1
|
Guadagni A, Barone S, Alfano AI, Pelliccia S, Bello I, Panza E, Summa V, Brindisi M. Tackling triple negative breast cancer with HDAC inhibitors: 6 is the isoform! Eur J Med Chem 2024; 279:116884. [PMID: 39321690 DOI: 10.1016/j.ejmech.2024.116884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Triple negative breast cancer (TNBC) is a highly aggressive breast cancer subtype characterized by the lack in the expression of estrogen and progesterone receptors, and human epidermal growth factor receptors 2. TNBC stands out among other breast cancers subtypes for its high aggressiveness and invasiveness, and for the limited therapeutic options available, which justify the poor survival rates registered for this breast cancer subtype. Compelling new evidence pointed out the role of epigenetic modifications in cancer, prompting tumor cell uncontrolled proliferation, epithelial-to-mesenchymal transition, and metastatic events. In this review we showcase the latest evidence supporting the involvement of histone deacetylase 6 (HDAC6) in cancer pathways strictly related to TNBC subtype, also tracking the latest advancements in the identification of novel HDAC6 inhibitors which showed efficacy in TNBC models, offering insights into the potential of targeting this key epigenetic player as an innovative therapeutic option for the treatment of TNBC.
Collapse
Affiliation(s)
- Anna Guadagni
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Simona Barone
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Antonella Ilenia Alfano
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Sveva Pelliccia
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Ivana Bello
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Elisabetta Panza
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Vincenzo Summa
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Margherita Brindisi
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy.
| |
Collapse
|
2
|
Rana PS, Ignatz-Hoover JJ, Kim BG, Malek E, Federov Y, Adams D, Chan T, Driscoll JJ. HDAC6 Inhibition Releases HR23B to Activate Proteasomes, Expand the Tumor Immunopeptidome and Amplify T-cell Antimyeloma Activity. CANCER RESEARCH COMMUNICATIONS 2024; 4:1517-1532. [PMID: 38747592 PMCID: PMC11188874 DOI: 10.1158/2767-9764.crc-23-0528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/14/2024] [Accepted: 05/09/2024] [Indexed: 06/19/2024]
Abstract
Proteasomes degrade intracellular proteins to generate antigenic peptides that are recognized by the adaptive immune system and promote anticancer immunity. However, tumors subvert the antigen presentation machinery to escape immunosurveillance. We hypothesized that proteasome activation could concomitantly increase antigen abundance and diversity in multiple myeloma cells. High-throughput screens revealed that histone deacetylase 6 (HDAC6) inhibitors activated proteasomes to unmask neoantigens and amplify the tumor-specific antigenic landscape. Treatment of patient CD138+ cells with HDAC6 inhibitors significantly promoted the antimyeloma activity of autologous CD8+ T cells. Pharmacologic blockade and genetic ablation of the HDAC6 ubiquitin-binding domain released HR23B, which shuttles ubiquitinylated cargo to proteasomes, while silencing HDAC6 or HR23B in multiple myeloma cells abolished the effect of HDAC6 inhibitors on proteasomes, antigen presentation, and T-cell cytotoxicity. Taken together, our results demonstrate the paradigm-shifting translational impact of proteasome activators to expand the myeloma immunopeptidome and have revealed novel, actionable antigenic targets for T cell-directed immunotherapy. SIGNIFICANCE The elimination of therapy-resistant tumor cells remains a major challenge in the treatment of multiple myeloma. Our study identifies and functionally validates agents that amplify MHC class I-presented antigens and pave the way for the development of proteasome activators as immune adjuvants to enhance immunotherapeutic responses in patients with multiple myeloma.
Collapse
Affiliation(s)
- Priyanka S. Rana
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - James J. Ignatz-Hoover
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Byung-Gyu Kim
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Ehsan Malek
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
- Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Yuriy Federov
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Small Molecule Drug Discovery Core, Case Western Reserve University, Cleveland, Ohio
| | - Drew Adams
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Small Molecule Drug Discovery Core, Case Western Reserve University, Cleveland, Ohio
| | - Timothy Chan
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio
| | - James J. Driscoll
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
- Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
3
|
Duda J, Thomas SN. Interactions of Histone Deacetylase 6 with DNA Damage Repair Factors Strengthen its Utility as a Combination Drug Target in High-Grade Serous Ovarian Cancer. ACS Pharmacol Transl Sci 2023; 6:1924-1933. [PMID: 38107255 PMCID: PMC10723650 DOI: 10.1021/acsptsci.3c00215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 12/19/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the deadliest gynecologic malignancy in women. The low survival rate is largely due to drug resistance. Approximately 80% of patients who initially respond to treatment relapse and become drug-resistant. The lack of effective second-line therapeutics remains a substantial challenge for BRCA-1/2 wild-type HGSOC patients. Histone Deacetylases (HDACs) are promising targets in HGSOC treatment; however, the mechanism and efficacy of HDAC inhibitors are understudied in HGSOC. In order to consider HDACs as a treatment target, an improved understanding of their function within HGSOC is required. This includes elucidating HDAC6-specific protein-protein interactions. In this study, we carried out substrate trapping followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) to elucidate HDAC6 catalytic domain (CD)-specific interactors in the context of BRCA-1/2 wild-type HGSOC. Overall, this study identified new HDAC6 substrates that may be unique to HGSOC. The HDAC6-CD1 mutant condition contained the largest number of significant proteins compared to the CD2 mutant and the CD1/2 mutant conditions, suggesting the HDAC6-CD1 domain has catalytic activity that is independent of CD2. Among the identified substrates were proteins involved in DNA damage repair including PARP proteins. These findings further justify the use of HDAC inhibitors as a combination treatment with platinum chemotherapy agents and PARP inhibitors in HGSOC.
Collapse
Affiliation(s)
- Jolene
M. Duda
- Department
of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Stefani N. Thomas
- Department
of Laboratory Medicine and Pathology, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Behrouj H, Vakili O, Sadeghdoust A, Aligolighasemabadi N, Khalili P, Zamani M, Mokarram P. Epigenetic regulation of autophagy in coronavirus disease 2019 (COVID-19). Biochem Biophys Rep 2022; 30:101264. [PMID: 35469237 PMCID: PMC9021360 DOI: 10.1016/j.bbrep.2022.101264] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/22/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has become the most serious global public health issue in the past two years, requiring effective therapeutic strategies. This viral infection is a contagious disease caused by new coronaviruses (nCoVs), also called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Autophagy, as a highly conserved catabolic recycling process, plays a significant role in the growth and replication of coronaviruses (CoVs). Therefore, there is great interest in understanding the mechanisms that underlie autophagy modulation. The modulation of autophagy is a very complex and multifactorial process, which includes different epigenetic alterations, such as histone modifications and DNA methylation. These mechanisms are also known to be involved in SARS-CoV-2 replication. Thus, molecular understanding of the epigenetic pathways linked with autophagy and COVID-19, could provide novel therapeutic targets for COVID-19 eradication. In this context, the current review highlights the role of epigenetic regulation of autophagy in controlling COVID-19, focusing on the potential therapeutic implications.
Collapse
Affiliation(s)
- Hamid Behrouj
- Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Adel Sadeghdoust
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Aligolighasemabadi
- Department of Internal Medicine, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parnian Khalili
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Iran
| |
Collapse
|
5
|
Varga JK, Diffley K, Welker Leng KR, Fierke CA, Schueler-Furman O. Structure-based prediction of HDAC6 substrates validated by enzymatic assay reveals determinants of promiscuity and detects new potential substrates. Sci Rep 2022; 12:1788. [PMID: 35110592 PMCID: PMC8810773 DOI: 10.1038/s41598-022-05681-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/17/2022] [Indexed: 01/25/2023] Open
Abstract
Histone deacetylases play important biological roles well beyond the deacetylation of histone tails. In particular, HDAC6 is involved in multiple cellular processes such as apoptosis, cytoskeleton reorganization, and protein folding, affecting substrates such as ɑ-tubulin, Hsp90 and cortactin proteins. We have applied a biochemical enzymatic assay to measure the activity of HDAC6 on a set of candidate unlabeled peptides. These served for the calibration of a structure-based substrate prediction protocol, Rosetta FlexPepBind, previously used for the successful substrate prediction of HDAC8 and other enzymes. A proteome-wide screen of reported acetylation sites using our calibrated protocol together with the enzymatic assay provide new peptide substrates and avenues to novel potential functional regulatory roles of this promiscuous, multi-faceted enzyme. In particular, we propose novel regulatory roles of HDAC6 in tumorigenesis and cancer cell survival via the regulation of EGFR/Akt pathway activation. The calibration process and comparison of the results between HDAC6 and HDAC8 highlight structural differences that explain the established promiscuity of HDAC6.
Collapse
Affiliation(s)
- Julia K Varga
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, POB 12272, 9112102, Jerusalem, Israel
| | - Kelsey Diffley
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI, 48109, USA
| | - Katherine R Welker Leng
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI, 48109, USA
| | - Carol A Fierke
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI, 48109, USA
- Department of Biochemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, POB 12272, 9112102, Jerusalem, Israel.
| |
Collapse
|
6
|
Liu P, Xiao J, Wang Y, Song X, Huang L, Ren Z, Kitazato K, Wang Y. Posttranslational modification and beyond: interplay between histone deacetylase 6 and heat-shock protein 90. Mol Med 2021; 27:110. [PMID: 34530730 PMCID: PMC8444394 DOI: 10.1186/s10020-021-00375-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/07/2021] [Indexed: 12/17/2022] Open
Abstract
Posttranslational modification (PTM) and regulation of protein stability are crucial to various biological processes. Histone deacetylase 6 (HDAC6), a unique histone deacetylase with two functional catalytic domains (DD1 and DD2) and a ZnF-UBP domain (ubiquitin binding domain, BUZ), regulates a number of biological processes, including gene expression, cell motility, immune response, and the degradation of misfolded proteins. In addition to the deacetylation of histones, other nonhistone proteins have been identified as substrates for HDAC6. Hsp90, a molecular chaperone that is a critical modulator of cell signaling, is one of the lysine deacetylase substrates of HDAC6. Intriguingly, as one of the best-characterized regulators of Hsp90 acetylation, HDAC6 is the client protein of Hsp90. In addition to regulating Hsp90 at the post-translational modification level, HDAC6 also regulates Hsp90 at the gene transcription level. HDAC6 mainly regulates the Hsp90-HSF1 complex through the ZnF-UBP domain, thereby promoting the HSF1 entry into the nucleus and activating gene transcription. The mutual interaction between HDAC6 and Hsp90 plays an important role in the regulation of protein stability, cell migration, apoptosis and other functions. Plenty of of studies have indicated that blocking HDAC6/Hsp90 has a vital regulatory role in multifarious diseases, mainly in cancers. Therefore, developing inhibitors or drugs against HDAC6/Hsp90 becomes a promising development direction. Herein, we review the current knowledge on molecular regulatory mechanisms based on the interaction of HDAC6 and Hsp90 and inhibition of HDAC6 and/or Hsp90 in oncogenesis and progression, antiviral and immune-related diseases and other vital biological processes.
Collapse
Affiliation(s)
- Ping Liu
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Ji Xiao
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Yiliang Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Xiaowei Song
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Lianzhou Huang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhe Ren
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Kaio Kitazato
- Department of Clinical Research Pharmacy, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| | - Yifei Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.
| |
Collapse
|
7
|
Li Y, Yang G, Yang C, Tang P, Chen J, Zhang J, Liu J, Ouyang L. Targeting Autophagy-Related Epigenetic Regulators for Cancer Drug Discovery. J Med Chem 2021; 64:11798-11815. [PMID: 34378389 DOI: 10.1021/acs.jmedchem.1c00579] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Existing evidence has demonstrated that epigenetic modifications (including DNA methylation, histone modifications, and microRNAs), which are associated with the occurrence and development of tumors, can directly or indirectly regulate autophagy. In particular, nuclear events induced by several epigenetic regulators can regulate the autophagic process and expression levels of tumor-associated genes, thereby promoting tumor progression. Tumor-associated microRNAs, including oncogenic and tumor-suppressive microRNAs, are of great significance to autophagy during tumor progression. Targeting autophagy with emerging epigenetic drugs is expected to be a promising therapeutic strategy for human tumors. From this perspective, we aim to summarize the role of epigenetic modification in the autophagic process and the underlying molecular mechanisms of tumorigenesis. Furthermore, the regulatory efficacy of epigenetic drugs on the autophagic process in tumors is also summarized. This perspective may provide a theoretical basis for the combined treatment of epigenetic drugs/autophagy mediators in tumors.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Gaoxia Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Chengcan Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Pan Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Juncheng Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jie Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
8
|
Li J, Tian L, Jing Z, Guo Z, Nan P, Liu F, Zou S, Yang L, Xie X, Zhu Y, Zhao Y, Sun W, Sun Y, Zhao X. Cytoplasmic RAD23B interacts with CORO1C to synergistically promote colorectal cancer progression and metastasis. Cancer Lett 2021; 516:13-27. [PMID: 34062216 DOI: 10.1016/j.canlet.2021.05.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Colorectal cancers (CRCs) are characterized by diffuse infiltration of tumor cells into the regional lymph nodes and metastasis to distant organs, and its highly invasive nature contributes to disease recurrence and poor outcomes. The molecular mechanisms underlying CRC cell invasion remain incompletely understood. Here, we identified the upregulation of DNA damage repair-related protein RAD23B in CRC cells and tissues and showed that it associates with coronin 1C or coronin 3 (CORO1C) to facilitate invasion. We found that knockdown of RAD23B expression significantly inhibited the proliferation, invasion, and migration abilities of CRC cells both in vitro and in vivo, and suppressed the talin1/2/integrin/FAK/RhoA/Rac1/CORO1C signaling pathways. Interestingly, RAD23B interacted and co-localized with CORO1C, and CORO1C aggregated toward the margin of cancer cells in both CRC cells and tissues when RAD23B overexpressed. Mechanistically, overexpression of RAD23B and/or CORO1C further increased invadopodia formation and matrix degradation in SW480 and HCT8 CRC cells. Conversely, silencing of RAD23B expression suppressed tumorigenesis and liver metastasis in xenotransplant murine models. Furthermore, we found that RAD23B was significantly overexpressed in tumor tissues (n = 720) compared to adjacent non-tumor tissues (n = 694) of patients with CRC. Finally, we identified a strong correlation between higher levels of cytoplasmic expression of RAD23B, and poor prognosis and liver metastasis in CRC patients. Taken together, our data highlight a novel RAD23B-CORO1C signaling axis in CRC cell invasion and metastasis that may be of clinical significance.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lusong Tian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zongpan Jing
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhengguang Guo
- Core Facility of Instruments, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Peng Nan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuangmei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lijun Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiufeng Xie
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ying Zhu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Sun
- Core Facility of Instruments, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yulin Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xiaohang Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
9
|
Kulka LAM, Fangmann PV, Panfilova D, Olzscha H. Impact of HDAC Inhibitors on Protein Quality Control Systems: Consequences for Precision Medicine in Malignant Disease. Front Cell Dev Biol 2020; 8:425. [PMID: 32582706 PMCID: PMC7291789 DOI: 10.3389/fcell.2020.00425] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/07/2020] [Indexed: 12/21/2022] Open
Abstract
Lysine acetylation is one of the major posttranslational modifications (PTM) in human cells and thus needs to be tightly regulated by the writers of this process, the histone acetyl transferases (HAT), and the erasers, the histone deacetylases (HDAC). Acetylation plays a crucial role in cell signaling, cell cycle control and in epigenetic regulation of gene expression. Bromodomain (BRD)-containing proteins are readers of the acetylation mark, enabling them to transduce the modification signal. HDAC inhibitors (HDACi) have been proven to be efficient in hematologic malignancies with four of them being approved by the FDA. However, the mechanisms by which HDACi exert their cytotoxicity are only partly resolved. It is likely that HDACi alter the acetylation pattern of cytoplasmic proteins, contributing to their anti-cancer potential. Recently, it has been demonstrated that various protein quality control (PQC) systems are involved in recognizing the altered acetylation pattern upon HDACi treatment. In particular, molecular chaperones, the ubiquitin proteasome system (UPS) and autophagy are able to sense the structurally changed proteins, providing additional targets. Recent clinical studies of novel HDACi have proven that proteins of the UPS may serve as biomarkers for stratifying patient groups under HDACi regimes. In addition, members of the PQC systems have been shown to modify the epigenetic readout of HDACi treated cells and alter proteostasis in the nucleus, thus contributing to changing gene expression profiles. Bromodomain (BRD)-containing proteins seem to play a potent role in transducing the signaling process initiating apoptosis, and many clinical trials are under way to test BRD inhibitors. Finally, it has been demonstrated that HDACi treatment leads to protein misfolding and aggregation, which may explain the effect of panobinostat, the latest FDA approved HDACi, in combination with the proteasome inhibitor bortezomib in multiple myeloma. Therefore, proteins of these PQC systems provide valuable targets for precision medicine in cancer. In this review, we give an overview of the impact of HDACi treatment on PQC systems and their implications for malignant disease. We exemplify the development of novel HDACi and how affected proteins belonging to PQC can be used to determine molecular signatures and utilized in precision medicine.
Collapse
Affiliation(s)
- Linda Anna Michelle Kulka
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Pia-Victoria Fangmann
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Diana Panfilova
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Heidi Olzscha
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
10
|
xCT Inhibition Increases Sensitivity to Vorinostat in a ROS-Dependent Manner. Cancers (Basel) 2020; 12:cancers12040827. [PMID: 32235498 PMCID: PMC7226257 DOI: 10.3390/cancers12040827] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/30/2022] Open
Abstract
As histone deacetylase inhibitors (HDACIs) have limited efficacy against solid tumors, we investigated whether and how oxidative stress is involved in sensitivity to HDACIs to develop a novel therapeutic option of HDACIs treatment. We first tested whether a reduction of the antioxidant glutathione (GSH) by glutamine deprivation affects sensitivity to a commercially available HDACI vorinostat and reactive oxygen species (ROS) accumulation. Next we investigated the relationship between a glutamate-cystine transporter xCT and the efficacy of vorinostat using siRNA of xCT and bioinformatic analyses. Finally, we verified the combinatory effects of vorinostat and the xCT inhibitor salazosulfapyridine (SASP) on ROS accumulation, cell death induction, and colony formation. Glutamine deprivation increased vorinostat-mediated cell death with ROS accumulation. Genetic ablation of xCT improved the efficacy of vorinostat, consistent with the results of public data analyses demonstrating that xCT expressions positively correlate with insensitivity to HDACIs in many types of cancer cell lines. Vorinostat caused ROS accumulation when combined with SASP, possibly resulting in synergistic ferroptosis. Our study provides a novel mechanistic insight into the mechanism underlying sensitivity to HDACIs involving xCT, suggesting xCT to be a promising predictive marker of HDACIs and rationalizing combinatory therapy of HDACIs with xCT inhibitors to induce ferroptosis.
Collapse
|
11
|
Epigenetic Control of Autophagy in Cancer Cells: A Key Process for Cancer-Related Phenotypes. Cells 2019; 8:cells8121656. [PMID: 31861179 PMCID: PMC6952790 DOI: 10.3390/cells8121656] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/19/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Although autophagy is a well-known and extensively described cell pathway, numerous studies have been recently interested in studying the importance of its regulation at different molecular levels, including the translational and post-translational levels. Therefore, this review focuses on the links between autophagy and epigenetics in cancer and summarizes the. following: (i) how ATG genes are regulated by epigenetics, including DNA methylation and post-translational histone modifications; (ii) how epidrugs are able to modulate autophagy in cancer and to alter cancer-related phenotypes (proliferation, migration, invasion, tumorigenesis, etc.) and; (iii) how epigenetic enzymes can also regulate autophagy at the protein level. One noteable observation was that researchers most often reported conclusions about the regulation of the autophagy flux, following the use of epidrugs, based only on the analysis of LC3B-II form in treated cells. However, it is now widely accepted that an increase in LC3B-II form could be the consequence of an induction of the autophagy flux, as well as a block in the autophagosome-lysosome fusion. Therefore, in our review, all the published results describing a link between epidrugs and autophagy were systematically reanalyzed to determine whether autophagy flux was indeed increased, or inhibited, following the use of these potentially new interesting treatments targeting the autophagy process. Altogether, these recent data strongly support the idea that the determination of autophagy status could be crucial for future anticancer therapies. Indeed, the use of a combination of epidrugs and autophagy inhibitors could be beneficial for some cancer patients, whereas, in other cases, an increase of autophagy, which is frequently observed following the use of epidrugs, could lead to increased autophagy cell death.
Collapse
|
12
|
Huang Z, Xia Y, Hu K, Zeng S, Wu L, Liu S, Zhi C, Lai M, Chen D, Xie L, Yuan Z. Histone deacetylase 6 promotes growth of glioblastoma through the MKK7/JNK/c-Jun signaling pathway. J Neurochem 2019; 152:221-234. [PMID: 31390677 DOI: 10.1111/jnc.14849] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022]
Abstract
Histone deacetylase 6 (HDAC6) activity contributes to the malignant proliferation, invasion, and migration of glioma cells (GCs), but the molecular mechanisms underlying the processes remains elusive. Here, we reported that HDAC6 inhibition by Ricolinostat (ACY-1215) or CAY10603 led to a remarkable decrease in the phosphorylation of c-Jun N-terminal kinase (JNK) and c-Jun, which preceded its suppressive effects on glioma cell growth. Further investigation showed that these effects resulted from HDAC6 inhibitor-induced suppression of MAPK kinase 7 (MKK7), which was identified to be critical for JNK activation and exerts the oncogenic roles in GCs. Selectively silencing HDAC6 by siRNAs had the same responses, whereas transient transfections expressing HDAC6 promoted MKK7 expression. Interestingly, by performing Q-PCR, HDAC6 inhibition did not cause a down-regulation of MKK7 mRNA level, whereas the suppressive effects on MKK7 protein can be efficiently blocked by the proteasomal inhibitor MG132. As a further test, elevating MKK7-JNK activity was sufficient to rescue HDAC6 inhibitor-mediated-suppressive effects on c-Jun activation and the malignant features. The suppression of both MKK7 expression and JNK/c-Jun activities was involved in the tumor-growth inhibitory effects induced by CAY10603 in U87-xenograft mice. Collectively, our findings provide new insights into the molecular mechanism of glioma malignancy regarding HDAC6 in the selective regulation of MKK7 expression and JNK/c-Jun activity. MKK7 protein stability critically depends on HDAC6 activity, and inhibition of HDAC6 probably presents a potential strategy for suppressing the oncogenic roles of MKK7/JNK/c-Jun axis in GCs.
Collapse
Affiliation(s)
- Ziyan Huang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Yong Xia
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Kunhua Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key laboratory of Brain Function and Disease, Guangzhou, China
| | - Shulian Zeng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Liqiang Wu
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Sisi Liu
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Cheng Zhi
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Miaoling Lai
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Danmin Chen
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Longchang Xie
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Zhongmin Yuan
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China.,Guangdong Province Key laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
13
|
Olzscha H. Posttranslational modifications and proteinopathies: how guardians of the proteome are defeated. Biol Chem 2019; 400:895-915. [DOI: 10.1515/hsz-2018-0458] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/13/2019] [Indexed: 01/15/2023]
Abstract
Abstract
Protein folding is one of the fundamental processes in life and therefore needs to be tightly regulated. Many cellular quality control systems are in place to ensure that proteostasis is optimally adjusted for a changing environment, facilitating protein folding, translocation and degradation. These systems include the molecular chaperones and the major protein degradation systems, namely the ubiquitin proteasome system and autophagy. However, the capacity of the quality control systems can be exhausted and protein misfolding and aggregation, including the formation of amyloids, can occur as a result of ageing, mutations or exogenous influences. There are many known diseases in which protein misfolding and aggregation can be the underlying cause of the pathological condition; these are referred to as proteinopathies. Over the last decade, it has become clear that posttranslational modifications can govern and modulate protein folding, and that aberrant posttranslational modifications can cause or contribute to proteinopathies. This review provides an overview of protein folding and misfolding and the role of the major protein quality control systems. It focusses on different posttranslational modifications and gives examples of how these posttranslational modifications can alter protein folding and cause or accompany proteinopathies.
Collapse
Affiliation(s)
- Heidi Olzscha
- Institut für Physiologische Chemie , Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg , Hollystr. 1 , D-06114 Halle/Saale , Germany
| |
Collapse
|
14
|
Butler RM, McKenzie RC, Jones CL, Flanagan CE, Woollard WJ, Demontis M, Ferreira S, Tosi I, John S, Whittaker SJ, Mitchell TJ. Contribution of STAT3 and RAD23B in Primary Sézary Cells to Histone Deacetylase Inhibitor FK228 Resistance. J Invest Dermatol 2019; 139:1975-1984.e2. [PMID: 30910759 DOI: 10.1016/j.jid.2019.03.1130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/23/2019] [Accepted: 03/06/2019] [Indexed: 01/12/2023]
Abstract
FK228 (romidepsin) and suberoylanilide hydroxamic acid (vorinostat) are histone deacetylase inhibitors (HDACi) approved by the US Food and Drug Administration for cutaneous T-cell lymphoma (CTCL), including the leukemic subtype Sézary syndrome. This study investigates RAD23B and STAT3 gene perturbations in a large cohort of primary Sézary cells and the effect of FK228 treatment on tyrosine phosphorylation of STAT3 (pYSTAT3) and RAD23B expression. We report RAD23B copy number variation in 10% (12/119, P ≤ 0.01) of SS patients, associated with reduced mRNA expression (P = 0.04). RAD23B knockdown in a CTCL cell line led to a reduction in FK228-induced apoptosis. Histone deacetylase inhibitor treatment significantly reduced pYSTAT3 in primary Sézary cells and was partially mediated by RAD23B. A distinct pattern of RAD23B-pYSTAT3 co-expression in primary Sézary cells was detected. Critically, Sézary cells harboring the common STAT3 Y640F variant were less sensitive to FK228-induced apoptosis and exogenous expression of STAT3 Y640F, and D661Y conferred partial resistance to STAT3 transcriptional inhibition by FK228 (P ≤ 0.0024). These findings suggest that RAD23B and STAT3 gene perturbations could reduce sensitivity to histone deacetylase inhibitors in SS patients.
Collapse
Affiliation(s)
- Rosie M Butler
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Robert C McKenzie
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Christine L Jones
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Charlotte E Flanagan
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Wesley J Woollard
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Maria Demontis
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Silvia Ferreira
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Isabella Tosi
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Susan John
- Department of Immunology, Infection and Inflammatory Disease, King's College London, Guy's Hospital, London, UK
| | - Sean J Whittaker
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Tracey J Mitchell
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK.
| |
Collapse
|
15
|
Liu Z, Cai Y, Yang Y, Li A, Bi R, Wang L, Shen X, Wang W, Jia Y, Yu B, Cao B, Cui W, Wei P, Zhou X. Activation of MET signaling by HDAC6 offers a rationale for a novel ricolinostat and crizotinib combinatorial therapeutic strategy in diffuse large B-cell lymphoma. J Pathol 2018; 246:141-153. [DOI: 10.1002/path.5108] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 04/26/2018] [Accepted: 05/30/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Zebing Liu
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
- Department of Pathology, Renji Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai PR China
| | - Ying Cai
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
- Department of Pathology; Wuxi People's Hospital Affiliated to Nanjing Medical University; Wuxi Jiangsu PR China
| | - Yu Yang
- Scientific Research Center, Shanghai Public Health Clinical Center; Fudan University; Shanghai PR China
| | - Anqi Li
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Rui Bi
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Lisha Wang
- Michigan Center for Translational Pathology; University of Michigan Medical School; Ann Arbor MI USA
| | - Xiaohan Shen
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Weige Wang
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Yijun Jia
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Baohua Yu
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Bing Cao
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Wenli Cui
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Ping Wei
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
| | - Xiaoyan Zhou
- Department of Pathology; Fudan University Shanghai Cancer Center; Shanghai PR China
- Department of Oncology, Shanghai Medical College; Fudan University; Shanghai PR China
- Institute of Pathology; Fudan University; Shanghai PR China
| |
Collapse
|
16
|
Fujii K, Suzuki N, Jimura N, Idogawa M, Kondo T, Iwatsuki K, Kanekura T. HSP72 functionally inhibits the anti-neoplastic effects of HDAC inhibitors. J Dermatol Sci 2018; 90:82-89. [PMID: 29395577 DOI: 10.1016/j.jdermsci.2018.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/18/2017] [Accepted: 01/09/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND The anticancer effects of histone deacetylase inhibitors (HDACi) vary between patients, and their molecular mechanisms remain poorly understood. Previously, we have identified heat shock 70 kDa protein 1A (HSPA1A, also known as HSP72) as the most overexpressed protein in valproic acid (VPA)-resistant cell lines. KNK437, an inhibitor of heat shock proteins, enhanced the cytotoxic effects of not only VPA but also vorinostat, another HDACi. However, the mechanisms underlying the role of HSP72 in resistance against HDACi remain largely unknown. OBJECTIVE The purpose of this study was to identify the mechanisms underlying the role of HSP72 in HDACi resistance. METHODS We established an HSP72-overexpressing Jurkat cell line and used it to assess the functional role of HSP72 following treatment with the HDACi vorinostat and VPA. RESULTS HDACi-induced apoptosis, assessed using annexin V assays, sub-G1 fraction analysis, and PARP cleavage, was significantly lower in HSP72-overexpressing cells than in control cells. The HDACi-induced upregulation in caspase-3, -8, and -9 activity, as well as the HDACi-induced reduction in mitochondrial membrane potential, were also suppressed following HSP72 overexpression. The basal expression levels of Bcl-2, phosphorylated Bad, and XIAP increased in HSP72-overexpressing cells, whereas HDACi-induced Bid truncation and the suppression of Bad expression. Furthermore, vorinostat-induced histone hyperacetylation was also diminished in HSP72-overexpressing cells. CONCLUSION These findings clearly demonstrate that HSP72 inhibits HDACi-induced apoptosis.
Collapse
Affiliation(s)
- Kazuyasu Fujii
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, Japan.
| | - Norihiro Suzuki
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nozomi Jimura
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, Japan
| | - Masashi Idogawa
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Keiji Iwatsuki
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuro Kanekura
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, Japan
| |
Collapse
|
17
|
Histone deacetylase 9 regulates breast cancer cell proliferation and the response to histone deacetylase inhibitors. Oncotarget 2017; 7:19693-708. [PMID: 26930713 PMCID: PMC4991412 DOI: 10.18632/oncotarget.7564] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 01/23/2016] [Indexed: 01/13/2023] Open
Abstract
Histone lysine acetylation is an epigenetic mark regulated by histone acetyltransferases and histone deacetylases (HDAC) which plays an important role in tumorigenesis. In this study, we observed a strong overexpression of class IIa HDAC9, at the mRNA and protein levels, in the most aggressive human breast cancer cell lines (i.e. in basal breast cancer cells vs luminal ones or in malignant vs begnin MCF10A breast epithelial cell lines). HDAC9 overexpression was associated with higher rates of gene transcription and increased epigenetic marks on the HDAC9 promoter. Ectopic expression of HDAC9 in MCF7 luminal breast cancer cells led to an increase in cell proliferation and to a decrease in apoptosis. These effects were associated with a deregulated expression of several genes controlled by HDAC inhibitors such as CDKN1A, BAX and TNFRSF10A. Inversely, knock-down of HDAC9 expression in MDA-MB436 basal breast cancer cells reduced cell proliferation. Moreover, high HDAC9 expression decreased the efficacy of HDAC inhibitors to reduce cell proliferation and to regulate CDKN1A gene expression. Interestingly, the gene encoding the transcription factor SOX9 was identified by a global transcriptomic approach as an HDAC9 target gene. In stably transfected MCF7 cells, SOX9 silencing significantly decreased HDAC9 mitogenic activity. Finally, in a large panel of breast cancer biopsies, HDAC9 expression was significantly increased in tumors of the basal subtype, correlated with SOX9 expression and associated with poor prognosis. Altogether, these results indicate that HDAC9 is a key factor involved in mammary carcinogenesis and in the response to HDAC inhibitors.
Collapse
|
18
|
Nikolova T, Kiweler N, Krämer OH. Interstrand Crosslink Repair as a Target for HDAC Inhibition. Trends Pharmacol Sci 2017; 38:822-836. [PMID: 28687272 DOI: 10.1016/j.tips.2017.05.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/29/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022]
Abstract
DNA interstrand crosslinks (ICLs) covalently connect complementary DNA strands. Consequently, DNA replication and transcription are hampered, DNA damage responses (DDR) are initiated, and cell death is triggered. Therefore, drugs inducing ICLs are effective against rapidly growing cancer cells. However, tumors engage a complicated enzymatic machinery to repair and survive ICLs. Several factors, including the post-translational acetylation/deacetylation of lysine residues within proteins, control this network. Histone deacetylases (HDACs) modulate the expression and functions of DNA repair proteins which remove ICLs and control the accessibility of chromatin. Accordingly, histone deacetylase inhibitors (HDACi) are small, pharmacologically and clinically relevant molecules that sensitize cancer cells to ICL inducers. We discuss the mechanism of ICL repair and targets of HDACi within this pathway.
Collapse
Affiliation(s)
- Teodora Nikolova
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany.
| | - Nicole Kiweler
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany
| | - Oliver H Krämer
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany.
| |
Collapse
|
19
|
Nebbioso A, Carafa V, Conte M, Tambaro FP, Abbondanza C, Martens J, Nees M, Benedetti R, Pallavicini I, Minucci S, Garcia-Manero G, Iovino F, Lania G, Ingenito C, Belsito Petrizzi V, Stunnenberg HG, Altucci L. c-Myc Modulation and Acetylation Is a Key HDAC Inhibitor Target in Cancer. Clin Cancer Res 2017; 23:2542-2555. [PMID: 27358484 DOI: 10.1158/1078-0432.ccr-15-2388] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 05/03/2016] [Accepted: 06/09/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Histone deacetylase inhibitors (HDACi) are promising anticancer drugs. Although some HDACi have entered the clinic, the mechanism(s) underlying their tumor selectivity are poorly understood.Experimental Design and Results: Using gene expression analysis, we define a core set of six genes commonly regulated in acute myeloid leukemia (AML) blasts and cell lines. MYC, the most prominently modulated, is preferentially altered in leukemia. Upon HDACi treatment, c-Myc is acetylated at lysine 323 and its expression decreases, leading to TRAIL activation and apoptosis. c-Myc binds to the TRAIL promoter on the proximal GC box through SP1 or MIZ1, impairing TRAIL activation. HDACi exposure triggers TRAIL expression, altering c-Myc-TRAIL binding. These events do not occur in normal cells. Excitingly, this inverse correlation between TRAIL and c-Myc is supported by HDACi treatment ex vivo of AML blasts and primary human breast cancer cells. The predictive value of c-Myc to HDACi responsiveness is confirmed in vivo in AML patients undergoing HDACi-based clinical trials.Conclusions: Collectively, our findings identify a key role for c-Myc in TRAIL deregulation and as a biomarker of the anticancer action of HDACi in AML. The potential improved patient stratification could pave the way toward personalized therapies. Clin Cancer Res; 23(10); 2542-55. ©2016 AACR.
Collapse
Affiliation(s)
- Angela Nebbioso
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy.
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud University, Nijmegen Center for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Vincenzo Carafa
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | | | - Francesco Paolo Tambaro
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ciro Abbondanza
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | - Joost Martens
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud University, Nijmegen Center for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Matthias Nees
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Rosaria Benedetti
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | - Isabella Pallavicini
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | - Francesco Iovino
- Dipartimento Scienze Anestesiologiche, Chirurgiche e dell'Emergenza, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy
| | - Gabriella Lania
- Institute of Genetics and Biophysics (IGB) 'Adriano Buzzati Traverso', Naples, Italy
| | | | | | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud University, Nijmegen Center for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Lucia Altucci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Università degli Studi della Campania 'L. Vanvitelli', Naples, Italy.
- Institute of Genetics and Biophysics (IGB) 'Adriano Buzzati Traverso', Naples, Italy
| |
Collapse
|
20
|
Yokoi M, Hanaoka F. Two mammalian homologs of yeast Rad23, HR23A and HR23B, as multifunctional proteins. Gene 2017; 597:1-9. [DOI: 10.1016/j.gene.2016.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
|
21
|
Personalized therapy for hepatocellular carcinoma: Where are we now? Cancer Treat Rev 2016; 45:77-86. [DOI: 10.1016/j.ctrv.2016.02.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 12/14/2022]
|
22
|
Stahl M, Gore SD, Vey N, Prebet T. Lost in translation? Ten years of development of histone deacetylase inhibitors in acute myeloid leukemia and myelodysplastic syndromes. Expert Opin Investig Drugs 2016; 25:307-17. [DOI: 10.1517/13543784.2016.1146251] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
23
|
Falkenberg KJ, Newbold A, Gould CM, Luu J, Trapani JA, Matthews GM, Simpson KJ, Johnstone RW. A genome scale RNAi screen identifies GLI1 as a novel gene regulating vorinostat sensitivity. Cell Death Differ 2016; 23:1209-18. [PMID: 26868908 DOI: 10.1038/cdd.2015.175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 12/28/2022] Open
Abstract
Vorinostat is an FDA-approved histone deacetylase inhibitor (HDACi) that has proven clinical success in some patients; however, it remains unclear why certain patients remain unresponsive to this agent and other HDACis. Constitutive STAT (signal transducer and activator of transcription) activation, overexpression of prosurvival Bcl-2 proteins and loss of HR23B have been identified as potential biomarkers of HDACi resistance; however, none have yet been used to aid the clinical utility of HDACi. Herein, we aimed to further elucidate vorinostat-resistance mechanisms through a functional genomics screen to identify novel genes that when knocked down by RNA interference (RNAi) sensitized cells to vorinostat-induced apoptosis. A synthetic lethal functional screen using a whole-genome protein-coding RNAi library was used to identify genes that when knocked down cooperated with vorinostat to induce tumor cell apoptosis in otherwise resistant cells. Through iterative screening, we identified 10 vorinostat-resistance candidate genes that sensitized specifically to vorinostat. One of these vorinostat-resistance genes was GLI1, an oncogene not previously known to regulate the activity of HDACi. Treatment of vorinostat-resistant cells with the GLI1 small-molecule inhibitor, GANT61, phenocopied the effect of GLI1 knockdown. The mechanism by which GLI1 loss of function sensitized tumor cells to vorinostat-induced apoptosis is at least in part through interactions with vorinostat to alter gene expression in a manner that favored apoptosis. Upon GLI1 knockdown and vorinostat treatment, BCL2L1 expression was repressed and overexpression of BCL2L1 inhibited GLI1-knockdown-mediated vorinostat sensitization. Taken together, we present the identification and characterization of GLI1 as a new HDACi resistance gene, providing a strong rationale for development of GLI1 inhibitors for clinical use in combination with HDACi therapy.
Collapse
Affiliation(s)
- K J Falkenberg
- Cancer Therapeutics Program, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia
| | - A Newbold
- Cancer Therapeutics Program, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia
| | - C M Gould
- Victorian Centre for Functional Genomics, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia
| | - J Luu
- Victorian Centre for Functional Genomics, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia
| | - J A Trapani
- Cancer Immunology Program, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - G M Matthews
- Cancer Therapeutics Program, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - K J Simpson
- Victorian Centre for Functional Genomics, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - R W Johnstone
- Cancer Therapeutics Program, The Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC 3002, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
24
|
Angelika Ihle M, Merkelbach-Bruse S, Hartmann W, Bauer S, Ratner N, Sonobe H, Nishio J, Larsson O, Åman P, Pedeutour F, Taguchi T, Wardelmann E, Buettner R, Schildhaus HU. HR23b expression is a potential predictive biomarker for HDAC inhibitor treatment in mesenchymal tumours and is associated with response to vorinostat. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 2:59-71. [PMID: 27499916 PMCID: PMC4907056 DOI: 10.1002/cjp2.35] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/13/2015] [Accepted: 12/17/2015] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDAC) are key players in epigenetic regulation of gene expression and HDAC inhibitor (HDACi) treatment seems to be a promising anticancer therapy in many human tumours, including soft tissue sarcomas. HR23b has been shown to be a potential biomarker for sensitivity to HDACi therapy in cutaneous T‐cell lymphoma and hepatocellular carcinoma. We aimed to evaluate HR23b as a candidate biomarker for HDACi response in sarcomas and gastrointestinal stromal tumours (GIST). Therefore, HR23b expression was analysed comprehensively by western blot in sarcoma and GIST cell lines covering all major clinically relevant subtypes. MTT assay and ApoTox‐GloTM Triplex assay were performed after treatment with vorinostat, belinostat, mocetinostat and entinostat. HR23b protein expression was measured under HDACi treatment. Furthermore, HR23b expression levels were immunohistochemically determined in a large set of 523 clinical samples from sarcoma and GIST patients. Western blot analyses showed that sarcomas differ significantly in their expression of HR23b protein. All HDACi were able to regulate proliferation and apoptosis in vitro. Sensitivity to vorinostat correlated significantly with HR23b protein expression. Immunohistochemical prevalence screening in clinical samples of relevant adult‐type tumours revealed that 12.5% of sarcomas (among them malignant peripheral nerve sheath tumours, pleomorphic liposarcomas, leiomyosarcomas, dedifferentiated liposarcomas, synovial sarcomas and angiosarcomas) and 23.2% of GIST show high HR23b expression. Therefore, HDACi have antiproliferative and proapoptotic effects in sarcomas depending on the expression level of HR23b. These findings suggest that HR23b represents a candidate biomarker for HDACi sensitivity in certain sarcoma types and in GIST.
Collapse
Affiliation(s)
| | | | - Wolfgang Hartmann
- Institute of Pathology, University Hospital CologneCologneGermany; Gerhard Domagk Institute of Pathology, University Hospital MünsterMünsterGermany
| | - Sebastian Bauer
- Sarcoma Center, West German Cancer Center, University of Essen Essen Germany
| | - Nancy Ratner
- US Department of Pediatrics Cincinnati Children's Hospital Medical Centre Cincinnati OH USA
| | - Hiroshi Sonobe
- Department of Laboratory Medicine Chugoku Central Hospital Fukuyama Hiroshima Japan
| | - Jun Nishio
- Faculty of Medicine, Department of Orthopaedic Surgery Fukuoka University Fukuoka Japan
| | - Olle Larsson
- Department of Oncology and Pathology The Karolinska Institute Stockholm Sweden
| | - Pierre Åman
- Sahlgrenska Cancer Centre, University of Gothenburg Gothenburg Sweden
| | - Florence Pedeutour
- Faculty of Medicine, Laboratory of Genetics of Solid Tumours Institute for Research on Cancer and Aging Nice France
| | - Takahiro Taguchi
- Division of Human Health & Medical Science, Graduate School of Kuroshio Science Kochi University Nankoku Kochi Japan
| | - Eva Wardelmann
- Institute of Pathology, University Hospital CologneCologneGermany; Gerhard Domagk Institute of Pathology, University Hospital MünsterMünsterGermany
| | | | - Hans-Ulrich Schildhaus
- Institute of Pathology, University Hospital CologneCologneGermany; Institute of Pathology, University Hospital GöttingenGöttingenGermany
| |
Collapse
|
25
|
Loss of RAD-23 Protects Against Models of Motor Neuron Disease by Enhancing Mutant Protein Clearance. J Neurosci 2016; 35:14286-306. [PMID: 26490867 DOI: 10.1523/jneurosci.0642-15.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Misfolded proteins accumulate and aggregate in neurodegenerative disease. The existence of these deposits reflects a derangement in the protein homeostasis machinery. Using a candidate gene screen, we report that loss of RAD-23 protects against the toxicity of proteins known to aggregate in amyotrophic lateral sclerosis. Loss of RAD-23 suppresses the locomotor deficit of Caenorhabditis elegans engineered to express mutTDP-43 or mutSOD1 and also protects against aging and proteotoxic insults. Knockdown of RAD-23 is further neuroprotective against the toxicity of SOD1 and TDP-43 expression in mammalian neurons. Biochemical investigation indicates that RAD-23 modifies mutTDP-43 and mutSOD1 abundance, solubility, and turnover in association with altering the ubiquitination status of these substrates. In human amyotrophic lateral sclerosis spinal cord, we find that RAD-23 abundance is increased and RAD-23 is mislocalized within motor neurons. We propose a novel pathophysiological function for RAD-23 in the stabilization of mutated proteins that cause neurodegeneration. SIGNIFICANCE STATEMENT In this work, we identify RAD-23, a component of the protein homeostasis network and nucleotide excision repair pathway, as a modifier of the toxicity of two disease-causing, misfolding-prone proteins, SOD1 and TDP-43. Reducing the abundance of RAD-23 accelerates the degradation of mutant SOD1 and TDP-43 and reduces the cellular content of the toxic species. The existence of endogenous proteins that act as "anti-chaperones" uncovers new and general targets for therapeutic intervention.
Collapse
|
26
|
Abstract
Lysine acetylation in proteins is one of the most abundant posttranslational modifications in eukaryotic cells. The dynamic homeostasis of lysine acetylation and deacetylation is dictated by the action of histone acetyltransferases (HAT) and histone deacetylases (HDAC). Important substrates for HATs and HDACs are histones, where lysine acetylation generally leads to an open and transcriptionally active chromatin conformation. Histone deacetylation forces the compaction of the chromatin with subsequent inhibition of transcription and reduced gene expression. Unbalanced HAT and HDAC activity, and therefore aberrant histone acetylation, has been shown to be involved in tumorigenesis and progression of malignancy in different types of cancer. Therefore, the development of HDAC inhibitors (HDIs) as therapeutic agents against cancer is of great interest. However, treatment with HDIs can also affect the acetylation status of many other non-histone proteins which play a role in different pathways including angiogenesis, cell cycle progression, autophagy and apoptosis. These effects have led HDIs to become anticancer agents, which can initiate apoptosis in tumor cells. Hematological malignancies in particular are responsive to HDIs, and four HDIs have already been approved as anticancer agents. There is a strong interest in finding adequate biomarkers to predict the response to HDI treatment. This chapter provides information on how to assess HDAC activity in vitro and determine the potency of HDIs on different HDACs. It also gives information on how to analyze cellular markers following HDI treatment and to analyze tissue biopsies from HDI-treated patients. Finally, a protocol is provided on how to detect HDI sensitivity determinants in human cells, based on a pRetroSuper shRNA screen upon HDI treatment.
Collapse
Affiliation(s)
- Heidi Olzscha
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Mina E Bekheet
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Semira Sheikh
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Nicholas B La Thangue
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7DQ, UK.
| |
Collapse
|
27
|
Haase M, Fitze G. HSP90AB1: Helping the good and the bad. Gene 2015; 575:171-86. [PMID: 26358502 DOI: 10.1016/j.gene.2015.08.063] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/30/2015] [Accepted: 08/27/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Michael Haase
- Department of Pediatric Surgery, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| | - Guido Fitze
- Department of Pediatric Surgery, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|
28
|
Abstract
Autophagy is a catabolic process whereby cytosolic components and organelles are degraded to recycle key cellular materials. It is a constitutive process required for proper tissue homoeostasis but can be rapidly regulated by a variety of stimuli (for example, nutrient starvation and chemotherapeutic agents). JMY is a DNA damage-responsive p53 cofactor and actin nucleator important for cell survival and motility. Here we show that JMY regulates autophagy through its actin nucleation activity. JMY contains an LC3-interacting region, which is necessary to target JMY to the autophagosome where it enhances the autophagy maturation process. In autophagosomes, the integrity of the WH2 domains allows JMY to promote actin nucleation, which is required for efficient autophagosome formation. Thus our results establish a direct role for actin nucleation mediated by WH2 domain proteins that reside at the autophagosome. Autophagy is a catabolic process whereby cellular components are degraded by the autophagosome, but the role of the actin cytoskeleton is not clear. Here Coutts and La Thangue show that the actin nucleator JMY is recruited to the autophagosome via binding LC3, and promotes actin nucleation that is required for autophagosome maturation.
Collapse
Affiliation(s)
- Amanda S Coutts
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, off Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Nicholas B La Thangue
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, off Roosevelt Drive, Oxford OX3 7DQ, UK
| |
Collapse
|
29
|
Villaruz LC, Huang G, Romkes M, Kirkwood JM, Buch SC, Nukui T, Flaherty KT, Lee SJ, Wilson MA, Nathanson KL, Benos PV, Tawbi HA. MicroRNA expression profiling predicts clinical outcome of carboplatin/paclitaxel-based therapy in metastatic melanoma treated on the ECOG-ACRIN trial E2603. Clin Epigenetics 2015; 7:58. [PMID: 26052356 PMCID: PMC4457092 DOI: 10.1186/s13148-015-0092-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/27/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Carboplatin/paclitaxel (CP), with or without sorafenib, result in objective response rates of 18-20 % in unselected chemotherapy-naïve patients. Molecular predictors of survival and response to CP-based chemotherapy in metastatic melanoma (MM) are critical to improving the therapeutic index. Intergroup trial E2603 randomized MM patients to CP with or without sorafenib. Expression data were collected from pre-treatment formalin-fixed paraffin-embedded (FFPE) tumor tissues from 115 of 823 patients enrolled on E2603. The selected patients were balanced across treatment arms, BRAF status, and clinical outcome. We generated data using Nanostring array (microRNA (miRNA) expression) and DNA-mediated annealing, selection, extension and ligation (DASL)/Illumina microarrays (HT12 v4) (mRNA expression) with protocols optimized for FFPE samples. Integrative computational analysis was performed using a novel Tree-guided Recursive Cluster Selection (T-ReCS) [1] algorithm to select the most informative features/genes, followed by TargetScan miRNA target prediction (Human v6.2) and mirConnX [2] for network inference. RESULTS T-ReCS identified PLXNB1 as negatively associated with progression-free survival (PFS) and miR-659-3p as the primary miRNA associated positively with PFS. miR-659-3p was differentially expressed based on PFS but not based on treatment arm, BRAF or NRAS status. Dichotomized by median PFS (less vs greater than 4 months), miR-659-3p expression was significantly different. High miR-659-3p expression distinguished patients with responsive disease (complete or partial response) from patients with stable disease. miR-659-3p predicted gene targets include NFIX, which is a transcription factor known to interact with c-Jun and AP-1 in the context of developmental processes and disease. CONCLUSIONS This novel integrative analysis implicates miR-659-3p as a candidate predictive biomarker for MM patients treated with platinum-based chemotherapy and may serve to improve patient selection.
Collapse
Affiliation(s)
- Liza C Villaruz
- University of Pittsburgh Cancer Institute, Pittsburgh, PA USA
| | - Grace Huang
- University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Marjorie Romkes
- University of Pittsburgh Cancer Institute, Pittsburgh, PA USA
| | - John M Kirkwood
- University of Pittsburgh Cancer Institute, Pittsburgh, PA USA
| | - Shama C Buch
- University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Tomoko Nukui
- University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Keith T Flaherty
- University of Pennsylvania, Philadelphia, PA USA.,Massachusetts General Hospital, Boston, MA USA
| | | | - Melissa A Wilson
- University of Pennsylvania, Philadelphia, PA USA.,New York University, New York, NY USA
| | | | | | - Hussein A Tawbi
- University of Pittsburgh Cancer Institute, Pittsburgh, PA USA
| |
Collapse
|
30
|
Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov 2014; 13:673-91. [PMID: 25131830 DOI: 10.1038/nrd4360] [Citation(s) in RCA: 1171] [Impact Index Per Article: 117.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epigenetic aberrations, which are recognized as key drivers of several human diseases, are often caused by genetic defects that result in functional deregulation of epigenetic proteins, their altered expression and/or their atypical recruitment to certain gene promoters. Importantly, epigenetic changes are reversible, and epigenetic enzymes and regulatory proteins can be targeted using small molecules. This Review discusses the role of altered expression and/or function of one class of epigenetic regulators--histone deacetylases (HDACs)--and their role in cancer, neurological diseases and immune disorders. We highlight the development of small-molecule HDAC inhibitors and their use in the laboratory, in preclinical models and in the clinic.
Collapse
|
31
|
Grundy GJ, Moulding HA, Caldecott KW, Rulten SL. One ring to bring them all--the role of Ku in mammalian non-homologous end joining. DNA Repair (Amst) 2014; 17:30-8. [PMID: 24680220 DOI: 10.1016/j.dnarep.2014.02.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 02/25/2014] [Indexed: 12/26/2022]
Abstract
The repair of DNA double strand breaks is essential for cell survival and several conserved pathways have evolved to ensure their rapid and efficient repair. The non-homologous end joining pathway is initiated when Ku binds to the DNA break site. Ku is an abundant nuclear heterodimer of Ku70 and Ku80 with a toroidal structure that allows the protein to slide over the broken DNA end and bind with high affinity. Once locked into placed, Ku acts as a tool-belt to recruit multiple interacting proteins, forming one or more non-homologous end joining complexes that act in a regulated manner to ensure efficient repair of DNA ends. Here we review the structure and functions of Ku and the proteins with which it interacts during non-homologous end joining.
Collapse
Affiliation(s)
- Gabrielle J Grundy
- Genome Damage and Stability Centre, Science Park Road, Falmer, Brighton BN1 9RQ, UK.
| | - Hayley A Moulding
- School of Biochemistry, Medical Sciences, University Walk, Bristol BS8 1TD, UK
| | - Keith W Caldecott
- Genome Damage and Stability Centre, Science Park Road, Falmer, Brighton BN1 9RQ, UK.
| | - Stuart L Rulten
- Genome Damage and Stability Centre, Science Park Road, Falmer, Brighton BN1 9RQ, UK.
| |
Collapse
|
32
|
Chan SL, Yeo W. Development of systemic therapy for hepatocellular carcinoma at 2013: updates and insights. World J Gastroenterol 2014; 20:3135-45. [PMID: 24696599 PMCID: PMC3964385 DOI: 10.3748/wjg.v20.i12.3135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/23/2013] [Accepted: 01/19/2014] [Indexed: 02/06/2023] Open
Abstract
A growing number of multi-targeted tyrosine kinase inhibitor (TKI) has undergone testing for hepatocellular carcinoma (HCC). Unfortunately, this enthusiasm has recently been discouraged by a number of negative phase III studies on several anti-angiogenic TKIs in HCC. Several postulations have been made to account for this phenomenon, namely the plateau effects of anti-angiogenesis approach, the heterogeneity of HCC in terms of background hepatitis/cirrhosis and tumor biology, as well as the way how clinical trials are designed. Regardless of the underlying reasons, these results suggested that alternative strategies are necessary to further develop systemic therapy for HCC. Several new strategies are currently evaluated: for examples, molecular agents with activities against targets other than vascular endothelial growth factor receptor are being evaluated in on-going clinical trials. In addition, different approaches of targeted agents in combination with various treatment modalities, such as concurrently with another molecular agent, cytotoxic chemotherapy or transarterial chemoembolization, are being developed. This review aims to give a summary on the results of recently released clinical trials on TKIs, followed by discussion on some of the potential novel agents and combinational approaches. Future directions for testing innovative systemic agents for HCC will also be discussed.
Collapse
|
33
|
Roca H, Pande M, Huo JS, Hernandez J, Cavalcoli JD, Pienta KJ, McEachin RC. A bioinformatics approach reveals novel interactions of the OVOL transcription factors in the regulation of epithelial - mesenchymal cell reprogramming and cancer progression. BMC SYSTEMS BIOLOGY 2014; 8:29. [PMID: 24612742 PMCID: PMC4008156 DOI: 10.1186/1752-0509-8-29] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 03/03/2014] [Indexed: 01/10/2023]
Abstract
Background Mesenchymal to Epithelial Transition (MET) plasticity is critical to cancer progression, and we recently showed that the OVOL transcription factors (TFs) are critical regulators of MET. Results of that work also posed the hypothesis that the OVOLs impact MET in a range of cancers. We now test this hypothesis by developing a model, OVOL Induced MET (OI-MET), and sub-model (OI-MET-TF), to characterize differential gene expression in MET common to prostate cancer (PC) and breast cancer (BC). Results In the OI-MET model, we identified 739 genes differentially expressed in both the PC and BC models. For this gene set, we found significant enrichment of annotation for BC, PC, cancer, and MET, as well as regulation of gene expression by AP1, STAT1, STAT3, and NFKB1. Focusing on the target genes for these four TFs plus the OVOLs, we produced the OI-MET-TF sub-model, which shows even greater enrichment for these annotations, plus significant evidence of cooperation among these five TFs. Based on known gene/drug interactions, we prioritized targets in the OI-MET-TF network for follow-on analysis, emphasizing the clinical relevance of this work. Reflecting these results back to the OI-MET model, we found that binding motifs for the TF pair AP1/MYC are more frequent than expected and that the AP1/MYC pair is significantly enriched in binding in cancer models, relative to non-cancer models, in these promoters. This effect is seen in both MET models (solid tumors) and in non-MET models (leukemia). These results are consistent with our hypothesis that the OVOLs impact cancer susceptibility by regulating MET, and extend the hypothesis to include mechanisms not specific to MET. Conclusions We find significant evidence of the OVOL, AP1, STAT1, STAT3, and NFKB1 TFs having important roles in MET, and more broadly in cancer. We prioritize known gene/drug targets for follow-up in the clinic, and we show that the AP1/MYC TF pair is a strong candidate for intervention.
Collapse
Affiliation(s)
| | | | | | | | | | - Kenneth J Pienta
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| | | |
Collapse
|
34
|
Abstract
Epigenetic enzymes are often dysregulated in human tumors through mutation, altered expression, or inappropriate recruitment to certain loci. The identification of these enzymes and their partner proteins has driven the rapid development of small-molecule inhibitors that target the cancer epigenome. Herein, we discuss the influence of aberrantly regulated histone deacetylases (HDACs) in tumorigenesis. We examine HDAC inhibitors (HDACis) targeting class I, II, and IV HDACs that are currently under development for use as anticancer agents following the FDA approval of two HDACis, vorinostat and romidepsin.
Collapse
|
35
|
Anne M, Sammartino D, Barginear MF, Budman D. Profile of panobinostat and its potential for treatment in solid tumors: an update. Onco Targets Ther 2013; 6:1613-24. [PMID: 24265556 PMCID: PMC3833618 DOI: 10.2147/ott.s30773] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The histone deacetylase (HDAC) inhibitors have emerged as novel therapies for cancer. Panobinostat (LBH 589, Novartis Pharmaceuticals) is a pan-deacetylase inhibitor that is being evaluated in both intravenous and oral formulations across multiple tumor types. Comparable to the other HDACs, panobinostat leads to hyperacetylation of histones and other intracellular proteins, allowing for the expression of otherwise repressed genes, leading to inhibition of cellular proliferation and induction of apoptosis in malignant cells. Panobinostat, analogous to other HDAC inhibitors, also induces apoptosis by directly activating cellular death receptor pathways. Preclinical data suggests that panobinostat has inhibitory activity at nanomolar concentrations and appears to be the most potent clinically available HDAC inhibitor. Here we review the current status of panobinostat and discuss its role in the treatment of solid tumors.
Collapse
Affiliation(s)
- Madhurima Anne
- Monter Cancer Center, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| | - Daniel Sammartino
- Department of Medicine, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| | - Myra F Barginear
- Monter Cancer Center, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| | - Daniel Budman
- Monter Cancer Center, Hofstra North Shore-LIJ School of Medicine, Lake Success, NY, USA
| |
Collapse
|