51
|
HDAC7 promotes the oncogenicity of nasopharyngeal carcinoma cells by miR-4465-EphA2 signaling axis. Cell Death Dis 2020; 11:322. [PMID: 32376822 PMCID: PMC7203158 DOI: 10.1038/s41419-020-2521-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022]
Abstract
HDAC7 plays a crucial role in cancers, and is the main drug target of several HDAC inhibitors. However, the role and mechanism of HDAC7 in nasopharyngeal carcinoma (NPC) are still unclear. In this study, we observed that HDAC7 was significantly upregulated in the NPC tissues relative to normal nasopharyngeal mucosa (NNM) tissues, HDAC7 expression levels were positively correlated with NPC progression and negatively correlated with patient prognosis, and HDAC7 knockdown dramatically inhibited the in vitro proliferation, migration, and invasion of NPC cells, and the growth of NPC xenografts in mice, indicating the HDAC7 promotes the oncogenicity of NPC. Mechanistically, HDAC7 promoted the in vitro proliferation, migration, and invasion of NPC cells by upregulating EphA2, in which miR-4465 mediated HDAC7-regulating EphA2, a direct target gene of miR-4465. We further showed that miR-4465 was significantly downregulated in the NPC tissues relative to NNM tissues, and inhibited the in vitro proliferation, migration, and invasion of NPC cells by targeting EphA2 expression. Moreover, we observed that the expressions of HDAC7, miR-4465, and EphA2 in NPC tissues were correlated. The results suggest that HDAC7 promotes the oncogenicity of NPC by downregulating miR-4465 and subsequently upregulating EphA2, highlighting HDAC7 as a potential therapeutic target for NPC.
Collapse
|
52
|
Li H, Li X, Lin H, Gong J. High HDAC9 is associated with poor prognosis and promotes malignant progression in pancreatic ductal adenocarcinoma. Mol Med Rep 2020; 21:822-832. [PMID: 31974610 PMCID: PMC6947911 DOI: 10.3892/mmr.2019.10869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 11/15/2019] [Indexed: 01/09/2023] Open
Abstract
Histone deacetylase 9 (HDAC9) is involved in a variety of malignant tumors, and leads to malignant tumor development and poor prognosis. However, the association between HDAC9 expression, and the prognosis and clinicopathological features of patients with pancreatic ductal adenocarcinoma (PDAC) remains unclear. The present study used reverse transcription‑quantitative PCR, western blotting and immunohistochemistry to detect the expression level of HDAC9 in PDAC tumors and cell lines. The Kaplan‑Meier method and Pearson's χ2 test were applied to evaluate the prognostic impact of HDAC9. The present study investigated the effect of HDAC9 on the biological function of PDAC cells. The present results indicated that HDAC9 was highly expressed in PDAC tissue and PDAC cell lines (P<0.05). HDAC9 expression level in tumor tissues was negatively associated with tumor size (P=0.026), T stage (P=0.014) and N stage (P=0.004). Kaplan‑Meier analysis suggested that patients with high HDAC9 had shorter recurrence‑free survival (RFS; P=0.017) and disease‑specific survival (DSS; P=0.022). Moreover, the present results suggested that T stage, N stage and HDAC9 expression level were independent predictive factors for RFS and DSS in patients with PDAC. In addition, silencing HDAC9 significantly inhibited the proliferation and migration of PDAC cells. The present results indicated that high expression levels of HDAC9 were associated with tumor progression and poor prognosis; thus, HDAC9 may serve as a prognostic predictor of PDAC.
Collapse
Affiliation(s)
- He Li
- Department of Hepatobiliary Surgery, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, P.R. China
| | - Xiaocheng Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Huapeng Lin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
53
|
Aberuyi N, Rahgozar S, Ghodousi ES, Ghaedi K. Drug Resistance Biomarkers and Their Clinical Applications in Childhood Acute Lymphoblastic Leukemia. Front Oncol 2020; 9:1496. [PMID: 32010613 PMCID: PMC6978753 DOI: 10.3389/fonc.2019.01496] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022] Open
Abstract
Biomarkers are biological molecules found in body fluids or tissues, which can be considered as indications of a normal or abnormal process, or of a condition or disease. There are various types of biomarkers based on their application and molecular alterations. Treatment-sensitivity or drug resistance biomarkers include prognostic and predictive molecules with utmost importance in selecting appropriate treatment protocols and improving survival rates. Acute lymphoblastic leukemia (ALL) is the most prevalent hematological malignancy diagnosed in children with nearly 80% cure rate. Despite the favorable survival rates of childhood ALL (chALL), resistance to chemotherapeutic agents and, as a consequence, a dismal prognosis develops in a significant number of patients. Therefore, there are urgent needs to have robust, sensitive, and disease-specific molecular prognostic and predictive biomarkers, which could allow better risk classification and then better clinical results. In this article, we review the currently known drug resistance biomarkers, including somatic or germ line nucleic acids, epigenetic alterations, protein expressions and metabolic variations. Moreover, biomarkers with potential clinical applications are discussed.
Collapse
Affiliation(s)
- Narges Aberuyi
- Division of Cellular and Molecular Biology, Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Soheila Rahgozar
- Division of Cellular and Molecular Biology, Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Elaheh Sadat Ghodousi
- Division of Cellular and Molecular Biology, Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Kamran Ghaedi
- Division of Cellular and Molecular Biology, Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| |
Collapse
|
54
|
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: 1.5] [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
|
55
|
Liang Z, Feng A, Shim H. MicroRNA-30c-regulated HDAC9 mediates chemoresistance of breast cancer. Cancer Chemother Pharmacol 2020; 85:413-423. [PMID: 31907648 DOI: 10.1007/s00280-019-04024-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/20/2019] [Indexed: 01/05/2023]
Abstract
PURPOSE Although histone deacetylase (HDAC) inhibitors have been shown to effectively induce the inhibition of proliferation and migration in breast cancer, the mechanism of HDAC9's contribution to chemoresistance remains poorly understood. The aim of this study was to investigate the role of miR-30c-regulated HDAC9 in chemoresistance of breast cancer and to determine the potential of selective inhibition of HDAC9 in sensitizing resistant breast cancer cells to chemotherapy. METHODS Expression levels of HDAC9 and miR-30c were measured in breast cancer cells and tissues using quantitative PCR analysis. The effect of selective inhibition of HDAC9 on sensitizing MDR cells to chemotherapy was assessed. MiR-30c/HDAC9 pathways' potential to mediate chemoresistance was analyzed. RESULTS Our studies show that HDAC9 was significantly up-regulated in chemoresistant breast cancer cell lines compared to a chemosensitive cell line and was inversely correlated with the levels of miR-30c. MiR-30c mimics and HDAC9 inhibitors reversed the chemoresistance of multidrug-resistant breast cancer cells. CONCLUSIONS These results indicate that the mechanism of chemoresistance reversal with selective HDAC inhibition was partially realized by regulating miR-30c via directly targeting HDAC9. Our findings suggest that the miR-30c/HDAC9 signaling axis could be a novel and potential therapeutic target in chemoresistant breast cancer.
Collapse
Affiliation(s)
- Zhongxing Liang
- Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| | - Amber Feng
- Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
56
|
Ye F, Huang J, Wang H, Luo C, Zhao K. Targeting epigenetic machinery: Emerging novel allosteric inhibitors. Pharmacol Ther 2019; 204:107406. [PMID: 31521697 DOI: 10.1016/j.pharmthera.2019.107406] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2019] [Indexed: 12/13/2022]
Abstract
Epigenetics has emerged as an extremely exciting fast-growing area of biomedical research in post genome era. Epigenetic dysfunction is tightly related with various diseases such as cancer and aging related degeneration, potentiating epigenetics modulators as important therapeutics targets. Indeed, inhibitors of histone deacetylase and DNA methyltransferase have been approved for treating blood tumor malignancies, whereas inhibitors of histone methyltransferase and histone acetyl-lysine recognizer bromodomain are in clinical stage. However, it remains a great challenge to discover potent and selective inhibitors by targeting catalytic site, as the same subfamily of epigenetic enzymes often share high sequence identity and very conserved catalytic core pocket. It is well known that epigenetic modifications are usually carried out by multi-protein complexes, and activation of catalytic subunit is often tightly regulated by other interactive protein component, especially in disease conditions. Therefore, it is not unusual that epigenetic complex machinery may exhibit allosteric regulation site induced by protein-protein interactions. Targeting allosteric site emerges as a compelling alternative strategy to develop epigenetic drugs with enhanced druggability and pharmacological profiles. In this review, we highlight recent progress in the development of allosteric inhibitors for epigenetic complexes through targeting protein-protein interactions. We also summarized the status of clinical applications of those inhibitors. Finally, we provide perspectives of future novel allosteric epigenetic machinery modulators emerging from otherwise undruggable single protein target.
Collapse
Affiliation(s)
- Fei Ye
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, China; College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jing Huang
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China; Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, China; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Cheng Luo
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China; Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, South Dong Qing Road, Guizhou 550025, China.
| | - Kehao Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, China; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| |
Collapse
|
57
|
Asfaha Y, Schrenk C, Alves Avelar LA, Hamacher A, Pflieger M, Kassack MU, Kurz T. Recent advances in class IIa histone deacetylases research. Bioorg Med Chem 2019; 27:115087. [PMID: 31561937 DOI: 10.1016/j.bmc.2019.115087] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/25/2019] [Accepted: 09/03/2019] [Indexed: 12/16/2022]
Abstract
Epigenetic control plays an important role in gene regulation through chemical modifications of DNA and post-translational modifications of histones. An essential post-translational modification is the histone acetylation/deacetylation-process which is regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). The mammalian zinc dependent HDAC family is subdivided into three classes: class I (HDACs 1-3, 8), class II (IIa: HDACs 4, 5, 7, 9; IIb: HDACs 6, 10) and class IV (HDAC 11). In this review, recent studies on the biological role and regulation of class IIa HDACs as well as their contribution in neurodegenerative diseases, immune disorders and cancer will be presented. Furthermore, the development, synthesis, and future perspectives of selective class IIa inhibitors will be highlighted.
Collapse
Affiliation(s)
- Yodita Asfaha
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Christian Schrenk
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Leandro A Alves Avelar
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Alexandra Hamacher
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Marc Pflieger
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Matthias U Kassack
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| | - Thomas Kurz
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
58
|
Xu G, Li N, Zhang Y, Zhang J, Xu R, Wu Y. MicroRNA-383-5p inhibits the progression of gastric carcinoma via targeting HDAC9 expression. ACTA ACUST UNITED AC 2019; 52:e8341. [PMID: 31365693 PMCID: PMC6668961 DOI: 10.1590/1414-431x20198341] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 05/07/2019] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs), as post-transcriptional regulators, have been reported to be involved in the initiation and progression of various types of cancer, including gastric cancer (GC). The present study aimed to investigate the role of miR-383-5p in gastric carcinogenesis. Cell viability was analyzed using CCK-8 kit. Annexin V-fluorescein isothiocyanate/propidium iodide double staining was used to evaluate cell apoptosis. The expression levels of miR-383-5p and histone deacetylase 9 (HDAC9) mRNA in GC tissues and cell lines were analyzed using RT-qPCR. The protein expression of HDAC9 was detected by western blotting. We found that HDAC9 was up-regulated and miR-383-5p was down-regulated in GC tissues and cell lines. High HDAC9 expression or low miR-383-5p expression was closely related to poor prognosis and metastasis in GC patients. HDAC9 knockout or miR-383-5p mimics led to growth inhibition and increased apoptosis in AGS and SGC-7901 cells. More importantly, we validated that miR-383-5p as a post-transcriptional regulator inhibited HDAC9 expression and was inversely correlated with HDAC9 expression in GC tissues. miR-383-5p had the opposite effects to HDAC9 in gastric carcinogenesis. miR-383-5p played an important role in gastric carcinogenesis, and it is one of the important mechanisms to regulate oncogenic HDAC9 in GC, which might be helpful in the development of novel therapeutic strategies for the treatment of GC.
Collapse
Affiliation(s)
- Gang Xu
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Na Li
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Yan Zhang
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Jinbiao Zhang
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Rui Xu
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| | - Yanling Wu
- Department of Oncology, Chinese PLA No.148 Hospital, Zibo, Shandong, China
| |
Collapse
|
59
|
Revisiting Histone Deacetylases in Human Tumorigenesis: The Paradigm of Urothelial Bladder Cancer. Int J Mol Sci 2019; 20:ijms20061291. [PMID: 30875794 PMCID: PMC6471041 DOI: 10.3390/ijms20061291] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/24/2022] Open
Abstract
Urinary bladder cancer is a common malignancy, being characterized by substantial patient mortality and management cost. Its high somatic-mutation frequency and molecular heterogeneity usually renders tumors refractory to the applied regimens. Hitherto, methotrexate-vinblastine-adriamycin-cisplatin and gemcitabine-cisplatin represent the backbone of systemic chemotherapy. However, despite the initial chemosensitivity, the majority of treated patients will eventually develop chemoresistance, which severely reduces their survival expectancy. Since chromatin regulation genes are more frequently mutated in muscle-invasive bladder cancer, as compared to other epithelial tumors, targeted therapies against chromatin aberrations in chemoresistant clones may prove beneficial for the disease. “Acetyl-chromatin” homeostasis is regulated by the opposing functions of histone acetyltransferases (HATs) and histone deacetylases (HDACs). The HDAC/SIRT (super-)family contains 18 members, which are divided in five classes, with each family member being differentially expressed in normal urinary bladder tissues. Since a strong association between irregular HDAC expression/activity and tumorigenesis has been previously demonstrated, we herein attempt to review the accumulated published evidences that implicate HDACs/SIRTs as critical regulators in urothelial bladder cancer. Moreover, the most extensively investigated HDAC inhibitors (HDACis) are also analyzed, and the respective clinical trials are also described. Interestingly, it seems that HDACis should be preferably used in drug-combination therapeutic schemes, including radiation.
Collapse
|
60
|
Abstract
Cancer can be identified as a chaotic cell state, which breaks the rules that govern growth and reproduction, with main characteristics such as uncontrolled division, invading other tissues, usurping resources, and eventually killing its host. It was once believed that cancer is caused by a progressive series of genetic aberrations, and certain mutations of genes, including oncogenes and tumor suppressor genes, have been identified as the cause of cancer. However, piling evidence suggests that epigenetic modifications working in concert with genetic mechanisms to regulate transcriptional activity are dysregulated in many diseases, including cancer. Cancer epigenetics explain a wide range of heritable changes in gene expression, which do not come from any alteration in DNA sequences. Aberrant DNA methylation, histone modifications, and expression of long non-coding RNAs (lncRNAs) are key epigenetic mechanisms associated with tumor initiation, cancer progression, and metastasis. Within the past decade, cancer epigenetics have enabled us to develop novel biomarkers and therapeutic target for many types of cancers. In this review, we will summarize the major epigenetic changes involved in cancer biology along with clinical and preclinical results developed as novel cancer therapeutics.
Collapse
Affiliation(s)
- Jong Woo Park
- Research Center for Epigenome Regulation, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jeung-Whan Han
- Research Center for Epigenome Regulation, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
61
|
Gao Y, Liu B, Feng L, Sun B, He S, Yang Y, Wu G, E G, Liu C, Gao Y, Zhang E, Zhu B. Targeting JUN, CEBPB, and HDAC3: A Novel Strategy to Overcome Drug Resistance in Hypoxic Glioblastoma. Front Oncol 2019; 9:33. [PMID: 30775317 PMCID: PMC6367651 DOI: 10.3389/fonc.2019.00033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 01/14/2019] [Indexed: 11/23/2022] Open
Abstract
Hypoxia is a predominant feature in glioblastoma (GBM) and contributes greatly to its drug resistance. However, the molecular mechanisms which are responsible for the development of the resistant phenotype of GBM under hypoxic conditions remain unclear. To analyze the key pathways promoting therapy resistance in hypoxic GBM, we utilized the U87-MG cell line as a human GBM cell model and the human brain HEB cell line as a non-neoplastic brain cell model. These cell lines were cultured in the presence of 21, 5, and 1% O2 for 24 h. We detected the changes in transcriptional profiling and analyzed the biological processes and functional interactions for the genes with different expression levels under different hypoxia conditions. The results indicated that those alterations of U87-MG cells presented specific transcriptional signature in response to diverse hypoxia levels. Gene ontology analysis revealed that the genes related to the DNA replication and cell cycle were suppressed, while the genes involved in tissue and system development to promote cancer development were activated following hypoxia. Moreover, functional interaction analysis suggested that the epigenetic regulator HDAC3 and the transcriptional factors CEBPB and JUN played a central role in organ and system developmental process pathway. Previous studies reported the global alterations caused by activation of HDAC3, CEBPB, and JUN could form the molecular basis of the resistance to chemotherapy and radiation therapy of hypoxic GBM. In our study, the significant growth inhibitory effect of temozolomide on hypoxic GBM cells could be promoted under downregulation of these genes. The experiment suggested that HDAC3, CEBPB, and JUN were closely involved in the drug-resistance phenotype of hypoxic GBM. In summary, we profiled the hypoxia-dependent changes in the transcriptome of the U87-MG cell line and the human brain cell line HEB to identify the transcriptional signatures of U87-MG cells and elucidate the role of hypoxia in the drug-resistant phenotype of GBM. Furthermore, we identified three key genes and explored their important roles in the drug resistance of hypoxic GBM.
Collapse
Affiliation(s)
- Yixing Gao
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Bao Liu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Lan Feng
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Binda Sun
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Shu He
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Yidong Yang
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Gang Wu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Guoji E
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Chang Liu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Yuqi Gao
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Erlong Zhang
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Bo Zhu
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
62
|
Biray Avci C, Goker Bagca B, Tetik Vardarli A, Saydam G, Gunduz C. Epigenetic modifications in chronic myeloid leukemia cells through ruxolitinib treatment. J Cell Biochem 2018; 120:4555-4563. [PMID: 30260022 DOI: 10.1002/jcb.27744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 08/31/2018] [Indexed: 12/28/2022]
Abstract
Chronic myeloid leukemia is a clonal malignancy of hematopoietic stem cell that is characterized by the occurrence of t(9;22)(q34;q11.2) translocation, named Philadelphia chromosome. Ruxolitinib is a powerful Janus tyrosine kinase 1 and 2 inhibitor that is used for myelofibrosis treatment. DNA-histone connection mediates a wide range of genes that code methylation, demethylation, acetylation, deacetylation, ubiquitination, and phosphorylation enzymes. Epigenetic modifications regulate chromatin compactness, which plays pivotal roles in critical biological processes including the transcriptional activity and cell proliferation as well as various pathological mechanisms, including CML. This study is aimed to determine the alterations of the expression levels of epigenetic modification-related genes after ruxolitinib treatment. Total RNA was isolated from K-562 cells treated with the IC50 value of ruxolitinib and untreated K-562 control cells. A reverse transcription procedure was performed for complementary DNA synthesis, and gene expressions were detected by real-time polymerase chain reaction compared with the untreated cells. Ruxolitinib treatment caused a significant alteration in the expression levels of epigenetic regulation-related genes in K-562 cells. Our novel results suggested that ruxolitinib has inhibitor effects on epigenetic modification-regulator genes.
Collapse
Affiliation(s)
- Cigir Biray Avci
- Department of Medical Biology, Medical Faculty, Ege University, Izmir, Turkey
| | - Bakiye Goker Bagca
- Department of Medical Biology, Medical Faculty, Ege University, Izmir, Turkey
| | - Asli Tetik Vardarli
- Department of Medical Biology, Medical Faculty, Ege University, Izmir, Turkey
| | - Guray Saydam
- Department of Internal Medicine, Division of Haematology, Medical Faculty, Ege University, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Medical Faculty, Ege University, Izmir, Turkey
| |
Collapse
|
63
|
Karabulutoglu M, Finnon R, Imaoka T, Friedl AA, Badie C. Influence of diet and metabolism on hematopoietic stem cells and leukemia development following ionizing radiation exposure. Int J Radiat Biol 2018; 95:452-479. [PMID: 29932783 DOI: 10.1080/09553002.2018.1490042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE The review aims to discuss the prominence of dietary and metabolic regulators in maintaining hematopoietic stem cell (HSC) function, long-term self-renewal, and differentiation. RESULTS Most adult stem cells are preserved in a quiescent, nonmotile state in vivo which acts as a "protective state" for stem cells to reduce endogenous stress provoked by DNA replication and cellular respiration as well as exogenous environmental stress. The dynamic balance between quiescence, self-renewal and differentiation is critical for supporting a functional blood system throughout life of an organism. Stress-conditions, for example ionizing radiation exposure can trigger the blood forming HSCs to proliferate and migrate through extramedullary tissues to expand the number of HSCs and increase hematopoiesis. In addition, a wealth of investigation validated that deregulation of this balance plays a critical pathogenic role in various different hematopoietic diseases including the leukemia development. CONCLUSION The review summarizes the current knowledge on how alterations in dietary and metabolic factors could alter the risk of leukemia development following ionizing radiation exposure by inhibiting or even reversing the leukemic progression. Understanding the influence of diet, metabolism, and epigenetics on radiation-induced leukemogenesis may lead to the development of practical interventions to reduce the risk in exposed populations.
Collapse
Affiliation(s)
- Melis Karabulutoglu
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK.,b CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
| | - Rosemary Finnon
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| | - Tatsuhiko Imaoka
- c Department of Radiation Effects Research, National Institute of Radiological Sciences , National Institutes for Quantum and Radiological Science and Technology , Chiba , Japan
| | - Anna A Friedl
- d Department of Radiation Oncology , University Hospital, LMU Munich , Munich , Germany
| | - Christophe Badie
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| |
Collapse
|
64
|
Vega-García N, Malatesta R, Estella C, Pérez-Jaume S, Esperanza-Cebollada E, Torrebadell M, Català A, Gassiot S, Berrueco R, Ruiz-Llobet A, Alonso-Saladrigues A, Mesegué M, Pont-Martí S, Rives S, Camós M. Paediatric patients with acute leukaemia andKMT2A (MLL)rearrangement show a distinctive expression pattern of histone deacetylases. Br J Haematol 2018; 182:542-553. [DOI: 10.1111/bjh.15436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Nerea Vega-García
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Roberta Malatesta
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Camino Estella
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Sara Pérez-Jaume
- Developmental Tumor Biology Laboratory; Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Elena Esperanza-Cebollada
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Montserrat Torrebadell
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
| | - Albert Català
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Susanna Gassiot
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Rubén Berrueco
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Anna Ruiz-Llobet
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Anna Alonso-Saladrigues
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Montserrat Mesegué
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Sandra Pont-Martí
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
| | - Susana Rives
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
- Paediatric Haematology and Oncology Department; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
| | - Mireia Camós
- Haematology Laboratory; Hospital Sant Joan de Déu Barcelona; University of Barcelona; Barcelona Spain
- Institut de Recerca Hospital Sant Joan de Déu Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER); Instituto de Salud Carlos III; Madrid Spain
| |
Collapse
|
65
|
Salgado E, Bian X, Feng A, Shim H, Liang Z. HDAC9 overexpression confers invasive and angiogenic potential to triple negative breast cancer cells via modulating microRNA-206. Biochem Biophys Res Commun 2018; 503:1087-1091. [PMID: 29936177 DOI: 10.1016/j.bbrc.2018.06.120] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/20/2018] [Indexed: 12/31/2022]
Abstract
Triple negative breast cancer (TNBC) is among the most aggressive breast cancer subtypes with poor prognosis. The purpose of this study is to better understand the molecular basis of TNBC as well as develop new therapeutic strategies. Our results demonstrate that HDAC9 is overexpressed in TNBC compared to non-TNBC cell lines and tissues and is inversely proportional with miR-206 expression levels. We show that HDAC9 selective inhibition blocked the invasion of TNBC cells in vitro and repressed the angiogenesis shown via in vivo Matrigel plug assays. Subsequent HDAC9 siRNA knockdown was then shown to restore miR-206 while also decreasing VEGF and MAPK3 levels. Furthermore, the inhibition of miR-206 neutralized the action of HDAC9 siRNA on decreasing VEGF and MAPK3 levels. This study highlights HDAC9 as a mediator of cell invasion and angiogenesis in TNBC cells through VEGF and MAPK3 by modulating miR-206 expression and suggests that selective inhibition of HDAC9 may be an efficient route for TNBC therapy.
Collapse
Affiliation(s)
- Eric Salgado
- Molecular and Systems Pharmacology Graduate Studies Program, Emory University, Atlanta, GA, 30322, USA; Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA
| | - Xuehai Bian
- Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA; Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Amber Feng
- Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA
| | - Hyunsuk Shim
- Molecular and Systems Pharmacology Graduate Studies Program, Emory University, Atlanta, GA, 30322, USA; Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| | - Zhongxing Liang
- Department of Radiation Oncology, Emory University, Atlanta, GA, 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
66
|
Singh AK, Bishayee A, Pandey AK. Targeting Histone Deacetylases with Natural and Synthetic Agents: An Emerging Anticancer Strategy. Nutrients 2018; 10:E731. [PMID: 29882797 PMCID: PMC6024317 DOI: 10.3390/nu10060731] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer initiation and progression are the result of genetic and/or epigenetic alterations. Acetylation-mediated histone/non-histone protein modification plays an important role in the epigenetic regulation of gene expression. Histone modification is controlled by the balance between histone acetyltransferase and (HAT) and histone deacetylase (HDAC) enzymes. Imbalance between the activities of these two enzymes is associated with various forms of cancer. Histone deacetylase inhibitors (HDACi) regulate the activity of HDACs and are being used in cancer treatment either alone or in combination with other chemotherapeutic drugs/radiotherapy. The Food and Drug Administration (FDA) has already approved four compounds, namely vorinostat, romidepsin, belinostat, and panobinostat, as HDACi for the treatment of cancer. Several other HDACi of natural and synthetic origin are under clinical trial for the evaluation of efficiency and side-effects. Natural compounds of plant, fungus, and actinomycetes origin, such as phenolics, polyketides, tetrapeptide, terpenoids, alkaloids, and hydoxamic acid, have been reported to show potential HDAC-inhibitory activity. Several HDACi of natural and dietary origin are butein, protocatechuic aldehyde, kaempferol (grapes, green tea, tomatoes, potatoes, and onions), resveratrol (grapes, red wine, blueberries and peanuts), sinapinic acid (wine and vinegar), diallyl disulfide (garlic), and zerumbone (ginger). HDACi exhibit their antitumor effect by the activation of cell cycle arrest, induction of apoptosis and autophagy, angiogenesis inhibition, increased reactive oxygen species generation causing oxidative stress, and mitotic cell death in cancer cells. This review summarizes the HDACs classification, their aberrant expression in cancerous tissue, structures, sources, and the anticancer mechanisms of HDACi, as well as HDACi that are either FDA-approved or under clinical trials.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Department of Biochemistry, University of Allahabad, Allahabad 211 002, Uttar Pradesh, India.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA.
| | - Abhay K Pandey
- Department of Biochemistry, University of Allahabad, Allahabad 211 002, Uttar Pradesh, India.
| |
Collapse
|
67
|
Huang Y, Jian W, Zhao J, Wang G. Overexpression of HDAC9 is associated with poor prognosis and tumor progression of breast cancer in Chinese females. Onco Targets Ther 2018; 11:2177-2184. [PMID: 29713186 PMCID: PMC5909784 DOI: 10.2147/ott.s164583] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Breast cancer represents a serious health issue among females. HDAC9 has been identified as an oncogene in human cancers. This study sought to assess the prognostic value and the biologic function of HDAC9 in breast cancer patients. METHODS Expression of HDAC9 in breast cancer tissues and cells was evaluated by quantitative real-time polymerase chain reaction. Kaplan-Meier survival analysis and Cox regression assay were conducted to explore the prognostic significance of HDAC9. Cell experiments were performed to investigate the effects of HDAC9 on the biologic behaviors of breast cancer cells. RESULTS Expression of HDAC9 was significantly upregulated in both cancerous tissues and cells compared with the normal controls (all P<0.05). Overexpression of HDAC9 was correlated with lymph node metastasis (P=0.021) and TNM stage (P=0.004). Patients with high HDAC9 had poor overall survival compared to those with low levels of HDAC9 (log-rank P<0.05). Elevated HDAC9 was found to be an independent prognostic factor for the patients (hazard ratio=2.996, 95% CI=1.611-5.572, P=0.001). According to the cell experiments, tumor cell proliferation, migration and invasion were suppressed by knockdown of HDAC9. CONCLUSION All data demonstrated that overexpression of HDAC9 serves as a prognostic biomarker and may be involved in the tumor progression of breast cancer.
Collapse
Affiliation(s)
- Yixiang Huang
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Wei Jian
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Junyong Zhao
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Gang Wang
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| |
Collapse
|
68
|
Abstract
Acute leukemias are hematologic malignancies with aggressive behavior especially in adult population. With the introduction of new gene expression and sequencing technologies there have been advances in the knowledge of the genetic landscape of acute leukemias. A more detailed analysis allows for the identification of additional alterations in epigenetic regulators that have a profound impact in cellular biology without changes in DNA sequence. These epigenetic alterations disturb the physiological balance between gene activation and gene repression and contribute to aberrant gene expression, contributing significantly to the leukemic pathogenesis and maintenance. We review epigenetic changes in acute leukemia in relation to what is known about their mechanism of action, their prognostic role and their potential use as therapeutic targets, with important implications for precision medicine.
Collapse
|
69
|
Hua WK, Qi J, Cai Q, Carnahan E, Ayala Ramirez M, Li L, Marcucci G, Kuo YH. HDAC8 regulates long-term hematopoietic stem-cell maintenance under stress by modulating p53 activity. Blood 2017; 130:2619-2630. [PMID: 29084772 PMCID: PMC5731083 DOI: 10.1182/blood-2017-03-771386] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 10/20/2017] [Indexed: 12/11/2022] Open
Abstract
The maintenance and functional integrity of long-term hematopoietic stem cells (LT-HSCs) is critical for lifelong hematopoietic regeneration. Histone deacetylases (HDACs) modulate acetylation of lysine residues, a protein modification important for regulation of numerous biological processes. Here, we show that Hdac8 is most highly expressed in the phenotypic LT-HSC population within the adult hematopoietic hierarchy. Using an Hdac8-floxed allele and a dual-fluorescence Cre reporter allele, largely normal hematopoietic differentiation capacity of Hdac8-deficient cells was observed. However, the frequency of phenotypic LT-HSC population was significantly higher shortly after Hdac8 deletion, and the expansion had shifted to the phenotypic multipotent progenitor population by 1 year. We show that Hdac8-deficient hematopoietic progenitors are compromised in colony-forming cell serial replating in vitro and long-term serial repopulating activity in vivo. Mechanistically, we demonstrate that the HDAC8 protein interacts with the p53 protein and modulates p53 activity via deacetylation. Hdac8-deficient LT-HSCs displayed hyperactivation of p53 and increased apoptosis under genotoxic and hematopoietic stress. Genetic inactivation of p53 reversed the increased apoptosis and elevated expression of proapoptotic targets Noxa and Puma seen in Hdac8-deleted LT-HSCs. Dramatically compromised hematopoietic recovery and increased lethality were seen in Hdac8-deficient mice challenged with serial 5-fluorouracil treatment. This hypersensitivity to hematopoietic ablation was completely rescued by inactivation of p53. Altogether, these results indicate that HDAC8 functions to modulate p53 activity to ensure LT-HSC maintenance and cell survival under stress.
Collapse
Affiliation(s)
- Wei-Kai Hua
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Jing Qi
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Qi Cai
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Emily Carnahan
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Maria Ayala Ramirez
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Ling Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Guido Marcucci
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Ya-Huei Kuo
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| |
Collapse
|
70
|
Pham HTT, Maurer B, Prchal-Murphy M, Grausenburger R, Grundschober E, Javaheri T, Nivarthi H, Boersma A, Kolbe T, Elabd M, Halbritter F, Pencik J, Kazemi Z, Grebien F, Hengstschläger M, Kenner L, Kubicek S, Farlik M, Bock C, Valent P, Müller M, Rülicke T, Sexl V, Moriggl R. STAT5BN642H is a driver mutation for T cell neoplasia. J Clin Invest 2017; 128:387-401. [PMID: 29200404 PMCID: PMC5749501 DOI: 10.1172/jci94509] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/05/2017] [Indexed: 01/07/2023] Open
Abstract
STAT5B is often mutated in hematopoietic malignancies. The most frequent STAT5B mutation, Asp642His (N642H), has been found in over 90 leukemia and lymphoma patients. Here, we used the Vav1 promoter to generate transgenic mouse models that expressed either human STAT5B or STAT5BN642H in the hematopoietic compartment. While STAT5B-expressing mice lacked a hematopoietic phenotype, the STAT5BN642H-expressing mice rapidly developed T cell neoplasms. Neoplasia manifested as transplantable CD8+ lymphoma or leukemia, indicating that the STAT5BN642H mutation drives cancer development. Persistent and enhanced levels of STAT5BN642H tyrosine phosphorylation in transformed CD8+ T cells led to profound changes in gene expression that were accompanied by alterations in DNA methylation at potential histone methyltransferase EZH2-binding sites. Aurora kinase genes were enriched in STAT5BN642H-expressing CD8+ T cells, which were exquisitely sensitive to JAK and Aurora kinase inhibitors. Together, our data suggest that JAK and Aurora kinase inhibitors should be further explored as potential therapeutics for lymphoma and leukemia patients with the STAT5BN642H mutation who respond poorly to conventional chemotherapy.
Collapse
Affiliation(s)
- Ha Thi Thanh Pham
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Maurer
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eva Grundschober
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tahereh Javaheri
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Harini Nivarthi
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Thomas Kolbe
- Biomodels Austria (Biat), University of Veterinary Medicine Vienna, Vienna, Austria.,IFA-Tulln, University of Natural Resources and Life Sciences, Tulln, Austria
| | - Mohamed Elabd
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Florian Halbritter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Jan Pencik
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Zahra Kazemi
- Medical University of Vienna, Vienna, Austria.,Center of Physiology and Pharmacology, Vienna, Austria
| | - Florian Grebien
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Markus Hengstschläger
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.,Unit of Pathology of Laboratory Animals, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Medical University of Vienna, Vienna, Austria.,Max Planck Institute for Informatics, Saarbrücken, Germany
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, and.,Ludwig Boltzmann-Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.,Medical University of Vienna, Vienna, Austria
| |
Collapse
|
71
|
Lernoux M, Schnekenburger M, Dicato M, Diederich M. Anti-cancer effects of naturally derived compounds targeting histone deacetylase 6-related pathways. Pharmacol Res 2017; 129:337-356. [PMID: 29133216 DOI: 10.1016/j.phrs.2017.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/02/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
Abstract
Alterations of the epigenetic machinery, affecting multiple biological functions, represent a major hallmark enabling the development of tumors. Among epigenetic regulatory proteins, histone deacetylase (HDAC)6 has emerged as an interesting potential therapeutic target towards a variety of diseases including cancer. Accordingly, this isoenzyme regulates many vital cellular regulatory processes and pathways essential to physiological homeostasis, as well as tumor multistep transformation involving initiation, promotion, progression and metastasis. In this review, we will consequently discuss the critical implications of HDAC6 in distinct mechanisms relevant to physiological and cancerous conditions, as well as the anticancer properties of synthetic, natural and natural-derived compounds through the modulation of HDAC6-related pathways.
Collapse
Affiliation(s)
- Manon Lernoux
- Laboratory of Molecular and Cellular Biology of Cancer, Kirchberg Hospital, 9, Edward Steichen Street, L-2540 Luxembourg, Luxembourg
| | - Michael Schnekenburger
- Laboratory of Molecular and Cellular Biology of Cancer, Kirchberg Hospital, 9, Edward Steichen Street, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratory of Molecular and Cellular Biology of Cancer, Kirchberg Hospital, 9, Edward Steichen Street, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, 08826, South Korea.
| |
Collapse
|
72
|
Lei Y, Liu L, Zhang S, Guo S, Li X, Wang J, Su B, Fang Y, Chen X, Ke H, Tao W. Hdac7 promotes lung tumorigenesis by inhibiting Stat3 activation. Mol Cancer 2017; 16:170. [PMID: 29126425 PMCID: PMC5681774 DOI: 10.1186/s12943-017-0736-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 10/20/2017] [Indexed: 11/29/2022] Open
Abstract
Background Lung cancer is the leading cause of cancer death worldwide. However, the molecular mechanisms underlying lung cancer development have not been fully understood. The functions of histone deacetylases (HDACs), a class of total eighteen proteins (HDAC1–11 and SIRT1–7 in mammals) that deacetylate histones and non-histone proteins, in cancers are largely unknown. Methods Hdac7+/−/K-Ras mice and HDAC7-depleted human lung cancer cell lines were used as models for studying the function of Hdac7 gene in lung cancer. Kaplan-Meier survival analysis was performed to explore the relationship between HDAC7 expression and prognosis of human lung cancers. Recombinant lentivirus-mediated in vivo gene expression or knockdown, Western blotting, and pull-down assay were applied to investigate the underlying molecular mechanism by which Hdac7 promotes lung tumorigenesis. Results The number and burden of lung tumor were dramatically reduced in Hdac7+/−/K-Ras mice compared to control K-Ras mice. Also, in Hdac7+/−/K-Ras mice, cell proliferation was significantly inhibited and apoptosis in lung tumors was greatly enhanced. Similarly, cell proliferation and anchorage-independent growth of human lung cancer cell lines expressing shHDAC7 were also significantly suppressed and apoptosis was dramatically elevated respectively. Mechanistic study revealed that Hdac7 mutation in mouse lung tumors or HDAC7 depletion in human tumor cell lines resulted in significantly enhanced acetylation and tyrosine-phosphorylation of Stat3 and HDAC7 protein directly interacted with and deacetylateed STAT3. The Hdac7 mutant-mediated inhibitory effects on lung tumorigenesis in mice and cell proliferation/soft agar colony formation of human lung cancer cell lines were respectively reversed by expressing dnStat3. Finally, the high HDAC7 mRNA level was found to be correlated with poor prognosis of human lung cancer patients. Conclusion Our study suggests that Hdac7 promotes lung tumorigenesis by inhibiting Stat3 activation via deacetylating Stat3 and may shed a light on the design of new therapeutic strategies for human lung cancer. Electronic supplementary material The online version of this article (10.1186/s12943-017-0736-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yubin Lei
- Obstetrics & Gynecology Hospital and State Key Laboratory of Genetic Engineering and Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Lingling Liu
- Obstetrics & Gynecology Hospital and State Key Laboratory of Genetic Engineering and Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shujing Zhang
- Obstetrics & Gynecology Hospital and State Key Laboratory of Genetic Engineering and Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shicheng Guo
- MOE Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoqing Li
- Obstetrics & Gynecology Hospital and State Key Laboratory of Genetic Engineering and Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- MOE Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Bo Su
- Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yuchao Fang
- Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaofeng Chen
- Huashan Hospital, Fudan University, Shanghai, China.
| | - Hengning Ke
- Cancer Research Institute, General Hospital, Ningxia Medical University, Yinchuan, China.
| | - Wufan Tao
- Obstetrics & Gynecology Hospital and State Key Laboratory of Genetic Engineering and Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China. .,Cancer Research Institute, General Hospital, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
73
|
Histone modifications: A review about the presence of this epigenetic phenomenon in carcinogenesis. Pathol Res Pract 2017; 213:1329-1339. [DOI: 10.1016/j.prp.2017.06.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/23/2017] [Accepted: 06/24/2017] [Indexed: 12/26/2022]
|
74
|
Jamalpour M, Li X, Cavelier L, Gustafsson K, Mostoslavsky G, Höglund M, Welsh M. Tumor SHB gene expression affects disease characteristics in human acute myeloid leukemia. Tumour Biol 2017; 39:1010428317720643. [DOI: 10.1177/1010428317720643] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Maria Jamalpour
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Xiujuan Li
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Lucia Cavelier
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Karin Gustafsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Gustavo Mostoslavsky
- Center for Regenerative Medicine (CReM), Department of Medicine, School of Medicine, Boston University, Boston, MA, USA
| | - Martin Höglund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael Welsh
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
75
|
Navarrete-Meneses MDP, Pérez-Vera P. Alteraciones epigenéticas en leucemia linfoblástica aguda. BOLETIN MEDICO DEL HOSPITAL INFANTIL DE MEXICO 2017; 74:243-264. [DOI: 10.1016/j.bmhimx.2017.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/04/2017] [Accepted: 02/08/2017] [Indexed: 12/22/2022] Open
|
76
|
|
77
|
Targeting HDAC3, a new partner protein of AKT in the reversal of chemoresistance in acute myeloid leukemia via DNA damage response. Leukemia 2017; 31:2761-2770. [PMID: 28462918 DOI: 10.1038/leu.2017.130] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/06/2017] [Accepted: 04/17/2017] [Indexed: 01/21/2023]
Abstract
Resistance to cytotoxic chemotherapy drugs remains as the major cause of treatment failure in acute myeloid leukemia. Histone deacetylases (HDAC) are important regulators to maintain chromatin structure and control DNA damage; nevertheless, how each HDAC regulates genome stability remains unclear, especially under genome stress conditions. Here, we identified a mechanism by which HDAC3 regulates DNA damage repair and mediates resistance to chemotherapy drugs. In addition to inducing DNA damage, chemotherapy drugs trigger upregulation of HDAC3 expression in leukemia cells. Using genetic and pharmacological approaches, we show that HDAC3 contributes to chemotherapy resistance by regulating the activation of AKT, a well-documented factor in drug resistance development. HDAC3 binds to AKT and deacetylates it at the site Lys20, thereby promoting the phosphorylation of AKT. Chemotherapy drug exposure enhances the interaction between HDAC3 and AKT, resulting in decrease in AKT acetylation and increase in AKT phosphorylation. Whereas HDAC3 depletion or inhibition abrogates these responses and meanwhile sensitizes leukemia cells to chemotoxicity-induced apoptosis. Importantly, in vivo HDAC3 suppression reduces leukemia progression and sensitizes MLL-AF9+ leukemia to chemotherapy. Our findings suggest that combination therapy with HDAC3 inhibitor and genotoxic agents may constitute a successful strategy for overcoming chemotherapy resistance.
Collapse
|
78
|
Wu C, Zhang D. Identification of early-stage lung adenocarcinoma prognostic signatures based on statistical modeling. Cancer Biomark 2017; 18:117-123. [PMID: 27935544 DOI: 10.3233/cbm-151368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Current staging methods are lack of precision in predicting prognosis of early-stage lung adenocarcinomas. OBJECTIVE We aimed to develop a gene expression signature to identify high- and low-risk groups of patients. METHODS We used the Bayesian Model Averaging algorithm to analyze the DNA microarray data from 442 lung adenocarcinoma patients from three independent cohorts, one of which was used for training. RESULTS The patients were assigned to either high- or low-risk groups based on the calculated risk scores based on the identified 25-gene signature. The prognostic power was evaluated using Kaplan-Meier analysis and the log-rank test. The testing sets were divided into two distinct groups with log-rank test p-values of 0.00601 and 0.0274 respectively. CONCLUSIONS Our results show that the prognostic models could successfully predict patients' outcome and serve as biomarkers for early-stage lung adenocarcinoma overall survival analysis.
Collapse
Affiliation(s)
- Chunxiao Wu
- Department of Thoracic Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.,Department of Thoracic Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Donglei Zhang
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China.,Department of Thoracic Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
79
|
Design, synthesis and anti-tumor activity study of novel histone deacetylase inhibitors containing isatin-based caps and o-phenylenediamine-based zinc binding groups. Bioorg Med Chem 2017; 25:2981-2994. [PMID: 28511906 DOI: 10.1016/j.bmc.2017.03.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/12/2017] [Accepted: 03/14/2017] [Indexed: 11/23/2022]
Abstract
As a hot topic of epigenetic studies, histone deacetylases (HDACs) are related to lots of diseases, especially cancer. Further researches indicated that different HDAC isoforms played various roles in a wide range of tumor types. Herein a novel series of HDAC inhibitors with isatin-based caps and o-phenylenediamine-based zinc binding groups have been designed and synthesized through scaffold hopping strategy. Among these compounds, the most potent compound 9n exhibited similar if not better HDAC inhibition and antiproliferative activities against multiple tumor cell lines compared with the positive control entinostat (MS-275). Additionally, compared with MS-275 (IC50 values for HDAC1, 2 and 3 were 0.163, 0.396 and 0.605µM, respectively), compound 9n with IC50 values of 0.032, 0.256 and 0.311µM for HDAC1, 2 and 3 respectively, showed a moderate HDAC1 selectivity.
Collapse
|
80
|
Yu Y, Cao F, Yu X, Zhou P, Di Q, Lei J, Tai Y, Wu H, Li X, Wang X, Zhang W, Li P, Li Y. The expression of HDAC7 in cancerous gastric tissues is positively associated with distant metastasis and poor patient prognosis. Clin Transl Oncol 2017; 19:1045-1054. [PMID: 28299580 DOI: 10.1007/s12094-017-1639-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 02/27/2017] [Indexed: 12/13/2022]
Abstract
PURPOSE To characterize the expression patterns of HDAC7 in patients with gastric cancer and evaluate the prognostic value of HDAC7 in gastric cancer. METHODS The expression of histone deacetylase 7 (HDAC7) was detected in paraffin-embedded gastric cancer samples from 86 patients by immunohistochemistry, and the differences in the expression of HDAC7 between cancerous and corresponding adjacent noncancerous tissues were compared using the Wilcoxon matched-pairs signed rank test. The correlation between HDAC7 expression and Ki-67 expression or clinicopathologic characteristics was evaluated using a Spearman rank correlation test. Prognostic outcomes that correlated with HDAC7 were examined using a Kaplan-Meier analysis and Cox proportional hazards model. Moreover, the effects of HDAC7 on the proliferation, migration and invasion of gastric cancer cells were investigated in vitro using human gastric carcinoma AGS cells. RESULTS We found that HDAC7 was downregulated in cancerous gastric tissues (P = 0.0019). However, the expression of HDAC7 in cancerous gastric tissues positively correlated with Ki-67 expression (P = 0.0325) and distant metastasis (P = 0.020). Moreover, overall survival was shorter for patients expressing higher levels of HDAC7 in cancerous tissues (P = 0.042). Mechanistically, the disruption of the HDAC7 gene attenuated the capacity of cell growth, migration and invasion and induced G0/G1 arrest in AGS cells. Conversely, forced ovperexpression of HDAC7 promoted cell growth, migration and invasion and G1/S transition in AGS cells. CONCLUSIONS These results indicate that high HDAC7 expression in cancerous gastric tissues correlates with distant metastasis and predicts a poor prognosis for patients with gastric cancer.
Collapse
Affiliation(s)
- Y Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - F Cao
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - X Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - P Zhou
- Department of Medical Laborotary, The First Affiliated Hospital, Nanchang University, 330006, Nanchang, China
| | - Q Di
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - J Lei
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - Y Tai
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - H Wu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - X Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - X Wang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China
| | - W Zhang
- Department of Medical Affairs, Renmin Hospital, Hubei University of Medicine, 442000, Shiyan, China
| | - P Li
- Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, China.
| | - Y Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Rd., 442000, Shiyan, China.
| |
Collapse
|
81
|
Abstract
In the past few years, it has become clear that mutations in epigenetic regulatory genes are common in human cancers. Therapeutic strategies are now being developed to target cancers with mutations in these genes using specific chemical inhibitors. In addition, a complementary approach based on the concept of synthetic lethality, which allows exploitation of loss-of-function mutations in cancers that are not targetable by conventional methods, has gained traction. Both of these approaches are now being tested in several clinical trials. In this Review, we present recent advances in epigenetic drug discovery and development, and suggest possible future avenues of investigation to drive progress in this area.
Collapse
|
82
|
Rastogi B, Kumar A, Raut SK, Panda NK, Rattan V, Joshi N, Khullar M. Downregulation of miR-377 Promotes Oral Squamous Cell Carcinoma Growth and Migration by Targeting HDAC9. Cancer Invest 2017; 35:152-162. [PMID: 28267394 DOI: 10.1080/07357907.2017.1286669] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
microRNAs are the post-transcriptional regulators implicated in the initiation and progression of various cancer types, including oral squamous cell carcinoma (OSCC). Here, we investigated the role of miR-377 in OSCC tumorigenesis. miR-377 expression was reduced in OSCC samples and cell line (UPCI-SCC-116), and was associated with patient survival. In vitro restoration of miR-377 repressed cell growth, induced apoptosis, and reduced cell migration. We identified HDAC9 as a target of miR-377 and found miR-377 to regulate HDAC9 and its pro-apoptotic target, NR4A1/Nur77. Our findings show that miR-377 targets HDAC9 pathway in OSCC, suggesting that miR-377-HDAC9 axis may provide a novel therapeutic target for OSCC therapy.
Collapse
Affiliation(s)
- Bhawna Rastogi
- a Department of Otolaryngology and Head and Neck Surgery , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| | - Amit Kumar
- b Department of Experimental Medicine and Biotechnology , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| | - Satish K Raut
- b Department of Experimental Medicine and Biotechnology , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| | - Naresh K Panda
- a Department of Otolaryngology and Head and Neck Surgery , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| | - Vidya Rattan
- c Department of Oral Health Sciences Centre , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| | - Nainesh Joshi
- b Department of Experimental Medicine and Biotechnology , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| | - Madhu Khullar
- b Department of Experimental Medicine and Biotechnology , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| |
Collapse
|
83
|
Zhang H, Shang YP, Chen HY, Li J. Histone deacetylases function as novel potential therapeutic targets for cancer. Hepatol Res 2017; 47:149-159. [PMID: 27457249 DOI: 10.1111/hepr.12757] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 05/29/2016] [Accepted: 05/31/2016] [Indexed: 12/12/2022]
Abstract
Diverse cellular functions, including tumor suppressor gene expression, DNA repair, cell proliferation and apoptosis, are regulated by histone acetylation and deacetylation. Histone deacetylases (HDACs) are enzymes involved in remodeling of chromatin by deacetylating the lysine residues. They play a pivotal role in epigenetic regulation of gene expression. Dysregulation of HDACs and aberrant chromatin acetylation and deacetylation have been implicated in the pathogenesis of various diseases, including cancer. Histone deacetylases have become a target for the development of drugs for treating cancer because of their major contribution to oncogenic cell transformation. Overexpression of HDACs correlates with tumorigenesis. Previous work showed that inhibition of HDACs results in apoptosis and the inhibition of cell proliferation in multiple cells. A significant number of HDAC inhibitors have been developed in the past decade. These inhibitors have strong anticancer effects in vitro and in vivo, inducing growth arrest, differentiation, and programmed cell death, inhibiting cell migration, invasion, and metastasis, and suppressing angiogenesis. In addition, HDAC-mediated deacetylation alters the transcriptional activity of nuclear transcription factors, including p53, E2F, c-Myc, and nuclear factor-κB, as well as the extracellular signal-regulated kinase1/2, phosphatidylinositol 3-kinase, Notch, and Wnt signaling pathways. This review highlights the role of HDACs in cancer pathogenesis and, more importantly, that HDACs are potential novel therapeutic targets.
Collapse
Affiliation(s)
- Hui Zhang
- Anhui Provincial Cancer Hospital and West Branch of Anhui Provincial Hospital
| | - Yu-Ping Shang
- Anhui Provincial Cancer Hospital and West Branch of Anhui Provincial Hospital
| | - Hong-Ying Chen
- Anhui Provincial Cancer Hospital and West Branch of Anhui Provincial Hospital
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
84
|
The Role of Histone Protein Modifications and Mutations in Histone Modifiers in Pediatric B-Cell Progenitor Acute Lymphoblastic Leukemia. Cancers (Basel) 2017; 9:cancers9010002. [PMID: 28054944 PMCID: PMC5295773 DOI: 10.3390/cancers9010002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/14/2016] [Accepted: 12/23/2016] [Indexed: 12/18/2022] Open
Abstract
While cancer has been long recognized as a disease of the genome, the importance of epigenetic mechanisms in neoplasia was acknowledged more recently. The most active epigenetic marks are DNA methylation and histone protein modifications and they are involved in basic biological phenomena in every cell. Their role in tumorigenesis is stressed by recent unbiased large-scale studies providing evidence that several epigenetic modifiers are recurrently mutated or frequently dysregulated in multiple cancers. The interest in epigenetic marks is especially due to the fact that they are potentially reversible and thus druggable. In B-cell progenitor acute lymphoblastic leukemia (BCP-ALL) there is a relative paucity of reports on the role of histone protein modifications (acetylation, methylation, phosphorylation) as compared to acute myeloid leukemia, T-cell ALL, or other hematologic cancers, and in this setting chromatin modifications are relatively less well studied and reviewed than DNA methylation. In this paper, we discuss the biomarker associations and evidence for a driver role of dysregulated global and loci-specific histone marks, as well as mutations in epigenetic modifiers in BCP-ALL. Examples of chromatin modifiers recurrently mutated/disrupted in BCP-ALL and associated with disease outcomes include MLL1, CREBBP, NSD2, and SETD2. Altered histone marks and histone modifiers and readers may play a particular role in disease chemoresistance and relapse. We also suggest that epigenetic regulation of B-cell differentiation may have parallel roles in leukemogenesis.
Collapse
|
85
|
Hutt DM, Roth DM, Marchal C, Bouchecareilh M. Using Histone Deacetylase Inhibitors to Analyze the Relevance of HDACs for Translation. Methods Mol Biol 2017; 1510:77-91. [PMID: 27761814 DOI: 10.1007/978-1-4939-6527-4_6] [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] [Indexed: 06/06/2023]
Abstract
Gene expression is regulated in part through the reversible acetylation of histones, by the action of histone acetyltransferases (HAT) and histone deacetylases (HDAC). HAT activity results in the addition of acetyl groups on the lysine residues of histone tails leading to decondensation of the chromatin, and increased gene transcription in general, whereas HDACs remove these acetyl groups, thus leading to an overall suppression of gene transcription. Recent evidence has elucidated that histones are not the only components of the proteome that are targeted by HATs and HDACs. A large number of nonhistone proteins undergo posttranslational acetylation. They include proteins involved in mRNA stability, protein localization and degradation, as well as protein-protein and protein-DNA interactions. In recent years, numerous studies have discovered increased HDAC expression and/or activity in numerous disease states, including cancer, where the upregulation of HDAC family members leads to dysregulation of genes and proteins involved in cell proliferation, cell cycle regulation, and apoptosis. These observations have pushed HDAC inhibitors (HDACi) to the forefront of therapeutic development of oncological conditions. HDACi, such as Vorinostat (Suberoylanilide hydroxamic acid (SAHA)), affect cancer cells in part by suppressing the translation of key proteins linked to tumorigenesis, such as cyclin D1 and hypoxia inducible factor 1 alpha (HIF-1α). Herein we describe methodologies to analyze the impact of the HDACi Vorinostat on HIF-1α translational regulation and downstream effectors.
Collapse
MESH Headings
- Acetylation
- Blotting, Western/methods
- Cell Line, Tumor
- Chromatin/chemistry
- Chromatin/drug effects
- Chromatin/metabolism
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Deferoxamine/pharmacology
- Eukaryotic Initiation Factor-3/antagonists & inhibitors
- Eukaryotic Initiation Factor-3/genetics
- Eukaryotic Initiation Factor-3/metabolism
- Gene Expression Regulation, Neoplastic
- Glycine/analogs & derivatives
- Glycine/pharmacology
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Histone Acetyltransferases/genetics
- Histone Acetyltransferases/metabolism
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylases/genetics
- Histone Deacetylases/metabolism
- Histones/genetics
- Histones/metabolism
- Humans
- Hydroxamic Acids/pharmacology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Leupeptins/pharmacology
- Protein Biosynthesis/drug effects
- Protein Processing, Post-Translational
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Vorinostat
Collapse
Affiliation(s)
- Darren M Hutt
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Daniela Martino Roth
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Christelle Marchal
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, Bordeaux, 33077, France
| | - Marion Bouchecareilh
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, Bordeaux, 33077, France.
| |
Collapse
|
86
|
Winters AC, Bernt KM. MLL-Rearranged Leukemias-An Update on Science and Clinical Approaches. Front Pediatr 2017; 5:4. [PMID: 28232907 PMCID: PMC5299633 DOI: 10.3389/fped.2017.00004] [Citation(s) in RCA: 265] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/09/2017] [Indexed: 12/18/2022] Open
Abstract
The mixed-lineage leukemia 1 (MLL1) gene (now renamed Lysine [K]-specific MethylTransferase 2A or KMT2A) on chromosome 11q23 is disrupted in a unique group of acute leukemias. More than 80 different partner genes in these fusions have been described, although the majority of leukemias result from MLL1 fusions with one of about six common partner genes. Approximately 10% of all leukemias harbor MLL1 translocations. Of these, two patient populations comprise the majority of cases: patients younger than 1 year of age at diagnosis (primarily acute lymphoblastic leukemias) and young- to-middle-aged adults (primarily acute myeloid leukemias). A much rarer subgroup of patients with MLL1 rearrangements develop leukemia that is attributable to prior treatment with certain chemotherapeutic agents-so-called therapy-related leukemias. In general, outcomes for all of these patients remain poor when compared to patients with non-MLL1 rearranged leukemias. In this review, we will discuss the normal biological roles of MLL1 and its fusion partners, how these roles are hypothesized to be dysregulated in the context of MLL1 rearrangements, and the clinical manifestations of this group of leukemias. We will go on to discuss the progress in clinical management and promising new avenues of research, which may lead to more effective targeted therapies for affected patients.
Collapse
Affiliation(s)
- Amanda C Winters
- Division of Pediatric Hematology/Oncology/BMT, University of Colorado School of Medicine and Children's Hospital Colorado , Aurora, CO , USA
| | - Kathrin M Bernt
- Division of Pediatric Hematology/Oncology/BMT, University of Colorado School of Medicine and Children's Hospital Colorado , Aurora, CO , USA
| |
Collapse
|
87
|
Gil VS, Bhagat G, Howell L, Zhang J, Kim CH, Stengel S, Vega F, Zelent A, Petrie K. Deregulated expression of HDAC9 in B cells promotes development of lymphoproliferative disease and lymphoma in mice. Dis Model Mech 2016; 9:1483-1495. [PMID: 27799148 PMCID: PMC5200892 DOI: 10.1242/dmm.023366] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022] Open
Abstract
Histone deacetylase 9 (HDAC9) is expressed in B cells, and its overexpression has been observed in B-lymphoproliferative disorders, including B-cell non-Hodgkin lymphoma (B-NHL). We examined HDAC9 protein expression and copy number alterations in primary B-NHL samples, identifying high HDAC9 expression among various lymphoma entities and HDAC9 copy number gains in 50% of diffuse large B-cell lymphoma (DLBCL). To study the role of HDAC9 in lymphomagenesis, we generated a genetically engineered mouse (GEM) model that constitutively expressed an HDAC9 transgene throughout B-cell development under the control of the immunoglobulin heavy chain (IgH) enhancer (Eμ). Here, we report that the Eμ-HDAC9 GEM model develops splenic marginal zone lymphoma and lymphoproliferative disease (LPD) with progression towards aggressive DLBCL, with gene expression profiling supporting a germinal center cell origin, as is also seen in human B-NHL tumors. Analysis of Eμ-HDAC9 tumors suggested that HDAC9 might contribute to lymphomagenesis by altering pathways involved in growth and survival, as well as modulating BCL6 activity and p53 tumor suppressor function. Epigenetic modifications play an important role in the germinal center response, and deregulation of the B-cell epigenome as a consequence of mutations and other genomic aberrations are being increasingly recognized as important steps in the pathogenesis of a variety of B-cell lymphomas. A thorough mechanistic understanding of these alterations will inform the use of targeted therapies for these malignancies. These findings strongly suggest a role for HDAC9 in B-NHL and establish a novel GEM model for the study of lymphomagenesis and, potentially, preclinical testing of therapeutic approaches based on histone deacetylase inhibitors. Summary: This study demonstrates that aberrant expression of HDAC9 in B cells promotes development of lymphoproliferative disease and lymphoma through altering expression of genes involved in the cell cycle and survival, and modulating the activity of key B-lineage factors such as BCL6 and p53.
Collapse
Affiliation(s)
- Veronica S Gil
- Division of Clinical Studies, Institute of Cancer Research, London SM2 5NG, UK
| | - Govind Bhagat
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Pathology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Louise Howell
- Division of Molecular Pathology, Institute of Cancer Research, London SM2 5NG, UK
| | - Jiyuan Zhang
- Department of Pathology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA.,Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Chae H Kim
- Division of Hematopathology, Sylvester Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Sven Stengel
- Division of Molecular Pathology, Institute of Cancer Research, London SM2 5NG, UK
| | - Francisco Vega
- Division of Hematopathology, Sylvester Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Arthur Zelent
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Kevin Petrie
- Department of Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| |
Collapse
|
88
|
Pruitt K. Molecular and Cellular Changes During Cancer Progression Resulting From Genetic and Epigenetic Alterations. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:3-47. [PMID: 27865461 DOI: 10.1016/bs.pmbts.2016.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumorigenesis is a complex process that involves a persistent dismantling of cellular safeguards and checkpoints. These molecular and cellular changes that accumulate over months or decades lead to a change in the fundamental identity of a cell as it transitions from normal to malignant. In this chapter, we will examine some of the molecular changes in the evolving relationship between the genome and epigenome and highlight some of the key changes that occur as normal cells progress to tumor cells. For many years tumorigenesis was almost exclusively attributed to mutations in protein-coding genes. This notion that mutations in protein-coding genes were a fundamental driver of tumorigenesis enabled the development of several novel therapeutics that targeted the mutant protein or overactive pathway responsible for driving a significant portion of the tumor growth. However, because many therapeutic challenges remained in the face of these advances, it was clear that other pieces to the puzzle had yet to be discovered. Advances in molecular and genomics techniques continued and the study of epigenetics began to expand and helped reshape the view that drivers of tumorigenesis extended beyond mutations in protein-coding genes. Studies in the field of epigenetics began to identify aberrant epigenetic marks which created altered chromatin structures and enabled protein expression in tissues that defied rules governing tissue-specificity. Not only were epigenetic alterations found to enable overexpression of proto-oncogenes, they also led to the silencing of tumor suppressor genes. With these discoveries, it became clear that tumor growth could be stimulated by much more than mutations in protein-coding genes. In fact, it became increasingly clear that much of the human genome, while transcribed, did not lead to proteins. This discovery further led to studies that began to uncover the role of noncoding RNAs in regulating chromatin structure, gene transcription, and tumor biology. In this chapter, some of the key alterations in the genome and epigenome will be explored, and some of the cancer therapies that were developed as a result of these discoveries will be discussed.
Collapse
Affiliation(s)
- K Pruitt
- Texas Tech University Health Sciences Center, Lubbock, TX, United States.
| |
Collapse
|
89
|
Li Y, Seto E. HDACs and HDAC Inhibitors in Cancer Development and Therapy. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a026831. [PMID: 27599530 DOI: 10.1101/cshperspect.a026831] [Citation(s) in RCA: 773] [Impact Index Per Article: 96.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the last several decades, it has become clear that epigenetic abnormalities may be one of the hallmarks of cancer. Posttranslational modifications of histones, for example, may play a crucial role in cancer development and progression by modulating gene transcription, chromatin remodeling, and nuclear architecture. Histone acetylation, a well-studied posttranslational histone modification, is controlled by the opposing activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). By removing acetyl groups, HDACs reverse chromatin acetylation and alter transcription of oncogenes and tumor suppressor genes. In addition, HDACs deacetylate numerous nonhistone cellular substrates that govern a wide array of biological processes including cancer initiation and progression. This review will discuss the role of HDACs in cancer and the therapeutic potential of HDAC inhibitors (HDACi) as emerging drugs in cancer treatment.
Collapse
Affiliation(s)
- Yixuan Li
- George Washington University Cancer Center, Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC 20037
| | - Edward Seto
- George Washington University Cancer Center, Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC 20037
| |
Collapse
|
90
|
Suzuki K, Okuno Y, Kawashima N, Muramatsu H, Okuno T, Wang X, Kataoka S, Sekiya Y, Hamada M, Murakami N, Kojima D, Narita K, Narita A, Sakaguchi H, Sakaguchi K, Yoshida N, Nishio N, Hama A, Takahashi Y, Kudo K, Kato K, Kojima S. MEF2D-BCL9 Fusion Gene Is Associated With High-Risk Acute B-Cell Precursor Lymphoblastic Leukemia in Adolescents. J Clin Oncol 2016; 34:3451-9. [PMID: 27507882 DOI: 10.1200/jco.2016.66.5547] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Acute lymphoblastic leukemia (ALL) makes up a significant proportion of all pediatric cancers, and relapsed ALL is a leading cause of cancer-associated deaths in children. Identification of risk factors and druggable molecular targets in ALL can lead to a better stratification of treatments and subsequent improvement in prognosis. PATIENTS AND METHODS We enrolled 59 children with relapsed or primary refractory ALL who were treated in our institutions. We primarily performed RNA sequencing (RNA-seq) using patients' leukemic cells to comprehensively detect gene fusions and analyze gene expression profiles. On the basis of results obtained by RNA-seq, we performed genetic validation, functional analysis, and in vitro drug sensitivity testing using patients' samples and an exogenous expression model. RESULTS We identified a total of 26 gene fusions in 22 patients by RNA-seq. Among these, 19 were nonrandom gene fusions already described in ALL, and four of the remaining seven involved identical combination of MEF2D and BCL9. All MEF2D-BCL9-positive patients had B-cell precursor immunophenotype and were characterized as being older in age, being resistant to chemotherapy, having very early relapse, and having leukemic blasts that mimic morphologically mature B-cell leukemia with markedly high expression of HDAC9. Exogenous expression of MEF2D-BCL9 in a B-cell precursor ALL cell line promoted cell growth, increased HDAC9 expression, and induced resistance to dexamethasone. Using a primary culture of leukemic blasts from a patient, we identified several molecular targeted drugs that conferred inhibitory effects in vitro. CONCLUSION A novel MEF2D-BCL9 fusion we identified characterizes a novel subset of pediatric ALL, predicts poor prognosis, and may be a candidate for novel molecular targeting.
Collapse
Affiliation(s)
- Kyogo Suzuki
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yusuke Okuno
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nozomu Kawashima
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hideki Muramatsu
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tatsuya Okuno
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Xinan Wang
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shinsuke Kataoka
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yuko Sekiya
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Motoharu Hamada
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Norihiro Murakami
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Daiei Kojima
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kotaro Narita
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Atsushi Narita
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hirotoshi Sakaguchi
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kimiyoshi Sakaguchi
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nao Yoshida
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nobuhiro Nishio
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Asahito Hama
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoshiyuki Takahashi
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kazuko Kudo
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Koji Kato
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan
| | - Seiji Kojima
- Kyogo Suzuki, Yusuke Okuno, Nozomu Kawashima, Hideki Muramatsu, Tatsuya Okuno, Xinan Wang, Shinsuke Kataoka, Yuko Sekiya, Motoharu Hamada, Norihiro Murakami, Daiei Kojima, Atsushi Narita, Nobuhiro Nishio, Asahito Hama, Yoshiyuki Takahashi, and Seiji Kojima, Nagoya University Graduate School of Medicine; Yusuke Okuno and Nobuhiro Nishio, Nagoya University Hospital; Kotaro Narita, Hirotoshi Sakaguchi, Nao Yoshida, and Koji Kato, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya; Kimiyoshi Sakaguchi, Hamamatsu University School of Medicine, Hamamatsu; and Kazuko Kudo, Fujita Health University School of Medicine, Toyoake, Japan.
| |
Collapse
|
91
|
Abnormal histone acetylation of CD8 + T cells in patients with severe aplastic anemia. Int J Hematol 2016; 104:540-547. [PMID: 27485471 DOI: 10.1007/s12185-016-2061-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/18/2016] [Accepted: 07/12/2016] [Indexed: 01/20/2023]
Abstract
Severe aplastic anemia (SAA) is a rare autoimmune disease characterized by severe pancytopenia and bone marrow failure, which is caused by activated T lymphocytes. In the present study, we evaluated histone H3 acetylation levels of bone marrow CD8+ T cells in SAA patients, and analyzed its correlation with clinical condition parameters. We found that the percentages of CD8+ T cell histone H3 acetylation in patients with untreated SAA, recovering SAA (R-SAA) and normal control, were 1.21 ± 0.08, 1.05 ± 0.36, and 1.00 ± 0.41, respectively, with no significant statistical differences. However, the amount of CD8+ T cell histone H3 acetylation from untreated SAA was 176.21 ± 32.22 μg/mg protein, which was significantly higher than that of complete response (CR)-SAA (104.29 ± 62.06 μg/mg protein) and normal control (133.94 ± 56.27 μg/mg protein) (P < 0.05) groups. Moreover, histone H3 acetylation amount of CD8+ T cell was significantly and negatively correlated with absolute neutrophil count, proportion of reticulocytes, ratio of CD4+ to CD8+ T cell in peripheral blood, and percentage of bone marrow erythroid (P < 0.05). To some extent, it also negatively correlated with hemoglobin level, platelet count, percentage of bone marrow granulocyte, and megakaryocyte count. Abnormal histone H3 acetylation of CD8+ T cells may thus play a role in the immune pathogenesis of SAA.
Collapse
|
92
|
Wang Z, Liu Y, Xue Y, Hu H, Ye J, Li X, Lu Z, Meng F, Liang S. Berberine acts as a putative epigenetic modulator by affecting the histone code. Toxicol In Vitro 2016; 36:10-17. [PMID: 27311644 DOI: 10.1016/j.tiv.2016.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 06/02/2016] [Accepted: 06/11/2016] [Indexed: 01/15/2023]
Abstract
Berberine, an isoquinoline plant alkaloid, exhibits a wide range of biochemical and pharmacological effects. However, the precise mechanism of these bioactivities remains poorly understood. In this study, we found significant similarity between berberine and two epigenetic modulators (CG-1521 and TSA). Reverse-docking using berberine as a ligand identified lysine-N-methyltransferase as a putative target of berberine. These findings suggested the potential role of berberine in epigenetic modulation. The results of PCR array analysis of epigenetic chromatin modification enzymes supported our hypothesis. Furthermore, the analysis showed that enzymes involved in histone acetylation and methylation were predominantly affected by treatment with berberine. Up-regulation of histone acetyltransferase CREBBP and EP300, histone deacetylase SIRT3, histone demethylase KDM6A as well as histone methyltransferase SETD7, and down-regulation of histone acetyltransferase HDAC8, histone methyltransferase WHSC1I, WHSC1II and SMYD3, in addition to 38 genes from histone clusters 1-3 were observed in berberine-treated cells using real-time PCR. In parallel, western blotting analyses revealed that the expression of H3K4me3, H3K27me3 and H3K36me3 proteins decreased with berberine treatment. These results were further confirmed in acute myelocytic leukemia (AML) cell lines HL-60/ADR and KG1-α. Taken together, this study suggests that berberine might modulate the expression of epigenetic regulators important for many downstream pathways, resulting in the variation of its bioactivities.
Collapse
Affiliation(s)
- Zhixiang Wang
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yuan Liu
- Department of Hematology, The Third Hospital of Nanchang City, Nanchang, China
| | - Yong Xue
- Jiangxi Science & Technology Research Center for Safety, Jiangxi, China
| | - Haiyan Hu
- Department of Oncology, 6th People's Hospital of Shanghai, Shanghai, China
| | - Jieyu Ye
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Xiaodong Li
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zhigang Lu
- Department of Blood Transfusion, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Fanyi Meng
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Shuang Liang
- Department of Neuroscience, Southern Medical University, Guangzhou, China.
| |
Collapse
|
93
|
Tandon N, Ramakrishnan V, Kumar SK. Clinical use and applications of histone deacetylase inhibitors in multiple myeloma. Clin Pharmacol 2016; 8:35-44. [PMID: 27226735 PMCID: PMC4866749 DOI: 10.2147/cpaa.s94021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The incorporation of various novel therapies has resulted in a significant survival benefit in newly diagnosed and relapsed patients with multiple myeloma (MM) over the past decade. Despite these advances, resistance to therapy leads to eventual relapse and fatal outcomes in the vast majority of patients. Hence, there is an unmet need for new safe and efficacious therapies for continued improvement in outcomes. Given the role of epigenetic aberrations in the pathogenesis and progression of MM and the success of histone deacetylase inhibitors (HDACi) in other malignancies, many HDACi have been tried in MM. Various preclinical studies helped us to understand the antimyeloma activity of different HDACi in MM as a single agent or in combination with conventional, novel, and immune therapies. The early clinical trials of HDACi depicted only modest single-agent activity, but recent studies have revealed encouraging clinical response rates in combination with other antimyeloma agents, especially proteasome inhibitors. This led to the approval of the combination of panobinostat and bortezomib for the treatment of relapsed/refractory MM patients with two prior lines of treatment by the US Food and Drug Administration. However, it remains yet to be defined how we can incorporate HDACi in the current therapeutic paradigms for MM that will help to achieve longer disease control and significant survival benefits. In addition, isoform-selective and/or class-selective HDAC inhibition to reduce unfavorable side effects needs further evaluation.
Collapse
Affiliation(s)
- Nidhi Tandon
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
94
|
San Jose-Eneriz E, Agirre X, Rodríguez-Otero P, Prosper F. Epigenetic regulation of cell signaling pathways in acute lymphoblastic leukemia. Epigenomics 2016; 5:525-38. [PMID: 24059799 DOI: 10.2217/epi.13.56] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous cancer that is characterized by rapid and uncontrolled proliferation of immature B- or T-lymphoid precursors. Although ALL has been regarded as a genetic disease for many years, the crucial importance of epigenetic alterations in leukemogenesis has become increasingly evident. Epigenetic mechanisms, which include DNA methylation and histone modifications, are critical for gene regulation during many key biological processes. Here, we review the cell signaling pathways that are regulated by DNA methylation or histone modifications in ALL. Recent studies have highlighted the fundamental role of these modifications in ALL development, and suggested that future investigation into the specific genes and pathways that are altered by epigenetic mechanisms can contribute to the development of novel drug-based therapies for ALL.
Collapse
Affiliation(s)
- Edurne San Jose-Eneriz
- Oncology Division, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | | | | |
Collapse
|
95
|
Zhang Y, Wu D, Xia F, Xian H, Zhu X, Cui H, Huang Z. Downregulation of HDAC9 inhibits cell proliferation and tumor formation by inducing cell cycle arrest in retinoblastoma. Biochem Biophys Res Commun 2016; 473:600-6. [PMID: 27033599 DOI: 10.1016/j.bbrc.2016.03.129] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 03/27/2016] [Indexed: 11/26/2022]
Abstract
Histone deacetylase 9 (HDAC9) is a member of class II HDACs, which regulates a wide variety of normal and abnormal physiological functions. Recently, HDAC9 has been found to be overexpressed in some types of human cancers. However, the role of HDAC9 in retinoblastoma remains unclear. In this study, we found that HDAC9 was commonly expressed in retinoblastoma tissues and HDAC9 was overexpressed in prognostically poor retinoblastoma patients. Through knocking down HDAC9 in Y79 and WERI-Rb-1 cells, the expression level of HDAC9 was found to be positively related to cell proliferation in vitro. Further investigation indicated that knockdown HDAC9 could significantly induce cell cycle arrest at G1 phase in retinoblastoma cells. Western blot assay showed downregulation of HDAC9 could significantly decrease cyclin E2 and CDK2 expression. Lastly, xenograft study in nude mice showed that downregulation of HDAC9 inhibited tumor growth and development in vivo. Therefore, our results suggest that HDAC9 could serve as a novel potential therapeutic target in the treatment of retinoblastoma.
Collapse
Affiliation(s)
- Yiting Zhang
- Medical School of Nanjing University, Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, China
| | - Dan Wu
- Medical School of Nanjing University, Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, China
| | - Fengjie Xia
- Medical School of Nanjing University, Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, China
| | - Hongyu Xian
- Medical School of Nanjing University, Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, China
| | - Xinyue Zhu
- Medical School of Nanjing University, Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Chongqing, 400716, China.
| | - Zhenping Huang
- Medical School of Nanjing University, Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, China.
| |
Collapse
|
96
|
Rastogi B, Raut SK, Panda NK, Rattan V, Radotra BD, Khullar M. Overexpression of HDAC9 promotes oral squamous cell carcinoma growth, regulates cell cycle progression, and inhibits apoptosis. Mol Cell Biochem 2016; 415:183-96. [PMID: 26992905 DOI: 10.1007/s11010-016-2690-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/12/2016] [Indexed: 12/18/2022]
Abstract
Histone deacetylases (HDACs) are a family of deacetylase enzymes that regulate the acetylation state of histones and a variety of other non-histone proteins including key oncogenic and tumor suppressor proteins, which modulates chromatin conformation, leading to regulation of gene expression. HDACs has been grouped into classes I-IV and histone deacetylase 9 (HDAC9) belongs to class IIa which exhibits tissue-specific expression. Recent reports have demonstrated both pro-oncogenic and tumor suppressive role for HDAC9 in different cancers; however, its role in OSCC remains elusive. Here, we investigated the role of HDAC9 in pathogenesis of oral squamous cell carcinoma (OSCC). Our data showed significantly increased mRNA and protein expression of HDAC9 in clinical OSCC samples and UPCI-SCC-116 cells as compared to normal counterpart. Kaplan-Meier analysis showed that the patients with high-level of HDAC9 expression had significantly reduced overall survival than those with low-level of HDAC9 expression (p = 0.034). Knockdown of HDAC9 using siRNA interference suppressed cell proliferation, increased apoptosis, and induced G0/G1 cell cycle arrest in UPCI-SCC-116 cells. Immunofluorescence analysis showed increased nuclear localization of HDAC9 in frozen OSCC sections, and indicative of active HDAC9 that may transcriptionally repress its downstream target genes. Subsequent investigation revealed that overexpression of HDAC9 contributes to OSCC carcinogenesis via targeting a transcription factor, MEF2D, and NR4A1/Nur77, a pro-apoptotic MEF2 target.
Collapse
Affiliation(s)
- Bhawna Rastogi
- Department of Otolaryngology and Head and Neck Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Satish K Raut
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Naresh K Panda
- Department of Otolaryngology and Head and Neck Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Vidya Rattan
- Department of Oral Health Sciences Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Bishan D Radotra
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| |
Collapse
|
97
|
Tedjaseputra A, Galli S, Ibrahim M, Harrison CN, McLornan DP. Histone deacetylase inhibitors in myeloproliferative neoplasms: current roles and future prospects. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1149467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
98
|
Qi J, Singh S, Hua WK, Cai Q, Chao SW, Li L, Liu H, Ho Y, McDonald T, Lin A, Marcucci G, Bhatia R, Huang WJ, Chang CI, Kuo YH. HDAC8 Inhibition Specifically Targets Inv(16) Acute Myeloid Leukemic Stem Cells by Restoring p53 Acetylation. Cell Stem Cell 2015; 17:597-610. [PMID: 26387755 PMCID: PMC4636961 DOI: 10.1016/j.stem.2015.08.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 06/15/2015] [Accepted: 08/04/2015] [Indexed: 11/28/2022]
Abstract
Acute myeloid leukemia (AML) is driven and sustained by leukemia stem cells (LSCs) with unlimited self-renewal capacity and resistance to chemotherapy. Mutation in the TP53 tumor suppressor is relatively rare in de novo AML; however, p53 can be regulated through post-translational mechanisms. Here, we show that p53 activity is inhibited in inv(16)(+) AML LSCs via interactions with the CBFβ-SMMHC (CM) fusion protein and histone deacetylase 8 (HDAC8). HDAC8 aberrantly deacetylates p53 and promotes LSC transformation and maintenance. HDAC8 deficiency or inhibition using HDAC8-selective inhibitors (HDAC8i) effectively restores p53 acetylation and activity. Importantly, HDAC8 inhibition induces apoptosis in inv(16)(+) AML CD34(+) cells, while sparing the normal hematopoietic stem cells. Furthermore, in vivo HDAC8i administration profoundly diminishes AML propagation and abrogates leukemia-initiating capacity of both murine and patient-derived LSCs. This study elucidates an HDAC8-mediated p53-inactivating mechanism promoting LSC activity and highlights HDAC8 inhibition as a promising approach to selectively target inv(16)(+) LSCs.
Collapse
Affiliation(s)
- Jing Qi
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Sandeep Singh
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Wei-Kai Hua
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Qi Cai
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | | | - Ling Li
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Hongjun Liu
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Yinwei Ho
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Tinisha McDonald
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Allen Lin
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Guido Marcucci
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Ravi Bhatia
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA
| | | | - Chung-I Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11574, Taiwan
| | - Ya-Huei Kuo
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Norbert Gehr and Family Leukemia Center, City of Hope Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
99
|
Zhang C, Zhong JF, Stucky A, Chen XL, Press MF, Zhang X. Histone acetylation: novel target for the treatment of acute lymphoblastic leukemia. Clin Epigenetics 2015; 7:117. [PMID: 26543507 PMCID: PMC4634719 DOI: 10.1186/s13148-015-0151-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/27/2015] [Indexed: 12/18/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) has been generally considered a genetic disease (disorder) with an aggressive tumor entity of highly proliferative malignant lymphoid cells. However, in recent years, significant advances have been made in the elucidation of the ALL-associated processes. Thus, we understand that histone acetylation is involved in the permanent changes of gene expression controlling ALL developmental outcomes. In this article, we will focus on histone acetylation associated with ALL, their implications as biomarkers for prognostic, and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 People's Republic of China
| | - Jiang F Zhong
- Department of Diagnostic Sciences & Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033 USA ; Department of Pediatric, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Andres Stucky
- Department of Diagnostic Sciences & Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033 USA ; Department of Pediatric, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Xue-Lian Chen
- Department of Diagnostic Sciences & Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033 USA ; Department of Pediatric, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Michael F Press
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 People's Republic of China
| |
Collapse
|
100
|
Jayaraman A, Jamil K, Khan HA. Identifying new targets in leukemogenesis using computational approaches. Saudi J Biol Sci 2015; 22:610-22. [PMID: 26288567 PMCID: PMC4537869 DOI: 10.1016/j.sjbs.2015.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/04/2015] [Accepted: 01/12/2015] [Indexed: 02/08/2023] Open
Abstract
There is a need to identify novel targets in Acute Lymphoblastic Leukemia (ALL), a hematopoietic cancer affecting children, to improve our understanding of disease biology and that can be used for developing new therapeutics. Hence, the aim of our study was to find new genes as targets using in silico studies; for this we retrieved the top 10% overexpressed genes from Oncomine public domain microarray expression database; 530 overexpressed genes were short-listed from Oncomine database. Then, using prioritization tools such as ENDEAVOUR, DIR and TOPPGene online tools, we found fifty-four genes common to the three prioritization tools which formed our candidate leukemogenic genes for this study. As per the protocol we selected thirty training genes from PubMed. The prioritized and training genes were then used to construct STRING functional association network, which was further analyzed using cytoHubba hub analysis tool to investigate new genes which could form drug targets in leukemia. Analysis of the STRING protein network built from these prioritized and training genes led to identification of two hub genes, SMAD2 and CDK9, which were not implicated in leukemogenesis earlier. Filtering out from several hundred genes in the network we also found MEN1, HDAC1 and LCK genes, which re-emphasized the important role of these genes in leukemogenesis. This is the first report on these five additional signature genes in leukemogenesis. We propose these as new targets for developing novel therapeutics and also as biomarkers in leukemogenesis, which could be important for prognosis and diagnosis.
Collapse
Affiliation(s)
- Archana Jayaraman
- Centre for Biotechnology and Bioinformatics, School of Life Sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), Secunderabad, Telangana, India
- Center for Biotechnology, Jawaharlal Nehru Technological University (JNTUH), Kukatpally, Hyderabad, Telangana, India
| | - Kaiser Jamil
- Centre for Biotechnology and Bioinformatics, School of Life Sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), Secunderabad, Telangana, India
- Corresponding author. at: Centre for Biotechnology and Bioinformatics, School of Life Sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), Buddha Bhawan, 6th Floor, M.G. Road, Secunderabad 500003, Telangana, India. Tel.: + 91 9676872626; fax: +91 40 27541551.
| | - Haseeb A. Khan
- Department of Biochemistry, College of Sciences, Bldg. 5, King Saud University, P.O. Box 2455, Riyadh, Saudi Arabia
| |
Collapse
|