1
|
Che H, Ding H, Jia X. circ_0080145 Enhances Imatinib Resistance of Chronic Myeloid Leukemia by Regulating miR-326/ PPFIA1 Axis. Cancer Biother Radiopharm 2024; 39:478-491. [PMID: 32598170 DOI: 10.1089/cbr.2020.3600] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Acquired multidrug resistance is often blamed for the failure of chemotherapy in patients with malignant tumors, including chronic myeloid leukemia (CML). In this study, the authors investigated the role of circular RNA 0080145 (circ_0080145) in imatinib (IM) resistance of CML. Materials and Methods: Quantitative real-time polymerase chain reaction was applied to measure the expression of circ_0080145, microRNA-326 (miR-326), and PTPRF interacting protein alpha 1 (PPFIA1) mRNA. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay was used to determine the half maximal inhibitory concentration (IC50) of IM and cell proliferation. Flow cytometry analysis was utilized to assess cell apoptosis. The levels of glucose uptake and lactate production were examined using specific kits. Protein levels were detected through Western blot assay. The targeting relationship between miR-326 and circ_0080145 or PPFIA1 was verified by dual-luciferase reporter assay. The murine xenograft model was constructed to investigate the effect of circ_0080145 in vivo. Results: circ_0080145 was upregulated in IM-resistant CML patients and cells. circ_0080145 silencing suppressed IM resistance, cell growth, and glycolysis and induced apoptosis in IM-resistant CML cells in vitro. Moreover, circ_0080145 knockdown blocked tumor growth and IM resistance in vivo. miR-326 was a target of circ_0080145, and miR-326 inhibition restored the effects of circ_0080145 silencing on cell progression and IM resistance. In addition, PPFIA1 was a target gene of miR-326. The suppressive roles in IM resistance, cell growth and glycolysis, and the promotional role in apoptosis mediated by miR-326 were abolished by PPFIA1 overexpression in IM-resistant CML cells. Conclusion: circ_0080145 contributes to IM resistance via modulating miR-326/PPFIA1 axis, which might provide a novel avenue for CML therapy.
Collapse
MESH Headings
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Drug Resistance, Neoplasm/genetics
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Mice
- Animals
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Male
- Female
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Xenograft Model Antitumor Assays
- Mice, Nude
- K562 Cells
- Cell Line, Tumor
Collapse
Affiliation(s)
- Hong Che
- Department of Hematology, Affiliated to Qingdao University Yuhuangding Hospital of Yantai, Shandong, China
| | - Hong Ding
- Department of Hematology, Affiliated to Qingdao University Yuhuangding Hospital of Yantai, Shandong, China
| | - Xizhen Jia
- Department of Operation Room, Affiliated to Qingdao University Yuhuangding Hospital of Yantai, Shandong, China
| |
Collapse
|
2
|
Wei D, Liang X, Huang M, Wang C, Ye Z, Zhang T, Zhang J. Targeting histone deacetylase 1 (HDAC1) in the bone marrow stromal cells revers imatinib resistance by modulating IL-6 in Ph + acute lymphoblastic leukemia. Ann Hematol 2024; 103:3015-3027. [PMID: 38847852 DOI: 10.1007/s00277-024-05830-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/31/2024] [Indexed: 07/28/2024]
Abstract
Bone marrow stromal cells (BMSCs) can promote the growth of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Histone deacetylases (HDACs) play essential roles in the proliferation and apoptosis resistance of Ph + ALL cells. In our previous study, inhibiting histone deacetylase 1 (HDAC1) decreases the proliferation of Ph + ALL cells. However, little is known regarding how HDAC1 in BMSCs of Ph + ALL patients affects the imatinib (IM) resistance. Therefore, the present work examined the roles of HDAC1 in BMSCs. Overexpression of HDAC1 was found in BMSCs of Ph + ALL patients with IM resistance. In addition, the Ph + ALL cell line SUP-B15 was co-cultured with BMSCs after lentivirus transfection for regulating HDAC1 expression. Knockdown of HDAC1 within BMSCs elevated the IM-mediated SUP-B15 cell apoptosis, while increasing HDAC1 expression had an opposite effect. IL-6 in BMSCs, which is an important factor for the microenvironment-associated chemoresistance, showed evident up-regulation in HDAC1-upregulated BMSCs and down-regulation in HDAC1-downregulated BMSCs. While recombinant IL-6 (rIL-6) can reversed the sensitivity of SUP-B15 cells to IM induced by downregulating HDAC1 expression in BMSCs. HDAC1 showed positive regulation on IL-6 transcription and secretion. Moreover, IL-6 secretion induced by HDAC1 in BMSCs might enhance IM resistance in Ph + ALL cells. With regard to the underlying molecular mechanism, NF-κB, an important signal responsible for IL-6 transcription in BMSCs, mediated the HDAC1-regulated IL-6 expression. Collectively, this study facilitated to develop HDAC1 inhibitors based not only the corresponding direct anti-Ph + ALL activity but also the regulation of bone marrow microenvironment.
Collapse
Affiliation(s)
- Danna Wei
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Xiaoling Liang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Meiling Huang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Caili Wang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Zhangmin Ye
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China
| | - Tianzhuo Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Jingrong Zhang
- Department of Pediatric Hematology, Guiyang Maternal and Child Health Care Hospital, Guiyang Children's Hospital, Guiyang, 550002, China.
| |
Collapse
|
3
|
Deng Y, Cheng Q, He J. HDAC inhibitors: Promising agents for leukemia treatment. Biochem Biophys Res Commun 2023; 680:61-72. [PMID: 37722346 DOI: 10.1016/j.bbrc.2023.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 09/20/2023]
Abstract
The essential role of epigenetic modification in the pathogenesis of a series of cancers have gradually been recognized. Histone deacetylase (HDACs), as well-known epigenetic modulators, are responsible for DNA repair, cell proliferation, differentiation, apoptosis and angiogenesis. Studies have shown that aberrant expression of HDACs is found in many cancer types. Thus, inhibition of HDACs has provided a promising therapeutic approach alternative for these patients. Since HDAC inhibitor (HDACi) vorinostat was first approved by the Food and Drug Administration (FDA) for treating cutaneous T-cell lymphoma (CTCL) in 2006, the combination of HDAC inhibitors with other molecules such as chemotherapeutic drugs has drawn much attention in current cancer treatment, especially in hematological malignancies therapy. Up to now, there have been more than twenty HDAC inhibitors investigated in clinic trials with five approvals being achieved. Indeed, Histone deacetylase inhibitors promote or enhance several different anticancer mechanisms and therefore are in evidence as potential antileukemia agents. In this review, we will focus on possible mechanisms by how HDAC inhibitors exert therapeutic benefit and their clinical utility in leukemia.
Collapse
Affiliation(s)
- Yun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Cheng
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jing He
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Fajardo-Orduña GR, Ledesma-Martínez E, Aguiñiga-Sanchez I, Weiss-Steider B, Santiago-Osorio E. Role of SIRT1 in Chemoresistant Leukemia. Int J Mol Sci 2023; 24:14470. [PMID: 37833921 PMCID: PMC10573076 DOI: 10.3390/ijms241914470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Leukemias of the AML, CML, and CLL types are the most common blood cancers worldwide, making them a major global public health problem. Furthermore, less than 24% of patients treated with conventional chemotherapy (low-risk patients) and 10-15% of patients ineligible for conventional chemotherapy (high-risk patients) survive five years. The low levels of survival are mainly due to toxicity and resistance to chemotherapy or other medication, the latter leading to relapse of the disease, which is the main obstacle to the treatment of leukemia. Drug resistance may include different molecular mechanisms, among which epigenetic regulators are involved. Silent information regulator 2 homolog 1 (SIRT1) is an epigenetic factor belonging to the sirtuin (SIRT) family known to regulate aspects of chromatin biology, genome stability, and metabolism, both in homeostasis processes and in different diseases, including cancer. The regulatory functions of SIRT1 in different biological processes and molecular pathways are dependent on the type and stage of the neoplasia; thus, it may act as both an oncogenic and tumor suppressor factor and may also participate in drug resistance. In this review, we explore the role of SIRT1 in drug-resistant leukemia and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Guadalupe Rosario Fajardo-Orduña
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, Faculty of High Studies Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.R.F.-O.)
| | - Edgar Ledesma-Martínez
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, Faculty of High Studies Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.R.F.-O.)
| | - Itzen Aguiñiga-Sanchez
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, Faculty of High Studies Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.R.F.-O.)
- Department of Biomedical Sciences, School of Medicine, Faculty of High Studies Zaragoza, National Autonomous University of Mexico, Mexico City 56410, Mexico
| | - Benny Weiss-Steider
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, Faculty of High Studies Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.R.F.-O.)
| | - Edelmiro Santiago-Osorio
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, Faculty of High Studies Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.R.F.-O.)
| |
Collapse
|
5
|
Karagiannis TC, Wall M, Ververis K, Pitsillou E, Tortorella SM, Wood PA, Rafehi H, Khurana I, Maxwell SS, Hung A, Vongsvivut J, El-Osta A. Characterization of K562 cells: uncovering novel chromosomes, assessing transferrin receptor expression, and probing pharmacological therapies. Cell Mol Life Sci 2023; 80:248. [PMID: 37578596 PMCID: PMC11072675 DOI: 10.1007/s00018-023-04905-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
Human erythroleukemic K562 cells represent the prototypical cell culture model of chronic myeloid leukemia (CML). The cells are pseudo-triploid and positive for the Philadelphia chromosome. Therefore, K562 cells have been widely used for investigating the BCR/ABL1 oncogene and the tyrosine kinase inhibitor, imatinib-mesylate. Further, K562 cells overexpress transferrin receptors (TfR) and have been used as a model for targeting cytotoxic therapies, via receptor-mediated endocytosis. Here, we have characterized K562 cells focusing on the karyotype of cells in prolonged culture, regulation of expression of TfR in wildtype (WT) and doxorubicin-resistant cells, and responses to histone deacetylase inhibition (HDACi). Karyotype analysis indicates novel chromosomes and gene expression analysis suggests a shift of cultured K562 cells away from patient-derived leukemic cells. We confirm the high expression of TfR on K562 cells using immunofluorescence and cell-surface receptor binding radioassays. Importantly, high TfR expression is observed in patient-derived cells, and we highlight the persistent expression of TfR following doxorubicin acquired resistance. Epigenetic analysis indicates that permissive histone acetylation and methylation at the promoter region regulates the transcription of TfR in K562 cells. Finally, we show relatively high expression of HDAC enzymes in K562 cells and demonstrate the chemotoxic effects of HDACi, using the FDA-approved hydroxamic acid, vorinostat. Together with a description of morphology, infrared spectral analysis, and examination of metabolic properties, we provide a comprehensive characterization of K562 cells. Overall, K562 cell culture systems remain widely used for the investigation of novel therapeutics for CML, which is particularly important in cases of imatinib-mesylate resistance.
Collapse
MESH Headings
- Humans
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- K562 Cells
- Fusion Proteins, bcr-abl/genetics
- Transferrin
- Pyrimidines/pharmacology
- Drug Resistance, Neoplasm/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Histone Deacetylases/metabolism
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Receptors, Transferrin/genetics
- Chromosomes/metabolism
- Mesylates/pharmacology
- Apoptosis
Collapse
Affiliation(s)
- Tom C Karagiannis
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia.
- Epigenomic Medicine Laboratory at prospED Training, Carlton, VIC, 3053, Australia.
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Epigenomic in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia.
| | - Meaghan Wall
- Victorian Cancer Cytogenetics Service, St Vincent's Hospital, Fitzroy, VIC, 3065, Australia
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Katherine Ververis
- Epigenomic Medicine Laboratory at prospED Training, Carlton, VIC, 3053, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Eleni Pitsillou
- Epigenomic Medicine Laboratory at prospED Training, Carlton, VIC, 3053, Australia
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Stephanie M Tortorella
- Epigenomic Medicine Laboratory at prospED Training, Carlton, VIC, 3053, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter A Wood
- Epigenomic Medicine Laboratory at prospED Training, Carlton, VIC, 3053, Australia
| | - Haloom Rafehi
- Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Ishant Khurana
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia
| | - Scott S Maxwell
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | | | - Assam El-Osta
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC, 3004, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30‑32 Ngan Shing Street, Sha Tin, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Biomedical Laboratory Science, Department of Technology, Faculty of Health, University College Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Chang YJ, Lin S, Kang ZF, Shen BJ, Tsai WH, Chen WC, Lu HP, Su YL, Chou SJ, Lin SY, Lin SW, Huang YJ, Wang HH, Chang CJ. Acetylation-Mimic Mutation of TRIM28-Lys304 to Gln Attenuates the Interaction with KRAB-Zinc-Finger Proteins and Affects Gene Expression in Leukemic K562 Cells. Int J Mol Sci 2023; 24:9830. [PMID: 37372979 DOI: 10.3390/ijms24129830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
TRIM28/KAP1/TIF1β is a crucial epigenetic modifier. Genetic ablation of trim28 is embryonic lethal, although RNAi-mediated knockdown in somatic cells yields viable cells. Reduction in TRIM28 abundance at the cellular or organismal level results in polyphenism. Posttranslational modifications such as phosphorylation and sumoylation have been shown to regulate TRIM28 activity. Moreover, several lysine residues of TRIM28 are subject to acetylation, but how acetylation of TRIM28 affects its functions remains poorly understood. Here, we report that, compared with wild-type TRIM28, the acetylation-mimic mutant TRIM28-K304Q has an altered interaction with Krüppel-associated box zinc-finger proteins (KRAB-ZNFs). The TRIM28-K304Q knock-in cells were created in K562 erythroleukemia cells by CRISPR-Cas9 (Clustered regularly interspaced short palindromic repeats/CRISPR-associated protein nuclease 9) gene editing method. Transcriptome analysis revealed that TRIM28-K304Q and TRIM28 knockout K562 cells had similar global gene expression profiles, yet the profiles differed considerably from wild-type K562 cells. The expression levels of embryonic-related globin gene and a platelet cell marker integrin-beta 3 were increased in TRIM28-K304Q mutant cells, indicating the induction of differentiation. In addition to the differentiation-related genes, many zinc-finger-proteins genes and imprinting genes were activated in TRIM28-K304Q cells; they were inhibited by wild-type TRIM28 via binding with KRAB-ZNFs. These results suggest that acetylation/deacetylation of K304 in TRIM28 constitutes a switch for regulating its interaction with KRAB-ZNFs and alters the gene regulation as demonstrated by the acetylation mimic TRIM28-K304Q.
Collapse
Affiliation(s)
- Yao-Jen Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Steven Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Zhi-Fu Kang
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Bin-Jon Shen
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Wen-Hai Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Wen-Ching Chen
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Hsin-Pin Lu
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Yu-Lun Su
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Shu-Jen Chou
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Shu-Yu Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Sheng-Wei Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Yin-Jung Huang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Hsin-Hui Wang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Pediatrics, Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ching-Jin Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
7
|
Zhu Y, Wang Z, Li Y, Peng H, Liu J, Zhang J, Xiao X. The Role of CREBBP/EP300 and Its Therapeutic Implications in Hematological Malignancies. Cancers (Basel) 2023; 15:cancers15041219. [PMID: 36831561 PMCID: PMC9953837 DOI: 10.3390/cancers15041219] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Disordered histone acetylation has emerged as a key mechanism in promoting hematological malignancies. CREB-binding protein (CREBBP) and E1A-binding protein P300 (EP300) are two key acetyltransferases and transcriptional cofactors that regulate gene expression by regulating the acetylation levels of histone proteins and non-histone proteins. CREBBP/EP300 dysregulation and CREBBP/EP300-containing complexes are critical for the initiation, progression, and chemoresistance of hematological malignancies. CREBBP/EP300 also participate in tumor immune responses by regulating the differentiation and function of multiple immune cells. Currently, CREBBP/EP300 are attractive targets for drug development and are increasingly used as favorable tools in preclinical studies of hematological malignancies. In this review, we summarize the role of CREBBP/EP300 in normal hematopoiesis and highlight the pathogenic mechanisms of CREBBP/EP300 in hematological malignancies. Moreover, the research basis and potential future therapeutic implications of related inhibitors were also discussed from several aspects. This review represents an in-depth insight into the physiological and pathological significance of CREBBP/EP300 in hematology.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Zi Wang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Yanan Li
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Jing Liu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| | - Ji Zhang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang 421001, China
- Correspondence: (J.Z.); (X.X.); Tel.: +86-734-8279050 (J.Z.); +86-731-84805449 (X.X.)
| | - Xiaojuan Xiao
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
- Correspondence: (J.Z.); (X.X.); Tel.: +86-734-8279050 (J.Z.); +86-731-84805449 (X.X.)
| |
Collapse
|
8
|
Alvespimycin Inhibits Heat Shock Protein 90 and Overcomes Imatinib Resistance in Chronic Myeloid Leukemia Cell Lines. Molecules 2023; 28:molecules28031210. [PMID: 36770876 PMCID: PMC9920317 DOI: 10.3390/molecules28031210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Heat shock protein 90 (HSP90) facilitates folding and stability and prevents the degradation of multiple client proteins. One of these HSP90 clients is BCR-ABL, the oncoprotein characteristic of chronic myeloid leukemia (CML) and the target of tyrosine kinase inhibitors, such as imatinib. Alvespimycin is an HSP90 inhibitor with better pharmacokinetic properties and fewer side effects than other similar drugs, but its role in overcoming imatinib resistance is not yet clarified. This work studied the therapeutic potential of alvespimycin in imatinib-sensitive (K562) and imatinib-resistant (K562-RC and K562-RD) CML cell lines. Metabolic activity was determined by the resazurin assay. Cell death, caspase activity, mitochondrial membrane potential, and cell cycle were evaluated by means of flow cytometry. Cell death was also analyzed by optical microscopy. HSPs expression levels were assessed by western blotting. Alvespimycin reduced metabolic activity in a time-, dose-, and cell line-dependent manner. Resistant cells were more sensitive to alvespimycin with an IC50 of 31 nM for K562-RC and 44 nM for K562-RD, compared to 50 nM for K562. This drug induced apoptosis via the mitochondrial pathway. In K562 cells, alvespimycin induced cell cycle arrest in G0/G1. As a marker of HSP90 inhibition, a significant increase in HSP70 expression was observed. Our results suggest that alvespimycin might be a new therapeutic approach to CML treatment, even in cases of resistance to imatinib.
Collapse
|
9
|
Liu P, Xiao J, Wang Y, Song X, Huang L, Ren Z, Kitazato K, Wang Y. Posttranslational modification and beyond: interplay between histone deacetylase 6 and heat-shock protein 90. Mol Med 2021; 27:110. [PMID: 34530730 PMCID: PMC8444394 DOI: 10.1186/s10020-021-00375-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/07/2021] [Indexed: 12/17/2022] Open
Abstract
Posttranslational modification (PTM) and regulation of protein stability are crucial to various biological processes. Histone deacetylase 6 (HDAC6), a unique histone deacetylase with two functional catalytic domains (DD1 and DD2) and a ZnF-UBP domain (ubiquitin binding domain, BUZ), regulates a number of biological processes, including gene expression, cell motility, immune response, and the degradation of misfolded proteins. In addition to the deacetylation of histones, other nonhistone proteins have been identified as substrates for HDAC6. Hsp90, a molecular chaperone that is a critical modulator of cell signaling, is one of the lysine deacetylase substrates of HDAC6. Intriguingly, as one of the best-characterized regulators of Hsp90 acetylation, HDAC6 is the client protein of Hsp90. In addition to regulating Hsp90 at the post-translational modification level, HDAC6 also regulates Hsp90 at the gene transcription level. HDAC6 mainly regulates the Hsp90-HSF1 complex through the ZnF-UBP domain, thereby promoting the HSF1 entry into the nucleus and activating gene transcription. The mutual interaction between HDAC6 and Hsp90 plays an important role in the regulation of protein stability, cell migration, apoptosis and other functions. Plenty of of studies have indicated that blocking HDAC6/Hsp90 has a vital regulatory role in multifarious diseases, mainly in cancers. Therefore, developing inhibitors or drugs against HDAC6/Hsp90 becomes a promising development direction. Herein, we review the current knowledge on molecular regulatory mechanisms based on the interaction of HDAC6 and Hsp90 and inhibition of HDAC6 and/or Hsp90 in oncogenesis and progression, antiviral and immune-related diseases and other vital biological processes.
Collapse
Affiliation(s)
- Ping Liu
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Ji Xiao
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Yiliang Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Xiaowei Song
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Lianzhou Huang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhe Ren
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Kaio Kitazato
- Department of Clinical Research Pharmacy, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| | - Yifei Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.
| |
Collapse
|
10
|
Lu T, Wei D, Yu K, Ma D, Xiong J, Fang Q, Wang J. Betulinic acid restores imatinib sensitivity in BCR-ABL1 kinase-independent, imatinib-resistant chronic myeloid leukemia by increasing HDAC3 ubiquitination and degradation. Ann N Y Acad Sci 2020; 1467:77-93. [PMID: 31930541 DOI: 10.1111/nyas.14298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/28/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022]
Abstract
Although imatinib (IM) has been demonstrated to be an efficient treatment in chronic myeloid leukemia (CML), some patients still experience IM resistance and disease relapse. Through in vitro studies, we observed that HDAC3 levels were elevated in BCR-ABL1 kinase-independent, IM-resistant primary cells from CML patients and in IM-resistant K562 (K562R) cells and that downregulation of HDAC3 could enhance IM efficacy in K562R cells. Furthermore, betulinic acid (BA), a lupane-type pentacyclic triterpenoid saponin isolated from birch trees, restored IM sensitivity in the BCR-ABL1 kinase-independent, IM-resistant primary cells and in K562R cells, as well as in primary CD34+ bone marrow cells from CML patients. We found that BA restored IM sensitivity through inhibition of HDAC3 accumulation in cells, and that this was mediated by BA-dependent ubiquitination and degradation of HDAC3. BA at low dosage significantly increased IM antitumor effects on murine xenografts bearing K562R cells and inhibited HDAC3 expression in tumor tissue. Our findings demonstrated that HDAC3 is an essential factor in BCR-ABL1 kinase-independent IM resistance, and that BA in combination with IM may be a novel treatment strategy for overcoming IM resistance in CML.
Collapse
Affiliation(s)
- Tingting Lu
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Department of Clinical Laboratory Centre, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China
| | - Danna Wei
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Kunlin Yu
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China.,Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Dan Ma
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Jie Xiong
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Jishi Wang
- Department of Haematology, Affiliated Hospital of Guizhou Medical University, Guizhou, China.,Hematological Institute of Guizhou Province, Guizhou, China.,Guizhou Province Hematopoietic Stem Cell Transplantation Centre and Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou, China
| |
Collapse
|
11
|
Epigenetic mechanisms underlying the therapeutic effects of HDAC inhibitors in chronic myeloid leukemia. Biochem Pharmacol 2019; 173:113698. [PMID: 31706847 DOI: 10.1016/j.bcp.2019.113698] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022]
Abstract
Chronic myeloid leukemia (CML) is a hematological disorder caused by the oncogenic BCR-ABL fusion protein in more than 90% of patients. Despite the striking improvements in the management of CML patients since the introduction of tyrosine kinase inhibitors (TKis), the appearance of TKi resistance and side effects lead to treatment failure, justifying the need of novel therapeutic approaches. Histone deacetylase inhibitors (HDACis), able to modulate gene expression patterns and important cellular signaling pathways through the regulation of the acetylation status of both histone and non-histone protein targets, have been reported to display promising anti-leukemic properties alone or in combination with TKis. This review summarizes pre-clinical and clinical studies that investigated the mechanisms underlying the anticancer potential of HDACis and discusses the rationale for a combination of HDACis with TKis as a therapeutic option in CML.
Collapse
|
12
|
Synergistic activity of imatinib and AR-42 against chronic myeloid leukemia cells mainly through HDAC1 inhibition. Life Sci 2018; 211:224-237. [DOI: 10.1016/j.lfs.2018.09.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/18/2018] [Accepted: 09/21/2018] [Indexed: 02/01/2023]
|
13
|
Wei D, Lu T, Ma D, Yu K, Li X, Chen B, Xiong J, Zhang T, Wang J. Heme oxygenase-1 reduces the sensitivity to imatinib through nonselective activation of histone deacetylases in chronic myeloid leukemia. J Cell Physiol 2018; 234:5252-5263. [PMID: 30256411 DOI: 10.1002/jcp.27334] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022]
Abstract
Resistance towards imatinib (IM) remains troublesome in treating many chronic myeloid leukemia (CML) patients. Heme oxygenase-1 (HO-1) is a key enzyme of antioxidative metabolism in association with cell resistance to apoptosis. Our previous studies have shown that overexpression of HO-1 resulted in resistance development to IM in CML cells, while the mechanism remains unclear. In the current study, the IM-resistant CML cells K562R indicated upregulation of some of the histone deacetylases (HDACs) compared with K562 cells. Therefore, we herein postulated HO-1 was associated with HDACs. Silencing HO-1 expression in K562R cells inhibited the expression of some HDACs, and the sensitivity to IM was increased. K562 cells transfected with HO-1 resisted IM and underwent obvious some HDACs. These findings related to the inhibitory effects of high HO-1 expression on the reactive oxygen species (ROS) signaling pathway that negatively regulated HDACs. Increased expression of HO-1 activated HDACs by inhibiting ROS production. In summary, HO-1, which is involved in the development of drug resistance in CML cells by regulating the expression of HDACs, is probably a novel target for improving CML therapy.
Collapse
MESH Headings
- Adult
- Antineoplastic Agents/pharmacology
- Drug Resistance, Neoplasm
- Enzyme Activation
- Female
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Heme Oxygenase-1/metabolism
- Histone Deacetylases/genetics
- Histone Deacetylases/metabolism
- Humans
- Imatinib Mesylate/pharmacology
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Middle Aged
- Protein Kinase Inhibitors/pharmacology
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Young Adult
Collapse
Affiliation(s)
- Danna Wei
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, PR, China
| | - Tingting Lu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, PR, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
| | - Kunlin Yu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, PR, China
| | - Xinyao Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, PR, China
| | - Bingqing Chen
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
| | - Ji Xiong
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
| | - Tianzhuo Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, PR, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, PR, China
- Department of Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Guiyang, PR, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, PR, China
| |
Collapse
|
14
|
Overexpression of miR-202 resensitizes imatinib resistant chronic myeloid leukemia cells through targetting Hexokinase 2. Biosci Rep 2018; 38:BSR20171383. [PMID: 29559564 PMCID: PMC5938424 DOI: 10.1042/bsr20171383] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 03/06/2018] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease which uniquely expresses a constitutively active tyrosine kinase, BCR/ABL. As a specific inhibitor of the BCR-ABL tyrosine kinase, imatinib becomes the first choice for the treatment of CML due to its high efficacy and low toxicity. However, the development of imatinib resistance limits the long-term treatment benefits of it in CML patients. In the present study, we aimed to investigate the roles of miR-202 in the regulation of imatinib sensitivity in CML cell lines and the possible mechanisms involved in this process. We found miR-202 was down-regulated in seven CML cell lines by quantitative reverse-transcription PCR (qRT-PCR) analysis. Overexpression of miR-202 significantly suppressed proliferation rates of CML cells. By establishing imatinib resistant cell lines originating from K562 and KU812 cells, we observed expressions of miR-202 were down-regulated by imatinib treatments and imatinib resistant CML cell lines exhibited lower level of miR-202. On the contrary, imatinib resistant CML cell lines displayed up-regulated glycolysis rate than sensitive cells with the evidence that glucose uptake, lactate production, and key glycolysis enzymes were elevated in imatinib resistant cells. Importantly, the imatinib resistant CML cell lines were more sensitive to glucose starvation and glycolysis inhibitors. In addition, we identified Hexokinase 2 (HK2) as a direct target of miR-202 in CML cell lines. Overexpression of miR-202 sensitized imatinib resistant CML through the miR-202-mediated glycolysis inhibition by targetting HK2. Finally, we provided the clinical relevance that miR-202 was down-regulated in CML patients and patients with lower miR-202 expression displayed higher HK2 expression. The present study will provide new aspects on the miRNA-modulated tyrosine kinase inhibitor (TKI) sensitivity in CML, contributing to the development of new therapeutic anticancer drugs.
Collapse
|
15
|
Yagi K, Shimada A, Sendo T. Pharmacological inhibition of JAK3 enhances the antitumor activity of imatinib in human chronic myeloid leukemia. Eur J Pharmacol 2018; 825:28-33. [DOI: 10.1016/j.ejphar.2018.02.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/11/2018] [Accepted: 02/14/2018] [Indexed: 12/25/2022]
|
16
|
The HDAC inhibitor SB939 overcomes resistance to BCR-ABL kinase Inhibitors conferred by the BIM deletion polymorphism in chronic myeloid leukemia. PLoS One 2017; 12:e0174107. [PMID: 28301600 PMCID: PMC5354438 DOI: 10.1371/journal.pone.0174107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/03/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic myeloid leukemia (CML) treatment has been improved by tyrosine kinase inhibitors (TKIs) such as imatinib mesylate (IM) but various factors can cause TKI resistance in patients with CML. One factor which contributes to TKI resistance is a germline intronic deletion polymorphism in the BCL2-like 11 (BIM) gene which impairs the expression of pro-apoptotic splice isoforms of BIM. SB939 (pracinostat) is a hydroxamic acid based HDAC inhibitor with favorable pharmacokinetic, physicochemical and pharmaceutical properties, and we investigated if this drug could overcome BIM deletion polymorphism-induced TKI resistance. We found that SB939 corrects BIM pre-mRNA splicing in CML cells with the BIM deletion polymorphism, and induces apoptotic cell death in CML cell lines and primary cells with the BIM deletion polymorphism. More importantly, SB939 both decreases the viability of CML cell lines and primary CML progenitors with the BIM deletion and restores TKI-sensitivity. Our results demonstrate that SB939 overcomes BIM deletion polymorphism-induced TKI resistance, and suggest that SB939 may be useful in treating CML patients with BIM deletion-associated TKI resistance.
Collapse
|
17
|
Arya D, Sachithanandan SP, Ross C, Palakodeti D, Li S, Krishna S. MiRNA182 regulates percentage of myeloid and erythroid cells in chronic myeloid leukemia. Cell Death Dis 2017; 8:e2547. [PMID: 28079885 PMCID: PMC5386378 DOI: 10.1038/cddis.2016.471] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022]
Abstract
The deregulation of lineage control programs is often associated with the progression of haematological malignancies. The molecular regulators of lineage choices in the context of tyrosine kinase inhibitor (TKI) resistance remain poorly understood in chronic myeloid leukemia (CML). To find a potential molecular regulator contributing to lineage distribution and TKI resistance, we undertook an RNA-sequencing approach for identifying microRNAs (miRNAs). Following an unbiased screen, elevated miRNA182-5p levels were detected in Bcr-Abl-inhibited K562 cells (CML blast crisis cell line) and in a panel of CML patients. Earlier, miRNA182-5p upregulation was reported in several solid tumours and haematological malignancies. We undertook a strategy involving transient modulation and CRISPR/Cas9 (clustered regularly interspersed short palindromic repeats)-mediated knockout of the MIR182 locus in CML cells. The lineage contribution was assessed by methylcellulose colony formation assay. The transient modulation of miRNA182-5p revealed a biased phenotype. Strikingly, Δ182 cells (homozygous deletion of MIR182 locus) produced a marked shift in lineage distribution. The phenotype was rescued by ectopic expression of miRNA182-5p in Δ182 cells. A bioinformatic analysis and Hes1 modulation data suggested that Hes1 could be a putative target of miRNA182-5p. A reciprocal relationship between miRNA182-5p and Hes1 was seen in the context of TK inhibition. In conclusion, we reveal a key role for miRNA182-5p in restricting the myeloid development of leukemic cells. We propose that the Δ182 cell line will be valuable in designing experiments for next-generation pharmacological interventions.
Collapse
Affiliation(s)
- Deepak Arya
- Cellular Organization and Signalling Group, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- Manipal University, Manipal, India
| | - Sasikala P Sachithanandan
- Cellular Organization and Signalling Group, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Cecil Ross
- Department of Medicine, St Johns Medical College and Hospitals, Bangalore, India
| | - Dasaradhi Palakodeti
- Stem Cells and Regeneration Group, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Shang Li
- Duke-NUS Graduate Medical School, Singapore
| | - Sudhir Krishna
- Cellular Organization and Signalling Group, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
18
|
Sunami Y, Araki M, Kan S, Ito A, Hironaka Y, Imai M, Morishita S, Ohsaka A, Komatsu N. Histone Acetyltransferase p300/CREB-binding Protein-associated Factor (PCAF) Is Required for All- trans-retinoic Acid-induced Granulocytic Differentiation in Leukemia Cells. J Biol Chem 2017; 292:2815-2829. [PMID: 28053092 DOI: 10.1074/jbc.m116.745398] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/30/2016] [Indexed: 01/01/2023] Open
Abstract
Differentiation therapy with all-trans-retinoic acid (ATRA) improves the treatment outcome of acute promyelocytic leukemia (APL); however, the molecular mechanism by which ATRA induces granulocytic differentiation remains unclear. We previously reported that the inhibition of the NAD-dependent histone deacetylase (HDAC) SIRT2 induces granulocytic differentiation in leukemia cells, suggesting the involvement of protein acetylation in ATRA-induced leukemia cell differentiation. Herein, we show that p300/CREB-binding protein-associated factor (PCAF), a histone acetyltransferase (HAT), is a prerequisite for ATRA-induced granulocytic differentiation in leukemia cells. We found that PCAF expression was markedly increased in leukemia cell lines (NB4 and HL-60) and primary APL cells during ATRA-induced granulocytic differentiation. Consistent with these results, the expression of PCAF was markedly up-regulated in the bone marrow cells of APL patients who received ATRA-containing chemotherapy. The knockdown of PCAF inhibited ATRA-induced granulocytic differentiation in leukemia cell lines and primary APL cells. Conversely, the overexpression of PCAF induced the expression of the granulocytic differentiation marker CD11b at the mRNA level. Acetylome analysis identified the acetylated proteins after ATRA treatment, and we found that histone H3, a known PCAF acetylation substrate, was preferentially acetylated by the ATRA treatment. Furthermore, we have demonstrated that PCAF is required for the acetylation of histone H3 on the promoter of ATRA target genes, such as CCL2 and FGR, and for the expression of these genes in ATRA-treated leukemia cells. These results strongly support our hypothesis that PCAF is induced and activated by ATRA, and the subsequent acetylation of PCAF substrates promotes granulocytic differentiation in leukemia cells. Targeting PCAF and its downstream acetylation targets could serve as a novel therapeutic strategy to overcome all subtypes of AML.
Collapse
Affiliation(s)
| | - Marito Araki
- Department of Transfusion Medicine and Stem Cell Regulation, and
| | - Shin Kan
- From the Department of Hematology.,Leading Center for the Development and Research of Cancer Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan and
| | - Akihiro Ito
- the Chemical Genetics Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | | - Misa Imai
- Leading Center for the Development and Research of Cancer Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan and
| | - Soji Morishita
- Department of Transfusion Medicine and Stem Cell Regulation, and
| | - Akimichi Ohsaka
- Department of Transfusion Medicine and Stem Cell Regulation, and
| | | |
Collapse
|
19
|
A NOX2/Egr-1/Fyn pathway delineates new targets for TKI-resistant malignancies. Oncotarget 2016; 6:23631-46. [PMID: 26136341 PMCID: PMC4695141 DOI: 10.18632/oncotarget.4604] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/12/2015] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase inhibitors (TKI) have improved CML response rates, and some are effective against resistance-promoting point mutations in BCR-ABL1. However, in the absence of point mutations, resistance still occurs. Here, we identify a novel pathway mediating resistance which connects p47phox, the organizer subunit of NADPH oxidase-2 (NOX2), with early growth response-1 (Egr-1) and the Src family kinase Fyn. We found up-regulation of p47phox, Egr-1, and Fyn mRNA and protein using paired isogenic CML cell lines and mined data. Isolation of CD34+ cells and tissue microarray staining from blast crisis CML patients confirmed in vivo over-expression of components of this pathway. Knockdown studies revealed that p47phox modulated reactive oxygen species and Egr-1 expression, which, in turn, controlled Fyn expression. Interestingly, Fyn knockdown sensitized TKI-resistant cells to dasatinib, a dual BCR-ABL1/Src inhibitor. Egr-1 knockdown had similar effects, indicating the utility of targeting Fyn expression over activation. Pointedly, p47phox knockdown also restored TKI-sensitivity, indicating that targeting the NOX2 complex can overcome resistance. The NOX2/Egr-1/Fyn pathway was also conserved within TKI-resistant EGFRΔIII-expressing glioblastoma and patient-derived glioblastoma stem cells. Thus, our findings suggest that targeting the NOX2/Egr-1/Fyn pathway may have clinical implications within multiple cancer types; particularly where efficacy of TKI is compromised.
Collapse
|
20
|
Distinct Dasatinib-Induced Mechanisms of Apoptotic Response and Exosome Release in Imatinib-Resistant Human Chronic Myeloid Leukemia Cells. Int J Mol Sci 2016; 17:531. [PMID: 27070592 PMCID: PMC4848987 DOI: 10.3390/ijms17040531] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/25/2016] [Accepted: 04/01/2016] [Indexed: 12/27/2022] Open
Abstract
Although dasatinib is effective in most imatinib mesylate (IMT)-resistant chronic myeloid leukemia (CML) patients, the underlying mechanism of its effectiveness in eliminating imatinib-resistant cells is only partially understood. This study investigated the effects of dasatinib on signaling mechanisms driving-resistance in imatinib-resistant CML cell line K562 (K562RIMT). Compared with K562 control cells, exsomal release, the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/ mammalian target of rapamycin (mTOR) signaling and autophagic activity were increased significantly in K562RIMT cells and mTOR-independent beclin-1/Vps34 signaling was shown to be involved in exosomal release in these cells. We found that Notch1 activation-mediated reduction of phosphatase and tensin homolog (PTEN) was responsible for the increased Akt/mTOR activities in K562RIMT cells and treatment with Notch1 γ-secretase inhibitor prevented activation of Akt/mTOR. In addition, suppression of mTOR activity by rapamycin decreased the level of activity of p70S6K, induced upregulation of p53 and caspase 3, and led to increase of apoptosis in K562RIMT cells. Inhibition of autophagy by spautin-1 or beclin-1 knockdown decreased exosomal release, but did not affect apoptosis in K562RIMT cells. In summary, in K562RIMT cells dasatinib promoted apoptosis through downregulation of Akt/mTOR activities, while preventing exosomal release and inhibiting autophagy by downregulating expression of beclin-1 and Vps34. Our findings reveal distinct dasatinib-induced mechanisms of apoptotic response and exosomal release in imatinib-resistant CML cells.
Collapse
|
21
|
Lee CR, Kang JA, Kim HE, Choi Y, Yang T, Park SG. Secretion of IL-1β from imatinib-resistant chronic myeloid leukemia cells contributes toBCR-ABLmutation-independent imatinib resistance. FEBS Lett 2016; 590:358-68. [DOI: 10.1002/1873-3468.12057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/18/2015] [Accepted: 01/03/2016] [Indexed: 01/29/2023]
Affiliation(s)
- Cho-Rong Lee
- School of Life Sciences; Gwangju Institute of Science and Technology; Korea
| | - Jung-Ah Kang
- School of Life Sciences; Gwangju Institute of Science and Technology; Korea
| | - Hye-Eun Kim
- School of Life Sciences; Gwangju Institute of Science and Technology; Korea
| | - Yegyun Choi
- School of Life Sciences; Gwangju Institute of Science and Technology; Korea
| | - Taewoo Yang
- School of Life Sciences; Gwangju Institute of Science and Technology; Korea
| | - Sung-Gyoo Park
- School of Life Sciences; Gwangju Institute of Science and Technology; Korea
| |
Collapse
|
22
|
Doxorubicin Differentially Induces Apoptosis, Expression of Mitochondrial Apoptosis-Related Genes, and Mitochondrial Potential in BCR-ABL1-Expressing Cells Sensitive and Resistant to Imatinib. BIOMED RESEARCH INTERNATIONAL 2015; 2015:673512. [PMID: 26618175 PMCID: PMC4649080 DOI: 10.1155/2015/673512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/28/2015] [Indexed: 02/01/2023]
Abstract
Imatinib resistance is an emerging problem in the therapy of chronic myeloid leukemia (CML). Because imatinib induces apoptosis, which may be coupled with mitochondria and DNA damage is a prototype apoptosis-inducing factor, we hypothesized that imatinib-sensitive and -resistant CML cells might differentially express apoptosis-related mitochondrially encoded genes in response to genotoxic stress. We investigated the effect of doxorubicin (DOX), a DNA-damaging anticancer drug, on apoptosis and the expression of the mitochondrial NADH dehydrogenase 3 (MT-ND3) and cytochrome b (MT-CYB) in model CML cells showing imatinib resistance caused by Y253H mutation in the BCR-ABL1 gene (253) or culturing imatinib-sensitive (S) cells in increasing concentrations of imatinib (AR). The imatinib-resistant 253 cells displayed higher sensitivity to apoptosis induced by 1 μM DOX and this was confirmed by an increased activity of executioner caspases 3 and 7 in those cells. Native mitochondrial potential was lower in imatinib-resistant cells than in their sensitive counterparts and DOX lowered it. MT-CYB mRNA expression in 253 cells was lower than that in S cells and 0.1 μM DOX kept this relationship. In conclusion, imatinib resistance may be associated with altered mitochondrial response to genotoxic stress, which may be further exploited in CML therapy in patients with imatinib resistance.
Collapse
|
23
|
Haura EB, Beg AA, Rix U, Antonia S. Charting Immune Signaling Proteomes En Route to New Therapeutic Strategies. Cancer Immunol Res 2015; 3:714-20. [PMID: 26081226 DOI: 10.1158/2326-6066.cir-15-0094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/01/2015] [Indexed: 01/12/2023]
Abstract
The activation state of an antitumor effector T cell in a tumor depends on the sum of all stimulatory signals and inhibitory signals that it receives in the tumor microenvironment. Accumulating data address the increasing complexity of these signals produced by a myriad of immune checkpoint molecules, cytokines, and metabolites. While reductionist experiments have identified key molecules and their importance in signaling, less clear is the integration of all these signals that allows T cells to guide their responses in health and in disease. Mass spectrometry-based proteomics is well poised to offer such insights, including monitoring emergence of resistance mechanisms to immunotherapeutics during treatments. A major application of this technology is in the discovery and characterization of small-molecule agents capable of enhancing the response to immunotherapeutic agents. Such an approach would reinvigorate small-molecule drug development aimed not at tumor cells but rather at tumor-resident T cells capable of producing dramatic and durable antitumor responses.
Collapse
Affiliation(s)
- Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Amer A Beg
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Scott Antonia
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
24
|
Tortorella SM, Hung A, Karagiannis TC. The implication of cancer progenitor cells and the role of epigenetics in the development of novel therapeutic strategies for chronic myeloid leukemia. Antioxid Redox Signal 2015; 22:1425-62. [PMID: 25366930 DOI: 10.1089/ars.2014.6096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Chronic myeloid leukemia (CML) involves the malignant transformation of hematopoietic stem cells, defined largely by the Philadelphia chromosome and expression of the breakpoint cluster region-Abelson (BCR-ABL) oncoprotein. Pharmacological tyrosine kinase inhibitors (TKIs), including imatinib mesylate, have overcome limitations in conventional treatment for the improved clinical management of CML. RECENT ADVANCES Accumulated evidence has led to the identification of a subpopulation of quiescent leukemia progenitor cells with stem-like self renewal properties that may initiate leukemogenesis, which are also shown to be present in residual disease due to their insensitivity to tyrosine kinase inhibition. CRITICAL ISSUES The characterization of quiescent leukemia progenitor cells as a unique cell population in CML pathogenesis has become critical with the complete elucidation of mechanisms involved in their survival independent of BCR-ABL that is important in the development of novel anticancer strategies. Understanding of these functional pathways in CML progenitor cells will allow for their selective therapeutic targeting. In addition, disease pathogenesis and drug responsiveness is also thought to be modulated by epigenetic regulatory mechanisms such as DNA methylation, histone acetylation, and microRNA expression, with a capacity to control CML-associated gene transcription. FUTURE DIRECTIONS A number of compounds in combination with TKIs are under preclinical and clinical investigation to assess their synergistic potential in targeting leukemic progenitor cells and/or the epigenome in CML. Despite the collective promise, further research is required in order to refine understanding, and, ultimately, advance antileukemic therapeutic strategies.
Collapse
Affiliation(s)
- Stephanie M Tortorella
- 1 Epigenomic Medicine, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct , Melbourne, Australia
| | | | | |
Collapse
|
25
|
de Oliveira GAP, Rangel LP, Costa DC, Silva JL. Misfolding, Aggregation, and Disordered Segments in c-Abl and p53 in Human Cancer. Front Oncol 2015; 5:97. [PMID: 25973395 PMCID: PMC4413674 DOI: 10.3389/fonc.2015.00097] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/10/2015] [Indexed: 01/31/2023] Open
Abstract
The current understanding of the molecular mechanisms that lead to cancer is not sufficient to explain the loss or gain of function in proteins related to tumorigenic processes. Among them, more than 100 oncogenes, 20-30 tumor-suppressor genes, and hundreds of genes participating in DNA repair and replication have been found to play a role in the origins of cancer over the last 25 years. The phosphorylation of serine, threonine, or tyrosine residues is a critical step in cellular growth and development and is achieved through the tight regulation of protein kinases. Phosphorylation plays a major role in eukaryotic signaling as kinase domains are found in 2% of our genes. The deregulation of kinase control mechanisms has disastrous consequences, often leading to gains of function, cell transformation, and cancer. The c-Abl kinase protein is one of the most studied targets in the fight against cancer and is a hotspot for drug development because it participates in several solid tumors and is the hallmark of chronic myelogenous leukemia. Tumor suppressors have the opposite effects. Their fundamental role in the maintenance of genomic integrity has awarded them a role as the guardians of DNA. Among the tumor suppressors, p53 is the most studied. The p53 protein has been shown to be a transcription factor that recognizes and binds to specific DNA response elements and activates gene transcription. Stress triggered by ionizing radiation or other mutagenic events leads to p53 phosphorylation and cell-cycle arrest, senescence, or programed cell death. The p53 gene is the most frequently mutated gene in cancer. Mutations in the DNA-binding domain are classified as class I or class II depending on whether substitutions occur in the DNA contact sites or in the protein core, respectively. Tumor-associated p53 mutations often lead to the loss of protein function, but recent investigations have also indicated gain-of-function mutations. The prion-like aggregation of mutant p53 is associated with loss-of-function, dominant-negative, and gain-of-function effects. In the current review, we focused on the most recent insights into the protein structure and function of the c-Abl and p53 proteins that will provide us guidance to understand the loss and gain of function of these misfolded tumor-associated proteins.
Collapse
Affiliation(s)
- Guilherme A. P. de Oliveira
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Biologia Estrutural e Bioimagem, Centro Nacional de Ressonância Magnética Nuclear Jiri Jonas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana P. Rangel
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Biologia Estrutural e Bioimagem, Centro Nacional de Ressonância Magnética Nuclear Jiri Jonas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielly C. Costa
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Biologia Estrutural e Bioimagem, Centro Nacional de Ressonância Magnética Nuclear Jiri Jonas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jerson L. Silva
- Programa de Biologia Estrutural, Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Biologia Estrutural e Bioimagem, Centro Nacional de Ressonância Magnética Nuclear Jiri Jonas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Jochems J, Teegarden SL, Chen Y, Boulden J, Challis C, Ben-Dor GA, Kim SF, Berton O. Enhancement of stress resilience through histone deacetylase 6-mediated regulation of glucocorticoid receptor chaperone dynamics. Biol Psychiatry 2015; 77:345-55. [PMID: 25442004 PMCID: PMC4297530 DOI: 10.1016/j.biopsych.2014.07.036] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/29/2014] [Accepted: 07/29/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Acetylation of heat shock protein 90 (Hsp90) regulates downstream hormone signaling via the glucocorticoid receptor (GR), but the role of this molecular mechanism in stress homeostasis is poorly understood. We tested whether acetylation of Hsp90 in the brain predicts and modulates the behavioral sequelae of a mouse model of social stress. METHODS Mice subjected to chronic social defeat stress were stratified into resilient and vulnerable subpopulations. Hypothalamic-pituitary-adrenal axis function was probed using a dexamethasone/corticotropin-releasing factor test. Measurements of Hsp90 acetylation, Hsp90-GR interactions, and GR translocation were performed in the dorsal raphe nucleus. To manipulate Hsp90 acetylation, we pharmacologically inhibited histone deacetylase 6, a known deacetylase of Hsp90, or overexpressed a point mutant that mimics the hyperacetylated state of Hsp90 at lysine K294. RESULTS Lower acetylated Hsp90, higher GR-Hsp90 association, and enhanced GR translocation were observed in dorsal raphe nucleus of vulnerable mice after chronic social defeat stress. Administration of ACY-738, a histone deacetylase 6-selective inhibitor, led to Hsp90 hyperacetylation in brain and in neuronal culture. In cell-based assays, ACY-738 increased the relative association of Hsp90 with FK506 binding protein 51 versus FK506 binding protein 52 and inhibited hormone-induced GR translocation. This effect was replicated by overexpressing the acetylation-mimic point mutant of Hsp90. In vivo, ACY-738 promoted resilience to chronic social defeat stress, and serotonin-selective viral overexpression of the acetylation-mimic mutant of Hsp90 in raphe neurons reproduced the behavioral effect of ACY-738. CONCLUSIONS Hyperacetylation of Hsp90 is a predictor and causal molecular determinant of stress resilience in mice. Brain-penetrant histone deacetylase 6 inhibitors increase Hsp90 acetylation and modulate GR chaperone dynamics offering a promising strategy to curtail deleterious socioaffective effects of stress and glucocorticoids.
Collapse
Affiliation(s)
- Jeanine Jochems
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Sarah L Teegarden
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Yong Chen
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Janette Boulden
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Collin Challis
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Gabriel A Ben-Dor
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Sangwon F Kim
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia PA 19104-3403
| | - Olivier Berton
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia Pennsylvania..
| |
Collapse
|
27
|
Wang W, Zhang J, Li Y, Yang X, He Y, Li T, Ren F, Zhang J, Lin R. Divalproex sodium enhances the anti-leukemic effects of imatinib in chronic myeloid leukemia cells partly through SIRT1. Cancer Lett 2014; 356:791-9. [PMID: 25449787 DOI: 10.1016/j.canlet.2014.10.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 10/28/2014] [Accepted: 10/28/2014] [Indexed: 11/29/2022]
Abstract
Imatinib (IM) represents a breakthrough in the treatment of chronic myeloid leukemia (CML) by inhibiting the activity of Bcr-Abl tyrosine kinase. However, many patients exhibit resistance to IM in the clinic. Recent studies have indicated that sirtuin 1 (SIRT1), a class III histone deacetylase (HDAC), plays an important role in leukemogenesis. In addition, some HDAC inhibitors are being tested to determine their anti-cancer activities in clinical trials. Divalproex sodium (DVPX), a first-line treatment for epilepsy, is also a HDAC inhibitor. However, it is unclear whether the anti-leukemic effects of IM in combination with DVPX on CML cells are related to SIRT1. The aim of this study was to investigate the effects of IM in combination with DVPX on cell viability, apoptosis, and cell cycle arrest in CML cells and to explore the underlying mechanisms. It was found that DVPX enhanced IM-induced cell growth inhibition, apoptosis and cell cycle arrest in K562-S and K562-G cells. Surprisingly, the level of p-Bcr-Abl was similar in K562-S and K562-G cells. Moreover, IM combined with DVPX had no effects on the phosphorylation of Bcr-Abl and its downstream target STAT5. Further study revealed that SIRT1 expression was higher in K562-G cells compared with K562-S cells. DVPX enhanced the inhibitory effect of IM on SIRT1 expression in K562-S and K562-G cells. Furthermore, knockdown of SIRT1 promoted apoptosis of K562-G cells treated with IM and DVPX. These results indicate that DVPX may increase the sensitivity of CML cells to IM and reverse IM resistance by regulating SIRT1 expression.
Collapse
Affiliation(s)
- Weirong Wang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China; Laboratory Animal Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jianfeng Zhang
- Department of Pharmacy, Eighth Hospital of Xi'an City, Xi'an, Shaanxi 710061, China
| | - Yanxiang Li
- Department of Pharmacology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaofeng Yang
- Department of Pharmacology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanhao He
- Department of Pharmacology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Tingting Li
- Department of Pharmacology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Feng Ren
- Department of Pharmacology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Rong Lin
- Department of Pharmacology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
28
|
Zhu L, Qi J, Chiao CYC, Zhang Q, Porco JA, Faller DV, Dai Y. Identification of a novel polyprenylated acylphloroglucinol‑derived SIRT1 inhibitor with cancer‑specific anti-proliferative and invasion-suppressing activities. Int J Oncol 2014; 45:2128-36. [PMID: 25189993 PMCID: PMC4203335 DOI: 10.3892/ijo.2014.2639] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/10/2014] [Indexed: 01/03/2023] Open
Abstract
SIRT1, a class III histone deacetylase, plays a critical role in regulating cancer cell growth, migration and invasion, which makes it a potential target for cancer therapeutics. In this study, we screened derivatives of several groups of natural products and identified a novel SIRT1 inhibitor JQ-101, a synthetic derivative of the polyprenylated acylphloroglucinol (PPAP) natural products, with an IC(50) for SIRT1 of 30 µM in vitro, with 5-fold higher activity for SIRT1 vs. SIRT2. Exposure of tumor cells to JQ-101 significantly enhanced acetylation of p53 and histone H4K16 at known sites of SIRT1 deacetylation, validating SIRT1 as its cellular target. JQ-101 suppressed cancer cell growth and survival by targeting SIRT1, and also exhibited selective cytotoxicity towards a panel of human tumor cell lines, while producing no toxicity in two normal human cell types at comparable concentrations. JQ-101 induced both apoptosis and cell senescence, and suppressed cancer cell invasion in vitro. In summary, we have identified JQ-101 as a new SIRT1 inhibitor which may have potential application in cancer treatment through its ability to induce tumor cell apoptosis and senescence and suppress cancer cell invasion.
Collapse
Affiliation(s)
- Lijia Zhu
- Cancer Research Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ji Qi
- Department of Chemistry, Center for Chemical Methodology and Library Development (CMLD-BU), Boston University, Boston, MA 02215, USA
| | - Christine Ya-Chi Chiao
- Cancer Research Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Qiang Zhang
- Department of Chemistry, Center for Chemical Methodology and Library Development (CMLD-BU), Boston University, Boston, MA 02215, USA
| | - John A Porco
- Department of Chemistry, Center for Chemical Methodology and Library Development (CMLD-BU), Boston University, Boston, MA 02215, USA
| | - Douglas V Faller
- Cancer Research Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Yan Dai
- Cancer Research Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
29
|
Regulatory role of the 90-kDa-heat-shock protein (Hsp90) and associated factors on gene expression. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:71-87. [DOI: 10.1016/j.bbagrm.2013.12.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 12/23/2013] [Accepted: 12/26/2013] [Indexed: 12/31/2022]
|
30
|
Sinclair A, Latif AL, Holyoake TL. Targeting survival pathways in chronic myeloid leukaemia stem cells. Br J Pharmacol 2014; 169:1693-707. [PMID: 23517124 DOI: 10.1111/bph.12183] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/11/2013] [Accepted: 02/19/2013] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Chronic myeloid leukaemia (CML) is a clonal myeloproliferative disorder characterized by the presence of a fusion oncogene BCR-ABL, which encodes a protein with constitutive TK activity. The implementation of tyrosine kinase inhibitors (TKIs) marked a major advance in CML therapy; however, there are problems with current treatment. For example, relapse occurs when these drugs are discontinued in the majority of patients who have achieved a complete molecular response on TKI and these agents are less effective in patients with mutations in the BCR-ABL kinase domain. Importantly, TKI can effectively target proliferating mature cells, but do not eradicate quiescent leukaemic stem cells (LSCs), therefore allowing disease persistence despite treatment. It is essential that alternative strategies are used to target the LSC population. BCR-ABL activation is responsible for the modulation of different signalling pathways, which allows the LSC fraction to evade cell death. Several pathways have been shown to be modulated by BCR-ABL, including PI3K/AKT/mTOR, JAK-STAT and autophagy signalling pathways. Targeting components of these survival pathways, alone or in combination with TKI, therefore represents an attractive potential therapeutic approach for targeting the LSC. However, many pathways are also active in normal stem cells. Therefore, potential targets must be validated to effectively eradicate CML stem cells while sparing normal counterparts. This review summarizes the main pathways modulated in CML stem cells, the recent developments and the use of novel drugs to target components in these pathways which may be used to target the LSC population. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- A Sinclair
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | | | | |
Collapse
|
31
|
García-Sanz R, Ocio EM. Novel treatment regimens for Waldenström’s macroglobulinemia. Expert Rev Hematol 2014; 3:339-50. [DOI: 10.1586/ehm.10.19] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
32
|
MicroRNA-124 (miR-124) regulates Ku70 expression and is correlated with neuronal death induced by ischemia/reperfusion. J Mol Neurosci 2013; 52:148-55. [PMID: 24166354 DOI: 10.1007/s12031-013-0155-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 10/15/2013] [Indexed: 12/13/2022]
Abstract
MicroRNAs are small, non-coding RNA molecules that regulate gene expression, and miR-124 is the most abundant miRNA in the brain. Studies have shown that miR-124 is clearly reduced in the ischemic brain after stroke; however, the role of miR-124 after stroke is less well studied. Using TargetScan, MicroCosm Targets version 5, and microRNA.org databases, we identified miR-124 as a possible regulator of the DNA repair protein Ku70. We validated that Ku70 is a target for miR-124 with a luciferase reporter activity assay. Moreover, adult rats subjected to focal cerebral ischemia exhibited a substantial reduction of miR-124 expression, which was inversely upregulated by Ku70 expression. In vivo treatment with miR-124 antagomir effectively enhanced Ku70 mRNA and protein levels in the ischemic region. Furthermore, knockdown of cerebral miR-124 reduced cell death and infarct size and improved neurological outcomes. Our data demonstrate that miR-124 is an endogenous regulator of Ku70 that improves ischemia/reperfusion (I/R)-induced brain injury and dysfunction.
Collapse
|
33
|
Jabbour EJ, Cortes JE, Kantarjian HM. Resistance to tyrosine kinase inhibition therapy for chronic myelogenous leukemia: a clinical perspective and emerging treatment options. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2013; 13:515-529. [PMID: 23890944 PMCID: PMC4160831 DOI: 10.1016/j.clml.2013.03.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 03/06/2013] [Indexed: 01/12/2023]
Abstract
The development of tyrosine kinase inhibitors (TKIs) has led to extended lifespans for many patients with chronic myelogenous leukemia (CML). However, 20% to 30% of patients fail to respond, respond suboptimally, or experience disease relapse after treatment with imatinib. A key factor is drug resistance. The molecular mechanisms implicated in this resistance include those that involve upregulation or mutation of BCR-ABL kinase and those that are BCR-ABL independent. The clinical consequences of these molecular mechanisms of resistance for disease pathogenesis remain open for debate. This review summarizes the molecular mechanisms and clinical consequences of TKI resistance and addresses the current and future treatment approaches for patients with TKI-resistant CML.
Collapse
Affiliation(s)
- Elias J Jabbour
- The University of Texas, MD Anderson Cancer Center, Houston, TX.
| | | | | |
Collapse
|
34
|
Kosan C, Ginter T, Heinzel T, Krämer OH. STAT5 acetylation: Mechanisms and consequences for immunological control and leukemogenesis. JAKSTAT 2013; 2:e26102. [PMID: 24416653 PMCID: PMC3876427 DOI: 10.4161/jkst.26102] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 08/08/2013] [Accepted: 08/09/2013] [Indexed: 12/30/2022] Open
Abstract
The cytokine-inducible transcription factors signal transducer and activator of transcription 5A and 5B (STAT5A and STAT5B) are important for the proper development of multicellular eukaryotes. Disturbed signaling cascades evoking uncontrolled expression of STAT5 target genes are associated with cancer and immunological failure. Here, we summarize how STAT5 acetylation is integrated into posttranslational modification networks within cells. Moreover, we focus on how inhibitors of deacetylases and tyrosine kinases can correct leukemogenic signaling nodes involving STAT5. Such small molecules can be exploited in the fight against neoplastic diseases and immunological disorders.
Collapse
Affiliation(s)
- Christian Kosan
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany
| | - Torsten Ginter
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany
| | - Thorsten Heinzel
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany
| | - Oliver H Krämer
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany ; Institute of Toxicology; Medical Center of the University Mainz; Mainz, Germany
| |
Collapse
|
35
|
Kim WS, Lee MJ, Kim DH, Lee JE, Kim JI, Kim YC, Song MR, Park SG. 5′-OH-5-nitro-Indirubin oxime (AGM130), an Indirubin derivative, induces apoptosis of Imatinib-resistant chronic myeloid leukemia cells. Leuk Res 2013; 37:427-33. [DOI: 10.1016/j.leukres.2012.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 12/30/2012] [Accepted: 12/30/2012] [Indexed: 11/24/2022]
|
36
|
Lee HR, Kim MJ, Ha G, Kim SJ, Kim SH, Kang CD. Presence of Leukemia-maintaining Cells in Differentiation-resistant Fraction of K562 Chronic Myelogenous Leukemia. ACTA ACUST UNITED AC 2013. [DOI: 10.5352/jls.2013.23.2.197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
37
|
Karvela M, Helgason GV, Holyoake TL. Mechanisms and novel approaches in overriding tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Expert Rev Anticancer Ther 2012; 12:381-92. [PMID: 22369329 DOI: 10.1586/era.12.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic myeloid leukemia is a stem cell-initiated but progenitor-driven disease induced by the BCR-ABL oncogene. Tyrosine kinase inhibitors (TKIs) were introduced in the late 1990s and have revolutionized the management of chronic myeloid leukemia in chronic phase. The majority of patients can now expect to live a normal life as long as they continue to comply with TKI treatment. However, in a significant proportion of cases TKI resistance develops over time, requiring a switch of therapy. The most frequent mechanism for drug resistance is the development of kinase domain mutations that reduce or completely ablate drug efficacy. Fortunately, the last 10 years have seen an impressive array of new drugs, some modeled on the mechanism of action of imatinib, others employing more novel approaches, for these patients.
Collapse
Affiliation(s)
- Maria Karvela
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 OYN, UK
| | | | | |
Collapse
|
38
|
Dos Santos SC, Mira NP, Moreira AS, Sá-Correia I. Quantitative- and phospho-proteomic analysis of the yeast response to the tyrosine kinase inhibitor imatinib to pharmacoproteomics-guided drug line extension. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2012; 16:537-51. [PMID: 22775238 DOI: 10.1089/omi.2012.0012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Imatinib mesylate (IM) is a potent tyrosine kinase inhibitor used as front-line therapy in chronic myeloid leukemia, a disease caused by the oncogenic kinase Bcr-Abl. Although the clinical success of IM set a new paradigm in molecular-targeted therapy, the emergence of IM resistance is a clinically significant problem. In an effort to obtain new insights into the mechanisms of adaptation and tolerance to IM, as well as the signaling pathways potentially affected by this drug, we performed a two-dimensional electrophoresis-based quantitative- and phospho-proteomic analysis in the eukaryotic model Saccharomyces cerevisiae. We singled out proteins that were either differentially expressed or differentially phosphorylated in response to IM, using the phosphoselective dye Pro-Q(®) Diamond, and identified 18 proteins in total. Ten were altered only at the content level (mostly decreased), while the remaining 8 possessed IM-repressed phosphorylation. These 18 proteins are mainly involved in cellular carbohydrate processes (glycolysis/gluconeogenesis), translation, protein folding, ion homeostasis, and nucleotide and amino acid metabolism. Remarkably, all 18 proteins have human functional homologs. A role for HSP70 proteins in the response to IM, as well as decreased glycolysis as a metabolic marker of IM action are suggested, consistent with findings from studies in human cell lines. The previously-proposed effect of IM as an inhibitor of vacuolar H(+)-ATPase function was supported by the identification of an underexpressed protein subunit of this complex. Taken together, these findings reinforce the role of yeast as a valuable eukaryotic model for pharmacological studies and identification of new drug targets, with potential clinical implications in drug reassignment or line extension under a personalized medicine perspective.
Collapse
Affiliation(s)
- Sandra C Dos Santos
- Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Technical University of Lisbon, Lisbon, Portugal
| | | | | | | |
Collapse
|
39
|
Kusio-Kobialka M, Wolanin K, Podszywalow-Bartnicka P, Sikora E, Skowronek K, McKenna SL, Ghizzoni M, Dekker FJ, Piwocka K. Increased acetylation of lysine 317/320 of p53 caused by BCR-ABL protects from cytoplasmic translocation of p53 and mitochondria-dependent apoptosis in response to DNA damage. Apoptosis 2012; 17:950-63. [DOI: 10.1007/s10495-012-0739-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
40
|
Cervera E, Candelaria M, López-Navarro O, Labardini J, Gonzalez-Fierro A, Taja-Chayeb L, Cortes J, Gordillo-Bastidas D, Dueñas-González A. Epigenetic therapy with hydralazine and magnesium valproate reverses imatinib resistance in patients with chronic myeloid leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:207-12. [PMID: 22420986 DOI: 10.1016/j.clml.2012.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 01/10/2012] [Accepted: 01/20/2012] [Indexed: 01/07/2023]
Abstract
UNLABELLED The epigenetic drugs hydralazine and valproate were administered in a compassionate manner to 8 patients with chronic myeloid leukemia (CML) refractory to imatinib. Two patients had a complete hematologic response (25%),1 major cytogenetic response, 1 complete cytogenetic response (25% any cytogenetic response), and 3 (37.5%)stable disease. No grade 3 or 4 toxicity was observed. These results show the ability of epigenetic therapy to revert imatinib resistance. BACKGROUND Epigenetic alterations participate in the development of acquired resistance to imatinib, hence, the DNA methylation, and histone deacetylase inhibitors hydralazine and valproate, respectively, has the potential to overcome it. PATIENT AND METHODS A series of 8 patients with chronic myeloid leukemia (CML) refractory to imatinib mesylate with no access to second-generation tyrosine kinase inhibitors were treated with hydralazine and valproate in a compassionate manner. Clinical efficacy and safety of these drugs added to imatinib mesylate were evaluated. RESULTS Two patients were in the blast phase, 5 were in the accelerated phase, and 1 was in the chronic phase. All the patients continued with the same dose of imatinib that they had been receiving at the time of development of resistance, with a median dose of 600 mg daily (range, 400-800 mg). The median time from diagnosis of CML to the start of hydralazine and valproate was 53.6 months (range, 19-84 months). Two (25%) patients had a complete hematologic response, one (12.5%) had an major cytogenetic response, and one (12.5%) had a complete cytogenetic response. Three (37.5%) patients had stable disease, and only one (12.5%) patient failed to respond. At a median follow-up time of 18 months (range, 3-18 months), the median survival had not been reached, and the projected overall survival was 63%. All the patients had mild neurologic toxicity, including distal tremor and somnolence. No grade 3 or 4 toxicity was observed. CONCLUSIONS Our results suggest that the epigenetic drugs hydralazine and valproate revert the resistance to imatinib in patients with CML.
Collapse
MESH Headings
- Adult
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Benzamides
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic/drug effects
- Female
- Follow-Up Studies
- Humans
- Hydralazine/administration & dosage
- Hydralazine/adverse effects
- Hydralazine/therapeutic use
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Male
- Middle Aged
- Piperazines/administration & dosage
- Piperazines/adverse effects
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Pyrimidines/administration & dosage
- Pyrimidines/adverse effects
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Treatment Outcome
- Valproic Acid/administration & dosage
- Valproic Acid/adverse effects
- Valproic Acid/therapeutic use
Collapse
Affiliation(s)
- Eduardo Cervera
- Hematology Department, Instituto Nacional de Cancerología, México City, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Myeloid hematological malignancies are among the epigenetically best characterized neoplasms. The comparatively low number of recurring balanced and unbalanced chromosomal abnormalities as well as common genetic mutations has enabled scientists to relate epigenetic states to these. The ease of accessing malignant cells through bone marrow aspiration has certainly contributed to the fast expansion of knowledge. Even so, the clinical and pathogenetic relevance of epigenetic changes is still not known, and the field will certainly evolve very fast with the development of new analytic techniques. The first example of successful epigenetic therapy is seen in myeloid malignancies, in the high-risk myelodysplastic syndromes (MDS) which are routinely treated with the demethylating agent azacytidine.This chapter will concentrate on describing the epigenetic changes in acute myeloid leukemia (AML), chronic myeloid leukemia (CML) and MDS. An overview of clinical relevance and epigenetic therapeutic approaches is also made.
Collapse
Affiliation(s)
- Stefan Deneberg
- Center of Hematology, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
42
|
Mechanisms of Resistance to Hsp90 Inhibitor Drugs: A Complex Mosaic Emerges. Pharmaceuticals (Basel) 2011; 4:1400-1422. [PMID: 27721330 PMCID: PMC4060131 DOI: 10.3390/ph4111400] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/04/2011] [Accepted: 10/17/2011] [Indexed: 01/07/2023] Open
Abstract
The molecular chaperone Hsp90 holds great promise as a cancer drug target, despite some of the initial clinical trials of Hsp90 inhibitor drugs having not lived up to expectation. Effective use of these drugs will benefit greatly from a much more detailed understanding of the factors that contribute to resistance, whether intrinsic or acquired. We review how cell culture studies have revealed a number of different mechanisms whereby cells can be rendered less susceptible to the effects of Hsp90 inhibitor treatment. A major influence is Hsp90 inhibition causing strong induction of the heat shock response, a stress response that increases cellular levels of prosurvival chaperones such as Hsp27 and Hsp70. Another problem seems to be that these inhibitors do not always access the Hsp90 proteins of the mitochondrion, forms of Hsp90 that—in cancer cells—are operating to suppress apoptosis. It should be possible to overcome these drawbacks through the appropriate drug redesign or with the combinatorial use of an Hsp90 inhibitor with a drug that targets either heat shock factor or the chaperone Hsp70. Still though, cells will often differ in the key antiapoptotic versus proapoptotic activities that are dependent on Hsp90, in the key steps in their apoptotic pathways responsive to Hsp90 inhibition or Hsp70 level, as well as the extents to which their survival is dependent on oncogenic tyrosine kinases that are clients of Hsp90. A systems approach will therefore often be required in order to establish the most prominent effects of Hsp90 inhibition in each type of cancer cell.
Collapse
|
43
|
Synergistic interactions between HDAC and sirtuin inhibitors in human leukemia cells. PLoS One 2011; 6:e22739. [PMID: 21818379 PMCID: PMC3144930 DOI: 10.1371/journal.pone.0022739] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 07/05/2011] [Indexed: 12/11/2022] Open
Abstract
Aberrant histone deacetylase (HDAC) activity is frequent in human leukemias. However, while classical, NAD+-independent HDACs are an established therapeutic target, the relevance of NAD+-dependent HDACs (sirtuins) in leukemia treatment remains unclear. Here, we assessed the antileukemic activity of sirtuin inhibitors and of the NAD+-lowering drug FK866, alone and in combination with traditional HDAC inhibitors. Primary leukemia cells, leukemia cell lines, healthy leukocytes and hematopoietic progenitors were treated with sirtuin inhibitors (sirtinol, cambinol, EX527) and with FK866, with or without addition of the HDAC inhibitors valproic acid, sodium butyrate, and vorinostat. Cell death was quantified by propidium iodide cell staining and subsequent flow-cytometry. Apoptosis induction was monitored by cell staining with FITC-Annexin-V/propidium iodide or with TMRE followed by flow-cytometric analysis, and by measuring caspase3/7 activity. Intracellular Bax was detected by flow-cytometry and western blotting. Cellular NAD+ levels were measured by enzymatic cycling assays. Bax was overexpressed by retroviral transduction. Bax and SIRT1 were silenced by RNA-interference. Sirtuin inhibitors and FK866 synergistically enhanced HDAC inhibitor activity in leukemia cells, but not in healthy leukocytes and hematopoietic progenitors. In leukemia cells, HDAC inhibitors were found to induce upregulation of Bax, a pro-apoptotic Bcl2 family-member whose translocation to mitochondria is normally prevented by SIRT1. As a result, leukemia cells become sensitized to sirtuin inhibitor-induced apoptosis. In conclusion, NAD+-independent HDACs and sirtuins cooperate in leukemia cells to avoid apoptosis. Combining sirtuin with HDAC inhibitors results in synergistic antileukemic activity that could be therapeutically exploited.
Collapse
|
44
|
Marino AM, Sofiadis A, Baryawno N, Johnsen JI, Larsson C, Vukojević V, Ekström TJ. Enhanced effects by 4-phenylbutyrate in combination with RTK inhibitors on proliferation in brain tumor cell models. Biochem Biophys Res Commun 2011; 411:208-12. [PMID: 21726539 DOI: 10.1016/j.bbrc.2011.06.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Accepted: 06/21/2011] [Indexed: 01/29/2023]
Abstract
We have investigated in vitro effects of anticancer therapy with the histone deacetylase inhibitor (HDACi) 4-phenylbutyrate (4-PB) combined with receptor tyrosine kinase inhibitors (RTKi) gefitinib or vandetanib on the survival of glioblastoma (U343MGa) and medulloblastoma (D324Med) cells. In comparison with individual effects of these drugs, combined treatment with gefitinib/4-PB or vandetanib/4-PB resulted in enhanced cell killing and reduced clonogenic survival in both cell lines. Our results suggest that combined treatment using HDACi and RTKi may beneficially affect the outcome of cancer therapy.
Collapse
Affiliation(s)
- Ana-Maria Marino
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
45
|
Buchi F, Pastorelli R, Ferrari G, Spinelli E, Gozzini A, Sassolini F, Bosi A, Tombaccini D, Santini V. Acetylome and phosphoproteome modifications in imatinib resistant chronic myeloid leukaemia cells treated with valproic acid. Leuk Res 2011; 35:921-31. [PMID: 21382639 DOI: 10.1016/j.leukres.2011.01.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 01/10/2011] [Accepted: 01/29/2011] [Indexed: 11/26/2022]
Abstract
Chronic myeloid leukaemia has a specific therapy: BCR/ABL inhibitor imatinib. Resistance due to BCR/ABL dependent and independent mechanisms is partially reversible by histone deacetylase inhibitors. We analysed by 2D-electrophoresis and anti-pan-acetylated and anti-phosphotyrosine immunoblots, followed by spot-matching and MALDI-TOF mass spectrometry, which proteome modifications would parallel restoration of sensitivity to imatinib by valproic acid (VPA). VPA plus imatinib significantly increased acetylation of HSP90 and hnRNP L and decreased phosphorylation of HSPs and hnRNPs in imatinib resistant cells. VPA was able to modify profoundly acetylome and phosphoproteome of CML cells, while reverting resistance to imatinib.
Collapse
Affiliation(s)
- Francesca Buchi
- Functional Unit of Haematology, Department of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Li QF, Yan J, Zhang K, Yang YF, Xiao FJ, Wu CT, Wang H, Wang LS. Bortezomib and sphingosine kinase inhibitor interact synergistically to induces apoptosis in BCR/ABl+ cells sensitive and resistant to STI571 through down-regulation Mcl-1. Biochem Biophys Res Commun 2011; 405:31-6. [DOI: 10.1016/j.bbrc.2010.12.111] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 12/21/2010] [Indexed: 11/30/2022]
|
47
|
Mithraprabhu S, Grigoriadis G, Khong T, Spencer A. Deactylase inhibition in myeloproliferative neoplasms. Invest New Drugs 2010; 28 Suppl 1:S50-7. [PMID: 21127942 PMCID: PMC3003795 DOI: 10.1007/s10637-010-9590-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 10/28/2010] [Indexed: 12/02/2022]
Abstract
Myeloproliferative neoplasms (MPN) are clonal haemopoietic progenitor cell disorders characterized by the proliferation of one or more of the haemopoietic lineages (myeloid, erythroid and/or megakaryocytic). The MPNs include eight haematological disorders: chronic myelogenous leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), primary myelofibrosis (PMF), systemic mastocytosis (SM), chronic eosinophilic leukemia, not otherwise specified (CEL, NOS), chronic neutrophilic leukemia (CNL), and unclassifiable MPN (MPN, U). Therapeutic interventions for MPNs include the use of tyrosine kinase inhibitors (TKIs) for BCR-ABL1(+) CML and JAK2 inhibitors for PV, ET and PMF. Histone deacetylase inhibitors (HDACi) are a novel class of drugs capable of altering the acetylation status of both histone and non-histone proteins, thereby affecting a repertoire of cellular functions in neoplastic cells including proliferation, differentiation, immune responses, angiogenesis and survival. Preliminary studies indicate that HDACi when used in combination with tyrosine kinase or JAK2 inhibitors may overcome resistance to the latter agents and enhance the pro-apoptotic effects on MPN cells. This review provides a review of pre-clinical and clinical studies that have explored the use of HDACi as potential therapeutics for MPNs.
Collapse
Affiliation(s)
- Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital/Monash University, Commercial Road, Melbourne, Victoria 3004 Australia
| | - George Grigoriadis
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital/Monash University, Commercial Road, Melbourne, Victoria 3004 Australia
- Burnet Institute, Commercial Road, Melbourne, Victoria 3004 Australia
- Department of Clinical Haematology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria 3004 Australia
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital/Monash University, Commercial Road, Melbourne, Victoria 3004 Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital/Monash University, Commercial Road, Melbourne, Victoria 3004 Australia
- Department of Clinical Haematology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria 3004 Australia
| |
Collapse
|
48
|
Abstract
Although only 5000 new cases of chronic myeloid leukemia (CML) were seen in the United States in 2009, this neoplasm continues to make scientific headlines year-after-year. Advances in understanding the molecular pathogenesis coupled with exciting developments in both drug design and development, targeting the initiating tyrosine kinase, have kept CML in the scientific limelight for more than a decade. Indeed, imatinib, a small-molecule inhibitor of the leukemia-initiating Bcr-Abl tyrosine kinase, has quickly become the therapeutic standard for newly diagnosed chronic phase-CML (CP-CML) patients. Yet, nearly one-third of patients will still have an inferior response to imatinib, either failing to respond to primary therapy or demonstrating progression after an initial response. Significant efforts geared toward understanding the molecular mechanisms of imatinib resistance have yielded valuable insights into the cellular biology of drug trafficking, enzyme structure and function, and the rational design of novel small molecule enzyme inhibitors. Indeed, new classes of kinase inhibitors have recently been investigated in imatinib-resistant CML. Understanding the pathogenesis of tyrosine kinase inhibitor resistance and the molecular rationale for the development of second and now third generation therapies for patients with CML will be keys to further disease control over the next 10 years.
Collapse
|
49
|
Rao KV, Iannucci A, Jabbour E. Current and Future Clinical Strategies in the Management of Chronic Myeloid Leukemia. Pharmacotherapy 2010. [DOI: 10.1592/phco.30.pt2.77s] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Zhou Q, Chaerkady R, Shaw PG, Kensler TW, Pandey A, Davidson NE. Screening for therapeutic targets of vorinostat by SILAC-based proteomic analysis in human breast cancer cells. Proteomics 2010; 10:1029-39. [PMID: 20049865 DOI: 10.1002/pmic.200900602] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Histone deacetylases (HDACs) play critical roles in silencing tumor suppressor genes. HDAC inhibitors reactivate tumor suppressor genes and inhibit tumor cell growth in vitro and in vivo, and several HDAC inhibitors are currently being evaluated in clinical trials for cancer therapy. A comprehensive analysis of proteins regulated by HDAC inhibitors would enhance our ability to define and characterize their essential therapeutic targets. Here, we employed stable isotope labeling with amino acids in cell culture-based quantitative proteomics to identify acetylated proteins in human breast cancer cells. Treatment with the clinically relevant HDAC inhibitor, suberoylanilide hydroxamic acid (vorinostat), induces lysine acetylation of 61 proteins in MDA-MB-231 human breast cancer cells. Suberoylanilide hydroxamic acid not only induces lysine acetylation in chromatin-associated proteins, but also acetylates previously unrecognized nonhistone proteins, including transcriptional factors and regulators, chaperones, cell structure proteins, and glycolytic enzymes in a time-dependent manner. Knowledge of the full repertoire of acetylated proteins will provide a foundation for further defining the functions of HDACs in cancer cells.
Collapse
Affiliation(s)
- Qun Zhou
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | |
Collapse
|