1
|
Sporbeck K, Haas ML, Pastor-Maldonado CJ, Schüssele DS, Hunter C, Takacs Z, Diogo de Oliveira AL, Franz-Wachtel M, Charsou C, Pfisterer SG, Gubas A, Haller PK, Knorr RL, Kaulich M, Macek B, Eskelinen EL, Simonsen A, Proikas-Cezanne T. The ABL-MYC axis controls WIPI1-enhanced autophagy in lifespan extension. Commun Biol 2023; 6:872. [PMID: 37620393 PMCID: PMC10449903 DOI: 10.1038/s42003-023-05236-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Human WIPI β-propellers function as PI3P effectors in autophagy, with WIPI4 and WIPI3 being able to link autophagy control by AMPK and TORC1 to the formation of autophagosomes. WIPI1, instead, assists WIPI2 in efficiently recruiting the ATG16L1 complex at the nascent autophagosome, which in turn promotes lipidation of LC3/GABARAP and autophagosome maturation. However, the specific role of WIPI1 and its regulation are unknown. Here, we discovered the ABL-ERK-MYC signalling axis controlling WIPI1. As a result of this signalling, MYC binds to the WIPI1 promoter and represses WIPI1 gene expression. When ABL-ERK-MYC signalling is counteracted, increased WIPI1 gene expression enhances the formation of autophagic membranes capable of migrating through tunnelling nanotubes to neighbouring cells with low autophagic activity. ABL-regulated WIPI1 function is relevant to lifespan control, as ABL deficiency in C. elegans increased gene expression of the WIPI1 orthologue ATG-18 and prolonged lifespan in a manner dependent on ATG-18. We propose that WIPI1 acts as an enhancer of autophagy that is physiologically relevant for regulating the level of autophagic activity over the lifespan.
Collapse
Affiliation(s)
- Katharina Sporbeck
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Maximilian L Haas
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Carmen J Pastor-Maldonado
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - David S Schüssele
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Catherine Hunter
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Zsuzsanna Takacs
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Institute of Molecular Biotechnology, A-1030, Vienna, Austria
| | - Ana L Diogo de Oliveira
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Chara Charsou
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Simon G Pfisterer
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00290, Helsinki, Finland
| | - Andrea Gubas
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Patricia K Haller
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Roland L Knorr
- Humboldt University of Berlin, Institute of Biology, D-10115, Berlin, Germany
- Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- International Research Frontiers Initiative, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Manuel Kaulich
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Boris Macek
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Eeva-Liisa Eskelinen
- Department of Biosciences, University of Helsinki, Fl-00790, Helsinki, Finland
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Tassula Proikas-Cezanne
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
| |
Collapse
|
2
|
Qian D, Liu H, Zhao L, Luo S, Walsh KM, Huang J, Li CY, Wei Q. A pleiotropic ATM variant (rs1800057 C>G) is associated with risk of multiple cancers. Carcinogenesis 2021; 43:60-66. [PMID: 34643693 DOI: 10.1093/carcin/bgab092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 10/02/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
ATM (ataxia-telangiectasia mutated) is an important cell-cycle checkpoint kinase required for cellular response to DNA damage. Activated by DNA double strand breaks, ATM regulates the activities of many downstream proteins involved in various carcinogenic events. Therefore, ATM or its genetic variants may have a pleiotropic effect in cancer development. We conducted a pleiotropic analysis to evaluate associations between genetic variants of ATM and risk of multiple cancers. With genotyping data extracted from previously published genome-wide association studies of various cancers, we performed multivariate logistic regression analysis, followed by a meta-analysis for each cancer site, to identify cancer risk-associated single-nucleotide polymorphisms (SNPs). In the ASSET two-sided analysis, we found that two ATM SNPs were significantly associated with risk of multiple cancers. One tagging SNP (rs1800057 C>G) was associated with risk of multiple cancers (two-sided P=5.27×10 -7). Because ATM rs1800057 is a missense variant, we also explored the intermediate phenotypes through which this variant may confer risk of multiple cancers and identified a possible immune-mediated effect of this variant. Our findings indicate that genetic variants of ATM may have a pleiotropic effect on cancer risk and thus provide an important insight into common mechanisms of carcinogenesis.
Collapse
Affiliation(s)
- Danwen Qian
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China.,Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lingling Zhao
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kyle M Walsh
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Neurosurgery, Duke University, Durham, NC 27710, USA
| | - Jiaoti Huang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of pathology, Duke University, Durham, NC 27710, USA
| | - Chuan-Yuan Li
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
3
|
Tripathi R, Liu Z, Jain A, Lyon A, Meeks C, Richards D, Liu J, He D, Wang C, Nespi M, Rymar A, Wang P, Wilson M, Plattner R. Combating acquired resistance to MAPK inhibitors in melanoma by targeting Abl1/2-mediated reactivation of MEK/ERK/MYC signaling. Nat Commun 2020; 11:5463. [PMID: 33122628 PMCID: PMC7596241 DOI: 10.1038/s41467-020-19075-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 09/29/2020] [Indexed: 12/20/2022] Open
Abstract
Metastatic melanoma remains an incurable disease for many patients due to the limited success of targeted and immunotherapies. BRAF and MEK inhibitors reduce metastatic burden for patients with melanomas harboring BRAF mutations; however, most eventually relapse due to acquired resistance. Here, we demonstrate that ABL1/2 kinase activities and/or expression are potentiated in cell lines and patient samples following resistance, and ABL1/2 drive BRAF and BRAF/MEK inhibitor resistance by inducing reactivation of MEK/ERK/MYC signaling. Silencing/inhibiting ABL1/2 blocks pathway reactivation, and resensitizes resistant cells to BRAF/MEK inhibitors, whereas expression of constitutively active ABL1/2 is sufficient to promote resistance. Significantly, nilotinib (2nd generation ABL1/2 inhibitor) reverses resistance, in vivo, causing prolonged regression of resistant tumors, and also, prevents BRAFi/MEKi resistance from developing in the first place. These data indicate that repurposing the FDA-approved leukemia drug, nilotinib, may be effective for prolonging survival for patients harboring BRAF-mutant melanomas.
Collapse
Affiliation(s)
- Rakshamani Tripathi
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Zulong Liu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Aditi Jain
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.,The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anastasia Lyon
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Christina Meeks
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Dana Richards
- Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Jinpeng Liu
- Biostatistics and Bioinformatics Shared Resource Facility, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Daheng He
- Biostatistics and Bioinformatics Shared Resource Facility, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Chi Wang
- Biostatistics and Bioinformatics Shared Resource Facility, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | | | | | - Peng Wang
- Department of Internal Medicine, University of Kentucky, College of Medicine, Lexington, KY, USA
| | - Melissa Wilson
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rina Plattner
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.
| |
Collapse
|
4
|
Van Den Borg R, Leonetti A, Tiseo M, Giovannetti E, Peters GJ. Novel targeted strategies to overcome resistance in small-cell lung cancer: focus on PARP inhibitors and rovalpituzumab tesirine. Expert Rev Anticancer Ther 2019; 19:461-471. [PMID: 31148500 DOI: 10.1080/14737140.2019.1624530] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/24/2019] [Indexed: 10/26/2022]
Abstract
Introduction: Small-cell lung cancer (SCLC) is a highly aggressive neuroendocrine tumour, and its outcome is strongly conditioned by the rapid onset of resistance to conventional chemotherapeutics. First-line treatment with a combination of platinum agents and topoisomerase inhibitors has been the standard of care for over 30 years, with disappointing clinical outcome caused by early-acquired chemoresistance. In this disheartening scenario, novel treatment strategies are being implemented in order to either revert or bypass resistance mechanisms. Areas covered: The general mechanism of action of the standard frontline treatment regimens for SCLC, as well as the known resistance mechanisms to these drugs, is reviewed. Moreover, we focus on the current preclinical and clinical evidence on the potential role of PARP inhibitors and rovalpituzumab tesirine (Rova-T) to tackle chemoresistance in SCLC. Expert opinion: Preliminary evidence supports PARP inhibitors and Rova-T as two promising approaches to either revert or bypass chemoresistance in SCLC, respectively. The identification of potential predictive biomarkers of response to these innovative treatments (SLFN11 and DLL3) has shortened the gap between SCLC and personalized targeted therapy. Further large-scale clinical studies are urgently needed for a better designation of PARP inhibitors and Rova-T in the therapeutic algorithm of SCLC patients.
Collapse
Affiliation(s)
- Robin Van Den Borg
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands
| | - Alessandro Leonetti
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands
- b Medical Oncology Unit , University Hospital of Parma , Parma , Italy
| | - Marcello Tiseo
- b Medical Oncology Unit , University Hospital of Parma , Parma , Italy
- c Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Elisa Giovannetti
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands
- d Cancer Pharmacology Lab , AIRC Start-Up Unit , Pisa , Italy
| | - Godefridus J Peters
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands
| |
Collapse
|
5
|
Sun R, Song Y, Li S, Ma Z, Deng X, Fu Q, Qu R, Ma S. Levo-tetrahydropalmatine Attenuates Neuron Apoptosis Induced by Cerebral Ischemia–Reperfusion Injury: Involvement of c-Abl Activation. J Mol Neurosci 2018; 65:391-399. [DOI: 10.1007/s12031-018-1063-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/27/2018] [Indexed: 11/29/2022]
|
6
|
Chipps E, Protzman A, Muhi MZ, Ando S, Calvet JP, Islam MR. Nuclear Localization Signal and p53 Binding Site in MAP/ERK Kinase Kinase 1 (MEKK1). J Cell Biochem 2016; 116:2903-14. [PMID: 26018553 DOI: 10.1002/jcb.25238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 05/19/2015] [Indexed: 12/18/2022]
Abstract
Previously, we showed that Mekk1 translocates to the nucleus, interacts with tumor suppressor protein p53, and co-represses PKD1 transcription via an atypical p53 binding site on the minimal PKD1 promoter (JBC 285:38,818-38,831, 2010). In this study, we report the mechanisms of Mekk1 nuclear transport and p53 binding. Using GFP-linked constitutively active-Mekk1 (CA-Mekk1) and a deletion strategy, we identified a nuclear localization signal (HRDVK) located at amino acid (aa) residues 1,349-1,353 in the C-terminal Mekk1 catalytic domain. Deletion of this sequence in CA-Mekk1 and full-length Mekk1 significantly reduced their nuclear translocation in both HEK293T and COS-1 cells. Using co-immunoprecipitation, we identified an adjacent sequence (GANLID, aa 1,354-1,360) in Mekk1 responsible for p53 binding. Deletion of this sequence markedly reduced the interaction of Mekk1 with p53. Mekk1 does not appear to affect phosphorylation of Ser15, located in the Mdm2 interaction site, or other Ser residues in p53. However, Mekk1 mediates p53 protein stability in the presence of Mdm2 and reduces p53 ubiquitination, suggesting an interference with Mdm2-mediated degradation of p53 by the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Elizabeth Chipps
- Laboratory of Biochemistry, Garrett-Strong Science Building 3100, 800 University Drive, Northwest Missouri State University, Maryville, Missouri, 64468
| | - April Protzman
- Laboratory of Biochemistry, Garrett-Strong Science Building 3100, 800 University Drive, Northwest Missouri State University, Maryville, Missouri, 64468
| | - M Zubayed Muhi
- Laboratory of Biochemistry, Garrett-Strong Science Building 3100, 800 University Drive, Northwest Missouri State University, Maryville, Missouri, 64468
| | - Shoko Ando
- Laboratory of Biochemistry, Garrett-Strong Science Building 3100, 800 University Drive, Northwest Missouri State University, Maryville, Missouri, 64468
| | - James P Calvet
- Department of Biochemistry and Molecular Biology and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, 66160
| | - M Rafiq Islam
- Laboratory of Biochemistry, Garrett-Strong Science Building 3100, 800 University Drive, Northwest Missouri State University, Maryville, Missouri, 64468
| |
Collapse
|
7
|
Ho IL, Kuo KL, Liu SH, Chang HC, Hsieh JT, Wu JT, Chiang CK, Lin WC, Tsai YC, Chou CT, Hsu CH, Pu YS, Shi CS, Huang KH. MLN4924 Synergistically Enhances Cisplatin-induced Cytotoxicity via JNK and Bcl-xL Pathways in Human Urothelial Carcinoma. Sci Rep 2015; 5:16948. [PMID: 26592553 PMCID: PMC4655337 DOI: 10.1038/srep16948] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 10/22/2015] [Indexed: 01/09/2023] Open
Abstract
Cisplatin-based chemotherapy is the primary treatment for metastatic bladder urothelial carcinoma. However, the response rate is only 40–65%. This study investigated the anti-tumor effect and underlying mechanisms of the combination of cisplatin and the NEDD8-activating enzyme inhibitor MLN4924 in human bladder urothelial carcinoma. The combination of cisplatin and MLN4924 exerted synergistic cytotoxicity on two high-grade bladder urothelial carcinoma cell lines, NTUB1 and T24 (combination index <1). MLN4924 also potentiated the cisplatin-induced apoptosis and activation of caspase-3 and -7, phospho-histone H2A.X and PARP. c-Jun N-terminal kinase (JNK) activation and a down-regulation of B-cell lymphoma-extra large (Bcl-xL) were also observed during cisplatin and MLN4924 treatment. Inhibition of JNK activation partially restored cell viability and Bcl-xL expression. Bcl-xL overexpression also rescued cell viability. MLN4924 significantly potentiated cisplatin-induced tumor suppression in urothelial carcinoma xenograft mice. In summary, MLN4924 synergistically enhanced the anti-tumor effect of cisplatin via an increase in DNA damage, JNK activation and down-regulation of Bcl-xL in urothelial carcinoma cells. These findings provide a new therapeutic strategy for the treatment of bladder cancer.
Collapse
Affiliation(s)
- I-Lin Ho
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| | - Kuan-Lin Kuo
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shing-Hwa- Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hong-Chiang Chang
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| | - Ju-Ton Hsieh
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| | - June-Tai Wu
- Graduate Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Chou Lin
- Department of Pathology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Chieh Tsai
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Tso Chou
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Hsun Hsu
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| | - Yeong-Shiau Pu
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Sheng Shi
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-How Huang
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
Zhu S, Pabla N, Tang C, He L, Dong Z. DNA damage response in cisplatin-induced nephrotoxicity. Arch Toxicol 2015; 89:2197-205. [PMID: 26564230 DOI: 10.1007/s00204-015-1633-3] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/29/2015] [Indexed: 01/17/2023]
Abstract
Cisplatin and its derivatives are widely used chemotherapeutic drugs for cancer treatment. However, they have debilitating side effects in normal tissues and induce ototoxicity, neurotoxicity, and nephrotoxicity. In kidneys, cisplatin preferentially accumulates in renal tubular cells causing tubular cell injury and death, resulting in acute kidney injury (AKI). Recent studies have suggested that DNA damage and the associated DNA damage response (DDR) are an important pathogenic mechanism of AKI following cisplatin treatment. Activation of DDR may lead to cell cycle arrest and DNA repair for cell survival or, in the presence of severe injury, kidney cell death. Modulation of DDR may provide novel renoprotective strategies for cancer patients undergoing cisplatin chemotherapy.
Collapse
Affiliation(s)
- Shiyao Zhu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Navjotsingh Pabla
- Departments of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Georgia Regents University and Charlie Norwood VA Medical Center, 1459 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
9
|
Krestnikova N, Stulpinas A, Imbrasaite A, Sinkeviciute G, Kalvelyte AV. JNK implication in adipocyte-like cell death induced by chemotherapeutic drug cisplatin. J Toxicol Sci 2015; 40:21-32. [PMID: 25560393 DOI: 10.2131/jts.40.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Recent evidence shows that tumor microenvironment containing heterogeneous cells may be involved in cancer initiation, growth and tumor cell response to anticancer therapy. Chemotherapy was designed to make toxic impact on malicious cells in organisms, however, the means to protect healthy cells against chemical toxicity are still unsuccessful. As known, the majority of tumor surrounding cells are cancer-associated adipocytes which influence cancer development, progression and treatment. Targeting the components of tumor microenvironment in combination with conventional cancer treatment may become an effective cancer therapy strategy. However, little is known about adipocyte death mechanisms during combined chemo- and targeted therapy. The importance of c-Jun-NH<inf>2</inf>-terminal kinase (JNK) signaling in tumor development and treatment has been demonstrated using various in vitro and in vivo cancer models. The aim of this study was to ascertain adipocyte viability during simultaneous stress kinase JNK inhibition and exposure to one of the most commonly used anticancer drugs cis-diamminedichloroplatinum II (cisplatin). Our model involved adipocyte-like cells (ADC) which were obtained during in vitro differentiation of adult rabbit muscle-derived stem cells. Cisplatin induced apoptotic cell death. During 24-hr cisplatin treatment gradual, strong and prolonged increase of both JNK and its target protein c-Jun phosphorylation was found in ADC. Pre-treatment of cells with SP600125 decreased cisplatin-induced activation of c-Jun and promoted apoptosis. Upregulation of proapoptotic Bax and downregulation of antiapoptotic Bcl-2 proteins were found to be regulated in JNK-dependent manner. Thus, the results prove the antiapoptotic role of activated JNK in adipocyte-like cells treated with cisplatin.
Collapse
Affiliation(s)
- Natalija Krestnikova
- Vilnius University Institute of Biochemistry, Department of Molecular Cell Biology, Lithuania
| | | | | | | | | |
Collapse
|
10
|
2-Methoxy-1,4-naphthoquinone (MNQ) induces apoptosis of A549 lung adenocarcinoma cells via oxidation-triggered JNK and p38 MAPK signaling pathways. Life Sci 2015; 135:158-64. [DOI: 10.1016/j.lfs.2015.03.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/23/2015] [Accepted: 03/20/2015] [Indexed: 11/21/2022]
|
11
|
Non-receptor tyrosine kinase inhibitors enhances β-cell survival by suppressing the PKCδ signal transduction pathway in streptozotocin – induced β-cell apoptosis. Cell Signal 2015; 27:1066-74. [DOI: 10.1016/j.cellsig.2015.01.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 01/12/2015] [Accepted: 01/17/2015] [Indexed: 11/17/2022]
|
12
|
Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol 2014; 740:364-78. [PMID: 25058905 PMCID: PMC4146684 DOI: 10.1016/j.ejphar.2014.07.025] [Citation(s) in RCA: 3699] [Impact Index Per Article: 336.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 02/06/2023]
Abstract
Cisplatin, cisplatinum, or cis-diamminedichloroplatinum (II), is a well-known chemotherapeutic drug. It has been used for treatment of numerous human cancers including bladder, head and neck, lung, ovarian, and testicular cancers. It is effective against various types of cancers, including carcinomas, germ cell tumors, lymphomas, and sarcomas. Its mode of action has been linked to its ability to crosslink with the purine bases on the DNA; interfering with DNA repair mechanisms, causing DNA damage, and subsequently inducing apoptosis in cancer cells. However, because of drug resistance and numerous undesirable side effects such as severe kidney problems, allergic reactions, decrease immunity to infections, gastrointestinal disorders, hemorrhage, and hearing loss especially in younger patients, other platinum-containing anti-cancer drugs such as carboplatin, oxaliplatin and others, have also been used. Furthermore, combination therapies of cisplatin with other drugs have been highly considered to overcome drug-resistance and reduce toxicity. This comprehensive review highlights the physicochemical properties of cisplatin and related platinum-based drugs, and discusses its uses (either alone or in combination with other drugs) for the treatment of various human cancers. A special attention is paid to its molecular mechanisms of action, and its undesirable side effects.
Collapse
Affiliation(s)
- Shaloam Dasari
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA
| | - Paul Bernard Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| |
Collapse
|
13
|
Zeng W, Zhang J, Qi M, Peng C, Su J, Chen X, Yuan Z. αNAC inhibition of the FADD-JNK axis plays anti-apoptotic role in multiple cancer cells. Cell Death Dis 2014; 5:e1282. [PMID: 24901053 PMCID: PMC4611707 DOI: 10.1038/cddis.2014.192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/30/2014] [Accepted: 03/31/2014] [Indexed: 01/19/2023]
Abstract
Nascent polypeptide-associated complex α (αNAC) is reportedly overexpressed in several types of cancers and regulates cell apoptosis under hypoxic conditions in HeLa cells. The aim of our study was to investigate the apoptotic function of αNAC in cancer progression. First, we observed the cellular effects of αNAC depletion. Mouse αNAC was used to restore the protein level and verify the effect. An Annexin V assay, a caspase activity reporter assay, an apoptotic molecular marker, and a colony formation assay were used as markers to investigate the mechanisms of cell death caused by αNAC depletion. The Cancer 10-pathway reporter assay was used to screen downstream pathways. PCR site-directed deletion based on the functional domains of αNAC was used to construct deletion mutants. Those functional domain deletion mutants were used to recover the apoptotic phenotype caused by αNAC depletion. Finally, the role of αNAC in TNF-related apoptosis-inducing ligand (TRAIL) treatment was investigated in vitro. We found that depletion of αNAC in multiple types of cancer cells induce typical apoptotic cell death. This anti-apoptotic function is mediated by the FADD/c-Jun N-terminal kinase pathway. Intact αNAC is required for the direct binding of FADD as well as its anti-apoptosis function. Either αNAC depletion or the deletion of the ubiquitin-associated domain of αNAC sensitizes L929 cancer cells to mTRAIL treatment. Our study revealed a αNAC anti-apoptotic function in multiple types of cancer cells and suggested its potential in cancer therapy.
Collapse
Affiliation(s)
- W Zeng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - J Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - M Qi
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - C Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - J Su
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - X Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Z Yuan
- 1] Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People's Republic of China [2] Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
14
|
Apigenin induces the apoptosis and regulates MAPK signaling pathways in mouse macrophage ANA-1 cells. PLoS One 2014; 9:e92007. [PMID: 24646936 PMCID: PMC3960135 DOI: 10.1371/journal.pone.0092007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 02/19/2014] [Indexed: 02/06/2023] Open
Abstract
Apigenin is a naturally occurring plant flavonoid that possesses antioxidant, anti-cancer and anti-inflammatory properties. However, there are few reports has been done on the ability of apigenin to induce apoptosis in macrophages. In this study, mouse macrophage ANA-1 cells were incubated with different concentrations of apigenin. The cell viability was determined by an MTT assay. The cell apoptosis were analyzed by flow cytometric analysis. Apoptosis were also analyzed using a TUNEL assay and a DNA ladder. The level of intracellular ROS was detected using a dichlorofluorescein -diacetate probe. The expression levels of apoptosis-related proteins were detected by western blot analysis. The results showed that apigenin decreased the viability of ANA-1 cells and induced apoptosis in a dose- and time-dependent manner. Apigenin increased the level of intracellular ROS, downregulated the expression of Bcl-2 and upregulated the expression of caspase-3 and caspase-8 in ANA-1 cells. Furthermore, apigenin downregulated the expression of phospho-ERK and phospho-JNK, upregulated the expression of phospho-p38 and had no significant effect on the expression of Bax, ERK, JNK and p38. The results suggested that apigenin induced cell apoptosis in mouse macrophage ANA-1 cells may via increasing intracellular ROS, regulating the MAPK pathway, and then inhibiting Bcl-2 expression.
Collapse
|
15
|
Pham TT, Angus SP, Johnson GL. MAP3K1: Genomic Alterations in Cancer and Function in Promoting Cell Survival or Apoptosis. Genes Cancer 2014; 4:419-26. [PMID: 24386504 DOI: 10.1177/1947601913513950] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 11/02/2013] [Indexed: 12/15/2022] Open
Abstract
MAP3K1 is a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family of serine/threonine kinases. MAP3K1 regulates JNK activation and is unique among human kinases in that it also encodes an E3 ligase domain that ubiquitylates c-Jun and ERK1/2. Full length MAP3K1 regulates cell migration and contributes to pro-survival signaling while its caspase 3-mediated cleavage generates a C-terminal kinase domain that promotes apoptosis. The critical function of MAP3K1 in cell fate decisions suggests that it may be a target for deregulation in cancer. Recent large-scale genomic studies have revealed that MAP3K1 copy number loss and somatic missense or nonsense mutations are observed in a significant number of different cancers, being most prominent in luminal breast cancer. The alteration of MAP3K1 in diverse cancer types demonstrates the importance of defining phenotypes for possible therapeutic targeting of tumor cell vulnerabilities created when MAP3K1 function is lost or gained.
Collapse
Affiliation(s)
- Trang T Pham
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Steven P Angus
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Gary L Johnson
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Sanchez-Arévalo Lobo VJ, Doni M, Verrecchia A, Sanulli S, Fagà G, Piontini A, Bianchi M, Conacci-Sorrell M, Mazzarol G, Peg V, Losa JH, Ronchi P, Ponzoni M, Eisenman RN, Doglioni C, Amati B. Dual regulation of Myc by Abl. Oncogene 2013; 32:5261-71. [PMID: 23318434 DOI: 10.1038/onc.2012.621] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 11/20/2012] [Accepted: 11/21/2012] [Indexed: 12/12/2022]
Abstract
The tyrosine kinase c-Abl (or Abl) and the prolyl-isomerase Pin1 cooperatively activate the transcription factor p73 by enhancing recruitment of the acetyltransferase p300. As the transcription factor c-Myc (or Myc) is a known target of Pin1 and p300, we hypothesized that it might be regulated in a similar manner. Consistent with this hypothesis, overexpression of Pin1 augmented the interaction of Myc with p300 and transcriptional activity. The action of Abl, however, was more complex than predicted. On one hand, Abl indirectly enhanced phosphorylation of Myc on Ser 62 and Thr 58, its association with Pin1 and p300 and its acetylation by p300. These effects of Abl were exerted through phosphorylation of substrate(s) other than Myc itself. On the other hand, Abl interacted with the C-terminal domain of Myc and phosphorylated up to five tyrosine residues in its N-terminus, the principal of which was Y74. Indirect immunofluorescence or immunohistochemical staining suggested that the Y74-phosphorylated form of Myc (Myc-pY74) localized to the cytoplasm and coexisted either with active Abl in a subset of mammary carcinomas or with Bcr-Abl in chronic myeloid leukemia. In all instances, Myc-pY74 constituted a minor fraction of the cellular Myc protein. Thus, our data unravel two potential effects of Abl on Myc: first, Abl signaling can indirectly augment acetylation of Myc by p300, and most likely also its transcriptional activity in the nucleus; second, Abl can directly phosphorylate Myc on tyrosine: the resulting form of Myc appears to be cytoplasmic, and its presence correlates with Abl activation in cancer.
Collapse
|
17
|
Pejchal J, Novotný J, Mařák V, Österreicher J, Tichý A, Vávrová J, Šinkorová Z, Zárybnická L, Novotná E, Chládek J, Babicová A, Kubelková K, Kuča K. Activation of p38 MAPK and expression of TGF-β1 in rat colon enterocytes after whole body γ-irradiation. Int J Radiat Biol 2012; 88:348-58. [DOI: 10.3109/09553002.2012.654044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
18
|
Non-substituted N-heteroaromatic selenosemicarbazone metal complexes induce apoptosis in cancer cells via activation of mitochondrial pathway. Eur J Med Chem 2011; 46:3734-47. [DOI: 10.1016/j.ejmech.2011.05.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Revised: 05/09/2011] [Accepted: 05/14/2011] [Indexed: 11/18/2022]
|
19
|
Bensimon A, Aebersold R, Shiloh Y. Beyond ATM: the protein kinase landscape of the DNA damage response. FEBS Lett 2011; 585:1625-39. [PMID: 21570395 DOI: 10.1016/j.febslet.2011.05.013] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 05/04/2011] [Accepted: 05/04/2011] [Indexed: 01/18/2023]
Abstract
The DNA of all organisms is constantly subjected to damaging agents, both exogenous and endogenous. One extremely harmful lesion is the double-strand break (DSB), which activates a massive signaling network - the DNA damage response (DDR). The chief activator of the DSB response is the ATM protein kinase, which phosphorylates numerous key players in its various branches. Recent phosphoproteomic screens have extended the scope of damage-induced phosphorylations beyond the direct ATM substrates. We review the evidence for the involvement of numerous other protein kinases in the DDR, obtained from documentation of specific pathways as well as high-throughput screens. The emerging picture of the protein phosphorylation landscape in the DDR broadens the current view on the role of this protein modification in the maintenance of genomic stability. Extensive cross-talk between many of these protein kinases forms an interlaced signaling network that spans numerous cellular processes. Versatile protein kinases in this network affect pathways that are different from those they have been identified with to date. The DDR appears to be one of the most extensive signaling responses to cellular stimuli.
Collapse
Affiliation(s)
- Ariel Bensimon
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | | | | |
Collapse
|
20
|
Larroque-Lombard AL, Todorova M, Qiyu Q, Jean-Claude B. Synthesis and studies on three-compartment flavone-containing combi-molecules designed to target EGFR, DNA, and MEK. Chem Biol Drug Des 2011; 77:309-18. [PMID: 21294849 DOI: 10.1111/j.1747-0285.2011.01098.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In order to induce a tandem targeting of EGFR, DNA, and MEK, we built complex combi-molecules containing an EGFR targeting quinazoline and an aminoethyltriazene moiety linking the entire molecule to PD98059. Two complex molecules were synthesized: one with a short aminoethyl spacer, AL232, and the other AL414 with a longer aminoethylaminoethyl spacer. AL414 was a more potent inhibitor of EGFR tyrosine kinase than AL232. Both combi-molecules blocked EGFR phosphorylation in whole cells and downregulated extracellular signaling-regulated kinases (ERK1,2). However, only AL414 was capable of inducing DNA damage. Thus, it was taken in vivo for metabolic analysis. The results showed that 3 h after injection, AL414 was hydrolyzed to an EGFR inhibitor FD105, which was further acetylated to FD105Ac, a more potent inhibitor of EGFR. The detected flavone derivative was PD98059 linked to the hydroxyalkyl moiety resulting from the decomposition of the alkyldiazonium species. Independent synthesis of the latter metabolite and further in vitro analysis showed that it was deprived of antiproliferative activity. The results in toto suggest that while AL414 is a three-compartment combi-molecule, only the EGFR and DNA targeting species can be released and the cleavage to the intact MEK inhibitor PD98059 was mitigated by the stability of the carbamate.
Collapse
Affiliation(s)
- Anne-Laure Larroque-Lombard
- Cancer Drug Research Laboratory, Department of Medicine, Division of Medical Oncology, McGill University Health Center/Royal Victoria Hospital, 687 Pine Avenue West Rm M-719, Montreal, Quebec H3A 1A1, Canada
| | | | | | | |
Collapse
|
21
|
Helbig L, Damrot J, Hülsenbeck J, Köberle B, Brozovic A, Osmak M, Fiket Z, Kaina B, Fritz G. Late activation of stress-activated protein kinases/c-Jun N-terminal kinases triggered by cisplatin-induced DNA damage in repair-defective cells. J Biol Chem 2011; 286:12991-3001. [PMID: 21324906 DOI: 10.1074/jbc.m110.190645] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Although stress-activated protein kinases/c-Jun N-terminal kinases (SAPK/JNK) are rapidly activated by genotoxins, the role of DNA damage in this response is not well defined. Here we show that the SEK1/MKK4-mediated dual phosphorylation of SAPK/JNK (Thr-183/Tyr-185) correlates with the level of cisplatin-DNA adducts at late times (16-24 h) after drug treatment in both human and mouse cells. Transfection of platinated plasmid DNA also caused SAPK/JNK activation. A defect in transcription-coupled nucleotide excision repair resting on a mutation in Cockayne syndrome group B protein promoted the late SAPK/JNK activation following cisplatin exposure. Signaling to SAPK/JNK was accompanied by activation of Ataxia telangiectasia mutated- and Rad3-related kinase, replication protein A, and checkpoint kinases as well as by the formation of DNA double strand breaks (DSBs). Ionizing radiation-induced DSBs did not provoke SAPK/JNK activation, and inhibition of transcription also failed to provoke this response. Late activation of SAPK/JNK stimulated by cisplatin-induced DNA lesions was reduced in the absence of specific DNA repair proteins, such as xeroderma pigmentosum protein C, pointing to an essential function of individual repair factors in DNA damage signaling to SAPK/JNK. Collectively, the data indicate that late SAPK/JNK activation is triggered by non-repaired cisplatin adducts in transcribed genes and involves replication-associated events, DSBs, tyrosine kinases, Rho GTPases, and specific repair factors.
Collapse
Affiliation(s)
- Lars Helbig
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Orrenius S, Nicotera P, Zhivotovsky B. Cell Death Mechanisms and Their Implications in Toxicology. Toxicol Sci 2010; 119:3-19. [DOI: 10.1093/toxsci/kfq268] [Citation(s) in RCA: 276] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
23
|
Basu A, Krishnamurthy S. Cellular responses to Cisplatin-induced DNA damage. J Nucleic Acids 2010; 2010:201367. [PMID: 20811617 PMCID: PMC2929606 DOI: 10.4061/2010/201367] [Citation(s) in RCA: 335] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 06/28/2010] [Indexed: 12/29/2022] Open
Abstract
Cisplatin is one of the most effective anticancer agents widely used in the treatment of solid tumors. It is generally considered as a cytotoxic drug which kills cancer cells by damaging DNA and inhibiting DNA synthesis. How cells respond to cisplatin-induced DNA damage plays a critical role in deciding cisplatin sensitivity. Cisplatin-induced DNA damage activates various signaling pathways to prevent or promote cell death. This paper summarizes our current understandings regarding the mechanisms by which cisplatin induces cell death and the bases of cisplatin resistance. We have discussed various steps, including the entry of cisplatin inside cells, DNA repair, drug detoxification, DNA damage response, and regulation of cisplatin-induced apoptosis by protein kinases. An understanding of how various signaling pathways regulate cisplatin-induced cell death should aid in the development of more effective therapeutic strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Alakananda Basu
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center and Institute for Cancer Research, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | - Soumya Krishnamurthy
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center and Institute for Cancer Research, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| |
Collapse
|
24
|
Selective GSK-3β inhibitors attenuate the cisplatin-induced cytotoxicity of auditory cells. Hear Res 2009; 257:53-62. [DOI: 10.1016/j.heares.2009.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 07/31/2009] [Accepted: 08/01/2009] [Indexed: 12/20/2022]
|
25
|
Gonfloni S, Di Tella L, Caldarola S, Cannata SM, Klinger FG, Di Bartolomeo C, Mattei M, Candi E, De Felici M, Melino G, Cesareni G. Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death. Nat Med 2009; 15:1179-85. [PMID: 19783996 DOI: 10.1038/nm.2033] [Citation(s) in RCA: 262] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 08/25/2009] [Indexed: 11/09/2022]
Abstract
Germ cells are sensitive to genotoxins, and ovarian failure and infertility are major side effects of chemotherapy in young patients with cancer. Here we describe the c-Abl-TAp63 pathway activated by chemotherapeutic DNA-damaging drugs in model human cell lines and in mouse oocytes and its role in cell death. In cell lines, upon cisplatin treatment, c-Abl phosphorylates TAp63 on specific tyrosine residues. Such modifications affect p63 stability and induce a p63-dependent activation of proapoptotic promoters. Similarly, in oocytes, cisplatin rapidly promotes TAp63 accumulation and eventually cell death. Treatment with the c-Abl kinase inhibitor imatinib counteracts these cisplatin-induced effects. Taken together, these data support a model in which signals initiated by DNA double-strand breaks are detected by c-Abl, which, through its kinase activity, modulates the p63 transcriptional output. Moreover, they suggest a new use for imatinib, aimed at preserving oocytes of the follicle reserve during chemotherapeutic treatments.
Collapse
|
26
|
Ma D, Warabi E, Yanagawa T, Kimura S, Harada H, Yamagata K, Ishii T. Peroxiredoxin I plays a protective role against cisplatin cytotoxicity through mitogen activated kinase signals. Oral Oncol 2009; 45:1037-43. [PMID: 19692293 DOI: 10.1016/j.oraloncology.2009.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 07/01/2009] [Accepted: 07/02/2009] [Indexed: 11/26/2022]
Abstract
The anticancer agent cis-diamminedichloroplatinum (cisplatin) is a first-line chemotherapeutic agent for oral cancer. Cell exposure to cisplatin is associated with increased oxidative stress and post-translational changes in components of apoptosis pathways, including p38 Mitogen-activated protein kinase (MAPK), c-Jun-NH2-kinase (JNK), and extracellular signal-regulated kinase (ERK). Peroxiredoxin (Prx) I is an oxidative stress-inducible protein expressed in many tissues and important for reducing reactive oxygen species in vivo; however, whether Prx I helps protect cells from cisplatin injury is unknown. In this report, we examined the effects of Prx I on cell sensitivity to cisplatin-induced apoptosis. Mouse embryo fibroblasts (MEFs) derived from Prx I-deficient mice showed increased cisplatin-induced apoptosis compared with wild-type MEFs. Cisplatin treatment also led to increased activation of p38 MAPK and JNK, and reduced ERK phosphorylation in Prx I-deficient MEFs compared with wild-type MEFs. Furthermore, JNK- and ERK-specific inhibitors protected the Prx I-deficient MEFs from cisplatin-induced apoptosis, but Prx I-deficient MEFs remained more sensitive than wild-type MEFs when treated with a p38 MAPK-specific inhibitor. These findings indicate that Prx I modulates the cisplatin-evoked activation of MAPKs that lead to apoptosis, and Prx I may thus represent a useful target as a protective therapy against cisplatin cytotoxicity.
Collapse
Affiliation(s)
- Dongmei Ma
- Molecular Cellular Physiology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Wang S, Gong Z, Chen R, Liu Y, Li A, Li G, Zhou J. JWA regulates XRCC1 and functions as a novel base excision repair protein in oxidative-stress-induced DNA single-strand breaks. Nucleic Acids Res 2009; 37:1936-50. [PMID: 19208635 PMCID: PMC2665235 DOI: 10.1093/nar/gkp054] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
JWA was recently demonstrated to be involved in cellular responses to environmental stress including oxidative stress. Although it was found that JWA protected cells from reactive oxygen species-induced DNA damage, upregulated base excision repair (BER) protein XRCC1 and downregulated PARP-1, the molecular mechanism of JWA in regulating the repair of DNA single-strand breaks (SSBs) is still unclear. Our present studies demonstrated that a reduction in JWA protein levels in cells resulted in a decrease of SSB repair capacity and hypersensitivity to DNA-damaging agents such as methyl methanesulfonate and hydrogen peroxide. JWA functioned as a repair protein by multi-interaction with XRCC1. On the one hand, JWA was translocated into the nucleus by the carrier protein XRCC1 and co-localized with XRCC1 foci after oxidative DNA damage. On the other hand, JWA via MAPK signaling pathway regulated nuclear factor E2F1, which further transcriptionally regulated XRCC1. In addition, JWA protected XRCC1 protein from ubiquitination and degradation by proteasome. These findings indicate that JWA may serve as a novel regulator of XRCC1 in the BER protein complex to facilitate the repair of DNA SSBs.
Collapse
Affiliation(s)
- Shouyu Wang
- Department of Molecular Cell Biology and Toxicology, Cancer Centre, School of Public Health, Nanjing Medical University, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
28
|
Ståhl S, Fung E, Adams C, Lengqvist J, Mörk B, Stenerlöw B, Lewensohn R, Lehtiö J, Zubarev R, Viktorsson K. Proteomics and pathway analysis identifies JNK signaling as critical for high linear energy transfer radiation-induced apoptosis in non-small lung cancer cells. Mol Cell Proteomics 2009; 8:1117-29. [PMID: 19168796 DOI: 10.1074/mcp.m800274-mcp200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During the past decade, we have witnessed an explosive increase in generation of large proteomics data sets, not least in cancer research. There is a growing need to extract and correctly interpret information from such data sets to generate biologically relevant hypotheses. A pathway search engine (PSE) has recently been developed as a novel tool intended to meet these requirements. Ionizing radiation (IR) is an anticancer treatment modality that triggers multiple signal transduction networks. In this work, we show that high linear energy transfer (LET) IR induces apoptosis in a non-small cell lung cancer cell line, U-1810, whereas low LET IR does not. PSE was applied to study changes in pathway status between high and low LET IR to find pathway candidates of importance for high LET-induced apoptosis. Such pathways are potential clinical targets, and they were further validated in vitro. We used an unsupervised shotgun proteomics approach where high resolution mass spectrometry coupled to nanoflow liquid chromatography determined the identity and relative abundance of expressed proteins. Based on the proteomics data, PSE suggested the JNK pathway (p = 6.10(-6)) as a key event in response to high LET IR. In addition, the Fas pathway was found to be activated (p = 3.10(-5)) and the p38 pathway was found to be deactivated (p = 0.001) compared with untreated cells. Antibody-based analyses confirmed that high LET IR caused an increase in phosphorylation of JNK. Moreover pharmacological inhibition of JNK blocked high LET-induced apoptotic signaling. In contrast, neither an activation of p38 nor a role for p38 in high LET IR-induced apoptotic signaling was found. We conclude that, in contrast to conventional low LET IR, high LET IR can trigger activation of the JNK pathway, which in turn is critical for induction of apoptosis in these cells. Thus PSE predictions were largely confirmed, and PSE was proven to be a useful hypothesis-generating tool.
Collapse
Affiliation(s)
- Sara Ståhl
- Department of Oncology/Pathology, Karolinska Biomics Center, Karolinska Institutet, S-17176 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kim HJ, So HS, Lee JH, Park C, Lee JB, Youn MJ, Kim SJ, Yang SH, Lee KM, Kwon KB, Park BH, Park R. Role of proinflammatory cytokines in cisplatin-induced vestibular hair cell damage. Head Neck 2009; 30:1445-56. [PMID: 18642321 DOI: 10.1002/hed.20892] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cisplatin causes the impairment of inner ear functions, including hearing and balance, through the involvement of a number of mechanisms. However, no laboratory studies have been performed on involvement of inflammation-related events in cisplatin-mediated vestibular dysfunction. METHODS We evaluated the secretion of proinflammatory cytokines and nuclear factor-kappaB (NF-kappaB) activation in cisplatin-treated UB/UE-1 utricular epithelial cells. We also employed immunohistochemistry to detect proinflammatory cytokines and NF-kappaB expression in cisplatin-injected mice. RESULTS Productions of proinflammatory cytokines significantly caused the death of UB/UE1 cells by cisplatin. Pharmacologic inhibition of mitogen-activated protein (MAP) kinase/ERK kinase-1 (MEK1) or extracellular signal-regulated kinase (ERK) significantly attenuated the death of UB/UE1 cells caused by cisplatin and proinflammatory cytokines. Immunohistochemical studies revealed an increase in the expression of proinflammatory cytokines and NF-kappaB in both the cristae ampullae and utricle of cisplatin-injected mice. CONCLUSIONS These results suggest that proinflammatory cytokines may play an important role in the pathogenesis of cisplatin-mediated vestibulo-toxicity.
Collapse
Affiliation(s)
- Hyung-Jin Kim
- Vestibulocochlear Research Center and Department of Microbiology, School of Medicine, Wonkwang University, Jeonbuk, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sánchez-Olea R, Calera MR, Degterev A. Molecular pathways involved in cell death after chemically induced DNA damage. EXS 2009; 99:209-30. [PMID: 19157063 DOI: 10.1007/978-3-7643-8336-7_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
DNA damage is at the center of the genesis, progression and treatment of cancer. We review here the molecular mechanisms of the DNA damage inducing small molecules most commonly used in cancer therapy. Cell cycle control and DNA repair mechanisms are known to be activated after DNA damage. Here, we revise recent discoveries related to the cell cycle control and DNA repair processes and how these findings are being utilized for the more efficient, powerful and selective therapies for cancer treatment.
Collapse
|
31
|
Hägerkvist R, Jansson L, Welsh N. Imatinib mesylate improves insulin sensitivity and glucose disposal rates in rats fed a high-fat diet. Clin Sci (Lond) 2008; 114:65-71. [PMID: 17868036 DOI: 10.1042/cs20070122] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The aim of the present study was to investigate whether imatinib affects insulin sensitivity and glucose disposal in HF (high-fat)-fed rats. Sprague-Dawley rats were fed either a standard pelleted rat food (low-fat diet) or an HF diet (60% fat) for 8 weeks. During the last 10 days of the HF diet regime, rats received saline alone or imatinib (50 or 100 mg/kg of body weight) daily by gavage. The higher dose of imatinib resulted in a decreased psoas fat pad weight in the HF-treated rats. Under euglycaemic hyperinsulinaemic clamp conditions, HF-fed rats exhibited increased insulin concentrations and decreased glucose disposal. The lower (50 mg/kg of body weight), but not the higher (100 mg/kg of body weight), dose of imatinib normalized insulin sensitivity and glucose disposal without affecting glucose metabolism in low-fat-fed rats. Hepatic glucose production at both fasting and hyperinsulinaemic conditions was only weakly affected by imatinib. We conclude that a moderate dose of imatinib efficiently counteracts HF-induced peripheral insulin resistance, and that further studies on the mechanisms by which imatinib increases insulin action in muscle and fat tissues might generate novel strategies for the treatment of Type 2 diabetes.
Collapse
Affiliation(s)
- Robert Hägerkvist
- Department of Medical Cell Biology, Uppsala University, S-75123 Uppsala, Sweden
| | | | | |
Collapse
|
32
|
Galan-Moya EM, Hernandez-Losa J, Aceves Luquero CI, de la Cruz-Morcillo MA, Ramírez-Castillejo C, Callejas-Valera JL, Arriaga A, Aranburo AF, Ramón y Cajal S, Silvio Gutkind J, Sánchez-Prieto R. c-Abl activates p38 MAPK independently of its tyrosine kinase activity: Implications in cisplatin-based therapy. Int J Cancer 2008; 122:289-97. [PMID: 17893873 DOI: 10.1002/ijc.23063] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Activation of p38 MAPK is a critical requisite for the therapeutics activity of the antitumor agent cisplatin. In this sense, a growing body of evidences supports the role of c-Abl as a major determinant of p38 MAPK activation, especially in response to genotoxic stress when triggered by cisplatin. Here, we demonstrate that p38 MAPK activation in response to cisplatin does not require the tyrosine kinase activity of c-Abl. Indeed, c-Abl can activate the p38 MAPK signaling pathway by a mechanism that is independent of its tyrosine kinase activity, but that instead involves the ability of c-Abl to increase the stability of MKK6. Similar results were obtained in chronic myeloid leukemia-derived cell lines, in which a chimeric Bcr/Abl protein mimics the effects of c-Abl overexpression on p38 MAPK activation. These findings may explain why a clinically used c-Abl inhibitor, imatinib mesylate, fails to inhibit the p38 MAPK pathway alone or in combination with cisplatin, and provide evidence of a novel signaling mechanism in which these antitumor agents act.
Collapse
|
33
|
Kim WJ, Rajasekaran B, Brown KD. MLH1- and ATM-dependent MAPK signaling is activated through c-Abl in response to the alkylator N-methyl-N'-nitro-N'-nitrosoguanidine. J Biol Chem 2007; 282:32021-31. [PMID: 17804421 DOI: 10.1074/jbc.m701451200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
N-Methyl-N'-nitro-N'-nitrosoguanidine (MNNG) is a DNA-methylating agent, and deficiency in mismatch repair (MMR) results in lack of sensitivity to this genotoxin (termed alkylation tolerance). A number of DNA damage response pathways are activated in a MMR-dependent manner following MNNG, and several also require ATM kinase activity. Here we show that activation of the transcription factor c-Jun is dependent upon both the MMR component MLH1 and ATM, but not ATR, in response to MNNG. In addition to c-Jun, the upstream MAPKs JNK and MKK4 are also activated in a MLH1- and ATM-dependent manner. We document that c-Jun activation is dependent on the MAPK kinase kinase MEKK1. Additionally, the tyrosine kinase c-Abl is required to activate this signaling cascade and forms a complex with MEKK1 and MLH1. This study indicates that an arm of DNA damage-activated MAPK signaling is activated in an MLH1- and ATM-dependent manner in response to MNNG and perhaps suggests that dysregulation of this signaling is responsible, in part, for alkylation tolerance.
Collapse
Affiliation(s)
- Wan-Ju Kim
- Department of Biochemistry and Molecular Biology and the University of Florida Shands Cancer Center, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | |
Collapse
|
34
|
Brozovic A, Osmak M. Activation of mitogen-activated protein kinases by cisplatin and their role in cisplatin-resistance. Cancer Lett 2007; 251:1-16. [DOI: 10.1016/j.canlet.2006.10.007] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Revised: 09/07/2006] [Accepted: 10/11/2006] [Indexed: 10/23/2022]
|
35
|
So H, Kim H, Lee JH, Park C, Kim Y, Kim E, Kim JK, Yun KJ, Lee KM, Lee HY, Moon SK, Lim DJ, Park R. Cisplatin cytotoxicity of auditory cells requires secretions of proinflammatory cytokines via activation of ERK and NF-kappaB. J Assoc Res Otolaryngol 2007; 8:338-55. [PMID: 17516123 PMCID: PMC2538433 DOI: 10.1007/s10162-007-0084-9] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Accepted: 04/23/2007] [Indexed: 12/17/2022] Open
Abstract
The ototoxicity of cisplatin, a widely used chemotherapeutic agent, involves a number of mechanisms, including perturbation of redox status, increase in lipid peroxidation, and formation of DNA adducts. In this study, we demonstrate that cisplatin increased the early immediate release and de novo synthesis of proinflammatory cytokines, including TNF-alpha, IL-1beta, and IL-6, through the activation of ERK and NF-kappaB in HEI-OC1 cells, which are conditionally immortalized cochlear cells that express hair cell markers. Both neutralization of proinflammatory cytokines and pharmacologic inhibition of ERK significantly attenuated the death of HEI-OC1 auditory cells caused by cisplatin and proinflammatory cytokines. We also observed a significant increase in the protein and mRNA levels of proinflammatory cytokines in both serum and cochleae of cisplatin-injected rats, which was suppressed by intraperitoneal injection of etanercept, an inhibitor of TNF-alpha. Immunohistochemical studies revealed that TNF-alpha expression was mainly located in the spiral ligament, spiral limbus, and the organ of Corti in the cochleae of cisplatin-injected rats. NF-kappaB protein expression, which overlapped with terminal deoxynucleotidyl transferase-mediated dUTP nick-end-labeling-positive signal, was very strong in specific regions of the cochleae, including the organ of Corti, spiral ligament, and stria vascularis. These results indicate that proinflammatory cytokines, especially TNF-alpha, play a central role in the pathophysiology of sensory hair cell damage caused by cisplatin.
Collapse
Affiliation(s)
- Hongseob So
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Department of Microbiology, Wonkwang University School of Medicine, 344-7 Shinyong-dong Iksan, Jeonbuk, 570-749 South Korea
| | - HyungJin Kim
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Division of Biological Sciences, Chonbuk National University, Jeonbuk, 561-756 South Korea
| | - Jeong-Han Lee
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Department of Microbiology, Wonkwang University School of Medicine, 344-7 Shinyong-dong Iksan, Jeonbuk, 570-749 South Korea
| | - Channy Park
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Department of Microbiology, Wonkwang University School of Medicine, 344-7 Shinyong-dong Iksan, Jeonbuk, 570-749 South Korea
| | - Yunha Kim
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Department of Microbiology, Wonkwang University School of Medicine, 344-7 Shinyong-dong Iksan, Jeonbuk, 570-749 South Korea
| | - Eunsook Kim
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Department of Microbiology, Wonkwang University School of Medicine, 344-7 Shinyong-dong Iksan, Jeonbuk, 570-749 South Korea
| | - Jin-Kyung Kim
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Department of Microbiology, Wonkwang University School of Medicine, 344-7 Shinyong-dong Iksan, Jeonbuk, 570-749 South Korea
| | - Ki-Jung Yun
- Department of Pathology, Wonkwang University School of Medicine, Jeonbuk, South Korea 570-749
| | - Kang-Min Lee
- Division of Biological Sciences, Chonbuk National University, Jeonbuk, 561-756 South Korea
| | - Haa-Yung Lee
- Gonda Department of Cell and Molecular Biology, House Ear Institute, Los Angeles, CA 90057 USA
| | - Sung-Kyun Moon
- Gonda Department of Cell and Molecular Biology, House Ear Institute, Los Angeles, CA 90057 USA
| | - David J. Lim
- Gonda Department of Cell and Molecular Biology, House Ear Institute, Los Angeles, CA 90057 USA
| | - Raekil Park
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Jeonbuk, 570-749 South Korea
- Department of Microbiology, Wonkwang University School of Medicine, 344-7 Shinyong-dong Iksan, Jeonbuk, 570-749 South Korea
| |
Collapse
|
36
|
Autret A, Martin-Latil S, Mousson L, Wirotius A, Petit F, Arnoult D, Colbère-Garapin F, Estaquier J, Blondel B. Poliovirus induces Bax-dependent cell death mediated by c-Jun NH2-terminal kinase. J Virol 2007; 81:7504-16. [PMID: 17494073 PMCID: PMC1933371 DOI: 10.1128/jvi.02690-06] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Poliovirus (PV) is the causal agent of paralytic poliomyelitis, a disease that involves the destruction of motor neurons associated with PV replication. In PV-infected mice, motor neurons die through an apoptotic process. However, mechanisms by which PV induces cell death in neuronal cells remain unclear. Here, we demonstrate that PV infection of neuronal IMR5 cells induces cytochrome c release from mitochondria and loss of mitochondrial transmembrane potential, both of which are evidence of mitochondrial outer membrane permeabilization. PV infection also activates Bax, a proapoptotic member of the Bcl-2 family; this activation involves its conformational change and its redistribution from the cytosol to mitochondria. Neutralization of Bax by vMIA protein expression prevents cytochrome c release, consistent with a contribution of PV-induced Bax activation to mitochondrial outer membrane permeabilization. Interestingly, we also found that c-Jun NH(2)-terminal kinase (JNK) is activated soon after PV infection and that the PV-cell receptor interaction alone is sufficient to induce JNK activation. Moreover, the pharmacological inhibition of JNK by SP600125 inhibits Bax activation and cytochrome c release. This is, to our knowledge, the first demonstration of JNK-mediated Bax-dependent apoptosis in PV-infected cells. Our findings contribute to our understanding of poliomyelitis pathogenesis at the cellular level.
Collapse
Affiliation(s)
- Arnaud Autret
- Biologie des Virus Entériques, Institut Pasteur, 28 rue du Docteur Roux, 75724 Paris cedex 15, France
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Qi H, Lin CP, Fu X, Wood LM, Liu AA, Tsai YC, Chen Y, Barbieri CM, Pilch DS, Liu LF. G-quadruplexes induce apoptosis in tumor cells. Cancer Res 2007; 66:11808-16. [PMID: 17178877 DOI: 10.1158/0008-5472.can-06-1225] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Several G-rich oligodeoxynucleotides (ODNs), which are capable of forming G-quadruplexes, have been shown to exhibit antiproliferative activity against tumor cell lines and antitumor activity in nude mice carrying prostate and breast tumor xenografts. However, the molecular basis for their antitumor activity remains unclear. In the current study, we showed that a variety of telomeric G-tail oligodeoxynucleotides (TG-ODNs) exhibited antiproliferative activity against many tumor cells in culture. Systematic mutational analysis of the TG-ODNs suggests that the antiproliferative activity depends on the G-quadruplex conformation of these TG-ODNs. TG-ODNs were also shown to induce poly(ADP-ribose) polymerase-1 cleavage, phosphatidylserine flipping, and caspase activation, indicative of induction of apoptosis. TG-ODN-induced apoptosis was largely ataxia telangiectasia mutated (ATM) dependent. Furthermore, TG-ODN-induced apoptosis was inhibited by the c-Jun NH(2)-terminal kinase (JNK) inhibitor SP600125. Indeed, TG-ODNs were shown to activate the JNK pathway in an ATM-dependent manner as evidenced by elevated phosphorylation of JNK and c-Jun. Interestingly, a number of G-quadruplex ODNs (GQ-ODN) derived from nontelomeric sequences also induced ATM/JNK-dependent apoptosis, suggesting a possible common mechanism of tumor cell killing by GQ-ODNs.
Collapse
Affiliation(s)
- Haiyan Qi
- Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kamath R, Jiang Z, Sun G, Yalowich JC, Baskaran R. c-Abl kinase regulates curcumin-induced cell death through activation of c-Jun N-terminal kinase. Mol Pharmacol 2007; 71:61-72. [PMID: 17021249 DOI: 10.1124/mol.106.026575] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Curcumin, a natural phenolic compound found in turmeric (Curcuma longa) exhibits anticancer properties, attributed to its antiproliferative and apoptosis-inducing activity. The ubiquitously expressed nonreceptor tyrosine kinase c-Abl regulates stress responses induced by oxidative agents such as ionizing radiation and H2O2. In this study, we show that c-Abl is an important component of the cell death response activated by curcumin and that Abl mediates this response partly through activation of c-Jun N-terminal kinase (JNK). Therefore, inhibition of Abl by STI571 [imatinib (Gleevec)] treatment or down-regulation of Abl expression through Abl-specific short-hairpin RNA (shRNA) diminished cell death induction and JNK activation. Highlighting the interdependent nature of the Abl and JNK signaling in the curcumin-induced cell death response, a JNK inhibitor [anthra(1,9-cd)pyrazol-6(2H)-one-1,9-pyrazoloanthrone (SP600125)] caused very little cell death inhibition in STI571-pretreated cells and in Abl shRNA-expressing cells. Moreover, treatment with Abl and JNK inhibitor alone or together caused similar levels of cell death inhibition. Although p53 induction in response to curcumin treatment is dependent on Abl, we found that Abl-->p53 signaling is not necessary for curcumin-induced cell death. Taken together, the results demonstrate the differential roles played by Abl-->p53 and Abl-->JNK signaling events in modulating the cell death response to curcumin.
Collapse
Affiliation(s)
- Ravindra Kamath
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, E1205 Biomedical Science Tower, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
39
|
Hägerkvist R, Sandler S, Mokhtari D, Welsh N. Amelioration of diabetes by imatinib mesylate (Gleevec): role of beta-cell NF-kappaB activation and anti-apoptotic preconditioning. FASEB J 2006; 21:618-28. [PMID: 17135364 DOI: 10.1096/fj.06-6910com] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
It was recently reported that tyrosine kinase inhibitor imatinib mesylate (Gleevec) improves Type 2 diabetes, possibly by decreasing insulin resistance. However, as both Type 2 and Type 1 diabetes are characterized by beta-cell dysfunction and death, we investigated whether imatinib counteracts diabetes by maintaining beta-cell function. We observed that imatinib counteracted diabetes in two animal models, the streptozotocin-injected mouse and the nonobese diabetes mouse, and that this was paralleled by a partial preservation of the beta-cell mass. In addition, imatinib decreased the death of human beta-cells in vitro when exposed to NO, cytokines, and streptozotocin. The imatinib effect was mimicked by siRNA-mediated knockdown of c-Abl mRNA. Imatinib enhanced beta-cell survival by promoting a state similar to ischemic preconditioning, as evidenced by NF-kappaB activation, increased NO and reactive oxygen species production, and depolarization of the inner mitochondrial membrane. Imatinib did not suppress islet cell death in the presence of an NF-kappaB inhibitor, suggesting that NF-kappaB activation is a necessary step in the antiapoptotic action of imatinib. We conclude that imatinib mediates beta-cell survival and that this could contribute to the beneficial effects observed in diabetes.
Collapse
Affiliation(s)
- Robert Hägerkvist
- Department of Medical Cell Biology, Uppsala University, Biomedicum, P.O. Box 571, SE-75123 Uppsala, Sweden
| | | | | | | |
Collapse
|
40
|
Abstract
Reactive oxygen species (ROS) play critical roles for the determination of cell fate by eliciting a wide variety of cellular responses, such as proliferation, differentiation and apoptosis. Many intracellular signaling pathways involved in such ROS-induced cellular responses are regulated by the intracellular redox state, which depends on the balance between the levels of oxidizing and reducing equivalents. Recently, increasing attention has been paid to the roles of thioredoxin (Trx) as a signaling intermediate beyond its intrinsic antioxidant activity. Especially, Trx participates in the control of the mitogen-activated protein kinase (MAPK) cascades through the redox state-dependent association and dissociation with apoptosis signal-regulating kinase 1 (ASK1), an upstream regulator of the cascades. This review highlights the current understanding of prototypical molecular mechanisms by which the redox signal is converted into the signaling through ROS-responsive protein kinases, with a special focus on the ASK1-Trx system. Understanding of such mechanisms may provide the basis for therapeutic interventions in redox-related diseases including various types of cancer.
Collapse
Affiliation(s)
- Go Fujino
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, CREST, Japan Science and Technology Corporation, and Strategic Approach to Drug Discovery and Development in Pharmaceutical Sciences, Tokyo, Japan
| | | | | | | |
Collapse
|
41
|
Raina D, Ahmad R, Kumar S, Ren J, Yoshida K, Kharbanda S, Kufe D. MUC1 oncoprotein blocks nuclear targeting of c-Abl in the apoptotic response to DNA damage. EMBO J 2006; 25:3774-83. [PMID: 16888623 PMCID: PMC1553184 DOI: 10.1038/sj.emboj.7601263] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Accepted: 07/07/2006] [Indexed: 11/08/2022] Open
Abstract
The nonreceptor c-Abl tyrosine kinase binds to cytosolic 14-3-3 proteins and is targeted to the nucleus in the apoptotic response to DNA damage. The MUC1 oncoprotein is overexpressed by most human carcinomas and blocks the induction of apoptosis by genotoxic agents. Using human carcinoma cells with gain and loss of MUC1 function, we show that nuclear targeting of c-Abl by DNA damage is abrogated by a MUC1-dependent mechanism. The results demonstrate that c-Abl phosphorylates MUC1 on Tyr-60 and forms a complex with MUC1 by binding of the c-Abl SH2 domain to the pTyr-60 site. Binding of MUC1 to c-Abl attenuates phosphorylation of c-Abl on Thr-735 and the interaction between c-Abl and cytosolic 14-3-3. We also show that expression of MUC1 with a mutation at Tyr-60 (i) disrupts the interaction between MUC1 and c-Abl, (ii) relieves the MUC1-induced block of c-Abl phosphorylation on Thr-735 and binding to 14-3-3, and (iii) attenuates the MUC1 antiapoptotic function. These findings indicate that MUC1 sequesters c-Abl in the cytoplasm and thereby inhibits apoptosis in the response to genotoxic anticancer agents.
Collapse
Affiliation(s)
- Deepak Raina
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rehan Ahmad
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Shailendra Kumar
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jian Ren
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kiyotsugu Yoshida
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Binny Street, Boston, MA 02115, USA. Tel.: +1 617 632 3141 Fax: +1 617 632 2934; E-mail:
| |
Collapse
|
42
|
Kim HJ, So HS, Lee JH, Lee JH, Park C, Park SY, Kim YH, Youn MJ, Kim SJ, Chung SY, Lee KM, Park R. Heme oxygenase-1 attenuates the cisplatin-induced apoptosis of auditory cells via down-regulation of reactive oxygen species generation. Free Radic Biol Med 2006; 40:1810-9. [PMID: 16678019 DOI: 10.1016/j.freeradbiomed.2006.01.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2005] [Revised: 01/10/2006] [Accepted: 01/13/2006] [Indexed: 10/25/2022]
Abstract
Heme oxygenase-1 (HO-1), the rate-limiting enzyme of heme catabolism, is known to modulate various cellular functions, including cytokine production, cell proliferation, and apoptosis, in stress-related conditions. However, the role of HO-1 in the auditory system remains elusive. Herein, we demonstrate that pharmacologic induction of HO-1 along with catalytic activation significantly suppressed apoptosis of HEI-OC1 cells induced by cisplatin. Studies of ectopic expression of pcDNA3-HO-1 and siRNA of HO-1 further revealed the protective role of HO-1 against cisplatin in HEI-OC1 cells. Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Furthermore, pharmacological induction of HO-1 completely prevented the destruction of outer hair cell arrays by cisplatin through a CO-dependent mechanism in organotrophic culture of the rat primary organ of Corti explants. These results suggest that HO-1 may serve as a safeguard of auditory sensory hair cells against a variety of challenges of oxidative stress, including noise trauma, presbycusis, and ototoxic drugs, respectively.
Collapse
Affiliation(s)
- Hyung-Jin Kim
- Vestibulocochlear Research Center (VCRC) and Department of Microbiology, Wonkwang University School of Medicine, 344-2 Shinyong-dong Iksan, Jeonbuk 570-749, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Stuart JR, Kawai H, Tsai KKC, Chuang EY, Yuan ZM. c-Abl regulates early growth response protein (EGR1) in response to oxidative stress. Oncogene 2006; 24:8085-92. [PMID: 16091742 DOI: 10.1038/sj.onc.1208953] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
c-Abl is a tyrosine kinase that can act as a regulator of cell growth and apoptosis in response to stress. Using cell lines expressing c-Abl in an inducible manner, we identified genes whose expression was regulated by c-Abl kinase activity. Microarray analysis indicated that Early Growth Response-1 (EGR1) gene expression is induced by c-Abl kinase activity, which was confirmed at the message and protein levels. Promoter mapping experiments revealed that c-Abl utilizes three distal serum response elements (SREs) in the EGR1 promoter, which are transactivated by mitogen/extracellular receptor kinase (MEK/ERK) signaling. PD 95089, a specific inhibitor of MEK/ERK signaling, attenuated c-Abl-mediated upregulation of EGR1 expression in a dose-dependent manner. Similar results were obtained by using a dominant-negative mutant of mitogen/extracellular kinase. Significantly, hydrogen peroxide-induced EGR1 expression appears to be mediated by c-Abl, as cells expressing dominant negative c-Abl, and c-Abl-/- murine embryonic fibroblasts, are completely defective in hydrogen peroxide-induced EGR1 expression. In addition, c-Abl-induced apoptosis is partially mitigated by EGR1 activity, as cells devoid of EGR1 expression undergo reduced rates of c-Abl-induced apoptosis. Together, these results indicate that c-Abl promotes the induction of EGR1 through the MEK/ERK pathway in regulating apoptotic response to oxidative stress.
Collapse
Affiliation(s)
- Jeremy R Stuart
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
44
|
So HS, Kim HJ, Lee JH, Lee JH, Park SY, Park C, Kim YH, Kim JK, Lee KM, Kim KS, Chung SY, Jang WC, Moon SK, Chung HT, Park RK. Flunarizine induces Nrf2-mediated transcriptional activation of heme oxygenase-1 in protection of auditory cells from cisplatin. Cell Death Differ 2006; 13:1763-75. [PMID: 16485034 DOI: 10.1038/sj.cdd.4401863] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We investigated the cytoprotective mechanisms of flunarizine in cisplatin-induced death of auditory cells. Concomitant with an increase in viability, treatment with flunarizine resulted in a marked dissociation of Nrf2/Keap1 and subsequent intranuclear translocation of Nrf2, which was mediated by PI3K-Akt signaling. Overexpression of Nrf2 protected cells from cisplatin along with transcriptional activation of ARE to generate heme oxygenase-1 (HO-1). Pretreatment with flunarizine predominantly increased the transcriptional activity of HO-1 among Nrf2-driven transcripts, including HO-1, NQO1, GCLC, GCLM, GST micro-1, and GSTA4. Furthermore, both pharmacological inhibition and siRNA transfection of HO-1 completely abolished the flunarizine-mediated protection of HEI-OC1 cells and the primary rat (P2) organ of Corti explants from cisplatin. These results suggest that Nrf2-driven transcriptional activation of ARE through PI3K-Akt signaling augments the generation of HO-1, which may be a critically important determinant in cellular response toward cisplatin and the cytoprotective effect of flunarizine against cisplatin.
Collapse
Affiliation(s)
- H-S So
- Vestibulocochlear Research Center & Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk 570-749, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ise T, Shimizu T, Lee EL, Inoue H, Kohno K, Okada Y. Roles of volume-sensitive Cl- channel in cisplatin-induced apoptosis in human epidermoid cancer cells. J Membr Biol 2006; 205:139-45. [PMID: 16362502 DOI: 10.1007/s00232-005-0779-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Indexed: 10/25/2022]
Abstract
The anti-cancer drug cisplatin induces apoptosis by damaging DNA. Since a stilbene-derivative blocker of Cl-/HCO3- exchangers and Cl- channels, SITS, is known to induce cisplatin resistance in a manner independent of intracellular pH and extracellular HCO3-, we investigated the relation between cisplatin-induced apoptosis and Cl- channel activity in human adenocarcinoma KB cells. A stilbene derivative, DIDS, reduced cisplatin-induced caspase-3 activation and cell death, which were detected over 18 h after treatment with cisplatin. DIDS was also found to reduce sensitivity of KB cells to 5-day exposure to cisplatin. Whole-cell patch-clamp recordings showed that KB cells functionally express volume-sensitive outwardly rectifying (VSOR) Cl- channels which are activated by osmotic cell swelling and sensitive to DIDS. Pretreatment of the cells with cisplatin for 12 h augmented the magnitude of VSOR Cl- current. Thus, it is concluded that cisplatin-induced cytotoxicity in KB cells is associated with augmented activity of a DIDS-sensitive VSOR Cl- channel and that blockade of this channel is, at least in part, responsible for cisplatin resistance induced by a stilbene derivative.
Collapse
Affiliation(s)
- T Ise
- Department of Molecular Biology, University of Occupational and Environmental Health, School of Medicine, Kitakyushu 807-8555, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Chu F, Barkinge J, Hawkins S, Gudi R, Salgia R, Kanteti PVS. Expression of Siva-1 protein or its putative amphipathic helical region enhances cisplatin-induced apoptosis in breast cancer cells: effect of elevated levels of BCL-2. Cancer Res 2005; 65:5301-9. [PMID: 15958577 DOI: 10.1158/0008-5472.can-04-3270] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
cis-Diaminedichloroplatinum (II) (cisplatin) is routinely used to treat various types of cancers; however, a significant number develop resistance. One of the underlying factors that contribute to cisplatin resistance is the elevated level of BCL-2 and/or BCL-XL, which promotes cell survival. A potential method of overcoming such resistance is to use a potentiator that is capable of neutralizing the antiapoptotic effects of BCL-2/BCL-XL, such as Siva-1. We previously cloned the proapoptotic protein Siva-1 and showed a possible role for it in both extrinsic and intrinsic apoptosis. Using an adenovirus-based expression system, we now show that Siva-1 can synergize with cisplatin in inducing apoptosis in MCF7 and MDA-MB-231 breast cancer cells. In an anchorage-independent clonogenicity assay, MCF7/caspase-3 cells stably expressing Siva-1, but not the control cells, showed a dramatic decrease in the number of colonies formed on one-time cisplatin treatment. Further, we show that the unique putative amphipathic helical region (SAH) in Siva-1 (amino acid residues 36-55) is necessary and sufficient for the observed enhancement in cisplatin-induced apoptosis by Siva-1. Although cisplatin treatment results in significant elevation in the expression of Fas ligand and intracellular p21 levels, expression of Siva-1 has no additional benefit. Instead, the enhancement in apoptosis seems to be due to activation of intrinsic pathway that involves caspase-9 activation. Moreover, Siva-1 augments cisplatin-mediated cell death in MCF7 cells stably expressing BCL-2. We therefore propose that Siva-1 or its SAH region can be used as a potentiator of cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Fei Chu
- Department of Microbiology and Immunology, University of Illinois at Chicago, 60612, USA
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Cisplatin, carboplatin and oxaliplatin are platinum-based drugs that are widely used in cancer chemotherapy. Platinum-DNA adducts, which are formed following uptake of the drug into the nucleus of cells, activate several cellular processes that mediate the cytotoxicity of these platinum drugs. This review focuses on recently discovered cellular pathways that are activated in response to cisplatin, including those involved in regulating drug uptake, the signalling of DNA damage, cell-cycle checkpoints and arrest, DNA repair and cell death. Such knowledge of the cellular processing of cisplatin adducts with DNA provides valuable clues for the rational design of more efficient platinum-based drugs as well as the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Dong Wang
- Department of Chemistry, Massachusetts Institute of Technology, Room 18-498, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
48
|
Cao C, Li Y, Leng Y, Li P, Ma Q, Kufe D. Ubiquitination and degradation of the Arg tyrosine kinase is regulated by oxidative stress. Oncogene 2005; 24:2433-40. [PMID: 15735735 DOI: 10.1038/sj.onc.1208454] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The c-Abl and Arg nonreceptor tyrosine kinases are activated in the response of cells to oxidative stress. The present studies demonstrate that treatment of cells with 0.1 mM H2O2 is associated with increased tyrosine phosphorylation of Arg and little effect on Arg levels. By contrast, exposure to 1.0 mM H2O2 decreased Arg phosphorylation. Treatment with 1.0 mM H2O2 was also associated with ubiquitination and degradation of Arg. The results show that Arg is stabilized in response to 0.1 mM H2O2 by autophosphorylation of Y-261, consistent with involvement of the Arg kinase function in regulating Arg levels. The results further demonstrate that c-Abl-mediated phosphorylation of Arg on Y-261 similarly confers Arg stabilization. In concert with these results, phosphorylation of Arg on Y-261 blocked H2O2-induced ubiquitination and thereby Arg degradation and inactivation. These findings demonstrate that Arg phosphorylation and degradation are differentially regulated by the degree of oxidative stress, and that Arg stability is conferred by phosphorylation of Y-261.
Collapse
Affiliation(s)
- Cheng Cao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | | | | | | | | | | |
Collapse
|
49
|
Raina D, Pandey P, Ahmad R, Bharti A, Ren J, Kharbanda S, Weichselbaum R, Kufe D. c-Abl tyrosine kinase regulates caspase-9 autocleavage in the apoptotic response to DNA damage. J Biol Chem 2005; 280:11147-51. [PMID: 15657060 DOI: 10.1074/jbc.m413787200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of the initiator caspase-9 is essential for induction of apoptosis by developmental signals, oncogenic transformation, and genotoxic stress. The c-Abl tyrosine kinase is also involved in the apoptotic response to DNA damage. The present results demonstrate that c-Abl binds directly to caspase-9. We show that c-Abl phosphorylates caspase-9 on Tyr-153 in vitro and in cells treated with DNA damaging agents. Moreover, inhibition of c-Abl with STI571 blocked DNA damage-induced autoprocessing of caspase-9 to the p35 subunit and activation of caspase-3. Caspase-9(Y153F) also attenuated DNA damage-induced processing of caspase-9 to p35, activation of caspase-3, and apoptosis. These findings indicate that caspase-9 autoprocessing is regulated by c-Abl in the apoptotic response to genotoxic stress.
Collapse
Affiliation(s)
- Deepak Raina
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Riechelmann H. Cellular and molecular mechanisms in environmental and occupational inhalation toxicology. GMS CURRENT TOPICS IN OTORHINOLARYNGOLOGY, HEAD AND NECK SURGERY 2004; 3:Doc02. [PMID: 22073044 PMCID: PMC3199796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The central issue of this review are inflammatory changes that take place in the mucous membranes of the respiratory tract as a result of inhaled pollutants. Of particular relevance are dusts, SO(2), ozone, aldehydes und volatile organic compounds. Bioorganic pollutants, especially fragments of bacteria and fungi, occur predominantly in indoor dusts. They activate the toll-like/IL-1 receptor and lead to the activation of the transcription factor NF-κB for the release of numerous proinflammatory cytokines. Metals are predominant in ambient air dust particles. They induce the release of reactive oxygen species that cause damage to lipids, proteins and the DNA of the cell. As well as NF-κB, transcription factors that foster proliferation are activated via stress activated protein kinases. Organic compounds such as polycyclic aromatic hydrocarbons and nitroso-compounds of incomplete combustion processes activate additional via the cytosolic arylhydrocarbon receptor for detoxification enzymes. Sulphur dioxide leads to acid stress, and ozone to oxidative stress of the cell. This is accompanied by the release of proinflammatory cytokines via stress activated protein kinases. Aldehydes and volatile organic compounds activate the vanilloid receptor of trigeminal nerve fibres and induce a hyperreactivity of the mucous membrane via the release of nerve growth factors. The mechanisms described work synergistically and lead to a chronic inflammatory reaction of the mucous membranes of the upper respiratory tract that is regularly demonstrable in inhabitants of western industrial nations. It is unclear whether we are dealing here with a physiological inflammation or with an at least partially avoidable result of chronic pollutant exposure.
Collapse
|