1
|
Kulbay M, Bernier-Parker N, Bernier J. The role of the DFF40/CAD endonuclease in genomic stability. Apoptosis 2021; 26:9-23. [PMID: 33387146 DOI: 10.1007/s10495-020-01649-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/18/2022]
Abstract
Maintenance of genomic stability in cells is primordial for cellular integrity and protection against tumor progression. Many factors such as ultraviolet light, oxidative stress, exposure to chemical reagents, particularly mutagens and radiation, can alter the integrity of the genome. Thus, human cells are equipped with many mechanisms that prevent these irreversible lesions in the genome, as DNA repair pathways, cell cycle checkpoints, and telomeric function. These mechanisms activate cellular apoptosis to maintain DNA stability. Emerging studies have proposed a new protein in the maintenance of genomic stability: the DNA fragmentation factor (DFF). The DFF40 is an endonuclease responsible of the oligonucleosomal fragmentation of the DNA during apoptosis. The lack of DFF in renal carcinoma cells induces apoptosis without oligonucleosomal fragmentation, which poses a threat to genetic information transfer between cancerous and healthy cells. In this review, we expose the link between the DFF and genomic instability as the source of disease development.
Collapse
Affiliation(s)
- Merve Kulbay
- INRS - Centre Armand-Frappier-Santé-Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada
- Department of Medicine, Université de Montréal, 2900 Blvd. Edouard Montpetit, Montreal, QC, Canada
| | - Nathan Bernier-Parker
- Toronto Animal Health Partners Emergency and Specialty Hospital, 1 Scarsdale Road, North York, ON, M3B 2R2, Canada
| | - Jacques Bernier
- INRS - Centre Armand-Frappier-Santé-Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada.
| |
Collapse
|
2
|
Yuan Y, Yang Z, Zou Q. MiRNA-145 Induces Apoptosis in a Gallbladder Carcinoma Cell Line by Targeting DFF45. Open Life Sci 2018; 13:227-235. [PMID: 33817087 PMCID: PMC7874708 DOI: 10.1515/biol-2018-0027] [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: 12/03/2017] [Accepted: 03/21/2018] [Indexed: 11/15/2022] Open
Abstract
Bakcground We measured expression of miRNA-145 in gallbladder carcinoma and its influence on propagation, invasion, and apoptosis of gallbladder carcinoma cells in vitro. Methods miRNA-145 expression was compared between normal gallbladder epithelial cells and GBS-SD (gallbladder series) cells using miRNA chip technology. Propagation, apoptosis, and invasion properties of each cell group were tested using MTT, a clone-formation assay, flow cytometry, Western blot, and Transwell assays. Results Expression of miRNA-145 was observed to be down-regulated and GBC-SD cell clones transiently transfected with hsa-miRNA-145 were substantially reduced compared with controls (p<0.01). We observed that GBC-SD cells transfected with hsa-miRNA-145 and double-positive (Annexin V and PI) for apoptosis were more numerous than controls. Moreover, GBC-SD cells over-expressing miRNA-145 had significantly greater expression of apoptosis-related protein, caspase-3. A Transwell assay confirmed that GBC-SD cells over-expressing miRNA-145 that migrated to the lower chamber were fewer compared with controls. Post-transcriptional regulation of gene expression was measured using dualluciferase reporter assays and data show that miRNA-145 facilitates the inhibition of GBC-SD cell growth and invasion while inducing apoptosis by targeting DFF45. Conclusion Thus, we speculate that miRNA-145 facilitates inhibition of GBC-SD cell growth and invasion while inducing apoptosis by targeting DFF45; however, miRNA-145 does not directly affect the GBC-SD cell cycle.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhulin Yang
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qiong Zou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
3
|
Ruirui K, Ray P, Yang M, Wen P, Zhu L, Liu J, Fushimi K, Kar A, Liu Y, He R, Kuo D, Wu JY. Alternative Pre-mRNA Splicing, Cell Death, and Cancer. Cancer Treat Res 2013; 158:181-212. [PMID: 24222359 DOI: 10.1007/978-3-642-31659-3_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Alternative splicing is one of the most powerful mechanisms for generating functionally distinct products from a single genetic loci and for fine-tuning gene activities at the post-transcriptional level. Alternative splicing plays important roles in regulating genes critical for cell death. These cell death genes encode death ligands, cell surface death receptors, intracellular death regulators, signal transduction molecules, and death executor enzymes such as caspases and nucleases. Alternative splicing of these genes often leads to the formation of functionally different products, some of which have antagonistic effects that are either cell death-promoting or cell death-preventing. Differential alternative splicing can affect expression, subcellular distribution, and functional activities of the gene products. Molecular defects in splicing regulation of cell death genes have been associated with cancer development and resistance to treatment. Studies using molecular, biochemical, and systems-based approaches have begun to reveal mechanisms underlying the regulation of alternative splicing of cell death genes. Systematic studies have begun to uncover the multi-level interconnected networks that regulate alternative splicing. A global picture of the complex mechanisms that regulate cell death genes at the pre-mRNA splicing level has thus begun to emerge.
Collapse
Affiliation(s)
- Kong Ruirui
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Iglesias-Guimarais V, Gil-Guiñon E, Gabernet G, García-Belinchón M, Sánchez-Osuna M, Casanelles E, Comella JX, Yuste VJ. Apoptotic DNA degradation into oligonucleosomal fragments, but not apoptotic nuclear morphology, relies on a cytosolic pool of DFF40/CAD endonuclease. J Biol Chem 2012; 287:7766-79. [PMID: 22253444 DOI: 10.1074/jbc.m111.290718] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Apoptotic cell death is characterized by nuclear fragmentation and oligonucleosomal DNA degradation, mediated by the caspase-dependent specific activation of DFF40/CAD endonuclease. Here, we describe how, upon apoptotic stimuli, SK-N-AS human neuroblastoma-derived cells show apoptotic nuclear morphology without displaying concomitant internucleosomal DNA fragmentation. Cytotoxicity afforded after staurosporine treatment is comparable with that obtained in SH-SY5Y cells, which exhibit a complete apoptotic phenotype. SK-N-AS cell death is a caspase-dependent process that can be impaired by the pan-caspase inhibitor q-VD-OPh. The endogenous inhibitor of DFF40/CAD, ICAD, is correctly processed, and dff40/cad cDNA sequence does not reveal mutations altering its amino acid composition. Biochemical approaches show that both SH-SY5Y and SK-N-AS resting cells express comparable levels of DFF40/CAD. However, the endonuclease is poorly expressed in the cytosolic fraction of healthy SK-N-AS cells. Despite this differential subcellular distribution of DFF40/CAD, we find no differences in the subcellular localization of both pro-caspase-3 and ICAD between the analyzed cell lines. After staurosporine treatment, the preferential processing of ICAD in the cytosolic fraction allows the translocation of DFF40/CAD from this fraction to a chromatin-enriched one. Therefore, the low levels of cytosolic DFF40/CAD detected in SK-N-AS cells determine the absence of DNA laddering after staurosporine treatment. In these cells DFF40/CAD cytosolic levels can be restored by the overexpression of their own endonuclease, which is sufficient to make them proficient at degrading their chromatin into oligonucleosome-size fragments after staurosporine treatment. Altogether, the cytosolic levels of DFF40/CAD are determinants in achieving a complete apoptotic phenotype, including oligonucleosomal DNA degradation.
Collapse
Affiliation(s)
- Victoria Iglesias-Guimarais
- Cell Death, Senescence, and Survival Group, Departament de Bioquimica i Biologia Molecular and Institut de Neurociencies, Facultat de Medicina, Universitat Autonoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Abstract
DNA fragmentation is a hallmark of apoptosis that is induced by apoptotic stimuli in various cell types. Apoptotic signal pathways, which eventually cause DNA fragmentation, are largely mediated by the family of cysteinyl aspartate-specific protease caspases. Caspases mediate apoptotic signal transduction by cleavage of apoptosis-implicated proteins and the caspases themselves. In the process of caspase activation, reversible protein phosphorylation plays an important role. The activation of various proteins is regulated by phosphorylation and dephosphorylation, both upstream and downstream of caspase activation. Many kinases/phosphatases are involved in the control of cell survival and death, including the mitogen-activated protein kinase signal transduction pathways. Reversible protein phosphorylation is involved in the widespread regulation of cellular signal transduction and apoptotic processes. Therefore, phosphatase/kinase inhibitors are commonly used as apoptosis inducers/inhibitors. Whether protein phosphorylation induces apoptosis depends on many factors, such as the type of phosphorylated protein, the degree of activation and the influence of other proteins. Phosphorylation signaling pathways are intricately interrelated; it was previously shown that either induction or inhibition of phosphorylation causes cell death. Determination of the relationship between protein and phosphorylation helps to reveal how apoptosis is regulated. Here we discuss DNA fragmentation and protein phosphorylation, focusing on caspase and serine/threonine protein phosphatase activation.
Collapse
Affiliation(s)
- Ikuko Kitazumi
- Bio Process Research and Development Laboratories, Kyowa Hakko Kirin Co. Ltd, Takasaki, Gunma, Japan
| | | |
Collapse
|
6
|
Neimanis S, Albig W, Doenecke D, Kahle J. Sequence elements in both subunits of the DNA fragmentation factor are essential for its nuclear transport. J Biol Chem 2007; 282:35821-30. [PMID: 17938174 DOI: 10.1074/jbc.m703110200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DNA cleavage is a biochemical hallmark of apoptosis. In humans, apoptotic DNA cleavage is executed by DNA fragmentation factor (DFF) 40. In proliferating cells DFF40 is expressed in the presence of its chaperone and inhibitor DFF45, which results in the formation of the DFF complex. Here, we present a systematic analysis of the nuclear import of the DFF complex. Our in vitro experiments demonstrate that the importin alpha/beta-heterodimer mediates the translocation of the DFF complex from the cytoplasm to the nucleus. Both DFF subunits interact directly with the importin alpha/beta-heterodimer. However, importin alpha/beta binds more tightly to the DFF complex compared with the individual subunits. Additionally, the isolated C-terminal regions of both DFF subunits together bind importin alpha/beta more strongly than the individual C termini. Our results from in vivo studies reveal that the C-terminal regions of both DFF subunits harbor nuclear localization signals. Furthermore, nuclear import of the DFF complex requires the C-terminal regions of both subunits. In more detail, one basic cluster in the C-terminal region of each subunit, DFF40 (RLKRK) and DFF45 (KRAR), is essential for nuclear accumulation of the DFF complex. Based on these findings two alternative models for the interaction of importin alpha/beta with the DFF complex are presented.
Collapse
Affiliation(s)
- Sonja Neimanis
- Institut für Biochemie und Molekulare Zellbiologie, Abteilung Molekularbiologie, Universität Göttingen, Humboldtallee 23, Göttingen 37073, Germany
| | | | | | | |
Collapse
|
7
|
Abstract
We have created new genomics tools for chromatin research by genetically engineering the human and mouse major apoptotic nucleases that are responsible for internucleosomal DNA cleavage, DNA fragmentation factor (DFF). Normally, in its inactive form, DFF is a heterodimer composed of a 45-kDa chaperone inhibitor subunit (DFF45 or ICAD), and a 40-kDa latent endonuclease subunit (DFF40 or CAD). Upon caspase-3 cleavage of DFF45, DFF40 forms active endonuclease homo-oligomers. Although Saccharomyces cerevisiae lacks DFF, expression of caspase-3 is lethal in this organism, but expression of the highly sequence-specific tobacco etch virus protease (TEVP) is harmless. Therefore, we inserted TEVP cleavage sites immediately downstream of the two caspase-3 cleavage sites within DFF45, generating a novel form of DFF (DFF-T) whose nuclease activity proved to be exclusively under the control of TEVP. We demonstrate that co-expression of TEVP and DFF-T under galactose control results in nucleosomal DNA laddering and cell death in S. cerevisiae. We also created synthetic DFF genes with optimized codons for high-level expression in Eschericia coli or S. cerevisiae. We further demonstrate the excellence of the synthetic gene products for in vitro mapping of the nucleosome positions and hypersensitive sites in specific genes such as the yeast PHO5.
Collapse
Affiliation(s)
- Fei Xiao
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA and Faculty of Biotechnology, University of Rzeszow, 36-100 Werynia, Poland
| | - Piotr Widlak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA and Faculty of Biotechnology, University of Rzeszow, 36-100 Werynia, Poland
| | - William T. Garrard
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA and Faculty of Biotechnology, University of Rzeszow, 36-100 Werynia, Poland
- *To whom correspondence should be addressed. +1 214 648 1924+1 214 648 1915
| |
Collapse
|
8
|
Widlak P, Garrard WT. Unique features of the apoptotic endonuclease DFF40/CAD relative to micrococcal nuclease as a structural probe for chromatin. Biochem Cell Biol 2007; 84:405-10. [PMID: 16936813 DOI: 10.1139/o06-063] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The gold standard for studies of nucleosomal chromatin structure for the past 30 years has been the enzyme micrococcal nuclease (MNase). During the course of our studies on the elucidation of the mechanism of action of the apoptotic nuclease DNA fragmentation factor-40 / caspase-activated deoxyribonuclease (DFF40/CAD) on naked DNA and chromatin substrates, it became clear that this enzyme is superior in certain respects to MNase for studying several aspects of chromatin structure. Here we review our published results supporting this statement. Relative to MNase, we have found that DFF40/CAD has the following properties: (i) it does not cut within nucleosomes to generate subnucleosomal DNA fragments; (ii) it is more specific for the linker regions between nucleosomes; (iii) it lacks exonuclease activity; (iv) it is specific for double-stranded DNA and makes exclusively double-stranded breaks; and (v) it attacks histone-H1-containing chromatin more efficiently. Taken together, these facts explain why DFF40/CAD generates sharper oligonucleosomal DNA ladders compared with those generated by MNase. We therefore recommend the following uses for DFF40/CAD for chromatin research: nucleosome isolation, chromatin-remodeling assays, repeat length measurements, and nucleosome-positioning assays along specific sequences. Other uses include footprinting assays of transcription factor positions, shearing chromatin for immunopreciptitation experiments (ChIP), and shearing DNA for recombinant DNA library preparation or for shotgun cloning for sequencing.
Collapse
Affiliation(s)
- Piotr Widlak
- Department of Molecular Biology, University of TX Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9148, USA
| | | |
Collapse
|
9
|
Joselin AP, Schulze-Osthoff K, Schwerk C. Loss of Acinus inhibits oligonucleosomal DNA fragmentation but not chromatin condensation during apoptosis. J Biol Chem 2006; 281:12475-84. [PMID: 16537548 DOI: 10.1074/jbc.m509859200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chromatin condensation and oligonucleosomal DNA fragmentation are the nuclear hallmarks of apoptosis. A proteolytic fragment of the apoptotic chromatin condensation inducer in the nucleus (Acinus), which is generated by caspase cleavage, has been implicated in mediating apoptotic chromatin condensation prior to DNA fragmentation. Acinus is also involved in mRNA splicing and a component of the apoptosis and splicing-associated protein (ASAP) complex. To study the role of Acinus for apoptotic nuclear alterations, we generated stable cell lines in which Acinus isoforms were knocked down by inducible and reversible RNA interference. We show that Acinus is not required for nuclear localization and interaction of the other ASAP subunits SAP18 and RNPS1; however, knockdown of Acinus leads to a reduction in cell growth. Most strikingly, down-regulation of Acinus did not inhibit apoptotic chromatin condensation either in intact cells or in a cell-free system. In contrast, although apoptosis proceeds rapidly, analysis of nuclear DNA from apoptotic Acinus knockdown cells shows inhibition of oligonucleosomal DNA fragmentation. Our results therefore suggest that Acinus is not involved in DNA condensation but rather point to a contribution of Acinus in internucleosomal DNA cleavage during programmed cell death.
Collapse
Affiliation(s)
- Alvin P Joselin
- Institute of Molecular Medicine, the University of Düsseldorf, 40225 Düsseldorf, Germany
| | | | | |
Collapse
|
10
|
Samejima K, Earnshaw WC. Trashing the genome: the role of nucleases during apoptosis. Nat Rev Mol Cell Biol 2005; 6:677-88. [PMID: 16103871 DOI: 10.1038/nrm1715] [Citation(s) in RCA: 226] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Two classes of nucleases degrade the cellular DNA during apoptosis. Cell-autonomous nucleases cleave DNA within the dying cell. They are not essential for apoptotic cell death or the life of the organism, but they might affect the efficiency of the process. By contrast, waste-management nucleases are essential for the life of the organism. In post-engulfment DNA degradation, the DNA of apoptotic cells is destroyed in lysosomes of the cells that have phagocytosed the corpses. Waste-management nucleases also destroy DNA that is released into the extracellular compartment. Here, we describe the complex group of nucleases that are involved in DNA destruction during apoptotic cell death.
Collapse
Affiliation(s)
- Kumiko Samejima
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Swann Building, King's Buildings, Mayfield Road, Edinburgh EH9 3JR, UK
| | | |
Collapse
|
11
|
Li X, Wang J, Manley JL. Loss of splicing factor ASF/SF2 induces G2 cell cycle arrest and apoptosis, but inhibits internucleosomal DNA fragmentation. Genes Dev 2005; 19:2705-14. [PMID: 16260492 PMCID: PMC1283963 DOI: 10.1101/gad.1359305] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ASF/SF2 is an SR protein splicing factor that participates in constitutive and alternative pre-mRNA splicing and is essential for cell viability. Using a genetically modified chicken B-cell line, DT40-ASF, we now show that ASF/SF2 inactivation results in a G2-phase cell cycle arrest and subsequent programmed cell death. However, although several hallmarks of apoptosis are apparent, internucleosomal DNA fragmentation was not detected. Furthermore, inactivation of ASF/SF2 also blocks DNA fragmentation normally induced by a variety of apoptotic stimuli. Notably, mRNA encoding the inhibitor of caspase-activated DNase-L (ICAD-L), which acts as an inhibitor as well as a chaperone of caspase-activated DNase (CAD), decreased in abundance, whereas the level of mRNA encoding ICAD-S, which has only inhibitory activity, increased upon ASF/SF2 depletion. Strikingly, expression of appropriate levels of exogenous human ICAD-L restored apoptotic DNA laddering in ASF/SF2-depleted cells. These results not only indicate that loss of an SR protein splicing factor can induce cell cycle arrest and apoptosis, but also illustrate the important role of ICAD and its regulation by alternative splicing in the process of apoptotic DNA fragmentation.
Collapse
Affiliation(s)
- Xialu Li
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | | | | |
Collapse
|
12
|
Widlak P, Garrard WT. Discovery, regulation, and action of the major apoptotic nucleases DFF40/CAD and endonuclease G. J Cell Biochem 2005; 94:1078-87. [PMID: 15723341 DOI: 10.1002/jcb.20409] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Toward the end of the 20th and beginning of the 21st centuries, clever in vitro biochemical complementation experiments and genetic screens from the laboratories of Xiaodong Wang, Shigekazu Nagata, and Ding Xue led to the discovery of two major apoptotic nucleases, termed DNA fragmentation factor (DFF) or caspase-activated DNase (CAD) and endonuclease G (Endo G). Both endonucleases attack chromatin to yield 3'-hydroxyl groups and 5'-phosphate residues, first at the level of 50-300 kb cleavage products and next at the level of internucleosomal DNA fragmentation, but these nucleases possess completely different cellular locations in normal cells and are regulated in vastly different ways. In non-apoptotic cells, DFF exists in the nucleus as a heterodimer, composed of a 45 kD chaperone and inhibitor subunit (DFF45) [also called inhibitor of CAD (ICAD-L)] and a 40 kD latent nuclease subunit (DFF40/CAD). Apoptotic activation of caspase-3 or -7 results in the cleavage of DFF45/ICAD and release of active DFF40/CAD nuclease. DFF40's nuclease activity is further activated by specific chromosomal proteins, such as histone H1, HMGB1/2, and topoisomerase II. DFF is regulated by multiple pre- and post-activation fail-safe steps, which include the requirements for DFF45/ICAD, Hsp70, and Hsp40 proteins to mediate appropriate folding during translation to generate a potentially activatable nuclease, and the synthesis in stoichiometric excess of the inhibitors (DFF45/35; ICAD-S/L). By contrast, Endo G resides in the mitochondrial intermembrane space in normal cells, and is released into the nucleus upon apoptotic disruption of mitochondrial membrane permeability in association with co-activators such as apoptosis-inducing factor (AIF). Understanding further regulatory check-points involved in safeguarding non-apoptotic cells against accidental activation of these nucleases remain as future challenges, as well as designing ways to selectively activate these nucleases in tumor cells.
Collapse
Affiliation(s)
- Piotr Widlak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | | |
Collapse
|
13
|
Schwerk C, Schulze-Osthoff K. Regulation of Apoptosis by Alternative Pre-mRNA Splicing. Mol Cell 2005; 19:1-13. [PMID: 15989960 DOI: 10.1016/j.molcel.2005.05.026] [Citation(s) in RCA: 423] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Revised: 02/22/2005] [Accepted: 05/05/2005] [Indexed: 12/15/2022]
Abstract
Apoptosis, a phenomenon that allows the regulated destruction and disposal of damaged or unwanted cells, is common to many cellular processes in multicellular organisms. In humans more than 200 proteins are involved in apoptosis, many of which are dysregulated or defective in human diseases including cancer. A large number of apoptotic factors are regulated via alternative splicing, a process that allows for the production of discrete protein isoforms with often distinct functions from a common mRNA precursor. The abundance of apoptosis genes that are alternatively spliced and the often antagonistic roles of the generated protein isoforms strongly imply that alternative splicing is a crucial mechanism for regulating life and death decisions. Importantly, modulation of isoform production of cell death proteins via pharmaceutical manipulation of alternative splicing may open up new therapeutic avenues for the treatment of disease.
Collapse
Affiliation(s)
- Christian Schwerk
- Institute of Molecular Medicine, University of Düsseldorf, Building 23.12, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | | |
Collapse
|
14
|
Pryde F, Khalili S, Robertson K, Selfridge J, Ritchie AM, Melton DW, Jullien D, Adachi Y. 53BP1 exchanges slowly at the sites of DNA damage and appears to require RNA for its association with chromatin. J Cell Sci 2005; 118:2043-55. [PMID: 15840649 DOI: 10.1242/jcs.02336] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
53BP1 protein is re-localized to the sites of DNA damage after ionizing radiation (IR) and is involved in DNA-damage-checkpoint signal transduction. We examined the dynamics of GFP-53BP1 in living cells. The protein starts to accumulate at the sites of DNA damage 2-3 minutes after damage induction. Fluorescence recovery after photobleaching experiments showed that GFP-53BP1 is highly mobile in non-irradiated cells. Upon binding to the IR-induced nuclear foci, the mobility of 53BP1 reduces greatly. The minimum (M) domain of 53BP1 essential for targeting to IR induced foci consists of residues 1220-1703. GFP-M protein forms foci in mouse embryonic fibroblast cells lacking functional endogenous 53BP1. The M domain contains a tandem repeat of Tudor motifs and an arginine- and glycine-rich domain (RG stretch), which are often found in proteins involved in RNA metabolism, the former being essential for targeting. RNase A treatment dissociates 53BP1 from IR-induced foci. In HeLa cells, dissociation of the M domain without the RG stretch by RNase A treatment can be restored by re-addition of nuclear RNA in the early stages of post-irradiation. 53BP1 immunoprecipitates contain some RNA molecules. Our results suggest a possible involvement of RNA in the binding of 53BP1 to chromatin damaged by IR.
Collapse
Affiliation(s)
- Fiona Pryde
- The Wellcome Trust Centre for Cell Biology, The Institute of Cell and Molecular Biology, The University of Edinburgh, The King's Buildings, Edinburgh, EH9 3JR, UK
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Korn C, Scholz SR, Gimadutdinow O, Lurz R, Pingoud A, Meiss G. Interaction of DNA Fragmentation Factor (DFF) with DNA Reveals an Unprecedented Mechanism for Nuclease Inhibition and Suggests That DFF Can Be Activated in a DNA-bound State. J Biol Chem 2005; 280:6005-15. [PMID: 15572351 DOI: 10.1074/jbc.m413035200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
DNA fragmentation factor (DFF) is a complex of the DNase DFF40 (CAD) and its chaperone/inhibitor DFF45 (ICAD-L) that can be activated during apoptosis to induce DNA fragmentation. Here, we demonstrate that DFF directly binds to DNA in vitro without promoting DNA cleavage. DNA binding by DFF is mediated by the nuclease subunit, which can also form stable DNA complexes after release from DFF. Recombinant and reconstituted DFF is catalytically inactive yet proficient in DNA binding, demonstrating that the nuclease subunit in DFF is inhibited in DNA cleavage but not in DNA binding, revealing an unprecedented mode of nuclease inhibition. Activation of DFF in the presence of naked DNA or isolated nuclei stimulates DNA degradation by released DFF40 (CAD). In transfected HeLa cells transiently expressed DFF associates with chromatin, suggesting that DFF could be activated during apoptosis in a DNA-bound state.
Collapse
Affiliation(s)
- Christian Korn
- Institute of Biochemistry, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Williams CL. The polybasic region of Ras and Rho family small GTPases: a regulator of protein interactions and membrane association and a site of nuclear localization signal sequences. Cell Signal 2003; 15:1071-80. [PMID: 14575862 DOI: 10.1016/s0898-6568(03)00098-6] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Many small GTPases in the Ras and Rho families have a C-terminal polybasic region (PBR) comprised of multiple lysines or arginines. The PBR controls diverse functions of these small GTPases, including their ability to associate with membranes, interact with specific proteins, and localize in subcellular compartments. Different signaling pathways mediated by Ras and Rho family members may converge when the small GTPases are directed by their PBRs to shared binding sites in specific proteins or at cell membranes. The PBR promotes the interactions of small GTPases with SmgGDS, which is a nucleocytoplasmic shuttling protein that stimulates guanine nucleotide exchange by small GTPases. The PBR of Rac1 was recently found to have a functional nuclear localization signal (NLS) sequence, which enhances the nuclear accumulation of protein complexes containing SmgGDS and Rac1. Sequence analysis demonstrates that canonical NLS sequences (K-K/R-x-K/R) are present in the PBRs of additional Ras and Rho family members, and are evolutionarily conserved across several phyla. These findings suggest that the PBR regulates the nucleocytoplasmic shuttling of some Ras and Rho family members when they are in protein complexes that are too large to diffuse through nuclear pores. These diverse functions of the PBR indicate its critical role in signaling by Ras and Rho family GTPases.
Collapse
Affiliation(s)
- Carol L Williams
- Molecular Pharmacology Laboratory, Guthrie Research Institute, One Guthrie Square, Sayre, PA 18840, USA.
| |
Collapse
|
17
|
Widlak P, Lanuszewska J, Cary RB, Garrard WT. Subunit structures and stoichiometries of human DNA fragmentation factor proteins before and after induction of apoptosis. J Biol Chem 2003; 278:26915-22. [PMID: 12748178 DOI: 10.1074/jbc.m303807200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DNA fragmentation factor (DFF) is one of the major endonucleases responsible for internucleosomal DNA cleavage during apoptosis. Understanding the regulatory checkpoints involved in safeguarding non-apoptotic cells against accidental activation of this nuclease is as important as elucidating its activation mechanisms during apoptosis. Here we address these issues by determining DFF native subunit structures and stoichiometries in human cells before and after induction of apoptosis using the technique of native pore-exclusion limit electrophoresis in combination with Western analyses. For comparison, we employed similar techniques with recombinant proteins in conjunction with atomic force microscopy. Before induction of apoptosis, the expression of DFF subunits varied widely among the cell types studied, and the chaperone/inhibitor subunits DFF45 and DFF35 unexpectedly existed primarily as monomers in vast excess of the latent nuclease subunit, DFF40, which was stoichiometrically associated with DFF45 to form heterodimers. DFF35 was exclusively cytoplasmic as a monomer. Nuclease activation upon caspase-3 cleavage of DFF45/DFF35 was accompanied by DFF40 homo-oligomer formation, with a tetramer being the smallest unit. Interestingly, intact DFF45 can inhibit nuclease activity by associating with these homo-oligomers without mediating their disassembly. We conclude that DFF nuclease is regulated by multiple pre- and post-activation fail-safe steps.
Collapse
Affiliation(s)
- Piotr Widlak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
18
|
Wu JY, Tang H, Havlioglu N. Alternative pre-mRNA splicing and regulation of programmed cell death. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2003; 31:153-85. [PMID: 12494766 DOI: 10.1007/978-3-662-09728-1_6] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- J Y Wu
- Department of Pediatrics and Department of Molecular Biology and Pharmacology, MPRB Rm3107, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | |
Collapse
|
19
|
Nagata S, Nagase H, Kawane K, Mukae N, Fukuyama H. Degradation of chromosomal DNA during apoptosis. Cell Death Differ 2003; 10:108-16. [PMID: 12655299 DOI: 10.1038/sj.cdd.4401161] [Citation(s) in RCA: 330] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Apoptosis is often accompanied by degradation of chromosomal DNA. CAD, caspase-activated DNase, was identified in 1998 as a DNase that is responsible for this process. In the last several years, mice deficient in the CAD system have been generated. Studies with these mice indicated that apoptotic DNA degradation occurs in two different systems. In one, the DNA fragmentation is carried out by CAD in the dying cells and in the other, by lysosomal DNase II after the dying cells are phagocytosed. Several other endonucleases have also been suggested as candidate effectors for the apoptotic degradation of chromosomal DNA. In this review, we will discuss the mechanism and role of DNA degradation during apoptosis.
Collapse
Affiliation(s)
- S Nagata
- Integrated Biology Laboratories, Graduate School of Frontier Science, Japan Science and Technology Corperation, Suita, Osaka.
| | | | | | | | | |
Collapse
|
20
|
Fukushima K, Kikuchi J, Koshiba S, Kigawa T, Kuroda Y, Yokoyama S. Solution structure of the DFF-C domain of DFF45/ICAD. A structural basis for the regulation of apoptotic DNA fragmentation. J Mol Biol 2002; 321:317-27. [PMID: 12144788 DOI: 10.1016/s0022-2836(02)00588-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
DFF45/ICAD has dual functions in the final stage of apoptosis, by acting as both a folding chaperone and a DNase inhibitor of DFF40/CAD. Here, we present the solution structure of the C-terminal domain of DFF45, which is essential for its chaperone-like activity. The structure of this domain (DFF-C) consists of four alpha helices, which are folded in a novel helix-packing arrangement. The 3D structure reveals a large cluster of negatively charged residues on the molecular surface of DFF-C. This observation suggests that charge complementation plays an important role in the interaction of DFF-C with the positively charged catalytic domain of DFF40, and thus for the chaperone activity of DFF45. The structure of DFF-C also provides a rationale for the loss of the chaperone activity in DFF35, a short isoform of DFF45. Indeed, in DFF35, the amino acid sequence is truncated in the middle of the second alpha helix constituting the structure of DFF-C, and thus both the hydrophobic core and the cluster of negative charges are disrupted.
Collapse
Affiliation(s)
- Kay Fukushima
- Protein Research Group, Genomic Sciences Center, RIKEN Yokohama Institute, 1-7-22, Suehiro-cho, Tsurumi, Yokohama, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Scholz SR, Korn C, Gimadutdinow O, Knoblauch M, Pingoud A, Meiss G. The effect of ICAD-S on the formation and intracellular distribution of a nucleolytically active caspase-activated DNase. Nucleic Acids Res 2002; 30:3045-51. [PMID: 12136086 PMCID: PMC135751 DOI: 10.1093/nar/gkf431] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We show here that co-expression of murine CAD with either ICAD-L or ICAD-S in Escherichia coli as well as mammalian cells leads to a functional DFF complex, which after caspase-3 activation releases a nucleolytically active DNase. The chaperone activity of ICAD-S is between one and two orders of magnitude less effective than that of ICAD-L, as deduced from cleavage experiments with different activated recombinant DFF complexes produced in E.coli. With nucleolytically active EGFP fusion proteins of CAD it is demonstrated that co-expression of ICAD-S, which lacks the C-terminal domain of ICAD-L, including the NLS, leads to a homogeneous intracellular distribution of the DNase in transfected cells, whereas co-expression of human or murine ICAD-L variants lacking the NLS leads to exclusion of EGFP-CAD from the nuclei in approximately 50% of cells. These results attribute a particular importance of the NLS in the long isoform of the inhibitor of CAD for nuclear accumulation of the DFF complex in living cells. It is concluded that ICAD-L and ICAD-S in vivo might function as tissue-specific modulators in the regulation of apoptotic DNA degradation by controlling not only the enzymatic activity but also the amount of CAD available in the nuclei of mammalian cells.
Collapse
Affiliation(s)
- Sebastian Richard Scholz
- Institut für Biochemie (FB 08), Justus-Liebig-Universität, Heinrich-Buff-Ring 58, D-35392 Giessen, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Nagata T, Kishi H, Liu QL, Matsuda T, Imanaka T, Tsukada K, Kang D, Muraguchi A. The regulation of DNAse activities in subcellular compartments of activated thymocytes. Immunology 2002; 105:399-406. [PMID: 11985660 PMCID: PMC1782686 DOI: 10.1046/j.1365-2567.2002.01347.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thymocytes expressing self-reactive T-cell receptors (TCR) are eliminated in the thymus through a TCR-mediated signal. This cell death signal (negative selection) generates nuclear morphological change and DNA fragmentation in thymocytes. However, the pathway leading to DNA fragmentation of thymocytes following TCR engagement remains obscure. In this study, we investigated the localization and function of caspase-activated DNAse (CAD) and its inhibitor (ICAD) in thymocytes prior to or after in vivo TCR stimulation. We showed that CAD and ICAD are co-localized in microsome, nuclei and cytosol in unstimulated thymocytes. Following in vivo TCR engagement, ICAD located in cytosol and microsome was degraded and the resulting activated CAD induced chromosomal DNA fragmentation. CAD present in cytosol and microsome of unstimulated thymocytes was activated by recombinant caspase-3, and microsomal CAD was released to the cytosol. These results demonstrate that TCR engagement of thymocytes induces caspase-3-dependent activation of CAD localized in both cytosol and microsome, leading to DNA fragmentation in harmony.
Collapse
Affiliation(s)
- Takuya Nagata
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical UniversityToyama, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical UniversityToyama, Japan
| | - Qin Li Liu
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical UniversityToyama, Japan
| | - Tadashi Matsuda
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical UniversityToyama, Japan
| | - Tsuneo Imanaka
- Second Department of Surgery, Faculty of Medicine, Toyama Medical and Pharmaceutical UniversityToyama, Japan
| | - Kazuhiro Tsukada
- Department of Biological Chemistry, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical UniversityToyama, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu UniversityFukuoka, Japan
| | - Atsushi Muraguchi
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical UniversityToyama, Japan
| |
Collapse
|
23
|
Affiliation(s)
- Steven Bassnett
- Department of Ophthalmology and Visual Sciences and of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
24
|
Samejima K, Tone S, Earnshaw WC. CAD/DFF40 nuclease is dispensable for high molecular weight DNA cleavage and stage I chromatin condensation in apoptosis. J Biol Chem 2001; 276:45427-32. [PMID: 11577114 DOI: 10.1074/jbc.m108844200] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DNA degradation during apoptotic execution generally occurs at two levels: early as high molecular weight (HMW) fragments and later on as oligonucleosomal fragments. Two nucleases, CAD/CPAN/DFF40 and endonuclease G, can digest nuclear chromatin to produce the oligonucleosomal fragments, and it has been suggested that CAD might be responsible for HMW DNA cleavage. To more clearly define the role of CAD in nuclear disassembly, we have generated CAD(-/-) sublines of chicken DT40 cells in which the entire CAD open reading frame has been deleted. These cells grow normally and undergo apoptosis with kinetics essentially identical to wild type cells. However, they fail to undergo detectable oligonucleosomal fragmentation, proving that CAD is essential for this stage of DNA cleavage, at least in DT40 cells. Other aspects of nuclear disassembly, including HMW DNA cleavage and early stage apoptotic chromatin condensation against the nuclear periphery proceed normally in the absence of CAD. However, the final stages of chromatin condensation and nuclear fragmentation do not occur. Our results demonstrate that CAD is required for complete disassembly of the nucleus during apoptosis and reveal the existence of one or more as yet unidentified second factors responsible for HMW DNA cleavage and the early stages of apoptotic chromatin condensation.
Collapse
Affiliation(s)
- K Samejima
- Wellcome Institute for Cell Biology, Institute of Cell and Molecular Biology, Swann Bldg., University of Edinburgh, Mayfield Rd., Edinburgh EH9 3JR, Scotland, United Kingdom
| | | | | |
Collapse
|
25
|
Caspase-activated DNase/DNA fragmentation factor 40 mediates apoptotic DNA fragmentation in transient cerebral ischemia and in neuronal cultures. J Neurosci 2001. [PMID: 11425895 DOI: 10.1523/jneurosci.21-13-04678.2001] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nuclear changes, including internucleosomal DNA fragmentation, are characteristic features of neuronal apoptosis resulting from transient cerebral ischemia and related brain insults for which the molecular mechanism has not been elucidated. Recent studies suggest that a caspase-3-mediated mechanism may be involved in the process of nuclear degradation in ischemic neurons. In this study, we cloned from rat brain a homolog cDNA encoding caspase-activated deoxyribonuclease (CAD)/DNA fragmentation factor 40 (DFF40), a 40 kDa nuclear enzyme that is activated by caspase-3 and promotes apoptotic DNA degradation. Subsequently, we investigated the role of CAD/DFF40 in the induction of internucleosomal DNA fragmentation in the hippocampus in a rat model of transient global ischemia and in primary neuronal cultures under ischemia-like conditions. At 8-72 hr after ischemia, CAD/DFF40 mRNA and protein were induced in the degenerating hippocampal CA1 neurons. CAD/DFF40 formed a heterodimeric complex in the nucleus with its natural inhibitor CAD (ICAD) and was activated after ischemia in a delayed manner (>24 hr) by caspase-3, which translocated into the nucleus and cleaved ICAD. Furthermore, an induced CAD/DFF40 activity was detected in nuclear extracts in both in vivo and in vitro models, and the DNA degradation activity of CAD/DFF40 was inhibited by purified ICAD protein. These results strongly suggest that CAD/DFF40 is the endogenous endonuclease that mediates caspase-3-dependent internucleosomal DNA degradation and related nuclear alterations in ischemic neurons.
Collapse
|
26
|
Sharif-Askari E, Alam A, Rhéaume E, Beresford PJ, Scotto C, Sharma K, Lee D, DeWolf WE, Nuttall ME, Lieberman J, Sékaly RP. Direct cleavage of the human DNA fragmentation factor-45 by granzyme B induces caspase-activated DNase release and DNA fragmentation. EMBO J 2001; 20:3101-13. [PMID: 11406587 PMCID: PMC150191 DOI: 10.1093/emboj/20.12.3101] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The protease granzyme B (GrB) plays a key role in the cytocidal activity during cytotoxic T lymphocyte (CTL)-mediated programmed cell death. Multiple caspases have been identified as direct substrates for GrB, suggesting that the activation of caspases constitutes an important event during CTL-induced cell death. However, recent studies have provided evidence for caspase-independent pathway(s) during CTL-mediated apoptosis. In this study, we demonstrate caspase-independent and direct cleavage of the 45 kDa unit of DNA fragmentation factor (DFF45) by GrB both in vitro and in vivo. Using a novel and selective caspase-3 inhibitor, we show the ability of GrB to process DFF45 directly and mediate DNA fragmentation in the absence of caspase-3 activity. Furthermore, studies with DFF45 mutants reveal that both caspase-3 and GrB share a common cleavage site, which is necessary and sufficient to induce DNA fragmentation in target cells during apoptosis. Together, our data suggest that CTLs possess alternative mechanism(s) for inducing DNA fragmentation without the requirement for caspases.
Collapse
Affiliation(s)
- Ehsan Sharif-Askari
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Antoine Alam
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Eric Rhéaume
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Paul J. Beresford
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Christian Scotto
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Kamal Sharma
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Dennis Lee
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Walter E. DeWolf
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Mark E. Nuttall
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Judy Lieberman
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Rafick-Pierre Sékaly
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| |
Collapse
|
27
|
Chen D, Stetler RA, Cao G, Pei W, O'Horo C, Yin XM, Chen J. Characterization of the rat DNA fragmentation factor 35/Inhibitor of caspase-activated DNase (Short form). The endogenous inhibitor of caspase-dependent DNA fragmentation in neuronal apoptosis. J Biol Chem 2000; 275:38508-17. [PMID: 10984476 DOI: 10.1074/jbc.m003906200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nuclear changes, including internucleosomal DNA fragmentation, are classical manifestations of apoptosis for which the biochemical mechanisms have not been fully elucidated, particularly in neuronal cells. We have cloned the rat DNA fragmentation factor 35/inhibitor of caspase-activated DNase (short form) (DFF35/ICAD(S)) and found it to be the predominant form of ICAD present in rodent brain cells as well as in many other types of cells. DFF35/ICAD(S) forms a functional complex with DFF40/caspase-activated DNase (CAD) in the nucleus, and when its caspase-resistant mutant is over-expressed, it inhibits the nuclease activity, internucleosomal DNA fragmentation, and nuclear fragmentation but not the shrinkage and condensation of the nucleus, in neuron-differentiated PC12 cells in response to apoptosis inducers. DFF40/CAD is found to be localized mainly in the nucleus, and during neuronal apoptosis, there is no evidence of further nuclear translocation of this molecule. It is further suggested that inactivation of DFF40/CAD-bound DFF35 and subsequent activation of DFF40/CAD during apoptosis of neuronal cells may not occur in the cytosol but rather in the nucleus through a novel mechanism that requires nuclear translocation of caspases. These results establish that DFF35/ICAD(S) is the endogenous inhibitor of DFF40/CAD and caspase-dependent apoptotic DNA fragmentation in neurons.
Collapse
Affiliation(s)
- D Chen
- Departments of Neurology, Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Faleiro L, Lazebnik Y. Caspases disrupt the nuclear-cytoplasmic barrier. J Cell Biol 2000; 151:951-9. [PMID: 11085998 PMCID: PMC2174353 DOI: 10.1083/jcb.151.5.951] [Citation(s) in RCA: 189] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2000] [Accepted: 10/16/2000] [Indexed: 11/22/2022] Open
Abstract
During apoptosis, caspases, a family of proteases, disassemble a cell by cleaving a set of proteins. Caspase-3 plays a major role in the dissassembly of the nucleus by processing several nuclear substrates. The question is how caspase-3 which is usually cytoplasmic, gains access to its nuclear targets. It was suggested that caspase-3 is actively transported to the nucleus through the nuclear pores. We found that caspase-9, which is activated earlier than caspase-3, directly or indirectly inactivates nuclear transport and increases the diffusion limit of the nuclear pores. This increase allows caspase-3 and other molecules that could not pass through the nuclear pores in living cells to enter or leave the nucleus during apoptosis by diffusion. Hence, caspase-9 contributes to cell disassembly by disrupting the nuclear cytoplasmic barrier.
Collapse
Affiliation(s)
- L Faleiro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 11724, USA
| | | |
Collapse
|
29
|
Ventelä S, Okabe M, Tanaka H, Nishimune Y, Toppari J, Parvinen M. Expression of green fluorescent protein under beta-actin promoter in living spermatogenic cells of the mouse: stage-specific regulation by FSH. INTERNATIONAL JOURNAL OF ANDROLOGY 2000; 23:236-42. [PMID: 10886427 DOI: 10.1046/j.1365-2605.2000.00237.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Spermatogenic cells from a mouse strain expressing enhanced green fluorescent protein (EGFP) under chicken beta-actin promoter were studied under living conditions to analyse stage- and cell-specific expression and hormonal regulation of the transgene. The isolated seminiferous tubules were examined by transillumination and the live cell squashes by phase contrast and fluorescence microscopy. FSH effects were measured in whole seminiferous tubules comparing stages I-VI, VII-VIII and IX-XII of the cycle. Beta-actin was highly expressed in spermatogonia, but almost no expression was found at early meiosis (leptotene spermatocytes). A gradual increase in translation of beta-actin was found during later stages of meiosis and early spermiogenesis, with a maximum in elongating spermatids. FSH increased the translation of beta-actin after 4 h and 24 h of incubation at stages I-VI, after 24 h at stages VII-VIII but not at stages IX-XII of the cycle. The results support the view that beta-actin plays a role in the nuclear elongation of spermatids and that its expression is regulated by FSH in a stage-specific fashion. Techniques used in this study give us new insight to study temporal and hormonal regulation of gene products in living spermatogenic cells.
Collapse
Affiliation(s)
- S Ventelä
- Department of Anatomy, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland.
| | | | | | | | | | | |
Collapse
|
30
|
Durrieu F, Samejima K, Fortune JM, Kandels-Lewis S, Osheroff N, Earnshaw WC. DNA topoisomerase IIalpha interacts with CAD nuclease and is involved in chromatin condensation during apoptotic execution. Curr Biol 2000; 10:923-6. [PMID: 10959840 DOI: 10.1016/s0960-9822(00)00620-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Apoptotic execution is characterized by dramatic changes in nuclear structure accompanied by cleavage of nuclear proteins by caspases (reviewed in [1]). Cell-free extracts have proved useful for the identification and functional characterization of activities involved in apoptotic execution [2-4] and for the identification of proteins cleaved by caspases [5]. More recent studies have suggested that nuclear disassembly is driven largely by factors activated downstream of caspases [6]. One such factor, the caspase-activated DNase, CAD/CPAN/DFF40 [4,7,8] (CAD) can induce apoptotic chromatin condensation in isolated HeLa cell nuclei in the absence of other cytosolic factors [6,8]. As chromatin condensation occurs even when CAD activity is inhibited, however, CAD cannot be the sole morphogenetic factor triggered by caspases [6]. Here we show that DNA topoisomerase IIalpha (Topo IIalpha), which is essential for both condensation and segregation of daughter chromosomes in mitosis [9], also functions during apoptotic execution. Simultaneous inhibition of Topo IIalpha and caspases completely abolishes apoptotic chromatin condensation. In addition, we show that CAD binds to Topo IIalpha, and that their association enhances the decatenation activity of Topo IIalpha in vitro.
Collapse
Affiliation(s)
- F Durrieu
- Institute of Cell and Molecular Biology, University of Edinburgh, UK
| | | | | | | | | | | |
Collapse
|
31
|
Lechardeur D, Drzymala L, Sharma M, Zylka D, Kinach R, Pacia J, Hicks C, Usmani N, Rommens JM, Lukacs GL. Determinants of the nuclear localization of the heterodimeric DNA fragmentation factor (ICAD/CAD). J Cell Biol 2000; 150:321-34. [PMID: 10908575 PMCID: PMC2180231 DOI: 10.1083/jcb.150.2.321] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Programmed cell death or apoptosis leads to the activation of the caspase-activated DNase (CAD), which degrades chromosomal DNA into nucleosomal fragments. Biochemical studies revealed that CAD forms an inactive heterodimer with the inhibitor of caspase-activated DNase (ICAD), or its alternatively spliced variant, ICAD-S, in the cytoplasm. It was initially proposed that proteolytic cleavage of ICAD by activated caspases causes the dissociation of the ICAD/CAD heterodimer and the translocation of active CAD into the nucleus in apoptotic cells. Here, we show that endogenous and heterologously expressed ICAD and CAD reside predominantly in the nucleus in nonapoptotic cells. Deletional mutagenesis and GFP fusion proteins identified a bipartite nuclear localization signal (NLS) in ICAD and verified the function of the NLS in CAD. The two NLSs have an additive effect on the nuclear targeting of the CAD-ICAD complex, whereas ICAD-S, lacking its NLS, appears to have a modulatory role in the nuclear localization of CAD. Staurosporine-induced apoptosis evoked the proteolysis and disappearance of endogenous and exogenous ICAD from the nuclei of HeLa cells, as monitored by immunoblotting and immunofluorescence microscopy. Similar phenomenon was observed in the caspase-3-deficient MCF7 cells upon expressing procaspase-3 transiently. We conclude that a complex mechanism, involving the recognition of the NLSs of both ICAD and CAD, accounts for the constitutive accumulation of CAD/ICAD in the nucleus, where caspase-3-dependent regulation of CAD activity takes place.
Collapse
Affiliation(s)
- Delphine Lechardeur
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
| | - Luke Drzymala
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
| | - Manu Sharma
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada M5G 1X8
| | - Danuta Zylka
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
| | - Robert Kinach
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
| | - Joanna Pacia
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
| | - Christopher Hicks
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
| | - Nawaid Usmani
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
| | - Johanna M. Rommens
- Program in Genetics and Genomics Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
- Department of Molecular and Medical Genetics, University of Toronto, Toronto, Ontario, Canada M5G 1X8
| | - Gergely L. Lukacs
- Program in Cell and Lung Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada M5G 1X8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada M5G 1X8
| |
Collapse
|