taken together, these results reveal, for the first time, that caspase-2 is involved in cell cycle promotion and AR activation. Given that prostate cancer cells depend on AR activity in order to survive, the fact that our data indicate that caspase-2 positively regulates AR activity suggests that caspase-2 has potential as a target in the treatment of prostate cancer.
Collapse
|
20
|
Ren K, Lu J, Porollo A, Du C. Tumor-suppressing function of caspase-2 requires catalytic site Cys-320 and site Ser-139 in mice. J Biol Chem 2012; 287:14792-802. [PMID: 22396545 DOI: 10.1074/jbc.m112.347625] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The multifunctional caspase-2 protein is involved in apoptosis, NF-κB regulation, and tumor suppression in mice. However, the mechanisms of caspase-2 responsible for tumor suppression remain unclear. Here we identified two sites of caspase-2, the catalytic Cys-320 site and the Ser-139 site, to be important for suppression of cellular transformation and tumorigenesis. Using SV40- and K-Ras-transformed caspase-2 KO mouse embryonic fibroblast cells reconstituted with expression of wild-type, catalytic dead (C320A), or Ser-139 (S139A) mutant caspase-2, we demonstrated that similar to caspase-2 deficiency, when Cys-320 and Ser-139 were mutated, caspase-2 lost its ability to inhibit cellular transformation and tumorigenesis. These mutant cells exhibited enhanced cell proliferation, elevated clonogenic activity, accelerated anchorage-independent growth, and transformation and were highly tumorigenic, rapidly producing large tumors in athymic nude mice. Investigation into the underlying mechanism showed that these two residues are needed for caspase-2 to suppress NF-κB activity, promote apoptosis, and sustain the G(2)/M checkpoint following DNA damage induction. In addition, tumors in nude mice derived from the two mutant cell lines had higher constitutive NF-κB activity and elevated expression of NF-κB targets of antiapoptotic proteins Bcl-xL, XIAP, and cIAP2. A reduction in caspase-2 mRNA was associated with multiple types of cancers in patients. Together, these observations suggest the combined functions of caspase-2 in suppressing NF-κB activation, promoting apoptosis, and sustaining G(2)/M checkpoint contribute to caspase-2 tumor-suppressing function and that caspase-2 may also impact tumor suppression in humans. These findings provide insight into tumor suppression at the cross-roads of apoptosis, cell cycle checkpoint, and NF-κB pathways.
Collapse
Affiliation(s)
- Keqin Ren
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | | | | | | |
Collapse
|
21
|
Sherman-Baust CA, Becker KG, Wood III WH, Zhang Y, Morin PJ. Gene expression and pathway analysis of ovarian cancer cells selected for resistance to cisplatin, paclitaxel, or doxorubicin. J Ovarian Res 2011; 4:21. [PMID: 22141344 PMCID: PMC3259089 DOI: 10.1186/1757-2215-4-21] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 12/05/2011] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Resistance to current chemotherapeutic agents is a major cause of therapy failure in ovarian cancer patients, but the exact mechanisms leading to the development of drug resistance remain unclear. METHODS To better understand mechanisms of drug resistance, and possibly identify novel targets for therapy, we generated a series of drug resistant ovarian cancer cell lines through repeated exposure to three chemotherapeutic drugs (cisplatin, doxorubicin, or paclitaxel), and identified changes in gene expression patterns using Illumina whole-genome expression microarrays. Validation of selected genes was performed by RT-PCR and immunoblotting. Pathway enrichment analysis using the KEGG, GO, and Reactome databases was performed to identify pathways that may be important in each drug resistance phenotype. RESULTS A total of 845 genes (p < 0.01) were found altered in at least one drug resistance phenotype when compared to the parental, drug sensitive cell line. Focusing on each resistance phenotype individually, we identified 460, 366, and 337 genes significantly altered in cells resistant to cisplatin, doxorubicin, and paclitaxel, respectively. Of the 845 genes found altered, only 62 genes were simultaneously altered in all three resistance phenotypes. Using pathway analysis, we found many pathways enriched for each resistance phenotype, but some dominant pathways emerged. The dominant pathways included signaling from the cell surface and cell movement for cisplatin resistance, proteasome regulation and steroid biosynthesis for doxorubicin resistance, and control of translation and oxidative stress for paclitaxel resistance. CONCLUSIONS Ovarian cancer cells develop drug resistance through different pathways depending on the drug used in the generation of chemoresistance. A better understanding of these mechanisms may lead to the development of novel strategies to circumvent the problem of drug resistance.
Collapse
Affiliation(s)
- Cheryl A Sherman-Baust
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore MD 21224, USA
| | - Kevin G Becker
- Research Resource Branch, National Institute on Aging, Baltimore MD 21224, USA
| | - William H Wood III
- Research Resource Branch, National Institute on Aging, Baltimore MD 21224, USA
| | - Yongqing Zhang
- Research Resource Branch, National Institute on Aging, Baltimore MD 21224, USA
| | - Patrice J Morin
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore MD 21224, USA
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| |
Collapse
|
22
|
Mijatovic SA, Timotijevic GS, Miljkovic DM, Radovic JM, Maksimovic-Ivanic DD, Dekanski DP, Stosic-Grujicic SD. Multiple antimelanoma potential of dry olive leaf extract. Int J Cancer 2011; 128:1955-65. [PMID: 20568104 DOI: 10.1002/ijc.25526] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Various constituents of the olive tree (Olea europaea) have been traditionally used in the treatment of infection, inflammation, prevention of chronic diseases, cardiovascular disorders and cancer. The anticancer potential of dry olive leaf extract (DOLE) represents the net effect of multilevel interactions between different biologically active compounds from the extract, cancer cells and conventional therapy. In this context, it was of primary interest to evaluate the influence of DOLE on progression of the highly malignant, immuno- and chemoresistant type of skin cancer-melanoma. DOLE significantly inhibited proliferation and subsequently restricted clonogenicity of the B16 mouse melanoma cell line in vitro. Moreover, late phase tumor treatment with DOLE significantly reduced tumor volume in a syngeneic strain of mice. DOLE-treated B16 cells were blocked in the G(0) /G(1) phase of the cell cycle, underwent early apoptosis and died by late necrosis. At the molecular level, the dying process started as caspase dependent, but finalized as caspase independent. In concordance, overexpression of antiapoptotic members of the Bcl-2 family, Bcl-2 and Bcl-XL, and diminished expression of their natural antagonists, Bim and p53, were observed. Despite molecular suppression of the proapoptotic process, DOLE successfully promoted cell death mainly through disruption of cell membrane integrity and late caspase-independent fragmentation of genetic material. Taken together, the results of this study indicate that DOLE possesses strong antimelanoma potential. When DOLE was applied in combination with different chemotherapeutics, various outcomes, including synergy and antagonism, were observed. This requires caution in the use of the extract as a supplementary antitumor therapeutic.
Collapse
Affiliation(s)
- Sanja A Mijatovic
- Department of Immunology, Institute for Biological Research, Belgrade University, Belgrade, Serbia.
| | | | | | | | | | | | | |
Collapse
|
23
|
Woldemichael GM, Turbyville TJ, Linehan WM, McMahon JB. Carminomycin I is an apoptosis inducer that targets the Golgi complex in clear cell renal carcinoma cells. Cancer Res 2011; 71:134-42. [PMID: 21199801 DOI: 10.1158/0008-5472.can-10-0757] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clear cell renal cell carcinoma (CCRCC) evolves due to mutations in the Von Hippel-Lindau (VHL) tumor suppressor gene. Although the loss of VHL enables survival and proliferation of CCRCC cells, it is also expected to introduce vulnerabilities that may be exploited for therapeutics discovery. To this end, we developed a high-throughput screen to identify small molecules derived from plants, microorganisms, and marine organisms to which CCRCC cells are sensitive. Screening over 8,000 compounds using this approach, we report here the identification of the microbially derived compound carminomycin I (CA) as an effective inhibitor of VHL-defective (VHL(-/-)) CCRCC cell proliferation. CA also induced apoptosis in CCRCC cells by a mechanism independent of p53 or hypoxia-inducible factor 2. We found that P-glycoprotein (P-gp) sequestered CA within the Golgi complex. Interestingly, Golgi sequestration was critical for the antiproliferative effects of CA and P-gp inhibitors abrogated this activity. Furthermore, CA induced cleavage of the Golgi protein p115 and the translocation of its C-terminal fragment to the nucleus. Finally, examination of the activity of the VHL-interacting Golgi protein, endoplasmic reticulum-Golgi intermediate compartment, ERGIC-53 showed that VHL could mediate protection from CA in CCRCC cells. Our natural product-based screening approach has revealed the P-gp-mediated localization of anticancer compounds within the Golgi in CCRCC cells as a potential strategy of targeting VHL-deficient CCRCC cells.
Collapse
Affiliation(s)
- Girma M Woldemichael
- Molecular Targets Laboratory, SAIC-Frederick, Inc., National Cancer Institute, Frederick, Maryland 21702, USA.
| | | | | | | |
Collapse
|
24
|
Duan Y, He X, Yang H, Ji Y, Tao T, Chen J, Hu L, Zhang F, Li X, Wang H, Shen A, Lu X. Cyclin D3/CDK11(p58) complex involved in Schwann cells proliferation repression caused by lipopolysaccharide. Inflammation 2010; 33:189-99. [PMID: 20066559 DOI: 10.1007/s10753-009-9173-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Schwann cells proliferation is the main characterize of kinds PNS inflammation diseases. It has been well documented that cyclin D3 /CDK11(p58) complex inhibits cell function through multiple mechanisms, but the mechanism of cyclin D3/CDK11(p58) complex exerts its repressive role in the Schwann cells proliferation remains to be identified. In the present investigation, we demonstrated that the expression of CDK11(p58) were upregulated in the inflammation caused by LPS, a main part of bacteria. Cyclin D3 and the 58-kDa isoform of cyclin-dependent kinase 11 (CDK11(p58)) interacted with each other mainly in nuclear region, repressed Schwann cells proliferation and induced cell apoptosis. Overexpression of CDK11(p58) expression might enhance this process, while silence of cyclin D3 reverting it. This work demonstrates for the first time the role of cyclin D3/CDK11(p58) complex in repressing the Schwann cells proliferation and inducing its apoptosis.
Collapse
Affiliation(s)
- Yinong Duan
- Laboratory Center, Affiliated Hospital of Nantong University, and Department of Parasitology and Microbiology, Medical College, Nantong University, 19 Qixiu Road, Nantong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cornils H, Stegert MR, Hergovich A, Hynx D, Schmitz D, Dirnhofer S, Hemmings BA. Ablation of the kinase NDR1 predisposes mice to the development of T cell lymphoma. Sci Signal 2010; 3:ra47. [PMID: 20551432 DOI: 10.1126/scisignal.2000681] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Defective apoptosis contributes to the development of various human malignancies. The kinases nuclear Dbf2-related 1 (NDR1) and NDR2 mediate apoptosis downstream of the tumor suppressor proteins RASSF1A (Ras association domain family member 1A) and MST1 (mammalian Ste20-like kinase 1). To further analyze the role of NDR1 in apoptosis, we generated NDR1-deficient mice. Although NDR1 is activated by both intrinsic and extrinsic proapoptotic stimuli, which indicates a role for NDR1 in regulating apoptosis, NDR1-deficient T cells underwent apoptosis in a manner similar to that of wild-type cells in response to different proapoptotic stimuli. Analysis of the abundances of NDR1 and NDR2 proteins revealed that loss of NDR1 was functionally compensated for by an increase in the abundance of NDR2 protein. Despite this compensation, NDR1(-/-) and NDR1(+/-) mice were more prone to the development of T cell lymphomas than were wild-type mice. Tumor development in mice and humans was accompanied by a decrease in the overall amounts of NDR proteins in T cell lymphoma samples. Thus, reduction in the abundance of NDR1 triggered a decrease in the total amount of both isoforms. Together, our data suggest that a reduction in the abundances of the NDR proteins results in defective responses to proapoptotic stimuli, thereby facilitating the development of tumors.
Collapse
Affiliation(s)
- Hauke Cornils
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058 Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Aberrations in proteins that control apoptosis and cell survival are common in cancer. These aberrations often reside in signalling proteins that control the activation of the apoptotic machinery or in the Bcl-2 family of proteins that control caspase activation. Recent evidence suggests that caspase 2, one of the most evolutionarily conserved caspases, may have multiple roles in the DNA damage response, cell cycle regulation and tumour suppression. These findings are unexpected and have important implications for our understanding of tumorigenesis and the treatment of cancer.
Collapse
Affiliation(s)
- Sharad Kumar
- Centre for Cancer Biology, Department of Haematology, SA Pathology, Frome Road, Adelaide, SA5000, Australia.
| |
Collapse
|
27
|
Kitevska T, Spencer DMS, Hawkins CJ. Caspase-2: controversial killer or checkpoint controller? Apoptosis 2009; 14:829-48. [PMID: 19479377 DOI: 10.1007/s10495-009-0365-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The caspases are an evolutionarily conserved family of cysteine proteases, with essential roles in apoptosis or inflammation. Caspase-2 was the second caspase to be cloned and it resembles the prototypical nematode caspase CED-3 more closely than any other mammalian protein. An absence of caspase-2-specific reagents and the subtle phenotype of caspase-2-deficient mice have hampered definition of the physiological role of caspase-2 and identification of factors regulating its activity. Although some data implicate caspase-2 in apoptotic pathways, a link with apoptosis has been less firmly established for caspase-2 than for some other caspases. Emerging evidence suggests that caspase-2 regulates the cell cycle and may act as a tumour suppressor. This article critically reviews the current state of knowledge regarding the biochemistry and biology of this controversial caspase.
Collapse
Affiliation(s)
- Tanja Kitevska
- Department of Biochemistry, La Trobe University, Bundoora, VIC 3086, Australia
| | | | | |
Collapse
|
28
|
Abstract
Apoptosis is mediated by the caspase family of proteases that act as effectors of cell death by cleaving many cellular substrates. Caspase-2 is one of the most evolutionarily conserved caspases, yet its physiological function has remained enigmatic because caspase-2-deficient mice develop normally and are viable. We report here that the caspase-2(-/-) mouse embryonic fibroblasts (MEFs) show increased proliferation. When transformed with E1A and Ras oncogenes, caspase-2(-/-) MEFs grew significantly faster than caspase-2(+/+) MEFs and formed more aggressive and accelerated tumors in nude mice. To assess whether the loss of caspase-2 predisposes animals to tumor development, we used the mouse Emu-Myc lymphoma model. Our findings suggest that loss of even a single allele of caspase-2 resulted in accelerated tumorigenesis, and this was further enhanced in caspase-2(-/-) mice. The caspase-2(-/-) cells showed resistance to apoptosis induced by chemotherapeutic drugs and DNA damage. Furthermore, caspase-2(-/-) MEFs had a defective apoptotic response to cell-cycle checkpoint regulation and showed abnormal cycling following gamma-irradiation. These data show that loss of caspase-2 results in an increased ability of cells to acquire a transformed phenotype and become malignant, indicating that caspase-2 is a tumor suppressor protein.
Collapse
|
29
|
Draper N, Bui M, Boulware DC, Lloyd M, Chiappori AA, Pledger WJ, Coppola D. Increased cyclin D3 expression significantly correlates with p27 nuclear positivity in gastrointestinal stromal tumors. Hum Pathol 2008; 39:1784-91. [DOI: 10.1016/j.humpath.2008.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2008] [Revised: 05/12/2008] [Accepted: 05/14/2008] [Indexed: 11/29/2022]
|
30
|
Krumschnabel G, Sohm B, Bock F, Manzl C, Villunger A. The enigma of caspase-2: the laymen's view. Cell Death Differ 2008; 16:195-207. [PMID: 19023332 DOI: 10.1038/cdd.2008.170] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Proteolysis of cellular substrates by caspases (cysteine-dependent aspartate-specific proteases) is one of the hallmarks of apoptotic cell death. Although the activation of apoptotic caspases is considered a 'late-stage' event in apoptosis signaling, past the commitment stage, one caspase family member, caspase-2, splits the cell death community into half - those searching for evidence of an apical initiator function of this molecule and those considering it as an amplifier of the apoptotic caspase cascade, at best, if relevant for apoptosis at all. This review screens past and present biochemical as well as genetic evidence for caspase-2 function in cell death signaling and beyond.
Collapse
Affiliation(s)
- G Krumschnabel
- Division of Developmental Immunology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | |
Collapse
|
31
|
Sidi S, Sanda T, Kennedy RD, Hagen AT, Jette CA, Hoffmans R, Pascual J, Imamura S, Kishi S, Amatruda JF, Kanki JP, Green DR, D'Andrea AA, Look AT. Chk1 suppresses a caspase-2 apoptotic response to DNA damage that bypasses p53, Bcl-2, and caspase-3. Cell 2008; 133:864-77. [PMID: 18510930 DOI: 10.1016/j.cell.2008.03.037] [Citation(s) in RCA: 260] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Revised: 01/25/2008] [Accepted: 03/24/2008] [Indexed: 12/20/2022]
Abstract
Evasion of DNA damage-induced cell death, via mutation of the p53 tumor suppressor or overexpression of prosurvival Bcl-2 family proteins, is a key step toward malignant transformation and therapeutic resistance. We report that depletion or acute inhibition of checkpoint kinase 1 (Chk1) is sufficient to restore gamma-radiation-induced apoptosis in p53 mutant zebrafish embryos. Surprisingly, caspase-3 is not activated prior to DNA fragmentation, in contrast to classical intrinsic or extrinsic apoptosis. Rather, an alternative apoptotic program is engaged that cell autonomously requires atm (ataxia telangiectasia mutated), atr (ATM and Rad3-related) and caspase-2, and is not affected by p53 loss or overexpression of bcl-2/xl. Similarly, Chk1 inhibitor-treated human tumor cells hyperactivate ATM, ATR, and caspase-2 after gamma-radiation and trigger a caspase-2-dependent apoptotic program that bypasses p53 deficiency and excess Bcl-2. The evolutionarily conserved "Chk1-suppressed" pathway defines a novel apoptotic process, whose responsiveness to Chk1 inhibitors and insensitivity to p53 and BCL2 alterations have important implications for cancer therapy.
Collapse
Affiliation(s)
- Samuel Sidi
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Nishizuka S, Ramalingam S, Spurrier B, Washburn FL, Krishna R, Honkanen P, Young L, Tsutomu S, Steeg PS, Austin J. Quantitative protein network monitoring in response to DNA damage. J Proteome Res 2008; 7:803-8. [PMID: 18173236 DOI: 10.1021/pr0702971] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Conventional molecular biology techniques have identified a large number of cell signaling pathways; however, the importance of these pathways often varies, depending on factors such as treatment type, dose, time after treatment, and cell type. Here, we describe a technique using "reverse-phase" protein lysate microarrays (RPAs) to acquire multiple dimensions of information on protein dynamics in response to DNA damage. Whole-cell lysates from three cellular stress treatments (IR, UV, and ADR) were collected at four doses per treatment, and each, in turn, at 10 time points, resulting in a single-slide RPA consisting of 10,240 features, including replicates. The dynamic molecular profile of 18 unique protein species was compared to phenotypic fate by FACS analysis for corresponding stress conditions. Our initial quantitative results in this new platform confirmed that (1) there is clear stress dose-response effect in p53 protein and (2) a comparison of the rates of increase of p21 and Cyclin D3/p53-Ser15 in response to DNA damage may be associated with the pattern of DNA content. This method, offering a quantitative time-course monitoring of protein expression levels, can provide an experimental reference for developing mathematical models of cell signaling dynamics. Although the present study focuses on the DNA damage-repair pathway, the technique is generally useful to the study of protein signaling.
Collapse
Affiliation(s)
- Satoshi Nishizuka
- Molecular Therapeutics Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mendelsohn AR. Interaction trap/two-hybrid system to identify loss-of-interaction mutant proteins. ACTA ACUST UNITED AC 2008; Chapter 20:Unit 20.8. [PMID: 18265346 DOI: 10.1002/0471142727.mb2008s65] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Protein-protein interactions play a critical role in biology. Disrupting a specific protein-protein interaction and studying the resulting phenotype can help elucidate the function of the interaction. A method to rapidly make and identify mutant proteins that no longer bind to a specific partner protein is described. The method takes advantage of the ease of the interaction trap/two-hybrid system and PCR mutagenesis. PCR fusion of the target protein to GFP is used to ensure the protein's open reading frame is not disrupted by mutagenesis. The resulting noninteracting mutant proteins usually result from a single missense mutation, which can easily be identified.
Collapse
|
34
|
Ji Y, Xiao F, Sun L, Qin J, Shi S, Yang J, Liu Y, Zhou D, Zhao J, Shen A. Increased expression of CDK11p58 and cyclin D3 following spinal cord injury in rats. Mol Cell Biochem 2007; 309:49-60. [PMID: 18008145 DOI: 10.1007/s11010-007-9642-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Accepted: 10/30/2007] [Indexed: 11/26/2022]
Abstract
Protein kinases are critical signalling molecules for normal cell growth and development. CDK11p58 is a p34cdc2-related protein kinase, and plays an important role in normal cell cycle progression. However its distribution and function in the central nervous system (CNS) lesion remain unclear. In this study, we mainly investigated the protein expression and cellular localization of CDK11 during spinal cord injury (SCI). Western blot analysis revealed that CDK11p58 was not detected in normal spinal cord. It gradually increased, reached a peak at 3 day after SCI, and then decreased. The protein expression of CDK11(p58) was further analyzed by immunohistochemistry. The variable immunostaining patterns of CDK11p58 were visualized at different periods of injury. Double immunofluorescence staining showed that CDK11 was co-expressed with NeuN, CNPase and GFAP. Co-localization of CDK11/active caspase-3 and CDK11/proliferating cell nuclear antigen (PCNA) were detected in some cells. Cyclin D3, which was associated with CDK11p58 and could enhance kinase activity, was detected in the normal and injured spinal cord. The cyclin D3 protein underwent a similar pattern with CDK11p58 during SCI. Double immunofluorescence staining indicated that CDK11 co-expressed with cyclin D3 in neurons and glial cells. Coimmunoprecipitation further showed that CDK11p58 and cyclin D3 interacted with each other in the damaged spinal cord. Thus, it is likely CDK11p58 and cyclin D3 could interact with each other after acute SCI. Another partner of CDK11p58 was beta-1,4-galactosyltransferase 1 (beta-1,4-GT 1). The co-localization of CDK11/beta-1,4-GT 1 in the damaged spinal cord was revealed by immunofluorescence analysis. The cyclin D3-CDK4 complexes were also present by coimmunoprecipitation analysis. Taken together, these data suggested that both CDK11 and cyclin D3 may play important roles in spinal cord pathophysiology.
Collapse
Affiliation(s)
- Yuhong Ji
- The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kim JY, Park OG, Lee JW, Lee YC. One- plus two-hybrid system, a novel yeast genetic selection for specific missense mutations disrupting protein/protein interactions. Mol Cell Proteomics 2007; 6:1727-40. [PMID: 17609197 DOI: 10.1074/mcp.m700079-mcp200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To facilitate analysis of protein/protein interaction interfaces, we devised a novel yeast genetic screening method, named the "one- plus two-hybrid system," for the efficient selection of missense mutations that specifically disrupt known protein/protein interactions. This system modifies the standard yeast two-hybrid system to allow the operation of dual reporter systems within the same cell. The one-hybrid system is first used to select the intact interacting partner (prey), resulting in the positive selection of informative missense mutants from a large library of randomly generated mutant alleles. Then in a second screening step, interaction-defective prey mutants for a given protein are selected using the two-hybrid reporter system among the isolated missense mutants. We used this method to characterize the interactions between unliganded nuclear receptors (NRs) and the conserved motif within the bipartite NR interaction domains (IDs) of the NR corepressor (N-CoR) and identified the specific residues of N-CoR-IDs required either generally for optimal NR binding or to interact with a particular NR. This efficient and rapid method should allow us to quickly analyze a large number of interaction interfaces.
Collapse
Affiliation(s)
- Ji Young Kim
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, South Korea
| | | | | | | |
Collapse
|
36
|
Jiang J, Wei Y, Liu D, Zhou J, Shen J, Chen X, Zhang S, Kong X, Gu J. E1AF promotes breast cancer cell cycle progression via upregulation of Cyclin D3 transcription. Biochem Biophys Res Commun 2007; 358:53-8. [PMID: 17467662 DOI: 10.1016/j.bbrc.2007.04.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Accepted: 04/10/2007] [Indexed: 10/23/2022]
Abstract
E1AF transcription factor, a member of Ets family, is deregulated in many tumors and widely known to play critical roles in tumor metastasis via directly binding to the promoter of genes involved in tumor migration and invasion. Here, we found that E1AF overexpression promoted breast cancer cell cycle progression and growth in vivo as well as the transcription of cell cycle-related protein Cyclin D3. And, the interference of Cyclin D3 expression by transfecting with Cyclin D3 RNAi inhibited the positive role of E1AF in cell cycle progression. We further showed that decreasing the expression of E1AF by E1AF RNAi reduced Cyclin D3 transcription and expression, and inhibited cell cycle progression that was abrogated by Cyclin D3 overexpression. Taken together, E1AF increases cell cycle progression via upregulation of Cyclin D3 transcription, which elicits a new mechanism of breast cancer growth and a new mechanism of Cyclin D3 transcription.
Collapse
Affiliation(s)
- Jianhai Jiang
- Key Laboratory of Glycoconjugates Research, Ministry of Public Health and Gene Research Center, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Smith L, Watson MB, O'Kane SL, Drew PJ, Lind MJ, Cawkwell L. The analysis of doxorubicin resistance in human breast cancer cells using antibody microarrays. Mol Cancer Ther 2006; 5:2115-20. [PMID: 16928833 DOI: 10.1158/1535-7163.mct-06-0190] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Doxorubicin is considered to be the most effective agent in the treatment of breast cancer patients. Unfortunately, resistance to this agent is common, representing a major obstacle to successful treatment. The identification of novel biomarkers that are able to predict treatment response may allow therapy to be tailored to individual patients. Antibody microarrays provide a powerful new technique, enabling the global comparative analysis of many proteins simultaneously. This technology may identify a panel of proteins to discriminate between drug-resistant and drug-sensitive samples. The Panorama Cell Signaling Antibody Microarray was exploited to analyze the MDA-MB-231 breast cancer cell line and a novel derivative, which displays significant resistance to doxorubicin at clinically relevant concentrations. The microarray comprised 224 antibodies selected from a variety of pathways, including apoptotic and cell signaling pathways. A standard >/=2.0-fold cutoff value was used to determine differentially expressed proteins. A decrease in the expression of mitogen-activated protein kinase-activated monophosphotyrosine (phosphorylated extracellular signal-regulated kinase; 2.8-fold decrease), cyclin D2 (2.5-fold decrease), cytokeratin 18 (2.5-fold decrease), cyclin B1 (2.4-fold decrease), and heterogeneous nuclear ribonucleoprotein m3-m4 (2.0-fold decrease) was associated with doxorubicin resistance. Western blotting was exploited to confirm results from the antibody microarray experiment. These results suggest that antibody microarrays can be used to identify novel biomarkers and further validation may reveal mechanisms of chemotherapy resistance and identify potential therapeutic targets. [Mol Cancer Ther 2006;5(8):2115-20].
Collapse
Affiliation(s)
- Laura Smith
- Cancer Biology Proteomics Group, Postgraduate Medical Institute of the University of Hill in association with the Hull-York Medical School, University of Hull, Hull, United Kingdom
| | | | | | | | | | | |
Collapse
|
38
|
Gianchandani EP, Brautigan DL, Papin JA. Systems analyses characterize integrated functions of biochemical networks. Trends Biochem Sci 2006; 31:284-91. [PMID: 16616498 DOI: 10.1016/j.tibs.2006.03.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Revised: 02/16/2006] [Accepted: 03/24/2006] [Indexed: 12/22/2022]
Abstract
Metabolic, regulatory and signaling pathways have been characterized in detail over the past century. As the amount of genomic, proteomic and metabolic data has increased, and the mathematical and analytical capabilities of interrogating these data have advanced, the overlapping roles of pathway constituents have been described. These developments reflect the truly integrated nature of subcellular biochemical networks. Systems analyses, including the reconstruction of stoichiometric networks, provide a key set of tools for quantifying overlap among the metabolic, regulatory and signaling functions of network components. Accounting for this integration is crucial for accurately describing the function of biochemical networks.
Collapse
Affiliation(s)
- Erwin P Gianchandani
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
39
|
Sun M, Wei Y, Yao L, Xie J, Chen X, Wang H, Jiang J, Gu J. Identification of extracellular signal-regulated kinase 3 as a new interaction partner of cyclin D3. Biochem Biophys Res Commun 2005; 340:209-14. [PMID: 16360641 DOI: 10.1016/j.bbrc.2005.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 12/01/2005] [Indexed: 12/13/2022]
Abstract
Cyclin D3, like cyclin D1 and D2 isoforms, is a crucial component of the core cell cycle machinery in mammalian cells. It also exhibits its unique properties in many other physiological processes. In the present study, using yeast two-hybrid screening, we identified ERK3, an atypical mitogen-activated protein kinase (MAPK), as a cyclin D3 binding partner. GST pull-down assays showed that cyclin D3 interacts directly and specifically with ERK3 in vitro. The binding of cyclin D3 and ERK3 was further confirmed in vivo by co-immunoprecipitation assay and confocal microscopic analysis. Moreover, carboxy-terminal extension of ERK3 was responsible for its association with intact cyclin D3. These findings further expand distinct roles of cyclin D3 and suggest the potential activity of ERK3 in cell proliferation.
Collapse
Affiliation(s)
- Maoyun Sun
- State Key Laboratory of Genetic Engineering and Gene Research Center, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Wei Y, Jiang J, Sun M, Chen X, Wang H, Gu J. ATF5 increases cisplatin-induced apoptosis through up-regulation of cyclin D3 transcription in HeLa cells. Biochem Biophys Res Commun 2005; 339:591-6. [PMID: 16300731 DOI: 10.1016/j.bbrc.2005.11.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Accepted: 11/10/2005] [Indexed: 10/25/2022]
Abstract
ATF5 transcription factor plays an essential role in hematopoietic and glioma cell survival and neuronal cell differentiation. Here, we report for the first time the pro-apoptosis role of ATF5 and identify Cyclin D3 as an ATF5-targeted apoptosis-related gene. The ectopic expression of ATF5 in HeLa cells could markedly increase cisplatin-induced apoptosis and the cleavage of Caspase-3, and induce Cyclin D3 mRNA expression via cooperation with E2F1 transcription factor. Moreover, the interference of Cyclin D3 expression by transfection with Cyclin D3 RNAi could protect cells from ATF5-mediated apoptosis induced by cisplatin, indicating the contribution of Cyclin D3 in ATF5-mediated apoptosis. Taken together, these results suggest that ATF5 increases cisplatin-induced apoptosis through up-regulation of Cyclin D3 transcription, which elicits survival signals in HeLa cells.
Collapse
Affiliation(s)
- Yuanyan Wei
- State Key Laboratory of Genetic Engineering and Gene Research Center, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Apoptosis, or programmed cell death, is an essential physiological process that plays a critical role in development and tissue homeostasis. The progress of apoptosis is regulated in an orderly way by a series of signal cascades under certain circumstances. The caspase-cascade system plays vital roles in the induction, transduction and amplification of intracellular apoptotic signals. Caspases, closely associated with apoptosis, are aspartate-specific cysteine proteases and members of the interleukin-1beta-converting enzyme family. The activation and function of caspases, involved in the delicate caspase-cascade system, are regulated by various kinds of molecules, such as the inhibitor of apoptosis protein, Bcl-2 family proteins, calpain, and Ca2+. Based on the latest research, the members of the caspase family, caspase-cascade system and caspase-regulating molecules involved in apoptosis are reviewed.
Collapse
Affiliation(s)
- Ting-Jun Fan
- College of Marine Life Sciences, Division of Life Science and Technology, Ocean University of China, Qingdao 266003, China.
| | | | | | | |
Collapse
|
42
|
Zhivotovsky B, Orrenius S. Caspase-2 function in response to DNA damage. Biochem Biophys Res Commun 2005; 331:859-67. [PMID: 15865942 DOI: 10.1016/j.bbrc.2005.03.191] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Indexed: 12/12/2022]
Abstract
Caspase-2 is one of the best conserved caspases across species. This enzyme is unique among caspases in that it has features of both initiator and effector caspases. Caspase-2 appears to be necessary for the onset of apoptosis triggered by several insults, including DNA damage, administration of TNF, and different pathogens and viruses. In several experimental systems, a link has been shown between the p53 family proteins and caspase-2 activation leading to cell death. In this review, current knowledge concerning the structure of this protease and its function in cell physiology and cell death, particularly cell death triggered by DNA damage, is summarized and discussed.
Collapse
Affiliation(s)
- Boris Zhivotovsky
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden.
| | | |
Collapse
|
43
|
Mashima T, Oh-hara T, Sato S, Mochizuki M, Sugimoto Y, Yamazaki K, Hamada JI, Tada M, Moriuchi T, Ishikawa Y, Kato Y, Tomoda H, Yamori T, Tsuruo T. p53-defective tumors with a functional apoptosome-mediated pathway: a new therapeutic target. J Natl Cancer Inst 2005; 97:765-77. [PMID: 15900046 DOI: 10.1093/jnci/dji133] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Although cancer cells appear to maintain the machinery for intrinsic apoptosis, defects in the pathway develop during malignant transformation, preventing apoptosis from occurring. How to specifically induce apoptosis in cancer cells remains unclear. METHODS We determined the apoptosome activity and p53 status of normal human cells and of lung, colon, stomach, brain, and breast cancer cells by measuring cytochrome c-dependent caspase activation and by DNA sequencing, respectively, and we used COMPARE analysis to identify apoptosome-specific agonists. We compared cell death, cytochrome c release, and caspase activation in NCI-H23 (lung cancer), HCT-15 (colon cancer), and SF268 (brain cancer) cells treated with Triacsin c, an inhibitor of acyl-CoA synthetase (ACS), or with vehicle. The cells were mock, transiently, or stably transfected with genes for Triacsin c-resistant ACSL5, dominant negative caspase-9, or apoptotic protease activating factor-1 knockdown. We measured ACS activity and levels of cardiolipin, a mitochondrial phospholipid, in mock and ACSL5-transduced SF268 cells. Nude mice carrying NCI-H23 xenograft tumors (n = 10) were treated with Triacsin c or vehicle, and xenograft tumor growth was assessed. Groups were compared using two-sided Student t tests. RESULTS Of 21 p53-defective tumor cell lines analyzed, 17 had higher apoptosome activity than did normal cells. Triacsin c selectively induced apoptosome-mediated death in tumor cells (caspase activity of Triacsin c-treated versus untreated SF268 cells; means = 1020% and 100%, respectively; difference = 920%, 95% CI = 900% to 940%; P<.001). Expression of ACSL5 suppressed Triacsin c-induced cytochrome c release and subsequent cell death (cell survival of Triacsin c-treated mock- versus ACSL5-transduced SF268 cells; means = 40% and 83%, respectively; difference = 43%, 95% CI = 39% to 47%; P<.001). ACS was also essential to the maintenance of cardiolipin levels. Finally, Triacsin c suppressed growth of xenograft tumors (relative tumor volume on day 21 of Triacsin c-treated versus untreated mice; means = 4.6 and 9.6, respectively; difference = 5.0, 95% CI = 2.1 to 7.9; P = .006). CONCLUSIONS Many p53-defective tumors retain activity of the apoptosome, which is therefore a potential target for cancer chemotherapy. Inhibition of ACS may be a novel strategy to induce the death of p53-defective tumor cells.
Collapse
Affiliation(s)
- Tetsuo Mashima
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Thorburn J, Moore F, Rao A, Barclay WW, Thomas LR, Grant KW, Cramer SD, Thorburn A. Selective inactivation of a Fas-associated death domain protein (FADD)-dependent apoptosis and autophagy pathway in immortal epithelial cells. Mol Biol Cell 2005; 16:1189-99. [PMID: 15635090 PMCID: PMC551484 DOI: 10.1091/mbc.e04-10-0906] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although evasion of apoptosis is thought to be required for the development of cancer, it is unclear which cell death pathways are evaded. We previously identified a novel epithelial cell death pathway that works in normal cells but is inactivated in tumor cells, implying that it may be targeted during tumor development. The pathway can be activated by the Fas-associated death domain (FADD) of the adaptor protein but is distinct from the known mechanism of FADD-induced apoptosis through caspase-8. Here, we show that a physiological signal (tumor necrosis factor-related apoptosis-inducing ligand) can kill normal epithelial cells through the endogenous FADD protein by using the novel FADD death domain pathway, which activates both apoptosis and autophagy. We also show that selective resistance to this pathway occurs when primary epithelial cells are immortalized and that this occurs through a mechanism that is independent of known events (telomerase activity, and loss of function of p53, Rb, INK4a, and ARF) that are associated with immortalization. These data identify a novel cell death pathway that combines apoptosis and autophagy and that is selectively inactivated at the earliest stages of epithelial cancer development.
Collapse
Affiliation(s)
- Jacqueline Thorburn
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Viktorsson K, Lewensohn R, Zhivotovsky B. Apoptotic Pathways and Therapy Resistance in Human Malignancies. Adv Cancer Res 2005; 94:143-96. [PMID: 16096001 DOI: 10.1016/s0065-230x(05)94004-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Apoptosis and necrosis are two morphologically distinct forms of cell death that are important for maintaining of cellular homeostasis. Almost all agents can provoke either response when applied to cells; however, the duration of treatment and the dose of the used agents determine which type of death (apoptosis or necrosis) is initiated. The response of tumors to chemo-, radio-, and hormone therapy or to treatment with biologically active agents may depend at least in part on the propensity of these tumors to undergo cell death. Some tumors, e.g., leukemias, small cell lung cancer, and seminomas, respond quickly to first-line therapy; this fast response is thought to result from induction of apoptosis. Solid tumors, on the other hand, usually respond slowly and less effectively, with cell death characterized not only by apoptosis but also by necrosis, or mitotic catastrophe. It is likely that resistance of tumors to treatment might be associated with defects in, or dysregulation of, different steps of the apoptotic pathways. Several attempts were undertaken to use the knowledge of these defects to design new drugs, which might either activate or re-activate the apoptotic machinery of tumor cells. Here we discuss the apoptotic pathways and their role in therapy resistance of human malignancies. Although such studies are still in progress, they offer great promise for future cancer therapy. We hope that some of these agents will turn out to be valuable additions to the future therapeutic arsenal, which will most probably include a combination of conventional cytotoxic drugs and molecular target-based pro-apoptotic drugs.
Collapse
Affiliation(s)
- Kristina Viktorsson
- Unit of Medical Radiobiology, Department of Oncology/Pathology, Cancer Center Karolinska, Karolinska Institute, S-171 76 Stockholm, Sweden
| | | | | |
Collapse
|
46
|
Shen X, Yang Y, Liu W, Sun M, Jiang J, Zong H, Gu J. Identification of the p28 subunit of eukaryotic initiation factor 3(eIF3k) as a new interaction partner of cyclin D3. FEBS Lett 2004; 573:139-46. [PMID: 15327989 DOI: 10.1016/j.febslet.2004.07.071] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/06/2004] [Accepted: 07/08/2004] [Indexed: 11/27/2022]
Abstract
Cyclin D3 is found to play a crucial role not only in progression through the G1 phase as a regulatory subunit of cyclin-dependent kinase 4 (CDK 4) and CDK 6, but also in many other aspects such as cell cycle, cell differentiation, transcriptional regulation and apoptosis. In this work, we screened a human fetal liver cDNA library using human cyclin D3 as bait and identified human eukaryotic initiation factor 3 p28 protein (eIF3k) as a partner of cyclin D3. The association of cyclin D3 with eIF3k was further confirmed by in vitro binding assay, in vivo coimmunoprecipitation, and confocal microscopic analysis. We found that cyclin D3 specifically interacted with eIF3k through its C-terminal domain. Immunofluorescence experiments showed that eIF3k distributed both in nucleus and cytoplasm and colocalized with cyclin D3. In addition, the cellular translation activity in HeLa cells was upregulated by cyclin D3 overexpression and the mRNA levels are constant. These data provide a new clue to our understanding of the cellular function of cyclin D3.
Collapse
Affiliation(s)
- Xiaoyun Shen
- State key laboratory of Genetic Engineering and Gene Research Center, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | | | | | | | | | | | | |
Collapse
|
47
|
Barr RK, Hopkins RM, Watt PM, Bogoyevitch MA. Reverse two-hybrid screening identifies residues of JNK required for interaction with the kinase interaction motif of JNK-interacting protein-1. J Biol Chem 2004; 279:43178-89. [PMID: 15271995 DOI: 10.1074/jbc.m405900200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The development of specific inhibitors for the c-Jun N-terminal kinase (JNK) family of mitogen-activated protein kinases (MAPKs) has been a recent research focus because of the association of JNK with cell death in conditions such as stroke and neurodegeneration. We have demonstrated previously the presence of critical inhibitory residues within an 11-mer peptide (TI-JIP) based on the sequence of JNK-interacting protein-1 (JIP-1). However, the corresponding region of JNK bound by this JIP-1-based peptide was unknown. To identify this region, we used a novel reverse two-hybrid approach with TI-JIP as bait. We screened a library of JNK1 mutants that had been generated by random PCR mutagenesis and found three mutants of JNK1 that failed to interact with TI-JIP. The mutations in JNK1 were L131R, R309W, and Y320H. Of these mutated residues, Leu-131 and Tyr-320 were located on a common face of the JNK protein close to other residues implicated previously in the interactions of MAPKs with substrates, phosphatases, and scaffolds. To test whether these JNK1 mutants were thus affected in their regulation, we evaluated their activation in mammalian cells in response to hyperosmolarity or cotransfection with a constitutively active upstream kinase or their direct phosphorylation by either MAPK kinase (MKK)4 or MKK7. In each situation, all three JNK mutants were not activated or phosphorylated to the same level as wild-type JNK. Therefore, the results of our unbiased reverse two-hybrid screening approach have identified residues of JNK responsible for binding JIP-1-based peptides as well as MKK4 or MKK7.
Collapse
Affiliation(s)
- Renae K Barr
- Cell Signalling Laboratory, Biochemistry and Molecular Biology, University of Western Australia, Crawley, Perth, Western Australia
| | | | | | | |
Collapse
|
48
|
Abstract
Unicellular organisms respond to the presence of DNA lesions by activating cell cycle checkpoint and repair mechanisms, while multicellular animals have acquired the further option of eliminating damaged cells by triggering apoptosis. Defects in DNA damage-induced apoptosis contribute to tumorigenesis and to the resistance of cancer cells to a variety of therapeutic agents. The intranuclear mechanisms that signal apoptosis after DNA damage overlap with those that initiate cell cycle arrest and DNA repair, and the early events in these pathways are highly conserved. In addition, multiple independent routes have recently been traced by which nuclear DNA damage can be signalled to the mitochondria, tipping the balance in favour of cell death rather than repair and survival. Here, we review current knowledge of nuclear DNA damage signalling, giving particular attention to interactions between these nuclear events and apoptotic processes in other intracellular compartments.
Collapse
Affiliation(s)
- Chris J Norbury
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | | |
Collapse
|
49
|
Cecconi F, Roth KA, Dolgov O, Munarriz E, Anokhin K, Gruss P, Salminen M. Apaf1-dependent programmed cell death is required for inner ear morphogenesis and growth. Development 2004; 131:2125-35. [PMID: 15105372 DOI: 10.1242/dev.01082] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During inner ear development programmed cell death occurs in specific areas of the otic epithelium but the significance of it and the molecules involved have remained unclear. We undertook an analysis of mouse mutants in which genes encoding apoptosis-associated molecules have been inactivated. Disruption of the Apaf1 gene led to a dramatic decrease in apoptosis in the inner ear epithelium, severe morphogenetic defects and a significant size reduction of the membranous labyrinth, demonstrating that an Apaf1-dependent apoptotic pathway is necessary for normal inner ear development. This pathway most probably operates through the apoptosome complex because caspase 9 mutant mice suffered similar defects. Inactivation of the Bcl2-like (Bcl2l) gene led to an overall increase in the number of cells undergoing apoptosis but did not cause any major morphogenetic defects. In contrast, decreased apoptosis was observed in specific locations that suffered from developmental deficits, indicating that proapoptotic isoform(s) produced from Bcl2l might have roles in inner ear development. In Apaf1-/-/Bcl2l-/-double mutant embryos, no cell death could be detected in the otic epithelium,demonstrating that the cell death regulated by the anti-apoptotic Bcl2l isoform, Bcl-XL in the otic epithelium is Apaf1-dependent. Furthermore, the otic vesicle failed to close completely in all double mutant embryos analyzed. These results indicate important roles for both Apaf1 and Bcl2l in inner ear development.
Collapse
Affiliation(s)
- Francesco Cecconi
- Dulbecco Telethon Institute, Department of Biology, University of Rome Tor Vergata, via della Ricerca Scientifica, 00133 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Castedo M, Perfettini JL, Roumier T, Valent A, Raslova H, Yakushijin K, Horne D, Feunteun J, Lenoir G, Medema R, Vainchenker W, Kroemer G. Mitotic catastrophe constitutes a special case of apoptosis whose suppression entails aneuploidy. Oncogene 2004; 23:4362-70. [PMID: 15048075 DOI: 10.1038/sj.onc.1207572] [Citation(s) in RCA: 236] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A conflict in cell cycle progression or DNA damage can lead to mitotic catastrophe when the DNA structure checkpoints are inactivated, for instance when the checkpoint kinase Chk2 is inhibited. Here we show that in such conditions, cells die during the metaphase of the cell cycle, as a result of caspase activation and subsequent mitochondrial damage. Molecular ordering of these phenomena reveals that mitotic catastrophe occurs in a p53-independent manner and involves a primary activation of caspase-2, upstream of cytochrome c release, followed by caspase-3 activation and chromatin condensation. Suppression of caspase-2 by RNA interference or pseudosubstrate inhibitors as well as blockade of the mitochondrial membrane permeabilization prevent the mitotic catastrophe and allow cells to further proceed the cell cycle beyond the metaphase, leading to asymmetric cell division. Heterokarya generated by the fusion of nonsynchronized cells can be driven to divide into three or more daughter cells when Chk2 and caspases are simultaneously inhibited. Such multipolar divisions, resulting from suppressed mitotic catastrophe, lead to the asymmetric distribution of cytoplasm (anisocytosis), DNA (anisokaryosis) and chromosomes (aneuploidy). Similarly, in a model of DNA damage-induced mitotic catastrophe, suppression of apoptosis leads to the generation of aneuploid cells. Our findings delineate a molecular pathway through which DNA damage, failure to arrest the cell cycle and inhibition of apoptosis can favor the occurrence of cytogenetic abnormalities that are likely to participate in oncogenesis.
Collapse
Affiliation(s)
- Maria Castedo
- CNRS-UMR 8125, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|